1
|
Ma X, Duan C, Wang X, Tao Y, Yang L, Teng Y, Pan Y, Zhang M, Xu J, Sheng J, Wang X, Jin P. Human gut microbiota adaptation to high-altitude exposure: longitudinal analysis over acute and prolonged periods. Microbiol Spectr 2025:e0291624. [PMID: 40257273 DOI: 10.1128/spectrum.02916-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 03/21/2025] [Indexed: 04/22/2025] Open
Abstract
This study investigated the longitudinal effects of acute (7-day) and prolonged (3-month) high-altitude exposure on gut microbiota in healthy adult males, addressing the limited data available in human populations. A cohort of 406 healthy adult males was followed, and fecal samples were collected at three time points: baseline at 800 m (406 samples), 7 days after ascending to 4,500 m (406 samples), and 2 weeks post-return to 800 m following 3 months at high altitude (186 samples). High-throughput 16S ribosomal DNA sequencing was employed to analyze microbiota composition and diversity. Results revealed significant changes in alpha- and beta-diversity, with acute high-altitude exposure inducing more pronounced effects compared to prolonged exposure. Specifically, acute exposure increased opportunistic pathogens (Ruminococcus and Oscillibacter) but decreased beneficial short-chain fatty acid producers (Faecalibacterium and Bifidobacterium). Notably, these changes in microbiota persisted even after returning to low altitude, indicating long-term remodeling. Functional analyses revealed substantial changes in metabolic pathways, suggesting microbiota-driven adaptations to energy utilization under high-altitude hypoxic conditions. In summary, acute high-altitude exposure caused dramatic changes in gut microbiota, while prolonged exposure led to structural and functional reshaping. These findings enhance our understanding of how high-altitude environments reshape gut microbiota. IMPORTANCE This study is the first to investigate the impact of high-altitude exposure on gut microbiota adaptation in a large-scale longitudinal cohort. It seeks to enhance understanding of how high-altitude environments reshape gut microbiota. Acute exposure to high altitude significantly affected both α-diversity and β-diversity of gut microbiota, with acute exposure causing more pronounced changes than prolonged adaptation, indicating temporary disruptions in microbial communities. Notable shifts in microbial abundance were observed, including increased levels of genera linked to hypoxic stress (e.g., Gemmiger, Ruminococcus, and Parabacteroides) and decreased levels of beneficial bacteria (e.g., Faecalibacterium, Roseburia, and Bifidobacterium), suggesting possible adverse health effects. Functional analysis indicated changes in metabolism-related pathways post-exposure, supporting the idea that high-altitude adaptations involve metabolic adjustments for energy management. These findings enhance understanding of high-altitude physiology, illustrating the role of gut microbiota in hypoxic health.
Collapse
Affiliation(s)
- Xianzong Ma
- Senior Department of Gastroenterology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
- Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| | | | - Xiaoying Wang
- Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yurong Tao
- Senior Department of Gastroenterology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
- Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Lang Yang
- Senior Department of Gastroenterology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
- Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yongsheng Teng
- Department of Gastroenterology, Chongqing General Hospital, Chongqing University, Chongqing, China
| | - Yuanming Pan
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Mingjie Zhang
- Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Junfeng Xu
- Senior Department of Gastroenterology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Jianqiu Sheng
- Senior Department of Gastroenterology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
- Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
- Medical School of Chinese PLA, Beijing, China
| | - Xin Wang
- Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Peng Jin
- Senior Department of Gastroenterology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
- Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
- Medical School of Chinese PLA, Beijing, China
| |
Collapse
|
2
|
Umar S, Yu W, Xuan H, Ahmed I, Zhong C, Morowitz M, Rogers MB, Attard MI, Sampath V. Neonatal gut microbiota succession in mice mapped over time, site, injury and single immunoglobulin interleukin-1 related receptor genotype. iScience 2025; 28:112243. [PMID: 40248118 PMCID: PMC12005339 DOI: 10.1016/j.isci.2025.112243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 01/23/2025] [Accepted: 03/13/2025] [Indexed: 04/19/2025] Open
Abstract
Microbial succession during postnatal gut development in mice is likely impacted by site of sampling, time, intestinal injury, and host genetics. We investigated this in wild-type and Sigirr transgenic mice that encode the p.Y168X mutation identified in a neonate with necrotizing enterocolitis (NEC). Temporal profiling of the ileal and colonic microbiome after birth to weaning revealed a clear pattern of progression from a less diverse, Proteobacteria/Escherichia_Shigella dominant community to a more diverse, Firmicutes/Bacteroidetes dominant community. Formula milk feeding, a risk factor for necrotizing enterocolitis, decreased Firmicutes and increased Proteobacteria leading to enrichment of bacterial genes denoting exaggerated glycolysis and increased production of acetate and lactate. Sigirr transgenic mice exhibited modest baseline differences in microbiota composition but exaggerated formula feeding-induced dysbiosis, mucosal inflammation, and villus injury. Postnatal intestinal microbiota succession in mice resembles human neonates and is shaped by developmental maturity, ileal vs. colonic sampling, formula feeding, and Sigirr genotype.
Collapse
Affiliation(s)
- Shahid Umar
- Department of Surgery, University of Kansas Medical Center, USA
| | - Wei Yu
- Department of Pediatrics/Neonatology, Children’s Mercy Hospital, Kansas City, USA
| | - Hao Xuan
- Department of Electrical Engineering and Computer Science, University of Kansas, USA
| | - Ishfaq Ahmed
- Department of Math, Science and Computer Technology, Kansas City Community College, USA
| | - Cuncong Zhong
- Department of Electrical Engineering and Computer Science, University of Kansas, USA
| | - Michael Morowitz
- Division of Pediatric General and Thoracic Surgery, University of Pittsburgh Children’s Hospital, Pittsburgh, PA, USA
| | - Mathew Brian Rogers
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, Canada
| | - Mark Ivan Attard
- Neonatal Unit, Aberdeen Maternity Hospital, Aberdeen AB25 2ZL, UK
| | - Venkatesh Sampath
- Department of Electrical Engineering and Computer Science, University of Kansas, USA
| |
Collapse
|
3
|
Salido RA, Zhao HN, McDonald D, Mannochio-Russo H, Zuffa S, Oles RE, Aron AT, El Abiead Y, Farmer S, González A, Martino C, Mohanty I, Parker CW, Patel L, Portal Gomes PW, Schmid R, Schwartz T, Zhu J, Barratt MR, Rubins KH, Chu H, Karouia F, Venkateswaran K, Dorrestein PC, Knight R. The International Space Station has a unique and extreme microbial and chemical environment driven by use patterns. Cell 2025; 188:2022-2041.e23. [PMID: 40020666 DOI: 10.1016/j.cell.2025.01.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 10/17/2024] [Accepted: 01/28/2025] [Indexed: 03/03/2025]
Abstract
Space habitation provides unique challenges in built environments isolated from Earth. We produced a 3D map of the microbes and metabolites throughout the United States Orbital Segment (USOS) of the International Space Station (ISS) with 803 samples collected during space flight, including controls. We find that the use of each of the nine sampled modules within the ISS strongly drives the microbiology and chemistry of the habitat. Relating the microbiology to other Earth habitats, we find that, as with human microbiota, built environment microbiota also align naturally along an axis of industrialization, with the ISS providing an extreme example of an industrialized environment. We demonstrate the utility of culture-independent sequencing for microbial risk monitoring, especially as the location of sequencing moves to space. The resulting resource of chemistry and microbiology in the space-built environment will guide long-term efforts to maintain human health in space for longer durations.
