MENU
The Electronic Scholarly Publishing Project: Providing world-wide, free access to classic scientific papers and other scholarly materials, since 1993.
More About: ESP | OUR CONTENT | THIS WEBSITE | WHAT'S NEW | WHAT'S HOT
ESP: PubMed Auto Bibliography 15 Jun 2025 at 01:51 Created:
Microbiome
It has long been known that every multicellular organism coexists with large prokaryotic ecosystems — microbiomes — that completely cover its surfaces, external and internal. Recent studies have shown that these associated microbiomes are not mere contamination, but instead have profound effects upon the function and fitness of the multicellular organism. We now know that all MCEs are actually functional composites, holobionts, composed of more prokaryotic cells than eukaryotic cells and expressing more prokaryotic genes than eukaryotic genes. A full understanding of the biology of "individual" eukaryotes will now depend on an understanding of their associated microbiomes.
Created with PubMed® Query: microbiome[tiab] NOT pmcbook NOT ispreviousversion
Citations The Papers (from PubMed®)
RevDate: 2025-06-14
CmpDate: 2025-06-14
Public Health Microbiome Curriculum: Looking Below the Tip of the Iceberg for Approaches to Population Health.
Microbial biotechnology, 18(6):e70160.
We discuss the opportunity for public health microbiome curricula to bridge the gaps in knowledge that exist between microbiome researchers and the lay public. We propose equipping public health professionals, important facilitators of public outreach and behaviour change, with three public health curriculum modules focused on breastfeeding, antibiotics and diet. These modules shift the focus from microbes as pathogens to potential partners in promoting health across the life course. Current public health messages cover only the 'tip of the iceberg' in exploring mechanisms, and this microbiome curriculum dives below the surface to provide fresh perspectives. These microbiome insights allow us to move beyond a focus on microbes as pathogens to understand the numerous collaborative roles played by the microbiome in producing health, and the upstream factors influencing the microbiome, thereby offering mechanistic insights that can be harnessed for public health education.
Additional Links: PMID-40515726
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40515726,
year = {2025},
author = {Melby, MK and Mylabathula, S and Azad, MB and Turner, S and Geva-Zatorsky, N and Tropini, C and Manus, MB and Blaser, M and Nichter, M},
title = {Public Health Microbiome Curriculum: Looking Below the Tip of the Iceberg for Approaches to Population Health.},
journal = {Microbial biotechnology},
volume = {18},
number = {6},
pages = {e70160},
doi = {10.1111/1751-7915.70160},
pmid = {40515726},
issn = {1751-7915},
support = {//Canadian Institute for Advanced Research/ ; },
mesh = {Humans ; *Public Health/education ; *Microbiota ; *Curriculum ; *Population Health ; Anti-Bacterial Agents/therapeutic use ; Breast Feeding ; },
abstract = {We discuss the opportunity for public health microbiome curricula to bridge the gaps in knowledge that exist between microbiome researchers and the lay public. We propose equipping public health professionals, important facilitators of public outreach and behaviour change, with three public health curriculum modules focused on breastfeeding, antibiotics and diet. These modules shift the focus from microbes as pathogens to potential partners in promoting health across the life course. Current public health messages cover only the 'tip of the iceberg' in exploring mechanisms, and this microbiome curriculum dives below the surface to provide fresh perspectives. These microbiome insights allow us to move beyond a focus on microbes as pathogens to understand the numerous collaborative roles played by the microbiome in producing health, and the upstream factors influencing the microbiome, thereby offering mechanistic insights that can be harnessed for public health education.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
*Public Health/education
*Microbiota
*Curriculum
*Population Health
Anti-Bacterial Agents/therapeutic use
Breast Feeding
RevDate: 2025-06-14
CmpDate: 2025-06-14
Probiotic-Based Approaches for Sustainable Control of Infectious Risk in Mass Transport: Current Data and Future Perspectives.
Microbial biotechnology, 18(6):e70177.
The built environments of high-traffic areas can play a significant role in the transmission of microorganisms and associated infections, sometimes favouring the selection of multidrug-resistant (MDR) organisms due to the excessive use of conventional disinfectants. Probiotic-based sanitation (PBS) was suggested as a novel alternative approach to control the infectious risk in crowded community environments due to its effectiveness in reducing fungal, bacterial, and viral pathogens in sanitary settings. PBS may thus trigger a paradigm shift from chemical to biological strategies in cleaning environments with high human occupancy, offering an ecological and economically sustainable alternative to conventional chemical disinfection. Providing robust data supporting the results reported so far, it has the potential to optimise bioburden control and infection prevention in mass transportation spaces. This review brings together existing research on PBS in mass transportation areas, pinpoints areas of lack of information, and explores its potential future uses, including the creation of probiotic-based materials for sustainable biocontrol in high-traffic areas.
Additional Links: PMID-40515692
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40515692,
year = {2025},
author = {Soffritti, I and D'Accolti, M and Bini, F and Mazziga, E and Volta, A and Bisi, M and Mazzacane, S and Caselli, E},
title = {Probiotic-Based Approaches for Sustainable Control of Infectious Risk in Mass Transport: Current Data and Future Perspectives.},
journal = {Microbial biotechnology},
volume = {18},
number = {6},
pages = {e70177},
doi = {10.1111/1751-7915.70177},
pmid = {40515692},
issn = {1751-7915},
mesh = {*Probiotics/administration & dosage ; Humans ; *Sanitation/methods ; *Infection Control/methods ; Transportation ; Disinfection/methods ; },
abstract = {The built environments of high-traffic areas can play a significant role in the transmission of microorganisms and associated infections, sometimes favouring the selection of multidrug-resistant (MDR) organisms due to the excessive use of conventional disinfectants. Probiotic-based sanitation (PBS) was suggested as a novel alternative approach to control the infectious risk in crowded community environments due to its effectiveness in reducing fungal, bacterial, and viral pathogens in sanitary settings. PBS may thus trigger a paradigm shift from chemical to biological strategies in cleaning environments with high human occupancy, offering an ecological and economically sustainable alternative to conventional chemical disinfection. Providing robust data supporting the results reported so far, it has the potential to optimise bioburden control and infection prevention in mass transportation spaces. This review brings together existing research on PBS in mass transportation areas, pinpoints areas of lack of information, and explores its potential future uses, including the creation of probiotic-based materials for sustainable biocontrol in high-traffic areas.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
*Probiotics/administration & dosage
Humans
*Sanitation/methods
*Infection Control/methods
Transportation
Disinfection/methods
RevDate: 2025-06-14
Iron retention coupled with trade-offs in localized symbiotic effects confers tolerance to combined iron deficiency and drought in soybean.
Journal of experimental botany pii:8162680 [Epub ahead of print].
Iron (Fe) and water availability are closely interlinked, with deficiencies in both adversely affecting soybean growth. However, the strategies employed by soybean to tolerate such conditions remain poorly understood. This study elucidates the interactions of host factors, and microbial associations using multi-omics approaches in Clark (tolerant) and Arisoy (sensitive) genotypes exposed to Fe deficiency and drought. Clark exhibited resilience to stress through sustained osmotic regulation, nutrient uptake, and photosynthetic activity, in contrast to Arisoy. Particularly, Fe retention in Clark, accompanied by the upregulation of ferritin-like proteins, may mitigate oxidative stress by reducing Fenton reactions. Furthermore, higher jasmonic and salicylic acid levels in Clark may contribute to its enhanced stress adaptation compared to Arisoy. RNA-seq analysis revealed 818 and 500 upregulated, along with 931 and 361 downregulated genes, in the roots of Clark and Arisoy, respectively, under stress. We observed the upregulation of symbiotic genes, such as Chalcone-flavonone isomerase 1 and SWEET10, accompanied by increased rhizosphere siderophore and root flavonoid in Clark. This indicates a significant role of microbes in mediating differential stress tolerance in soybean. Particularly, the combined stress led to distinct root and nodule microbiome dynamics, with Clark recruiting beneficial microbes such as Variovorax and Paecilomyces, whereas Arisoy exhibited the opposite pattern. In addition, Clark maintained nodule Bradyrhizobium and tissue nitrogen status, supported by ammonium retention and induction of Ammonium transporter 1 in the roots. Furthermore, in vitro compatibility between V. paradoxus and P. lilacinus suggests a synergistic interaction, with their localized signals benefiting Clark. Remarkably, enriched microbiomes significantly improved growth parameters, accompanied by elevated rhizosphere siderophore in sensitive genotypes under stress. This study is the first to uncover mechanisms of dual stress tolerance in soybean that may offer promising targets for breeding programs and microbiome-based biofertilizer strategies to improve combined stress tolerance in soybean and other legumes.
Additional Links: PMID-40515627
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40515627,
year = {2025},
author = {Hasan, MR and Thapa, A and Kabir, AH},
title = {Iron retention coupled with trade-offs in localized symbiotic effects confers tolerance to combined iron deficiency and drought in soybean.},
journal = {Journal of experimental botany},
volume = {},
number = {},
pages = {},
doi = {10.1093/jxb/eraf263},
pmid = {40515627},
issn = {1460-2431},
abstract = {Iron (Fe) and water availability are closely interlinked, with deficiencies in both adversely affecting soybean growth. However, the strategies employed by soybean to tolerate such conditions remain poorly understood. This study elucidates the interactions of host factors, and microbial associations using multi-omics approaches in Clark (tolerant) and Arisoy (sensitive) genotypes exposed to Fe deficiency and drought. Clark exhibited resilience to stress through sustained osmotic regulation, nutrient uptake, and photosynthetic activity, in contrast to Arisoy. Particularly, Fe retention in Clark, accompanied by the upregulation of ferritin-like proteins, may mitigate oxidative stress by reducing Fenton reactions. Furthermore, higher jasmonic and salicylic acid levels in Clark may contribute to its enhanced stress adaptation compared to Arisoy. RNA-seq analysis revealed 818 and 500 upregulated, along with 931 and 361 downregulated genes, in the roots of Clark and Arisoy, respectively, under stress. We observed the upregulation of symbiotic genes, such as Chalcone-flavonone isomerase 1 and SWEET10, accompanied by increased rhizosphere siderophore and root flavonoid in Clark. This indicates a significant role of microbes in mediating differential stress tolerance in soybean. Particularly, the combined stress led to distinct root and nodule microbiome dynamics, with Clark recruiting beneficial microbes such as Variovorax and Paecilomyces, whereas Arisoy exhibited the opposite pattern. In addition, Clark maintained nodule Bradyrhizobium and tissue nitrogen status, supported by ammonium retention and induction of Ammonium transporter 1 in the roots. Furthermore, in vitro compatibility between V. paradoxus and P. lilacinus suggests a synergistic interaction, with their localized signals benefiting Clark. Remarkably, enriched microbiomes significantly improved growth parameters, accompanied by elevated rhizosphere siderophore in sensitive genotypes under stress. This study is the first to uncover mechanisms of dual stress tolerance in soybean that may offer promising targets for breeding programs and microbiome-based biofertilizer strategies to improve combined stress tolerance in soybean and other legumes.},
}
RevDate: 2025-06-14
Microenvironmental Effects of a Non-Antibiotic Therapy for a Chronic Polymicrobial Infection Alter Microbial Physiology, Competition, and Virulence.
The ISME journal pii:8162675 [Epub ahead of print].
People with cystic fibrosis (pwCF) have reduced mucociliary clearance in their airways, leading to the build-up of thick, sticky mucus susceptible to opportunistic infection. A new treatment, comprised of three small molecule drugs called Elexacaftor/Tezacaftor/Ivacaftor (ETI), has improved mucociliary clearance and lung function in pwCF, but how this therapy alters lung infections is poorly understood. This study experimentally modeled the biochemical changes in airway mucus caused by ETI to determine its effect on the CF lung microbiome structure and function. We prepared Artificial Sputum Medium (ASM) with reduced primary carbon sources (amino acids, deoxyribonucleic acid DNA, and mucin) to mimic the effects of ETI on mucus biochemistry due to improved mucociliary clearance and reduced pulmonary inflammation. The control and modified ASM were inoculated with pure CF pathogens or mixed-species communities and then grown in oxic and anoxic conditions, followed by multi-omics data analysis. Although oxygen strongly altered the community structure, the nutrient depletions in ASM had little effect. Instead, the reduced carbon sources altered the physiology of the collective community and its individual pathogens. This included modified growth kinetics in addition to altered nitrogen and nucleotide metabolism. Under reduced amino acid concentrations, a known effect of ETI on the sputum metabolome, the production of both Pseudomonas aeruginosa's quinolones and rhamnolipids was significantly reduced. This indirect effect of ETI translates to reduced killing of competing pathogens and reduced toxicity to epithelial cells isolated from the airways of explanted human lung tissues. These findings indicate that ETI may provide further benefit to pwCF by reducing the competition and virulence of its principal pathogen and highlight how microenvironmental effects can have powerful impacts on polymicrobial infections.
Additional Links: PMID-40515620
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40515620,
year = {2025},
author = {González, CT and Martin, C and Crane, M and Gutierrez, K and Thomas, J and Remisoski, L and Okros, M and Fu, Y and Guzior, DV and Finkhouse, D and Bridges, C and Mielke, J and Querido, G and Padillo, L and Girgis, R and McClelland, M and Conrad, D and Li, X and Quinn, RA},
title = {Microenvironmental Effects of a Non-Antibiotic Therapy for a Chronic Polymicrobial Infection Alter Microbial Physiology, Competition, and Virulence.},
journal = {The ISME journal},
volume = {},
number = {},
pages = {},
doi = {10.1093/ismejo/wraf125},
pmid = {40515620},
issn = {1751-7370},
abstract = {People with cystic fibrosis (pwCF) have reduced mucociliary clearance in their airways, leading to the build-up of thick, sticky mucus susceptible to opportunistic infection. A new treatment, comprised of three small molecule drugs called Elexacaftor/Tezacaftor/Ivacaftor (ETI), has improved mucociliary clearance and lung function in pwCF, but how this therapy alters lung infections is poorly understood. This study experimentally modeled the biochemical changes in airway mucus caused by ETI to determine its effect on the CF lung microbiome structure and function. We prepared Artificial Sputum Medium (ASM) with reduced primary carbon sources (amino acids, deoxyribonucleic acid DNA, and mucin) to mimic the effects of ETI on mucus biochemistry due to improved mucociliary clearance and reduced pulmonary inflammation. The control and modified ASM were inoculated with pure CF pathogens or mixed-species communities and then grown in oxic and anoxic conditions, followed by multi-omics data analysis. Although oxygen strongly altered the community structure, the nutrient depletions in ASM had little effect. Instead, the reduced carbon sources altered the physiology of the collective community and its individual pathogens. This included modified growth kinetics in addition to altered nitrogen and nucleotide metabolism. Under reduced amino acid concentrations, a known effect of ETI on the sputum metabolome, the production of both Pseudomonas aeruginosa's quinolones and rhamnolipids was significantly reduced. This indirect effect of ETI translates to reduced killing of competing pathogens and reduced toxicity to epithelial cells isolated from the airways of explanted human lung tissues. These findings indicate that ETI may provide further benefit to pwCF by reducing the competition and virulence of its principal pathogen and highlight how microenvironmental effects can have powerful impacts on polymicrobial infections.},
}
RevDate: 2025-06-14
Ruegeria strains promote growth and morphogenesis of the giant coenocytic alga Bryopsis.
Journal of experimental botany pii:8162674 [Epub ahead of print].
An evolutionarily intriguing life form among extant organisms is the giant coenocyte, exemplified by green macroalgae in the order Bryopsidales. In these algae, cell separation does not follow nuclear division, resulting in a body composed of a single multinucleated cell. How a single cell grows to over 10 cm and undergoes characteristic morphogenesis without cell division or differentiation remains poorly understood. Macroalgae are known to associate with numerous microbes, and in some cases, these interactions influence algal cell division and differentiation. Here, we show that specific bacterial strains can promote the growth and morphogenesis of the coenocytic macroalga Bryopsis. Among >100 bacterial isolates obtained from Bryopsis, four strains belonging to the genus Ruegeria were found to accelerate the growth of the main axis and induce side-branch formation when co-cultured with the alga. The same effects were observed using conditioned seawater in which Ruegeria had been pre-cultured and subsequently removed. Seasonal microbiome analysis revealed that cultured Bryopsis associates with hundreds of bacterial species, exhibiting seasonal variations in community composition. However, Ruegeria was one of the few bacterial genera consistently associated with the cultured strain, suggesting a symbiotic relationship. Notably, although Ruegeria was not detected in Bryopsis strains isolated from other regions, its effects on growth and morphogenesis were observed in co-culture experiments. These findings suggest that Bryopsis, like multicellular macroalgae, utilises associated bacteria for growth and development without strict specificity.
Additional Links: PMID-40515619
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40515619,
year = {2025},
author = {Ochiai, KK and Goshima, G},
title = {Ruegeria strains promote growth and morphogenesis of the giant coenocytic alga Bryopsis.},
journal = {Journal of experimental botany},
volume = {},
number = {},
pages = {},
doi = {10.1093/jxb/eraf262},
pmid = {40515619},
issn = {1460-2431},
abstract = {An evolutionarily intriguing life form among extant organisms is the giant coenocyte, exemplified by green macroalgae in the order Bryopsidales. In these algae, cell separation does not follow nuclear division, resulting in a body composed of a single multinucleated cell. How a single cell grows to over 10 cm and undergoes characteristic morphogenesis without cell division or differentiation remains poorly understood. Macroalgae are known to associate with numerous microbes, and in some cases, these interactions influence algal cell division and differentiation. Here, we show that specific bacterial strains can promote the growth and morphogenesis of the coenocytic macroalga Bryopsis. Among >100 bacterial isolates obtained from Bryopsis, four strains belonging to the genus Ruegeria were found to accelerate the growth of the main axis and induce side-branch formation when co-cultured with the alga. The same effects were observed using conditioned seawater in which Ruegeria had been pre-cultured and subsequently removed. Seasonal microbiome analysis revealed that cultured Bryopsis associates with hundreds of bacterial species, exhibiting seasonal variations in community composition. However, Ruegeria was one of the few bacterial genera consistently associated with the cultured strain, suggesting a symbiotic relationship. Notably, although Ruegeria was not detected in Bryopsis strains isolated from other regions, its effects on growth and morphogenesis were observed in co-culture experiments. These findings suggest that Bryopsis, like multicellular macroalgae, utilises associated bacteria for growth and development without strict specificity.},
}
RevDate: 2025-06-14
CmpDate: 2025-06-14
Human Gut Microbes Produce EPA- and DHA-Derived Oxylipins, but not N-Acyl-Ethanolamines, From Fish Oil.
FASEB journal : official publication of the Federation of American Societies for Experimental Biology, 39(12):e70713.
We recently reported that human fecal microbiota colonized in a simulator of the human intestinal microbiome ecosystem (SHIME) process dietary oil from Buglossoides arvensis (Ahiflower), rich in the n-3 polyunsaturated fatty acid (PUFA) stearidonic acid, to the endocannabinoid (eCB)-like N-stearidonoyl-ethanolamine. Here, we assess if human fecal microbiota collected in summer and winter and inoculated in the SHIME (simulating four different intestinal sections; ileum, and ascending, transverse and descending colon) and then treated with fish oil (FO) rich in triglyceride-esterified docosahexaenoic and eicosapentaenoic acids (DHA and EPA, respectively) result in the formation of the corresponding anti-inflammatory and anticancer n-3 PUFA metabolites, including N-acylethanolamines and oxylipins. Effluents were collected every day and analyzed by LC-MS/MS for PUFA metabolites and bacterial and short-chain fatty acids (SCFA) composition during an initial 7-day treatment with vehicle and then a further 7-day treatment with FO (DHA and EPA in equal amounts, 910 mg/day each). A time-dependent increase of DHA and EPA was observed, particularly with the summer microbiota in all intestinal sections except for the duodenum, which does not contain gut microbiota. The formation of 17-hydroxy-docosahexaenoic acid from DHA, and 15-hydroxy- and 18-hydroxy-eicosapentaenoic acid from EPA was observed within the summer samples. However, no accumulation of N-docosahexaenoyl-ethanolamine or N-eicosapentaenoyl-ethanolamine, nor any other N-acyl-ethanolamines, was detected in any intestinal section and season. These data suggest that a human fecal microbiome cultivated in a SHIME processes DHA and EPA-containing triglycerides to oxylipins with known activity on the host, but not necessarily to N-acyl-ethanolamines.
Additional Links: PMID-40515551
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40515551,
year = {2025},
author = {Roussel, C and Lessard-Lord, J and Nallabelli, N and Muller, C and Flamand, N and Silvestri, C and Di Marzo, V},
title = {Human Gut Microbes Produce EPA- and DHA-Derived Oxylipins, but not N-Acyl-Ethanolamines, From Fish Oil.},
journal = {FASEB journal : official publication of the Federation of American Societies for Experimental Biology},
volume = {39},
number = {12},
pages = {e70713},
doi = {10.1096/fj.202500752RR},
pmid = {40515551},
issn = {1530-6860},
support = {//Canadian Government | Natural Sciences and Engineering Research Council of Canada (NSERC)/ ; //Canadian Government | CIHR | Institute of Nutrition, Metabolism and Diabetes (INMD)/ ; //Canadian Government | Social Sciences and Humanities Research Council of Canada (SSHRC)/ ; },
mesh = {Humans ; *Eicosapentaenoic Acid/metabolism ; *Docosahexaenoic Acids/metabolism ; *Gastrointestinal Microbiome/physiology ; *Fish Oils/metabolism ; *Ethanolamines/metabolism ; *Oxylipins/metabolism ; Feces/microbiology ; Male ; Female ; },
abstract = {We recently reported that human fecal microbiota colonized in a simulator of the human intestinal microbiome ecosystem (SHIME) process dietary oil from Buglossoides arvensis (Ahiflower), rich in the n-3 polyunsaturated fatty acid (PUFA) stearidonic acid, to the endocannabinoid (eCB)-like N-stearidonoyl-ethanolamine. Here, we assess if human fecal microbiota collected in summer and winter and inoculated in the SHIME (simulating four different intestinal sections; ileum, and ascending, transverse and descending colon) and then treated with fish oil (FO) rich in triglyceride-esterified docosahexaenoic and eicosapentaenoic acids (DHA and EPA, respectively) result in the formation of the corresponding anti-inflammatory and anticancer n-3 PUFA metabolites, including N-acylethanolamines and oxylipins. Effluents were collected every day and analyzed by LC-MS/MS for PUFA metabolites and bacterial and short-chain fatty acids (SCFA) composition during an initial 7-day treatment with vehicle and then a further 7-day treatment with FO (DHA and EPA in equal amounts, 910 mg/day each). A time-dependent increase of DHA and EPA was observed, particularly with the summer microbiota in all intestinal sections except for the duodenum, which does not contain gut microbiota. The formation of 17-hydroxy-docosahexaenoic acid from DHA, and 15-hydroxy- and 18-hydroxy-eicosapentaenoic acid from EPA was observed within the summer samples. However, no accumulation of N-docosahexaenoyl-ethanolamine or N-eicosapentaenoyl-ethanolamine, nor any other N-acyl-ethanolamines, was detected in any intestinal section and season. These data suggest that a human fecal microbiome cultivated in a SHIME processes DHA and EPA-containing triglycerides to oxylipins with known activity on the host, but not necessarily to N-acyl-ethanolamines.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
*Eicosapentaenoic Acid/metabolism
*Docosahexaenoic Acids/metabolism
*Gastrointestinal Microbiome/physiology
*Fish Oils/metabolism
*Ethanolamines/metabolism
*Oxylipins/metabolism
Feces/microbiology
Male
Female
RevDate: 2025-06-14
Review: Human Intestinal Barrier-Optimal Measurement and Effects of Diet in the Absence of Overt Inflammation or Ulceration.
Alimentary pharmacology & therapeutics [Epub ahead of print].
BACKGROUND: The intestinal barrier protects humans from potentially deleterious ingested material. It consists of several components: commensal organisms, mucus, and transepithelial pathways, and immune functions.
AIMS: To review, with major focus on human studies, the methods to measure intestinal barrier function in vivo and the impaired intestinal barrier function in non-inflammatory conditions, and the deleterious or beneficial effects of dietary components on the intestinal barrier.
METHODS: PUBMED literature search was intentionally focused, when possible, on human studies conducted in vivo.
RESULTS: Although many gastrointestinal, rheumatological, and degenerative neurological diseases are attributed to impaired intestinal barrier function, often termed "leaky gut," methods of accurate measurement of intestinal barrier function in vivo in humans are still being developed. In vivo measurements provide an overall assessment of barrier function at a whole organ level, whereas ex vivo or in vitro measurements using mucosal biopsies address mechanistic information at the cellular level. Several dietary components are detrimental to the barrier, including ethanol, fat, sugars, gliadin, food additives, emulsifiers, and microbial transglutaminase. Conversely, dietary components improving barrier function include fibre and metabolites such as short-chain fatty acids, anthocyanins, polyphenols, vitamins (such as A and D), zinc, specific amino acids (such as glutamine) and probiotics.
CONCLUSIONS: Currently, data are not exclusively from human studies, and research is needed to corroborate observations in animals or further validate in humans. There are several practical dietary approaches that can be instituted for restoration of the intestinal barrier in humans.
Additional Links: PMID-40515459
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40515459,
year = {2025},
author = {Camilleri, M},
title = {Review: Human Intestinal Barrier-Optimal Measurement and Effects of Diet in the Absence of Overt Inflammation or Ulceration.},
journal = {Alimentary pharmacology & therapeutics},
volume = {},
number = {},
pages = {},
doi = {10.1111/apt.70225},
pmid = {40515459},
issn = {1365-2036},
abstract = {BACKGROUND: The intestinal barrier protects humans from potentially deleterious ingested material. It consists of several components: commensal organisms, mucus, and transepithelial pathways, and immune functions.
AIMS: To review, with major focus on human studies, the methods to measure intestinal barrier function in vivo and the impaired intestinal barrier function in non-inflammatory conditions, and the deleterious or beneficial effects of dietary components on the intestinal barrier.
METHODS: PUBMED literature search was intentionally focused, when possible, on human studies conducted in vivo.
RESULTS: Although many gastrointestinal, rheumatological, and degenerative neurological diseases are attributed to impaired intestinal barrier function, often termed "leaky gut," methods of accurate measurement of intestinal barrier function in vivo in humans are still being developed. In vivo measurements provide an overall assessment of barrier function at a whole organ level, whereas ex vivo or in vitro measurements using mucosal biopsies address mechanistic information at the cellular level. Several dietary components are detrimental to the barrier, including ethanol, fat, sugars, gliadin, food additives, emulsifiers, and microbial transglutaminase. Conversely, dietary components improving barrier function include fibre and metabolites such as short-chain fatty acids, anthocyanins, polyphenols, vitamins (such as A and D), zinc, specific amino acids (such as glutamine) and probiotics.
CONCLUSIONS: Currently, data are not exclusively from human studies, and research is needed to corroborate observations in animals or further validate in humans. There are several practical dietary approaches that can be instituted for restoration of the intestinal barrier in humans.},
}
RevDate: 2025-06-13
Composition and rhythmic variations in the microbiome of Southwestern Atlantic corals.
Environmental microbiome, 20(1):71.
BACKGROUND: Diel and tidal rhythms can regulate the metabolism, physiology, behavior, and gene expression patterns of different organisms, with evidence of an integration on the circadian behavior of host species and their microbial community. Corals host a diverse and dynamic microbial community, with variable diversity and abundance across geographic and temporal scales. Within scleractinian corals, those that host endosymbiotic algae (i.e., zooxanthellate) display a diel variation in the oxygen levels, an oscillation in their internal environment that has the potential to influence its microbiome abundance and/or composition. Here we investigate in situ daily fluctuations on the microbial community of two zooxanthellate (Madracis decactis and Mussismilia hispida) and two azooxanthellate coral species (Tubastraea coccinea and T. tagusensis) along a 72-hour period.
RESULTS: Day and night alpha diversity values were similar for all species, with Ma. decactis hosting a significantly more diverse community. Similarly, there was no fluctuation in the microbiome composition at the Amplicon Sequence Variants (ASV) level between day and night within species, but all species were significantly different from each other. Interestingly, Mu. hispida, an endemic species to the Southwestern Atlantic, had a high proportion of unidentified microbial taxa at genus level, suggesting a species-specific microbiome community composed by unidentified taxa. Significant rhythmicity in the abundance of individual ASVs was observed for one ASV (genus Pseudoalteromonas) in T. tagusensis and one (genus Woeseia) in Ma. decactis, with 24 and 12-hour fluctuations, respectively. In addition, DESeq2 recovered 13 ASVs (four in Ma. decactis, two in Mu. hispida, six in T. coccinea, and one in T. tagusensis) with different abundances between day and night.
CONCLUSIONS: Results show divergent microbial communities when comparing zooxanthellate and azooxanthellate species, with few significant changes within a 24-hour period. Future studies should focus on metabolic pathways to better understand how the microbiome community can adjust to environmental changes within the coral host in short time scales.
Additional Links: PMID-40514749
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40514749,
year = {2025},
author = {Seiblitz, IGL and Capel, KCC and Oliveira, RR and Zilberberg, C and Cordeiro, CAMM and Francini, CLB and Zanotti, AA and Ciotti, AM and Kitahara, MV},
title = {Composition and rhythmic variations in the microbiome of Southwestern Atlantic corals.},
journal = {Environmental microbiome},
volume = {20},
number = {1},
pages = {71},
pmid = {40514749},
issn = {2524-6372},
support = {Finance Code 001//Coordenação de Aperfeiçoamento de Pessoal de Nível Superior- Brasil (CAPES)/ ; 2022/09789-5//Fundação de Amparo à Pesquisa do Estado de São Paulo/ ; 2020/15945-4//Fundação de Amparo à Pesquisa do Estado de São Paulo/ ; 2021/06866-6//Fundação de Amparo à Pesquisa do Estado de São Paulo/ ; 305274/2021-0//Conselho Nacional de Desenvolvimento Científico e Tecnológico/ ; },
abstract = {BACKGROUND: Diel and tidal rhythms can regulate the metabolism, physiology, behavior, and gene expression patterns of different organisms, with evidence of an integration on the circadian behavior of host species and their microbial community. Corals host a diverse and dynamic microbial community, with variable diversity and abundance across geographic and temporal scales. Within scleractinian corals, those that host endosymbiotic algae (i.e., zooxanthellate) display a diel variation in the oxygen levels, an oscillation in their internal environment that has the potential to influence its microbiome abundance and/or composition. Here we investigate in situ daily fluctuations on the microbial community of two zooxanthellate (Madracis decactis and Mussismilia hispida) and two azooxanthellate coral species (Tubastraea coccinea and T. tagusensis) along a 72-hour period.
RESULTS: Day and night alpha diversity values were similar for all species, with Ma. decactis hosting a significantly more diverse community. Similarly, there was no fluctuation in the microbiome composition at the Amplicon Sequence Variants (ASV) level between day and night within species, but all species were significantly different from each other. Interestingly, Mu. hispida, an endemic species to the Southwestern Atlantic, had a high proportion of unidentified microbial taxa at genus level, suggesting a species-specific microbiome community composed by unidentified taxa. Significant rhythmicity in the abundance of individual ASVs was observed for one ASV (genus Pseudoalteromonas) in T. tagusensis and one (genus Woeseia) in Ma. decactis, with 24 and 12-hour fluctuations, respectively. In addition, DESeq2 recovered 13 ASVs (four in Ma. decactis, two in Mu. hispida, six in T. coccinea, and one in T. tagusensis) with different abundances between day and night.
CONCLUSIONS: Results show divergent microbial communities when comparing zooxanthellate and azooxanthellate species, with few significant changes within a 24-hour period. Future studies should focus on metabolic pathways to better understand how the microbiome community can adjust to environmental changes within the coral host in short time scales.},
}
RevDate: 2025-06-13
Microbiome dynamics associated with Hematodinium sp. infection in Norway lobster (Nephrops norvegicus).
Animal microbiome, 7(1):62.
BACKGROUND: The parasite Hematodinium sp. causes morbidity and seasonal mortality events in more than 40 decapod species globally and therefore, it is now recognised as a significant threat to the future sustainability of shellfish fisheries and aquaculture worldwide. Among these, Norway lobster (Nephrops norvegicus), an important representative of the marine benthos and supporting the most valuable shellfish fishery in the UK, experience yearly seasonal Hematodinium sp. patent infections. Currently, little is known about the N. norvegicus microbiome and potential role during Hematodinium sp.
INFECTION: Therefore, in this study we investigated the microbiome dynamics of N. norvegicus associated with Hematodinium sp. infection and disease progression in the haemolymph and gut. N. norvegicus were sampled from the Clyde Sea Area, Scotland during the peak of the Hematodinium sp. patent infection. The presence and intensity of Hematodinium sp. infection were determined using the body colour method (BCM), pleopod method (PM), histology (heart, gonads, hepatopancreas, gills and muscle) and molecular tools (PCR).
RESULTS: Marked shifts in the bacterial richness of the haemolymph and significant alterations in the overall bacterial community composition of both tissues were observed in infected lobsters. These changes, observed even at subpatent levels of infection (only positive by PCR), indicate a prompt and persistent microbiome shift associated with Hematodinium sp.
INFECTION: Furthermore, smaller healthy animals (25.2 ± 1.20 mm CL) known to be particularly susceptible to high severity infection displayed a decreased microbiome richness in the haemolymph suggesting a potential link between the host microbiome and susceptibility to disease progression, a possibility that merits further research.
CONCLUSIONS: This study offers the first insights into the pathobiome of N. norvegicus due to Hematodinium sp. infection and disease that in turn provides a foundation for further studies on the pathogenesis of this important parasitic disease.
Additional Links: PMID-40514740
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40514740,
year = {2025},
author = {Martin, I and Elsheshtawy, A and Clokie, BGJ and MacKenzie, S and Bateman, KS and Bass, D and Stentiford, GD and Albalat, A},
title = {Microbiome dynamics associated with Hematodinium sp. infection in Norway lobster (Nephrops norvegicus).},
journal = {Animal microbiome},
volume = {7},
number = {1},
pages = {62},
pmid = {40514740},
issn = {2524-4671},
abstract = {BACKGROUND: The parasite Hematodinium sp. causes morbidity and seasonal mortality events in more than 40 decapod species globally and therefore, it is now recognised as a significant threat to the future sustainability of shellfish fisheries and aquaculture worldwide. Among these, Norway lobster (Nephrops norvegicus), an important representative of the marine benthos and supporting the most valuable shellfish fishery in the UK, experience yearly seasonal Hematodinium sp. patent infections. Currently, little is known about the N. norvegicus microbiome and potential role during Hematodinium sp.
INFECTION: Therefore, in this study we investigated the microbiome dynamics of N. norvegicus associated with Hematodinium sp. infection and disease progression in the haemolymph and gut. N. norvegicus were sampled from the Clyde Sea Area, Scotland during the peak of the Hematodinium sp. patent infection. The presence and intensity of Hematodinium sp. infection were determined using the body colour method (BCM), pleopod method (PM), histology (heart, gonads, hepatopancreas, gills and muscle) and molecular tools (PCR).
RESULTS: Marked shifts in the bacterial richness of the haemolymph and significant alterations in the overall bacterial community composition of both tissues were observed in infected lobsters. These changes, observed even at subpatent levels of infection (only positive by PCR), indicate a prompt and persistent microbiome shift associated with Hematodinium sp.
INFECTION: Furthermore, smaller healthy animals (25.2 ± 1.20 mm CL) known to be particularly susceptible to high severity infection displayed a decreased microbiome richness in the haemolymph suggesting a potential link between the host microbiome and susceptibility to disease progression, a possibility that merits further research.
CONCLUSIONS: This study offers the first insights into the pathobiome of N. norvegicus due to Hematodinium sp. infection and disease that in turn provides a foundation for further studies on the pathogenesis of this important parasitic disease.},
}
RevDate: 2025-06-13
Antarctic ice-free terrestrial microbial functional redundancy in core ecological functions and microhabitat-specific microbial taxa and adaptive strategy.
Environmental microbiome, 20(1):70.
BACKGROUND: Although ice-free terrestrial ecosystems in Antarctica cover only limited areas, they harbor diverse and metabolically active microbial communities. These ecosystems encompass distinct microhabitats such as mosses, lichens, and soils, each offering unique ecological niches. However, how different microbial taxa respond to microhabitat heterogeneity, ecological strategies such as functional redundancy and specialization contribute to adaptation in extreme environments remains underexplored. To address these questions, we employed high-throughput 16 S rRNA gene and ITS sequencing in combination with GeoChip-based functional gene profiling to assess the structure and functional potential of microbial communities across moss, lichen and soil microhabitats in Antarctic ice-free terrestrial ecosystem.
RESULTS: Microhabitat type has a greater influence on microbial community structure and function in the ice-free Antarctic terrestrial ecosystem than geographical location. Though all prokaryotic communities were dominated by Pseudomonadota, Nostoc and Endobacter were significantly enriched in the moss and lichen microhabitats, respectively. Meanwhile, all fungal communities were primarily dominated by Ascomycota and Basidiomycota, with Byssoloma and Usnea showing significant enrichment in the moss and lichen microhabitats, respectively. Despite these taxonomic differences, the three microhabitats show similar core ecological functions with widespread microbial functional redundancy. Nevertheless, clear microhabitat-specific functional specialization was suggested. For example, moss microhabitat was enriched in Pyoverdin_pvcC and Zeaxanthin_glucosyltransferase, sdhA, lichen microhabitat harbored higher levels of nhaA, nikC, vacuolar_iron_transport, mttB, glucoamylase, pel_Cdeg, pme_Cdeg, rgh, rgl, while soil microhabitat was enriched in 5f1_ppn and isopullulanase. Notably, genes involved in carotenoid biosynthesis were significantly more abundant in moss and lichen microhabitats than in soil microhabitat, indicating the adaptive capacity of symbiotic microorganisms to mitigate ultraviolet radiation and oxidative stress to protect their hosts.
CONCLUSIONS: Microbial communities associated with distinct microhabitats (i.e. mosses, lichens, and soils) in Antarctic ice-free terrestrial ecosystem exhibit both functional redundancy in core ecological functions and microhabitat-specific specialization in key microbial taxa and adaptive strategy.
Additional Links: PMID-40514722
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40514722,
year = {2025},
author = {Zhang, M and Xiao, Y and Song, Q and Li, Z},
title = {Antarctic ice-free terrestrial microbial functional redundancy in core ecological functions and microhabitat-specific microbial taxa and adaptive strategy.},
journal = {Environmental microbiome},
volume = {20},
number = {1},
pages = {70},
pmid = {40514722},
issn = {2524-6372},
support = {2022YFC280410//National Key Research and Development Program of China/ ; },
abstract = {BACKGROUND: Although ice-free terrestrial ecosystems in Antarctica cover only limited areas, they harbor diverse and metabolically active microbial communities. These ecosystems encompass distinct microhabitats such as mosses, lichens, and soils, each offering unique ecological niches. However, how different microbial taxa respond to microhabitat heterogeneity, ecological strategies such as functional redundancy and specialization contribute to adaptation in extreme environments remains underexplored. To address these questions, we employed high-throughput 16 S rRNA gene and ITS sequencing in combination with GeoChip-based functional gene profiling to assess the structure and functional potential of microbial communities across moss, lichen and soil microhabitats in Antarctic ice-free terrestrial ecosystem.
RESULTS: Microhabitat type has a greater influence on microbial community structure and function in the ice-free Antarctic terrestrial ecosystem than geographical location. Though all prokaryotic communities were dominated by Pseudomonadota, Nostoc and Endobacter were significantly enriched in the moss and lichen microhabitats, respectively. Meanwhile, all fungal communities were primarily dominated by Ascomycota and Basidiomycota, with Byssoloma and Usnea showing significant enrichment in the moss and lichen microhabitats, respectively. Despite these taxonomic differences, the three microhabitats show similar core ecological functions with widespread microbial functional redundancy. Nevertheless, clear microhabitat-specific functional specialization was suggested. For example, moss microhabitat was enriched in Pyoverdin_pvcC and Zeaxanthin_glucosyltransferase, sdhA, lichen microhabitat harbored higher levels of nhaA, nikC, vacuolar_iron_transport, mttB, glucoamylase, pel_Cdeg, pme_Cdeg, rgh, rgl, while soil microhabitat was enriched in 5f1_ppn and isopullulanase. Notably, genes involved in carotenoid biosynthesis were significantly more abundant in moss and lichen microhabitats than in soil microhabitat, indicating the adaptive capacity of symbiotic microorganisms to mitigate ultraviolet radiation and oxidative stress to protect their hosts.
CONCLUSIONS: Microbial communities associated with distinct microhabitats (i.e. mosses, lichens, and soils) in Antarctic ice-free terrestrial ecosystem exhibit both functional redundancy in core ecological functions and microhabitat-specific specialization in key microbial taxa and adaptive strategy.},
}
RevDate: 2025-06-13
Peripheral Immune-Inflammatory Pathways in Major Depressive Disorder, Bipolar Disorder, and Schizophrenia: Exploring Their Potential as Treatment Targets.
CNS drugs [Epub ahead of print].
Major depressive disorder (MDD), bipolar disorder (BD), and schizophrenia (SCZ) are major mental disorders linked to substantial morbidity. Traditional monoamine-based pharmacotherapies frequently produce inadequate outcomes for many patients. The elevated levels of treatment resistance require the exploration of new pharmacological targets. Evidence indicates that peripheral immune-inflammatory dysregulation, characterized by an imbalance between immunological responses and compensatory immune-regulatory systems (IRS/CIRS), together with increased oxidative and nitrosative stress (O&NS), significantly contributes to the pathogenesis of these disorders. This review examines IRS/CIRS/O&NS pathways as new drug targets and highlights novel pharmacological trials. Antiinflammatory drugs have been repurposed as augmentation strategies for the treatment of MDD/BD and SCZ, including nonsteroidal antiinflammatory medications, such as cyclooxygenase-2 (COX-2) inhibitors; cytokine-targeting biologics, such as tumor necrosis factor-α monoclonal antibodies; and minocycline, an antibiotic that attenuates neuroinflammation. N-acetylcysteine, curcumin, and omega-3 polyunsaturated fatty acids demonstrate some efficacy as augmentation therapies in MDD, likely by diminishing IRS activation and O&NS. Strategies aimed at the gut-brain axis and gut dysbiosis, including fecal microbiota transplantation, are under investigation for their capacity to restore immunological homeostasis by improving gut barrier integrity and microbiome composition. This review examines new potential therapeutic targets arising from recent discoveries in neuro-immune interactions and oxidative stress, including particular lymphocyte surface markers, the CIRS, and intracellular network molecules in both affective and psychotic disorders. The evidence underscores the clinical importance of immune-targeted augmentation treatments in psychiatric disorders and supports the ongoing development of these novel pharmacotherapies within a precision medicine paradigm.
Additional Links: PMID-40514640
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40514640,
year = {2025},
author = {Almulla, AF and Maes, M},
title = {Peripheral Immune-Inflammatory Pathways in Major Depressive Disorder, Bipolar Disorder, and Schizophrenia: Exploring Their Potential as Treatment Targets.},
journal = {CNS drugs},
volume = {},
number = {},
pages = {},
pmid = {40514640},
issn = {1179-1934},
support = {RA66/016//FF66 grant and a Sompoch Endowment Fund (Faculty of Medicine), MDCU/ ; BG-RRP-2.004-0007-С01//Strategic Research and Innovation Program for the Development of MU - PLOVDIV-(SRIPD-MUP)", Creation of a network of research higher schools, National plan for recovery and sustainability, European Union - NextGenerationEU/ ; },
abstract = {Major depressive disorder (MDD), bipolar disorder (BD), and schizophrenia (SCZ) are major mental disorders linked to substantial morbidity. Traditional monoamine-based pharmacotherapies frequently produce inadequate outcomes for many patients. The elevated levels of treatment resistance require the exploration of new pharmacological targets. Evidence indicates that peripheral immune-inflammatory dysregulation, characterized by an imbalance between immunological responses and compensatory immune-regulatory systems (IRS/CIRS), together with increased oxidative and nitrosative stress (O&NS), significantly contributes to the pathogenesis of these disorders. This review examines IRS/CIRS/O&NS pathways as new drug targets and highlights novel pharmacological trials. Antiinflammatory drugs have been repurposed as augmentation strategies for the treatment of MDD/BD and SCZ, including nonsteroidal antiinflammatory medications, such as cyclooxygenase-2 (COX-2) inhibitors; cytokine-targeting biologics, such as tumor necrosis factor-α monoclonal antibodies; and minocycline, an antibiotic that attenuates neuroinflammation. N-acetylcysteine, curcumin, and omega-3 polyunsaturated fatty acids demonstrate some efficacy as augmentation therapies in MDD, likely by diminishing IRS activation and O&NS. Strategies aimed at the gut-brain axis and gut dysbiosis, including fecal microbiota transplantation, are under investigation for their capacity to restore immunological homeostasis by improving gut barrier integrity and microbiome composition. This review examines new potential therapeutic targets arising from recent discoveries in neuro-immune interactions and oxidative stress, including particular lymphocyte surface markers, the CIRS, and intracellular network molecules in both affective and psychotic disorders. The evidence underscores the clinical importance of immune-targeted augmentation treatments in psychiatric disorders and supports the ongoing development of these novel pharmacotherapies within a precision medicine paradigm.},
}
RevDate: 2025-06-13
Murine maternal microbiome modifies adverse effects of protein undernutrition on offspring neurobehaviour.
Nature microbiology [Epub ahead of print].
Protein undernutrition results in impaired growth and neurobehavioural development in children. However, the impact of timing, environmental factors and maternal versus neonatal influences are unclear. Here, using a mouse model of fetal growth restriction where maternal protein intake is limited during pregnancy, we show that adult offspring exhibit cognitive and anxiety-like behavioural abnormalities. Cross-fostering newborn mice to dams previously exposed to either low protein or standard diet reveals that behavioural impairments in adult offspring require diet-induced conditioning of both fetal development and maternal peripartum physiology. Maternal gut microbiome diversity is reduced, maternal immune, milk, and serum metabolomic profiles are altered, and widespread changes in fetal brain transcriptomic and metabolomic profiles are observed, including subsets of microbiome-dependent metabolites. Finally, we show that dam treatment with a cocktail of ten diet- and microbiome-dependent metabolites results in differential effects on fetal development and postnatal behaviour. Our study highlights the impact of prenatal maternal protein undernutrition on offspring neurobehavioural trajectories and the role of the maternal microbiome.
Additional Links: PMID-40514563
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40514563,
year = {2025},
author = {Coley-O'Rourke, EJ and Lum, GR and Pronovost, GN and Yu, LW and Özcan, E and Yu, KB and McDermott, J and Chakhoyan, A and Goldman, E and Vuong, HE and Paramo, J and McCune, S and Sejane, K and Renwick, S and Bode, L and Chu, A and Calkins, KL and Hsiao, EY},
title = {Murine maternal microbiome modifies adverse effects of protein undernutrition on offspring neurobehaviour.},
journal = {Nature microbiology},
volume = {},
number = {},
pages = {},
pmid = {40514563},
issn = {2058-5276},
support = {1R01HD111079//U.S. Department of Health & Human Services | NIH | Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD)/ ; Graduate Research Fellowship//National Science Foundation (NSF)/ ; },
abstract = {Protein undernutrition results in impaired growth and neurobehavioural development in children. However, the impact of timing, environmental factors and maternal versus neonatal influences are unclear. Here, using a mouse model of fetal growth restriction where maternal protein intake is limited during pregnancy, we show that adult offspring exhibit cognitive and anxiety-like behavioural abnormalities. Cross-fostering newborn mice to dams previously exposed to either low protein or standard diet reveals that behavioural impairments in adult offspring require diet-induced conditioning of both fetal development and maternal peripartum physiology. Maternal gut microbiome diversity is reduced, maternal immune, milk, and serum metabolomic profiles are altered, and widespread changes in fetal brain transcriptomic and metabolomic profiles are observed, including subsets of microbiome-dependent metabolites. Finally, we show that dam treatment with a cocktail of ten diet- and microbiome-dependent metabolites results in differential effects on fetal development and postnatal behaviour. Our study highlights the impact of prenatal maternal protein undernutrition on offspring neurobehavioural trajectories and the role of the maternal microbiome.},
}
RevDate: 2025-06-13
CmpDate: 2025-06-13
Mucosal immune responses and intestinal microbiome associations in wild spotted hyenas (Crocuta crocuta).
Communications biology, 8(1):924.
Little is known about host-gut microbiome interactions within natural populations at the intestinal mucosa, the primary interface. We investigate associations between the intestinal microbiome and mucosal immune measures while controlling for host, social and ecological factors in 199 samples of 158 wild spotted hyenas (Crocuta crocuta) in the Serengeti National Park, Tanzania. We profile the microbiome composition using a multi-amplicon approach and measure faecal immunoglobulin A and mucin. Probabilistic models indicate that both immune measures predicted microbiome similarity among individuals in an age-dependent manner. These associations are the strongest within bacteria, intermediate within parasites, and weakest within fungi communities. Machine learning models accurately predicted both immune measures and identify the taxa driving these associations: symbiotic bacteria reported in humans and laboratory mice, unclassified bacteria, parasitic hookworms and fungi. These findings improve our understanding of the gut microbiome, its drivers, and interactions in wild populations under natural selection.
Additional Links: PMID-40514454
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40514454,
year = {2025},
author = {Veloso Soares, SP and Jarquín-Díaz, VH and Veiga, MM and Karl, S and Czirják, GÁ and Weyrich, A and Metzger, S and East, ML and Hofer, H and Heitlinger, E and Benhaiem, S and Ferreira, SCM},
title = {Mucosal immune responses and intestinal microbiome associations in wild spotted hyenas (Crocuta crocuta).},
journal = {Communications biology},
volume = {8},
number = {1},
pages = {924},
pmid = {40514454},
issn = {2399-3642},
support = {GRAKO 2046//Deutsche Forschungsgemeinschaft (German Research Foundation)/ ; DFG-Grako 2046//Deutsche Forschungsgemeinschaft (German Research Foundation)/ ; DFG-Grako 2046//Deutsche Forschungsgemeinschaft (German Research Foundation)/ ; GRK 2046//Deutsche Forschungsgemeinschaft (German Research Foundation)/ ; GRK 2046//Deutsche Forschungsgemeinschaft (German Research Foundation)/ ; GRK 2046//Deutsche Forschungsgemeinschaft (German Research Foundation)/ ; GRK 2046//Deutsche Forschungsgemeinschaft (German Research Foundation)/ ; GRK 2046//Deutsche Forschungsgemeinschaft (German Research Foundation)/ ; },
mesh = {Animals ; *Gastrointestinal Microbiome/immunology ; *Hyaenidae/microbiology/immunology ; *Immunity, Mucosal ; Feces/microbiology/chemistry ; Female ; Male ; Tanzania ; Bacteria/classification/genetics ; },
abstract = {Little is known about host-gut microbiome interactions within natural populations at the intestinal mucosa, the primary interface. We investigate associations between the intestinal microbiome and mucosal immune measures while controlling for host, social and ecological factors in 199 samples of 158 wild spotted hyenas (Crocuta crocuta) in the Serengeti National Park, Tanzania. We profile the microbiome composition using a multi-amplicon approach and measure faecal immunoglobulin A and mucin. Probabilistic models indicate that both immune measures predicted microbiome similarity among individuals in an age-dependent manner. These associations are the strongest within bacteria, intermediate within parasites, and weakest within fungi communities. Machine learning models accurately predicted both immune measures and identify the taxa driving these associations: symbiotic bacteria reported in humans and laboratory mice, unclassified bacteria, parasitic hookworms and fungi. These findings improve our understanding of the gut microbiome, its drivers, and interactions in wild populations under natural selection.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Animals
*Gastrointestinal Microbiome/immunology
*Hyaenidae/microbiology/immunology
*Immunity, Mucosal
Feces/microbiology/chemistry
Female
Male
Tanzania
Bacteria/classification/genetics
RevDate: 2025-06-13
CmpDate: 2025-06-13
Di (2-ethylhexyl) Phthalate decrease pregnancy rate via disrupting the microbe-gut-hypothalamic-pituitary-ovarian axis in mice.
NPJ biofilms and microbiomes, 11(1):107.
Di (2-ethylhexyl) Phthalate (DEHP), a widely used plasticizer and endocrine disruptor, poses risks to human health, particularly reproductive function. Using a mouse model, we investigated how DEHP exposure impacts the hypothalamic-pituitary-ovarian (HPO) axis through gut microbiome disruption. DEHP decreased pregnancy rates by impairing ovarian function, activating hypothalamic astrocytes, and increasing neuregulin 1 (NRG1) expression. NRG1 binding to astrocyte ErbB2 receptors elevated prostaglandin E2 (PGE2) and gonadotropin-releasing hormone (GnRH), disrupting HPO axis homeostasis. Additionally, DEHP altered gut microbiota, destabilized microbial networks, and impacted β-glucuronidase-related taxa, leading to hormone fluctuations and reduced fertility. This study highlights gut microbiome perturbations as a novel mechanism linking DEHP exposure to reproductive dysfunction. Our study provides novel insights concerning perturbations of the gut microbiome and HPO axis and their functions as a potential new mechanism by which DEHP exposes interferes with the reproductive function-related human health.
Additional Links: PMID-40514374
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40514374,
year = {2025},
author = {Xu, L and Yan, J and Yin, T and Pan, Y and Chen, M and Wang, X and Wu, L and Ding, H},
title = {Di (2-ethylhexyl) Phthalate decrease pregnancy rate via disrupting the microbe-gut-hypothalamic-pituitary-ovarian axis in mice.},
journal = {NPJ biofilms and microbiomes},
volume = {11},
number = {1},
pages = {107},
pmid = {40514374},
issn = {2055-5008},
support = {BK20230153//Natural Science Foundation of Jiangsu Province, China/ ; ZKX14035//Nanjing Medical Science and Technique Development Foundation/ ; },
mesh = {Animals ; Female ; *Gastrointestinal Microbiome/drug effects ; *Diethylhexyl Phthalate/toxicity ; Mice ; Pregnancy ; *Ovary/drug effects ; *Hypothalamo-Hypophyseal System/drug effects ; Gonadotropin-Releasing Hormone/metabolism ; Endocrine Disruptors/toxicity ; Astrocytes/drug effects/metabolism ; Bacteria/classification/genetics/drug effects ; },
abstract = {Di (2-ethylhexyl) Phthalate (DEHP), a widely used plasticizer and endocrine disruptor, poses risks to human health, particularly reproductive function. Using a mouse model, we investigated how DEHP exposure impacts the hypothalamic-pituitary-ovarian (HPO) axis through gut microbiome disruption. DEHP decreased pregnancy rates by impairing ovarian function, activating hypothalamic astrocytes, and increasing neuregulin 1 (NRG1) expression. NRG1 binding to astrocyte ErbB2 receptors elevated prostaglandin E2 (PGE2) and gonadotropin-releasing hormone (GnRH), disrupting HPO axis homeostasis. Additionally, DEHP altered gut microbiota, destabilized microbial networks, and impacted β-glucuronidase-related taxa, leading to hormone fluctuations and reduced fertility. This study highlights gut microbiome perturbations as a novel mechanism linking DEHP exposure to reproductive dysfunction. Our study provides novel insights concerning perturbations of the gut microbiome and HPO axis and their functions as a potential new mechanism by which DEHP exposes interferes with the reproductive function-related human health.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Animals
Female
*Gastrointestinal Microbiome/drug effects
*Diethylhexyl Phthalate/toxicity
Mice
Pregnancy
*Ovary/drug effects
*Hypothalamo-Hypophyseal System/drug effects
Gonadotropin-Releasing Hormone/metabolism
Endocrine Disruptors/toxicity
Astrocytes/drug effects/metabolism
Bacteria/classification/genetics/drug effects
RevDate: 2025-06-13
CmpDate: 2025-06-13
A novel approach to finding the compositional differences and biomarkers in gut microbiota in type 2 diabetic patients via meta-analysis, data-mining, and multivariate analysis.
Endocrinologia, diabetes y nutricion, 72(6):501561.
Type 2 diabetes mellitus (T2DM)-one of the fastest globally spreading diseases-is a chronic metabolic disorder characterized by elevated blood glucose levels. It has been suggested that the composition of gut microbiota plays key roles in the prevalence of T2DM. In this study, a novel approach of large-scale data mining and multivariate analysis of the gut microbiome of T2DM patients and healthy controls was conducted to find the key compositional differences in their microbiota and potential biomarkers of the disease.
METHODS: First, suitable datasets were identified (9 in total with 946 samples), analyzed, and their operational taxonomic units (OTUs) were computed by identical parameters to increase accuracy. The following OTUs were merged and compared based on their health status, and compositional differences detected. For biomarker identification, the OTUs were subjected to 9 different attribute weighting models. Additionally, OTUs were independently analyzed by multivariate algorithms (LEfSe test) to verify the realized biomarkers.
RESULTS: Overall, 23 genera and 4 phyla were identified as possible biomarkers. At genus level, the decrease of Bacteroides, Methanobrevibacter, Paraprevotella, and [Eubacterium] hallii group in T2DM and the increase of Prevotella, Megamonas, Megasphaera, Ligilactobacillus, and Lachnoclostridium were selected as biomarkers; and at phylum level, the increase of Synergistota and the decrease of Euryarchaeota, Desulfobacterota (Thermodesulfobacteriota), and Ptescibacteria.
CONCLUSION: This is the first study ever conducted to find the microbial compositional differences and biomarkers in T2DM using data mining models applied on a widespread metagenome dataset and verified by multivariate analysis.
Additional Links: PMID-40514168
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40514168,
year = {2025},
author = {Ebrahimi, F and Maleki, H and Ebrahimi, M and Beiki, AH},
title = {A novel approach to finding the compositional differences and biomarkers in gut microbiota in type 2 diabetic patients via meta-analysis, data-mining, and multivariate analysis.},
journal = {Endocrinologia, diabetes y nutricion},
volume = {72},
number = {6},
pages = {501561},
doi = {10.1016/j.endien.2025.501561},
pmid = {40514168},
issn = {2530-0180},
mesh = {*Diabetes Mellitus, Type 2/microbiology ; Humans ; *Gastrointestinal Microbiome ; *Data Mining ; Biomarkers/analysis ; Multivariate Analysis ; Male ; Middle Aged ; Female ; },
abstract = {Type 2 diabetes mellitus (T2DM)-one of the fastest globally spreading diseases-is a chronic metabolic disorder characterized by elevated blood glucose levels. It has been suggested that the composition of gut microbiota plays key roles in the prevalence of T2DM. In this study, a novel approach of large-scale data mining and multivariate analysis of the gut microbiome of T2DM patients and healthy controls was conducted to find the key compositional differences in their microbiota and potential biomarkers of the disease.
METHODS: First, suitable datasets were identified (9 in total with 946 samples), analyzed, and their operational taxonomic units (OTUs) were computed by identical parameters to increase accuracy. The following OTUs were merged and compared based on their health status, and compositional differences detected. For biomarker identification, the OTUs were subjected to 9 different attribute weighting models. Additionally, OTUs were independently analyzed by multivariate algorithms (LEfSe test) to verify the realized biomarkers.
RESULTS: Overall, 23 genera and 4 phyla were identified as possible biomarkers. At genus level, the decrease of Bacteroides, Methanobrevibacter, Paraprevotella, and [Eubacterium] hallii group in T2DM and the increase of Prevotella, Megamonas, Megasphaera, Ligilactobacillus, and Lachnoclostridium were selected as biomarkers; and at phylum level, the increase of Synergistota and the decrease of Euryarchaeota, Desulfobacterota (Thermodesulfobacteriota), and Ptescibacteria.
CONCLUSION: This is the first study ever conducted to find the microbial compositional differences and biomarkers in T2DM using data mining models applied on a widespread metagenome dataset and verified by multivariate analysis.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
*Diabetes Mellitus, Type 2/microbiology
Humans
*Gastrointestinal Microbiome
*Data Mining
Biomarkers/analysis
Multivariate Analysis
Male
Middle Aged
Female
RevDate: 2025-06-13
Exploring the mechanism of Carbonized Typhae Pollen in treating blood stasis syndrome through metabolic profiling: the synergistic effect of hemostasis without blood stasis.
Journal of ethnopharmacology pii:S0378-8741(25)00811-6 [Epub ahead of print].
Removing blood stasis and stopping bleeding traditional Chinese medicines (RBSB-TCM) formed a unique class of TCM, characterized by vasodilating, removing stasis and hemostatic effects. Carbonized Typhae Pollen (CTP), derived from Typhae Pollen (TP) through carbonization, has emerged as a particularly valuable therapeutic agent. It has been widely used in clinical practice to treat hemorrhagic disorders caused by blood stasis syndrome (BSS). However, the potential mechanism for CTP to achieve the dual synergistic effect of promoting blood flow and hemostasis remains unclear.
AIM OF THE STUDY: From the standpoint of metabolite profiles, this study attempts to investigate the fundamental mechanism of CTP in the elimination of blood stasis and the cessation of bleeding.
MATERIALS AND METHODS: First, chemical constituents, absorbed constituents and metabolites in rats following oral administration of CTP were identified by ultra-high performance liquid chromatography coupled with the quadrupole time-of-flight mass spectrometry (UHPLC-Q-TOF-MS) method combined with MetabolitePilot 2.0.4 software. Subsequently, the pharmacological effects of CTP were systematically investigated using rat models with BSS and zebrafish with cerebral hemorrhage. Specifically, the impact on coagulation function and histopathology in rats, as well as the effect on cerebral hemorrhage in zebrafish, were thoroughly evaluated. Untargeted metabolomics based on rat plasma was applied to analyze the metabolic profile changes, revealing the potential action mechanism. The underlying mechanism was furtherly confirmed by gut microbiome analysis and systemic molecular biology experiments.
RESULTS: 34 prototype chemicals and 71 metabolites from the liver, heart, spleen, lung, kidney, small intestine, uterus, and serum were found. CTP improved the abnormal coagulation system, promoted blood circulation, and reduced pathological damage caused by BSS. Plasma metabolomics revealed that BSS significantly altered bile acid (BA) metabolism and arachidonic acid (AA) metabolism. Gut microbiome analysis and fecal microbiota transplantation (FMT) experiments further demonstrated that CTP modulated the gut microbiota. This modulation promoted BA production and activated endothelial nitric oxide synthase (eNOS), leading to increased nitric oxide (NO) levels. These changes contributed to the therapeutic effect of CTP in removing blood stasis. Systemic molecular biology experiments showed that CTP activated key components of the AA metabolic pathway. It promoted PLCγ1 phosphorylation, increased intracellular Ca[2+] levels, and upregulated COX-2 expression. In addition, CTP enhanced the production of AA-related metabolites, including 6-keto-prostaglandin F1α (6-keto-PGF1α), prostaglandin E2 (PGE2), and thromboxane B2 (TXB2). It also increased the transcription of AA metabolism-related genes, such as PLCγ1, PTGS2a, PTGS2b, PTGIS, PTGES, TXBAS, and vWF.
CONCLUSIONS: CTP could promote the generation of AA metabolites through PLCγ1/Ca[2+]/COX-2 to stop bleeding, while also enhancing eNOS activity and NO synthesis through gut microbiota-bile acid axis to remove blood stasis. These two effect were balanced to achieve hemostasis without blood stasis.
Additional Links: PMID-40513921
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40513921,
year = {2025},
author = {Zhang, X and Sheng, N and Wang, Z and Cao, Y and Jiang, X and Yan, H and Cheng, F and Geng, T and Wei, K and Zhang, L and Gao, M and Zhou, G and Chen, P},
title = {Exploring the mechanism of Carbonized Typhae Pollen in treating blood stasis syndrome through metabolic profiling: the synergistic effect of hemostasis without blood stasis.},
journal = {Journal of ethnopharmacology},
volume = {},
number = {},
pages = {120124},
doi = {10.1016/j.jep.2025.120124},
pmid = {40513921},
issn = {1872-7573},
abstract = {Removing blood stasis and stopping bleeding traditional Chinese medicines (RBSB-TCM) formed a unique class of TCM, characterized by vasodilating, removing stasis and hemostatic effects. Carbonized Typhae Pollen (CTP), derived from Typhae Pollen (TP) through carbonization, has emerged as a particularly valuable therapeutic agent. It has been widely used in clinical practice to treat hemorrhagic disorders caused by blood stasis syndrome (BSS). However, the potential mechanism for CTP to achieve the dual synergistic effect of promoting blood flow and hemostasis remains unclear.
AIM OF THE STUDY: From the standpoint of metabolite profiles, this study attempts to investigate the fundamental mechanism of CTP in the elimination of blood stasis and the cessation of bleeding.
MATERIALS AND METHODS: First, chemical constituents, absorbed constituents and metabolites in rats following oral administration of CTP were identified by ultra-high performance liquid chromatography coupled with the quadrupole time-of-flight mass spectrometry (UHPLC-Q-TOF-MS) method combined with MetabolitePilot 2.0.4 software. Subsequently, the pharmacological effects of CTP were systematically investigated using rat models with BSS and zebrafish with cerebral hemorrhage. Specifically, the impact on coagulation function and histopathology in rats, as well as the effect on cerebral hemorrhage in zebrafish, were thoroughly evaluated. Untargeted metabolomics based on rat plasma was applied to analyze the metabolic profile changes, revealing the potential action mechanism. The underlying mechanism was furtherly confirmed by gut microbiome analysis and systemic molecular biology experiments.
RESULTS: 34 prototype chemicals and 71 metabolites from the liver, heart, spleen, lung, kidney, small intestine, uterus, and serum were found. CTP improved the abnormal coagulation system, promoted blood circulation, and reduced pathological damage caused by BSS. Plasma metabolomics revealed that BSS significantly altered bile acid (BA) metabolism and arachidonic acid (AA) metabolism. Gut microbiome analysis and fecal microbiota transplantation (FMT) experiments further demonstrated that CTP modulated the gut microbiota. This modulation promoted BA production and activated endothelial nitric oxide synthase (eNOS), leading to increased nitric oxide (NO) levels. These changes contributed to the therapeutic effect of CTP in removing blood stasis. Systemic molecular biology experiments showed that CTP activated key components of the AA metabolic pathway. It promoted PLCγ1 phosphorylation, increased intracellular Ca[2+] levels, and upregulated COX-2 expression. In addition, CTP enhanced the production of AA-related metabolites, including 6-keto-prostaglandin F1α (6-keto-PGF1α), prostaglandin E2 (PGE2), and thromboxane B2 (TXB2). It also increased the transcription of AA metabolism-related genes, such as PLCγ1, PTGS2a, PTGS2b, PTGIS, PTGES, TXBAS, and vWF.
CONCLUSIONS: CTP could promote the generation of AA metabolites through PLCγ1/Ca[2+]/COX-2 to stop bleeding, while also enhancing eNOS activity and NO synthesis through gut microbiota-bile acid axis to remove blood stasis. These two effect were balanced to achieve hemostasis without blood stasis.},
}
RevDate: 2025-06-13
Screening of potential probiotic Bifidobacteria from intestinal tract and its application in fermented milk.
Journal of dairy science pii:S0022-0302(25)00414-X [Epub ahead of print].
Bifidobacteria, a significant component of human gut microbiota, play a crucial role in maintaining human health. Studies have shown that the composition of gut microbiome is closely related to the geographical environment, ethnicity, and dietary structure of the host. Currently, most of the mainstream probiotics on the market are based on the gut microbiota characteristics of Western populations, which may not be completely suitable for the gut environment of Chinese people. Therefore, the isolation and screening of Bifidobacteria with probiotic functions in the intestinal tract is critically important to meet the demands of the Chinese market. In the present study, 216 Bifidobacterial strains were isolated and identified from 35 fecal samples collected from the city of Hohhot, Inner Mongolia, China. Further investigations were conducted to determine the probiotic potential of these strains. Among the screened strains, Bifidobacterium longum ssp. longum IMAU12449 was identified as the most promising probiotic strain. The survival rate of strain IMAU12449 was highest in the simulated artificial gastrointestinal fluid. The survival rate of strain IMAU12449 in artificial gastric fluid for 3 h was 62.38%, and the survival rate was 40.30% in the artificial intestinal fluid for 8 h. Furthermore, the strain exhibited excellent cell autoaggregation (72.79%) and surface hydrophobicity (45.66%). Strain IMAU12449 also exhibited a strong inhibitory effect on common pathogens. Safety evaluation revealed that the strain possessed 2 potential antibiotic resistance genes and no virulence factors. This strain exhibited γ-hemolysis in vitro. The advantages of a stable viable count, moderate acidity, and absence of whey precipitation were observed in fermented cow and soy milk processed by strain IMAU12449 combined with commercial starters. After storage at 4°C for 21 d, the viable cell count remained above 10[7] cfu/mL. In conclusion, a Bifidobacterial strain, IMAU12449, with favorable probiotic properties was identified in this study. This strain has demonstrated significant potential for application in fermented milk, and further research should be conducted to elucidate its role in promoting health.
Additional Links: PMID-40513864
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40513864,
year = {2025},
author = {Li, Y and Fan, Q and Dong, H and Chang, S and Liu, W},
title = {Screening of potential probiotic Bifidobacteria from intestinal tract and its application in fermented milk.},
journal = {Journal of dairy science},
volume = {},
number = {},
pages = {},
doi = {10.3168/jds.2024-25960},
pmid = {40513864},
issn = {1525-3198},
abstract = {Bifidobacteria, a significant component of human gut microbiota, play a crucial role in maintaining human health. Studies have shown that the composition of gut microbiome is closely related to the geographical environment, ethnicity, and dietary structure of the host. Currently, most of the mainstream probiotics on the market are based on the gut microbiota characteristics of Western populations, which may not be completely suitable for the gut environment of Chinese people. Therefore, the isolation and screening of Bifidobacteria with probiotic functions in the intestinal tract is critically important to meet the demands of the Chinese market. In the present study, 216 Bifidobacterial strains were isolated and identified from 35 fecal samples collected from the city of Hohhot, Inner Mongolia, China. Further investigations were conducted to determine the probiotic potential of these strains. Among the screened strains, Bifidobacterium longum ssp. longum IMAU12449 was identified as the most promising probiotic strain. The survival rate of strain IMAU12449 was highest in the simulated artificial gastrointestinal fluid. The survival rate of strain IMAU12449 in artificial gastric fluid for 3 h was 62.38%, and the survival rate was 40.30% in the artificial intestinal fluid for 8 h. Furthermore, the strain exhibited excellent cell autoaggregation (72.79%) and surface hydrophobicity (45.66%). Strain IMAU12449 also exhibited a strong inhibitory effect on common pathogens. Safety evaluation revealed that the strain possessed 2 potential antibiotic resistance genes and no virulence factors. This strain exhibited γ-hemolysis in vitro. The advantages of a stable viable count, moderate acidity, and absence of whey precipitation were observed in fermented cow and soy milk processed by strain IMAU12449 combined with commercial starters. After storage at 4°C for 21 d, the viable cell count remained above 10[7] cfu/mL. In conclusion, a Bifidobacterial strain, IMAU12449, with favorable probiotic properties was identified in this study. This strain has demonstrated significant potential for application in fermented milk, and further research should be conducted to elucidate its role in promoting health.},
}
RevDate: 2025-06-13
The Link Between Alcohol Consumption and Kidney Injury.
The American journal of pathology pii:S0002-9440(25)00193-2 [Epub ahead of print].
Alcohol consumption contributes to systemic organ dysfunction, but its direct effect on kidney health is unclear. Epidemiological studies show inconsistent findings due to reliance on conventional markers like serum creatinine (sCr) and blood urea nitrogen (BUN), which are insensitive to early chronic kidney disease (CKD) and influenced by factors such as muscle mass, diet, and hydration status. Experimental studies indicate that alcohol may directly exacerbate renal damage through mitochondrial dysfunction, oxidative stress, and inflammation. Furthermore, indirect effects from alcohol-induced altered intestinal permeability and microbiome, liver injury, microcirculatory/cardiac dysfunction and muscle damage may also facilitate kidney damage. Notably, alcohol-related liver disease can lead to hepatorenal syndrome, a severe form of kidney dysfunction driven by circulatory disturbances and systemic inflammation. This overview explores the adverse effects of alcohol misuse on kidney health and disease, emphasizing the need for comprehensive epidemiological studies with more sensitive kidney injury biomarkers. It also highlights the importance of using clinically relevant preclinical models to clarify the underlying mechanisms of alcohol-related kidney injury and to enhance our understanding of its long-term clinical consequences.
Additional Links: PMID-40513821
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40513821,
year = {2025},
author = {Yokus, B and Maccioni, L and Fu, L and Haskó, G and Nagy, LE and Gao, B and Pacher, P},
title = {The Link Between Alcohol Consumption and Kidney Injury.},
journal = {The American journal of pathology},
volume = {},
number = {},
pages = {},
doi = {10.1016/j.ajpath.2025.05.011},
pmid = {40513821},
issn = {1525-2191},
abstract = {Alcohol consumption contributes to systemic organ dysfunction, but its direct effect on kidney health is unclear. Epidemiological studies show inconsistent findings due to reliance on conventional markers like serum creatinine (sCr) and blood urea nitrogen (BUN), which are insensitive to early chronic kidney disease (CKD) and influenced by factors such as muscle mass, diet, and hydration status. Experimental studies indicate that alcohol may directly exacerbate renal damage through mitochondrial dysfunction, oxidative stress, and inflammation. Furthermore, indirect effects from alcohol-induced altered intestinal permeability and microbiome, liver injury, microcirculatory/cardiac dysfunction and muscle damage may also facilitate kidney damage. Notably, alcohol-related liver disease can lead to hepatorenal syndrome, a severe form of kidney dysfunction driven by circulatory disturbances and systemic inflammation. This overview explores the adverse effects of alcohol misuse on kidney health and disease, emphasizing the need for comprehensive epidemiological studies with more sensitive kidney injury biomarkers. It also highlights the importance of using clinically relevant preclinical models to clarify the underlying mechanisms of alcohol-related kidney injury and to enhance our understanding of its long-term clinical consequences.},
}
RevDate: 2025-06-13
Temporal dynamics of microbial communities in anaerobic digestion: Influence of temperature and feedstock composition on reactor performance and stability.
Water research, 284:123974 pii:S0043-1354(25)00882-6 [Epub ahead of print].
This study presents a novel multi-disciplinary approach, integrating explainable machine-learning with detailed chemical and biological analysis using microbial fermentation food wastewater to identify critical parameters influencing AD microbiome diversity and reactor performance over time. Our research addresses knowledge gaps on the resource recovery potential from food fermentation wastewater which remains largely unexplored. Eighteen continuous bioreactors were operated under varying temperature and process configuration (single vs. two stages) using wastewater stream derived from real-world microbial protein manufacturing. Detailed chemical analysis fingerprinted the fermentation-wastewater throughout the AD process including sugars, sugar alcohols and volatile fatty acids (VFAs). Significant elevations in Methanomassiliicoccus were correlated to high butyric acid concentrations (p < 0.05) and decreased biogas production, further elucidating the role of this newly discovered methanogen in AD. Machine-learning models predicting reactor performance achieved high accuracy based on operational parameters and microbial taxonomy (14.03 root mean squared error and 0.86 R[2]). Operational parameters were found to have the most substantial influence on chemical oxygen demand removal. Oscillibacter and Clostridium sensu stricto were highlighted as key factors impacting specific biogas yield for the first time. By integrating detailed chemical and biological fingerprinting with explainable machine learning models this research presents a novel approach to advance whole-systems AD understanding, offering insights of potential new biomarkers (Oscillibacter and Clostridium sensu stricto) for industrial applications of sustainable waste-to-energy systems.
Additional Links: PMID-40513460
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40513460,
year = {2025},
author = {Piercy, E and Sun, X and Ellis, PR and Taylor, M and Guo, M},
title = {Temporal dynamics of microbial communities in anaerobic digestion: Influence of temperature and feedstock composition on reactor performance and stability.},
journal = {Water research},
volume = {284},
number = {},
pages = {123974},
doi = {10.1016/j.watres.2025.123974},
pmid = {40513460},
issn = {1879-2448},
abstract = {This study presents a novel multi-disciplinary approach, integrating explainable machine-learning with detailed chemical and biological analysis using microbial fermentation food wastewater to identify critical parameters influencing AD microbiome diversity and reactor performance over time. Our research addresses knowledge gaps on the resource recovery potential from food fermentation wastewater which remains largely unexplored. Eighteen continuous bioreactors were operated under varying temperature and process configuration (single vs. two stages) using wastewater stream derived from real-world microbial protein manufacturing. Detailed chemical analysis fingerprinted the fermentation-wastewater throughout the AD process including sugars, sugar alcohols and volatile fatty acids (VFAs). Significant elevations in Methanomassiliicoccus were correlated to high butyric acid concentrations (p < 0.05) and decreased biogas production, further elucidating the role of this newly discovered methanogen in AD. Machine-learning models predicting reactor performance achieved high accuracy based on operational parameters and microbial taxonomy (14.03 root mean squared error and 0.86 R[2]). Operational parameters were found to have the most substantial influence on chemical oxygen demand removal. Oscillibacter and Clostridium sensu stricto were highlighted as key factors impacting specific biogas yield for the first time. By integrating detailed chemical and biological fingerprinting with explainable machine learning models this research presents a novel approach to advance whole-systems AD understanding, offering insights of potential new biomarkers (Oscillibacter and Clostridium sensu stricto) for industrial applications of sustainable waste-to-energy systems.},
}
RevDate: 2025-06-13
The oral microbiome and all-cause mortality in aUS population representative prospective cohort.
The Journal of infectious diseases pii:8162318 [Epub ahead of print].
No large studies have evaluated whether the human oral microbiome is directly associated with mortality. We evaluated prospective associations between the oral microbiome, measured using 16S rRNA gene sequencing, from participants aged 20-69 years in the 2009-2012 cycles of the National Health and Nutrition Examination Survey (NHANES) and all-cause mortality (N=7,721, representing ∼194 million individuals). Alpha diversity was inversely associated with mortality, and some significant associations were observed with the beta diversity matrices. Higher relative abundances of Granulicatella and Lactobacillus were associated with increased risk, while Bacteroides was associated with decreased all-cause mortality at the genus level. Results suggest oral bacterial communities may be important contributors to health and disease.
Additional Links: PMID-40513117
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40513117,
year = {2025},
author = {Vogtmann, E and Yano, Y and Shi, J and Wan, Y and Purandare, V and McLean, J and Li, S and Knight, R and Kahle, L and Hullings, AG and Hua, X and Graubard, BI and Gillison, ML and Caporaso, JG and Bokulich, NA and Blaser, MJ and Freedman, ND and Chaturvedi, AK and Abnet, CC},
title = {The oral microbiome and all-cause mortality in aUS population representative prospective cohort.},
journal = {The Journal of infectious diseases},
volume = {},
number = {},
pages = {},
doi = {10.1093/infdis/jiaf321},
pmid = {40513117},
issn = {1537-6613},
abstract = {No large studies have evaluated whether the human oral microbiome is directly associated with mortality. We evaluated prospective associations between the oral microbiome, measured using 16S rRNA gene sequencing, from participants aged 20-69 years in the 2009-2012 cycles of the National Health and Nutrition Examination Survey (NHANES) and all-cause mortality (N=7,721, representing ∼194 million individuals). Alpha diversity was inversely associated with mortality, and some significant associations were observed with the beta diversity matrices. Higher relative abundances of Granulicatella and Lactobacillus were associated with increased risk, while Bacteroides was associated with decreased all-cause mortality at the genus level. Results suggest oral bacterial communities may be important contributors to health and disease.},
}
RevDate: 2025-06-13
CmpDate: 2025-06-13
Enzymatic carbon-fluorine bond cleavage by human gut microbes.
Proceedings of the National Academy of Sciences of the United States of America, 122(24):e2504122122.
Fluorinated compounds are used for agrochemical, pharmaceutical, and numerous industrial applications, resulting in global contamination. In many molecules, fluorine is incorporated to enhance the half-life and improve bioavailability. Fluorinated compounds enter the human body through food, water, and xenobiotics including pharmaceuticals, exposing gut microbes to these substances. The human gut microbiota is known for its xenobiotic biotransformation capabilities, but it was not previously known whether gut microbial enzymes could break carbon-fluorine bonds, potentially altering the toxicity of these compounds. Here, through the development of a rapid, miniaturized fluoride detection assay for whole-cell screening, we identified active gut microbial defluorinases. We biochemically characterized enzymes from diverse human gut microbial classes including Clostridia, Bacilli, and Coriobacteriia, with the capacity to hydrolyze (di)fluorinated organic acids and a fluorinated amino acid. Whole-protein alanine scanning, molecular dynamics simulations, and chimeric protein design enabled the identification of a disordered C-terminal protein segment involved in defluorination activity. Domain swapping exclusively of the C-terminus conferred defluorination activity to a nondefluorinating dehalogenase. To advance our understanding of the structural and sequence differences between defluorinating and nondefluorinating dehalogenases, we trained machine learning models which identified protein termini as important features. Models trained on 41-amino acid segments from protein C termini alone predicted defluorination activity with 83% accuracy (compared to 95% accuracy based on full-length protein features). This work is relevant for therapeutic interventions and environmental and human health by uncovering specificity-determining signatures of fluorine biochemistry from the gut microbiome.
Additional Links: PMID-40512801
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40512801,
year = {2025},
author = {Probst, SI and Felder, FD and Poltorak, V and Mewalal, R and Blaby, IK and Robinson, SL},
title = {Enzymatic carbon-fluorine bond cleavage by human gut microbes.},
journal = {Proceedings of the National Academy of Sciences of the United States of America},
volume = {122},
number = {24},
pages = {e2504122122},
doi = {10.1073/pnas.2504122122},
pmid = {40512801},
issn = {1091-6490},
support = {2022-YIG-090//Helmut Horten Stiftung (Helmut Horten Foundation)/ ; PZPGP2_209124//Schweizerischer Nationalfonds zur Förderung der Wissenschaftlichen Forschung (SNF)/ ; PZPGP2_209124//Schweizerischer Nationalfonds zur Förderung der Wissenschaftlichen Forschung (SNF)/ ; N/A//Peter und Traudl Engelhorn Stiftung (Peter and Traudl Engelhorn Foundation)/ ; 230-2024//Uniscientia Foundation/ ; 23-2-ETH-047//Eidgenössische Technische Hochschule Zürich (ETH)/ ; },
mesh = {Humans ; *Gastrointestinal Microbiome/physiology ; *Carbon/metabolism/chemistry ; *Fluorine/metabolism/chemistry ; Halogenation ; *Bacterial Proteins/metabolism/chemistry/genetics ; *Hydrolases/metabolism/chemistry ; Molecular Dynamics Simulation ; },
abstract = {Fluorinated compounds are used for agrochemical, pharmaceutical, and numerous industrial applications, resulting in global contamination. In many molecules, fluorine is incorporated to enhance the half-life and improve bioavailability. Fluorinated compounds enter the human body through food, water, and xenobiotics including pharmaceuticals, exposing gut microbes to these substances. The human gut microbiota is known for its xenobiotic biotransformation capabilities, but it was not previously known whether gut microbial enzymes could break carbon-fluorine bonds, potentially altering the toxicity of these compounds. Here, through the development of a rapid, miniaturized fluoride detection assay for whole-cell screening, we identified active gut microbial defluorinases. We biochemically characterized enzymes from diverse human gut microbial classes including Clostridia, Bacilli, and Coriobacteriia, with the capacity to hydrolyze (di)fluorinated organic acids and a fluorinated amino acid. Whole-protein alanine scanning, molecular dynamics simulations, and chimeric protein design enabled the identification of a disordered C-terminal protein segment involved in defluorination activity. Domain swapping exclusively of the C-terminus conferred defluorination activity to a nondefluorinating dehalogenase. To advance our understanding of the structural and sequence differences between defluorinating and nondefluorinating dehalogenases, we trained machine learning models which identified protein termini as important features. Models trained on 41-amino acid segments from protein C termini alone predicted defluorination activity with 83% accuracy (compared to 95% accuracy based on full-length protein features). This work is relevant for therapeutic interventions and environmental and human health by uncovering specificity-determining signatures of fluorine biochemistry from the gut microbiome.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
*Gastrointestinal Microbiome/physiology
*Carbon/metabolism/chemistry
*Fluorine/metabolism/chemistry
Halogenation
*Bacterial Proteins/metabolism/chemistry/genetics
*Hydrolases/metabolism/chemistry
Molecular Dynamics Simulation
RevDate: 2025-06-13
Exploring the role of glucagon-like peptide-1 receptor agonists in critical illness: mechanisms, benefits, and clinical implications.
Current opinion in critical care pii:00075198-990000000-00277 [Epub ahead of print].
PURPOSE OF REVIEW: This review aims to synthesize current evidence on the expanding role of glucagon-like peptide-1 (GLP-1) receptor agonists (RAs) beyond glycemic control, particularly in the context of critical illness. With growing recognition of their anti-inflammatory, immunomodulatory, and organ-protective properties, GLP-1RAs show promise in managing complications such as sepsis, acute respiratory distress syndrome (ARDS), acute kidney injury, and multiorgan dysfunction. This review explores their mechanisms across vital systems - including cardiovascular, pulmonary, renal, hepatic, neuropsychiatric, and gut-lung-microbiome axes - highlighting preclinical and clinical data. By consolidating emerging findings, this review aims to inform future translational research and support the integration of GLP-1RAs into critical care strategies.
RECENT FINDINGS: Recent evidence highlights GLP-1RAs as promising adjuncts in critical illness due to their multiorgan protective effects. In cardiovascular systems, GLP-1RAs improve endothelial function, reduce ischemia-reperfusion injury, and modulate autonomic tone. In the CNS, they provide neuroprotection by reducing neuroinflammation and pyroptosis. In the lungs, they attenuate ARDS by decreasing cytokine production, enhancing surfactant secretion, and restoring alveolar-capillary integrity. Renally, they reduce acute injury and preserve filtration function. In the gut, GLP-1RAs modulate the microbiome, enhance barrier integrity, and lower systemic inflammation via the gut-lung axis. Additionally, they reduce hepatic inflammation and support pancreatic beta-cell survival, improving insulin sensitivity and metabolic stability in critical care.
SUMMARY: GLP-1 receptor agonists hold promise as multiorgan protective agents in critical illness, offering benefits beyond glucose control. Their anti-inflammatory, metabolic, and vaso-protective properties may be harnessed to prevent or attenuate organ failure, support recovery, and improve long-term outcomes in critically ill patients. Further clinical trials are warranted to define their safety, efficacy, and optimal application across ICU populations.
Additional Links: PMID-40512576
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40512576,
year = {2025},
author = {Jogani, VG and Mohamed Elfadil, O and Edakkanambeth Varayil, J and Mundi, MS},
title = {Exploring the role of glucagon-like peptide-1 receptor agonists in critical illness: mechanisms, benefits, and clinical implications.},
journal = {Current opinion in critical care},
volume = {},
number = {},
pages = {},
doi = {10.1097/MCC.0000000000001294},
pmid = {40512576},
issn = {1531-7072},
abstract = {PURPOSE OF REVIEW: This review aims to synthesize current evidence on the expanding role of glucagon-like peptide-1 (GLP-1) receptor agonists (RAs) beyond glycemic control, particularly in the context of critical illness. With growing recognition of their anti-inflammatory, immunomodulatory, and organ-protective properties, GLP-1RAs show promise in managing complications such as sepsis, acute respiratory distress syndrome (ARDS), acute kidney injury, and multiorgan dysfunction. This review explores their mechanisms across vital systems - including cardiovascular, pulmonary, renal, hepatic, neuropsychiatric, and gut-lung-microbiome axes - highlighting preclinical and clinical data. By consolidating emerging findings, this review aims to inform future translational research and support the integration of GLP-1RAs into critical care strategies.
RECENT FINDINGS: Recent evidence highlights GLP-1RAs as promising adjuncts in critical illness due to their multiorgan protective effects. In cardiovascular systems, GLP-1RAs improve endothelial function, reduce ischemia-reperfusion injury, and modulate autonomic tone. In the CNS, they provide neuroprotection by reducing neuroinflammation and pyroptosis. In the lungs, they attenuate ARDS by decreasing cytokine production, enhancing surfactant secretion, and restoring alveolar-capillary integrity. Renally, they reduce acute injury and preserve filtration function. In the gut, GLP-1RAs modulate the microbiome, enhance barrier integrity, and lower systemic inflammation via the gut-lung axis. Additionally, they reduce hepatic inflammation and support pancreatic beta-cell survival, improving insulin sensitivity and metabolic stability in critical care.
SUMMARY: GLP-1 receptor agonists hold promise as multiorgan protective agents in critical illness, offering benefits beyond glucose control. Their anti-inflammatory, metabolic, and vaso-protective properties may be harnessed to prevent or attenuate organ failure, support recovery, and improve long-term outcomes in critically ill patients. Further clinical trials are warranted to define their safety, efficacy, and optimal application across ICU populations.},
}
RevDate: 2025-06-13
The Role of Gut Microbiota in the Association Between Air Pollution and Cognitive Function in Older Adults.
Environmental health perspectives [Epub ahead of print].
BACKGROUND: Growing evidence links air pollution to cognitive dysfunction in older adults. The gut microbiome and circulating metabolites present an important yet unexplored pathway, given their crucial role in the gut-brain axis.
OBJECTIVES: We aimed to explore the potential roles of gut bacteria, fungi, microbial functional potentials, and circuiting metabolites in the association of residential PM2.5 and O3 exposures with cognitive dysfunction.
METHODS: We analyzed gut microbiome data from 1,027 older adults using metagenome and internal transcribed spacer sequencing to profile bacterial and fungal taxa, functional pathways, and enzyme abundances. Targeted metabolomics quantified 195 circulating metabolites, such as amino acids and organic acids. Annual average ambient PM2.5 and O3 exposures were estimated using satellite-based models. Cognitive outcomes, including mild cognitive impairment and cognitive decline, were assessed using the Mini-mental State Examination and Hasegawa dementia scale. Statistical analyses included Microbiome Multivariable Association with Linear Models (with a false discovery rate threshold of 0.25) for microbial associations and multivariate regression for metabolites and cognitive outcomes.
RESULTS: Higher PM2.5 and O3 exposures were associated with disturbances in microbial composition, altered taxonomic profiles (e.g., decreased abundances of Blautia obeum and Gordonibacter pamelaeae), and disrupted functional pathways, particularly those regulating 2-oxoglutarate. These findings were partially replicated in an independent population. Higher air pollution levels were associated with increased circulating levels of 2-oxoglutarate and L-glutamine (key metabolites in neurodegenerative progression), which were further linked to higher odds of concurrent mild cognitive impairment (OR: 1.39-1.56) and an increased 2-year risk of cognitive decline (OR: 1.26-1.37). These associations were partially mediated by air pollution-related changes in microbial anaerobic energy metabolism pathways, especially involving 2-oxoglutarate metabolism and the enzyme aspartate transaminase.
CONCLUSIONS: Our findings highlight the role of the gut microbiome and microbial metabolites in mediating the detrimental impact of air pollution on cognitive health in older adults, providing new insights into the underlying etiology for future hypothesis generation. https://doi.org/10.1289/EHP16515.
Additional Links: PMID-40512497
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40512497,
year = {2025},
author = {Qi, W and Kong, M and Meng, X and Sun, Z and Mei, Z and Pu, Y and Zhou, X and Wang, Q and Qiu, JG and Jiang, BH and Shen, J and Yuan, C and Ji, JS and Wang, X and Kan, H and Zheng, Y},
title = {The Role of Gut Microbiota in the Association Between Air Pollution and Cognitive Function in Older Adults.},
journal = {Environmental health perspectives},
volume = {},
number = {},
pages = {},
doi = {10.1289/EHP16515},
pmid = {40512497},
issn = {1552-9924},
abstract = {BACKGROUND: Growing evidence links air pollution to cognitive dysfunction in older adults. The gut microbiome and circulating metabolites present an important yet unexplored pathway, given their crucial role in the gut-brain axis.
OBJECTIVES: We aimed to explore the potential roles of gut bacteria, fungi, microbial functional potentials, and circuiting metabolites in the association of residential PM2.5 and O3 exposures with cognitive dysfunction.
METHODS: We analyzed gut microbiome data from 1,027 older adults using metagenome and internal transcribed spacer sequencing to profile bacterial and fungal taxa, functional pathways, and enzyme abundances. Targeted metabolomics quantified 195 circulating metabolites, such as amino acids and organic acids. Annual average ambient PM2.5 and O3 exposures were estimated using satellite-based models. Cognitive outcomes, including mild cognitive impairment and cognitive decline, were assessed using the Mini-mental State Examination and Hasegawa dementia scale. Statistical analyses included Microbiome Multivariable Association with Linear Models (with a false discovery rate threshold of 0.25) for microbial associations and multivariate regression for metabolites and cognitive outcomes.
RESULTS: Higher PM2.5 and O3 exposures were associated with disturbances in microbial composition, altered taxonomic profiles (e.g., decreased abundances of Blautia obeum and Gordonibacter pamelaeae), and disrupted functional pathways, particularly those regulating 2-oxoglutarate. These findings were partially replicated in an independent population. Higher air pollution levels were associated with increased circulating levels of 2-oxoglutarate and L-glutamine (key metabolites in neurodegenerative progression), which were further linked to higher odds of concurrent mild cognitive impairment (OR: 1.39-1.56) and an increased 2-year risk of cognitive decline (OR: 1.26-1.37). These associations were partially mediated by air pollution-related changes in microbial anaerobic energy metabolism pathways, especially involving 2-oxoglutarate metabolism and the enzyme aspartate transaminase.
CONCLUSIONS: Our findings highlight the role of the gut microbiome and microbial metabolites in mediating the detrimental impact of air pollution on cognitive health in older adults, providing new insights into the underlying etiology for future hypothesis generation. https://doi.org/10.1289/EHP16515.},
}
RevDate: 2025-06-13
Novel Approaches in Establishing Chemical Food Safety Based on the Detoxification Capacity of Probiotics and Postbiotics: A Critical Review.
Probiotics and antimicrobial proteins [Epub ahead of print].
Environmental and chemical contaminants from industrial and agricultural sources increasingly threaten food safety worldwide. These contaminants, including bacterial toxins (e.g., botulinum toxin), mycotoxins (e.g., aflatoxin, ochratoxin), pesticides, heavy metals, and microplastics, pose significant health and economic risks. Emerging evidence highlights that chronic exposure to such xenobiotics disrupts gut microbiota structure and function, adversely affecting host health. While traditional physicochemical detoxification methods exist, they often require impractical conditions. Biological detoxification via probiotics and their metabolites (postbiotics) has gained attention as a practical and cost-effective alternative. Numerous studies concur that probiotics can bind, biotransform, or sequester xenobiotics, thereby mitigating toxicity and restoring microbiota balance. However, variations in strain efficacy, binding mechanisms, and in vivo performance indicate areas needing further exploration. This review critically synthesizes current knowledge on probiotic-mediated detoxification mechanisms, their interaction with xenobiotics and the gut microbiome, and host responses, highlighting consensus, discrepancies, and gaps to inform future research and applications.
Additional Links: PMID-40512444
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40512444,
year = {2025},
author = {Abbasi, A and Sheykhsaran, E and Hosseinzadeh, N and Bazdar, M and Hamehjani, M and Aghapour, B and Shojaee-Aliabadi, S},
title = {Novel Approaches in Establishing Chemical Food Safety Based on the Detoxification Capacity of Probiotics and Postbiotics: A Critical Review.},
journal = {Probiotics and antimicrobial proteins},
volume = {},
number = {},
pages = {},
pmid = {40512444},
issn = {1867-1314},
abstract = {Environmental and chemical contaminants from industrial and agricultural sources increasingly threaten food safety worldwide. These contaminants, including bacterial toxins (e.g., botulinum toxin), mycotoxins (e.g., aflatoxin, ochratoxin), pesticides, heavy metals, and microplastics, pose significant health and economic risks. Emerging evidence highlights that chronic exposure to such xenobiotics disrupts gut microbiota structure and function, adversely affecting host health. While traditional physicochemical detoxification methods exist, they often require impractical conditions. Biological detoxification via probiotics and their metabolites (postbiotics) has gained attention as a practical and cost-effective alternative. Numerous studies concur that probiotics can bind, biotransform, or sequester xenobiotics, thereby mitigating toxicity and restoring microbiota balance. However, variations in strain efficacy, binding mechanisms, and in vivo performance indicate areas needing further exploration. This review critically synthesizes current knowledge on probiotic-mediated detoxification mechanisms, their interaction with xenobiotics and the gut microbiome, and host responses, highlighting consensus, discrepancies, and gaps to inform future research and applications.},
}
RevDate: 2025-06-13
Candida albicans: Insights into the Biology and Experimental Innovations of a Commonly Isolated Human Fungal Pathogen.
ACS infectious diseases [Epub ahead of print].
Candida albicans is an opportunistic pathogen associated with healthy humans. It is the major causative agent for superficial and invasive candidiasis in immunocompromised individuals globally. Lack of awareness toward fungal infections, poor disease management, and increasing drug resistance have contributed to the burden of C. albicans-associated diseases. C. albicans displays polymorphism, assisting in host interaction, virulence, and sustenance of the pathogen. Well-known polymorphic forms of C. albicans include yeast, hyphal, pseudohyphal, white, opaque, gray, GUT, and chlamydospores. While the predominant mode of propagation is via mitosis, the organism also displays a parasexual cycle. The pathogen's parasexual mating and genome plasticity introduce variants in the population, leading to adaptability and drug resistance. This organism can adopt robust pathogenic modules capable of destroying host tissues and forming drug-resistant biofilms. On the other hand, its ability to maintain an amicable interaction with the host immune system, coexisting microbiome, and adaptability to the host microenvironment makes it a successful commensal. Over the years, C. albicans has emerged as a useful model for understanding basic cell biology and drug resistance in fungal pathogens. Advancements in genetic manipulation tools have accelerated our understanding of these biological processes and eased the drug development process. This review provides comprehensive information about C. albicans and the tools available to investigate it. Studying its life cycle, disease epidemiology, mechanisms of infection, and host immune responses is crucial for developing effective therapeutic interventions.