Collapse
Affiliation(s)
- Rodolfo A Salido
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | - Haoqi Nina Zhao
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Daniel McDonald
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
| | - Helena Mannochio-Russo
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Simone Zuffa
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Renee E Oles
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA; Department of Pathology, University of California, San Diego, La Jolla, CA, USA
| | - Allegra T Aron
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA; Department of Chemistry and Biochemistry, University of Denver, Denver, CO, USA
| | - Yasin El Abiead
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Sawyer Farmer
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
| | - Antonio González
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
| | - Cameron Martino
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
| | - Ipsita Mohanty
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Ceth W Parker
- Biotechnology and Planetary Protection Group, Jet Propulsion Laboratory, California Institute of Technology, Pasadena, CA, USA
| | - Lucas Patel
- Bioinformatics and Systems Biology Program, University of California, San Diego, La Jolla, CA, USA; Medical Scientist Training Program, University of California, San Diego, La Jolla, CA, USA
| | - Paulo Wender Portal Gomes
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Robin Schmid
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Tara Schwartz
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
| | - Jennifer Zhu
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
| | | | | | - Hiutung Chu
- Department of Pathology, University of California, San Diego, La Jolla, CA, USA; Chiba University-UC San Diego Center for Mucosal Immunology, Allergy and Vaccines (CU-UCSD cMAV), La Jolla, CA, USA; Center for Microbiome Innovation, University of California, San Diego, La Jolla, CA, USA
| | - Fathi Karouia
- Blue Marble Space Institute of Science, Exobiology Branch, NASA Ames Research Center, Moffett Field, CA, USA; Space Research Within Reach, San Francisco, CA, USA; Center for Space Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Kasthuri Venkateswaran
- Biotechnology and Planetary Protection Group, Jet Propulsion Laboratory, California Institute of Technology, Pasadena, CA, USA
| | - Pieter C Dorrestein
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA; Center for Microbiome Innovation, University of California, San Diego, La Jolla, CA, USA; Department of Pharmacology, University of California, San Diego, La Jolla, CA, USA; Collaborative Mass Spectrometry Innovation Center, University of California, San Diego, La Jolla, CA, USA.
| | - Rob Knight
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA; Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA; Center for Microbiome Innovation, University of California, San Diego, La Jolla, CA, USA; Department of Computer Science and Engineering, University of California, San Diego, La Jolla, CA, USA; Halıcıoğlu Data Science Institute, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
4
|
Wang J, Yin J, Liu X, Liu Y, Jin X. Gut commensal bacterium Bacteroides vulgatus exacerbates helminth-induced cardiac fibrosis through succinate accumulation. PLoS Pathog 2025; 21:e1013069. [PMID: 40238740 PMCID: PMC12002503 DOI: 10.1371/journal.ppat.1013069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Accepted: 02/21/2025] [Indexed: 04/18/2025] Open
Abstract
Trichinella spiralis (Ts) is known to cause cardiac fibrosis, which is a critical precursor to various heart diseases, and its progression is influenced by metabolic changes. However, the metabolic mechanisms remain unclear. Here, we observed that Ts-infected mice exhibited cardiac fibrosis along with elevated succinate levels in the heart using metabolomic analysis. Administration of succinate exacerbated fibrosis during Ts infection, while deficiency in succinate receptor 1 (Sucnr1) alleviated the condition, highlighting the role of the succinate-Sucnr1 axis in fibrosis development. Furthermore, metagenomics sequencing showed that Ts-infected mice had a higher abundance ratio of succinate-producing bacteria to succinate-consuming bacteria in the intestines. Notably, the succinate-producer Bacteroides vulgatus was enriched in Ts group. Oral supplementation with B. vulgatus aggravated Ts-induced cardiac fibrosis. In summary, our findings underscore the succinate-Sucnr1 axis as a critical pathway in helminth-induced cardiac fibrosis and highlight the potential of targeting this axis for therapeutic interventions. This study presents novel insights into the gut-heart axis, revealing innovative strategies for managing cardiovascular complications associated with helminth infections.
Collapse
Affiliation(s)
- Jiaqi Wang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
- College of Animal Sciences, Jilin University, Changchun, China
| | - Jiali Yin
- The Second Hospital of Jilin University, Changchun, China
| | - Xiaolei Liu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Yi Liu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
- College of Food Science and Engineering, Jilin University, Changchun, China
| | - Xuemin Jin
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| |
Collapse
|
5
|
Gudnadottir U, Fransson E, Ljungman G, Wikman A, Vlieghe E, Engstrand L, Brusselaers N. Prenatal and Early Childhood Exposure to Proton Pump Inhibitors and Antibiotics and the Risk of Childhood Cancer: A Nationwide Population-Based Cohort Study. Drug Saf 2025; 48:375-388. [PMID: 39666165 PMCID: PMC11903606 DOI: 10.1007/s40264-024-01500-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/14/2024] [Indexed: 12/13/2024]
Abstract
BACKGROUND Our microbiome is established during infancy, a time important for later health and long-term effects. Proton pump inhibitors and antibiotics are regularly prescribed during pregnancy. Both drugs cause microbiome disturbance and have been associated with increased cancer risk in adults, but effects of these drugs on the growing foetus and infant remain understudied. AIM The aim of this study is to study the association between prenatal and early life proton pump inhibitor and antibiotics exposure and the risk of childhood cancer. METHODS This study is a retrospective population-based cohort design, using registry data on all births (n = 722,372) in Sweden between 2006 and 2016, according to the STROBE checklist. For women who had multiple children in the timeframe of the study, only the first child during the time period was included in the cohort. Exposure was defined as either ≥ 1 proton pump inhibitor or antibiotics prescription during pregnancy, or during the first 2 years of life. Outcome was defined as cancer at any time during the follow-up or cancer after the age of 2 years for early life exposure. Multivariable Cox proportional hazard models were used to calculate hazard ratios. RESULTS In total, 1091 (0.2%) children were diagnosed with malignant cancer during the follow-up. Prenatal exposure to proton pump inhibitors and antibiotics were not associated with an increased risk of cancer. Regarding early life exposure, proton pump inhibitors were associated with an increased risk of cancer at age two or older (adjusted hazard ratio [aHR] 3.68, 95% confidence interval [CI] 2.24-6.06). CONCLUSIONS We did not find evidence that prenatal proton pump inhibitors and antibiotics were associated with overall childhood cancer. However, proton pump inhibitors during early life were associated with an increased risk of childhood cancer, but indication on drug use was not available and confounding by indication may be present.