Additional Links: PMID-40512087
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40512087,
year = {2025},
author = {Sahoo, B and Goyal, R and Dutta, S and Joshi, P and Sanyal, K},
title = {Candida albicans: Insights into the Biology and Experimental Innovations of a Commonly Isolated Human Fungal Pathogen.},
journal = {ACS infectious diseases},
volume = {},
number = {},
pages = {},
doi = {10.1021/acsinfecdis.5c00079},
pmid = {40512087},
issn = {2373-8227},
abstract = {Candida albicans is an opportunistic pathogen associated with healthy humans. It is the major causative agent for superficial and invasive candidiasis in immunocompromised individuals globally. Lack of awareness toward fungal infections, poor disease management, and increasing drug resistance have contributed to the burden of C. albicans-associated diseases. C. albicans displays polymorphism, assisting in host interaction, virulence, and sustenance of the pathogen. Well-known polymorphic forms of C. albicans include yeast, hyphal, pseudohyphal, white, opaque, gray, GUT, and chlamydospores. While the predominant mode of propagation is via mitosis, the organism also displays a parasexual cycle. The pathogen's parasexual mating and genome plasticity introduce variants in the population, leading to adaptability and drug resistance. This organism can adopt robust pathogenic modules capable of destroying host tissues and forming drug-resistant biofilms. On the other hand, its ability to maintain an amicable interaction with the host immune system, coexisting microbiome, and adaptability to the host microenvironment makes it a successful commensal. Over the years, C. albicans has emerged as a useful model for understanding basic cell biology and drug resistance in fungal pathogens. Advancements in genetic manipulation tools have accelerated our understanding of these biological processes and eased the drug development process. This review provides comprehensive information about C. albicans and the tools available to investigate it. Studying its life cycle, disease epidemiology, mechanisms of infection, and host immune responses is crucial for developing effective therapeutic interventions.},
}
RevDate: 2025-06-13
Metabolome-microbiome modulations induced by ZIF8@ZIF67 supplementation in the midgut of Bombyx mori.
Insect molecular biology [Epub ahead of print].
Bombyx mori is an economically crucial lepidopteran insect, and its health is closely related to the gut microbiota and metabolic status. Our previous research confirmed that metal-organic frameworks nanocomposite ZIF8@ZIF67 supplementation can significantly enhance silkworm survival rates upon bacterial challenges without adversely affecting its growth or cocoon weight. To elucidate the mechanism, this study integrated non-targeted metabolomics and 16S rRNA sequencing to dissect ZIF8@ZIF67-driven midgut modulations. The results showed that ZIF8@ZIF67 supplementation led to significant metabolic shifts in the midgut of B. mori, with 96 metabolites up-regulated and 54 down-regulated in the negative ion model, and 117 up-regulated and 43 down-regulated in the positive ion model. Key pathways included glutathione metabolism, purine/pyrimidine metabolism, and histidine metabolism, which are involved in antioxidant defence, nucleotide and protein synthesis for growth and immunity regulation. Furthermore, the gut microbiota composition of B. mori was significantly altered after ZIF8@ZIF67 supplementation, with a marked increase in the relative abundance of Mammaliicoccus and Enterococcus. Lastly, the correlation between metabolites and microbiomes was analysed, including Enterococcus and 1-methylhistidine (r = 0.8895, p = 0.0001), Akkermansia and N-acetyl-d-galactosamine (r = 0.8674, p = 0.0003). These findings demonstrated that ZIF8@ZIF67 could optimise silkworm health by orchestrating metabolite-microbe interactions to enhance nutrient assimilation and oxidative stress resilience, while simultaneously activating pathways essential for growth regulation and immune function.
Additional Links: PMID-40512059
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40512059,
year = {2025},
author = {Sadiq, S and Shen, ZY and Jiao, X and Zhang, N and Wang, L and Xu, T and Wu, P and Khan, I},
title = {Metabolome-microbiome modulations induced by ZIF8@ZIF67 supplementation in the midgut of Bombyx mori.},
journal = {Insect molecular biology},
volume = {},
number = {},
pages = {},
doi = {10.1111/imb.13008},
pmid = {40512059},
issn = {1365-2583},
support = {32372948//National Natural Science Foundation of China/ ; BE2022355//Jiangsu Provincial Key Research and Development Program/ ; KYCX24_4142//Research Innovation Project of Postgraduate Students in Jiangsu Province/ ; //China Agriculture Research System of MOF and MARA/ ; },
abstract = {Bombyx mori is an economically crucial lepidopteran insect, and its health is closely related to the gut microbiota and metabolic status. Our previous research confirmed that metal-organic frameworks nanocomposite ZIF8@ZIF67 supplementation can significantly enhance silkworm survival rates upon bacterial challenges without adversely affecting its growth or cocoon weight. To elucidate the mechanism, this study integrated non-targeted metabolomics and 16S rRNA sequencing to dissect ZIF8@ZIF67-driven midgut modulations. The results showed that ZIF8@ZIF67 supplementation led to significant metabolic shifts in the midgut of B. mori, with 96 metabolites up-regulated and 54 down-regulated in the negative ion model, and 117 up-regulated and 43 down-regulated in the positive ion model. Key pathways included glutathione metabolism, purine/pyrimidine metabolism, and histidine metabolism, which are involved in antioxidant defence, nucleotide and protein synthesis for growth and immunity regulation. Furthermore, the gut microbiota composition of B. mori was significantly altered after ZIF8@ZIF67 supplementation, with a marked increase in the relative abundance of Mammaliicoccus and Enterococcus. Lastly, the correlation between metabolites and microbiomes was analysed, including Enterococcus and 1-methylhistidine (r = 0.8895, p = 0.0001), Akkermansia and N-acetyl-d-galactosamine (r = 0.8674, p = 0.0003). These findings demonstrated that ZIF8@ZIF67 could optimise silkworm health by orchestrating metabolite-microbe interactions to enhance nutrient assimilation and oxidative stress resilience, while simultaneously activating pathways essential for growth regulation and immune function.},
}
RevDate: 2025-06-13
Correlation with viruses enhances network complexity and stability of co-occurrence prokaryotes across the oceans.
mSystems [Epub ahead of print].
UNLABELLED: Viruses play critical roles in shaping microbial distribution, organization, and ecological functions. However, the mechanisms behind the association and structuring of microbiomes with viruses still remain largely unknown. To understand how viruses affect the assembly of marine microbiomes, the network complexity and stability of prokaryotic microbiomes with/without viruses were analyzed and compared using the Tara Ocean Project data set, including samples collected from the Indian Ocean (IO), South Pacific Ocean (SPO), North Pacific Ocean (NPO), Mediterranean Sea, and South Atlantic Ocean. The network complexity of prokaryotic communities was substantially different across oceans, being high in the IO and low in the SPO and NPO. Network complexity and stability of marine prokaryotic microbial communities were enhanced by the influence of viruses: microbiomes associated with viruses were more complex, had a high modularity, and were robust with low fragmentation and vulnerability and higher compositional stability than prokaryotic microbiomes without an association with viruses. Results presented here shed light on how the associations with viruses influenced microbial assembly and their co-occurrence pattern and stability across the oceans. Overall, given such changed environmental characteristics across the oceans, stable and resilient microbiome networks imply that biotic associations with viruses play a critical role in maintaining the integrity and resilience of marine microbiomes and their ecological functions.
IMPORTANCE: This study represents the first detailed research on the effects of viruses on the organization of marine prokaryotic microbiomes at the global scale. Biotic factors and environmental heterogeneity directly and indirectly affect microbiome and viral co-occurrence with varied strength. Network complexity and stability of marine microbial communities were enhanced by the influence of viruses. Thus, given such dynamic environmental gradients in the ocean, stable and persistent microbiome networks suggest that biotic associations with viruses play a critical role in maintaining the integrity and resilience of marine microbiomes and influencing the function of marine ecosystems.
Additional Links: PMID-40511943
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40511943,
year = {2025},
author = {Wang, B and Liang, Y and Lian, K and Zhang, C and Han, M and Wang, M and Shao, H and McMinn, A and Wang, H},
title = {Correlation with viruses enhances network complexity and stability of co-occurrence prokaryotes across the oceans.},
journal = {mSystems},
volume = {},
number = {},
pages = {e0053925},
doi = {10.1128/msystems.00539-25},
pmid = {40511943},
issn = {2379-5077},
abstract = {UNLABELLED: Viruses play critical roles in shaping microbial distribution, organization, and ecological functions. However, the mechanisms behind the association and structuring of microbiomes with viruses still remain largely unknown. To understand how viruses affect the assembly of marine microbiomes, the network complexity and stability of prokaryotic microbiomes with/without viruses were analyzed and compared using the Tara Ocean Project data set, including samples collected from the Indian Ocean (IO), South Pacific Ocean (SPO), North Pacific Ocean (NPO), Mediterranean Sea, and South Atlantic Ocean. The network complexity of prokaryotic communities was substantially different across oceans, being high in the IO and low in the SPO and NPO. Network complexity and stability of marine prokaryotic microbial communities were enhanced by the influence of viruses: microbiomes associated with viruses were more complex, had a high modularity, and were robust with low fragmentation and vulnerability and higher compositional stability than prokaryotic microbiomes without an association with viruses. Results presented here shed light on how the associations with viruses influenced microbial assembly and their co-occurrence pattern and stability across the oceans. Overall, given such changed environmental characteristics across the oceans, stable and resilient microbiome networks imply that biotic associations with viruses play a critical role in maintaining the integrity and resilience of marine microbiomes and their ecological functions.
IMPORTANCE: This study represents the first detailed research on the effects of viruses on the organization of marine prokaryotic microbiomes at the global scale. Biotic factors and environmental heterogeneity directly and indirectly affect microbiome and viral co-occurrence with varied strength. Network complexity and stability of marine microbial communities were enhanced by the influence of viruses. Thus, given such dynamic environmental gradients in the ocean, stable and persistent microbiome networks suggest that biotic associations with viruses play a critical role in maintaining the integrity and resilience of marine microbiomes and influencing the function of marine ecosystems.},
}
RevDate: 2025-06-13
The cell wall hydrolase MltG is essential to maintain cell wall homeostasis of Enterococcus faecalis.
Journal of bacteriology [Epub ahead of print].
UNLABELLED: Infections caused by enterococci are increasingly prevalent and difficult to treat due to multidrug resistance. Enterococcus faecalis exhibits intrinsic resistance toward cephalosporins, which inhibit the final step of peptidoglycan (PG) synthesis. Intrinsic resistance requires multiple factors in the PG synthesis pathway and at least two cell-wall-stress signal transduction systems; however, the complete molecular mechanism of enterococcal cephalosporin resistance remains to be elucidated. MltG, a predicted PG hydrolase, is thought to process nascent strands of PG, suggesting that MltG might play an important role in enterococcal cell wall homeostasis and potentially cephalosporin resistance. Here, we demonstrate that enterococcal MltG cleaves nascent PG. An E. faecalis mutant lacking MltG exhibits several related phenotypes in the absence of exogenous stress: a marked growth defect, a loss of cell wall integrity, a reduction in PG synthesis, and activation of two cell-wall-stress signal transduction systems that drive elevated cephalosporin resistance. Together, these results are consistent with the model that MltG promotes proper cell wall homeostasis in E. faecalis, and further reveal that the enzymatic activity of MltG is not necessary for it to perform this function-instead, the LysM (putative PG-binding) domain of MltG plays the critical role. Nevertheless, the enzymatic activity of MltG does impact cephalosporin resistance, because a catalytically inactive MltG variant leads to elevated resistance. Collectively, our findings represent the first description of MltG function in E. faecalis and point to at least two distinct roles for MltG in PG homeostasis and cephalosporin resistance.
IMPORTANCE: Enterococcus faecalis is an opportunistic pathogen that colonizes the human gut microbiome. Infections caused by E. faecalis are increasingly prevalent and difficult to treat due to the multidrug resistance exhibited toward common clinical antibiotics. A thorough understanding of the mechanisms used by E. faecalis to maintain cell wall homeostasis will serve as a foundation for future development of new therapeutics that disable enterococcal resistance to cell-wall-active antibiotics and may reveal new vulnerabilities that could be exploited by novel antimicrobials. Here, we demonstrate that the MltG peptidoglycan hydrolase is essential for enterococcal cell wall homeostasis, but that the enzymatic activity of MltG is not required for this role. Instead, the enzymatic activity of MltG impacts intrinsic resistance toward cephalosporins.
Additional Links: PMID-40511937
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40511937,
year = {2025},
author = {Knotek, AAU and Kristich, CJ},
title = {The cell wall hydrolase MltG is essential to maintain cell wall homeostasis of Enterococcus faecalis.},
journal = {Journal of bacteriology},
volume = {},
number = {},
pages = {e0005625},
doi = {10.1128/jb.00056-25},
pmid = {40511937},
issn = {1098-5530},
abstract = {UNLABELLED: Infections caused by enterococci are increasingly prevalent and difficult to treat due to multidrug resistance. Enterococcus faecalis exhibits intrinsic resistance toward cephalosporins, which inhibit the final step of peptidoglycan (PG) synthesis. Intrinsic resistance requires multiple factors in the PG synthesis pathway and at least two cell-wall-stress signal transduction systems; however, the complete molecular mechanism of enterococcal cephalosporin resistance remains to be elucidated. MltG, a predicted PG hydrolase, is thought to process nascent strands of PG, suggesting that MltG might play an important role in enterococcal cell wall homeostasis and potentially cephalosporin resistance. Here, we demonstrate that enterococcal MltG cleaves nascent PG. An E. faecalis mutant lacking MltG exhibits several related phenotypes in the absence of exogenous stress: a marked growth defect, a loss of cell wall integrity, a reduction in PG synthesis, and activation of two cell-wall-stress signal transduction systems that drive elevated cephalosporin resistance. Together, these results are consistent with the model that MltG promotes proper cell wall homeostasis in E. faecalis, and further reveal that the enzymatic activity of MltG is not necessary for it to perform this function-instead, the LysM (putative PG-binding) domain of MltG plays the critical role. Nevertheless, the enzymatic activity of MltG does impact cephalosporin resistance, because a catalytically inactive MltG variant leads to elevated resistance. Collectively, our findings represent the first description of MltG function in E. faecalis and point to at least two distinct roles for MltG in PG homeostasis and cephalosporin resistance.
IMPORTANCE: Enterococcus faecalis is an opportunistic pathogen that colonizes the human gut microbiome. Infections caused by E. faecalis are increasingly prevalent and difficult to treat due to the multidrug resistance exhibited toward common clinical antibiotics. A thorough understanding of the mechanisms used by E. faecalis to maintain cell wall homeostasis will serve as a foundation for future development of new therapeutics that disable enterococcal resistance to cell-wall-active antibiotics and may reveal new vulnerabilities that could be exploited by novel antimicrobials. Here, we demonstrate that the MltG peptidoglycan hydrolase is essential for enterococcal cell wall homeostasis, but that the enzymatic activity of MltG is not required for this role. Instead, the enzymatic activity of MltG impacts intrinsic resistance toward cephalosporins.},
}
RevDate: 2025-06-13
Live Malassezia strains from the mucosa of patients with ulcerative colitis: pathogenic potential and environmental adaptations.
mBio [Epub ahead of print].
UNLABELLED: The human gut contains a diverse range of microorganisms, including bacteria, viruses, protozoa, and fungi. Although research has predominantly focused on bacterial populations, emerging evidence highlights the significant role of the gut mycobiota, particularly in the context of inflammatory bowel disease (IBD), including ulcerative colitis (UC) and Crohn's disease. This study investigates the intestinal mucosal mycobiota of UC patients, aiming to isolate and characterize live Malassezia fungal strains from the gut mucosa. Our analysis confirmed the presence of Malassezia in the intestinal mucosal layer of UC patients, with live Malassezia globosa strains being successfully isolated. As Malassezia is generally associated with the skin, the genomic and transcriptomic profiles and virulence of the M. globosa gut isolates were compared with those of the skin isolates. While both gut and skin isolates of M. globosa shared high genomic similarity, transcriptomic analysis revealed distinct responses to oxygen levels, suggesting niche-specific adaptation. Compared with the skin isolates, the gut isolates exhibited higher virulence in a dextran sulfate sodium-induced mouse colitis model, resulting in more severe disease, reduced survival rates, and elevated proinflammatory cytokine levels in the host. Our findings highlight the potential role of M. globosa in the pathogenesis of IBD and underscore the importance of niche-specific adaptations in fungal virulence.
IMPORTANCE: Malassezia fungi predominantly reside on human skin and are associated with several skin diseases, such as seborrheic dermatitis. They have also been implicated in various other diseases, including inflammatory bowel disease (IBD). While Malassezia DNA has been detected in many fungal microbiome studies using fecal samples, no previous research had isolated live Malassezia strains from the gut or confirmed that live Malassezia cells reside within the gut environment. In this study, we successfully isolated live Malassezia globosa strains from the gut mucosal surface of ulcerative colitis patients and compared them to M. globosa skin isolates. Our results revealed significant differences in pathogenicity between the gut and skin isolates and suggest the important role of M. globosa in the gut and its involvement in IBD.
Additional Links: PMID-40511923
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40511923,
year = {2025},
author = {Cho, Y-J and Shin, SY and Yang, J and Kim, HK and Rintarhat, P and Park, M and Sung, M and Lagree, K and Underhill, DM and Lee, D-W and Choi, S and Choi, CH and Yang, C-S and Jung, WH},
title = {Live Malassezia strains from the mucosa of patients with ulcerative colitis: pathogenic potential and environmental adaptations.},
journal = {mBio},
volume = {},
number = {},
pages = {e0140025},
doi = {10.1128/mbio.01400-25},
pmid = {40511923},
issn = {2150-7511},
abstract = {UNLABELLED: The human gut contains a diverse range of microorganisms, including bacteria, viruses, protozoa, and fungi. Although research has predominantly focused on bacterial populations, emerging evidence highlights the significant role of the gut mycobiota, particularly in the context of inflammatory bowel disease (IBD), including ulcerative colitis (UC) and Crohn's disease. This study investigates the intestinal mucosal mycobiota of UC patients, aiming to isolate and characterize live Malassezia fungal strains from the gut mucosa. Our analysis confirmed the presence of Malassezia in the intestinal mucosal layer of UC patients, with live Malassezia globosa strains being successfully isolated. As Malassezia is generally associated with the skin, the genomic and transcriptomic profiles and virulence of the M. globosa gut isolates were compared with those of the skin isolates. While both gut and skin isolates of M. globosa shared high genomic similarity, transcriptomic analysis revealed distinct responses to oxygen levels, suggesting niche-specific adaptation. Compared with the skin isolates, the gut isolates exhibited higher virulence in a dextran sulfate sodium-induced mouse colitis model, resulting in more severe disease, reduced survival rates, and elevated proinflammatory cytokine levels in the host. Our findings highlight the potential role of M. globosa in the pathogenesis of IBD and underscore the importance of niche-specific adaptations in fungal virulence.
IMPORTANCE: Malassezia fungi predominantly reside on human skin and are associated with several skin diseases, such as seborrheic dermatitis. They have also been implicated in various other diseases, including inflammatory bowel disease (IBD). While Malassezia DNA has been detected in many fungal microbiome studies using fecal samples, no previous research had isolated live Malassezia strains from the gut or confirmed that live Malassezia cells reside within the gut environment. In this study, we successfully isolated live Malassezia globosa strains from the gut mucosal surface of ulcerative colitis patients and compared them to M. globosa skin isolates. Our results revealed significant differences in pathogenicity between the gut and skin isolates and suggest the important role of M. globosa in the gut and its involvement in IBD.},
}
RevDate: 2025-06-13
Atomic Ce-Induced Adaptive Synergism for Self-Optimized Multi-Enzymatic Nanozyme Design for Soil Amendment.
Small (Weinheim an der Bergstrasse, Germany) [Epub ahead of print].
Single-atom nanozymes (SAzymes) offer exceptional promise as next-generation substitutes for natural enzymes due to its superior catalytic performance. However, single active sites of SAzymes have predominantly been limited to mimicking a monofunctional enzymatic activity, which hinders their adaptability in complex biological systems. Herein, an innovative Ce-induced adaptive synergism is reported, which allows Ce single-atom to activate Ni-Fe dual-site adaptive synergy for boosting multi-enzymatic activities of Ce-modified nanozyme (Ce-MOF), along with finely tuned reactive oxygen species (ROS) production. In-situ spectroscopic studies and theoretical calculations reveal that Ce induces spin-state polarization of Ni and Fe centers, promoting the generation of bound Ni/Fe = O intermediates that modulate ROS pathways depending on substrate and pH. This adaptive synergism enables continuous structural adaptation of the dual-metal active sites, enhancing both catalytic efficiency and selectivity. Functionally, Ce-MOF exhibits potent antifungal activity against Fusarium solani in vitro without the introduction of exogenous O2 or H2O2. Meanwhile, soil experiments demonstrate effective inhibition of phytopathogen colonization and a notable recovery of beneficial microbial communities via selective production of ROS. Furthermore, the amended soil treated with Ce-MOF flourishes Panax ginseng growth and provides a promising solution for sustainable plant cultivation via rhizosphere microbiome regulation.
Additional Links: PMID-40511703
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40511703,
year = {2025},
author = {Li, Q and Cha, Y and Zhan, Y and Miao, X and He, G and Lv, P and Wang, L and Li, W and Chen, C},
title = {Atomic Ce-Induced Adaptive Synergism for Self-Optimized Multi-Enzymatic Nanozyme Design for Soil Amendment.},
journal = {Small (Weinheim an der Bergstrasse, Germany)},
volume = {},
number = {},
pages = {e2503939},
doi = {10.1002/smll.202503939},
pmid = {40511703},
issn = {1613-6829},
support = {82204558//National Natural Science Foundation of China/ ; YDZJ202401074ZYTS//Natural Science Foundation of Jilin Province/ ; },
abstract = {Single-atom nanozymes (SAzymes) offer exceptional promise as next-generation substitutes for natural enzymes due to its superior catalytic performance. However, single active sites of SAzymes have predominantly been limited to mimicking a monofunctional enzymatic activity, which hinders their adaptability in complex biological systems. Herein, an innovative Ce-induced adaptive synergism is reported, which allows Ce single-atom to activate Ni-Fe dual-site adaptive synergy for boosting multi-enzymatic activities of Ce-modified nanozyme (Ce-MOF), along with finely tuned reactive oxygen species (ROS) production. In-situ spectroscopic studies and theoretical calculations reveal that Ce induces spin-state polarization of Ni and Fe centers, promoting the generation of bound Ni/Fe = O intermediates that modulate ROS pathways depending on substrate and pH. This adaptive synergism enables continuous structural adaptation of the dual-metal active sites, enhancing both catalytic efficiency and selectivity. Functionally, Ce-MOF exhibits potent antifungal activity against Fusarium solani in vitro without the introduction of exogenous O2 or H2O2. Meanwhile, soil experiments demonstrate effective inhibition of phytopathogen colonization and a notable recovery of beneficial microbial communities via selective production of ROS. Furthermore, the amended soil treated with Ce-MOF flourishes Panax ginseng growth and provides a promising solution for sustainable plant cultivation via rhizosphere microbiome regulation.},
}
RevDate: 2025-06-14
CmpDate: 2025-06-13
Exploring the systemic impacts of urinary tract infection-specific antibiotic treatments on the gut microbiome, metabolome, and intestinal morphology in rats.
PeerJ, 13:e19486.
The gut microbiota is a dynamic ecosystem that plays a crucial role in host metabolism, immune system regulation, and protection against pathogens. An imbalanced gut microbiota has been associated with various diseases such as inflammatory bowel disease, metabolic disorders, and autoimmune diseases. Antibiotic therapy can disrupt the balance of the microbiome, making it essential to understand the specific effects of these antibiotics on gut microbiota and related host function. This study aims to systematically investigate the impact of UTI-specific antibiotic treatment on the gut microbiota of rats. By analyzing changes in microbial composition and their effects on host metabolism and intestinal structure, we seek to provide new insights into the broader consequences of antibiotic intervention on host-microbiota interactions. The research findings indicate that antibiotic treatment leads to a significant disruption in microbial diversity and metabolic characteristics, as well as notable histological changes in the intestinal mucosa. These results highlight the need for cautious use of antibiotics, considering their extensive effects beyond antimicrobial activity. Future research should focus on strategies to mitigate these impacts, potentially through targeted antibiotic therapies or probiotics, to better balance treatment efficacy with health preservation.
Additional Links: PMID-40511381
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40511381,
year = {2025},
author = {Guo, H and Zhou, X and Li, Z and Zhi, J and Fu, C and Liu, X and Wu, Y and Liu, F and Feng, N},
title = {Exploring the systemic impacts of urinary tract infection-specific antibiotic treatments on the gut microbiome, metabolome, and intestinal morphology in rats.},
journal = {PeerJ},
volume = {13},
number = {},
pages = {e19486},
pmid = {40511381},
issn = {2167-8359},
mesh = {Animals ; *Gastrointestinal Microbiome/drug effects ; *Anti-Bacterial Agents/pharmacology/adverse effects ; Rats ; *Metabolome/drug effects ; *Urinary Tract Infections/drug therapy/microbiology ; *Intestinal Mucosa/drug effects/pathology ; *Intestines/drug effects/pathology ; Male ; Rats, Sprague-Dawley ; },
abstract = {The gut microbiota is a dynamic ecosystem that plays a crucial role in host metabolism, immune system regulation, and protection against pathogens. An imbalanced gut microbiota has been associated with various diseases such as inflammatory bowel disease, metabolic disorders, and autoimmune diseases. Antibiotic therapy can disrupt the balance of the microbiome, making it essential to understand the specific effects of these antibiotics on gut microbiota and related host function. This study aims to systematically investigate the impact of UTI-specific antibiotic treatment on the gut microbiota of rats. By analyzing changes in microbial composition and their effects on host metabolism and intestinal structure, we seek to provide new insights into the broader consequences of antibiotic intervention on host-microbiota interactions. The research findings indicate that antibiotic treatment leads to a significant disruption in microbial diversity and metabolic characteristics, as well as notable histological changes in the intestinal mucosa. These results highlight the need for cautious use of antibiotics, considering their extensive effects beyond antimicrobial activity. Future research should focus on strategies to mitigate these impacts, potentially through targeted antibiotic therapies or probiotics, to better balance treatment efficacy with health preservation.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Animals
*Gastrointestinal Microbiome/drug effects
*Anti-Bacterial Agents/pharmacology/adverse effects
Rats
*Metabolome/drug effects
*Urinary Tract Infections/drug therapy/microbiology
*Intestinal Mucosa/drug effects/pathology
*Intestines/drug effects/pathology
Male
Rats, Sprague-Dawley
RevDate: 2025-06-14
Sweet Shield: Enhancing Oral Health with Bioactive HETAFU Candies in Adults - A Dose-Dependent Study.
Journal of pharmacy & bioallied sciences, 17(Suppl 1):S133-S136.
Functional foods incorporating probiotics, essential oils, and DHA, PhosphatidylSerine have been explored as innovative approaches to improving oral health. This study evaluates the effects of consuming two versus five HETAFU candies daily on oral microbial levels over eight weeks. To compare the impact of moderate (two candies/day) and higher (five candies/day) HETAFU candy consumption on oral microbial populations, focusing on Streptococcus mutans, Lactobacillus, Actinomycetes, and Candida. A total of 200 participants were randomly divided into two groups: Group 1 consumed two candies daily, and Group 2 consumed five candies daily. Microbial levels were measured at baseline, 2 weeks, 4 weeks, and 8 weeks using microbiological techniques. Statistical analysis included Z scores and P values to determine the significance of microbial changes between groups. Both groups showed significant reductions in Streptococcus mutans and Candida, with a concurrent decrease in Lactobacillus and Actinomycetes levels over time. However, Group 2 demonstrated greater microbial shifts, indicating enhanced modulation of the oral microbiome with higher candy consumption. Higher consumption of HETAFU candies (five/day) provides superior benefits in reducing pathogenic microbes and promoting beneficial bacteria compared to moderate consumption (two/day). This suggests a dose-dependent relationship, with increased intake offering additional protective effects against oral microbial imbalances. Functional foods like HETAFU candies represent a promising adjunct to oral hygiene practices.
Additional Links: PMID-40511069
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40511069,
year = {2025},
author = {Lakkoju, S and Bolisetty, D and Kumar, R},
title = {Sweet Shield: Enhancing Oral Health with Bioactive HETAFU Candies in Adults - A Dose-Dependent Study.},
journal = {Journal of pharmacy & bioallied sciences},
volume = {17},
number = {Suppl 1},
pages = {S133-S136},
pmid = {40511069},
issn = {0976-4879},
abstract = {Functional foods incorporating probiotics, essential oils, and DHA, PhosphatidylSerine have been explored as innovative approaches to improving oral health. This study evaluates the effects of consuming two versus five HETAFU candies daily on oral microbial levels over eight weeks. To compare the impact of moderate (two candies/day) and higher (five candies/day) HETAFU candy consumption on oral microbial populations, focusing on Streptococcus mutans, Lactobacillus, Actinomycetes, and Candida. A total of 200 participants were randomly divided into two groups: Group 1 consumed two candies daily, and Group 2 consumed five candies daily. Microbial levels were measured at baseline, 2 weeks, 4 weeks, and 8 weeks using microbiological techniques. Statistical analysis included Z scores and P values to determine the significance of microbial changes between groups. Both groups showed significant reductions in Streptococcus mutans and Candida, with a concurrent decrease in Lactobacillus and Actinomycetes levels over time. However, Group 2 demonstrated greater microbial shifts, indicating enhanced modulation of the oral microbiome with higher candy consumption. Higher consumption of HETAFU candies (five/day) provides superior benefits in reducing pathogenic microbes and promoting beneficial bacteria compared to moderate consumption (two/day). This suggests a dose-dependent relationship, with increased intake offering additional protective effects against oral microbial imbalances. Functional foods like HETAFU candies represent a promising adjunct to oral hygiene practices.},
}
RevDate: 2025-06-14
Navigating the Vaginal Milieu During Perimenopause: A Narrative Review of Physiological Changes and Clinical Implications.
Journal of pharmacy & bioallied sciences, 17(Suppl 1):S92-S95.
Perimenopause represents a transitional phase in a woman's life, marked by significant hormonal and physiological changes that profoundly impact the vaginal milieu. These changes can lead to a variety of symptoms, collectively known as genitourinary syndrome of menopause, affecting a woman's sexual health, quality of life, and overall well-being. This narrative review aims to explore the underlying physiological alterations in the vaginal environment during perimenopause, their clinical implications, and current therapeutic approaches. By highlighting the challenges and emerging strategies in managing these changes, this review provides insights into personalized treatment options for improving women's health during this crucial period.
Additional Links: PMID-40511019
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40511019,
year = {2025},
author = {Tiwari, M and Acharya, N and Mahakarkar, M},
title = {Navigating the Vaginal Milieu During Perimenopause: A Narrative Review of Physiological Changes and Clinical Implications.},
journal = {Journal of pharmacy & bioallied sciences},
volume = {17},
number = {Suppl 1},
pages = {S92-S95},
pmid = {40511019},
issn = {0976-4879},
abstract = {Perimenopause represents a transitional phase in a woman's life, marked by significant hormonal and physiological changes that profoundly impact the vaginal milieu. These changes can lead to a variety of symptoms, collectively known as genitourinary syndrome of menopause, affecting a woman's sexual health, quality of life, and overall well-being. This narrative review aims to explore the underlying physiological alterations in the vaginal environment during perimenopause, their clinical implications, and current therapeutic approaches. By highlighting the challenges and emerging strategies in managing these changes, this review provides insights into personalized treatment options for improving women's health during this crucial period.},
}
RevDate: 2025-06-14
The Role of Skin Dysbiosis and Quorum Sensing in Atopic Dermatitis.
JID innovations : skin science from molecules to population health, 5(4):100377.
The skin microbiome plays a crucial role in the pathogenesis of atopic dermatitis (AD), a chronic inflammatory skin disorder strongly associated with microbial dysbiosis, particularly Staphylococcus aureus colonization. However, the mechanisms linking S aureus to AD remain insufficiently understood. This review explores the impact of the quorum-sensing (QS) system, particularly the accessory gene regulator Agr, in AD development and progression. By examining key microbial-host interactions, we provide insights into how QS influences skin inflammation and dysbiosis. Furthermore, we discuss the potential of microbiome-targeted therapeutic strategies to mitigate or prevent AD, highlighting their implications for future research and clinical applications.
Additional Links: PMID-40510906
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40510906,
year = {2025},
author = {Okamoto, H and Li, S and Nakamura, Y},
title = {The Role of Skin Dysbiosis and Quorum Sensing in Atopic Dermatitis.},
journal = {JID innovations : skin science from molecules to population health},
volume = {5},
number = {4},
pages = {100377},
pmid = {40510906},
issn = {2667-0267},
abstract = {The skin microbiome plays a crucial role in the pathogenesis of atopic dermatitis (AD), a chronic inflammatory skin disorder strongly associated with microbial dysbiosis, particularly Staphylococcus aureus colonization. However, the mechanisms linking S aureus to AD remain insufficiently understood. This review explores the impact of the quorum-sensing (QS) system, particularly the accessory gene regulator Agr, in AD development and progression. By examining key microbial-host interactions, we provide insights into how QS influences skin inflammation and dysbiosis. Furthermore, we discuss the potential of microbiome-targeted therapeutic strategies to mitigate or prevent AD, highlighting their implications for future research and clinical applications.},
}
RevDate: 2025-06-13
Editorial: Statistical approaches, applications, and software for longitudinal microbiome data analysis and microbiome multi-omics data integration.
Frontiers in genetics, 16:1624791.
Additional Links: PMID-40510813
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40510813,
year = {2025},
author = {Li, H and Chen, J and Fenger, M and Jiang, Y},
title = {Editorial: Statistical approaches, applications, and software for longitudinal microbiome data analysis and microbiome multi-omics data integration.},
journal = {Frontiers in genetics},
volume = {16},
number = {},
pages = {1624791},
pmid = {40510813},
issn = {1664-8021},
}
RevDate: 2025-06-13
CmpDate: 2025-06-13
Characterization of the salivary microbiome in healthy individuals under fatigue status.
Frontiers in cellular and infection microbiology, 15:1506723.
BACKGROUND & AIMS: Limited understanding exists regarding the characteristics and biological significance of the salivary microbiome in healthy individuals experiencing physiological fatigue. This study aimed to delineate the structural and functional alterations in the salivary microbiome of healthy individuals undergoing physiological fatigue compared to energetic controls, and to explore its potential as a biomarker for fatigue status.
METHODS: A cohort of 7 healthy individuals experiencing acute physiological fatigue (induced by prolonged study and confirmed via electroencephalography; Fatigue group, FTG) and 63 energetic healthy controls (Energetic group, ENG) were enrolled. Saliva samples were collected, from which microbial DNA was extracted. The V3-V4 hypervariable region of the 16S rRNA gene was subsequently sequenced using high-throughput technology. Bioinformatics analyses encompassed assessment of alpha and beta diversity, identification of differential taxa using Linear discriminant analysis Effect Size (LEfSe) with multi-method cross-validation, construction of microbial co-occurrence networks, and screening of fatigue-associated biomarker genera via the Boruta-SHAP algorithm. Microbial community phenotypes and potential functional pathways were predicted using BugBase and PICRUSt2, respectively.
RESULTS: The FTG group exhibited significantly diminished alpha diversity (Simpson index, p=0.01071) relative to the ENG group. Beta diversity analysis demonstrated significant dissimilarities in microbial community structure between the groups (p<0.05). Taxonomic profiling revealed a significant enrichment in the relative abundance of potential periodontopathogenic genera, including Streptococcus and Filifactor, within the FTG group, concomitantly with a significant depletion of health-associated genera such as Rothia and Neisseria. A predictive model constructed using the Boruta-SHAP algorithm, based on 15 key genera, effectively discriminated between fatigue and non-fatigue states, achieving an area under the receiver operating characteristic curve (AUC) of 0.948. Phenotypic predictions indicated a significant increase in the proportion of bacteria harboring Mobile Genetic Elements (MGEs) (p=0.048), alongside significant reductions in the proportion of aerobic bacteria (p=0.006) and biofilm-forming capacity (p=0.002) in the FTG group. Functional pathway analysis (PICRUSt2) revealed an enrichment of pathways such as "Neuroactive ligand-receptor interaction" in the FTG group, whereas pathways pertinent to energy metabolism (e.g., Citrate cycle (TCA cycle), Oxidative phosphorylation) and amino acid metabolism (e.g., Phenylalanine metabolism, Histidine metabolism) were significantly enriched in the ENG group.
CONCLUSION: This study provides novel evidence that physiological fatigue induces significant structural and functional alterations in the salivary microbiome of healthy individuals. These perturbations include diminished microbial diversity, disrupted community architecture, enrichment of potential opportunistic pathogens, and marked shifts in key metabolic pathways, particularly those governing neuroactivity and energy metabolism. These findings suggest that the salivary microbiome may be implicated in the physiological regulation of fatigue, potentially via an "oral-microbiome-brain axis," and underscore its potential as a source of non-invasive biomarkers for assessing fatigue status. Further mechanistic investigations are warranted to elucidate these interactions.
Additional Links: PMID-40510800
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40510800,
year = {2025},
author = {Peng, X and Han, N and Gong, Y and He, L and Xu, Y and Xiao, D and Zhang, T and Qiang, Y and Li, X and Zhang, W and Zhang, J},
title = {Characterization of the salivary microbiome in healthy individuals under fatigue status.},
journal = {Frontiers in cellular and infection microbiology},
volume = {15},
number = {},
pages = {1506723},
pmid = {40510800},
issn = {2235-2988},
mesh = {Humans ; *Saliva/microbiology ; *Microbiota ; RNA, Ribosomal, 16S/genetics ; Male ; Adult ; Female ; *Fatigue/microbiology ; Healthy Volunteers ; Biomarkers/analysis ; *Bacteria/classification/genetics/isolation & purification ; DNA, Bacterial/genetics ; Young Adult ; High-Throughput Nucleotide Sequencing ; Computational Biology ; Sequence Analysis, DNA ; Middle Aged ; },
abstract = {BACKGROUND & AIMS: Limited understanding exists regarding the characteristics and biological significance of the salivary microbiome in healthy individuals experiencing physiological fatigue. This study aimed to delineate the structural and functional alterations in the salivary microbiome of healthy individuals undergoing physiological fatigue compared to energetic controls, and to explore its potential as a biomarker for fatigue status.
METHODS: A cohort of 7 healthy individuals experiencing acute physiological fatigue (induced by prolonged study and confirmed via electroencephalography; Fatigue group, FTG) and 63 energetic healthy controls (Energetic group, ENG) were enrolled. Saliva samples were collected, from which microbial DNA was extracted. The V3-V4 hypervariable region of the 16S rRNA gene was subsequently sequenced using high-throughput technology. Bioinformatics analyses encompassed assessment of alpha and beta diversity, identification of differential taxa using Linear discriminant analysis Effect Size (LEfSe) with multi-method cross-validation, construction of microbial co-occurrence networks, and screening of fatigue-associated biomarker genera via the Boruta-SHAP algorithm. Microbial community phenotypes and potential functional pathways were predicted using BugBase and PICRUSt2, respectively.
RESULTS: The FTG group exhibited significantly diminished alpha diversity (Simpson index, p=0.01071) relative to the ENG group. Beta diversity analysis demonstrated significant dissimilarities in microbial community structure between the groups (p<0.05). Taxonomic profiling revealed a significant enrichment in the relative abundance of potential periodontopathogenic genera, including Streptococcus and Filifactor, within the FTG group, concomitantly with a significant depletion of health-associated genera such as Rothia and Neisseria. A predictive model constructed using the Boruta-SHAP algorithm, based on 15 key genera, effectively discriminated between fatigue and non-fatigue states, achieving an area under the receiver operating characteristic curve (AUC) of 0.948. Phenotypic predictions indicated a significant increase in the proportion of bacteria harboring Mobile Genetic Elements (MGEs) (p=0.048), alongside significant reductions in the proportion of aerobic bacteria (p=0.006) and biofilm-forming capacity (p=0.002) in the FTG group. Functional pathway analysis (PICRUSt2) revealed an enrichment of pathways such as "Neuroactive ligand-receptor interaction" in the FTG group, whereas pathways pertinent to energy metabolism (e.g., Citrate cycle (TCA cycle), Oxidative phosphorylation) and amino acid metabolism (e.g., Phenylalanine metabolism, Histidine metabolism) were significantly enriched in the ENG group.
CONCLUSION: This study provides novel evidence that physiological fatigue induces significant structural and functional alterations in the salivary microbiome of healthy individuals. These perturbations include diminished microbial diversity, disrupted community architecture, enrichment of potential opportunistic pathogens, and marked shifts in key metabolic pathways, particularly those governing neuroactivity and energy metabolism. These findings suggest that the salivary microbiome may be implicated in the physiological regulation of fatigue, potentially via an "oral-microbiome-brain axis," and underscore its potential as a source of non-invasive biomarkers for assessing fatigue status. Further mechanistic investigations are warranted to elucidate these interactions.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
*Saliva/microbiology
*Microbiota
RNA, Ribosomal, 16S/genetics
Male
Adult
Female
*Fatigue/microbiology
Healthy Volunteers
Biomarkers/analysis
*Bacteria/classification/genetics/isolation & purification
DNA, Bacterial/genetics
Young Adult
High-Throughput Nucleotide Sequencing
Computational Biology
Sequence Analysis, DNA
Middle Aged
RevDate: 2025-06-13
Association between oral microbiome diversity and kidney stones: a cross-sectional study.
Frontiers in microbiology, 16:1600961.
BACKGROUND: Kidney stones are a common urologic disorder that imposes a significant burden on global public health. This study aimed to determine the association between oral microbiome diversity and kidney stones.
METHODS: The data for this study came from the National Health and Nutrition Examination Survey 2009-2012 survey cycle. Use of alpha diversity to assess oral microbiome diversity. Multivariate logistic regression modeling was used to assess the association between different alpha-diversity indicators and kidney stones. Subgroup analyses and interaction tests were used to assess the stability of the association between alpha-diversity and kidney stones. Restricted cubic spline plots were used to assess non-linear associations and dose-response relationships.
RESULTS: The study included 5,870 eligible participants with a mean age of 43.74 years at baseline. After adjusting for all covariates, the observed oral microbiome diversity was significantly negatively associated with the risk of kidney stones (P < 0.05). Subgroup analyses showed that oral microbiome diversity was negatively associated with the risk of kidney stones in certain populations, particularly among those aged 40-60 years, men, obese, with moderate to high cardiovascular health scores, smokers, and those without hypertension. Restricted cubic spline analysis suggested a significant non-linear negative correlation between the Shannon and Simpson diversity indices and the risk of kidney stones (p for non-linear < 0.05). Since our study was a cross-sectional design, the main limitation was the inability to prove causality.
CONCLUSIONS: In this study, we found an inverse relationship between oral microbiome diversity and kidney stone risk observed in alpha diversity. This reveals the complexity of host-microbiome interactions, and further mechanistic studies are necessary to elucidate these complex roles in the future.
Additional Links: PMID-40510677
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40510677,
year = {2025},
author = {Zhang, Z and Guan, S and Chen, L and Jiang, F and Dong, H and Chen, Z and Lv, L and Song, H and Sun, W and He, D and Jiang, S and Tian, F},
title = {Association between oral microbiome diversity and kidney stones: a cross-sectional study.},
journal = {Frontiers in microbiology},
volume = {16},
number = {},
pages = {1600961},
pmid = {40510677},
issn = {1664-302X},
abstract = {BACKGROUND: Kidney stones are a common urologic disorder that imposes a significant burden on global public health. This study aimed to determine the association between oral microbiome diversity and kidney stones.
METHODS: The data for this study came from the National Health and Nutrition Examination Survey 2009-2012 survey cycle. Use of alpha diversity to assess oral microbiome diversity. Multivariate logistic regression modeling was used to assess the association between different alpha-diversity indicators and kidney stones. Subgroup analyses and interaction tests were used to assess the stability of the association between alpha-diversity and kidney stones. Restricted cubic spline plots were used to assess non-linear associations and dose-response relationships.
RESULTS: The study included 5,870 eligible participants with a mean age of 43.74 years at baseline. After adjusting for all covariates, the observed oral microbiome diversity was significantly negatively associated with the risk of kidney stones (P < 0.05). Subgroup analyses showed that oral microbiome diversity was negatively associated with the risk of kidney stones in certain populations, particularly among those aged 40-60 years, men, obese, with moderate to high cardiovascular health scores, smokers, and those without hypertension. Restricted cubic spline analysis suggested a significant non-linear negative correlation between the Shannon and Simpson diversity indices and the risk of kidney stones (p for non-linear < 0.05). Since our study was a cross-sectional design, the main limitation was the inability to prove causality.
CONCLUSIONS: In this study, we found an inverse relationship between oral microbiome diversity and kidney stone risk observed in alpha diversity. This reveals the complexity of host-microbiome interactions, and further mechanistic studies are necessary to elucidate these complex roles in the future.},
}
RevDate: 2025-06-13
Rhinoceromics: a multi-amplicon study with clinical markers to transferrin saturation levels in ex-situ black rhinoceros (Diceros bicornis michaeli).
Frontiers in microbiology, 16:1515939.
Iron overload disorder (IOD) is a common condition in ex-situ black rhinoceroses (Diceros bicornis), although it has not been reported in the wild. This study aimed to gain a deeper understanding of the relationship between 25-hydroxy vitamin D [25(OH)D], inflammatory markers, insulin levels, the gut microbiome, dietary components, and transferrin saturation (TS) in ex-situ black rhinoceroses. Blood and fecal samples from 11 black rhinoceroses at five different European zoological institutions were monitored over a 1-year period. Inflammatory markers such as interleukin 6 (IL-6), serum amyloid A (SAA), interferon γ (IFN-γ), and tumor necrosis factor α (TNF-α) were analyzed. Our study corroborates the findings of previous research, which demonstrated that insulin, inflammatory markers, and TS% are higher in ex-situ black rhinoceroses compared to published wild ranges. Our data show no correlations between insulin, 25(OH)D, TS%, inflammatory markers, or short-chain fatty acids (SFCAs). Serum 25(OH)D exhibited significantly higher levels in summer than in winter. Transferrin saturation was influenced by age, which is consistent with previous studies. The microbiome did not differ significantly among individuals, institutions, sex, or season, unlike the mycobiome, which exhibited significant differences across institutions. The impact of the mycobiome differences on the physiology of the animals could not be determined from this study.
Additional Links: PMID-40510676
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40510676,
year = {2025},
author = {Bruins-van Sonsbeek, LGR and Verschuren, MCM and Kaal, S and Lindenburg, PW and Rodenburg, KCW and Clauss, M and Speksnijder, AGCL and Rutten, VPMG and Bonnet, BFJ and Wittink, F},
title = {Rhinoceromics: a multi-amplicon study with clinical markers to transferrin saturation levels in ex-situ black rhinoceros (Diceros bicornis michaeli).},
journal = {Frontiers in microbiology},
volume = {16},
number = {},
pages = {1515939},
pmid = {40510676},
issn = {1664-302X},
abstract = {Iron overload disorder (IOD) is a common condition in ex-situ black rhinoceroses (Diceros bicornis), although it has not been reported in the wild. This study aimed to gain a deeper understanding of the relationship between 25-hydroxy vitamin D [25(OH)D], inflammatory markers, insulin levels, the gut microbiome, dietary components, and transferrin saturation (TS) in ex-situ black rhinoceroses. Blood and fecal samples from 11 black rhinoceroses at five different European zoological institutions were monitored over a 1-year period. Inflammatory markers such as interleukin 6 (IL-6), serum amyloid A (SAA), interferon γ (IFN-γ), and tumor necrosis factor α (TNF-α) were analyzed. Our study corroborates the findings of previous research, which demonstrated that insulin, inflammatory markers, and TS% are higher in ex-situ black rhinoceroses compared to published wild ranges. Our data show no correlations between insulin, 25(OH)D, TS%, inflammatory markers, or short-chain fatty acids (SFCAs). Serum 25(OH)D exhibited significantly higher levels in summer than in winter. Transferrin saturation was influenced by age, which is consistent with previous studies. The microbiome did not differ significantly among individuals, institutions, sex, or season, unlike the mycobiome, which exhibited significant differences across institutions. The impact of the mycobiome differences on the physiology of the animals could not be determined from this study.},
}
RevDate: 2025-06-13
Impact of the soil layer on the soil microbial diversity and composition of Pinus yunnanensis at the Ailao Mountains subtropical forest.
Frontiers in microbiology, 16:1558906.
Microbial communities residing in forest soils play crucial roles in decomposing organic matter and recycling nutrients, making these ecosystems one of the most diverse habitats on Earth. However, the composition and function of these complex and diverse microbiomes across different soil layers remain largely unknown. In this study, we collected soil samples from various layers and analysed the bacterial and fungal community compositions in experimental forest ecosystems using sequencing techniques. Our findings revealed that the soil layer was the primary factor influencing microbial communities, whereas sampling season had only a marginal effect. The most prevalent bacterial phyla and fungal classes were Acidobacteria, Actinobacteria, Armatimonadetes, Bacteroidetes, Firmicutes, Planctomycetes, Proteobacteria, Verrucomicrobia, and Agaricomycetes. Owing to the heterogeneity of the soil layer environment, we observed distinct patterns in the bacterial and fungal microbiomes across different layers. Moreover, the soil layer affected the network complexity, with fungi exhibiting higher complexity in the upper layer, whereas bacteria showed the opposite trend. Additionally, the dominant bacterial and fungal taxa across all soil layers belonged predominantly to Acidobacteria and Agaricomycetes, respectively. These findings underscore the significance of soil layers in shaping soil microbial communities and highlight the composition and co-occurrence patterns of the microbial communities within these layers.
Additional Links: PMID-40510675
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40510675,
year = {2025},
author = {Qiao, H and Zeng, Q and Martin, F and Wang, Q},
title = {Impact of the soil layer on the soil microbial diversity and composition of Pinus yunnanensis at the Ailao Mountains subtropical forest.},
journal = {Frontiers in microbiology},
volume = {16},
number = {},
pages = {1558906},
pmid = {40510675},
issn = {1664-302X},
abstract = {Microbial communities residing in forest soils play crucial roles in decomposing organic matter and recycling nutrients, making these ecosystems one of the most diverse habitats on Earth. However, the composition and function of these complex and diverse microbiomes across different soil layers remain largely unknown. In this study, we collected soil samples from various layers and analysed the bacterial and fungal community compositions in experimental forest ecosystems using sequencing techniques. Our findings revealed that the soil layer was the primary factor influencing microbial communities, whereas sampling season had only a marginal effect. The most prevalent bacterial phyla and fungal classes were Acidobacteria, Actinobacteria, Armatimonadetes, Bacteroidetes, Firmicutes, Planctomycetes, Proteobacteria, Verrucomicrobia, and Agaricomycetes. Owing to the heterogeneity of the soil layer environment, we observed distinct patterns in the bacterial and fungal microbiomes across different layers. Moreover, the soil layer affected the network complexity, with fungi exhibiting higher complexity in the upper layer, whereas bacteria showed the opposite trend. Additionally, the dominant bacterial and fungal taxa across all soil layers belonged predominantly to Acidobacteria and Agaricomycetes, respectively. These findings underscore the significance of soil layers in shaping soil microbial communities and highlight the composition and co-occurrence patterns of the microbial communities within these layers.},
}
RevDate: 2025-06-13
Intermittent fasting inhibits the development of colorectal cancer in APC [Min/+] mice through gut microbiota and its related metabolites.
Frontiers in microbiology, 16:1563224.
BACKGROUND AND OBJECTIVES: Intermittent fasting is an emerging dietary approach, but its specific role in colorectal cancer (CRC) is not yet clear. In this study, we investigated the relationship between intermittent fasting and colorectal development in mice.
METHODS: First, APC [Min/+] mouse models (a spontaneous model of colorectal cancer) were subjected to intermittent fasting intervention (2 days/week) with regular monitoring of body weight changes. Subsequently, 16S rRNA sequencing and untargeted metabolomics were employed to analyze alterations in fecal microbial community structure and metabolic profiles following the fasting intervention. Tumor development was quantitatively assessed by enumerating CRC lesions using HE staining, while histopathological evaluation was performed to determine the degree of neoplastic progression. Concurrently, western blotting was conducted to examine the expression levels of intestinal barrier function-related proteins. Finally, in vitro validation experiments, including colony formation assay and transwell invasion assay, were performed to investigate the effects of the key microbial metabolite isovaleric acid on the proliferative and invasive capacities of CRC cells.
RESULTS: Intermittent fasting significantly reduced tumor incidence by approximately 50% compared to the control group (1.25 ± 0.38 vs 2.50 ± 0.38 tumors/mouse, P = 0.017) and markedly attenuated tumor progression. 16S rRNA sequencing analysis revealed significant enrichment of two key bacterial genera, Alistipes (P = 0.030) and Odoribacter (P = 0.030), along with a significant reduction in fecal isovaleric acid levels (P < 0.05) in the intermittent fasting group. Furthermore, intermittent fasting effectively controlled body weight gain (P < 0.05) and significantly improved intestinal barrier function (P < 0.05). In vitro experiments further demonstrated that isovaleric acid directly promoted CRC cell proliferation (P < 0.05) and enhanced their invasive capacity (P < 0.05).
CONCLUSION: Intermittent fasting suppresses CRC development in mice through its effects on gut microbiota and related metabolites.
Additional Links: PMID-40510674
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40510674,
year = {2025},
author = {Chen, J and Su, R and He, Y and Chen, J},
title = {Intermittent fasting inhibits the development of colorectal cancer in APC [Min/+] mice through gut microbiota and its related metabolites.},
journal = {Frontiers in microbiology},
volume = {16},
number = {},
pages = {1563224},
pmid = {40510674},
issn = {1664-302X},
abstract = {BACKGROUND AND OBJECTIVES: Intermittent fasting is an emerging dietary approach, but its specific role in colorectal cancer (CRC) is not yet clear. In this study, we investigated the relationship between intermittent fasting and colorectal development in mice.
METHODS: First, APC [Min/+] mouse models (a spontaneous model of colorectal cancer) were subjected to intermittent fasting intervention (2 days/week) with regular monitoring of body weight changes. Subsequently, 16S rRNA sequencing and untargeted metabolomics were employed to analyze alterations in fecal microbial community structure and metabolic profiles following the fasting intervention. Tumor development was quantitatively assessed by enumerating CRC lesions using HE staining, while histopathological evaluation was performed to determine the degree of neoplastic progression. Concurrently, western blotting was conducted to examine the expression levels of intestinal barrier function-related proteins. Finally, in vitro validation experiments, including colony formation assay and transwell invasion assay, were performed to investigate the effects of the key microbial metabolite isovaleric acid on the proliferative and invasive capacities of CRC cells.
RESULTS: Intermittent fasting significantly reduced tumor incidence by approximately 50% compared to the control group (1.25 ± 0.38 vs 2.50 ± 0.38 tumors/mouse, P = 0.017) and markedly attenuated tumor progression. 16S rRNA sequencing analysis revealed significant enrichment of two key bacterial genera, Alistipes (P = 0.030) and Odoribacter (P = 0.030), along with a significant reduction in fecal isovaleric acid levels (P < 0.05) in the intermittent fasting group. Furthermore, intermittent fasting effectively controlled body weight gain (P < 0.05) and significantly improved intestinal barrier function (P < 0.05). In vitro experiments further demonstrated that isovaleric acid directly promoted CRC cell proliferation (P < 0.05) and enhanced their invasive capacity (P < 0.05).
CONCLUSION: Intermittent fasting suppresses CRC development in mice through its effects on gut microbiota and related metabolites.},
}
RevDate: 2025-06-13
Plant developmental stage drives the assembly and functional adaptability of endophytic microbial communities.
Frontiers in microbiology, 16:1492141.
INTRODUCTION: The seeds of Amorphophallus muelleri represent a unique category of herbaceous seeds that arise from triploid apomixis. They necessitate an exceptionally protracted maturation phase of 8 months, followed by a dormancy period of 4 months, before they can germinate and give rise to fully formed new plants. Currently, the connection between endophytic microbial communities in A. muelleri seeds and the host plant's development is largely unexplored.
METHODS: Herein, we analyzed the temporal dynamics of the endophytic bacterial and fungal communities from seed germination to seedling establishment (seven stages) through amplicon sequencing.
RESULTS AND DISCUSSION: The results showed that plant developmental stage explained the large variation in endophytic bacterial and fungal communities in A. muelleri and that multiple microbial attributes (e.g., α, β-diversity, community composition, and bacterial and fungal ecological networks) are driven by the developmental state of A. muelleri. Metagenomic analyses further indicated that the four stages after rooting have higher microbial functional diversity. Microbial functional genes involved in cell wall/membrane/envelope biogenesis, inorganic ion transport and metabolism, and carbon degradation were abundant in A. muelleri seeds from Stage 1 to Stage 3 (before rooting). From Stage 4 to Stage 7 (after rooting), microbial functional genes involved in the carbon, nitrogen and phosphorus cycles, starch and sucrose metabolism, and energy production and conversion were more abundant. Coincidentally, more abundant Proteobacteria, and Basidiomycota taxa related to carbon degradation were found in stages 1-3, while more Allorhizobium-Neorhizobium-Pararhizobium-Rhizobium and Stenotrophomonas taxa associated with nitrogen cycling and plant growth promotion were observed in stages 4-7. These findings have greatly improved our basic understanding of the assembly and functional adaptability of the endophytic microbiome during A. muelleri plant development and are helpful for the mining, development and utilization of functional microbial resources.
Additional Links: PMID-40510672
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40510672,
year = {2025},
author = {Yang, M and Wang, J and Qi, Y and Gao, P and Li, L and Guo, J and Zhao, Y and Liu, J and Chen, Z and Zhao, J and Yu, L},
title = {Plant developmental stage drives the assembly and functional adaptability of endophytic microbial communities.},
journal = {Frontiers in microbiology},
volume = {16},
number = {},
pages = {1492141},
pmid = {40510672},
issn = {1664-302X},
abstract = {INTRODUCTION: The seeds of Amorphophallus muelleri represent a unique category of herbaceous seeds that arise from triploid apomixis. They necessitate an exceptionally protracted maturation phase of 8 months, followed by a dormancy period of 4 months, before they can germinate and give rise to fully formed new plants. Currently, the connection between endophytic microbial communities in A. muelleri seeds and the host plant's development is largely unexplored.
METHODS: Herein, we analyzed the temporal dynamics of the endophytic bacterial and fungal communities from seed germination to seedling establishment (seven stages) through amplicon sequencing.
RESULTS AND DISCUSSION: The results showed that plant developmental stage explained the large variation in endophytic bacterial and fungal communities in A. muelleri and that multiple microbial attributes (e.g., α, β-diversity, community composition, and bacterial and fungal ecological networks) are driven by the developmental state of A. muelleri. Metagenomic analyses further indicated that the four stages after rooting have higher microbial functional diversity. Microbial functional genes involved in cell wall/membrane/envelope biogenesis, inorganic ion transport and metabolism, and carbon degradation were abundant in A. muelleri seeds from Stage 1 to Stage 3 (before rooting). From Stage 4 to Stage 7 (after rooting), microbial functional genes involved in the carbon, nitrogen and phosphorus cycles, starch and sucrose metabolism, and energy production and conversion were more abundant. Coincidentally, more abundant Proteobacteria, and Basidiomycota taxa related to carbon degradation were found in stages 1-3, while more Allorhizobium-Neorhizobium-Pararhizobium-Rhizobium and Stenotrophomonas taxa associated with nitrogen cycling and plant growth promotion were observed in stages 4-7. These findings have greatly improved our basic understanding of the assembly and functional adaptability of the endophytic microbiome during A. muelleri plant development and are helpful for the mining, development and utilization of functional microbial resources.},
}
RevDate: 2025-06-13
CmpDate: 2025-06-13
Gut Microbiota in Lactose Intolerance: A Mendelian Randomization Study on Microbial Mechanisms and Potential Links to Tumor Inflammatory Microenvironments.
Mediators of inflammation, 2025:8181816.
Background: Previous observational studies have suggested an association between the composition of the intestinal microbiome and lactose intolerance (LI). However, the causal direction remains unclear. This study utilized Mendelian randomization (MR) to rigorously evaluate the potential causal link between the gut microbiome and LI. Methods: Genome-wide association studies (GWASs) summary statistics for gut microbiota and LI were sourced from previously published GWAS studies. Multiple methods, such as Simple mode, MR-Egger regression, weighted median, inverse variance-weighted (IVW), and weighted model, were used to determine the causal relationship between gut microbiota and LI. To validate the primary findings from the MR analyses, several sensitivity analyses were conducted. Furthermore, a reverse MR analysis was executed on bacterial taxa previously identified to have a potential causal link with LI risk, aiming to evaluate the possibility of reverse causation. Results: The IVW results revealed that the genus Lachnospiraceae UCG008 (OR = 0.584, 95%CI 0.356-0.958, p=0.0330), genus Eubacterium hallii group (OR = 0.467, 95% CI 0.242-0.899, p=0.023), and genus Ruminococcus gauvreauii group (OR = 0.506, 95% CI 0.2653-0.968, p=0.039) have a protective effect against LI. In contrast, the genus Holdemania (OR = 1.86, 95% CI 1.105-3.131, p=0.0194) displayed a predisposing effect. Sensitivity analyses did not detect any outlier single-nucleotide polymorphisms (SNPs). Further analyses reinforced the association between specific gut microbiota compositions and LI. No evidence suggested reverse causality between LI and the bacterial taxa identified in the reverse MR analysis. Conclusions: From a genetic standpoint, this MR study indicates a causal relationship between variations in gut microbiota composition and LI. This not only underscores the potential of gut microbiota-centric treatments for LI but also provides a foundation for exploring the role of gut microbiota in LI development. Further study of the mechanism of Lachnospiraceae in the treatment of IL is conducive to the discovery of new therapeutic targets for IL.
Additional Links: PMID-40510587
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40510587,
year = {2025},
author = {Xie, Y and Cao, Q and Huang, Z and Zou, X},
title = {Gut Microbiota in Lactose Intolerance: A Mendelian Randomization Study on Microbial Mechanisms and Potential Links to Tumor Inflammatory Microenvironments.},
journal = {Mediators of inflammation},
volume = {2025},
number = {},
pages = {8181816},
pmid = {40510587},
issn = {1466-1861},
mesh = {*Gastrointestinal Microbiome/genetics/physiology ; Humans ; *Mendelian Randomization Analysis/methods ; *Lactose Intolerance/microbiology/genetics ; Genome-Wide Association Study ; Polymorphism, Single Nucleotide/genetics ; *Inflammation/microbiology ; },
abstract = {Background: Previous observational studies have suggested an association between the composition of the intestinal microbiome and lactose intolerance (LI). However, the causal direction remains unclear. This study utilized Mendelian randomization (MR) to rigorously evaluate the potential causal link between the gut microbiome and LI. Methods: Genome-wide association studies (GWASs) summary statistics for gut microbiota and LI were sourced from previously published GWAS studies. Multiple methods, such as Simple mode, MR-Egger regression, weighted median, inverse variance-weighted (IVW), and weighted model, were used to determine the causal relationship between gut microbiota and LI. To validate the primary findings from the MR analyses, several sensitivity analyses were conducted. Furthermore, a reverse MR analysis was executed on bacterial taxa previously identified to have a potential causal link with LI risk, aiming to evaluate the possibility of reverse causation. Results: The IVW results revealed that the genus Lachnospiraceae UCG008 (OR = 0.584, 95%CI 0.356-0.958, p=0.0330), genus Eubacterium hallii group (OR = 0.467, 95% CI 0.242-0.899, p=0.023), and genus Ruminococcus gauvreauii group (OR = 0.506, 95% CI 0.2653-0.968, p=0.039) have a protective effect against LI. In contrast, the genus Holdemania (OR = 1.86, 95% CI 1.105-3.131, p=0.0194) displayed a predisposing effect. Sensitivity analyses did not detect any outlier single-nucleotide polymorphisms (SNPs). Further analyses reinforced the association between specific gut microbiota compositions and LI. No evidence suggested reverse causality between LI and the bacterial taxa identified in the reverse MR analysis. Conclusions: From a genetic standpoint, this MR study indicates a causal relationship between variations in gut microbiota composition and LI. This not only underscores the potential of gut microbiota-centric treatments for LI but also provides a foundation for exploring the role of gut microbiota in LI development. Further study of the mechanism of Lachnospiraceae in the treatment of IL is conducive to the discovery of new therapeutic targets for IL.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
*Gastrointestinal Microbiome/genetics/physiology
Humans
*Mendelian Randomization Analysis/methods
*Lactose Intolerance/microbiology/genetics
Genome-Wide Association Study
Polymorphism, Single Nucleotide/genetics
*Inflammation/microbiology
RevDate: 2025-06-13
The early-life gut microbiome in common pediatric diseases: roles and therapeutic implications.
Frontiers in nutrition, 12:1597206.
The early-life gut microbiome has been increasingly recognized as a contributing factor for pediatric health and diseases. Studies have reported that the human gut microbiota colonization commences at birth and progresses over the course of the first three years of life, until it reaches a mature and stable diversity and composition. During this critical window, the gut microbiome is vulnerably subjected to environmental factors, leading to transient microbial reprogramming and functional changes. The dynamic early-life intestinal microbiota is frequently manipulated by environmental factors, which impact the composition and function of the gut microflora, hence confer to short-and/or long-term health outcomes extending to adulthood. Evidence has shown that the imbalanced gut microbial community early in life is associated with several childhood diseases and disorders, such as inflammatory bowel diseases, allergies, attention-deficit/hyperactivity disorder and pediatric obesity. Manipulating the early-life intestinal microbes can either ameliorate or impair host's immunological and metabolic responses, impacting overall health conditions later in life. This narrative review article discusses the recent understanding and implications of the early-life gut microbiome in common pediatric diseases and potential intervention approaches.
Additional Links: PMID-40510495
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40510495,
year = {2025},
author = {Bankole, T and Li, Y},
title = {The early-life gut microbiome in common pediatric diseases: roles and therapeutic implications.},
journal = {Frontiers in nutrition},
volume = {12},
number = {},
pages = {1597206},
pmid = {40510495},
issn = {2296-861X},
abstract = {The early-life gut microbiome has been increasingly recognized as a contributing factor for pediatric health and diseases. Studies have reported that the human gut microbiota colonization commences at birth and progresses over the course of the first three years of life, until it reaches a mature and stable diversity and composition. During this critical window, the gut microbiome is vulnerably subjected to environmental factors, leading to transient microbial reprogramming and functional changes. The dynamic early-life intestinal microbiota is frequently manipulated by environmental factors, which impact the composition and function of the gut microflora, hence confer to short-and/or long-term health outcomes extending to adulthood. Evidence has shown that the imbalanced gut microbial community early in life is associated with several childhood diseases and disorders, such as inflammatory bowel diseases, allergies, attention-deficit/hyperactivity disorder and pediatric obesity. Manipulating the early-life intestinal microbes can either ameliorate or impair host's immunological and metabolic responses, impacting overall health conditions later in life. This narrative review article discusses the recent understanding and implications of the early-life gut microbiome in common pediatric diseases and potential intervention approaches.},
}
RevDate: 2025-06-13
Partially hydrolyzed guar gum ingestion suppresses atopic dermatitis-like symptoms through prebiotic effect in mice.
Journal of clinical biochemistry and nutrition, 76(3):280-288.
Growing knowledge reveals the association between the gut microbiome and skin, rendering the gut microbiome an appealing potential therapeutic target for atopic dermatitis (AD). In this study, we assessed the effect of partially hydrolyzed guar gum (PHGG) on AD-like symptoms induced by topical 1-Chloro-2,4-dinitrobenzene (DNCB) in BALB/c mice. Four weeks of PHGG feeding prevented the loss of epidermal barrier integrity and epithelial hyperplasia in the AD lesion (p<0.05, effect size >0.80), indicating a reduction in AD-like symptoms. According to the postulated mechanism, PHGG ingestion modulates the gut microbiome resulting in enhanced butyrate production (p<0.05). Butyrate suppresses Th2 function in gut immunity, which is believed to have significance in systemic immune regulation. The lowering of blood Th2 cytokines (IL-4 and IL-10, p<0.05) in the PHGG-fed group confirmed the existence of such a pathway, and butyrate can possibly be considered to have an indirect involvement in the suppression of Th2 immune response in the AD lesions. These findings encourage support for an association between gut microbiome and skin through the immune system, implying that daily PHGG ingestion may be beneficial for suppressing AD symptoms across the gut-immune-skin axis.
Additional Links: PMID-40510389
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40510389,
year = {2025},
author = {Morishima, S and Abe, A and Okamoto, S and Kapoor, MP and Matsuura, S and Kuriya, K and Ozeki, M and Nishio, M and Miura, H and Inoue, R},
title = {Partially hydrolyzed guar gum ingestion suppresses atopic dermatitis-like symptoms through prebiotic effect in mice.},
journal = {Journal of clinical biochemistry and nutrition},
volume = {76},
number = {3},
pages = {280-288},
pmid = {40510389},
issn = {0912-0009},
abstract = {Growing knowledge reveals the association between the gut microbiome and skin, rendering the gut microbiome an appealing potential therapeutic target for atopic dermatitis (AD). In this study, we assessed the effect of partially hydrolyzed guar gum (PHGG) on AD-like symptoms induced by topical 1-Chloro-2,4-dinitrobenzene (DNCB) in BALB/c mice. Four weeks of PHGG feeding prevented the loss of epidermal barrier integrity and epithelial hyperplasia in the AD lesion (p<0.05, effect size >0.80), indicating a reduction in AD-like symptoms. According to the postulated mechanism, PHGG ingestion modulates the gut microbiome resulting in enhanced butyrate production (p<0.05). Butyrate suppresses Th2 function in gut immunity, which is believed to have significance in systemic immune regulation. The lowering of blood Th2 cytokines (IL-4 and IL-10, p<0.05) in the PHGG-fed group confirmed the existence of such a pathway, and butyrate can possibly be considered to have an indirect involvement in the suppression of Th2 immune response in the AD lesions. These findings encourage support for an association between gut microbiome and skin through the immune system, implying that daily PHGG ingestion may be beneficial for suppressing AD symptoms across the gut-immune-skin axis.},
}
RevDate: 2025-06-13
Gut microbiome signatures predict 5-ASA efficacy in ulcerative colitis.
iScience, 28(6):112568.
Ulcerative colitis (UC) prevalence is rising globally, yet fewer than 50% of patients achieve mucosal healing (MH) with first-line 5-aminosalicylic acid (5-ASA) therapy. We aimed to identify microbial signatures that could predict the treatment efficacy of 5-ASA. Active UC patients on standardized 5-ASA treatment were prospectively enrolled. Shotgun metagenomic sequencing was performed to identify the taxonomic and functional profiles before and after treatment. Six species were enriched in the effective group and 3 species in the ineffective group at baseline. Faecalibacterium prausnitzii, Blautia massiliensis, and Phascolarctobacterium faecium were consistently depleted in the ineffective group at both time points. A random forest model based on these three species predicted ineffective 5-ASA treatment with area under the curve (AUC) = 0.80 (validation in the Inflammatory Bowel Disease Multi'omics Database [IBDMDB]: AUC = 0.82, specificity = 0.88, negative predictive value [NPV] = 0.70, and positive predictive value [PPV] = 0.80). Gut microbiome signatures have potential to serve as non-invasive predictors for ineffective 5-ASA treatment in UC.
Additional Links: PMID-40510115
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40510115,
year = {2025},
author = {Dang, Y and Xu, X and Ma, J and Zhou, M and Xu, C and Huang, X and Xu, F and Wang, Z and Shi, H and Zhang, S},
title = {Gut microbiome signatures predict 5-ASA efficacy in ulcerative colitis.},
journal = {iScience},
volume = {28},
number = {6},
pages = {112568},
pmid = {40510115},
issn = {2589-0042},
abstract = {Ulcerative colitis (UC) prevalence is rising globally, yet fewer than 50% of patients achieve mucosal healing (MH) with first-line 5-aminosalicylic acid (5-ASA) therapy. We aimed to identify microbial signatures that could predict the treatment efficacy of 5-ASA. Active UC patients on standardized 5-ASA treatment were prospectively enrolled. Shotgun metagenomic sequencing was performed to identify the taxonomic and functional profiles before and after treatment. Six species were enriched in the effective group and 3 species in the ineffective group at baseline. Faecalibacterium prausnitzii, Blautia massiliensis, and Phascolarctobacterium faecium were consistently depleted in the ineffective group at both time points. A random forest model based on these three species predicted ineffective 5-ASA treatment with area under the curve (AUC) = 0.80 (validation in the Inflammatory Bowel Disease Multi'omics Database [IBDMDB]: AUC = 0.82, specificity = 0.88, negative predictive value [NPV] = 0.70, and positive predictive value [PPV] = 0.80). Gut microbiome signatures have potential to serve as non-invasive predictors for ineffective 5-ASA treatment in UC.},
}
RevDate: 2025-06-13
CmpDate: 2025-06-13
[Changes of intestinal microflora in patients with colorectal benign and malignant tumors in high altitude area and comparison with the normal population in low altitude area].
Beijing da xue xue bao. Yi xue ban = Journal of Peking University. Health sciences, 57(3):578-583.
OBJECTIVE: To analyze the changes of intestinal flora in patients with benign and malignant colorectal tumors in high altitude areas and to compare them with the normal population in low altitude areas.
METHODS: The clinical data of 61 patients who underwent colonoscopy in the People' s Hospital of Tibet Autonomous Region from 2020 to 2022 were collected as the high altitude group. According to the colonoscopy results, they were divided into control group (29 cases), non-adenomatous polyp group (12 cases), adenoma group (10 cases), colorectal cancer group (10 cases). 17 patients who had negative colonoscopy results in the Peking University Third Hospital during the same period were collected as the low altitude control group. Before bowel preparation for colonoscopy, the fecal samples were collected. Then the DNA of bacteria in the fecal samples was extracted. The V3-V4 variable region of the 16S rRNA gene was PCR amplified and high-throughput sequenced. The species diversity of fecal flora was analyzed.
RESULTS: Alpha diversity analysis showed that the species diversity of samples from the high altitude colorectal cancer group differed statistically from that of the high altitude non-adenomatous polyp group and the low altitude control group, and the species diversity of colorectal samples from the high altitude colorectal cancer group was higher than that of the other two groups. While beta diversity showed no significant difference among the five groups. Differences were found in phylum level analysis that the abundance of Actinobacteriota in the low altitude control group was significantly lower than those in each group of the high altitude area, while the abundance of Actinobacteriota in the colorectal cancer group was significantly lower than those in the other 3 groups of the high altitude area. Differences were found in genus level analysis that the abundance of Bacteroides, Phascclarctobacterium and Lachnoclostridium in the low altitude control group was significantly higher than those of all the groups in the high altitude area; the abundance of Blautia and Collinsella in the high altitude control group was the highest. Lactobacillus was not detected in the low altitude control group, while there was a highly significant difference (P < 0.05) in the level of Lactobacillus in the four groups of high altitude area, and the abundance of Lactobacillus was significantly higher in the control group than those of the other three groups. In the four groups of samples at high altitude, the abundance of Bifidobacterium decreased significantly, while the abundance of Christensenellaceae_R-7_group increased significantly.
CONCLUSION: Compared with the high and low altitude controls, the diversity and abundance of intestinal flora in patients with colorectal benign and malignant tumors at high altitudes are different. And the abundance of species are also diffe-rent at the phylum and genus levels, suggesting that altitude factors may have some influence on intestinal flora.
Additional Links: PMID-40509837
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40509837,
year = {2025},
author = {Han, D and Ciren, Y and Li, Q and Li, J},
title = {[Changes of intestinal microflora in patients with colorectal benign and malignant tumors in high altitude area and comparison with the normal population in low altitude area].},
journal = {Beijing da xue xue bao. Yi xue ban = Journal of Peking University. Health sciences},
volume = {57},
number = {3},
pages = {578-583},
pmid = {40509837},
issn = {1671-167X},
mesh = {Humans ; *Altitude ; *Colorectal Neoplasms/microbiology ; *Gastrointestinal Microbiome ; Feces/microbiology ; Male ; Female ; Middle Aged ; Tibet ; RNA, Ribosomal, 16S/genetics ; Colonoscopy ; Adult ; Aged ; Adenoma/microbiology ; },
abstract = {OBJECTIVE: To analyze the changes of intestinal flora in patients with benign and malignant colorectal tumors in high altitude areas and to compare them with the normal population in low altitude areas.
METHODS: The clinical data of 61 patients who underwent colonoscopy in the People' s Hospital of Tibet Autonomous Region from 2020 to 2022 were collected as the high altitude group. According to the colonoscopy results, they were divided into control group (29 cases), non-adenomatous polyp group (12 cases), adenoma group (10 cases), colorectal cancer group (10 cases). 17 patients who had negative colonoscopy results in the Peking University Third Hospital during the same period were collected as the low altitude control group. Before bowel preparation for colonoscopy, the fecal samples were collected. Then the DNA of bacteria in the fecal samples was extracted. The V3-V4 variable region of the 16S rRNA gene was PCR amplified and high-throughput sequenced. The species diversity of fecal flora was analyzed.
RESULTS: Alpha diversity analysis showed that the species diversity of samples from the high altitude colorectal cancer group differed statistically from that of the high altitude non-adenomatous polyp group and the low altitude control group, and the species diversity of colorectal samples from the high altitude colorectal cancer group was higher than that of the other two groups. While beta diversity showed no significant difference among the five groups. Differences were found in phylum level analysis that the abundance of Actinobacteriota in the low altitude control group was significantly lower than those in each group of the high altitude area, while the abundance of Actinobacteriota in the colorectal cancer group was significantly lower than those in the other 3 groups of the high altitude area. Differences were found in genus level analysis that the abundance of Bacteroides, Phascclarctobacterium and Lachnoclostridium in the low altitude control group was significantly higher than those of all the groups in the high altitude area; the abundance of Blautia and Collinsella in the high altitude control group was the highest. Lactobacillus was not detected in the low altitude control group, while there was a highly significant difference (P < 0.05) in the level of Lactobacillus in the four groups of high altitude area, and the abundance of Lactobacillus was significantly higher in the control group than those of the other three groups. In the four groups of samples at high altitude, the abundance of Bifidobacterium decreased significantly, while the abundance of Christensenellaceae_R-7_group increased significantly.
CONCLUSION: Compared with the high and low altitude controls, the diversity and abundance of intestinal flora in patients with colorectal benign and malignant tumors at high altitudes are different. And the abundance of species are also diffe-rent at the phylum and genus levels, suggesting that altitude factors may have some influence on intestinal flora.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
*Altitude
*Colorectal Neoplasms/microbiology
*Gastrointestinal Microbiome
Feces/microbiology
Male
Female
Middle Aged
Tibet
RNA, Ribosomal, 16S/genetics
Colonoscopy
Adult
Aged
Adenoma/microbiology
RevDate: 2025-06-13
Skin microbiome analysis of a junctional epidermolysis bullosa patient treated with genetically modified stem cells.
Journal der Deutschen Dermatologischen Gesellschaft = Journal of the German Society of Dermatology : JDDG [Epub ahead of print].
BACKGROUND AND OBJECTIVE: Junctional epidermolysis bullosa (JEB) is a subtype of epidermolysis bullosa caused by mutations in the LAMB3 gene. We treated a patient with JEB using genetically corrected autologous epidermal cultures retrovirally transduced with the functional LAMB3 gene sequence. The objective of this study was to analyze the skin microbiome of this patient, with a particular focus on transgenic skin, and to compare the findings to the skin microbiome of healthy controls and patients with atopic dermatitis and well-documented microbial dysbiosis.
PATIENTS AND METHODS: Skin microbiome analysis was performed on a JEB patient 72 months after combined gene and stem cell therapy. Skin swabs from age-matched healthy controls and atopic dermatitis patients were included from the ProRaD study of CK-CARE.
RESULTS: The transgenic skin had comparably high relative and absolute Staphylococcus (S.) aureus abundance to blistering and non-blistering skin of the JEB patient, while the total bacterial load was lower. In blistering skin of the JEB patient, higher bacterial load was driven by S. aureus.
CONCLUSIONS: Our investigation confirms a unique microbiome composition in JEB, characterized by S. aureus driven bacterial overgrowth. The dysbiosis was not reversed in transgenic, non-blistering skin areas. However, the transgenic skin demonstrates stability in an environment of bacterial dysbiosis.
Additional Links: PMID-40509691
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40509691,
year = {2025},
author = {Dermietzel, A and Tosun, B and Nguyen, M and Wessel, K and Rauer, L and Neumann, AU and Hirsch, T and , and Traidl-Hoffmann, C and Reiger, M and Hülpüsch, C and Kueckelhaus, M},
title = {Skin microbiome analysis of a junctional epidermolysis bullosa patient treated with genetically modified stem cells.},
journal = {Journal der Deutschen Dermatologischen Gesellschaft = Journal of the German Society of Dermatology : JDDG},
volume = {},
number = {},
pages = {},
doi = {10.1111/ddg.15776},
pmid = {40509691},
issn = {1610-0387},
abstract = {BACKGROUND AND OBJECTIVE: Junctional epidermolysis bullosa (JEB) is a subtype of epidermolysis bullosa caused by mutations in the LAMB3 gene. We treated a patient with JEB using genetically corrected autologous epidermal cultures retrovirally transduced with the functional LAMB3 gene sequence. The objective of this study was to analyze the skin microbiome of this patient, with a particular focus on transgenic skin, and to compare the findings to the skin microbiome of healthy controls and patients with atopic dermatitis and well-documented microbial dysbiosis.
PATIENTS AND METHODS: Skin microbiome analysis was performed on a JEB patient 72 months after combined gene and stem cell therapy. Skin swabs from age-matched healthy controls and atopic dermatitis patients were included from the ProRaD study of CK-CARE.
RESULTS: The transgenic skin had comparably high relative and absolute Staphylococcus (S.) aureus abundance to blistering and non-blistering skin of the JEB patient, while the total bacterial load was lower. In blistering skin of the JEB patient, higher bacterial load was driven by S. aureus.
CONCLUSIONS: Our investigation confirms a unique microbiome composition in JEB, characterized by S. aureus driven bacterial overgrowth. The dysbiosis was not reversed in transgenic, non-blistering skin areas. However, the transgenic skin demonstrates stability in an environment of bacterial dysbiosis.},
}
RevDate: 2025-06-13
Protective Properties of the White Button Mushroom, Agaricus bisporus, in a Mouse Model of Colitis.
Molecular nutrition & food research [Epub ahead of print].
Previous work demonstrated the promising immunomodulatory potential of a naturally derived whole mushroom powder (WMP) from the white button mushroom, Agaricus bisporus. Here, we further investigate the protective properties of WMP in a mouse model of colitis. An in vitro digested WMP (IVD-WMP) reduced permeability of intestinal epithelial Caco-2 and HT-29-MTX cell monolayers to FITC dextran. In vivo, WMP orally administered to mice as a pretreatment before induction of dextran sulfate sodium (DSS)-induced colitis. Though statistically significant decreases in disease scores were not reported, we observed an antiinflammatory and antioxidative stress profile in the colon. Additionally, 16S ribosomal RNA (16S rRNA) microbiome analysis revealed differences in bacterial abundance associated with WMP pretreatment, including a decrease in Allobaculum species associated with inflammatory bowel disease. In a DSS-colitis recovery model, WMP promoted recovery as evidenced by improved weight gain, reduced stool scores, reduced IL-1β levels, and myeloperoxidase (MPO) activity in colonic tissue. This work demonstrates the health benefits associated with the consumption of the white button mushroom, including support of intestinal barrier integrity combined with antioxidant and antiinflammatory activity.
Additional Links: PMID-40509652
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40509652,
year = {2025},
author = {Dempsey, E and Walsh, AM and Yadav, S and Wilson, J and Sheedy, FJ and Corr, SC},
title = {Protective Properties of the White Button Mushroom, Agaricus bisporus, in a Mouse Model of Colitis.},
journal = {Molecular nutrition & food research},
volume = {},
number = {},
pages = {e70133},
doi = {10.1002/mnfr.70133},
pmid = {40509652},
issn = {1613-4133},
support = {EPSPD/2022/130//Irish Research Council Enterprise Partnership Scheme (Postdoctoral)/ ; 19/FFP/6499//Science Foundation Ireland Frontiers for the Future Programme/ ; },
abstract = {Previous work demonstrated the promising immunomodulatory potential of a naturally derived whole mushroom powder (WMP) from the white button mushroom, Agaricus bisporus. Here, we further investigate the protective properties of WMP in a mouse model of colitis. An in vitro digested WMP (IVD-WMP) reduced permeability of intestinal epithelial Caco-2 and HT-29-MTX cell monolayers to FITC dextran. In vivo, WMP orally administered to mice as a pretreatment before induction of dextran sulfate sodium (DSS)-induced colitis. Though statistically significant decreases in disease scores were not reported, we observed an antiinflammatory and antioxidative stress profile in the colon. Additionally, 16S ribosomal RNA (16S rRNA) microbiome analysis revealed differences in bacterial abundance associated with WMP pretreatment, including a decrease in Allobaculum species associated with inflammatory bowel disease. In a DSS-colitis recovery model, WMP promoted recovery as evidenced by improved weight gain, reduced stool scores, reduced IL-1β levels, and myeloperoxidase (MPO) activity in colonic tissue. This work demonstrates the health benefits associated with the consumption of the white button mushroom, including support of intestinal barrier integrity combined with antioxidant and antiinflammatory activity.},
}
RevDate: 2025-06-13
Nutrient Availability and Pathogen Clearance Impact Microbiome Composition in a Gnotobiotic Kimchi Model.
Foods (Basel, Switzerland), 14(11): pii:foods14111948.
Kimchi is a fermented Korean food typically made with napa cabbage, garlic, radish, ginger, and chili pepper. It is becoming increasingly popular due to its flavor, high fiber content, and purported probiotic benefits. The microbial ecology of the fermentation community has been extensively studied, though what's less understood is how its microbial community changes when nutrients or pathogens are introduced. To study this, we used gnotobiotic cabbage media inoculated with a kimchi starter culture as a model system. These inoculated samples were exposed to E. coli or Bacillus cereus, with or without added nutrients in the form of tryptic soy broth (TSB). We tracked pH, colony-forming units (CFUs), and community composition over time. We also used Oxford Nanopore sequencing to analyze the 16S rRNA gene (V4-V9), followed by use of the Emu algorithm for taxonomic assignments. As expected, LABs suppressed pathogens, but this effect was weaker early on in the nutrient-rich condition. Pathogen exposure changed the overall community-Lactobacillus species became more common, and Leuconostoc mesenteroides less so. Interestingly, adding nutrients alone caused similar microbial shifts to those seen with pathogen exposure. This could suggest that nutrient levels have a larger impact on the final microbiome structure than direct microbial competition. Together, these findings suggest that monitoring total microbial composition, and not just the presence of pathogens, may be important for ensuring kimchi fermentation reproducibility.
Additional Links: PMID-40509476
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40509476,
year = {2025},
author = {Bemis, DH and Camphausen, CE and Liu, E and Dantus, JJ and Navarro, JA and Dykstra, KL and Paltrowitz, LA and Dzhelmach, M and Joerg, M and Tamelessio, P and Belenky, P},
title = {Nutrient Availability and Pathogen Clearance Impact Microbiome Composition in a Gnotobiotic Kimchi Model.},
journal = {Foods (Basel, Switzerland)},
volume = {14},
number = {11},
pages = {},
doi = {10.3390/foods14111948},
pmid = {40509476},
issn = {2304-8158},
abstract = {Kimchi is a fermented Korean food typically made with napa cabbage, garlic, radish, ginger, and chili pepper. It is becoming increasingly popular due to its flavor, high fiber content, and purported probiotic benefits. The microbial ecology of the fermentation community has been extensively studied, though what's less understood is how its microbial community changes when nutrients or pathogens are introduced. To study this, we used gnotobiotic cabbage media inoculated with a kimchi starter culture as a model system. These inoculated samples were exposed to E. coli or Bacillus cereus, with or without added nutrients in the form of tryptic soy broth (TSB). We tracked pH, colony-forming units (CFUs), and community composition over time. We also used Oxford Nanopore sequencing to analyze the 16S rRNA gene (V4-V9), followed by use of the Emu algorithm for taxonomic assignments. As expected, LABs suppressed pathogens, but this effect was weaker early on in the nutrient-rich condition. Pathogen exposure changed the overall community-Lactobacillus species became more common, and Leuconostoc mesenteroides less so. Interestingly, adding nutrients alone caused similar microbial shifts to those seen with pathogen exposure. This could suggest that nutrient levels have a larger impact on the final microbiome structure than direct microbial competition. Together, these findings suggest that monitoring total microbial composition, and not just the presence of pathogens, may be important for ensuring kimchi fermentation reproducibility.},
}
RevDate: 2025-06-13
Fermented Dairy Products as Precision Modulators of Gut Microbiota and Host Health: Mechanistic Insights, Clinical Evidence, and Future Directions.
Foods (Basel, Switzerland), 14(11): pii:foods14111946.
Dairy products-encompassing yogurt, kefir, cheese, and cultured milk beverages-are emerging as versatile, food-based modulators of gut microbiota and host physiology. This review synthesizes mechanistic insights demonstrating how live starter cultures and their fermentation-derived metabolites (short-chain fatty acids, bioactive peptides, and exopolysaccharides) act synergistically to enhance microbial diversity, reinforce epithelial barrier integrity via upregulation of tight-junction proteins, and modulate immune signaling. Clinical evidence supports significant improvements in metabolic parameters (fasting glucose, lipid profiles, blood pressure) and reductions in systemic inflammation across metabolic syndrome, hypertension, and IBS cohorts. We highlight critical modulatory factors-including strain specificity, host enterotypes and FUT2 genotype, fermentation parameters, and matrix composition-that govern probiotic engraftment, postbiotic yield, and therapeutic efficacy. Despite promising short-term outcomes, current studies are limited by heterogeneous designs and brief intervention periods, underscoring the need for long-term, adaptive trials and integrative multi-omics to establish durability and causality. Looking forward, precision nutrition frameworks that harness baseline microbiota profiling, host genetics, and data-driven fermentation design will enable bespoke fermented dairy formulations, transforming these traditional foods into next-generation functional matrices for targeted prevention and management of metabolic, inflammatory, and neuroimmune disorders.
Additional Links: PMID-40509473
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40509473,
year = {2025},
author = {Gao, Y and Liu, Y and Ma, T and Liang, Q and Sun, J and Wu, X and Song, Y and Nie, H and Huang, J and Mu, G},
title = {Fermented Dairy Products as Precision Modulators of Gut Microbiota and Host Health: Mechanistic Insights, Clinical Evidence, and Future Directions.},
journal = {Foods (Basel, Switzerland)},
volume = {14},
number = {11},
pages = {},
doi = {10.3390/foods14111946},
pmid = {40509473},
issn = {2304-8158},
support = {32302032//National Natural Science Foundation of China/ ; 2022YFD2101503//National Key Research and Development Program/ ; 2020GXNSFBA297083//Natural Science Foundation of Guangxi, China/ ; },
abstract = {Dairy products-encompassing yogurt, kefir, cheese, and cultured milk beverages-are emerging as versatile, food-based modulators of gut microbiota and host physiology. This review synthesizes mechanistic insights demonstrating how live starter cultures and their fermentation-derived metabolites (short-chain fatty acids, bioactive peptides, and exopolysaccharides) act synergistically to enhance microbial diversity, reinforce epithelial barrier integrity via upregulation of tight-junction proteins, and modulate immune signaling. Clinical evidence supports significant improvements in metabolic parameters (fasting glucose, lipid profiles, blood pressure) and reductions in systemic inflammation across metabolic syndrome, hypertension, and IBS cohorts. We highlight critical modulatory factors-including strain specificity, host enterotypes and FUT2 genotype, fermentation parameters, and matrix composition-that govern probiotic engraftment, postbiotic yield, and therapeutic efficacy. Despite promising short-term outcomes, current studies are limited by heterogeneous designs and brief intervention periods, underscoring the need for long-term, adaptive trials and integrative multi-omics to establish durability and causality. Looking forward, precision nutrition frameworks that harness baseline microbiota profiling, host genetics, and data-driven fermentation design will enable bespoke fermented dairy formulations, transforming these traditional foods into next-generation functional matrices for targeted prevention and management of metabolic, inflammatory, and neuroimmune disorders.},
}
RevDate: 2025-06-13
Characteristics and Functions of Different Intestinal Segments in Juvenile Greater Amberjack (Seriola dumerili).
Animals : an open access journal from MDPI, 15(11): pii:ani15111672.
The greater amberjack (Seriola dumerili), a key species in marine aquaculture, relies heavily on its intestine for nutrient absorption and immune function. However, the structural and functional specialization of its intestinal segments remains poorly understood. In this study, we divided the intestine of S. dumerili into foregut, midgut, and hindgut, and conducted a multi-omics analysis integrating histological staining (H&E/AB-PAS), digestive enzyme assays, transcriptome sequencing, and 16S rRNA microbiota profiling to characterize structural, functional, molecular, and microbial differences across intestinal segments. Histological examinations revealed that brush border microvillus length, muscle layer thickness, and folding height were significantly greater in the foregut and hindgut compared to the midgut, while mucus and goblet cell density was higher in the foregut and midgut. Digestive enzyme assays showed that lipase activity peaked in the foregut, α-amylase in the midgut, and protease in the midgut and hindgut. Alkaline phosphatase (AKP) and acid phosphatase (ACP) activities were highest in the foregut and midgut. Immune-related enzyme activities (SOD (Superoxide dismutase), GSH-Px (Glutathione peroxidase), T-AOC (Total Antioxidant Capacity)) were elevated and MDA levels were lower in the midgut, indicating its role as the primary immune site. Transcriptome analysis identified segment-specific expression of nutrient transporters, such as slc6a19b (hindgut, protein), apoa1b (foregut, lipid), and slc37a4 (midgut, carbohydrate). Microbiome analysis revealed Ruminococcus dominance in the foregut (lipid digestion) and Prevotella, Bifidobacterium, and Lactobacillus enrichment in the midgut (carbohydrate metabolism and immunity). These findings highlight functional zonation in S. dumerili: the foregut specializes in lipid digestion, the midgut in carbohydrate metabolism and immunity, and the hindgut in protein digestion. This study provides foundational insights for optimizing aquaculture practices and advancing research in nutrition, immunology, and disease modeling in S. dumerili.
Additional Links: PMID-40509138
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40509138,
year = {2025},
author = {Zhu, K and Jiang, M and Yan, M and Huang, Y and Yang, T and Zhu, C},
title = {Characteristics and Functions of Different Intestinal Segments in Juvenile Greater Amberjack (Seriola dumerili).},
journal = {Animals : an open access journal from MDPI},
volume = {15},
number = {11},
pages = {},
doi = {10.3390/ani15111672},
pmid = {40509138},
issn = {2076-2615},
abstract = {The greater amberjack (Seriola dumerili), a key species in marine aquaculture, relies heavily on its intestine for nutrient absorption and immune function. However, the structural and functional specialization of its intestinal segments remains poorly understood. In this study, we divided the intestine of S. dumerili into foregut, midgut, and hindgut, and conducted a multi-omics analysis integrating histological staining (H&E/AB-PAS), digestive enzyme assays, transcriptome sequencing, and 16S rRNA microbiota profiling to characterize structural, functional, molecular, and microbial differences across intestinal segments. Histological examinations revealed that brush border microvillus length, muscle layer thickness, and folding height were significantly greater in the foregut and hindgut compared to the midgut, while mucus and goblet cell density was higher in the foregut and midgut. Digestive enzyme assays showed that lipase activity peaked in the foregut, α-amylase in the midgut, and protease in the midgut and hindgut. Alkaline phosphatase (AKP) and acid phosphatase (ACP) activities were highest in the foregut and midgut. Immune-related enzyme activities (SOD (Superoxide dismutase), GSH-Px (Glutathione peroxidase), T-AOC (Total Antioxidant Capacity)) were elevated and MDA levels were lower in the midgut, indicating its role as the primary immune site. Transcriptome analysis identified segment-specific expression of nutrient transporters, such as slc6a19b (hindgut, protein), apoa1b (foregut, lipid), and slc37a4 (midgut, carbohydrate). Microbiome analysis revealed Ruminococcus dominance in the foregut (lipid digestion) and Prevotella, Bifidobacterium, and Lactobacillus enrichment in the midgut (carbohydrate metabolism and immunity). These findings highlight functional zonation in S. dumerili: the foregut specializes in lipid digestion, the midgut in carbohydrate metabolism and immunity, and the hindgut in protein digestion. This study provides foundational insights for optimizing aquaculture practices and advancing research in nutrition, immunology, and disease modeling in S. dumerili.},
}
RevDate: 2025-06-13
A Preliminary Investigation of the Gastrointestinal Bacterial Microbiomes of Barred Owls (Strix varia) Admitted to a Wildlife Hospital.
Animals : an open access journal from MDPI, 15(11): pii:ani15111643.