Collapse
Affiliation(s)
- Unnur Gudnadottir
- Department of Microbiology, Tumour and Cell Biology (MTC), Centre for Translational Microbiome Research, Karolinska Institutet, Tomtebodavägen 16, Solna, 171 65, Stockholm, Sweden.
| | - Emma Fransson
- Department of Microbiology, Tumour and Cell Biology (MTC), Centre for Translational Microbiome Research, Karolinska Institutet, Tomtebodavägen 16, Solna, 171 65, Stockholm, Sweden
- Department of Women's and Children's health, Uppsala University, Sjukhusvägen 7, 753 09, Uppsala, Sweden
| | - Gustaf Ljungman
- Department of Women's and Children's health, Uppsala University, Sjukhusvägen 7, 753 09, Uppsala, Sweden
| | - Anna Wikman
- Department of Women's and Children's health, Uppsala University, Sjukhusvägen 7, 753 09, Uppsala, Sweden
| | - Erika Vlieghe
- Department of Family Medicine and Population Health, Global Health Institute, University of Antwerp, 2610, Antwerp, Belgium
| | - Lars Engstrand
- Department of Microbiology, Tumour and Cell Biology (MTC), Centre for Translational Microbiome Research, Karolinska Institutet, Tomtebodavägen 16, Solna, 171 65, Stockholm, Sweden
| | - Nele Brusselaers
- Department of Microbiology, Tumour and Cell Biology (MTC), Centre for Translational Microbiome Research, Karolinska Institutet, Tomtebodavägen 16, Solna, 171 65, Stockholm, Sweden
- Department of Family Medicine and Population Health, Global Health Institute, University of Antwerp, 2610, Antwerp, Belgium
- Department of Public Health and Primary Care, Ghent University, 9000, Ghent, Belgium
| |
Collapse
|
6
|
Abdolmaleky HM, Nohesara S, Zhou JR, Thiagalingam S. Epigenetics in evolution and adaptation to environmental challenges: pathways for disease prevention and treatment. Epigenomics 2025; 17:317-333. [PMID: 39948759 PMCID: PMC11970782 DOI: 10.1080/17501911.2025.2464529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 02/04/2025] [Indexed: 04/02/2025] Open
Abstract
Adaptation to challenging environmental conditions is crucial for the survival/fitness of all organisms. Alongside genetic mutations that provide adaptive potential during environmental challenges, epigenetic modifications offer dynamic, reversible, and rapid mechanisms for regulating gene expression in response to environmental changes in both evolution and daily life, without altering DNA sequences or relying on accidental favorable mutations. The widespread conservation of diverse epigenetic mechanisms - like DNA methylation, histone modifications, and RNA interference across diverse species, including plants - underscores their significance in evolutionary biology. Remarkably, environmentally induced epigenetic alterations are passed to daughter cells and inherited transgenerationally through germline cells, shaping offspring phenotypes while preserving adaptive epigenetic memory. Throughout anthropoid evolution, epigenetic modifications have played crucial roles in: i) suppressing transposable elements and viral genomes intruding into the host genome; ii) inactivating one of the X chromosomes in female cells to balance gene dosage; iii) genetic imprinting to ensure expression from one parental allele; iv) regulating functional alleles to compensate for dysfunctional ones; and v) modulating the epigenome and transcriptome in response to influence from the gut microbiome among other functions. Understanding the interplay between environmental factors and epigenetic processes may provide valuable insights into developmental plasticity, evolutionary dynamics, and disease susceptibility.
Collapse
Affiliation(s)
- Hamid Mostafavi Abdolmaleky
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Medicine, Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Shabnam Nohesara
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Jin-Rong Zhou
- Nutrition/Metabolism Laboratory, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Sam Thiagalingam
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Pathology & Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| |
Collapse
|
7
|
Yang J, Cha Y, Oh SY. Habitat prevails over host sex in influencing mycobiome structure of terrestrial isopod, Armadillidium vulgare. Microbiol Spectr 2025:e0217224. [PMID: 40162752 DOI: 10.1128/spectrum.02172-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 03/07/2025] [Indexed: 04/02/2025] Open
Abstract
The terrestrial isopod, a crustacean order that has successfully transitioned from aquatic to land ecosystem, functions as a soil bioengineer and plays a crucial role in ecological decomposition. While there has been comprehensive documentation of bacterial associations with isopods, suggesting their contribution to digestive processes, the fungal component of isopods remains a less-explored dimension. Expanding our fundamental exploration into terrestrial isopods, we investigated whether the fungal community in Armadillidium vulgare is more significantly influenced by host sex or habitat using high-throughput sequencing-based internal transcribed spacer region amplification. Our findings revealed that the isopod mycobiome structure and taxonomic composition are predominantly shaped by the host's habitat rather than its sex. While the fungal richness of the mycobiome varied based on isopod sex, the co-occurrence network exhibited distinct variations dependent on both habitat and host sex. Finally, we observed a complete overlap of core mycobiomes based on isopod sex and identified fungal residents that are consistently distributed in a manner specific to habitat. In conclusion, we assert that community assembly of the isopod mycobiome in an untouched ecosystem is more significantly influenced by location than sex. We contend that this offers a fundamental groundwork, laying the cornerstone for future investigations into terrestrial isopods-pivotal agents in ecological decomposition within ecosystems.IMPORTANCEThis study addresses a significant knowledge gap in the mycobiome of terrestrial isopods, an area that has received limited scientific attention despite extensive research on bacterial associations within these organisms. Using high-throughput sequencing, this study demonstrates that the habitat of Armadillidium vulgare exerts a more pronounced influence on the composition of its mycobiome compared with host sex. By examining variations in community structure, diversity, co-occurrence patterns, and identifying core mycobiomes and specialist taxa based on isopod location, this study provides crucial foundational data. These findings are essential for advancing future research on the ecological and evolutionary dynamics of fungal communities in terrestrial isopods.