Research on the gut microbiome, which includes microbial communities and genetic material in the gastrointestinal tract, has revealed essential roles beyond digestion, such as immune regulation, metabolism, and homeostasis. However, studies on birds-key ecosystem members-remain limited. Injured wild birds admitted to wildlife hospitals often receive antibiotics that can alter gut microbiota, leading to dysbiosis and promoting antimicrobial-resistant (AMR) bacteria. This study examined how hospitalization and antibiotics influence the cloacal microbiota of barred owls admitted for fracture repair. A total of 17 cloacal swab samples were analyzed using next-generation sequencing targeting 16S rRNA and AMR genes. Across all samples, Bacillota (Firmicutes), Actinomycetota (Actinobacteria), and Pseudomonadota (Proteobacteria) were the most abundant phyla. In non-antibiotic-treated owls, alpha and beta diversity showed no significant changes between admission and release; however, antibiotic-treated owls exhibited significant diversity shifts in these parameters at release. AMR genes were detected in most samples at admission, with some increasing significantly during hospitalization, suggesting an impact of antibiotic exposure. These findings provide insights into how antibiotics used in wildlife rehabilitation affect host microbiota and contribute to AMR gene dissemination.
Additional Links: PMID-40509108
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40509108,
year = {2025},
author = {Rhim, H and Aguilar, MG and Boykin, KL and Zapanta, K and Krumbeck, JA and Mitchell, MA},
title = {A Preliminary Investigation of the Gastrointestinal Bacterial Microbiomes of Barred Owls (Strix varia) Admitted to a Wildlife Hospital.},
journal = {Animals : an open access journal from MDPI},
volume = {15},
number = {11},
pages = {},
doi = {10.3390/ani15111643},
pmid = {40509108},
issn = {2076-2615},
support = {PG007872//Louisiana State University/ ; Foundation//Fluker Farms/ ; },
abstract = {Research on the gut microbiome, which includes microbial communities and genetic material in the gastrointestinal tract, has revealed essential roles beyond digestion, such as immune regulation, metabolism, and homeostasis. However, studies on birds-key ecosystem members-remain limited. Injured wild birds admitted to wildlife hospitals often receive antibiotics that can alter gut microbiota, leading to dysbiosis and promoting antimicrobial-resistant (AMR) bacteria. This study examined how hospitalization and antibiotics influence the cloacal microbiota of barred owls admitted for fracture repair. A total of 17 cloacal swab samples were analyzed using next-generation sequencing targeting 16S rRNA and AMR genes. Across all samples, Bacillota (Firmicutes), Actinomycetota (Actinobacteria), and Pseudomonadota (Proteobacteria) were the most abundant phyla. In non-antibiotic-treated owls, alpha and beta diversity showed no significant changes between admission and release; however, antibiotic-treated owls exhibited significant diversity shifts in these parameters at release. AMR genes were detected in most samples at admission, with some increasing significantly during hospitalization, suggesting an impact of antibiotic exposure. These findings provide insights into how antibiotics used in wildlife rehabilitation affect host microbiota and contribute to AMR gene dissemination.},
}
RevDate: 2025-06-13
Postbiotic Lactiplantibacillus plantarum CECT 9161 Influences the Canine Oral Metagenome and Reduces Plaque Biofilm Formation.
Animals : an open access journal from MDPI, 15(11): pii:ani15111615.
Periodontal diseases are highly prevalent in dogs and intricately interconnected with the composition and functional attributes of the oral microbiota. The demand for non-invasive interventions to support oral health presents an opportunity for functional ingredients. The novel postbiotic heat-treated (HT) Lactiplantibacillus plantarum CECT 9161 inhibited growth and biofilm formation of oral microorganisms in vitro. The in vitro growth of saliva-derived biofilms was also inhibited and revealed microbiome modulation. Two doses of the postbiotic (LOW: 5 mg dog/day, HIGH: 25 mg/dog/day) were assessed in a placebo-controlled, double-blinded, 57-day clinical trial involving 60 dogs. Associations were found between the postbiotic, reduced plaque formation, and modulation of the oral microbiome, including increased abundance of genes involved in denitrification, heme and catechol biosynthesis, and oxidative stress reduction. The results suggest that HT Lactiplantibacillus plantarum CECT 9161 may support oral health in dogs by modifying the microbiome of supragingival plaque and reducing plaque formation.
Additional Links: PMID-40509083
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40509083,
year = {2025},
author = {Florit-Ruiz, A and Rago, L and Rojas, A and Guzelkhanova, B and Pont-Beltran, A and Lamelas, A and Solaz-Fuster, MC and Martinez-Blanch, JF and López, ME and García-Lainez, G and Rosier, BT and Day, R and Rubio, T and Batchelor, R and Nixon, SL},
title = {Postbiotic Lactiplantibacillus plantarum CECT 9161 Influences the Canine Oral Metagenome and Reduces Plaque Biofilm Formation.},
journal = {Animals : an open access journal from MDPI},
volume = {15},
number = {11},
pages = {},
doi = {10.3390/ani15111615},
pmid = {40509083},
issn = {2076-2615},
support = {N/A//Archer Daniels Midland (United States)/ ; },
abstract = {Periodontal diseases are highly prevalent in dogs and intricately interconnected with the composition and functional attributes of the oral microbiota. The demand for non-invasive interventions to support oral health presents an opportunity for functional ingredients. The novel postbiotic heat-treated (HT) Lactiplantibacillus plantarum CECT 9161 inhibited growth and biofilm formation of oral microorganisms in vitro. The in vitro growth of saliva-derived biofilms was also inhibited and revealed microbiome modulation. Two doses of the postbiotic (LOW: 5 mg dog/day, HIGH: 25 mg/dog/day) were assessed in a placebo-controlled, double-blinded, 57-day clinical trial involving 60 dogs. Associations were found between the postbiotic, reduced plaque formation, and modulation of the oral microbiome, including increased abundance of genes involved in denitrification, heme and catechol biosynthesis, and oxidative stress reduction. The results suggest that HT Lactiplantibacillus plantarum CECT 9161 may support oral health in dogs by modifying the microbiome of supragingival plaque and reducing plaque formation.},
}
RevDate: 2025-06-13
Unlocking the Potential of Paper Mulberry Powder in Cherry Valley Ducks: Impacts on Growth, Serum Biochemistry, and Cecum Microbiome.
Animals : an open access journal from MDPI, 15(11): pii:ani15111602.
This study investigates the effects of incorporating paper mulberry (Broussonetia papyrifera L.) powder into the diets of Cherry Valley ducks on growth performance, serum biochemistry, and the gut microbiome. A total of 350 14-day-old male Cherry Valley ducks were randomly assigned to five groups receiving diets with 0%, 4%, 6%, 8%, and 10% paper mulberry powder for 42 days. Growth performance, meat quality, serum immunity, and cecal microbial composition were assessed. The results showed no significant differences in average daily feed intake and feed conversion ratio among treatments, with the 6% paper mulberry group showing the highest average daily gain (79.73 g) (p < 0.05). Meat quality parameters, including color, drip loss, cooking loss, and shear force, were not significantly affected by paper mulberry powder supplementation, while the 8% paper mulberry group showed the highest pH24 value (5.47) (p < 0.05). Serum biochemistry revealed increased total protein (G0, G4, G6, G8, and G10: 41.50, 44.47, 45.58, 45.67, and 45.85 g/L, respectively), albumin (G0, G4, G6, G8, and G10: 18.61, 19.56, 20.29, 20.2, and 20.39 g/L, respectively), total cholesterol (G0, G4, G6, G8, and G10: 5.31, 4.96, 5.37, 5.53, and 5.59 mmol/L, respectively), and high-density lipoprotein cholesterol (HDL) in ducks fed 6%, 8%, and 10% paper mulberry powder, with lower alanine aminotransferase (ALT), aspartate aminotransferase (AST), and total bilirubin (TBI) in the 8% and 10% groups (p < 0.05). Cecal microbial diversity was enhanced with paper mulberry powder, particularly in the 6% group, which showed increased Bacteroides abundance (p < 0.05). Supplementing duck diets with 6% paper mulberry powder increased average daily gain, without adversely affecting meat quality and health, suggesting its potential as a sustainable feed ingredient in the duck meat industry.
Additional Links: PMID-40509068
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40509068,
year = {2025},
author = {Xiong, Y and Tang, C and Wang, X and Wang, Y and Yang, F},
title = {Unlocking the Potential of Paper Mulberry Powder in Cherry Valley Ducks: Impacts on Growth, Serum Biochemistry, and Cecum Microbiome.},
journal = {Animals : an open access journal from MDPI},
volume = {15},
number = {11},
pages = {},
doi = {10.3390/ani15111602},
pmid = {40509068},
issn = {2076-2615},
support = {32201473//National Natural Science Foundation of China/ ; },
abstract = {This study investigates the effects of incorporating paper mulberry (Broussonetia papyrifera L.) powder into the diets of Cherry Valley ducks on growth performance, serum biochemistry, and the gut microbiome. A total of 350 14-day-old male Cherry Valley ducks were randomly assigned to five groups receiving diets with 0%, 4%, 6%, 8%, and 10% paper mulberry powder for 42 days. Growth performance, meat quality, serum immunity, and cecal microbial composition were assessed. The results showed no significant differences in average daily feed intake and feed conversion ratio among treatments, with the 6% paper mulberry group showing the highest average daily gain (79.73 g) (p < 0.05). Meat quality parameters, including color, drip loss, cooking loss, and shear force, were not significantly affected by paper mulberry powder supplementation, while the 8% paper mulberry group showed the highest pH24 value (5.47) (p < 0.05). Serum biochemistry revealed increased total protein (G0, G4, G6, G8, and G10: 41.50, 44.47, 45.58, 45.67, and 45.85 g/L, respectively), albumin (G0, G4, G6, G8, and G10: 18.61, 19.56, 20.29, 20.2, and 20.39 g/L, respectively), total cholesterol (G0, G4, G6, G8, and G10: 5.31, 4.96, 5.37, 5.53, and 5.59 mmol/L, respectively), and high-density lipoprotein cholesterol (HDL) in ducks fed 6%, 8%, and 10% paper mulberry powder, with lower alanine aminotransferase (ALT), aspartate aminotransferase (AST), and total bilirubin (TBI) in the 8% and 10% groups (p < 0.05). Cecal microbial diversity was enhanced with paper mulberry powder, particularly in the 6% group, which showed increased Bacteroides abundance (p < 0.05). Supplementing duck diets with 6% paper mulberry powder increased average daily gain, without adversely affecting meat quality and health, suggesting its potential as a sustainable feed ingredient in the duck meat industry.},
}
RevDate: 2025-06-13
Characterisation of the Gastrointestinal Microbiome of Green Sea Turtles (Chelonia mydas): A Systematic Review.
Animals : an open access journal from MDPI, 15(11): pii:ani15111594.
The gut microbiome of sea turtles is essential for their ecological resilience and adaptation to environmental stressors. We hypothesised that different gut microbial profiles existed between green sea turtles kept in captivity and those in the wild. The aim of this systematic review was to determine dominant bacterial phyla in the gut microbiomes of wild and captive green sea turtles. Comparison of the top four bacterial phyla revealed that Bacillota was the most abundant phylum in captive turtles (40.9-87.5%), but it only ranked second (3.5-57.8%) in wild turtles. Bacteroidota had comparable relative abundance in captive (8.7-45.6%) and wild (3.6-43.1%) populations. By contrast, the relative abundance of Pseudomonadota was higher in wild turtles (6.2-68.1%) compared to the captive population (0.1-6.6%). Verrucomicrobiota was less prevalent in wild and captive populations, with relative abundances ranging from 0.28 to 5.4% and 2.3 to 7.2%, respectively. These findings highlight a putative gut microbial shift between wild and captive green sea turtle populations. This shift may be shaped by variations in environmental factors in captivity or the wild. Nonetheless, the significance of these putative changes is still unknown; the potential to use microbial shifts to guide management, rehabilitation, and conservation of green sea turtles is promising, but remains limited.
Additional Links: PMID-40509060
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40509060,
year = {2025},
author = {Ghafoor, D and Hayakijkosol, O and Ewels, C and Kinobe, R},
title = {Characterisation of the Gastrointestinal Microbiome of Green Sea Turtles (Chelonia mydas): A Systematic Review.},
journal = {Animals : an open access journal from MDPI},
volume = {15},
number = {11},
pages = {},
doi = {10.3390/ani15111594},
pmid = {40509060},
issn = {2076-2615},
abstract = {The gut microbiome of sea turtles is essential for their ecological resilience and adaptation to environmental stressors. We hypothesised that different gut microbial profiles existed between green sea turtles kept in captivity and those in the wild. The aim of this systematic review was to determine dominant bacterial phyla in the gut microbiomes of wild and captive green sea turtles. Comparison of the top four bacterial phyla revealed that Bacillota was the most abundant phylum in captive turtles (40.9-87.5%), but it only ranked second (3.5-57.8%) in wild turtles. Bacteroidota had comparable relative abundance in captive (8.7-45.6%) and wild (3.6-43.1%) populations. By contrast, the relative abundance of Pseudomonadota was higher in wild turtles (6.2-68.1%) compared to the captive population (0.1-6.6%). Verrucomicrobiota was less prevalent in wild and captive populations, with relative abundances ranging from 0.28 to 5.4% and 2.3 to 7.2%, respectively. These findings highlight a putative gut microbial shift between wild and captive green sea turtle populations. This shift may be shaped by variations in environmental factors in captivity or the wild. Nonetheless, the significance of these putative changes is still unknown; the potential to use microbial shifts to guide management, rehabilitation, and conservation of green sea turtles is promising, but remains limited.},
}
RevDate: 2025-06-13
Acute Effect of Short-Term Benzocaine Anesthesia on the Skin Mucus Microbiome of Atlantic salmon (Salmo salar).
Animals : an open access journal from MDPI, 15(11): pii:ani15111566.
Routine aquaculture practices such as capture, transportation, and handling can disrupt the relationship between commensal and opportunistic bacteria in the fish skin microbiome. Anesthetic baths are a common welfare practice in aquaculture to reduce stress during handling. However, to date, no studies assessed the effect of anesthetics on bacterial communities in fish skin mucus. This study is the first to evaluate the influence of benzocaine, a widely used anesthetic, on the skin mucus bacterial microbiome of Atlantic salmon reared in a recirculating aquaculture system (RAS). Using Illumina high-throughput 16S rRNA gene sequencing, we found that bacterial richness and diversity were significantly reduced in skin mucus samples from fish with anesthesia (ANE) when compared with those without anesthesia (CTR). The predominant bacterial classes in both groups were Gammaproteobacteria (54.1-62.6%) and Betaproteobacteria (22.6-22.9%). However, significant dissimilarities in beta diversity were observed between the bacterial community structure of salmon skin mucus samples from ANE and CTR. These findings demonstrate that benzocaine exposure alters skin mucus microbiome of Atlantic salmon potentially leading to dysbiosis. This study also provides baseline information on the bacterial communities of Atlantic salmon skin mucus microbiome in an RAS. As no temporal resampling was performed, the duration and persistence of these changes remain unknown and warrant further investigation.
Additional Links: PMID-40509032
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40509032,
year = {2025},
author = {Martins, P and Pimentel, T and Ribeiro, N and Calado, R},
title = {Acute Effect of Short-Term Benzocaine Anesthesia on the Skin Mucus Microbiome of Atlantic salmon (Salmo salar).},
journal = {Animals : an open access journal from MDPI},
volume = {15},
number = {11},
pages = {},
doi = {10.3390/ani15111566},
pmid = {40509032},
issn = {2076-2615},
support = {UIDP/50017/2020+UIDB/50017/2020+LA/P/0094/2020//Centro de Estudos Ambientais e Marinhos/ ; },
abstract = {Routine aquaculture practices such as capture, transportation, and handling can disrupt the relationship between commensal and opportunistic bacteria in the fish skin microbiome. Anesthetic baths are a common welfare practice in aquaculture to reduce stress during handling. However, to date, no studies assessed the effect of anesthetics on bacterial communities in fish skin mucus. This study is the first to evaluate the influence of benzocaine, a widely used anesthetic, on the skin mucus bacterial microbiome of Atlantic salmon reared in a recirculating aquaculture system (RAS). Using Illumina high-throughput 16S rRNA gene sequencing, we found that bacterial richness and diversity were significantly reduced in skin mucus samples from fish with anesthesia (ANE) when compared with those without anesthesia (CTR). The predominant bacterial classes in both groups were Gammaproteobacteria (54.1-62.6%) and Betaproteobacteria (22.6-22.9%). However, significant dissimilarities in beta diversity were observed between the bacterial community structure of salmon skin mucus samples from ANE and CTR. These findings demonstrate that benzocaine exposure alters skin mucus microbiome of Atlantic salmon potentially leading to dysbiosis. This study also provides baseline information on the bacterial communities of Atlantic salmon skin mucus microbiome in an RAS. As no temporal resampling was performed, the duration and persistence of these changes remain unknown and warrant further investigation.},
}
RevDate: 2025-06-13
Effects of Grazing in a Low Deciduous Forest on Rumen Microbiota and Volatile Fatty Acid Production in Lambs.
Animals : an open access journal from MDPI, 15(11): pii:ani15111565.
The aim of the present study was to evaluate the effect of grazing the low deciduous forest (LDF) vegetation on the diversity of the rumen microbiome in growing lambs and its relationship with volatile fatty acid (VFA) profiles. After a 35-day indoor acclimatization (stabilization period), the lambs were assigned to two groups: housed (CG, n = 4) and grazing (EG, n = 4). The grazing lambs had a 14-day habituation period in the LDF (4 h/day) and a further 30 grazing days when fodder intake was observed. Ruminal samples were collected at the end of the stabilization, on day 14 post-stabilization (14DPS), and on day 44 post-stabilization (44DPS). The ruminal butyrate concentration showed a progressive decrease of approximately 23% over the time (p = 0.0130). The qualitative composition (p = 0.001) and relative proportions of bacteria (p = 0.004) in EG-44DPS exhibited a greater diversity, with 107 total genera and 19 unique, significant abundances in 13 genera with a higher presence of Bacteroidales_RF16_group, Lachnospiraceae_ND3007_group, and WCHB1-41. Moreover, significant functional profiles are associated with key metabolic pathways in bacteria and are interconnected by the need to generate energy and biosynthetic precursors and to manage available nitrogen and carbon. Finally, eight bacterial genera were identified as biomarkers correlated with the increase in VFA in EG-44DPS.
Additional Links: PMID-40509031
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40509031,
year = {2025},
author = {Ávila-Cervantes, R and González-Pech, P and Sandoval-Castro, C and Torres-Acosta, F and Ramos-Zapata, J and Galicia-Jiménez, M and Pacheco-Arjona, R},
title = {Effects of Grazing in a Low Deciduous Forest on Rumen Microbiota and Volatile Fatty Acid Production in Lambs.},
journal = {Animals : an open access journal from MDPI},
volume = {15},
number = {11},
pages = {},
doi = {10.3390/ani15111565},
pmid = {40509031},
issn = {2076-2615},
abstract = {The aim of the present study was to evaluate the effect of grazing the low deciduous forest (LDF) vegetation on the diversity of the rumen microbiome in growing lambs and its relationship with volatile fatty acid (VFA) profiles. After a 35-day indoor acclimatization (stabilization period), the lambs were assigned to two groups: housed (CG, n = 4) and grazing (EG, n = 4). The grazing lambs had a 14-day habituation period in the LDF (4 h/day) and a further 30 grazing days when fodder intake was observed. Ruminal samples were collected at the end of the stabilization, on day 14 post-stabilization (14DPS), and on day 44 post-stabilization (44DPS). The ruminal butyrate concentration showed a progressive decrease of approximately 23% over the time (p = 0.0130). The qualitative composition (p = 0.001) and relative proportions of bacteria (p = 0.004) in EG-44DPS exhibited a greater diversity, with 107 total genera and 19 unique, significant abundances in 13 genera with a higher presence of Bacteroidales_RF16_group, Lachnospiraceae_ND3007_group, and WCHB1-41. Moreover, significant functional profiles are associated with key metabolic pathways in bacteria and are interconnected by the need to generate energy and biosynthetic precursors and to manage available nitrogen and carbon. Finally, eight bacterial genera were identified as biomarkers correlated with the increase in VFA in EG-44DPS.},
}
RevDate: 2025-06-13
Soil-Gradient-Derived Bacterial Synthetic Communities Enhance Drought Tolerance in Quercus pubescens and Sorbus domestica Seedlings.
Plants (Basel, Switzerland), 14(11): pii:plants14111659.
Climate-change-induced drought threatens forest restoration by limiting seedling establishment. To address this, we developed synthetic bacterial communities (SynComs) tailored to support drought tolerance in two Mediterranean tree species, Quercus pubescens and Sorbus domestica. Bacteria were isolated from forest soil exposed to long-term drought, sampling across soil depths and root-associated compartments. We selected strains with key plant-beneficial traits, including exopolysaccharide (EPS) production, hormone synthesis (auxin, ABA), siderophore release, and osmotic tolerance. SynComs were assembled based on functional complementarity and ecological origin. Biofilm assays showed that even weak individual producers could enhance community-level performance. After initial screening on Arabidopsis thaliana, the most and least effective SynComs were tested on Q. pubescens and S. domestica seedlings. Compared to controls, the best-performing SynComs reduced the proportion of drought-symptomatic seedlings by 47% in Q. pubescens and 71% in S. domestica, outperforming single-strain inoculants. Notably, EPS-rich SynCom B aligned with the conservative root traits of Q. pubescens, while hormone-rich SynCom F matched the acquisitive strategy of S. domestica. Predictive modeling identified bacterial identity and symptom timing as key predictors of drought resilience. Our results highlight the value of matching microbial traits with plant strategies and drought context for climate-smart forest restoration.
Additional Links: PMID-40508333
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40508333,
year = {2025},
author = {Aleksieienko, I and Fernandes Hertel, M and Reilhan, J and de Castro, M and Légeret, B and Caixeta Oliveira, H and Reiter, IM and Santaella, C},
title = {Soil-Gradient-Derived Bacterial Synthetic Communities Enhance Drought Tolerance in Quercus pubescens and Sorbus domestica Seedlings.},
journal = {Plants (Basel, Switzerland)},
volume = {14},
number = {11},
pages = {},
doi = {10.3390/plants14111659},
pmid = {40508333},
issn = {2223-7747},
support = {101094587//HORIZON EUROPE INFRA-2022-TECH project 'PHENET'/ ; 88887.712065/2022-00//Coordenação de Aperfeicoamento de Pessoal de Nível Superior/ ; ANR-20-EBI5-0008-07//Agence Nationale de la Recherche/ ; ANR-20-EBI5-0008-07//2019-2020 BiodivERsA+ joint call, BiodivClim ERA-Net COFUND, Fundação Araucária/Secretaria de Estado da Ciência, Tecnologia e Ensino Superior do Paraná (NAPI Biodiversidade), FAPESP (BR), ANR (FR), Federal Ministry of Education and Research (DE)/ ; Sv 946/10 - 41880PL//CAPES-COFECUB/ ; },
abstract = {Climate-change-induced drought threatens forest restoration by limiting seedling establishment. To address this, we developed synthetic bacterial communities (SynComs) tailored to support drought tolerance in two Mediterranean tree species, Quercus pubescens and Sorbus domestica. Bacteria were isolated from forest soil exposed to long-term drought, sampling across soil depths and root-associated compartments. We selected strains with key plant-beneficial traits, including exopolysaccharide (EPS) production, hormone synthesis (auxin, ABA), siderophore release, and osmotic tolerance. SynComs were assembled based on functional complementarity and ecological origin. Biofilm assays showed that even weak individual producers could enhance community-level performance. After initial screening on Arabidopsis thaliana, the most and least effective SynComs were tested on Q. pubescens and S. domestica seedlings. Compared to controls, the best-performing SynComs reduced the proportion of drought-symptomatic seedlings by 47% in Q. pubescens and 71% in S. domestica, outperforming single-strain inoculants. Notably, EPS-rich SynCom B aligned with the conservative root traits of Q. pubescens, while hormone-rich SynCom F matched the acquisitive strategy of S. domestica. Predictive modeling identified bacterial identity and symptom timing as key predictors of drought resilience. Our results highlight the value of matching microbial traits with plant strategies and drought context for climate-smart forest restoration.},
}
RevDate: 2025-06-13
Application of a Synthetic Microbial Community to Enhance Pepper Resistance Against Phytophthora capsici.
Plants (Basel, Switzerland), 14(11): pii:plants14111625.
Pepper (Capsicum annuum) production faces significant challenges from soil-borne pathogens, particularly Phytophthora capsici, which induces root rot and damping-off diseases. Management of this pathogen remains challenging owing to the scarcity of resistant cultivars and the ineffectiveness of chemical control methods. A single strain has been used to prevent pathogenic disease, and this approach limits the exploration of consortia comprising different genera. In this study, we isolated five bacterial strains (Bacillus sp. T3, Flavobacterium anhuiense T4, Cytobacillus firmus T8, Streptomyces roseicoloratus T14, and Pseudomonas frederiksbergensis A6) from the rhizosphere of healthy pepper plants. We then applied this 5-isolate synthetic microbial community (SynCom) to Capsicum annuum to evaluate its efficacy in improving pepper resilience against P. capsici. The SynCom members exhibited phosphate solubilization, indole-3-acetic acid production, catalase activity, siderophore synthesis, and strong antagonism against P. capsici. The SynCom reduced disease severity and enhanced the growth of pepper plants. Furthermore, the beneficial genera such as Bacillus, Fusicolla, and Trichoderma, significantly increased in the rhizosphere of pepper after the application of the SynCom. Microbial functional prediction analysis revealed that these microbial shifts were associated with nitrogen cycling and pathogen suppression. Our SynCom approach demonstrates the effectiveness of microbial consortia in promoting the growth of pathogen-infected plants by reprogramming the microbial community in the rhizosphere.
Additional Links: PMID-40508300
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40508300,
year = {2025},
author = {Bashizi, TF and Kim, MJ and Lim, K and Lee, G and Tagele, SB and Shin, JH},
title = {Application of a Synthetic Microbial Community to Enhance Pepper Resistance Against Phytophthora capsici.},
journal = {Plants (Basel, Switzerland)},
volume = {14},
number = {11},
pages = {},
doi = {10.3390/plants14111625},
pmid = {40508300},
issn = {2223-7747},
support = {2021R1A6C101A416//Korea Basic Science Institute/ ; },
abstract = {Pepper (Capsicum annuum) production faces significant challenges from soil-borne pathogens, particularly Phytophthora capsici, which induces root rot and damping-off diseases. Management of this pathogen remains challenging owing to the scarcity of resistant cultivars and the ineffectiveness of chemical control methods. A single strain has been used to prevent pathogenic disease, and this approach limits the exploration of consortia comprising different genera. In this study, we isolated five bacterial strains (Bacillus sp. T3, Flavobacterium anhuiense T4, Cytobacillus firmus T8, Streptomyces roseicoloratus T14, and Pseudomonas frederiksbergensis A6) from the rhizosphere of healthy pepper plants. We then applied this 5-isolate synthetic microbial community (SynCom) to Capsicum annuum to evaluate its efficacy in improving pepper resilience against P. capsici. The SynCom members exhibited phosphate solubilization, indole-3-acetic acid production, catalase activity, siderophore synthesis, and strong antagonism against P. capsici. The SynCom reduced disease severity and enhanced the growth of pepper plants. Furthermore, the beneficial genera such as Bacillus, Fusicolla, and Trichoderma, significantly increased in the rhizosphere of pepper after the application of the SynCom. Microbial functional prediction analysis revealed that these microbial shifts were associated with nitrogen cycling and pathogen suppression. Our SynCom approach demonstrates the effectiveness of microbial consortia in promoting the growth of pathogen-infected plants by reprogramming the microbial community in the rhizosphere.},
}
RevDate: 2025-06-13
Functional Roles of the Seagrass (Zostera marina) Holobiont Change with Plant Development.
Plants (Basel, Switzerland), 14(11): pii:plants14111584.
Seagrass meadows play a critical role in biogeochemical cycling, especially in nitrogen and sulphur processes, driven by their associated microbiome. This study provides a novel functional analysis of microbial communities in seagrass (Zostera marina) rhizosphere and endosphere, comparing seedlings and mature plants. While nitrogen-fixing bacteria are more abundant in seedlings, mature plants exhibit greater microbial diversity and stability. Sediment samples show higher microbial diversity than roots, suggesting distinct niche environments in seagrass roots. Key microbial taxa (sulphur-oxidizing and nitrogen-cycling bacteria) were observed across developmental stages, with rapid establishment in seedlings aiding survival in sulphide-rich, anoxic sediments. Chromatiales, which oxidize sulphur, are hypothesized to support juvenile plant growth by mitigating sulphide toxicity, a key stressor in early development. Additionally, sulfate-reducing bacteria (SRB), though potentially harmful due to H2S production, may also aid in nitrogen fixation by producing ammonium. The study underscores the dynamic relationship between seagrass and its microbiome, especially the differences in microbial community structure and function between juvenile and mature plants. The study emphasizes the need for a deeper understanding of microbial roles within the seagrass holobiont to aid with Blue Carbon stores and to improve restoration success, particularly for juvenile plants struggling to establish effective microbiomes.
Additional Links: PMID-40508259
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40508259,
year = {2025},
author = {Gorvel, S and Walter, B and Taylor, JD and Unsworth, RKF},
title = {Functional Roles of the Seagrass (Zostera marina) Holobiont Change with Plant Development.},
journal = {Plants (Basel, Switzerland)},
volume = {14},
number = {11},
pages = {},
doi = {10.3390/plants14111584},
pmid = {40508259},
issn = {2223-7747},
abstract = {Seagrass meadows play a critical role in biogeochemical cycling, especially in nitrogen and sulphur processes, driven by their associated microbiome. This study provides a novel functional analysis of microbial communities in seagrass (Zostera marina) rhizosphere and endosphere, comparing seedlings and mature plants. While nitrogen-fixing bacteria are more abundant in seedlings, mature plants exhibit greater microbial diversity and stability. Sediment samples show higher microbial diversity than roots, suggesting distinct niche environments in seagrass roots. Key microbial taxa (sulphur-oxidizing and nitrogen-cycling bacteria) were observed across developmental stages, with rapid establishment in seedlings aiding survival in sulphide-rich, anoxic sediments. Chromatiales, which oxidize sulphur, are hypothesized to support juvenile plant growth by mitigating sulphide toxicity, a key stressor in early development. Additionally, sulfate-reducing bacteria (SRB), though potentially harmful due to H2S production, may also aid in nitrogen fixation by producing ammonium. The study underscores the dynamic relationship between seagrass and its microbiome, especially the differences in microbial community structure and function between juvenile and mature plants. The study emphasizes the need for a deeper understanding of microbial roles within the seagrass holobiont to aid with Blue Carbon stores and to improve restoration success, particularly for juvenile plants struggling to establish effective microbiomes.},
}
RevDate: 2025-06-13
CmpDate: 2025-06-13
α-Synuclein Pathology in Synucleinopathies: Mechanisms, Biomarkers, and Therapeutic Challenges.
International journal of molecular sciences, 26(11): pii:ijms26115405.
Parkinson's disease and related synucleinopathies, including dementia with Lewy bodies and multiple system atrophy, are characterised by the pathological aggregation of the α-synuclein (aSyn) protein in neuronal and glial cells, leading to cellular dysfunction and neurodegeneration. This review synthesizes knowledge of aSyn biology, including its structure, aggregation mechanisms, cellular interactions, and systemic influences. We highlight the structural diversity of aSyn aggregates, ranging from oligomers to fibrils, their strain-like properties, and their prion-like propagation. While the role of prion-like mechanisms in disease progression remains a topic of ongoing debate, these processes may contribute to the clinical heterogeneity of synucleinopathies. Dysregulation of protein clearance pathways, including chaperone-mediated autophagy and the ubiquitin-proteasome system, exacerbates aSyn accumulation, while post-translational modifications influence its toxicity and aggregation propensity. Emerging evidence suggests that immune responses and alterations in the gut microbiome are key modulators of aSyn pathology, linking peripheral processes-particularly those of intestinal origin-to central neurodegeneration. Advances in biomarker development, such as cerebrospinal fluid assays, post-translationally modified aSyn, and real-time quaking-induced conversion technology, hold promise for early diagnosis and disease monitoring. Furthermore, positron emission tomography imaging and conformation-specific antibodies offer innovative tools for visualising and targeting aSyn pathology in vivo. Despite significant progress, challenges remain in accurately modelling human synucleinopathies, as existing animal and cellular models capture only specific aspects of the disease. This review underscores the need for more reliable aSyn biomarkers to facilitate the development of effective treatments. Achieving this goal requires an interdisciplinary approach integrating genetic, epigenetic, and environmental insights.
Additional Links: PMID-40508212
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40508212,
year = {2025},
author = {Arias-Carrión, O and Guerra-Crespo, M and Padilla-Godínez, FJ and Soto-Rojas, LO and Manjarrez, E},
title = {α-Synuclein Pathology in Synucleinopathies: Mechanisms, Biomarkers, and Therapeutic Challenges.},
journal = {International journal of molecular sciences},
volume = {26},
number = {11},
pages = {},
doi = {10.3390/ijms26115405},
pmid = {40508212},
issn = {1422-0067},
mesh = {Humans ; *alpha-Synuclein/metabolism/chemistry/genetics ; Biomarkers/metabolism ; *Synucleinopathies/metabolism/pathology/therapy/diagnosis ; Animals ; Parkinson Disease/metabolism/pathology ; Protein Aggregation, Pathological/metabolism ; },
abstract = {Parkinson's disease and related synucleinopathies, including dementia with Lewy bodies and multiple system atrophy, are characterised by the pathological aggregation of the α-synuclein (aSyn) protein in neuronal and glial cells, leading to cellular dysfunction and neurodegeneration. This review synthesizes knowledge of aSyn biology, including its structure, aggregation mechanisms, cellular interactions, and systemic influences. We highlight the structural diversity of aSyn aggregates, ranging from oligomers to fibrils, their strain-like properties, and their prion-like propagation. While the role of prion-like mechanisms in disease progression remains a topic of ongoing debate, these processes may contribute to the clinical heterogeneity of synucleinopathies. Dysregulation of protein clearance pathways, including chaperone-mediated autophagy and the ubiquitin-proteasome system, exacerbates aSyn accumulation, while post-translational modifications influence its toxicity and aggregation propensity. Emerging evidence suggests that immune responses and alterations in the gut microbiome are key modulators of aSyn pathology, linking peripheral processes-particularly those of intestinal origin-to central neurodegeneration. Advances in biomarker development, such as cerebrospinal fluid assays, post-translationally modified aSyn, and real-time quaking-induced conversion technology, hold promise for early diagnosis and disease monitoring. Furthermore, positron emission tomography imaging and conformation-specific antibodies offer innovative tools for visualising and targeting aSyn pathology in vivo. Despite significant progress, challenges remain in accurately modelling human synucleinopathies, as existing animal and cellular models capture only specific aspects of the disease. This review underscores the need for more reliable aSyn biomarkers to facilitate the development of effective treatments. Achieving this goal requires an interdisciplinary approach integrating genetic, epigenetic, and environmental insights.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
*alpha-Synuclein/metabolism/chemistry/genetics
Biomarkers/metabolism
*Synucleinopathies/metabolism/pathology/therapy/diagnosis
Animals
Parkinson Disease/metabolism/pathology
Protein Aggregation, Pathological/metabolism
RevDate: 2025-06-13
CmpDate: 2025-06-13
Colonizing Bacteria Aggravate Inflammation, Cytotoxicity and Immune Defense During Influenza A Virus Infection.
International journal of molecular sciences, 26(11): pii:ijms26115364.
A diverse bacterial community colonizes the respiratory system, including commensals such as Staphylococcus epidermidis (S. epidermidis) and Streptococcus salivarius (S. salivarius), as well as facultative pathogens like Staphylococcus aureus (S. aureus). This study aimed to establish a colonized cell culture model to investigate the impact of these bacteria on influenza A virus (IAV) infection. Respiratory epithelial cells were exposed to S. epidermidis, S. salivarius, or S. aureus, using either live or heat-inactivated bacteria, followed by IAV infection. Cell integrity was assessed microscopically, cytotoxicity was measured via LDH assay, and inflammatory responses were analyzed through cytokine expression. Additionally, macrophage function was examined in response to bacterial colonization and IAV infection. While commensals maintained epithelial integrity for 48 h, S. aureus induced severe cell damage and death. The most pronounced epithelial destruction was caused by coinfection with S. aureus and IAV. Notably, commensals did not confer protection against IAV but instead enhanced epithelial inflammation. These effects were dependent on live bacteria, as inactivated bacteria had no impact. However, prior exposure to S. epidermidis and S. salivarius improved macrophage-mediated immune responses against IAV. These findings suggest that while individual commensals do not directly protect epithelial cells, they may contribute to immune training and enhance lung defense mechanisms.
Additional Links: PMID-40508173
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40508173,
year = {2025},
author = {Giebeler, L and Ehrhardt, C and Häder, A and Lauf, T and Deinhardt-Emmer, S and Löffler, B},
title = {Colonizing Bacteria Aggravate Inflammation, Cytotoxicity and Immune Defense During Influenza A Virus Infection.},
journal = {International journal of molecular sciences},
volume = {26},
number = {11},
pages = {},
doi = {10.3390/ijms26115364},
pmid = {40508173},
issn = {1422-0067},
support = {LPI-BT1 and BT2//BMBF/ ; EXC 2051-Project-ID 390713860 and SFB 1278/2, D02//Deutsche Forschungsgemeinschaft/ ; },
mesh = {Humans ; *Influenza A virus/immunology ; *Inflammation/microbiology/immunology ; Macrophages/immunology/microbiology ; Staphylococcus aureus/immunology ; Animals ; *Influenza, Human/immunology/microbiology/virology ; Epithelial Cells/microbiology/virology/immunology ; Staphylococcus epidermidis/immunology ; Cytokines/metabolism ; Coinfection/microbiology/immunology ; *Orthomyxoviridae Infections/immunology/microbiology ; A549 Cells ; },
abstract = {A diverse bacterial community colonizes the respiratory system, including commensals such as Staphylococcus epidermidis (S. epidermidis) and Streptococcus salivarius (S. salivarius), as well as facultative pathogens like Staphylococcus aureus (S. aureus). This study aimed to establish a colonized cell culture model to investigate the impact of these bacteria on influenza A virus (IAV) infection. Respiratory epithelial cells were exposed to S. epidermidis, S. salivarius, or S. aureus, using either live or heat-inactivated bacteria, followed by IAV infection. Cell integrity was assessed microscopically, cytotoxicity was measured via LDH assay, and inflammatory responses were analyzed through cytokine expression. Additionally, macrophage function was examined in response to bacterial colonization and IAV infection. While commensals maintained epithelial integrity for 48 h, S. aureus induced severe cell damage and death. The most pronounced epithelial destruction was caused by coinfection with S. aureus and IAV. Notably, commensals did not confer protection against IAV but instead enhanced epithelial inflammation. These effects were dependent on live bacteria, as inactivated bacteria had no impact. However, prior exposure to S. epidermidis and S. salivarius improved macrophage-mediated immune responses against IAV. These findings suggest that while individual commensals do not directly protect epithelial cells, they may contribute to immune training and enhance lung defense mechanisms.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
*Influenza A virus/immunology
*Inflammation/microbiology/immunology
Macrophages/immunology/microbiology
Staphylococcus aureus/immunology
Animals
*Influenza, Human/immunology/microbiology/virology
Epithelial Cells/microbiology/virology/immunology
Staphylococcus epidermidis/immunology
Cytokines/metabolism
Coinfection/microbiology/immunology
*Orthomyxoviridae Infections/immunology/microbiology
A549 Cells
RevDate: 2025-06-13
CmpDate: 2025-06-13
Microbiome Engineering for Biotherapeutic in Alzheimer's Disease Through the Gut-Brain Axis: Potentials and Limitations.
International journal of molecular sciences, 26(11): pii:ijms26115351.
Alzheimer's disease (AD) is a neurodegenerative condition characterized by considerable cognitive decline and functional impairment, primarily due to the progressive alteration of neurons, microglia, and astrocytes. Pathological manifestations of AD include the loss of synaptic plasticity, reduction in synaptic strength by amyloid-beta, aggregation, and neurotoxicity from tau protein post-translational modifications, all contributing to the disruption of neural networks. Despite its current pharmacological treatment for AD, different approaches to treat such disease are being developed, from a microbiome perspective. The microbiome encompasses a diverse microorganism, including beneficial bacteria that create a positive impact to diminish AD pathogenesis. Growing evidence suggests that probiotic, prebiotic, synbiotic, and postbiotics can positively modulate the gut-brain axis, reducing systemic inflammation, restoring neurotransmitter balance, and improving gut health, thereby possibly mitigating AD pathogenesis. Moreover, there is paraprobiotics as the most recently developed biotherapeutic with beneficial effects. This review explores the correlation between AD and gut-brain axis as a novel biotherapeutic target. The underlying mechanism of the microbiota-gut-brain axis in AD is examined. Novel insights into the current applications as potential treatment and its limitations are highlighted.
Additional Links: PMID-40508160
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40508160,
year = {2025},
author = {Renesteen, E and Boyajian, JL and Islam, P and Kassab, A and Abosalha, A and Makhlouf, S and Santos, M and Chen, H and Shum-Tim, C and Prakash, S},
title = {Microbiome Engineering for Biotherapeutic in Alzheimer's Disease Through the Gut-Brain Axis: Potentials and Limitations.},
journal = {International journal of molecular sciences},
volume = {26},
number = {11},
pages = {},
doi = {10.3390/ijms26115351},
pmid = {40508160},
issn = {1422-0067},
support = {252743//CIHR/ ; //Indonesia Endowment Fund for Education from the Ministry of Finance of the Republic of Indonesia/ ; 335999//Fonds de Recherche du Québec - Santé (FRQS)/ ; 2020-245622//Islamic Development Bank Scholarship/ ; //The Ministry of Higher Education of the Arab Republic of Egypt/ ; 504933//Canadian Graduate Scholarship-Masters from the Natural Sciences and Engineering Research Council (NSERC)/ ; 351337//Fonds de Recherche du Québec - Santé (FRQS)/ ; },
mesh = {Humans ; *Alzheimer Disease/therapy/microbiology/metabolism ; *Gastrointestinal Microbiome ; *Brain/metabolism ; Probiotics/therapeutic use ; Animals ; Prebiotics/administration & dosage ; *Brain-Gut Axis ; *Biological Therapy/methods ; },
abstract = {Alzheimer's disease (AD) is a neurodegenerative condition characterized by considerable cognitive decline and functional impairment, primarily due to the progressive alteration of neurons, microglia, and astrocytes. Pathological manifestations of AD include the loss of synaptic plasticity, reduction in synaptic strength by amyloid-beta, aggregation, and neurotoxicity from tau protein post-translational modifications, all contributing to the disruption of neural networks. Despite its current pharmacological treatment for AD, different approaches to treat such disease are being developed, from a microbiome perspective. The microbiome encompasses a diverse microorganism, including beneficial bacteria that create a positive impact to diminish AD pathogenesis. Growing evidence suggests that probiotic, prebiotic, synbiotic, and postbiotics can positively modulate the gut-brain axis, reducing systemic inflammation, restoring neurotransmitter balance, and improving gut health, thereby possibly mitigating AD pathogenesis. Moreover, there is paraprobiotics as the most recently developed biotherapeutic with beneficial effects. This review explores the correlation between AD and gut-brain axis as a novel biotherapeutic target. The underlying mechanism of the microbiota-gut-brain axis in AD is examined. Novel insights into the current applications as potential treatment and its limitations are highlighted.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
*Alzheimer Disease/therapy/microbiology/metabolism
*Gastrointestinal Microbiome
*Brain/metabolism
Probiotics/therapeutic use
Animals
Prebiotics/administration & dosage
*Brain-Gut Axis
*Biological Therapy/methods
RevDate: 2025-06-13
CmpDate: 2025-06-13
Quorum Signaling Molecules: Interactions Between Plants and Associated Pathogens.
International journal of molecular sciences, 26(11): pii:ijms26115235.
The morphogenesis and defense evolution of plants are intricately linked to soil microbial community dynamics, where beneficial and pathogenic bacteria regulate ecosystem stability through chemical signaling. A microbial communication mechanism known as quorum sensing (QS), which affects population density, virulence, and biofilm formation, substantially impacts plant development and immune responses. However, plants have developed strategies to detect and manipulate QS signals, enabling bidirectional interactions that influence both plant physiology and the balance of the microbiome. In this review, QS signals from bacteria, fungi, and nematodes are systematically examined, emphasizing their recognition by plant receptors, downstream signaling pathways, and the activation of defense responses. Most significantly, attention is given to the role of fungal and nematode QS molecules in modulating plant microbe interactions. By elucidating these communication networks, we highlight their potential applications in sustainable agriculture, offering novel insights into crop health management and ecosystem resilience.
Additional Links: PMID-40508052
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40508052,
year = {2025},
author = {Zheng, X and Liu, J and Wang, X},
title = {Quorum Signaling Molecules: Interactions Between Plants and Associated Pathogens.},
journal = {International journal of molecular sciences},
volume = {26},
number = {11},
pages = {},
doi = {10.3390/ijms26115235},
pmid = {40508052},
issn = {1422-0067},
support = {202201BC070004//The Science and Technology Major Project from Yunnan Province/ ; 32370138//The National Natural Science Foundation of China/ ; 32260029//The National Natural Science Foundation of China/ ; 202201AT070089//The Applied Basic Research Foundation of Yunnan Province/ ; 2023YFD1400013//The National Key Research and Development Program/ ; },
mesh = {*Quorum Sensing ; *Plants/microbiology/metabolism/parasitology ; Signal Transduction ; Fungi/pathogenicity/metabolism ; Animals ; Bacteria/pathogenicity/metabolism ; Nematoda/physiology ; *Host-Pathogen Interactions ; Plant Diseases/microbiology ; Microbiota ; },
abstract = {The morphogenesis and defense evolution of plants are intricately linked to soil microbial community dynamics, where beneficial and pathogenic bacteria regulate ecosystem stability through chemical signaling. A microbial communication mechanism known as quorum sensing (QS), which affects population density, virulence, and biofilm formation, substantially impacts plant development and immune responses. However, plants have developed strategies to detect and manipulate QS signals, enabling bidirectional interactions that influence both plant physiology and the balance of the microbiome. In this review, QS signals from bacteria, fungi, and nematodes are systematically examined, emphasizing their recognition by plant receptors, downstream signaling pathways, and the activation of defense responses. Most significantly, attention is given to the role of fungal and nematode QS molecules in modulating plant microbe interactions. By elucidating these communication networks, we highlight their potential applications in sustainable agriculture, offering novel insights into crop health management and ecosystem resilience.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
*Quorum Sensing
*Plants/microbiology/metabolism/parasitology
Signal Transduction
Fungi/pathogenicity/metabolism
Animals
Bacteria/pathogenicity/metabolism
Nematoda/physiology
*Host-Pathogen Interactions
Plant Diseases/microbiology
Microbiota
RevDate: 2025-06-13
CmpDate: 2025-06-13
Impact of DNA Extraction and 16S rRNA Gene Amplification Strategy on Microbiota Profiling of Faecal Samples.
International journal of molecular sciences, 26(11): pii:ijms26115226.
High-throughput 16S rRNA metagenomic sequencing has advanced our understanding of the gut microbiome, but its reliability depends on upstream processes such as DNA extraction and bacterial library preparation. In this study, we evaluated the impact of three different DNA extraction methods (a manual method with an ad hoc-designed pre-extraction phase (PE-QIA), and two automated magnetic bead-based methods (T180H and TAT132H)) and two bacterial library preparation protocols (home brew and VeriFi) on the 16S rRNA-based metagenomic profiling of faecal samples. T180H and TAT132H produced significantly higher DNA concentrations than PE-QIA, whereas TAT132H yielded DNA of lower purity compared to the others. In the taxonomic analysis, PE-QIA provided a balanced recovery of Gram-positive and Gram-negative bacteria, TAT132H was enriched in Gram-positive taxa, and T180H was enriched in Gram-negative taxa. An analysis of Microbial Community Standard (MOCK) samples showed that PE-QIA and T180H were more accurate than TAT132H. Finally, the VeriFi method yielded higher amplicon concentrations and sequence counts than the home brew protocol, despite the high level of chimeras. In conclusion, a robust performance in terms of DNA yield, purity, and taxonomic representation was obtained by PE-QIA and T180H. Furthermore, it was found that the impact of PCR-based steps on gut microbiota profiling can be minimized by an accurate bioinformatic pipeline.
Additional Links: PMID-40508035
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40508035,
year = {2025},
author = {Toto, F and Scanu, M and Gramegna, M and Putignani, L and Del Chierico, F},
title = {Impact of DNA Extraction and 16S rRNA Gene Amplification Strategy on Microbiota Profiling of Faecal Samples.},
journal = {International journal of molecular sciences},
volume = {26},
number = {11},
pages = {},
doi = {10.3390/ijms26115226},
pmid = {40508035},
issn = {1422-0067},
support = {Current Research funds//Italian Ministry of Health/ ; n.a.//Technogenetics S.p.A./ ; },
mesh = {*RNA, Ribosomal, 16S/genetics ; *Feces/microbiology ; Humans ; *DNA, Bacterial/genetics/isolation & purification ; *Gastrointestinal Microbiome/genetics ; Metagenomics/methods ; High-Throughput Nucleotide Sequencing/methods ; *Microbiota/genetics ; Bacteria/genetics/classification ; Metagenome ; },
abstract = {High-throughput 16S rRNA metagenomic sequencing has advanced our understanding of the gut microbiome, but its reliability depends on upstream processes such as DNA extraction and bacterial library preparation. In this study, we evaluated the impact of three different DNA extraction methods (a manual method with an ad hoc-designed pre-extraction phase (PE-QIA), and two automated magnetic bead-based methods (T180H and TAT132H)) and two bacterial library preparation protocols (home brew and VeriFi) on the 16S rRNA-based metagenomic profiling of faecal samples. T180H and TAT132H produced significantly higher DNA concentrations than PE-QIA, whereas TAT132H yielded DNA of lower purity compared to the others. In the taxonomic analysis, PE-QIA provided a balanced recovery of Gram-positive and Gram-negative bacteria, TAT132H was enriched in Gram-positive taxa, and T180H was enriched in Gram-negative taxa. An analysis of Microbial Community Standard (MOCK) samples showed that PE-QIA and T180H were more accurate than TAT132H. Finally, the VeriFi method yielded higher amplicon concentrations and sequence counts than the home brew protocol, despite the high level of chimeras. In conclusion, a robust performance in terms of DNA yield, purity, and taxonomic representation was obtained by PE-QIA and T180H. Furthermore, it was found that the impact of PCR-based steps on gut microbiota profiling can be minimized by an accurate bioinformatic pipeline.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
*RNA, Ribosomal, 16S/genetics
*Feces/microbiology
Humans
*DNA, Bacterial/genetics/isolation & purification
*Gastrointestinal Microbiome/genetics
Metagenomics/methods
High-Throughput Nucleotide Sequencing/methods
*Microbiota/genetics
Bacteria/genetics/classification
Metagenome
RevDate: 2025-06-13
CmpDate: 2025-06-13
Gut Microbiota Dysbiosis in Endometriosis: A Potential Link to Inflammation and Disease Progression.
International journal of molecular sciences, 26(11): pii:ijms26115144.
Endometriosis is a complex gynaecological disorder characterised by the presence of endometrial-like tissue outside the uterus, leading to chronic inflammation, pain, and infertility. Recent research suggests that gut microbiota may play a crucial role in the pathogenesis and progression of endometriosis by modulating immune responses and oestrogen metabolism. This study investigates the intestinal microbiota composition in women with endometriosis and its potential as a disease diagnosis and severity biomarker. Stool samples from nine patients diagnosed with endometriosis were analysed using the GI Effects[®] Comprehensive Stool Profile test. The tests revealed significant dysbiosis, particularly an altered Firmicutes/Bacteroidetes ratio and increased levels of Bacteroidetes. Inflammatory markers, including β-glucuronidase and secretory IgA, were also elevated, suggesting a potential link between gut microbiota and systemic inflammation in endometriosis. While our findings align with previous studies, further research with larger cohorts is necessary to validate these observations. Understanding the role of the microbiome in endometriosis could open new avenues for noninvasive diagnostic tools in endometriosis and microbiota-targeted therapies.
Additional Links: PMID-40507956
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40507956,
year = {2025},
author = {Baușic, AIG and Scurtu, F and Manu, A and Matasariu, DR and Brătilă, E},
title = {Gut Microbiota Dysbiosis in Endometriosis: A Potential Link to Inflammation and Disease Progression.},
journal = {International journal of molecular sciences},
volume = {26},
number = {11},
pages = {},
doi = {10.3390/ijms26115144},
pmid = {40507956},
issn = {1422-0067},
support = {"PUBLISH NOT PERISH" - 2025//Carol Davila University of Medicine and Pharmacy/ ; },
mesh = {Humans ; Female ; *Endometriosis/microbiology/pathology ; *Dysbiosis/microbiology/complications ; *Gastrointestinal Microbiome ; Adult ; Disease Progression ; *Inflammation/microbiology ; Biomarkers ; Feces/microbiology ; },
abstract = {Endometriosis is a complex gynaecological disorder characterised by the presence of endometrial-like tissue outside the uterus, leading to chronic inflammation, pain, and infertility. Recent research suggests that gut microbiota may play a crucial role in the pathogenesis and progression of endometriosis by modulating immune responses and oestrogen metabolism. This study investigates the intestinal microbiota composition in women with endometriosis and its potential as a disease diagnosis and severity biomarker. Stool samples from nine patients diagnosed with endometriosis were analysed using the GI Effects[®] Comprehensive Stool Profile test. The tests revealed significant dysbiosis, particularly an altered Firmicutes/Bacteroidetes ratio and increased levels of Bacteroidetes. Inflammatory markers, including β-glucuronidase and secretory IgA, were also elevated, suggesting a potential link between gut microbiota and systemic inflammation in endometriosis. While our findings align with previous studies, further research with larger cohorts is necessary to validate these observations. Understanding the role of the microbiome in endometriosis could open new avenues for noninvasive diagnostic tools in endometriosis and microbiota-targeted therapies.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
Female
*Endometriosis/microbiology/pathology
*Dysbiosis/microbiology/complications
*Gastrointestinal Microbiome
Adult
Disease Progression
*Inflammation/microbiology
Biomarkers
Feces/microbiology
RevDate: 2025-06-13
CmpDate: 2025-06-13
Synbiotic Supplementation Attenuates Doxorubicin-Induced Oxidative Stress and Inflammation in the Gut-Heart Axis of Chemotherapy-Treated Mice.
International journal of molecular sciences, 26(11): pii:ijms26115136.
The gut microbiome supports immune health and influences gut and heart functions through the gut-heart axis. Synbiotics (SBT), combining probiotics and prebiotics, help restore microbiome balance. Chemotherapy often disrupts this balance, leading to adverse effects on the gut and heart. This study explores the potential of SBT supplementation in reducing heart and gut inflammation caused by doxorubicin (DOX) chemotherapy. The gut microbiome plays a vital role in immune health, and metabolites produced by gut bacteria contribute to physiological functions through the gut-heart axis. Chemotherapy drugs often disrupt these processes, leading to adverse effects on internal organs. Using 24 ICR male mice divided into four groups, the experiment assessed the impact of SBT on DOX-induced damage. Results indicated that DOX treatment significantly worsened survival rates, physical performance, heart function, and gut microbiome stability. However, co-treatment with SBT improved these markers, suggesting that SBT may help mitigate chemotherapy-induced side effects in cancer patients.
Additional Links: PMID-40507947
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40507947,
year = {2025},
author = {Cheng, CF and Nguyen, TKN and Shen, SC and Chen, BY and Wu, YB and Liang, HJ and Wu, CH},
title = {Synbiotic Supplementation Attenuates Doxorubicin-Induced Oxidative Stress and Inflammation in the Gut-Heart Axis of Chemotherapy-Treated Mice.},
journal = {International journal of molecular sciences},
volume = {26},
number = {11},
pages = {},
doi = {10.3390/ijms26115136},
pmid = {40507947},
issn = {1422-0067},
support = {N/A//This study was partly funded by Industry-Academic Cooperation Project from ARJIL Pharmaceu-ticals LLC, TTY Biopharm Co., Ltd., and Shiun Chao Biopharm Co., Ltd./ ; },
mesh = {Animals ; *Doxorubicin/adverse effects ; *Synbiotics/administration & dosage ; *Gastrointestinal Microbiome/drug effects ; Mice ; *Oxidative Stress/drug effects ; Male ; *Inflammation/chemically induced/metabolism ; Mice, Inbred ICR ; *Heart/drug effects ; Dietary Supplements ; Probiotics ; },
abstract = {The gut microbiome supports immune health and influences gut and heart functions through the gut-heart axis. Synbiotics (SBT), combining probiotics and prebiotics, help restore microbiome balance. Chemotherapy often disrupts this balance, leading to adverse effects on the gut and heart. This study explores the potential of SBT supplementation in reducing heart and gut inflammation caused by doxorubicin (DOX) chemotherapy. The gut microbiome plays a vital role in immune health, and metabolites produced by gut bacteria contribute to physiological functions through the gut-heart axis. Chemotherapy drugs often disrupt these processes, leading to adverse effects on internal organs. Using 24 ICR male mice divided into four groups, the experiment assessed the impact of SBT on DOX-induced damage. Results indicated that DOX treatment significantly worsened survival rates, physical performance, heart function, and gut microbiome stability. However, co-treatment with SBT improved these markers, suggesting that SBT may help mitigate chemotherapy-induced side effects in cancer patients.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Animals
*Doxorubicin/adverse effects
*Synbiotics/administration & dosage
*Gastrointestinal Microbiome/drug effects
Mice
*Oxidative Stress/drug effects
Male
*Inflammation/chemically induced/metabolism
Mice, Inbred ICR
*Heart/drug effects
Dietary Supplements
Probiotics
RevDate: 2025-06-13
CmpDate: 2025-06-13
Investigating the Role of Gut Microbiota in the Pathogenesis and Progression of Rheumatoid Arthritis in a Collagen-Induced Arthritis Mouse Model.
International journal of molecular sciences, 26(11): pii:ijms26115099.
Rheumatoid arthritis (RA) is a chronic systemic autoimmune disorder whose precise etiology remains unclear, though growing evidence implicates gut microbiota in its pathogenesis. This study aimed to investigate the role of gut microbiota in the onset and progression of RA by employing fecal microbiota transplantation (FMT) in a collagen-induced arthritis (CIA) mouse model using DBA/1J and Aire[-]/[-] strains. Mice received FMT from healthy donors, treatment-naïve RA patients, or treated RA patients in relapse, followed by assessment of microbiota composition via 16S rRNA sequencing, arthritis severity scoring, histological evaluations, and systemic inflammatory markers. The findings revealed distinct microbiota clustering patterns post-FMT across experimental groups, highlighting strain-specific colonization effects. Notably, genera such as Bifidobacterium and Paraprevotella correlated positively with arthritis severity in DBA/1J mice, whereas Corynebacterium, Enterorhabdus, and Odoribacter exhibited negative correlations, suggesting potential protective roles. Despite these microbial differences, minor variations in arthritis scores, paw inflammation, or systemic inflammation were observed among FMT groups. This indicates that although gut microbiota alterations are associated with RA pathogenesis, further investigation with larger cohorts and comprehensive sequencing approaches is essential to elucidate the therapeutic potential of microbiome modulation in autoimmune diseases.
Additional Links: PMID-40507919
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40507919,
year = {2025},
author = {Belvončíková, P and Macáková, K and Tóthová, N and Babál, P and Tarabčáková, L and Gardlík, R},
title = {Investigating the Role of Gut Microbiota in the Pathogenesis and Progression of Rheumatoid Arthritis in a Collagen-Induced Arthritis Mouse Model.},
journal = {International journal of molecular sciences},
volume = {26},
number = {11},
pages = {},
doi = {10.3390/ijms26115099},
pmid = {40507919},
issn = {1422-0067},
support = {APVV-21-0370//Slovak Research and Development Agency/ ; VEGA 1/0706/25//Ministry of Education, Science, Research and Sport of the Slovak Republic/ ; },
mesh = {Animals ; *Gastrointestinal Microbiome ; *Arthritis, Rheumatoid/microbiology/pathology/therapy/etiology ; *Arthritis, Experimental/microbiology/pathology/therapy ; Mice ; Disease Models, Animal ; RNA, Ribosomal, 16S/genetics ; Disease Progression ; Humans ; Mice, Inbred DBA ; Male ; Fecal Microbiota Transplantation ; Female ; },
abstract = {Rheumatoid arthritis (RA) is a chronic systemic autoimmune disorder whose precise etiology remains unclear, though growing evidence implicates gut microbiota in its pathogenesis. This study aimed to investigate the role of gut microbiota in the onset and progression of RA by employing fecal microbiota transplantation (FMT) in a collagen-induced arthritis (CIA) mouse model using DBA/1J and Aire[-]/[-] strains. Mice received FMT from healthy donors, treatment-naïve RA patients, or treated RA patients in relapse, followed by assessment of microbiota composition via 16S rRNA sequencing, arthritis severity scoring, histological evaluations, and systemic inflammatory markers. The findings revealed distinct microbiota clustering patterns post-FMT across experimental groups, highlighting strain-specific colonization effects. Notably, genera such as Bifidobacterium and Paraprevotella correlated positively with arthritis severity in DBA/1J mice, whereas Corynebacterium, Enterorhabdus, and Odoribacter exhibited negative correlations, suggesting potential protective roles. Despite these microbial differences, minor variations in arthritis scores, paw inflammation, or systemic inflammation were observed among FMT groups. This indicates that although gut microbiota alterations are associated with RA pathogenesis, further investigation with larger cohorts and comprehensive sequencing approaches is essential to elucidate the therapeutic potential of microbiome modulation in autoimmune diseases.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Animals
*Gastrointestinal Microbiome
*Arthritis, Rheumatoid/microbiology/pathology/therapy/etiology
*Arthritis, Experimental/microbiology/pathology/therapy
Mice
Disease Models, Animal
RNA, Ribosomal, 16S/genetics
Disease Progression
Humans
Mice, Inbred DBA
Male
Fecal Microbiota Transplantation
Female
RevDate: 2025-06-13
CmpDate: 2025-06-13
Roseburia intestinalis Modulates Immune Responses by Inducing M1 Macrophage Polarization.
International journal of molecular sciences, 26(11): pii:ijms26115049.
In recent years, the gut microbiome has been recognized as one influential factor in cancer development. Particularly in colorectal cancer (CRC), several studies observed a major imbalance of the intestinal microbiota, marked by a reduction in beneficial bacterial species, such as Roseburia intestinalis, and an increase in opportunistic pathobionts, like Peptostreptococcus stomatis. We previously observed that specific Eubacteriales, including R. intestinalis, were significantly reduced in CRC patients and have a potent anti-tumor immune effect when applied as oral monotherapy in mice. Here, we investigate the molecular mechanism of R. intestinalis on various cell types in vitro, highlighting its potential therapeutic value in CRC. Co-culture experiments with macrophages demonstrated that R. intestinalis exposure induced an increase in the M1 phenotype and decreased the M2 phenotype, suggesting macrophage-polarizing properties of these bacteria. R. intestinalis also triggered a gene expression profile resembling M1 macrophages and led to distinct chemokine and cytokine secretion in cancer cells, suggesting an immune-activating environment. However, we did not observe direct cytotoxic effects in cancer cells. Our research provides insights into the potential of R. intestinalis to activate immune responses, supporting further investigation into its therapeutic role in CRC. These findings underscore the need for deeper studies on the bacterium's impact on CRC pathogenesis and treatment.
Additional Links: PMID-40507861
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40507861,
year = {2025},
author = {Bircher, A and Katkeviciute, E and Morsy, Y and Lang, S and Montalban-Arques, A and Scharl, M},
title = {Roseburia intestinalis Modulates Immune Responses by Inducing M1 Macrophage Polarization.},
journal = {International journal of molecular sciences},
volume = {26},
number = {11},
pages = {},
doi = {10.3390/ijms26115049},
pmid = {40507861},
issn = {1422-0067},
support = {KFS-5372-08-2021-R//Swiss Cancer Research Foundation/ ; NA//Fondazione San Salvatore/ ; NA//Holcim Foundation/ ; },
mesh = {*Macrophages/immunology/microbiology/metabolism ; Humans ; Animals ; Colorectal Neoplasms/immunology/microbiology ; *Clostridiales/immunology ; Mice ; Gastrointestinal Microbiome/immunology ; *Macrophage Activation/immunology ; Cytokines/metabolism ; Cell Line, Tumor ; Coculture Techniques ; },
abstract = {In recent years, the gut microbiome has been recognized as one influential factor in cancer development. Particularly in colorectal cancer (CRC), several studies observed a major imbalance of the intestinal microbiota, marked by a reduction in beneficial bacterial species, such as Roseburia intestinalis, and an increase in opportunistic pathobionts, like Peptostreptococcus stomatis. We previously observed that specific Eubacteriales, including R. intestinalis, were significantly reduced in CRC patients and have a potent anti-tumor immune effect when applied as oral monotherapy in mice. Here, we investigate the molecular mechanism of R. intestinalis on various cell types in vitro, highlighting its potential therapeutic value in CRC. Co-culture experiments with macrophages demonstrated that R. intestinalis exposure induced an increase in the M1 phenotype and decreased the M2 phenotype, suggesting macrophage-polarizing properties of these bacteria. R. intestinalis also triggered a gene expression profile resembling M1 macrophages and led to distinct chemokine and cytokine secretion in cancer cells, suggesting an immune-activating environment. However, we did not observe direct cytotoxic effects in cancer cells. Our research provides insights into the potential of R. intestinalis to activate immune responses, supporting further investigation into its therapeutic role in CRC. These findings underscore the need for deeper studies on the bacterium's impact on CRC pathogenesis and treatment.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
*Macrophages/immunology/microbiology/metabolism
Humans
Animals
Colorectal Neoplasms/immunology/microbiology
*Clostridiales/immunology
Mice
Gastrointestinal Microbiome/immunology
*Macrophage Activation/immunology
Cytokines/metabolism
Cell Line, Tumor
Coculture Techniques
RevDate: 2025-06-13
CmpDate: 2025-06-13
Exploring the Healing Powers of Histatins: From Oral Health to Therapeutics.
International journal of molecular sciences, 26(11): pii:ijms26115019.
Histatin peptides are a family of small histidine-rich cationic polypeptides produced by two genes, HTN1 and HTN3. They are found in salivary secretions from the parotid, sublingual, and submandibular salivary glands. These peptides undergo proteolytic cleavages to produce different histatin fragments which play multiple roles including wound healing, maintenance of enamel, and regulation of balance in the oral microbiome. In this review, we explored the expression, structural characteristics, and metal-ion-binding capacities of these peptides and how their functions are modulated by their structure. We also provide here an insight into the potential use of histatins as biomarkers and therapeutic peptides in the management of oral and non-oral diseases including cancer. Potential gaps in the current understanding of histatins that warrant further research have also been highlighted.
Additional Links: PMID-40507844
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40507844,
year = {2025},
author = {Usman, S and You, Y and Waseem, A},
title = {Exploring the Healing Powers of Histatins: From Oral Health to Therapeutics.},
journal = {International journal of molecular sciences},
volume = {26},
number = {11},
pages = {},
doi = {10.3390/ijms26115019},
pmid = {40507844},
issn = {1422-0067},
mesh = {Humans ; *Histatins/chemistry/metabolism/therapeutic use/genetics/pharmacology ; *Wound Healing ; *Oral Health ; Animals ; },
abstract = {Histatin peptides are a family of small histidine-rich cationic polypeptides produced by two genes, HTN1 and HTN3. They are found in salivary secretions from the parotid, sublingual, and submandibular salivary glands. These peptides undergo proteolytic cleavages to produce different histatin fragments which play multiple roles including wound healing, maintenance of enamel, and regulation of balance in the oral microbiome. In this review, we explored the expression, structural characteristics, and metal-ion-binding capacities of these peptides and how their functions are modulated by their structure. We also provide here an insight into the potential use of histatins as biomarkers and therapeutic peptides in the management of oral and non-oral diseases including cancer. Potential gaps in the current understanding of histatins that warrant further research have also been highlighted.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
*Histatins/chemistry/metabolism/therapeutic use/genetics/pharmacology
*Wound Healing
*Oral Health
Animals
RevDate: 2025-06-13
CmpDate: 2025-06-13
A Comparison of the Effects of Milk, Yogurt, and Cheese on Insulin Sensitivity, Hepatic Steatosis, and Gut Microbiota in Diet-Induced Obese Male Mice.
International journal of molecular sciences, 26(11): pii:ijms26115026.
The effects of low-fat dairy products on insulin resistance (IR), hepatic steatosis, and gut microbiota composition in high-fat diet (HFD)-fed obese mice were examined. C57BL/6 male mice (n = 16/group) were fed a high-fat diet (HFD, 45% fat) or HFD supplemented with either fat-free milk (MILK), fat-free yogurt (YOG), or reduced-fat (19% milk fat) cheddar cheese (CHE) at 10% of the total energy intake for 8 weeks. Body weight, fat mass, liver lipids, and metabolic enzymes were evaluated. Compared with HFD, MILK reduced homeostatic assessment of insulin resistance along with increased hepatic insulin signaling and decreased hepatic gluconeogenic enzymes. YOG and MILK decreased hepatic triacylglycerol content and lipid droplet size, while CHE had no effect. In the liver, MILK and YOG downregulated de novo lipogenesis enzymes. In MILK, fat oxidation capacity was elevated. Compared with HFD, liver lipidomic analysis in MILK and YOG revealed unique profiles of decreased proinflammatory lipid species, including ceramides. Dairy feeding elicited an increase in beneficial bacteria, such as Streptococcus in YOG and Anaero-tignum in MILK, as shown by 16S rRNA sequencing of gut microbiota. In conclusion, the ability of milk and yogurt to reduce hepatic steatosis in HFD mice may be explained, at least in part, by the regulation of the gut microbiome and liver lipidome.
Additional Links: PMID-40507838
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40507838,
year = {2025},
author = {Yuzbashian, E and Fernando, DN and Jacobs, RL and Lesker, TR and Strowig, T and Ussar, S and Chan, CB},
title = {A Comparison of the Effects of Milk, Yogurt, and Cheese on Insulin Sensitivity, Hepatic Steatosis, and Gut Microbiota in Diet-Induced Obese Male Mice.},
journal = {International journal of molecular sciences},
volume = {26},
number = {11},
pages = {},
doi = {10.3390/ijms26115026},
pmid = {40507838},
issn = {1422-0067},
mesh = {Animals ; *Gastrointestinal Microbiome ; *Yogurt ; Male ; Mice ; *Milk ; Diet, High-Fat/adverse effects ; *Insulin Resistance ; *Cheese ; *Obesity/metabolism/etiology/microbiology ; Mice, Inbred C57BL ; Liver/metabolism ; *Fatty Liver/metabolism/etiology ; Mice, Obese ; Lipid Metabolism ; },
abstract = {The effects of low-fat dairy products on insulin resistance (IR), hepatic steatosis, and gut microbiota composition in high-fat diet (HFD)-fed obese mice were examined. C57BL/6 male mice (n = 16/group) were fed a high-fat diet (HFD, 45% fat) or HFD supplemented with either fat-free milk (MILK), fat-free yogurt (YOG), or reduced-fat (19% milk fat) cheddar cheese (CHE) at 10% of the total energy intake for 8 weeks. Body weight, fat mass, liver lipids, and metabolic enzymes were evaluated. Compared with HFD, MILK reduced homeostatic assessment of insulin resistance along with increased hepatic insulin signaling and decreased hepatic gluconeogenic enzymes. YOG and MILK decreased hepatic triacylglycerol content and lipid droplet size, while CHE had no effect. In the liver, MILK and YOG downregulated de novo lipogenesis enzymes. In MILK, fat oxidation capacity was elevated. Compared with HFD, liver lipidomic analysis in MILK and YOG revealed unique profiles of decreased proinflammatory lipid species, including ceramides. Dairy feeding elicited an increase in beneficial bacteria, such as Streptococcus in YOG and Anaero-tignum in MILK, as shown by 16S rRNA sequencing of gut microbiota. In conclusion, the ability of milk and yogurt to reduce hepatic steatosis in HFD mice may be explained, at least in part, by the regulation of the gut microbiome and liver lipidome.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Animals
*Gastrointestinal Microbiome
*Yogurt
Male
Mice
*Milk
Diet, High-Fat/adverse effects
*Insulin Resistance
*Cheese
*Obesity/metabolism/etiology/microbiology
Mice, Inbred C57BL
Liver/metabolism
*Fatty Liver/metabolism/etiology
Mice, Obese
Lipid Metabolism
RevDate: 2025-06-13
CmpDate: 2025-06-13
Skin Microbiome and Radiation-Induced Skin Injury: Unraveling the Relationship, Mechanisms, and Therapeutic Implications.
International journal of molecular sciences, 26(11): pii:ijms26115022.
Radiotherapy (RT) is a treatment method commonly used in oncology. A vast majority of patients undergoing RT suffer from radiation-induced skin injury (RISI), which results from complex biochemical reactions in the irradiated skin. Current strategies for preventing and managing RISI are insufficient for achieving full skin regeneration. Multiple studies have shown that alterations in the skin microbiome correlate with the development and severity of RISI. These studies suggest that dysbiosis is a crucial factor in promoting radiation-associated dermatitis. Targeting the skin microbiota presents a potential therapeutic approach that could significantly improve the quality of life for patients undergoing RT. This review aims to present current findings on the interplay between the skin microbiome and radiation-induced skin damage as well as to discuss potential therapeutic strategies for preventing and mitigating this condition.
Additional Links: PMID-40507833
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40507833,
year = {2025},
author = {Bratborska, AW and Głuszak, P and Joks, M and Kaźmierska, J and Pazdrowski, J and Polańska, A and Jain, S and Yadav, H and Masternak, MM and Dańczak-Pazdrowska, A},
title = {Skin Microbiome and Radiation-Induced Skin Injury: Unraveling the Relationship, Mechanisms, and Therapeutic Implications.},
journal = {International journal of molecular sciences},
volume = {26},
number = {11},
pages = {},
doi = {10.3390/ijms26115022},
pmid = {40507833},
issn = {1422-0067},
mesh = {Humans ; *Skin/microbiology/radiation effects/pathology ; *Microbiota/radiation effects ; Animals ; *Radiodermatitis/microbiology/therapy/etiology ; Dysbiosis/microbiology ; *Radiation Injuries/microbiology ; Radiotherapy/adverse effects ; Skin Microbiome ; },
abstract = {Radiotherapy (RT) is a treatment method commonly used in oncology. A vast majority of patients undergoing RT suffer from radiation-induced skin injury (RISI), which results from complex biochemical reactions in the irradiated skin. Current strategies for preventing and managing RISI are insufficient for achieving full skin regeneration. Multiple studies have shown that alterations in the skin microbiome correlate with the development and severity of RISI. These studies suggest that dysbiosis is a crucial factor in promoting radiation-associated dermatitis. Targeting the skin microbiota presents a potential therapeutic approach that could significantly improve the quality of life for patients undergoing RT. This review aims to present current findings on the interplay between the skin microbiome and radiation-induced skin damage as well as to discuss potential therapeutic strategies for preventing and mitigating this condition.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
*Skin/microbiology/radiation effects/pathology
*Microbiota/radiation effects
Animals
*Radiodermatitis/microbiology/therapy/etiology
Dysbiosis/microbiology
*Radiation Injuries/microbiology
Radiotherapy/adverse effects
Skin Microbiome
RevDate: 2025-06-13
CmpDate: 2025-06-13
The MDA-MB-231 Breast Cancer Cell Secretomes Modify Metabolomes of Pseudomonas aeruginosa Breast Microbiome.
International journal of molecular sciences, 26(11): pii:ijms26115003.
Breast cancer (BC) is globally becoming a great challenge, being both the most diagnosed cancer and the leading cause of death in women. In addition to cancer cells, many bacteria co-inhabit BC, which differ in type and number from the resident microbiota found in healthy breast tissue. While many reports have demonstrated the ability of different bacteria to dysregulate BC's metabolites, the reciprocal effect of these metabolites on the bacterial microbiota has not yet been investigated. Herein, we assess the effect of conditioned media (CM) from a triple-negative BC cell line (MDA-MB-231) on the metabolic profile of Pseudomonas aeruginosa (P. aeruginosa), an important breast resident Gram-negative bacteria that influence oncogenesis. Optical density and scanning electron microscopes were used to assess the impact of MDA-MB-231-CM (BC-CM) on P. aeruginosa growth and morphological changes, respectively. In addition, liquid chromatography-high-resolution mass spectrometry was used to identify metabolic changes in P. aeruginosa and their secretomes in response to the BC-CM. The BC-CM significantly suppressed the growth of P. aeruginosa in the log phase and induced concentration-dependent cytopathological changes in their cell walls. The metabolites of P. aeruginosa were dysregulated considerably depending on the time of exposure to the BC-CM. When treated with the BC-CM, P. aeruginosa induced the purine alkaloid spliceostatin (FR901464), a prominent antitumor metabolite. The BC-CM also promoted other P. aeruginosa metabolites such as amino acids, phosphoribosyl-AMP, 2-aminoacetophenone, pyochelin I, guanosine monophosphate, riboflavin, and terpenoids, which are capable of interfering with oncogenesis. Nine of the significantly identified metabolites from the 0-3 h comparison and four of those identified from the 0-6 h comparison have potential roles in influencing cancer cell behavior. Our findings demonstrate the ability of triple-negative BC-CM not only to alter the growth and morphology of P. aeruginosa but also to modulate their metabolic profile. A better understanding of the influence of BC on certain resident breast microbiomes, such as P. aeruginosa, may open a new therapeutic intervention opportunity for the treatment of cancer.
Additional Links: PMID-40507816
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40507816,
year = {2025},
author = {AlDawsari, M and Al-Ansari, MM and AlMalki, RH and Rahman, AMA and Al-Alwan, M},
title = {The MDA-MB-231 Breast Cancer Cell Secretomes Modify Metabolomes of Pseudomonas aeruginosa Breast Microbiome.},
journal = {International journal of molecular sciences},
volume = {26},
number = {11},
pages = {},
doi = {10.3390/ijms26115003},
pmid = {40507816},
issn = {1422-0067},
support = {RAC#2240005 and 2220015//King Faisal Specialist Hospital & Research Centre/ ; },
mesh = {Humans ; *Pseudomonas aeruginosa/metabolism/drug effects/growth & development ; Female ; Cell Line, Tumor ; *Metabolome/drug effects ; *Breast Neoplasms/microbiology/metabolism ; *Microbiota ; Culture Media, Conditioned/pharmacology ; *Breast/microbiology ; },
abstract = {Breast cancer (BC) is globally becoming a great challenge, being both the most diagnosed cancer and the leading cause of death in women. In addition to cancer cells, many bacteria co-inhabit BC, which differ in type and number from the resident microbiota found in healthy breast tissue. While many reports have demonstrated the ability of different bacteria to dysregulate BC's metabolites, the reciprocal effect of these metabolites on the bacterial microbiota has not yet been investigated. Herein, we assess the effect of conditioned media (CM) from a triple-negative BC cell line (MDA-MB-231) on the metabolic profile of Pseudomonas aeruginosa (P. aeruginosa), an important breast resident Gram-negative bacteria that influence oncogenesis. Optical density and scanning electron microscopes were used to assess the impact of MDA-MB-231-CM (BC-CM) on P. aeruginosa growth and morphological changes, respectively. In addition, liquid chromatography-high-resolution mass spectrometry was used to identify metabolic changes in P. aeruginosa and their secretomes in response to the BC-CM. The BC-CM significantly suppressed the growth of P. aeruginosa in the log phase and induced concentration-dependent cytopathological changes in their cell walls. The metabolites of P. aeruginosa were dysregulated considerably depending on the time of exposure to the BC-CM. When treated with the BC-CM, P. aeruginosa induced the purine alkaloid spliceostatin (FR901464), a prominent antitumor metabolite. The BC-CM also promoted other P. aeruginosa metabolites such as amino acids, phosphoribosyl-AMP, 2-aminoacetophenone, pyochelin I, guanosine monophosphate, riboflavin, and terpenoids, which are capable of interfering with oncogenesis. Nine of the significantly identified metabolites from the 0-3 h comparison and four of those identified from the 0-6 h comparison have potential roles in influencing cancer cell behavior. Our findings demonstrate the ability of triple-negative BC-CM not only to alter the growth and morphology of P. aeruginosa but also to modulate their metabolic profile. A better understanding of the influence of BC on certain resident breast microbiomes, such as P. aeruginosa, may open a new therapeutic intervention opportunity for the treatment of cancer.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
*Pseudomonas aeruginosa/metabolism/drug effects/growth & development
Female
Cell Line, Tumor
*Metabolome/drug effects
*Breast Neoplasms/microbiology/metabolism
*Microbiota
Culture Media, Conditioned/pharmacology
*Breast/microbiology
RevDate: 2025-06-13
CmpDate: 2025-06-13
Biological Actions of Bile Acids via Cell Surface Receptors.
International journal of molecular sciences, 26(11): pii:ijms26115004.
Bile acids (BAs) are synthesized in the liver from cholesterol and are subsequently conjugated with glycine and taurine. In the intestine, bile acids undergo various modifications, such as deconjugation, dehydrogenation, oxidation, and epimerization by the gut microbiota. These bile acids are absorbed in the intestine and transported to the liver as well as the systemic circulation. BAs can activate many types of receptors, including nuclear receptors and cell surface receptors. By activating these receptors, BAs can exert various effects on the metabolic, immune, and nervous systems. Recently, the detailed structure of TGR5, the major plasma membrane receptor for BAs, was elucidated, revealing a putative second BA binding site along with the orthosteric binding site. Furthermore, BAs act as ligands for bitter taste receptors and the Leukemia inhibitory factor receptor. In addition, the Mas-related, G-protein-coupled receptor X4 interacts with receptor activity-modifying proteins. Thus, a variety of cell surface receptors are associated with BAs, and BAs are thought to have very complex activities. This review focuses on recent advances regarding cell surface receptors for bile acids and the biological actions they mediate.
Additional Links: PMID-40507815
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40507815,
year = {2025},
author = {Kiriyama, Y and Tokumaru, H and Sadamoto, H and Nochi, H},
title = {Biological Actions of Bile Acids via Cell Surface Receptors.},
journal = {International journal of molecular sciences},
volume = {26},
number = {11},
pages = {},
doi = {10.3390/ijms26115004},
pmid = {40507815},
issn = {1422-0067},
mesh = {*Bile Acids and Salts/metabolism/chemistry ; Humans ; Animals ; Receptors, G-Protein-Coupled/metabolism/chemistry ; *Receptors, Cell Surface/metabolism/chemistry ; },
abstract = {Bile acids (BAs) are synthesized in the liver from cholesterol and are subsequently conjugated with glycine and taurine. In the intestine, bile acids undergo various modifications, such as deconjugation, dehydrogenation, oxidation, and epimerization by the gut microbiota. These bile acids are absorbed in the intestine and transported to the liver as well as the systemic circulation. BAs can activate many types of receptors, including nuclear receptors and cell surface receptors. By activating these receptors, BAs can exert various effects on the metabolic, immune, and nervous systems. Recently, the detailed structure of TGR5, the major plasma membrane receptor for BAs, was elucidated, revealing a putative second BA binding site along with the orthosteric binding site. Furthermore, BAs act as ligands for bitter taste receptors and the Leukemia inhibitory factor receptor. In addition, the Mas-related, G-protein-coupled receptor X4 interacts with receptor activity-modifying proteins. Thus, a variety of cell surface receptors are associated with BAs, and BAs are thought to have very complex activities. This review focuses on recent advances regarding cell surface receptors for bile acids and the biological actions they mediate.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
*Bile Acids and Salts/metabolism/chemistry
Humans
Animals
Receptors, G-Protein-Coupled/metabolism/chemistry
*Receptors, Cell Surface/metabolism/chemistry
RevDate: 2025-06-13
CmpDate: 2025-06-13
SkinDuo[TM] as a Targeted Probiotic Therapy: Shifts in Skin Microbiota and Clinical Outcomes in Acne Patients.
International journal of molecular sciences, 26(11): pii:ijms26115000.
Acne vulgaris is a common dermatological condition strongly associated with disruptions in the skin microbiota, specifically involving key species such as Cutibacterium acnes and Staphylococcus epidermidis. This study investigates the efficacy of SkinDuo[TM], a topical probiotic containing Lactiplantibacillus plantarum, in modulating the skin microbiota and improving clinical outcomes in patients with acne vulgaris. Over a 4-week to 8-week observational study period, microbial composition and diversity shifts were analyzed using full-length 16S rRNA sequencing. Patient responses were categorized into "good" responders (showing significant clinical improvement) and "no_change" responders (with minimal or no improvement). SkinDuo[TM] treatment resulted in lower post-treatment Cutibacterium acnes abundance in the "good" group compared to the "no_change" group. The "good" group maintained a stable level of alpha diversity following treatment. In contrast, the "no_change" group exhibited a marked reduction in microbial diversity. Beta diversity analysis revealed distinct clustering patterns associated with improved clinical outcomes. These findings suggest that the preservation of microbial richness and evenness may serve as a potential biomarker for positive response to probiotic therapy. This study highlights the potential of SkinDuo[TM] to restore microbial balance and alleviate acne symptoms, contributing to the growing body of evidence supporting microbiome-based therapeutic strategies in dermatology.
Additional Links: PMID-40507814
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40507814,
year = {2025},
author = {Biazzo, M and Pinzauti, D and Podrini, C},
title = {SkinDuo[TM] as a Targeted Probiotic Therapy: Shifts in Skin Microbiota and Clinical Outcomes in Acne Patients.},
journal = {International journal of molecular sciences},
volume = {26},
number = {11},
pages = {},
doi = {10.3390/ijms26115000},
pmid = {40507814},
issn = {1422-0067},
support = {R&I-2022-019L//Xjenza Malta/ ; },
mesh = {Humans ; *Probiotics/therapeutic use/administration & dosage ; *Acne Vulgaris/microbiology/therapy/drug therapy ; *Skin/microbiology ; *Microbiota/drug effects ; Male ; Female ; Treatment Outcome ; RNA, Ribosomal, 16S/genetics ; Adult ; Young Adult ; Adolescent ; Propionibacterium acnes ; Skin Microbiome ; },
abstract = {Acne vulgaris is a common dermatological condition strongly associated with disruptions in the skin microbiota, specifically involving key species such as Cutibacterium acnes and Staphylococcus epidermidis. This study investigates the efficacy of SkinDuo[TM], a topical probiotic containing Lactiplantibacillus plantarum, in modulating the skin microbiota and improving clinical outcomes in patients with acne vulgaris. Over a 4-week to 8-week observational study period, microbial composition and diversity shifts were analyzed using full-length 16S rRNA sequencing. Patient responses were categorized into "good" responders (showing significant clinical improvement) and "no_change" responders (with minimal or no improvement). SkinDuo[TM] treatment resulted in lower post-treatment Cutibacterium acnes abundance in the "good" group compared to the "no_change" group. The "good" group maintained a stable level of alpha diversity following treatment. In contrast, the "no_change" group exhibited a marked reduction in microbial diversity. Beta diversity analysis revealed distinct clustering patterns associated with improved clinical outcomes. These findings suggest that the preservation of microbial richness and evenness may serve as a potential biomarker for positive response to probiotic therapy. This study highlights the potential of SkinDuo[TM] to restore microbial balance and alleviate acne symptoms, contributing to the growing body of evidence supporting microbiome-based therapeutic strategies in dermatology.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
*Probiotics/therapeutic use/administration & dosage
*Acne Vulgaris/microbiology/therapy/drug therapy
*Skin/microbiology
*Microbiota/drug effects
Male
Female
Treatment Outcome
RNA, Ribosomal, 16S/genetics
Adult
Young Adult
Adolescent
Propionibacterium acnes
Skin Microbiome
RevDate: 2025-06-13
Corporal Composition and Gut Microbiome Modification Through Exclusion Dietary Intervention in Adult Patients with Crohn's Disease: Protocol for a Prospective, Interventional, Controlled, Randomized Clinical Trial.
Journal of clinical medicine, 14(11): pii:jcm14113998.
Background: Crohn's disease (CD) is an inflammatory bowel disease in which there is an alteration in the homeostasis and functionality of the intestinal mucosa accompanied by a dysbiosis of the commensal microbiota. The analysis of different dietary strategies to achieve CD remission and reduce gastrointestinal symptoms concludes that it is necessary to restrict the intake of ultra-processed products and to promote the consumption of those with anti-inflammatory effects that improve intestinal permeability and dysbiosis. Methods: Based on previous studies conducted in other cohorts, mainly pediatric, we propose an experimental, prospective, randomized study in patients with active CD who do not show improvement with conventional pharmacological treatment. The control group will receive standard nutritional recommendations while the intervention group will be prescribed an exclusion diet supplemented with enteral nutrition. Results: Patients in the intervention group are expected to exhibit increased lean body mass and reduced visceral fat, as measured by bioelectrical impedance analysis (BIA), alongside higher rates of clinical remission (CDAI), decreased inflammatory markers, and improved gut microbiota composition. Additionally, improvements in health-related quality of life are anticipated, as assessed by validated questionnaires. Conclusions: In the present project, we plan to conduct a detailed study to determine the potential of the exclusion diet for the treatment and remission of CD in adult patients, with the hypothesis that this nutritional intervention will be able to modify and improve intestinal dysbiosis, inflammatory status, and clinical and body composition markers in these patients.
Additional Links: PMID-40507757
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40507757,
year = {2025},
author = {Cano-Mármol, RP and Fernández-Ruiz, VE and Martínez-Pascual, C and Ros-Madrid, I and Martín-Pozuelo, G and Oliva-Bolarín, A and Martínez-Sánchez, MA and Egea-Valenzuela, J and Núñez-Sánchez, MÁ and Ramos-Molina, B and Ruiz-Alcaraz, AJ and Ferrer-Gómez, M},
title = {Corporal Composition and Gut Microbiome Modification Through Exclusion Dietary Intervention in Adult Patients with Crohn's Disease: Protocol for a Prospective, Interventional, Controlled, Randomized Clinical Trial.},
journal = {Journal of clinical medicine},
volume = {14},
number = {11},
pages = {},
doi = {10.3390/jcm14113998},
pmid = {40507757},
issn = {2077-0383},
abstract = {Background: Crohn's disease (CD) is an inflammatory bowel disease in which there is an alteration in the homeostasis and functionality of the intestinal mucosa accompanied by a dysbiosis of the commensal microbiota. The analysis of different dietary strategies to achieve CD remission and reduce gastrointestinal symptoms concludes that it is necessary to restrict the intake of ultra-processed products and to promote the consumption of those with anti-inflammatory effects that improve intestinal permeability and dysbiosis. Methods: Based on previous studies conducted in other cohorts, mainly pediatric, we propose an experimental, prospective, randomized study in patients with active CD who do not show improvement with conventional pharmacological treatment. The control group will receive standard nutritional recommendations while the intervention group will be prescribed an exclusion diet supplemented with enteral nutrition. Results: Patients in the intervention group are expected to exhibit increased lean body mass and reduced visceral fat, as measured by bioelectrical impedance analysis (BIA), alongside higher rates of clinical remission (CDAI), decreased inflammatory markers, and improved gut microbiota composition. Additionally, improvements in health-related quality of life are anticipated, as assessed by validated questionnaires. Conclusions: In the present project, we plan to conduct a detailed study to determine the potential of the exclusion diet for the treatment and remission of CD in adult patients, with the hypothesis that this nutritional intervention will be able to modify and improve intestinal dysbiosis, inflammatory status, and clinical and body composition markers in these patients.},
}
RevDate: 2025-06-13
A Review of the Influence of Prebiotics, Probiotics, Synbiotics, and Postbiotics on the Human Gut Microbiome and Intestinal Integrity.
Journal of clinical medicine, 14(11): pii:jcm14113673.
OBJECTIVE: This review aims to comprehensively evaluate the current evidence on the role of prebiotics, probiotics, synbiotics, and postbiotics-collectively referred to as "biotics"-in modulating the human gut microbiota and enhancing intestinal epithelial integrity.
FINDINGS: Biotics exert their beneficial effects through several mechanisms, including by promoting the growth of beneficial microbes, producing short-chain fatty acids (SCFAs), strengthening the gut barrier, and regulating immune responses. Prebiotics selectively stimulate beneficial bacteria, probiotics introduce live microorganisms with therapeutic functions, synbiotics combine the strengths of both, and postbiotics offer non-viable microbial components and metabolites that mimic probiotic benefits with enhanced safety profiles. Each type of biotic demonstrates unique and complementary effects across a range of conditions, such as inflammatory bowel disease, irritable bowel syndrome, obesity, constipation, and antibiotic-associated diarrhea.
IMPLICATIONS: As disruptions in the gut microbiota and intestinal barrier are increasingly linked to chronic and immune-mediated diseases, leveraging biotics offers promising avenues for personalized nutrition, preventive healthcare, and adjunct therapies. The integration of biotics into clinical and dietary strategies may significantly contribute to improving gastrointestinal and systemic health.
Additional Links: PMID-40507435
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40507435,
year = {2025},
author = {Smolinska, S and Popescu, FD and Zemelka-Wiacek, M},
title = {A Review of the Influence of Prebiotics, Probiotics, Synbiotics, and Postbiotics on the Human Gut Microbiome and Intestinal Integrity.},
journal = {Journal of clinical medicine},
volume = {14},
number = {11},
pages = {},
doi = {10.3390/jcm14113673},
pmid = {40507435},
issn = {2077-0383},
abstract = {OBJECTIVE: This review aims to comprehensively evaluate the current evidence on the role of prebiotics, probiotics, synbiotics, and postbiotics-collectively referred to as "biotics"-in modulating the human gut microbiota and enhancing intestinal epithelial integrity.
FINDINGS: Biotics exert their beneficial effects through several mechanisms, including by promoting the growth of beneficial microbes, producing short-chain fatty acids (SCFAs), strengthening the gut barrier, and regulating immune responses. Prebiotics selectively stimulate beneficial bacteria, probiotics introduce live microorganisms with therapeutic functions, synbiotics combine the strengths of both, and postbiotics offer non-viable microbial components and metabolites that mimic probiotic benefits with enhanced safety profiles. Each type of biotic demonstrates unique and complementary effects across a range of conditions, such as inflammatory bowel disease, irritable bowel syndrome, obesity, constipation, and antibiotic-associated diarrhea.
IMPLICATIONS: As disruptions in the gut microbiota and intestinal barrier are increasingly linked to chronic and immune-mediated diseases, leveraging biotics offers promising avenues for personalized nutrition, preventive healthcare, and adjunct therapies. The integration of biotics into clinical and dietary strategies may significantly contribute to improving gastrointestinal and systemic health.},
}
RevDate: 2025-06-13
Impaired Overall Survival of Melanoma Patients Due to Antibiotic Use Prior to Immune Checkpoint Inhibitor Therapy: Systematic Review and Meta-Analysis.
Cancers, 17(11): pii:cancers17111872.
Background: The gut microbiome plays a pivotal role in shaping systemic immunity and modulating anti-tumor responses. Preclinical and clinical studies have shown that higher gut microbial diversity and the presence of specific commensal taxa correlate with improved responses to immune checkpoint inhibitors (ICI) in melanoma. Conversely, broad-spectrum antibiotics can induce dysbiosis, reducing T cell activation and cytokine production, and have been linked to diminished ICI efficacy in several cancer types. Methods: We conducted a systematic review and meta-analysis of seven retrospective cohorts (total n = 5213) comparing overall survival in cutaneous melanoma (CM) patients who did or did not receive systemic antibiotics within six weeks before ICI initiation. From each study, we extracted hazard ratios (HRs) for death, antibiotic-to-ICI interval, ICI regimen (PD-1 monotherapy vs. PD-1 + CTLA-4 combination), cohort size, and country. Pooled log-HRs were estimated under fixed-effect and random-effects (REML) models. Statistical heterogeneity was quantified by Cochran's Q and I[2] statistics, and τ[2]. We performed leave-one-out sensitivity analyses, generated a Baujat plot to identify influential studies, applied trim-and-fill to assess publication bias, and ran meta-regressions for regimen, antibiotic timing, sample size, and geography. Results: Under the fixed-effect model, antibiotic exposure corresponded to a pooled HR of 1.26 (95% CI 1.13-1.41; p < 0.001). The random-effects model yielded a pooled HR of 1.55 (95% CI 1.21-1.98; p = 0.0005) with substantial heterogeneity (Q = 25.1; I[2] = 76%). Prediction intervals (0.78-3.06) underscored between-study variability. Leave-one-out analyses produced HRs from 1.50 to 1.75, confirming robustness, and the Baujat plot highlighted two cohorts as primary heterogeneity drivers. Trim-and-fill adjusted the HR to 1.46 (95% CI 1.08-1.97). In subgroup analyses, combination therapy studies (k = 4) showed a pooled HR of ~1.9 (I[2] = 58%) versus ~1.3 (I[2] = 79%) for monotherapy. Meta-regression attributed the largest variance to the regimen (R[2] = 32%; β(monotherapy) = -0.35; p = 0.13). Conclusions: Pre-ICI antibiotic use in CM is consistently associated with a 26-55% increase in mortality risk, particularly with PD-1 + CTLA-4 combinations, reinforcing the mechanistic link between microbiome integrity and ICI success. Looking ahead, integrating prospective microbiome profiling into clinical trials will be critical to personalize ICI therapy, clarify causality, and identify microbial biomarkers for optimal treatment selection. Prospective, microbiome-integrated trials promise to refine melanoma immunotherapy by tailoring antibiotic stewardship and microbial interventions to enhance patient outcomes.