Collapse
Affiliation(s)
- Jiho Yang
- Gyeongnam Bio and Anti-aging Core Facility Center, Changwon National University, Changwon, South Korea
| | - Yehyeon Cha
- Department of Biology and Microbiology, Changwon National University, Changwon, South Korea
| | - Seung-Yoon Oh
- Gyeongnam Bio and Anti-aging Core Facility Center, Changwon National University, Changwon, South Korea
- Department of Biology and Microbiology, Changwon National University, Changwon, South Korea
- Department of Biology and Chemistry, Changwon National University, Changwon, South Korea
| |
Collapse
|
8
|
McDonnell KJ. Operationalizing Team Science at the Academic Cancer Center Network to Unveil the Structure and Function of the Gut Microbiome. J Clin Med 2025; 14:2040. [PMID: 40142848 PMCID: PMC11943358 DOI: 10.3390/jcm14062040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 02/28/2025] [Accepted: 03/05/2025] [Indexed: 03/28/2025] Open
Abstract
Oncologists increasingly recognize the microbiome as an important facilitator of health as well as a contributor to disease, including, specifically, cancer. Our knowledge of the etiologies, mechanisms, and modulation of microbiome states that ameliorate or promote cancer continues to evolve. The progressive refinement and adoption of "omic" technologies (genomics, transcriptomics, proteomics, and metabolomics) and utilization of advanced computational methods accelerate this evolution. The academic cancer center network, with its immediate access to extensive, multidisciplinary expertise and scientific resources, has the potential to catalyze microbiome research. Here, we review our current understanding of the role of the gut microbiome in cancer prevention, predisposition, and response to therapy. We underscore the promise of operationalizing the academic cancer center network to uncover the structure and function of the gut microbiome; we highlight the unique microbiome-related expert resources available at the City of Hope of Comprehensive Cancer Center as an example of the potential of team science to achieve novel scientific and clinical discovery.
Collapse
Affiliation(s)
- Kevin J McDonnell
- Center for Precision Medicine, Department of Medical Oncology & Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| |
Collapse
|
9
|
Deng AQ, Yue SY, Niu D, Zhang DD, Hou BB, Zhang L, Liang CZ, Du HX. The role of microbiota in the chronic prostatitis/chronic pelvis pain syndrome: a review. Front Microbiol 2025; 16:1488732. [PMID: 40143861 PMCID: PMC11937130 DOI: 10.3389/fmicb.2025.1488732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 02/24/2025] [Indexed: 03/28/2025] Open
Abstract
Chronic prostatitis/Chronic pelvis pain syndrome (CP/CPPS), a kind of frequent urinary condition among adult males, has caused a lot of inconvenience to patients in life, whose pathogenesis is unclear. Current evidence suggests that it is most likely to be an autoimmune disease. Symbiotic microbes, a highly diverse biological community that harbors trillions of microbes in each region of the human body, have gradually made people realize their important role in immune regulation, material metabolism, and health maintenance. In recent years, increasing studies have shown a connection between microbiota and CP/CPPS. In view of this, we performed this review to summarize the literature pertaining to microbiota and its association with the pathophysiological mechanism of CP/CPPS. In addition, we gleaned the latest progress in the therapeutic strategy of CP/CPPS that related to microbiota regulation in order to offer new perspectives on the management of CP/CPPS.
Collapse
Affiliation(s)
- An-Qi Deng
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Hefei, Anhui, China
- The Second Clinical Medical School, Anhui Medical University, Hefei, Anhui, China
| | - Shao-Yu Yue
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Hefei, Anhui, China
| | - Di Niu
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Hefei, Anhui, China
| | - Dan-Dan Zhang
- Clinical College of Anhui Medical University, Hefei, Anhui, China
| | - Bing-Bing Hou
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Hefei, Anhui, China
| | - Li Zhang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Hefei, Anhui, China
| | - Chao-Zhao Liang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Hefei, Anhui, China
| | - He-Xi Du
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Hefei, Anhui, China
| |
Collapse
|
10
|
Zhang M, Qin Z, Huang C, Liang B, Zhang X, Sun W. The gut microbiota modulates airway inflammation in allergic asthma through the gut-lung axis related immune modulation: A review. BIOMOLECULES & BIOMEDICINE 2025; 25:727-738. [PMID: 39465678 PMCID: PMC11959394 DOI: 10.17305/bb.2024.11280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 10/20/2024] [Accepted: 10/20/2024] [Indexed: 10/29/2024]
Abstract
The human gut microbiota is a vast and complex microbial community. According to statistics, the number of bacteria residing in the human intestinal tract is approximately ten times that of total human cells, with over 1000 different species. The interaction between the gut microbiota and various organ tissues plays a crucial role in the pathogenesis of local and systemic diseases, exerting a significant influence on disease progression. The relationship between the gut microbiota and intestinal diseases, along with its connection to the pulmonary immune environment and the development of lung diseases, is commonly referred to as the "gut-lung axis." The incidence of bronchial asthma is rising globally. With ongoing research on gut microbiota, it is widely believed that intestinal microorganisms and their metabolic products directly or indirectly participate in the occurrence and development of asthma. Based on the gut-lung axis, this review examines recent research suggesting that the intestinal microbiota can influence the occurrence and progression of allergic asthma through the modulation of cytokine immune balance and mucosal integrity. Though the precise immune pathways or microbial species influencing asthma through the gut-lung axis are still under exploration, summarizing the immune modulation through the gut-lung axis in allergic asthma may provide insights for the clinical management of the condition.
Collapse
Affiliation(s)
- Meng Zhang
- Department of Gastroenterology, People’s Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province, China
| | - Ziwen Qin
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| | - Chuanjun Huang
- Department of Respiratory and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province, China
| | - Bin Liang
- Department of Respiratory and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province, China
| | - Xiuqing Zhang
- Department of Radiology, Dongying City Dongying District People’s Hospital, Dongying, Shandong Province, China
| | - Weitao Sun
- Department of Respiratory Medicine, Dongying City Dongying District People’s Hospital, Dongying, Shandong Province, China
| |
Collapse
|
11
|
Huang YJ. The Microbiome in Asthma Heterogeneity: The Role of Multi-Omic Investigations. Immunol Rev 2025; 330:e70015. [PMID: 40072031 PMCID: PMC11899502 DOI: 10.1111/imr.70015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 02/13/2025] [Accepted: 02/28/2025] [Indexed: 03/15/2025]
Abstract
Asthma is one of the most prevalent and extensively studied chronic respiratory conditions, yet the heterogeneity of asthma remains biologically puzzling. Established factors like exogenous exposures and treatment adherence contribute to variability in asthma risk and clinical outcomes. It is also clear that the endogenous factors of genetics and immune system response patterns play key roles in asthma. Despite significant existing knowledge in the above, divergent clinical trajectories and outcomes are still observed, even among individuals with similar risk profiles, biomarkers, and optimal medical management. This suggests uncaptured biological interactions that contribute to asthma's heterogeneity, for which the role of host microbiota has lately attracted much research attention. This review will highlight recent evidence in this area, focusing on bedside-to-bench investigations that have leveraged omic technologies to uncover microbiome links to asthma outcomes and immunobiology. Studies centered on the respiratory system and the use of multi-omics are noted in particular. These represent a new generation of reverse-translational investigations revealing potential functional crosstalk in host microbiomes that may drive phenotypic heterogeneity in chronic diseases like asthma. Multi-omic data offer a wide lens into ecosystem interactions within a host. This informs new hypotheses and experimental work to elucidate mechanistic pathways for unresolved asthma endotypes. Further incorporation of multi-omics into patient-centered investigations can yield new insights that hopefully lead to even more precise, microbiome-informed strategies to reduce asthma burden.
Collapse
Affiliation(s)
- Yvonne J. Huang
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, Department of Microbiology and ImmunologyUniversity of Michigan Medical SchoolAnn ArborMichiganUSA
| |
Collapse
|
12
|
Dasinger JH, Abais-Battad JM, McCrorey MK, Van Beusecum JP. Recent advances on immunity and hypertension: the new cells on the kidney block. Am J Physiol Renal Physiol 2025; 328:F301-F315. [PMID: 39853324 DOI: 10.1152/ajprenal.00309.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 11/26/2024] [Accepted: 01/20/2025] [Indexed: 01/26/2025] Open
Abstract
Over the past 50 years, the contribution of the immune system has been identified in the development of hypertension and renal injury. Both human and experimental animal models of hypertension have demonstrated that innate and adaptive immune cells, along with their cytokines and chemokines, modulate blood pressure fluctuations and end organ renal damage. Numerous cell types of the innate immune system, specifically monocytes, macrophages, and dendritic cells, present antigenic peptides to T cells, promoting inflammation and the elevation of blood pressure. These T cells and other adaptive immune cells migrate to vascular and tubular cells of the kidney and promote end-organ fibrosis, damage, and ultimately hypertensive injury. Through the development of high-throughput screening, novel renal and immune cell subsets have been identified as possible contributors and regulators of renal injury and hypertension. In this review, we will consider classical immunological cells and their contribution to renal inflammation, and novel cell subsets, including renal stromal cells, that could potentially shed new light on renal injury and hypertension. Finally, we will discuss how interorgan inflammation contributes to the development of hypertension and hypertension-related multiorgan damage, and explore the clinical implications of the immunological components of renal injury and hypertension.
Collapse
Affiliation(s)
- John Henry Dasinger
- Department of Physiology, School of Medicine, Tulane University, New Orleans, Louisiana, United States
| | - Justine M Abais-Battad
- Department of Physiology, Medical College of Georgia, August University, Augusta, Georgia, United States
| | - Marice K McCrorey
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Justin P Van Beusecum
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States
- Department of Research and Development, Ralph H. Johnson VA Healthcare System, Charleston, South Carolina, United States
| |
Collapse
|
13
|
Petrut SM, Bragaru AM, Munteanu AE, Moldovan AD, Moldovan CA, Rusu E. Gut over Mind: Exploring the Powerful Gut-Brain Axis. Nutrients 2025; 17:842. [PMID: 40077713 PMCID: PMC11901622 DOI: 10.3390/nu17050842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 02/18/2025] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
Background: The human gastrointestinal tract is home to a wide variety of microorganisms. For some decades now, bacteria known as probiotics have been added to various foods because of their beneficial effects for human health. Evidence indicates that probiotics significantly regulate gut microbiota, which is vital for digestion, metabolism, immune function, and mental health. Methods: We conducted a narrative review of available original research published in PubMed for the past ten years focusing on recent advancements that provide a thorough understanding of the relationship between the gastrointestinal system and the brain. Results: Recent advances in research have focused on the importance of gut microbiota in influencing mental health. The microbiota-gut-brain axis is a complex, bidirectional communication network linking the central nervous system and the gastrointestinal tract, which highlights how the gut and brain are deeply interconnected and influence each other in ways that affect our overall health, emotions, and behavior. This powerful link is a major area of research as scientists discover more about how gut health can impact mental well-being. Conclusions: A comprehensive understanding of microbiota composition and mechanisms involved in these interactions between the gut and the brain could shape future medical and therapeutic approaches. It would balance scientific explanation with clinical relevance, offering insights into how understanding the brain-gut axis can revolutionize our approach to treating mental health and gastrointestinal disorders.
Collapse
Affiliation(s)
- Stefana-Maria Petrut
- Department of Preclinical Sciences, Faculty of Medicine, Titu Maiorescu University, 031593 Bucharest, Romania; (S.-M.P.); (E.R.)
| | - Alexandra Maria Bragaru
- Doctoral School of Medicine, Titu Maiorescu University of Bucharest, 040317 Bucharest, Romania; (A.M.B.); (A.-D.M.)
| | - Alice Elena Munteanu
- Department of Medico-Surgical and Prophylactic Sciences, Faculty of Medicine, Titu Maiorescu University, 031593 Bucharest, Romania;
- Department of Cardiology, “Dr. Carol Davila” Central Military Emergency University Hospital, 010825 Bucharest, Romania
| | - Adina-Diana Moldovan
- Doctoral School of Medicine, Titu Maiorescu University of Bucharest, 040317 Bucharest, Romania; (A.M.B.); (A.-D.M.)