Additional Links: PMID-40507352
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40507352,
year = {2025},
author = {Gambichler, T and Weyer-Fahlbusch, SS and Overbeck, J and Abu Rached, N and Becker, JC and Susok, L},
title = {Impaired Overall Survival of Melanoma Patients Due to Antibiotic Use Prior to Immune Checkpoint Inhibitor Therapy: Systematic Review and Meta-Analysis.},
journal = {Cancers},
volume = {17},
number = {11},
pages = {},
doi = {10.3390/cancers17111872},
pmid = {40507352},
issn = {2072-6694},
abstract = {Background: The gut microbiome plays a pivotal role in shaping systemic immunity and modulating anti-tumor responses. Preclinical and clinical studies have shown that higher gut microbial diversity and the presence of specific commensal taxa correlate with improved responses to immune checkpoint inhibitors (ICI) in melanoma. Conversely, broad-spectrum antibiotics can induce dysbiosis, reducing T cell activation and cytokine production, and have been linked to diminished ICI efficacy in several cancer types. Methods: We conducted a systematic review and meta-analysis of seven retrospective cohorts (total n = 5213) comparing overall survival in cutaneous melanoma (CM) patients who did or did not receive systemic antibiotics within six weeks before ICI initiation. From each study, we extracted hazard ratios (HRs) for death, antibiotic-to-ICI interval, ICI regimen (PD-1 monotherapy vs. PD-1 + CTLA-4 combination), cohort size, and country. Pooled log-HRs were estimated under fixed-effect and random-effects (REML) models. Statistical heterogeneity was quantified by Cochran's Q and I[2] statistics, and τ[2]. We performed leave-one-out sensitivity analyses, generated a Baujat plot to identify influential studies, applied trim-and-fill to assess publication bias, and ran meta-regressions for regimen, antibiotic timing, sample size, and geography. Results: Under the fixed-effect model, antibiotic exposure corresponded to a pooled HR of 1.26 (95% CI 1.13-1.41; p < 0.001). The random-effects model yielded a pooled HR of 1.55 (95% CI 1.21-1.98; p = 0.0005) with substantial heterogeneity (Q = 25.1; I[2] = 76%). Prediction intervals (0.78-3.06) underscored between-study variability. Leave-one-out analyses produced HRs from 1.50 to 1.75, confirming robustness, and the Baujat plot highlighted two cohorts as primary heterogeneity drivers. Trim-and-fill adjusted the HR to 1.46 (95% CI 1.08-1.97). In subgroup analyses, combination therapy studies (k = 4) showed a pooled HR of ~1.9 (I[2] = 58%) versus ~1.3 (I[2] = 79%) for monotherapy. Meta-regression attributed the largest variance to the regimen (R[2] = 32%; β(monotherapy) = -0.35; p = 0.13). Conclusions: Pre-ICI antibiotic use in CM is consistently associated with a 26-55% increase in mortality risk, particularly with PD-1 + CTLA-4 combinations, reinforcing the mechanistic link between microbiome integrity and ICI success. Looking ahead, integrating prospective microbiome profiling into clinical trials will be critical to personalize ICI therapy, clarify causality, and identify microbial biomarkers for optimal treatment selection. Prospective, microbiome-integrated trials promise to refine melanoma immunotherapy by tailoring antibiotic stewardship and microbial interventions to enhance patient outcomes.},
}
RevDate: 2025-06-13
The Knowledge Gap in Gut Microbiome Characterization in Early-Onset Colorectal Cancer Patients: A Systematic Scoping Review.
Cancers, 17(11): pii:cancers17111863.
BACKGROUND/OBJECTIVES: Over the past two decades, the incidence of early-onset colorectal cancer (EoCRC) has been increasing, although its underlying causes remain unclear. Gut microbiome is known to play a role in carcinogenesis of colorectal cancer. This scoping review aims to systematically map and synthetize current evidence on gut microbiome characterization in EoCRC (vs. late-onset colorectal cancer (LoCRC) and healthy individuals), describe the methodology used, and identify knowledge gaps to inform and guide future research.
METHODS: This systematic scoping review followed the Joanna Briggs Institute (JBI) methodology for scoping reviews. Searches were conducted in PubMed, Web of Science, and Scopus between January and February 2025. Two reviewers independently screened and selected the studies. One reviewer extracted the relevant information, using an adapted version of the JBI template.
RESULTS: Seven studies met eligibility criteria. Compared to healthy young adults, EoCRC patients had a predominance of lower α diversity, different β diversity, and greater abundance of Flavonifractor plautii, Akkermansia muciniphila, Bacteroides, and Fusobacteria. Comparisons with LoCRC showed that EoCRC had distinct β diversity and a higher abundance in Fusobacterium, Akkermansia, Bacteroides, and Actinomyces. Only three studies correlated the microbiota composition of EoCRC with clinicopathology features and suggested positive associations between Fusobacterium abundance, rectal tumors and lower survival and Akkermansia abundance with body mass index (BMI) ≥ 25 kg/m[2], rectal EoCRC, and better survival.
CONCLUSIONS: There is a lack of large, methodologically robust studies linking gut microbiota with clinicopathological, lifestyle, and tumor molecular features in EoCRC. Our review highlights critical knowledge gaps, the need for standardized methodologies, and key areas for future investigation.
Additional Links: PMID-40507344
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40507344,
year = {2025},
author = {Gomes de Sousa, R and Guerreiro, CS and Santos, I and Cravo, M},
title = {The Knowledge Gap in Gut Microbiome Characterization in Early-Onset Colorectal Cancer Patients: A Systematic Scoping Review.},
journal = {Cancers},
volume = {17},
number = {11},
pages = {},
doi = {10.3390/cancers17111863},
pmid = {40507344},
issn = {2072-6694},
abstract = {BACKGROUND/OBJECTIVES: Over the past two decades, the incidence of early-onset colorectal cancer (EoCRC) has been increasing, although its underlying causes remain unclear. Gut microbiome is known to play a role in carcinogenesis of colorectal cancer. This scoping review aims to systematically map and synthetize current evidence on gut microbiome characterization in EoCRC (vs. late-onset colorectal cancer (LoCRC) and healthy individuals), describe the methodology used, and identify knowledge gaps to inform and guide future research.
METHODS: This systematic scoping review followed the Joanna Briggs Institute (JBI) methodology for scoping reviews. Searches were conducted in PubMed, Web of Science, and Scopus between January and February 2025. Two reviewers independently screened and selected the studies. One reviewer extracted the relevant information, using an adapted version of the JBI template.
RESULTS: Seven studies met eligibility criteria. Compared to healthy young adults, EoCRC patients had a predominance of lower α diversity, different β diversity, and greater abundance of Flavonifractor plautii, Akkermansia muciniphila, Bacteroides, and Fusobacteria. Comparisons with LoCRC showed that EoCRC had distinct β diversity and a higher abundance in Fusobacterium, Akkermansia, Bacteroides, and Actinomyces. Only three studies correlated the microbiota composition of EoCRC with clinicopathology features and suggested positive associations between Fusobacterium abundance, rectal tumors and lower survival and Akkermansia abundance with body mass index (BMI) ≥ 25 kg/m[2], rectal EoCRC, and better survival.
CONCLUSIONS: There is a lack of large, methodologically robust studies linking gut microbiota with clinicopathological, lifestyle, and tumor molecular features in EoCRC. Our review highlights critical knowledge gaps, the need for standardized methodologies, and key areas for future investigation.},
}
RevDate: 2025-06-13
CmpDate: 2025-06-13
Function of Yogurt Fermented with the Lactococcus lactis 11/19-B1 Strain in Improving the Lipid Profile and Intestinal Microbiome in Hemodialysis Patients.
Nutrients, 17(11): pii:nu17111931.
BACKGROUND/OBJECTIVES: The number of chronic kidney disease (CKD) patients is increasing in Japan, and this population is at high risk of death from cardiovascular and cerebrovascular diseases. Therefore, prevention of arteriosclerosis as a common underlying cause of these diseases is required. In this study, we examined whether 11/19-B1 yogurt, which has been proven to reduce serum low-density lipoprotein (LDL) levels, can decrease the serum levels of indoxylsulfate and trimethylamine-N-oxide (TMAO), which are produced by intestinal microbiota and known to cause arteriosclerosis, through improving dysbiosis in hemodialysis patients.
METHODS: Nineteen dialysis patients consumed 50 g of 11/19-B1 yogurt daily for 8 weeks, and changes in serum lipid profile and uremic toxin levels, intestinal microbiome, as well as the frequency of bowel movement and stool characteristics were observed.
RESULTS: The results demonstrated that an intake of yogurt decreased serum LDL 99.3 to 88.5 (p = 0.049) and indoxylsulfate in seven of nine subjects with previously high concentrations, and improved stool characteristics as estimated by the Bristle stool score, although decreased HDL and no beneficial effect on serum TMAO was observed.
CONCLUSIONS: These results may suggest that the ingestion of 11/19-B1 yogurt provides a preventative effect against the progression of atherosclerosis and renal dysfunction.
Additional Links: PMID-40507200
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40507200,
year = {2025},
author = {Suzuki, Y and Ishioka, K and Nakamura, T and Miyazaki, N and Marubashi, S and Suzutani, T},
title = {Function of Yogurt Fermented with the Lactococcus lactis 11/19-B1 Strain in Improving the Lipid Profile and Intestinal Microbiome in Hemodialysis Patients.},
journal = {Nutrients},
volume = {17},
number = {11},
pages = {},
doi = {10.3390/nu17111931},
pmid = {40507200},
issn = {2072-6643},
mesh = {Humans ; *Yogurt/microbiology ; *Gastrointestinal Microbiome ; *Renal Dialysis ; Male ; Female ; Middle Aged ; Aged ; *Renal Insufficiency, Chronic/therapy/blood ; *Fermentation ; *Lipids/blood ; Indican/blood ; Methylamines/blood ; Dysbiosis ; Probiotics ; },
abstract = {BACKGROUND/OBJECTIVES: The number of chronic kidney disease (CKD) patients is increasing in Japan, and this population is at high risk of death from cardiovascular and cerebrovascular diseases. Therefore, prevention of arteriosclerosis as a common underlying cause of these diseases is required. In this study, we examined whether 11/19-B1 yogurt, which has been proven to reduce serum low-density lipoprotein (LDL) levels, can decrease the serum levels of indoxylsulfate and trimethylamine-N-oxide (TMAO), which are produced by intestinal microbiota and known to cause arteriosclerosis, through improving dysbiosis in hemodialysis patients.
METHODS: Nineteen dialysis patients consumed 50 g of 11/19-B1 yogurt daily for 8 weeks, and changes in serum lipid profile and uremic toxin levels, intestinal microbiome, as well as the frequency of bowel movement and stool characteristics were observed.
RESULTS: The results demonstrated that an intake of yogurt decreased serum LDL 99.3 to 88.5 (p = 0.049) and indoxylsulfate in seven of nine subjects with previously high concentrations, and improved stool characteristics as estimated by the Bristle stool score, although decreased HDL and no beneficial effect on serum TMAO was observed.
CONCLUSIONS: These results may suggest that the ingestion of 11/19-B1 yogurt provides a preventative effect against the progression of atherosclerosis and renal dysfunction.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
*Yogurt/microbiology
*Gastrointestinal Microbiome
*Renal Dialysis
Male
Female
Middle Aged
Aged
*Renal Insufficiency, Chronic/therapy/blood
*Fermentation
*Lipids/blood
Indican/blood
Methylamines/blood
Dysbiosis
Probiotics
RevDate: 2025-06-13
CmpDate: 2025-06-13
Potential Therapeutic Targets for Androgenetic Alopecia (AGA) in Obese Individuals as Revealed by a Gut Microbiome Analysis: A Mendelian Randomization Study.
Nutrients, 17(11): pii:nu17111892.
Objective: This study aimed to investigate the role of the gut microbiome in androgenetic alopecia (AGA) among obese individuals using Mendelian randomization (MR), and to identify potential therapeutic targets for mitigating AGA in this population. Methods: Genomic data for 412 gut microbiomes, AGA, and obesity were obtained from genome-wide association studies (GWAS). Bidirectional MR was performed using inverse variance weighted (IVW) as the primary analysis method, complemented by sensitivity analyses. Potential therapeutic targets within the gut microbiome associated with AGA in obese individuals were identified. Results: Two gut microbiomes were identified as having a significant impact on obese individuals with AGA. Specifically, the abundance of the sulfoglycolysis pathway in gut bacteria was found to significantly increase the risk of both obesity and AGA. In contrast, the abundance of the de novo biosynthesis of the adenosine ribonucleotide pathway in gut bacteria was associated with a significant increase in the risk of obesity but a significant decrease in the risk of AGA. Conclusions: The abundance of gut bacterial pathways, including sulfoglycolysis and the de novo biosynthesis of adenosine ribonucleotides, can serve as potential therapeutic targets for managing obesity-associated AGA. These findings offer a novel research direction for the development of innovative diagnostic and treatment strategies for patients with obesity and AGA.
Additional Links: PMID-40507161
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40507161,
year = {2025},
author = {Li, Y and Liao, X and Tang, S and Wang, Q and Lin, H and Yu, X and Xiao, Y and Tao, X and Zhong, T},
title = {Potential Therapeutic Targets for Androgenetic Alopecia (AGA) in Obese Individuals as Revealed by a Gut Microbiome Analysis: A Mendelian Randomization Study.},
journal = {Nutrients},
volume = {17},
number = {11},
pages = {},
doi = {10.3390/nu17111892},
pmid = {40507161},
issn = {2072-6643},
mesh = {Humans ; *Gastrointestinal Microbiome/genetics ; *Alopecia/microbiology/genetics/therapy ; Mendelian Randomization Analysis ; *Obesity/complications/microbiology/genetics ; Genome-Wide Association Study ; Male ; Female ; Adult ; },
abstract = {Objective: This study aimed to investigate the role of the gut microbiome in androgenetic alopecia (AGA) among obese individuals using Mendelian randomization (MR), and to identify potential therapeutic targets for mitigating AGA in this population. Methods: Genomic data for 412 gut microbiomes, AGA, and obesity were obtained from genome-wide association studies (GWAS). Bidirectional MR was performed using inverse variance weighted (IVW) as the primary analysis method, complemented by sensitivity analyses. Potential therapeutic targets within the gut microbiome associated with AGA in obese individuals were identified. Results: Two gut microbiomes were identified as having a significant impact on obese individuals with AGA. Specifically, the abundance of the sulfoglycolysis pathway in gut bacteria was found to significantly increase the risk of both obesity and AGA. In contrast, the abundance of the de novo biosynthesis of the adenosine ribonucleotide pathway in gut bacteria was associated with a significant increase in the risk of obesity but a significant decrease in the risk of AGA. Conclusions: The abundance of gut bacterial pathways, including sulfoglycolysis and the de novo biosynthesis of adenosine ribonucleotides, can serve as potential therapeutic targets for managing obesity-associated AGA. These findings offer a novel research direction for the development of innovative diagnostic and treatment strategies for patients with obesity and AGA.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
*Gastrointestinal Microbiome/genetics
*Alopecia/microbiology/genetics/therapy
Mendelian Randomization Analysis
*Obesity/complications/microbiology/genetics
Genome-Wide Association Study
Male
Female
Adult
RevDate: 2025-06-13
CmpDate: 2025-06-13
The Influence of Dietary Factors on Melanoma Development and Progression: A Comprehensive Review.
Nutrients, 17(11): pii:nu17111891.
Melanoma is an aggressive cutaneous malignancy with increasing global incidence and high metastatic potential. While ultraviolet (UV) radiation remains the primary environmental risk factor, emerging evidence suggests that dietary factors may influence melanoma risk, progression, and treatment outcomes. This comprehensive review examines the impact of dietary components, including fats, vitamins, minerals, antioxidants, bioactive compounds, and the gut microbiome, on melanoma pathogenesis. The current literature indicates that diets rich in polyunsaturated fatty acids (PUFAs), antioxidants, and plant-based bioactive compounds may confer protective effects against melanoma by modulating oxidative stress, inflammation, and immune response. Additionally, the gut microbiome plays a critical role in melanoma progression and immunotherapy response, with dietary patterns influencing microbial composition and, consequently, host immunity. Despite these promising associations, research remains limited, and findings across studies are inconsistent, preventing the establishment of definitive dietary guidelines for melanoma prevention and management. Future research should focus on large-scale prospective studies to elucidate the mechanisms underlying the dietary influences on melanoma and determine evidence-based nutritional strategies. Understanding the interplay between diet, immune modulation, and gut microbiome composition represents a promising avenue for advancing melanoma prevention and treatment strategies.
Additional Links: PMID-40507159
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40507159,
year = {2025},
author = {Watson, AE and Yusuf, N},
title = {The Influence of Dietary Factors on Melanoma Development and Progression: A Comprehensive Review.},
journal = {Nutrients},
volume = {17},
number = {11},
pages = {},
doi = {10.3390/nu17111891},
pmid = {40507159},
issn = {2072-6643},
mesh = {Humans ; *Melanoma/etiology/prevention & control/pathology ; Gastrointestinal Microbiome ; *Diet/adverse effects ; Disease Progression ; *Skin Neoplasms/etiology/prevention & control/pathology ; Antioxidants/administration & dosage ; Risk Factors ; Oxidative Stress ; },
abstract = {Melanoma is an aggressive cutaneous malignancy with increasing global incidence and high metastatic potential. While ultraviolet (UV) radiation remains the primary environmental risk factor, emerging evidence suggests that dietary factors may influence melanoma risk, progression, and treatment outcomes. This comprehensive review examines the impact of dietary components, including fats, vitamins, minerals, antioxidants, bioactive compounds, and the gut microbiome, on melanoma pathogenesis. The current literature indicates that diets rich in polyunsaturated fatty acids (PUFAs), antioxidants, and plant-based bioactive compounds may confer protective effects against melanoma by modulating oxidative stress, inflammation, and immune response. Additionally, the gut microbiome plays a critical role in melanoma progression and immunotherapy response, with dietary patterns influencing microbial composition and, consequently, host immunity. Despite these promising associations, research remains limited, and findings across studies are inconsistent, preventing the establishment of definitive dietary guidelines for melanoma prevention and management. Future research should focus on large-scale prospective studies to elucidate the mechanisms underlying the dietary influences on melanoma and determine evidence-based nutritional strategies. Understanding the interplay between diet, immune modulation, and gut microbiome composition represents a promising avenue for advancing melanoma prevention and treatment strategies.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
*Melanoma/etiology/prevention & control/pathology
Gastrointestinal Microbiome
*Diet/adverse effects
Disease Progression
*Skin Neoplasms/etiology/prevention & control/pathology
Antioxidants/administration & dosage
Risk Factors
Oxidative Stress
RevDate: 2025-06-13
CmpDate: 2025-06-13
In Search of Healthy Ageing: A Microbiome-Based Precision Nutrition Approach for Type 2 Diabetes Prevention.
Nutrients, 17(11): pii:nu17111877.
Background/Objectives: Type 2 diabetes (T2D) is a leading cause of morbidity and mortality worldwide and in Spain, particularly in the elderly population, affecting healthy ageing. Nutritional strategies are key to its prevention. The gut microbiota is also implicated in T2D and can be modulated by nutrition. We hypothesize that precision nutrition through microbiota modulation may help prevent T2D. This article aims to (1) describe a gut microbiota bacterial profile associated with T2D prevention, (2) provide precision nutrition tools to optimize this profile, (3) analyze how overweight influences the microbiota composition and precision nutrition response, and (4) address the technical challenges of microbiome-based precision nutrition clinical implementation to prevent T2D. Methods: A review of gut microbiota associated with T2D prevention was conducted. 13 healthy Spanish participants over 62 with optimal blood glucose levels (7 normal weight and 6 overweight) underwent a 3-month precision nutrition intervention to optimize T2D-preventive gut microbiota using a bioinformatics food recommendation system, Phymofood (EP22382095). Fecal microbiota was analyzed pre- and post-intervention using full-length 16S rRNA gene amplification, MinION sequencing, and NCBI taxonomic classification. Results: 31 potentially preventive bacteria against T2D were selected. The intervention increased the relative abundance of beneficial genera (Butyrivibrio and Faecalibacterium) and species (Eshraghiella crossota, and Faecalibacterium prausnitzii). The overweight influenced microbiota composition and intervention response. Conclusions: A gut microbiota profile associated with T2D prevention was identified, and precision nutrition could increase the relative abundance of beneficial bacteria. Confounding factors such as overweight should be considered when designing microbiome-based precision nutrition interventions. These results contribute to a better understanding of the microbiota associated with T2D prevention and address technical challenges for clinical implementation in future healthy ageing strategies.
Additional Links: PMID-40507144
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40507144,
year = {2025},
author = {González, A and Fullaondo, A and Odriozola, A},
title = {In Search of Healthy Ageing: A Microbiome-Based Precision Nutrition Approach for Type 2 Diabetes Prevention.},
journal = {Nutrients},
volume = {17},
number = {11},
pages = {},
doi = {10.3390/nu17111877},
pmid = {40507144},
issn = {2072-6643},
support = {IT1547-22//Basque Government, Department of Education/ ; },
mesh = {Humans ; *Diabetes Mellitus, Type 2/prevention & control/microbiology ; *Gastrointestinal Microbiome/physiology ; Aged ; Female ; Middle Aged ; Male ; *Healthy Aging ; *Precision Medicine/methods ; Spain ; Feces/microbiology ; Overweight/microbiology ; },
abstract = {Background/Objectives: Type 2 diabetes (T2D) is a leading cause of morbidity and mortality worldwide and in Spain, particularly in the elderly population, affecting healthy ageing. Nutritional strategies are key to its prevention. The gut microbiota is also implicated in T2D and can be modulated by nutrition. We hypothesize that precision nutrition through microbiota modulation may help prevent T2D. This article aims to (1) describe a gut microbiota bacterial profile associated with T2D prevention, (2) provide precision nutrition tools to optimize this profile, (3) analyze how overweight influences the microbiota composition and precision nutrition response, and (4) address the technical challenges of microbiome-based precision nutrition clinical implementation to prevent T2D. Methods: A review of gut microbiota associated with T2D prevention was conducted. 13 healthy Spanish participants over 62 with optimal blood glucose levels (7 normal weight and 6 overweight) underwent a 3-month precision nutrition intervention to optimize T2D-preventive gut microbiota using a bioinformatics food recommendation system, Phymofood (EP22382095). Fecal microbiota was analyzed pre- and post-intervention using full-length 16S rRNA gene amplification, MinION sequencing, and NCBI taxonomic classification. Results: 31 potentially preventive bacteria against T2D were selected. The intervention increased the relative abundance of beneficial genera (Butyrivibrio and Faecalibacterium) and species (Eshraghiella crossota, and Faecalibacterium prausnitzii). The overweight influenced microbiota composition and intervention response. Conclusions: A gut microbiota profile associated with T2D prevention was identified, and precision nutrition could increase the relative abundance of beneficial bacteria. Confounding factors such as overweight should be considered when designing microbiome-based precision nutrition interventions. These results contribute to a better understanding of the microbiota associated with T2D prevention and address technical challenges for clinical implementation in future healthy ageing strategies.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
*Diabetes Mellitus, Type 2/prevention & control/microbiology
*Gastrointestinal Microbiome/physiology
Aged
Female
Middle Aged
Male
*Healthy Aging
*Precision Medicine/methods
Spain
Feces/microbiology
Overweight/microbiology
RevDate: 2025-06-13
CmpDate: 2025-06-13
Cooked Bean (Phaseolus vulgaris L.) Consumption Alters Bile Acid Metabolism in a Mouse Model of Diet-Induced Metabolic Dysfunction: Proof-of-Concept Investigation.
Nutrients, 17(11): pii:nu17111827.
Background/Objectives: Metabolic dysregulation underlies a myriad of chronic diseases, including metabolic dysfunction-associated steatotic liver disease (MASLD) and obesity, and bile acids emerge as an important mediator in their etiology. Weight control by improving diet quality is the standard of care in prevention and control of these metabolic diseases. Inclusion of pulses, such as common bean, is an affordable yet neglected approach to improving diet quality and metabolic outcomes. Thus, this study evaluated the possibility that common bean alters bile acid metabolism in a health-beneficial manner. Methods: Using biospecimens from several similarly designed studies, cecal content, feces, liver tissue, and plasma samples from C57BL/6 mice fed an obesogenic diet lacking (control) or containing cooked common bean were subjected to total bile acid analysis and untargeted metabolomics. RNA-seq, qPCR, and Western blot assays of liver tissue complemented the bile acid analyses. Microbial composition and predicted function in the cecal contents were evaluated using 16S rRNA gene amplicon and shotgun metagenomic sequencing. Results: Bean-fed mice had increased cecal bile acid content and excreted more bile acids per gram of feces. Consistent with these effects, increased synthesis of bile acids in the liver was observed. Microbial composition and capacity to metabolize bile acids were markedly altered by bean, with greater prominence of secondary bile acid metabolites in bean-fed mice, i.e., microbial metabolites of chenodeoxycholate/lithocholate increased while metabolites of hyocholate were reduced. Conclusions: In rendering mice resistant to obesogenic diet-induced MASLD and obesity, cooked bean consumption sequesters bile acids, increasing their hepatic synthesis and enhancing their diversity through microbial metabolism. Bean-induced changes in bile acid metabolism have potential to improve dyslipidemia.
Additional Links: PMID-40507096
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40507096,
year = {2025},
author = {Lutsiv, T and Fitzgerald, VK and Neil, ES and McGinley, JN and Hussan, H and Thompson, HJ},
title = {Cooked Bean (Phaseolus vulgaris L.) Consumption Alters Bile Acid Metabolism in a Mouse Model of Diet-Induced Metabolic Dysfunction: Proof-of-Concept Investigation.},
journal = {Nutrients},
volume = {17},
number = {11},
pages = {},
doi = {10.3390/nu17111827},
pmid = {40507096},
issn = {2072-6643},
support = {58-3060-8-031//USDA ARS/ ; 2020-05206//National Institute for Food and Agriculture:/ ; },
mesh = {Animals ; *Bile Acids and Salts/metabolism ; Mice, Inbred C57BL ; *Phaseolus ; Mice ; Male ; Liver/metabolism ; Disease Models, Animal ; Feces/chemistry ; Cecum/microbiology/metabolism ; Gastrointestinal Microbiome ; Cooking ; Obesity/metabolism ; Metabolomics ; *Diet ; *Metabolic Diseases/etiology/metabolism ; Diet, High-Fat/adverse effects ; },
abstract = {Background/Objectives: Metabolic dysregulation underlies a myriad of chronic diseases, including metabolic dysfunction-associated steatotic liver disease (MASLD) and obesity, and bile acids emerge as an important mediator in their etiology. Weight control by improving diet quality is the standard of care in prevention and control of these metabolic diseases. Inclusion of pulses, such as common bean, is an affordable yet neglected approach to improving diet quality and metabolic outcomes. Thus, this study evaluated the possibility that common bean alters bile acid metabolism in a health-beneficial manner. Methods: Using biospecimens from several similarly designed studies, cecal content, feces, liver tissue, and plasma samples from C57BL/6 mice fed an obesogenic diet lacking (control) or containing cooked common bean were subjected to total bile acid analysis and untargeted metabolomics. RNA-seq, qPCR, and Western blot assays of liver tissue complemented the bile acid analyses. Microbial composition and predicted function in the cecal contents were evaluated using 16S rRNA gene amplicon and shotgun metagenomic sequencing. Results: Bean-fed mice had increased cecal bile acid content and excreted more bile acids per gram of feces. Consistent with these effects, increased synthesis of bile acids in the liver was observed. Microbial composition and capacity to metabolize bile acids were markedly altered by bean, with greater prominence of secondary bile acid metabolites in bean-fed mice, i.e., microbial metabolites of chenodeoxycholate/lithocholate increased while metabolites of hyocholate were reduced. Conclusions: In rendering mice resistant to obesogenic diet-induced MASLD and obesity, cooked bean consumption sequesters bile acids, increasing their hepatic synthesis and enhancing their diversity through microbial metabolism. Bean-induced changes in bile acid metabolism have potential to improve dyslipidemia.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Animals
*Bile Acids and Salts/metabolism
Mice, Inbred C57BL
*Phaseolus
Mice
Male
Liver/metabolism
Disease Models, Animal
Feces/chemistry
Cecum/microbiology/metabolism
Gastrointestinal Microbiome
Cooking
Obesity/metabolism
Metabolomics
*Diet
*Metabolic Diseases/etiology/metabolism
Diet, High-Fat/adverse effects
RevDate: 2025-06-13
CmpDate: 2025-06-13
The Effect of Probiotics on Health in Pregnancy and Infants: A Randomized, Double-Blind, Placebo-Controlled Trial.
Nutrients, 17(11): pii:nu17111825.
Background/Objectives: There is growing interest in the benefits of probiotic supplementation during pregnancy and lactation, but evidence supporting the beneficial effects for mother-infant dyads remains scarce. This study assessed the effects of probiotic supplementation on infection frequency and immunity in pregnant women and infants, and on microbiome establishment during the first month of life. Methods: At 28 weeks of gestation, 180 healthy pregnant women were randomized to receive either a placebo (n = 90) or a probiotic supplement (n = 90), Prenatis™, containing 5 billion CFU/day of Lacticaseibacillus rhamnosus Rosell[®]-11 and Bifidobacterium bifidum HA-132. Results: There was a significantly lower number of women with one or more infections during the study in the probiotics group (8 vs. 18, p = 0.05) and a trend for a lower number of infections during pregnancy (primary outcome) in the probiotics group (p = 0.07). Regarding infants, a lower number of days with infections during the first month of life was observed in the probiotics group (4.7 days on average vs. 10.5 days, p = 0.03). The vaginal microbiota composition during pregnancy and after childbirth showed no significant differences between groups while the infants' gut microbiome demonstrated a significantly higher abundance/prevalence of beneficial taxa in the probiotics group. The benefits conferred by probiotics were especially notable when considering birth by C-section. Probiotics promoted the vertical transmission of beneficial species and the induction of a highly interconnected microbiota, structured around key species. Conclusions: Probiotic supplementation during the third trimester of pregnancy and lactation is a valid strategy for conferring benefits to mothers and infants.
Additional Links: PMID-40507094
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40507094,
year = {2025},
author = {Binda, S and Chow-Shi-Yée, M and El Salti, S and Auclair-Ouellet, N and Oula, ML and Carton, T and Leuillet, S and Tomassi, D and Hemmings, R and Kadoch, IJ},
title = {The Effect of Probiotics on Health in Pregnancy and Infants: A Randomized, Double-Blind, Placebo-Controlled Trial.},
journal = {Nutrients},
volume = {17},
number = {11},
pages = {},
doi = {10.3390/nu17111825},
pmid = {40507094},
issn = {2072-6643},
mesh = {Humans ; Female ; *Probiotics/administration & dosage ; Pregnancy ; Double-Blind Method ; Adult ; Infant, Newborn ; Gastrointestinal Microbiome ; Infant ; Lacticaseibacillus rhamnosus ; Vagina/microbiology ; Dietary Supplements ; Bifidobacterium bifidum ; Young Adult ; },
abstract = {Background/Objectives: There is growing interest in the benefits of probiotic supplementation during pregnancy and lactation, but evidence supporting the beneficial effects for mother-infant dyads remains scarce. This study assessed the effects of probiotic supplementation on infection frequency and immunity in pregnant women and infants, and on microbiome establishment during the first month of life. Methods: At 28 weeks of gestation, 180 healthy pregnant women were randomized to receive either a placebo (n = 90) or a probiotic supplement (n = 90), Prenatis™, containing 5 billion CFU/day of Lacticaseibacillus rhamnosus Rosell[®]-11 and Bifidobacterium bifidum HA-132. Results: There was a significantly lower number of women with one or more infections during the study in the probiotics group (8 vs. 18, p = 0.05) and a trend for a lower number of infections during pregnancy (primary outcome) in the probiotics group (p = 0.07). Regarding infants, a lower number of days with infections during the first month of life was observed in the probiotics group (4.7 days on average vs. 10.5 days, p = 0.03). The vaginal microbiota composition during pregnancy and after childbirth showed no significant differences between groups while the infants' gut microbiome demonstrated a significantly higher abundance/prevalence of beneficial taxa in the probiotics group. The benefits conferred by probiotics were especially notable when considering birth by C-section. Probiotics promoted the vertical transmission of beneficial species and the induction of a highly interconnected microbiota, structured around key species. Conclusions: Probiotic supplementation during the third trimester of pregnancy and lactation is a valid strategy for conferring benefits to mothers and infants.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
Female
*Probiotics/administration & dosage
Pregnancy
Double-Blind Method
Adult
Infant, Newborn
Gastrointestinal Microbiome
Infant
Lacticaseibacillus rhamnosus
Vagina/microbiology
Dietary Supplements
Bifidobacterium bifidum
Young Adult
RevDate: 2025-06-13
CmpDate: 2025-06-13
A Comprehensive Scoping Review on Diet and Nutrition in Relation to Long COVID-19 Symptoms and Recovery.
Nutrients, 17(11): pii:nu17111802.
Background/Objectives: Long COVID-19 is characterized by persistent symptoms lasting three months or more following SARS-CoV-2 infection. Nutrition has emerged as a modifiable factor influencing recovery trajectories and symptom burden; however, existing evidence remains fragmented across diverse study designs and populations. This scoping review synthesized global evidence on the role of diet and nutrition in managing long COVID-19 symptoms and supporting recovery. Methods: Following PRISMA-ScR and Joanna Briggs Institute guidelines for scoping reviews, we searched major biomedical databases for studies published between 2020 and 2025. Eligible studies examined dietary intake, nutritional status, or nutrition-related interventions in adults with long COVID-19. Results: After duplicates were removed, 1808 records were screened, resulting in 50 studies that met the inclusion criteria-27 intervention studies and 23 observational studies. Nutritional exposures included micronutrients (e.g., vitamins D, K2), amino acids (e.g., L-arginine), multinutrient formulations, microbiota-targeted therapies (e.g., probiotics, synbiotics), nutritional status, diet quality, and whole-diet patterns (e.g., the Mediterranean diet). Approximately 76% of studies reported improvements in long COVID-19-related symptoms such as fatigue, mood disturbances, physical function, and markers of inflammation. Conclusions: Diet and nutrition may support long COVID-19 recovery by targeting inflammation and the gut microbiome to alleviate symptoms and improve functional outcomes. Well-powered trials of whole-diet approaches, combined with targeted supplementation, are needed to confirm their potential as scalable, accessible tools for post-COVID-19 recovery and management.
Additional Links: PMID-40507071
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40507071,
year = {2025},
author = {Bigman, G and Rusu, ME and Shelawala, N and Sorkin, JD and Beamer, BA and Ryan, AS},
title = {A Comprehensive Scoping Review on Diet and Nutrition in Relation to Long COVID-19 Symptoms and Recovery.},
journal = {Nutrients},
volume = {17},
number = {11},
pages = {},
doi = {10.3390/nu17111802},
pmid = {40507071},
issn = {2072-6643},
mesh = {Humans ; *COVID-19/complications/physiopathology/diet therapy ; *Nutritional Status ; *Diet ; SARS-CoV-2 ; Post-Acute COVID-19 Syndrome ; Adult ; },
abstract = {Background/Objectives: Long COVID-19 is characterized by persistent symptoms lasting three months or more following SARS-CoV-2 infection. Nutrition has emerged as a modifiable factor influencing recovery trajectories and symptom burden; however, existing evidence remains fragmented across diverse study designs and populations. This scoping review synthesized global evidence on the role of diet and nutrition in managing long COVID-19 symptoms and supporting recovery. Methods: Following PRISMA-ScR and Joanna Briggs Institute guidelines for scoping reviews, we searched major biomedical databases for studies published between 2020 and 2025. Eligible studies examined dietary intake, nutritional status, or nutrition-related interventions in adults with long COVID-19. Results: After duplicates were removed, 1808 records were screened, resulting in 50 studies that met the inclusion criteria-27 intervention studies and 23 observational studies. Nutritional exposures included micronutrients (e.g., vitamins D, K2), amino acids (e.g., L-arginine), multinutrient formulations, microbiota-targeted therapies (e.g., probiotics, synbiotics), nutritional status, diet quality, and whole-diet patterns (e.g., the Mediterranean diet). Approximately 76% of studies reported improvements in long COVID-19-related symptoms such as fatigue, mood disturbances, physical function, and markers of inflammation. Conclusions: Diet and nutrition may support long COVID-19 recovery by targeting inflammation and the gut microbiome to alleviate symptoms and improve functional outcomes. Well-powered trials of whole-diet approaches, combined with targeted supplementation, are needed to confirm their potential as scalable, accessible tools for post-COVID-19 recovery and management.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
*COVID-19/complications/physiopathology/diet therapy
*Nutritional Status
*Diet
SARS-CoV-2
Post-Acute COVID-19 Syndrome
Adult
RevDate: 2025-06-13
CmpDate: 2025-06-13
Cultural and Molecular Factors Predisposed to Non-Alcoholic Fatty Liver Disease and Type 2 Diabetes Mellitus.
Nutrients, 17(11): pii:nu17111797.
There is an exponential increase in the global prevalence of non-alcoholic fatty liver disease (NAFLD) in all populations. The objective of this review is to examine how different cultures and molecular entities influence the progression of NAFLD. Research databases, including PubMed, Scopus, the American Diabetes Association, the American Liver Foundation, and Diabetes UK, were used to retrieve information. Our data analysis showed that cultural norms shape the perceptions of health, illness, and mortality, thus influencing how individuals view themselves and express their experiences and may also affect decisions related to treatment and healthcare. Cultural competence, the ability to understand and navigate cultural differences, is essential for eliciting patient and practitioner perspectives and integrating this understanding into diagnostic and treatment plans. By acknowledging and respecting a patient's cultural background, healthcare providers can foster trust, improve care quality, enhance acceptance of diagnoses, and boost treatment adherence. Although cultural factors play a crucial role in the progression of NAFLD, the disease is also shaped by genetic predispositions, molecular mechanisms, and comorbidities. Molecular pathways involved in the development and progression of NAFLD include alterations in lipid metabolism, insulin signaling, insulin resistance, oxidative stress, defective gut microbiome, and inflammation. This study concludes that a combination of cultural preferences and molecular factors has contributed to the worldwide exponential rise in the prevalence of NAFLD, which in turn has led to an increase in the prevalence of comorbidities such as cardiovascular diseases, diabetes mellitus, and metabolic syndrome.
Additional Links: PMID-40507067
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40507067,
year = {2025},
author = {George, H and Permata, FS and D'Souza, CM and Adeghate, EA},
title = {Cultural and Molecular Factors Predisposed to Non-Alcoholic Fatty Liver Disease and Type 2 Diabetes Mellitus.},
journal = {Nutrients},
volume = {17},
number = {11},
pages = {},
doi = {10.3390/nu17111797},
pmid = {40507067},
issn = {2072-6643},
support = {G00003388; G00002809; G00003627, 31M374//United Arab Emirates University/ ; },
mesh = {Humans ; *Non-alcoholic Fatty Liver Disease/epidemiology/ethnology/genetics/etiology ; *Diabetes Mellitus, Type 2/ethnology/epidemiology/etiology ; Prevalence ; *Culture ; Insulin Resistance ; Genetic Predisposition to Disease ; },
abstract = {There is an exponential increase in the global prevalence of non-alcoholic fatty liver disease (NAFLD) in all populations. The objective of this review is to examine how different cultures and molecular entities influence the progression of NAFLD. Research databases, including PubMed, Scopus, the American Diabetes Association, the American Liver Foundation, and Diabetes UK, were used to retrieve information. Our data analysis showed that cultural norms shape the perceptions of health, illness, and mortality, thus influencing how individuals view themselves and express their experiences and may also affect decisions related to treatment and healthcare. Cultural competence, the ability to understand and navigate cultural differences, is essential for eliciting patient and practitioner perspectives and integrating this understanding into diagnostic and treatment plans. By acknowledging and respecting a patient's cultural background, healthcare providers can foster trust, improve care quality, enhance acceptance of diagnoses, and boost treatment adherence. Although cultural factors play a crucial role in the progression of NAFLD, the disease is also shaped by genetic predispositions, molecular mechanisms, and comorbidities. Molecular pathways involved in the development and progression of NAFLD include alterations in lipid metabolism, insulin signaling, insulin resistance, oxidative stress, defective gut microbiome, and inflammation. This study concludes that a combination of cultural preferences and molecular factors has contributed to the worldwide exponential rise in the prevalence of NAFLD, which in turn has led to an increase in the prevalence of comorbidities such as cardiovascular diseases, diabetes mellitus, and metabolic syndrome.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
*Non-alcoholic Fatty Liver Disease/epidemiology/ethnology/genetics/etiology
*Diabetes Mellitus, Type 2/ethnology/epidemiology/etiology
Prevalence
*Culture
Insulin Resistance
Genetic Predisposition to Disease
RevDate: 2025-06-13
CmpDate: 2025-06-13
Age-Dependent Gut Microbiome Dysbiosis in Autism Spectrum Disorder and the Role of Key Bacterial Ratios.
Nutrients, 17(11): pii:nu17111775.
Background/Objectives: Autism spectrum disorder (ASD) has a wide-ranging impact on individuals' quality of life and development, and there is a critical need for greater awareness, early intervention, and comprehensive support strategies to effectively address the unique needs of those affected by ASD. Recent studies highlight the gut microbiome's potential role in modulating ASD symptoms via the gut-brain axis, but specific microbial biomarkers remain unclear. This study aims to investigate differences in gut microbiota composition between ASD patients and neurotypical controls in a novel approach, specifically assessing ratios of Firmicutes/Bacteroidetes (F/B), Actinobacteria/Proteobacteria (A/P), and Prevotella/Bacteroides (P/B) as potential biomarkers. Methods: We analyzed gut microbiome samples from 302 Bulgarian children and adolescents diagnosed with ASD (aged 2-19 years). Microbial ratios (F/B, A/P, and P/B) were calculated and compared against previously reported reference meta-analytic means from European neurotypical populations. The statistical significance of deviations was assessed using parametric (t-tests), non-parametric (Wilcoxon signed-rank tests), and proportion-based (binomial tests) methods. Effect sizes were quantified using Cohen's d. Significant differences between ASD cases and neurotypical reference values were observed across several age groups. Results: Notably, children with ASD demonstrated significantly lower F/B and A/P ratios, with the youngest cohort (0-4 years) exhibiting the greatest differences. Deviations in the P/B ratio varied across age groups, with a significant elevation in the oldest group (≥10 years). Collectively, ASD cases consistently exhibited microbiota profiles indicative of dysbiosis. Conclusions: Our findings support gut microbiome dysbiosis as a potential biomarker for ASD, highlighting significantly altered bacterial ratios compared to neurotypical controls. These microbiome shifts could reflect early-life disruptions influencing neurodevelopment. Future studies should adopt longitudinal and mechanistic approaches to elucidate causal relationships and evaluate therapeutic microbiome modulation strategies.
Additional Links: PMID-40507042
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40507042,
year = {2025},
author = {Kadiyska, T and Vassilev, D and Tourtourikov, I and Ciurinskiene, S and Madzharova, D and Savcheva, M and Stoynev, N and Mileva-Popova, R and Tafradjiiska-Hadjiolova, R and Mitev, V},
title = {Age-Dependent Gut Microbiome Dysbiosis in Autism Spectrum Disorder and the Role of Key Bacterial Ratios.},
journal = {Nutrients},
volume = {17},
number = {11},
pages = {},
doi = {10.3390/nu17111775},
pmid = {40507042},
issn = {2072-6643},
support = {D-116/29.05.2024//Medical University of Sofia/ ; },
mesh = {Humans ; *Gastrointestinal Microbiome ; *Autism Spectrum Disorder/microbiology ; *Dysbiosis/microbiology ; Child ; Adolescent ; Child, Preschool ; Male ; Female ; Young Adult ; Age Factors ; Feces/microbiology ; Biomarkers ; Case-Control Studies ; },
abstract = {Background/Objectives: Autism spectrum disorder (ASD) has a wide-ranging impact on individuals' quality of life and development, and there is a critical need for greater awareness, early intervention, and comprehensive support strategies to effectively address the unique needs of those affected by ASD. Recent studies highlight the gut microbiome's potential role in modulating ASD symptoms via the gut-brain axis, but specific microbial biomarkers remain unclear. This study aims to investigate differences in gut microbiota composition between ASD patients and neurotypical controls in a novel approach, specifically assessing ratios of Firmicutes/Bacteroidetes (F/B), Actinobacteria/Proteobacteria (A/P), and Prevotella/Bacteroides (P/B) as potential biomarkers. Methods: We analyzed gut microbiome samples from 302 Bulgarian children and adolescents diagnosed with ASD (aged 2-19 years). Microbial ratios (F/B, A/P, and P/B) were calculated and compared against previously reported reference meta-analytic means from European neurotypical populations. The statistical significance of deviations was assessed using parametric (t-tests), non-parametric (Wilcoxon signed-rank tests), and proportion-based (binomial tests) methods. Effect sizes were quantified using Cohen's d. Significant differences between ASD cases and neurotypical reference values were observed across several age groups. Results: Notably, children with ASD demonstrated significantly lower F/B and A/P ratios, with the youngest cohort (0-4 years) exhibiting the greatest differences. Deviations in the P/B ratio varied across age groups, with a significant elevation in the oldest group (≥10 years). Collectively, ASD cases consistently exhibited microbiota profiles indicative of dysbiosis. Conclusions: Our findings support gut microbiome dysbiosis as a potential biomarker for ASD, highlighting significantly altered bacterial ratios compared to neurotypical controls. These microbiome shifts could reflect early-life disruptions influencing neurodevelopment. Future studies should adopt longitudinal and mechanistic approaches to elucidate causal relationships and evaluate therapeutic microbiome modulation strategies.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Humans
*Gastrointestinal Microbiome
*Autism Spectrum Disorder/microbiology
*Dysbiosis/microbiology
Child
Adolescent
Child, Preschool
Male
Female
Young Adult
Age Factors
Feces/microbiology
Biomarkers
Case-Control Studies
RevDate: 2025-06-13
The Lung Microbiome and Its Impact on Obstructive Sleep Apnea: A Diagnostic Frontier.
Diagnostics (Basel, Switzerland), 15(11): pii:diagnostics15111431.
Obstructive sleep apnea (OSA), a prevalent disorder characterized by recurrent upper airway collapse, is increasingly recognized as a systemic inflammatory condition influenced by microbial dysregulation. Emerging evidence underscores the lung microbiome as a mediator in OSA pathophysiology, where dysbiotic shifts driven by intermittent hypoxia, oxidative stress and mechanical airway trauma amplify inflammatory cascades and perpetuate respiratory instability. This review synthesizes current knowledge on the bidirectional interplay between OSA and lung microbial communities. It aims to highlight how hypoxia-induced alterations in microbial ecology disrupt immune homeostasis, while inflammation-driven mucosal injury fosters pathogenic colonization. Clinical correlations between specific taxa like Streptococcus and Prevotella, and disease severity, suggest microbial signatures as novel biomarkers for OSA progression and treatment response. Furthermore, oxidative stress markers and pro-inflammatory cytokines emerge as potential diagnostic tools that bridge microbial dysbiosis with sleep-related outcomes. However, challenges persist in sampling standardization of the low-biomass lower airways, as well as in causative mechanisms linking microbial dysbiosis to OSA pathophysiology. By integrating microbial ecology with precision sleep medicine, this paradigm shift promises to transform OSA management from mechanical stabilization to holistic ecosystem restoration.
Additional Links: PMID-40507003
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40507003,
year = {2025},
author = {Karkala, A and Kotoulas, SC and Tzinas, A and Massa, E and Mouloudi, E and Gkakou, F and Pataka, A},
title = {The Lung Microbiome and Its Impact on Obstructive Sleep Apnea: A Diagnostic Frontier.},
journal = {Diagnostics (Basel, Switzerland)},
volume = {15},
number = {11},
pages = {},
doi = {10.3390/diagnostics15111431},
pmid = {40507003},
issn = {2075-4418},
abstract = {Obstructive sleep apnea (OSA), a prevalent disorder characterized by recurrent upper airway collapse, is increasingly recognized as a systemic inflammatory condition influenced by microbial dysregulation. Emerging evidence underscores the lung microbiome as a mediator in OSA pathophysiology, where dysbiotic shifts driven by intermittent hypoxia, oxidative stress and mechanical airway trauma amplify inflammatory cascades and perpetuate respiratory instability. This review synthesizes current knowledge on the bidirectional interplay between OSA and lung microbial communities. It aims to highlight how hypoxia-induced alterations in microbial ecology disrupt immune homeostasis, while inflammation-driven mucosal injury fosters pathogenic colonization. Clinical correlations between specific taxa like Streptococcus and Prevotella, and disease severity, suggest microbial signatures as novel biomarkers for OSA progression and treatment response. Furthermore, oxidative stress markers and pro-inflammatory cytokines emerge as potential diagnostic tools that bridge microbial dysbiosis with sleep-related outcomes. However, challenges persist in sampling standardization of the low-biomass lower airways, as well as in causative mechanisms linking microbial dysbiosis to OSA pathophysiology. By integrating microbial ecology with precision sleep medicine, this paradigm shift promises to transform OSA management from mechanical stabilization to holistic ecosystem restoration.},
}
RevDate: 2025-06-12
CmpDate: 2025-06-12
The gut microbiome and metabolomic alterations underlying colitis-induced encephalopathy in mice: mechanistic insight.
Behavioral and brain functions : BBF, 21(1):17.