- MedLife SA, 010719 Bucharest, Romania
| | - Cosmin-Alec Moldovan
- Department of Medico-Surgical and Prophylactic Sciences, Faculty of Medicine, Titu Maiorescu University, 031593 Bucharest, Romania;
- Department of General Surgery, Witting Clinical Hospital, 010243 Bucharest, Romania
| | - Elena Rusu
- Department of Preclinical Sciences, Faculty of Medicine, Titu Maiorescu University, 031593 Bucharest, Romania; (S.-M.P.); (E.R.)
| |
Collapse
|
14
|
Ke H, Yao H, Wei P. Advances in research on gut microbiota and allergic diseases in children. CURRENT RESEARCH IN MICROBIAL SCIENCES 2025; 8:100362. [PMID: 40123594 PMCID: PMC11930230 DOI: 10.1016/j.crmicr.2025.100362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/25/2025] Open
Abstract
Epidemiological studies indicate a rising prevalence of allergic diseases, now recognized as a major global public health concern. In children, the progression of these diseases often follows the "atopic march," beginning with eczema, followed by food allergies, allergic rhinitis, and asthma. Recent research has linked gut microbiota dysbiosis to the development of allergic diseases in children. The gut microbiota, a crucial component of human health, plays a vital role in maintaining overall well-being, highlighting its potential in preventing and modifying the course of allergic diseases. This review examines the relationship between childhood allergic diseases and gut microbiota, drawing on the latest evidence. We first elaborated the concepts of allergic diseases and gut microbiota, followed by a discussion of the developmental trajectory of the gut microbiota in healthy children. This review further explored the richness, diversity, and composition of the gut microbiota, as well as specific microbial taxa associated with allergic disease. Lastly, we discussed the current status and future potential of probiotic interventions in managing pediatric allergic diseases.
Collapse
Affiliation(s)
- Heng Ke
- Department of Otolaryngology, The Children's Hospital of Chongqing Medical University, Chongqing, PR China
| | - Hongbing Yao
- Department of Otolaryngology, The Children's Hospital of Chongqing Medical University, Chongqing, PR China
| | - Ping Wei
- Department of Otolaryngology, The Children's Hospital of Chongqing Medical University, Chongqing, PR China
| |
Collapse
|
15
|
Han Y, Teng TM, Han J, Kim HS. Antibiotic-associated changes in Akkermansia muciniphila alter its effects on host metabolic health. MICROBIOME 2025; 13:48. [PMID: 39920776 PMCID: PMC11804010 DOI: 10.1186/s40168-024-02023-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 12/19/2024] [Indexed: 02/09/2025]
Abstract
BACKGROUND Altered gut microbiota has emerged as a major contributing factor to the etiology of chronic conditions in humans. Antibiotic exposure, historically dating back to the mass production of penicillin in the early 1940s, has been proposed as a primary contributor to the cumulative alteration of microbiota over generations. However, the mechanistic link between the antibiotics-altered microbiota and chronic conditions remains unclear. RESULTS In this study, we discovered that variants of the key beneficial gut microbe, Akkermansia muciniphila, were selected upon exposure to penicillin. These variants had mutations in the promoter of a TEM-type β-lactamase gene or pur genes encoding the de novo purine biosynthesis pathway, and they exhibited compromised abilities to mitigate host obesity in a murine model. Notably, variants of A. muciniphila are prevalent in the human microbiome worldwide. CONCLUSIONS These findings highlight a previously unknown mechanism through which antibiotics influence host health by affecting the beneficial capacities of the key gut microbes. Furthermore, the global prevalence of A. muciniphila variants raises the possibility that these variants contribute to global epidemics of chronic conditions, warranting further investigations in human populations. Video Abstract.
Collapse
Affiliation(s)
- Yumin Han
- Division of Biosystems & Biomedical Sciences, College of Health Sciences, 145 Anam-Ro, Seongbuk-Gu, Seoul, 02841, Korea
| | - Teh Min Teng
- Division of Biosystems & Biomedical Sciences, College of Health Sciences, 145 Anam-Ro, Seongbuk-Gu, Seoul, 02841, Korea
| | - Juwon Han
- Division of Biosystems & Biomedical Sciences, College of Health Sciences, 145 Anam-Ro, Seongbuk-Gu, Seoul, 02841, Korea
| | - Heenam Stanley Kim
- Division of Biosystems & Biomedical Sciences, College of Health Sciences, 145 Anam-Ro, Seongbuk-Gu, Seoul, 02841, Korea.
| |
Collapse
|
16
|
Bijnens K, Thijs S, Alfano R, McAmmond B, Van Hamme J, Artois T, Plusquin M, Vangronsveld J, Smeets K. Impact of host physiology and external stressors on the bacterial community of Schmidtea mediterranea. Sci Rep 2025; 15:4398. [PMID: 39910204 PMCID: PMC11799148 DOI: 10.1038/s41598-025-86920-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 01/15/2025] [Indexed: 02/07/2025] Open
Abstract
To fully comprehend host-microorganism interactions, it is crucial to understand the composition and diversity of the microbiome, as well as the factors that shape these characteristics. We investigated microbiome variation using the freshwater planarian Schmidtea mediterranea, an invertebrate model in regeneration biology and (eco-)toxicology, by exposing the organisms to various controlled conditions. The microbiome composition exhibited high variability, with most of the bacteria belonging to the Betaproteobacteria. Among the diverse microbial communities, a few genera, such as Curvibacter, were consistently present, but exhibited significant alterations in response to changing conditions. The relative abundance of Curvibacter fluctuated during the regeneration process, initially increasing before returning to a composition similar to the beginning situation. After applying external stress, the relative abundance of Curvibacter and other genera decreased. Variation over time, between different origin laboratories and between individuals, showed that additional, yet to-be-identified, factors of variation are present. Taking all results together, our study provides a solid basis for future research focusing on bacterial functionality in planarians and other invertebrates.
Collapse
Affiliation(s)
- Karolien Bijnens
- Centre for Environmental Sciences, Zoology, Biodiversity and Toxicology, Hasselt University, Diepenbeek, Belgium
| | - Sofie Thijs
- Centre for Environmental Sciences, Environmental Biology, Hasselt University, Diepenbeek, Belgium
| | - Rossella Alfano
- Centre for Environmental Sciences, Epidemiology, Hasselt University, Diepenbeek, Belgium
| | - Breanne McAmmond
- Department of Biological Sciences, Thompson Rivers University, Kamloops, BC, Canada
| | - Jonathan Van Hamme
- Department of Biological Sciences, Thompson Rivers University, Kamloops, BC, Canada
| | - Tom Artois
- Centre for Environmental Sciences, Zoology, Biodiversity and Toxicology, Hasselt University, Diepenbeek, Belgium
| | - Michelle Plusquin
- Centre for Environmental Sciences, Epidemiology, Hasselt University, Diepenbeek, Belgium
| | - Jaco Vangronsveld
- Centre for Environmental Sciences, Environmental Biology, Hasselt University, Diepenbeek, Belgium
- Department of Plant Physiology and Biophysics, Institute of Biology and Biotechnology, Maria Skłodowska-Curie University, Lublin, Poland
| | - Karen Smeets
- Centre for Environmental Sciences, Zoology, Biodiversity and Toxicology, Hasselt University, Diepenbeek, Belgium.
| |
Collapse
|
17
|
Castellanos-Ruiz D, Ojeda-Borbolla JG, Ruiz-García OV, Peña-Corona SI, Martínez-Peña AA, Ibarra-Rubio ME, Gavilanes-Ruiz M, Mendoza-Rodríguez CA. Uterine Microbiota and Bisphenols: Novel Influencers in Reproductive Health. J Xenobiot 2025; 15:26. [PMID: 39997369 PMCID: PMC11856463 DOI: 10.3390/jox15010026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 01/10/2025] [Accepted: 01/24/2025] [Indexed: 02/26/2025] Open
Abstract
Infertility affects 8-12% of couples worldwide, and 30-75% of preclinical pregnancy losses are due to a failure during the implantation process. Exposure to endocrine disruptors, like bisphenols, among others, has been associated with the increase in infertility observed in the past decades. An increase in infertility has correlated with exposure to endocrine disruptors like bisphenols. The uterus harbors its own microbiota, and changes in this microbiota have been linked to several gynecological conditions, including reproductive failure. There are no studies on the effects of bisphenols on the uterine-microbiota composition, but some inferences can be gleaned by looking at the gut. Bisphenols can alter the gut microbiota, and the molecular mechanism by which gut microbiota regulates intestinal permeability involves Toll-like receptors (TLRs) and tight junction (TJ) proteins. TJs participate in embryo implantation in the uterus, but bisphenol exposure disrupts the expression and localization of TJ proteins. The aim of this review is to summarize the current knowledge on the microbiota of the female reproductive tract (FRT), its association with different reproductive diseases-particularly reproductive failure-the effects of bisphenols on microbiota composition and reproductive health, and the molecular mechanisms regulating uterine-microbiota interactions crucial for embryo implantation. This review also highlights existing knowledge gaps and outlines research needs for future risk assessments regarding the effects of bisphenols on reproduction.
Collapse
Affiliation(s)
- Dafne Castellanos-Ruiz
- Facultad de Química, Departamento de Biología, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico (M.E.I.-R.)
| | - J. Gerardo Ojeda-Borbolla
- Facultad de Química, Departamento de Biología, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico (M.E.I.-R.)
| | - Olga V. Ruiz-García
- Facultad de Química, Departamento de Biología, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico (M.E.I.-R.)
| | - Sheila I. Peña-Corona
- Facultad de Química, Departamento de Farmacia, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Annia A. Martínez-Peña
- División de Ciencias de la Salud, Universidad Intercontinental, A. C., Ciudad de México 14420, Mexico
| | - María Elena Ibarra-Rubio
- Facultad de Química, Departamento de Biología, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico (M.E.I.-R.)
| | - Marina Gavilanes-Ruiz
- Facultad de Química, Departamento de Bioquímica, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - C. Adriana Mendoza-Rodríguez
- Facultad de Química, Departamento de Biología, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico (M.E.I.-R.)
| |
Collapse
|
18
|
Kumar P, Kumar A, Kumar V. Role of Microbiota-Derived Metabolites in Prostate Cancer Inflammation and Progression. Cell Biochem Funct 2025; 43:e70050. [PMID: 39891389 DOI: 10.1002/cbf.70050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 12/25/2024] [Accepted: 01/20/2025] [Indexed: 02/03/2025]
Abstract
Prostate cancer (PCa) is the most commonly detected malignancy in men worldwide. PCa is a slow-growing cancer with the absence of symptoms at early stages. The pathogenesis has not been entirely understood including the key risk factors related to PCa development like diet and microbiota derived metabolites. Microbiota may influence the host's immunological responses, inflammatory responses, and metabolic pathways, which may be crucial for the development and metastasis. Similarly, short-chain fatty acids, methylamines, hippurate, bile acids, and other metabolites generated by microbiota may have potential roles in cancer inflammation and progression of cancer. Most studies have focused on the role of metabolites and their pathways involved in chronic inflammation, tumor initiation, proliferation, and progression. In summary, the review discusses the role of microbiota and microbial-derived metabolite-built strategies in inflammation and progression of the PCa.
Collapse
Affiliation(s)
- Pradeep Kumar
- Department of NMR, All India Institute of Medical Sciences, New Delhi, India
| | - Anil Kumar
- Gene Regulation Laboratory, National Institute of Immunology, New Delhi, India
| | - Virendra Kumar
- Department of NMR, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
19
|
Hu D, Li X, Duan X, Yang L, Luo B, Wang L, Hu Z, Zhou Y, Qian P. Recombinant Saccharomyces cerevisiae EBY100/pYD1-FaeG: a candidate for an oral subunit vaccine against F4+ ETEC infection. Appl Environ Microbiol 2025; 91:e0181724. [PMID: 39601541 PMCID: PMC11784076 DOI: 10.1128/aem.01817-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 11/04/2024] [Indexed: 11/29/2024] Open
Abstract
Diarrheal diseases attributable to multidrug-resistant F4+ enterotoxigenic Escherichia coli (ETEC) are escalating in severity, posing significant risks to the health and safety of both humans and animals. This study used Saccharomyces cerevisiae EBY100 to display the FaeG subunit of F4 colonizing factor as an oral vaccine against F4+ ETEC infection. Mice were orally immunized twice with 108 CFU of EBY100/pYD1-FaeG, followed by a challenge with F4+ ETEC EC6 on day 7 post-immunization. The results showed that the recombinant strain EBY100/pYD1-FaeG orally enhanced the growth of the small intestine villi, significantly boosted the expression of tight junction proteins (ZO-1, Occludin, MUC2, and Claudin) (P < 0.05), and modulated the gut microbiota composition. Additionally, immunization with EBY100/pYD1-FaeG also upregulated the levels of IL-2, IL-4, and IFN-γ in the intestines of mice (P < 0.01), while serum IgG and fecal sIgA titer significantly increased (P < 0.05). These immune responses enhanced the capacity to fight against ETEC, leading to an increased survival rate of mice and relieved damage to tissues and organs of mice infection. In summary, the study suggested that the recombinant Saccharomyces cerevisiae EBY100/pYD1-FaeG could effectively stimulate the immune response and generate specific antibodies against F4+ ETEC, showing its potential to serve as a subunit oral vaccine candidate for preventing F4+ ETEC infection.IMPORTANCEThe multidrug-resistant F4+ enterotoxigenic Escherichia coli (ETEC) strains are the primary clinical pathogens responsible for post-weaning diarrhea in piglets, resulting in substantial economic losses in the pig farming industry. In the study, we developed an oral vaccine candidate, Saccharomyces cerevisiae EBY100/pYD1-FaeG, to prevent diarrhea caused by multidrug-resistant F4+ ETEC. Oral administration of EBY100/pYD1-FaeG significantly enhanced immune responses, improved intestinal health, and provided protection against F4+ ETEC infection in mice. This approach offers a potential application prospect for preventing F4+ ETEC infections that lead to post-weaning diarrhea in clinical settings and provides a promising solution for addressing the growing threat of antibiotic resistance in bacterial pathogens.