BACKGROUND: In addition to classical gastrointestinal symptoms, patients with inflammatory bowel disease (IBD) often exhibit neurological manifestations, such as mood disorders and cognitive dysfunctions, which are frequently overlooked. However, the potential pathogenesis of IBD-related encephalopathy remains unclear, and few studies have explored the influence of interactions between the gut microbiota and the host gut-brain metabolome on the emergence of brain diseases in IBD mice. In this study, we conducted a comprehensive analysis of gut microbiome and metabolome alterations in dextran sulfate sodium salt (DSS)-induced IBD mice compared to control mice, focusing on colonic contents and hippocampal tissue. Our aim was to investigate the putative mechanisms underlying the microbiota-gut-brain axis in IBD-induced encephalopathy.
RESULTS: IBD mice showed depression-like behaviors and cognitive deficits. Metabolic profiling revealed distinct patterns in the colonic contents and hippocampal areas of IBD mice, marked by decreased energy metabolism, amino acid levels, short-chain fatty acids (SCFAs), and choline metabolism. These metabolic changes were negatively associated with the abundance of Bacteroides, Turicibacter, Ruminococcus, and Akkermansia, while Desulfovibrio and Lactobacillus showed positive correlations.
CONCLUSIONS: This study identifies unique microbial and gut-brain metabolite signatures associated with DSS-induced changes and offers new metabolic insights into the microbiota-gut-brain axis in IBD-related brain disorders. It highlights the potential of targeting gut microbiota to modulate host metabolism as a therapeutic approach for IBD-related neurological complications.
Additional Links: PMID-40506791
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40506791,
year = {2025},
author = {Cai, A and Shen, D and Xiong, Q and Li, S and Qiu, C and Li, L and Chen, Z and Lin, X and Yao, Q and Zhang, Y and Chen, R and Kou, L},
title = {The gut microbiome and metabolomic alterations underlying colitis-induced encephalopathy in mice: mechanistic insight.},
journal = {Behavioral and brain functions : BBF},
volume = {21},
number = {1},
pages = {17},
pmid = {40506791},
issn = {1744-9081},
support = {Y20210133//Wenzhou Science and Technology Bureau/ ; 2024ZYC-A10//Zhejiang Medical Association Clinical Medical Research special fund project/ ; 2023KY907//Zhejiang Medical and Health Science and Technology Program/ ; },
mesh = {Animals ; *Gastrointestinal Microbiome/physiology ; Mice ; *Colitis/metabolism/complications/chemically induced/microbiology ; Male ; Hippocampus/metabolism ; *Metabolome/physiology ; *Brain Diseases/metabolism/microbiology/etiology ; Dextran Sulfate ; Colon/metabolism ; *Inflammatory Bowel Diseases/metabolism/complications/microbiology ; Disease Models, Animal ; Mice, Inbred C57BL ; Brain/metabolism ; Metabolomics ; Fatty Acids, Volatile/metabolism ; Brain-Gut Axis/physiology ; },
abstract = {BACKGROUND: In addition to classical gastrointestinal symptoms, patients with inflammatory bowel disease (IBD) often exhibit neurological manifestations, such as mood disorders and cognitive dysfunctions, which are frequently overlooked. However, the potential pathogenesis of IBD-related encephalopathy remains unclear, and few studies have explored the influence of interactions between the gut microbiota and the host gut-brain metabolome on the emergence of brain diseases in IBD mice. In this study, we conducted a comprehensive analysis of gut microbiome and metabolome alterations in dextran sulfate sodium salt (DSS)-induced IBD mice compared to control mice, focusing on colonic contents and hippocampal tissue. Our aim was to investigate the putative mechanisms underlying the microbiota-gut-brain axis in IBD-induced encephalopathy.
RESULTS: IBD mice showed depression-like behaviors and cognitive deficits. Metabolic profiling revealed distinct patterns in the colonic contents and hippocampal areas of IBD mice, marked by decreased energy metabolism, amino acid levels, short-chain fatty acids (SCFAs), and choline metabolism. These metabolic changes were negatively associated with the abundance of Bacteroides, Turicibacter, Ruminococcus, and Akkermansia, while Desulfovibrio and Lactobacillus showed positive correlations.
CONCLUSIONS: This study identifies unique microbial and gut-brain metabolite signatures associated with DSS-induced changes and offers new metabolic insights into the microbiota-gut-brain axis in IBD-related brain disorders. It highlights the potential of targeting gut microbiota to modulate host metabolism as a therapeutic approach for IBD-related neurological complications.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Animals
*Gastrointestinal Microbiome/physiology
Mice
*Colitis/metabolism/complications/chemically induced/microbiology
Male
Hippocampus/metabolism
*Metabolome/physiology
*Brain Diseases/metabolism/microbiology/etiology
Dextran Sulfate
Colon/metabolism
*Inflammatory Bowel Diseases/metabolism/complications/microbiology
Disease Models, Animal
Mice, Inbred C57BL
Brain/metabolism
Metabolomics
Fatty Acids, Volatile/metabolism
Brain-Gut Axis/physiology
RevDate: 2025-06-12
CmpDate: 2025-06-12
Underground guardians: how collagen and chitin amendments shape soil microbiome structure and function for Meloidogyne enterolobii control.
Microbiome, 13(1):141.
BACKGROUND: The emergence of the guava root-knot nematode (Meloidogyne enterolobii) poses a significant threat to tomato yields globally. This study evaluated the impact of collagen and chitin soil amendments on soil microbial composition and function (fungal and bacterial communities) and their effects on tomato plant health and M. enterolobii infection under standard (5000 eggs plant[-1]) and high (50,000 eggs plant[-1]) inoculum pressure. Conducted in a greenhouse setting, the study investigated the effectiveness of these amendments in nurturing beneficial microbial communities across both native and agricultural soils.
RESULTS: Both collagen and chitin were effective in reducing nematode egg counts by up to 66% and 84% under standard and high inoculum pressure, respectively, and enhanced plant health parameters (biomass and chlorophyll content). Moreover, a microbiome shift led to an increase in bacterial (Kitasatospora, Bacillus, and Streptomyces) and fungal (Phialemonium) genera, known for their chitinase, collagenase, and plant-parasitic nematode control. Among the microbes, Streptomyces spp. were found among the core microbiome and associated with a lower disease incidence assessed through a phenotype-OTU network analysis (PhONA). Under standard inoculum, higher metabolite expression was observed with amino acids representing a majority among the metabolite groups.
CONCLUSIONS: The findings highlight the potential of collagen and chitin to mitigate M. enterolobii infection by fostering beneficial soil microbial communities. Video Abstract.
Additional Links: PMID-40506747
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40506747,
year = {2025},
author = {Pasche, JM and Sawlani, R and Buttrós, VH and Desaeger, J and Garrett, KA and Martins, SJ},
title = {Underground guardians: how collagen and chitin amendments shape soil microbiome structure and function for Meloidogyne enterolobii control.},
journal = {Microbiome},
volume = {13},
number = {1},
pages = {141},
pmid = {40506747},
issn = {2049-2618},
support = {2021-68013-33758//National Institute of Food and Agriculture/ ; },
mesh = {*Chitin/pharmacology ; Animals ; *Soil Microbiology ; *Tylenchoidea/drug effects/pathogenicity ; Solanum lycopersicum/parasitology/growth & development/microbiology ; *Collagen/pharmacology ; Soil/chemistry ; *Microbiota/drug effects ; *Plant Diseases/parasitology/prevention & control ; Bacteria/classification/genetics/isolation & purification ; Fungi/classification/isolation & purification/genetics ; Plant Roots/parasitology ; },
abstract = {BACKGROUND: The emergence of the guava root-knot nematode (Meloidogyne enterolobii) poses a significant threat to tomato yields globally. This study evaluated the impact of collagen and chitin soil amendments on soil microbial composition and function (fungal and bacterial communities) and their effects on tomato plant health and M. enterolobii infection under standard (5000 eggs plant[-1]) and high (50,000 eggs plant[-1]) inoculum pressure. Conducted in a greenhouse setting, the study investigated the effectiveness of these amendments in nurturing beneficial microbial communities across both native and agricultural soils.
RESULTS: Both collagen and chitin were effective in reducing nematode egg counts by up to 66% and 84% under standard and high inoculum pressure, respectively, and enhanced plant health parameters (biomass and chlorophyll content). Moreover, a microbiome shift led to an increase in bacterial (Kitasatospora, Bacillus, and Streptomyces) and fungal (Phialemonium) genera, known for their chitinase, collagenase, and plant-parasitic nematode control. Among the microbes, Streptomyces spp. were found among the core microbiome and associated with a lower disease incidence assessed through a phenotype-OTU network analysis (PhONA). Under standard inoculum, higher metabolite expression was observed with amino acids representing a majority among the metabolite groups.
CONCLUSIONS: The findings highlight the potential of collagen and chitin to mitigate M. enterolobii infection by fostering beneficial soil microbial communities. Video Abstract.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
*Chitin/pharmacology
Animals
*Soil Microbiology
*Tylenchoidea/drug effects/pathogenicity
Solanum lycopersicum/parasitology/growth & development/microbiology
*Collagen/pharmacology
Soil/chemistry
*Microbiota/drug effects
*Plant Diseases/parasitology/prevention & control
Bacteria/classification/genetics/isolation & purification
Fungi/classification/isolation & purification/genetics
Plant Roots/parasitology
RevDate: 2025-06-12
Selective recruitment of beneficial microbes in the rhizosphere of maize affected by microbial inoculants, farming practice, and seasonal variations.
Environmental microbiome, 20(1):69.
BACKGROUND: Plant beneficial microorganisms as inoculants can improve crop performance, but factors affecting their impact on plant performance under field conditions remain unclear, thereby limiting their use in farming. Here, we investigated how farming practices (e.g., tillage and N-fertilization intensity) and growing seasons influenced the impact of a beneficial microorganism consortium (BMc: Trichoderma, Bacillus, and Pseudomonas strains) in maize and affected the rhizosphere competence of each BMc strain. In addition, we tested whether the consortium affects the resident rhizosphere microbiome and crop performance. In two growing seasons (2020 and 2021), we assessed how BMc inoculation affects maize growth, nutritional status, gene expression, and rhizosphere microbiome under different farming practices at the flowering stage.
RESULTS: Inoculated strains successfully colonized the maize rhizosphere independently of farming practice. BMc inoculation improved plant growth and iron uptake in 2020, regardless of farming practice. These effects co-occurred with lower precipitation levels in 2020 compared to 2021. BMc inoculation reduced the expression of several stress-related genes in maize in 2020 under drought. An increased iron uptake by the BMc-inoculated plants was observed in 2020 and was associated with the upregulation of the gene ZmNAS3, which is linked to iron uptake. Therefore, BMc inoculation mitigated the drought impact on maize. The microbial rhizosphere communities were altered by BMc inoculation in both years, but patterns of responder taxa differed between seasons. Metagenome analysis revealed that more genes (e.g., genes encoding biosurfactants and siderophores) were enriched in the rhizosphere of BMc-inoculated plants in 2020 than in 2021. Moreover, we identified bacterial and fungal taxa positively associated with maize iron uptake. The relative abundance of these iron uptake-associated bacterial and fungal taxa significantly increased due to BMc inoculation in 2020, while they showed overall higher relative abundances in 2021, independently of BMc inoculation. We mapped the sequences of these iron-associated taxa to publicly available genomes and verified the occurrence of various plant beneficial traits in several mapped genomes.
CONCLUSIONS: Overall, we show that the growing season determined the effect of BMc inoculation on maize plants by shaping microbiome composition and function in the maize rhizosphere more than farming practice. These findings highlight the importance of the complex interplay between microbial inoculants and the resident rhizosphere microorganisms under abiotic stress conditions.
Additional Links: PMID-40506735
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40506735,
year = {2025},
author = {Kampouris, ID and Kuhl-Nagel, T and Behr, JH and Sommermann, L and Babin, D and Francioli, D and Zrenner, R and Kublik, S and Schloter, M and Ludewig, U and Smalla, K and Neumann, G and Grosch, R and Geistlinger, J},
title = {Selective recruitment of beneficial microbes in the rhizosphere of maize affected by microbial inoculants, farming practice, and seasonal variations.},
journal = {Environmental microbiome},
volume = {20},
number = {1},
pages = {69},
pmid = {40506735},
issn = {2524-6372},
abstract = {BACKGROUND: Plant beneficial microorganisms as inoculants can improve crop performance, but factors affecting their impact on plant performance under field conditions remain unclear, thereby limiting their use in farming. Here, we investigated how farming practices (e.g., tillage and N-fertilization intensity) and growing seasons influenced the impact of a beneficial microorganism consortium (BMc: Trichoderma, Bacillus, and Pseudomonas strains) in maize and affected the rhizosphere competence of each BMc strain. In addition, we tested whether the consortium affects the resident rhizosphere microbiome and crop performance. In two growing seasons (2020 and 2021), we assessed how BMc inoculation affects maize growth, nutritional status, gene expression, and rhizosphere microbiome under different farming practices at the flowering stage.
RESULTS: Inoculated strains successfully colonized the maize rhizosphere independently of farming practice. BMc inoculation improved plant growth and iron uptake in 2020, regardless of farming practice. These effects co-occurred with lower precipitation levels in 2020 compared to 2021. BMc inoculation reduced the expression of several stress-related genes in maize in 2020 under drought. An increased iron uptake by the BMc-inoculated plants was observed in 2020 and was associated with the upregulation of the gene ZmNAS3, which is linked to iron uptake. Therefore, BMc inoculation mitigated the drought impact on maize. The microbial rhizosphere communities were altered by BMc inoculation in both years, but patterns of responder taxa differed between seasons. Metagenome analysis revealed that more genes (e.g., genes encoding biosurfactants and siderophores) were enriched in the rhizosphere of BMc-inoculated plants in 2020 than in 2021. Moreover, we identified bacterial and fungal taxa positively associated with maize iron uptake. The relative abundance of these iron uptake-associated bacterial and fungal taxa significantly increased due to BMc inoculation in 2020, while they showed overall higher relative abundances in 2021, independently of BMc inoculation. We mapped the sequences of these iron-associated taxa to publicly available genomes and verified the occurrence of various plant beneficial traits in several mapped genomes.
CONCLUSIONS: Overall, we show that the growing season determined the effect of BMc inoculation on maize plants by shaping microbiome composition and function in the maize rhizosphere more than farming practice. These findings highlight the importance of the complex interplay between microbial inoculants and the resident rhizosphere microorganisms under abiotic stress conditions.},
}
RevDate: 2025-06-12
CmpDate: 2025-06-12
Metagenomic insight into the vaginal microbiome in women infected with HPV 16 and 18.
NPJ biofilms and microbiomes, 11(1):105.
Human papillomavirus (HPV) 16 and 18 (HPV 16/18) account for over 70% of cervical cancer (CC) cases, yet their interaction with the vaginal microbiome (VM) remains unclear. This study explored the association between high-risk HPV types (HR-HPVs), VM composition and bacterial function using shotgun metagenomic sequencing. In early-stage cervical lesions, the HPV 16/18 group showed reduced Lactobacillus-dominant community state types compared to other HR-HPVs, while invasive CC exhibited increased pathogenic bacteria, including Streptococcus agalactiae, Fannyhessea vaginae, and Sneathia vaginalis. The VM associated with HPV 16/18 was enriched in immune response and inflammation pathways, whereas other HR-HPVs were linked to cellular metabolism and hormonal signaling. Notably, HPV 16/18 exhibited stronger bacterial-fungal correlations, indicating shifts in the microbial community. Furthermore, 137 metagenome-assembled genomes provided insights into unique microbial genomic signatures. Our study links VM differences with HPV 16/18 oncogenic potential across cervical lesion stages, urging further research for better diagnostics and treatments.
Additional Links: PMID-40506497
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40506497,
year = {2025},
author = {Jung, DR and Choi, Y and Jeong, M and Singh, V and Jeon, SY and Seo, I and Park, NJ and Lee, YH and Park, JY and Han, HS and Shin, JH and Chong, GO},
title = {Metagenomic insight into the vaginal microbiome in women infected with HPV 16 and 18.},
journal = {NPJ biofilms and microbiomes},
volume = {11},
number = {1},
pages = {105},
pmid = {40506497},
issn = {2055-5008},
mesh = {Female ; Humans ; *Vagina/microbiology/virology ; *Human papillomavirus 16/genetics/isolation & purification ; *Papillomavirus Infections/virology/microbiology ; *Microbiota/genetics ; Metagenomics ; *Human papillomavirus 18/genetics/isolation & purification ; *Bacteria/classification/genetics/isolation & purification ; Uterine Cervical Neoplasms/virology/microbiology ; Metagenome ; Adult ; Middle Aged ; },
abstract = {Human papillomavirus (HPV) 16 and 18 (HPV 16/18) account for over 70% of cervical cancer (CC) cases, yet their interaction with the vaginal microbiome (VM) remains unclear. This study explored the association between high-risk HPV types (HR-HPVs), VM composition and bacterial function using shotgun metagenomic sequencing. In early-stage cervical lesions, the HPV 16/18 group showed reduced Lactobacillus-dominant community state types compared to other HR-HPVs, while invasive CC exhibited increased pathogenic bacteria, including Streptococcus agalactiae, Fannyhessea vaginae, and Sneathia vaginalis. The VM associated with HPV 16/18 was enriched in immune response and inflammation pathways, whereas other HR-HPVs were linked to cellular metabolism and hormonal signaling. Notably, HPV 16/18 exhibited stronger bacterial-fungal correlations, indicating shifts in the microbial community. Furthermore, 137 metagenome-assembled genomes provided insights into unique microbial genomic signatures. Our study links VM differences with HPV 16/18 oncogenic potential across cervical lesion stages, urging further research for better diagnostics and treatments.},
}
MeSH Terms:
show MeSH Terms
hide MeSH Terms
Female
Humans
*Vagina/microbiology/virology
*Human papillomavirus 16/genetics/isolation & purification
*Papillomavirus Infections/virology/microbiology
*Microbiota/genetics
Metagenomics
*Human papillomavirus 18/genetics/isolation & purification
*Bacteria/classification/genetics/isolation & purification
Uterine Cervical Neoplasms/virology/microbiology
Metagenome
Adult
Middle Aged
RevDate: 2025-06-12
Detection of Volatile Organic Compounds as an emerging strategy for Parkinson's disease diagnosis and monitoring.
NPJ Parkinson's disease, 11(1):161.
Growing evidence suggests that specific volatile organic compound (VOC) profiles may reflect key pathophysiological processes in Parkinson's disease (PD), including alterations in the microbiome, metabolism, and oxidative stress. Identifying reliable VOC biomarkers could enable non-invasive tests for early diagnosis, disease monitoring, and therapy evaluation. This review examines VOC analysis in biological matrices such as breath, skin, and stool, outlining current research and future applications in PD. We evaluate analytical techniques based on sensitivity, specificity, and clinical applicability. Additionally, we classify VOCs identified in previous studies alongside their proposed biological origins. Special attention is given to short-chain fatty acids, produced by the gut microbiome, a novel target in PD research. Our findings highlight the need for larger cohort studies and standardized protocols to advance VOC-based diagnostics in PD. Understanding the interplay between VOCs and PD may facilitate biomarker discovery, enhancing non-invasive diagnostic strategies and personalized disease management.
Additional Links: PMID-40506424
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40506424,
year = {2025},
author = {Belluomo, I and Tarazi, M and Lao-Kaim, NP and Tai, YF and Spanel, P and Hanna, GB},
title = {Detection of Volatile Organic Compounds as an emerging strategy for Parkinson's disease diagnosis and monitoring.},
journal = {NPJ Parkinson's disease},
volume = {11},
number = {1},
pages = {161},
pmid = {40506424},
issn = {2373-8057},
abstract = {Growing evidence suggests that specific volatile organic compound (VOC) profiles may reflect key pathophysiological processes in Parkinson's disease (PD), including alterations in the microbiome, metabolism, and oxidative stress. Identifying reliable VOC biomarkers could enable non-invasive tests for early diagnosis, disease monitoring, and therapy evaluation. This review examines VOC analysis in biological matrices such as breath, skin, and stool, outlining current research and future applications in PD. We evaluate analytical techniques based on sensitivity, specificity, and clinical applicability. Additionally, we classify VOCs identified in previous studies alongside their proposed biological origins. Special attention is given to short-chain fatty acids, produced by the gut microbiome, a novel target in PD research. Our findings highlight the need for larger cohort studies and standardized protocols to advance VOC-based diagnostics in PD. Understanding the interplay between VOCs and PD may facilitate biomarker discovery, enhancing non-invasive diagnostic strategies and personalized disease management.},
}
RevDate: 2025-06-12
Potential role of intratumoral Fusobacterium nucleatum and interleukin-1 beta in breast cancer cell growth.
Journal of microbiology, immunology, and infection = Wei mian yu gan ran za zhi pii:S1684-1182(25)00119-7 [Epub ahead of print].
BACKGROUND: It has been shown that the human breast harbors a rich and diverse microbiome, with significant differences observed between tumor tissue and normal breast tissue. Recently, Fusobacterium nucleatum (F. nucleatum) has been shown to affect breast cancer growth, but the underlying mechanism remains enigmatic.
METHODS: Breast cancer tissues were obtained from clinical patients and analyzed for the microbiome composition using 16S rDNA sequencing and qPCR. Both serum and intratumoral cytokine levels were measured to assess their correlation with intratumoral F. nucleatum. Breast cancer cell lines and patient-derived cancer cells were infected with different strains of F. nucleatum, followed by different analyses. Additionally, peripheral blood mononuclear cells (PBMCs) were isolated from healthy individuals to investigate the immunoregulatory effect of F. nucleatum.
RESULTS: Our results identified a higher abundance of F. nucleatum in breast cancer tissue compared to adjacent normal breast tissue, which strongly correlated with intratumoral IL-1β levels. In vitro studies confirmed this correlation, demonstrating that infection of breast cancer cells with F. nucleatum promotes tumor growth. Further investigation suggested that F. nucleatum induces IL-1β secretion in both breast cancer cells and PBMCs, but only IL-1β secreted by breast cancer cells stimulates cancer cell growth. Inhibition of NLRP3 reversed the growth-promoting effect of F. nucleatum on breast cancer cells.
CONCLUSION: Our results demonstrate the role of F. nucleatum in stimulating breast cancer cell growth. Therefore, targeting intratumoral F. nucleatum could provide a promising therapeutic approach to combat breast cancer.
Additional Links: PMID-40506319
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40506319,
year = {2025},
author = {Chang, CM and Lam, LY and Lam, HYP and Kao, PY and Hsu, ST and Wu, WJ and Chang, KC and Huang, CY},
title = {Potential role of intratumoral Fusobacterium nucleatum and interleukin-1 beta in breast cancer cell growth.},
journal = {Journal of microbiology, immunology, and infection = Wei mian yu gan ran za zhi},
volume = {},
number = {},
pages = {},
doi = {10.1016/j.jmii.2025.05.008},
pmid = {40506319},
issn = {1995-9133},
abstract = {BACKGROUND: It has been shown that the human breast harbors a rich and diverse microbiome, with significant differences observed between tumor tissue and normal breast tissue. Recently, Fusobacterium nucleatum (F. nucleatum) has been shown to affect breast cancer growth, but the underlying mechanism remains enigmatic.
METHODS: Breast cancer tissues were obtained from clinical patients and analyzed for the microbiome composition using 16S rDNA sequencing and qPCR. Both serum and intratumoral cytokine levels were measured to assess their correlation with intratumoral F. nucleatum. Breast cancer cell lines and patient-derived cancer cells were infected with different strains of F. nucleatum, followed by different analyses. Additionally, peripheral blood mononuclear cells (PBMCs) were isolated from healthy individuals to investigate the immunoregulatory effect of F. nucleatum.
RESULTS: Our results identified a higher abundance of F. nucleatum in breast cancer tissue compared to adjacent normal breast tissue, which strongly correlated with intratumoral IL-1β levels. In vitro studies confirmed this correlation, demonstrating that infection of breast cancer cells with F. nucleatum promotes tumor growth. Further investigation suggested that F. nucleatum induces IL-1β secretion in both breast cancer cells and PBMCs, but only IL-1β secreted by breast cancer cells stimulates cancer cell growth. Inhibition of NLRP3 reversed the growth-promoting effect of F. nucleatum on breast cancer cells.
CONCLUSION: Our results demonstrate the role of F. nucleatum in stimulating breast cancer cell growth. Therefore, targeting intratumoral F. nucleatum could provide a promising therapeutic approach to combat breast cancer.},
}
RevDate: 2025-06-12
Cystic Fibrosis Microbiome-directed Antibiotic Therapy Trial in Exacerbations Results Stratified (CFMATTERS): Results of a multi-centre randomised controlled trial.
The European respiratory journal pii:13993003.02443-2024 [Epub ahead of print].
BACKGROUND: This study explores the effectiveness and safety of microbiome-directed-antimicrobial-therapy versus usual-antimicrobial-therapy in adult cystic fibrosis pulmonary exacerbations.
METHODS: A multi-centre two-arm parallel randomised control trial conducted across Europe/North-America enrolled 223 participants (January 2015 - August 2017). All participants were chronically colonised with Pseudomonas aeruginosa and were randomised 1:1 into two study-arms. The "usual-therapy group" received 2-weeks of IV ceftazidime 3g thrice-daily (for allergies: aztreonam 2g thrice-daily) and tobramycin 5-10mg·kg[-1] once-daily. The "microbiome-directed group" received the same usual-therapy plus an additional antibiotic with greatest presumed activity against the 2nd, 3rd and 4th most abundant genera present in the sputum microbiome, selected by a Consensus Expert Treatment Panel. The primary outcome was change in percentage of predicted FEV1 (ppFEV1) at 14 days post initiation of antibiotics. Secondary outcomes examined ppFEV1 at 7 days, 28 days, and 3 months; time-to-next exacerbation; symptom burden at 7 days; Health Related Quality of Life (HRQoL) at 28 days; and number of exacerbations and IV antibiotic days at 12 months.
RESULTS: 149 participants had an eligible exacerbation (usual-therapy n=83, microbiome-directed therapy n=66). There was no difference between the groups for ppFEV1 at day 14 (-1.1%, 95%CI -3.9 to 1.7; p=0.46), or ppFEV1 measured at other time-points, or for time-to-next exacerbation (microbiome-directed versus usual-therapy Hazard Ratio 0.91 [95%CI 0.60 to 1.38; p=0.66]). The microbiome-directed group trended to have more IV days (median 42 versus 28; p=0.08) and more subsequent exacerbations (median 3 versus 2; p=0.044) the following year. There were no appreciable differences in symptom burden; however, HRQoL sub-scores were consistently worse in the microbiome-directed group (-4.3 points versus usual therapy (95%CI -8.3 to -0.3, p=0.033).
CONCLUSION: The addition of a third antibiotic based on sputum microbiome sequencing analysis did not result in improved clinical outcomes.
Additional Links: PMID-40506211
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40506211,
year = {2025},
author = {Plant, BJ and Einarsson, GG and Deasy, KF and Dahly, D and Singh, PK and Barry, PJ and Goss, CH and Fajac, I and Vagg, T and Durieu, I and Flanagan, E and O'Callaghan, G and Martin, C and Burgel, PR and Haworth, CS and Floto, RA and Downey, DG and Dupont, LJ and Jones, AM and Elborn, JS and Eustace, JA and Mall, MA and Tunney, MM},
title = {Cystic Fibrosis Microbiome-directed Antibiotic Therapy Trial in Exacerbations Results Stratified (CFMATTERS): Results of a multi-centre randomised controlled trial.},
journal = {The European respiratory journal},
volume = {},
number = {},
pages = {},
doi = {10.1183/13993003.02443-2024},
pmid = {40506211},
issn = {1399-3003},
abstract = {BACKGROUND: This study explores the effectiveness and safety of microbiome-directed-antimicrobial-therapy versus usual-antimicrobial-therapy in adult cystic fibrosis pulmonary exacerbations.
METHODS: A multi-centre two-arm parallel randomised control trial conducted across Europe/North-America enrolled 223 participants (January 2015 - August 2017). All participants were chronically colonised with Pseudomonas aeruginosa and were randomised 1:1 into two study-arms. The "usual-therapy group" received 2-weeks of IV ceftazidime 3g thrice-daily (for allergies: aztreonam 2g thrice-daily) and tobramycin 5-10mg·kg[-1] once-daily. The "microbiome-directed group" received the same usual-therapy plus an additional antibiotic with greatest presumed activity against the 2nd, 3rd and 4th most abundant genera present in the sputum microbiome, selected by a Consensus Expert Treatment Panel. The primary outcome was change in percentage of predicted FEV1 (ppFEV1) at 14 days post initiation of antibiotics. Secondary outcomes examined ppFEV1 at 7 days, 28 days, and 3 months; time-to-next exacerbation; symptom burden at 7 days; Health Related Quality of Life (HRQoL) at 28 days; and number of exacerbations and IV antibiotic days at 12 months.
RESULTS: 149 participants had an eligible exacerbation (usual-therapy n=83, microbiome-directed therapy n=66). There was no difference between the groups for ppFEV1 at day 14 (-1.1%, 95%CI -3.9 to 1.7; p=0.46), or ppFEV1 measured at other time-points, or for time-to-next exacerbation (microbiome-directed versus usual-therapy Hazard Ratio 0.91 [95%CI 0.60 to 1.38; p=0.66]). The microbiome-directed group trended to have more IV days (median 42 versus 28; p=0.08) and more subsequent exacerbations (median 3 versus 2; p=0.044) the following year. There were no appreciable differences in symptom burden; however, HRQoL sub-scores were consistently worse in the microbiome-directed group (-4.3 points versus usual therapy (95%CI -8.3 to -0.3, p=0.033).
CONCLUSION: The addition of a third antibiotic based on sputum microbiome sequencing analysis did not result in improved clinical outcomes.},
}
RevDate: 2025-06-12
Milk Composition Changes and Alterations in Bacteria, Serum, and Gut Metabolome Over Time in Lactating Naks and Simmental Cows.
Animal bioscience pii:ab.25.0109 [Epub ahead of print].
OBJECTIVE: This study aimed to elucidate the mechanisms underlying milk composition divergence between naks (female yaks) and Simmental cows (S-cows) by integrating longitudinal multi-omics analyses of gut microbiota and metabolomes.
METHODS: We determined the gut microbiota and metabolites of both species over a 54-day period (day 26 to 80 of lactation), with ten naks and ten S-cows. Gut microbiota dynamics were assessed via 16S rRNA sequencing, while serum and fecal metabolomes were profiled using UHPLC-MS/MS. Statistical analyses included Wilcoxon rank-sum tests, LEfSe (LDA > 2, p < 0.05), and Spearman correlations (r > 0.70).
RESULTS: Milk yield was lesser (0.53-0.91 vs. 2.07-3.88 kg/d) but concentrations of fat (5.63-6.30% vs. 3.30-3.74%), protein (5.66-6.30% vs. 3.39-3.74%), and conjugated linoleic acid(CLA) (1.74-2.35% vs. 1.40-1.75%) were greater (p < 0.001) in nak than S-cow milk. Species-specific microbial signatures emerged. In naks, the g-Family-XIII-AD3011-group and g-norank-Ruminococcaceae were correlated with bile acid metabolism and CLA synthesis via 13-hydroxyoctadecadienoic acid transport. Additionally, the naks gut had a greater concentration of 13-hydroxyoctadecadienoic acid, a precursor of CLA, which may be transported to mammary cells via phosphatidylcholine and converted to CLA under the catalysis of fatty acid desaturase2(FADS2). S-cows harbored g-Succinivibrio and g-Eubacterium-ruminantium-group, which are linked to galactose utilization and mTOR-mediated amino acid allocation. Metabolomics revealed naks-enriched steroid biosynthesis and taurine pathways (FDR < 0.05), while S-cows exhibited a lactating network associated with greater milk yield.
CONCLUSIONS: Host-specific gut microbiota mediated nutrient allocation trade-offs. Naks optimized lipid-rich milk through bile acid and CLA metabolic networks, whereas S-cows enhanced yield via microbial-galactose synergies. This research underscores the pivotal role of the gut microbiome in mediating milk composition and suggests that microbiome manipulation could be a promising strategy to enhance milk quality in ruminants.
Additional Links: PMID-40506032
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40506032,
year = {2025},
author = {Wang, R and Yunxiang, X and Degen, A and Han, X and Zhao, X and Zhang, Q and Huang, Y and Bai, B and Yang, Y and Liu, S and Xue, Y and Hao, L},
title = {Milk Composition Changes and Alterations in Bacteria, Serum, and Gut Metabolome Over Time in Lactating Naks and Simmental Cows.},
journal = {Animal bioscience},
volume = {},
number = {},
pages = {},
doi = {10.5713/ab.25.0109},
pmid = {40506032},
issn = {2765-0189},
abstract = {OBJECTIVE: This study aimed to elucidate the mechanisms underlying milk composition divergence between naks (female yaks) and Simmental cows (S-cows) by integrating longitudinal multi-omics analyses of gut microbiota and metabolomes.
METHODS: We determined the gut microbiota and metabolites of both species over a 54-day period (day 26 to 80 of lactation), with ten naks and ten S-cows. Gut microbiota dynamics were assessed via 16S rRNA sequencing, while serum and fecal metabolomes were profiled using UHPLC-MS/MS. Statistical analyses included Wilcoxon rank-sum tests, LEfSe (LDA > 2, p < 0.05), and Spearman correlations (r > 0.70).
RESULTS: Milk yield was lesser (0.53-0.91 vs. 2.07-3.88 kg/d) but concentrations of fat (5.63-6.30% vs. 3.30-3.74%), protein (5.66-6.30% vs. 3.39-3.74%), and conjugated linoleic acid(CLA) (1.74-2.35% vs. 1.40-1.75%) were greater (p < 0.001) in nak than S-cow milk. Species-specific microbial signatures emerged. In naks, the g-Family-XIII-AD3011-group and g-norank-Ruminococcaceae were correlated with bile acid metabolism and CLA synthesis via 13-hydroxyoctadecadienoic acid transport. Additionally, the naks gut had a greater concentration of 13-hydroxyoctadecadienoic acid, a precursor of CLA, which may be transported to mammary cells via phosphatidylcholine and converted to CLA under the catalysis of fatty acid desaturase2(FADS2). S-cows harbored g-Succinivibrio and g-Eubacterium-ruminantium-group, which are linked to galactose utilization and mTOR-mediated amino acid allocation. Metabolomics revealed naks-enriched steroid biosynthesis and taurine pathways (FDR < 0.05), while S-cows exhibited a lactating network associated with greater milk yield.
CONCLUSIONS: Host-specific gut microbiota mediated nutrient allocation trade-offs. Naks optimized lipid-rich milk through bile acid and CLA metabolic networks, whereas S-cows enhanced yield via microbial-galactose synergies. This research underscores the pivotal role of the gut microbiome in mediating milk composition and suggests that microbiome manipulation could be a promising strategy to enhance milk quality in ruminants.},
}
RevDate: 2025-06-12
Investigating acupuncture therapy in depression: mechanisms of synaptic plasticity regulation.
Neuroscience pii:S0306-4522(25)00687-6 [Epub ahead of print].
Depression is a severe heterogeneous mental illness that is highly co-morbid with other mental and somatic disorders. It poses a significant healthcare burden on both individuals and society. Currently, the use of single-target antidepressants exhibits suboptimal efficacy with significant adverse effects. Acupuncture has been advocated as a practical and effective treatment for depression, due to its low adverse effects rate compared to antidepressant medication. Currently, several studies have shown that acupuncture treatment for depression primarily involves multiple therapeutic mechanisms, including the regulation of specific gene expression, neuropeptide and neurotransmitter release, increasing the expression of neurotrophic factors, suppressing hyperactivity of the hypothalamic-pituitary-adrenal (HPA) axis, attenuating inflammatory responses, and restoring gut microbiota balance. These therapeutic effects involve the regulation of critical signaling pathways, including the cAMP-responsive element binding protein (CREB) signaling pathway, mitogen-activated protein kinases (MAPK) signaling pathway, mechanistic target of rapamycin (mTOR) signaling pathway, and toll-like receptors (TLR) signaling pathway. Notably, depression-associated molecular mechanisms and signaling pathway dysregulations are closely linked to impaired neural and synaptic plasticity. Acupuncture synergistically modulates the neuro-immune-microbiome multidimensional network and integrates crosstalk among key pathways such as CREB, thereby systemically restoring synaptic plasticity. This multi-dimensional integrative mechanism likely underlies its therapeutic superiority over single-target antidepressants. This review aims to elucidate how acupuncture restores cerebral synaptic plasticity by rectifying depression-related systemic dysfunctions and signaling pathway abnormalities, which will advance our understanding of its regulatory potential in depression treatment and inform the development of precision therapeutic strategies.
Additional Links: PMID-40506009
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40506009,
year = {2025},
author = {Si, Y and Ma, W and Zhang, Q and Zhang, Y and An, J and Zhang, M and Fu, Y and Yu, Y and Zhang, H and Fang, Y and Zhang, D},
title = {Investigating acupuncture therapy in depression: mechanisms of synaptic plasticity regulation.},
journal = {Neuroscience},
volume = {},
number = {},
pages = {},
doi = {10.1016/j.neuroscience.2025.06.005},
pmid = {40506009},
issn = {1873-7544},
abstract = {Depression is a severe heterogeneous mental illness that is highly co-morbid with other mental and somatic disorders. It poses a significant healthcare burden on both individuals and society. Currently, the use of single-target antidepressants exhibits suboptimal efficacy with significant adverse effects. Acupuncture has been advocated as a practical and effective treatment for depression, due to its low adverse effects rate compared to antidepressant medication. Currently, several studies have shown that acupuncture treatment for depression primarily involves multiple therapeutic mechanisms, including the regulation of specific gene expression, neuropeptide and neurotransmitter release, increasing the expression of neurotrophic factors, suppressing hyperactivity of the hypothalamic-pituitary-adrenal (HPA) axis, attenuating inflammatory responses, and restoring gut microbiota balance. These therapeutic effects involve the regulation of critical signaling pathways, including the cAMP-responsive element binding protein (CREB) signaling pathway, mitogen-activated protein kinases (MAPK) signaling pathway, mechanistic target of rapamycin (mTOR) signaling pathway, and toll-like receptors (TLR) signaling pathway. Notably, depression-associated molecular mechanisms and signaling pathway dysregulations are closely linked to impaired neural and synaptic plasticity. Acupuncture synergistically modulates the neuro-immune-microbiome multidimensional network and integrates crosstalk among key pathways such as CREB, thereby systemically restoring synaptic plasticity. This multi-dimensional integrative mechanism likely underlies its therapeutic superiority over single-target antidepressants. This review aims to elucidate how acupuncture restores cerebral synaptic plasticity by rectifying depression-related systemic dysfunctions and signaling pathway abnormalities, which will advance our understanding of its regulatory potential in depression treatment and inform the development of precision therapeutic strategies.},
}
RevDate: 2025-06-12
Gut Microbiota and Behavioral Ontogeny in Autism Spectrum Disorder: A Pathway to Therapeutic Innovations.
Physiology & behavior pii:S0031-9384(25)00190-8 [Epub ahead of print].
Autism Spectrum Disorder (ASD) is a multifaceted neurodevelopmental condition characterized by deficits in social communication, repetitive behaviors, and restricted interests. Emerging evidence suggests that gut-brain axis a dynamic, bidirectional communication network between gut microbiota and central nervous system, is critical in shaping behavioral ontogeny in ASD. Dysbiosis of gut microbiota, commonly observed in individuals with ASD, has been associated with alterations in neurodevelopmental trajectories and symptom severity. Furthermore, disturbances in maternal microbiome during pregnancy are increasingly recognized as key factors influencing fetal brain development, potentially heightening risk of ASD and behavioral manifestations. Mechanistic research reveals that gut-derived metabolites modulate blood-brain barrier integrity, neuroinflammatory processes, and neuronal circuit formation, contributing to behavioral outcomes. These findings emphasize gut microbiota's profound influence on emergence and progression of ASD-related behaviors. Promising therapeutic strategies, including probiotics, prebiotics, fecal microbiota transplantation, and dietary interventions, have demonstrated potential in modulating the gut microbiome and improving behavioral symptoms in ASD. However, challenges such as individual variability in microbiome composition, limited clinical evidence, and an incomplete understanding of causative mechanisms remain significant barriers to clinical translation. This review explores the interplay between gut microbiota and ASD-associated behaviors, focusing on key mechanisms such as microbial regulation of neurotransmitter production, immune signaling, and neuroinflammation. It further highlights gut microbiota's potential as a modifiable factor influencing neurodevelopmental and behavioral outcomes in ASD. By advancing our understanding of gut-brain axis, we can pave the way for personalized and targeted interventions aimed at improving behavioral ontogeny and developmental trajectories in individuals with ASD.
Additional Links: PMID-40505847
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40505847,
year = {2025},
author = {Yadav, A and Tadas, M and Kale, M and Wankhede, N and Umekar, M and Kotagale, N and Taksande, B},
title = {Gut Microbiota and Behavioral Ontogeny in Autism Spectrum Disorder: A Pathway to Therapeutic Innovations.},
journal = {Physiology & behavior},
volume = {},
number = {},
pages = {114989},
doi = {10.1016/j.physbeh.2025.114989},
pmid = {40505847},
issn = {1873-507X},
abstract = {Autism Spectrum Disorder (ASD) is a multifaceted neurodevelopmental condition characterized by deficits in social communication, repetitive behaviors, and restricted interests. Emerging evidence suggests that gut-brain axis a dynamic, bidirectional communication network between gut microbiota and central nervous system, is critical in shaping behavioral ontogeny in ASD. Dysbiosis of gut microbiota, commonly observed in individuals with ASD, has been associated with alterations in neurodevelopmental trajectories and symptom severity. Furthermore, disturbances in maternal microbiome during pregnancy are increasingly recognized as key factors influencing fetal brain development, potentially heightening risk of ASD and behavioral manifestations. Mechanistic research reveals that gut-derived metabolites modulate blood-brain barrier integrity, neuroinflammatory processes, and neuronal circuit formation, contributing to behavioral outcomes. These findings emphasize gut microbiota's profound influence on emergence and progression of ASD-related behaviors. Promising therapeutic strategies, including probiotics, prebiotics, fecal microbiota transplantation, and dietary interventions, have demonstrated potential in modulating the gut microbiome and improving behavioral symptoms in ASD. However, challenges such as individual variability in microbiome composition, limited clinical evidence, and an incomplete understanding of causative mechanisms remain significant barriers to clinical translation. This review explores the interplay between gut microbiota and ASD-associated behaviors, focusing on key mechanisms such as microbial regulation of neurotransmitter production, immune signaling, and neuroinflammation. It further highlights gut microbiota's potential as a modifiable factor influencing neurodevelopmental and behavioral outcomes in ASD. By advancing our understanding of gut-brain axis, we can pave the way for personalized and targeted interventions aimed at improving behavioral ontogeny and developmental trajectories in individuals with ASD.},
}
RevDate: 2025-06-12
Gut microbiome contributes to 6PPD-Quinone induced cognitive impairment through PI3K/Akt signaling.
Toxicology pii:S0300-483X(25)00176-3 [Epub ahead of print].
Studies show that N-(1,3-dimethylbutyl)-N'-phenyl-p-phenylenediamine-quinone (6PPD-Q) accumulates in the central nervous system, but its role in cognitive impairment and underlying mechanisms remain unclear. Morris water maze assay revealed that 6-PPDQ significantly impairs cognitive function, particularly learning and memory. HE staining revealed alterations in the hippocampal DG and CA3 regions of exposed mice, including sparse cell arrangement, blurred boundaries, nuclear condensation, and a reduction in Nissl bodies. Fecal microbiota transplantation from 6-PPDQ-exposed mice to normal mice induced cognitive deficits and hippocampal pathological damage. Western Blot assay showed that 6-PPDQ exposure resulted in inhibition of PI3K/AKT signaling. Moreover, blunted PI3K/AKT signaling was observed in mice transplanted with 6-PPDQ-associated mice fecal microbiota. Further analysis of 16S rDNA assay identified a total of 30 differential bacteria at the genus level, including 8 upregulated bacteria such as g_Helicobacter and 22 downregulated bacteria such as g_Prevotellaceae_NK3B31_group. In conclusion, this study uncovers gut microbiome mediates 6PPD-Q-induced cognitive impairment through inhibiting of PI3K/Akt signaling, and provides a basis for further investigation into gut microbiome's protective effects on 6-PPDQ-induced nervous system injury.
Additional Links: PMID-40505719
Publisher:
PubMed:
Citation:
show bibtex listing
hide bibtex listing
@article {pmid40505719,
year = {2025},
author = {Zhu, X and Xu, Y},
title = {Gut microbiome contributes to 6PPD-Quinone induced cognitive impairment through PI3K/Akt signaling.},
journal = {Toxicology},
volume = {},
number = {},
pages = {154217},
doi = {10.1016/j.tox.2025.154217},
pmid = {40505719},
issn = {1879-3185},
abstract = {Studies show that N-(1,3-dimethylbutyl)-N'-phenyl-p-phenylenediamine-quinone (6PPD-Q) accumulates in the central nervous system, but its role in cognitive impairment and underlying mechanisms remain unclear. Morris water maze assay revealed that 6-PPDQ significantly impairs cognitive function, particularly learning and memory. HE staining revealed alterations in the hippocampal DG and CA3 regions of exposed mice, including sparse cell arrangement, blurred boundaries, nuclear condensation, and a reduction in Nissl bodies. Fecal microbiota transplantation from 6-PPDQ-exposed mice to normal mice induced cognitive deficits and hippocampal pathological damage. Western Blot assay showed that 6-PPDQ exposure resulted in inhibition of PI3K/AKT signaling. Moreover, blunted PI3K/AKT signaling was observed in mice transplanted with 6-PPDQ-associated mice fecal microbiota. Further analysis of 16S rDNA assay identified a total of 30 differential bacteria at the genus level, including 8 upregulated bacteria such as g_Helicobacter and 22 downregulated bacteria such as g_Prevotellaceae_NK3B31_group. In conclusion, this study uncovers gut microbiome mediates 6PPD-Q-induced cognitive impairment through inhibiting of PI3K/Akt signaling, and provides a basis for further investigation into gut microbiome's protective effects on 6-PPDQ-induced nervous system injury.},
}
▼ ▼ LOAD NEXT 100 CITATIONS
ESP Quick Facts
ESP Origins
In the early 1990's, Robert Robbins was a faculty member at Johns Hopkins, where he directed the informatics core of GDB — the human gene-mapping database of the international human genome project. To share papers with colleagues around the world, he set up a small paper-sharing section on his personal web page. This small project evolved into The Electronic Scholarly Publishing Project.
ESP Support
In 1995, Robbins became the VP/IT of the Fred Hutchinson Cancer Research Center in Seattle, WA. Soon after arriving in Seattle, Robbins secured funding, through the ELSI component of the US Human Genome Project, to create the original ESP.ORG web site, with the formal goal of providing free, world-wide access to the literature of classical genetics.
ESP Rationale
Although the methods of molecular biology can seem almost magical to the uninitiated, the original techniques of classical genetics are readily appreciated by one and all: cross individuals that differ in some inherited trait, collect all of the progeny, score their attributes, and propose mechanisms to explain the patterns of inheritance observed.
ESP Goal
In reading the early works of classical genetics, one is drawn, almost inexorably, into ever more complex models, until molecular explanations begin to seem both necessary and natural. At that point, the tools for understanding genome research are at hand. Assisting readers reach this point was the original goal of The Electronic Scholarly Publishing Project.
ESP Usage
Usage of the site grew rapidly and has remained high. Faculty began to use the site for their assigned readings. Other on-line publishers, ranging from The New York Times to Nature referenced ESP materials in their own publications. Nobel laureates (e.g., Joshua Lederberg) regularly used the site and even wrote to suggest changes and improvements.
ESP Content
When the site began, no journals were making their early content available in digital format. As a result, ESP was obliged to digitize classic literature before it could be made available. For many important papers — such as Mendel's original paper or the first genetic map — ESP had to produce entirely new typeset versions of the works, if they were to be available in a high-quality format.
ESP Help
Early support from the DOE component of the Human Genome Project was critically important for getting the ESP project on a firm foundation. Since that funding ended (nearly 20 years ago), the project has been operated as a purely volunteer effort. Anyone wishing to assist in these efforts should send an email to Robbins.
ESP Plans
With the development of methods for adding typeset side notes to PDF files, the ESP project now plans to add annotated versions of some classical papers to its holdings. We also plan to add new reference and pedagogical material. We have already started providing regularly updated, comprehensive bibliographies to the ESP.ORG site.
ESP Picks from Around the Web (updated 28 JUL 2024 )
Old Science
Weird Science
Treating Disease with Fecal Transplantation
Fossils of miniature humans (hobbits) discovered in Indonesia
Paleontology
Dinosaur tail, complete with feathers, found preserved in amber.
Astronomy
Mysterious fast radio burst (FRB) detected in the distant universe.
Big Data & Informatics
Big Data: Buzzword or Big Deal?
Hacking the genome: Identifying anonymized human subjects using publicly available data.