Collapse
Affiliation(s)
- Dayue Hu
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
- The Cooperative Innovation Centre for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Xiangmin Li
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
- The Cooperative Innovation Centre for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Xiaochao Duan
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
- The Cooperative Innovation Centre for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Liuyue Yang
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
- The Cooperative Innovation Centre for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Baizhi Luo
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
- The Cooperative Innovation Centre for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Linkang Wang
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
- The Cooperative Innovation Centre for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Zihui Hu
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
- The Cooperative Innovation Centre for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Yang Zhou
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
- The Cooperative Innovation Centre for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Ping Qian
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
- The Cooperative Innovation Centre for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
20
|
Benitez AJ, Tanes C, Friedman ES, Zackular JP, Ford E, Gerber JS, DeRusso PA, Kelly A, Li H, Elovitz MA, Wu GD, Zemel B, Bittinger K. Antibiotic exposure is associated with minimal gut microbiome perturbations in healthy term infants. MICROBIOME 2025; 13:21. [PMID: 39856742 PMCID: PMC11761179 DOI: 10.1186/s40168-024-01999-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 12/05/2024] [Indexed: 01/27/2025]
Abstract
BACKGROUND The evolving infant gut microbiome influences host immune development and later health outcomes. Early antibiotic exposure could impact microbiome development and contribute to poor outcomes. Here, we use a prospective longitudinal birth cohort of n = 323 healthy term African American children to determine the association between antibiotic exposure and the gut microbiome through shotgun metagenomics sequencing as well as bile acid profiles through liquid chromatography-mass spectrometry. RESULTS Stool samples were collected at ages 4, 12, and 24 months for antibiotic-exposed (n = 170) and unexposed (n = 153) participants. A short-term substudy (n = 39) collected stool samples at first exposure, and over 3 weeks following antibiotics initiation. Antibiotic exposure (predominantly amoxicillin) was associated with minimal microbiome differences, whereas all tested taxa were modified by breastfeeding. In the short-term substudy, we observed microbiome differences only in the first 2 weeks following antibiotics initiation, mainly a decrease in Bifidobacterium bifidum. The differences did not persist a month after antibiotic exposure. Four species were associated with infant age. Antibiotic exposure was not associated with an increase in antibiotic resistance gene abundance or with differences in microbiome-derived fecal bile acid composition. CONCLUSIONS Short-term and long-term gut microbiome perturbations by antibiotic exposure were detectable but substantially smaller than those associated with breastfeeding and infant age.
Collapse
Affiliation(s)
- Alain J Benitez
- Division of Gastroenterology, Hepatology, and Nutrition, The Children's Hospital of Philadelphia, Philadelphia, PA, 19146, USA
- Department of Pediatrics, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Ceylan Tanes
- Division of Gastroenterology, Hepatology, and Nutrition, The Children's Hospital of Philadelphia, Philadelphia, PA, 19146, USA
| | - Elliot S Friedman
- Division of Gastroenterology and Hepatology, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Joseph P Zackular
- Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, and The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Division of Protective Immunity, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Eileen Ford
- Division of Endocrinology and Diabetes, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jeffrey S Gerber
- Department of Pediatrics, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Division of Infectious Diseases, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Biomedical and Health Informatics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Patricia A DeRusso
- Division of Gastroenterology, Hepatology, and Nutrition, The Children's Hospital of Philadelphia, Philadelphia, PA, 19146, USA
- Department of Pediatrics, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Andrea Kelly
- Department of Pediatrics, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Division of Endocrinology and Diabetes, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Hongzhe Li
- Department of Biostatistics, Informatics, and Epidemiology, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Michal A Elovitz
- Women's Biomedical Research Institute, Icahn School of Medicine, New York, NY, USA
| | - Gary D Wu
- Division of Gastroenterology and Hepatology, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Babette Zemel
- Division of Gastroenterology, Hepatology, and Nutrition, The Children's Hospital of Philadelphia, Philadelphia, PA, 19146, USA
- Department of Pediatrics, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Kyle Bittinger
- Division of Gastroenterology, Hepatology, and Nutrition, The Children's Hospital of Philadelphia, Philadelphia, PA, 19146, USA.
- Department of Pediatrics, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
21
|
Armetta J, Li SS, Vaaben TH, Vazquez-Uribe R, Sommer MOA. Metagenome-guided culturomics for the targeted enrichment of gut microbes. Nat Commun 2025; 16:663. [PMID: 39809763 PMCID: PMC11733127 DOI: 10.1038/s41467-024-55668-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 12/19/2024] [Indexed: 01/16/2025] Open
Abstract
The gut microbiome significantly impacts human health, yet cultivation challenges hinder its exploration. Here, we combine deep whole-metagenome sequencing with culturomics to selectively enrich for taxa and functional capabilities of interest. Using a modified commercial base medium, 50 growth modifications were evaluated, spanning antibiotics, physico-chemical conditions, and bioactive compounds. Whole-metagenome sequencing identified medium additives, like caffeine, that enhance taxa often associated with healthier subjects (e.g., Lachnospiraceae, Oscillospiraceae, Ruminococcaceae). We also explore the impact of modifications on the composition of cultured communities and establish a link between medium preference and microbial phylogeny. Leveraging these insights, we demonstrate that combinations of media modifications can further enhance the targeted enrichment of taxa and metabolic functions, such as Collinsella aerofaciens, or strains harboring biochemical pathways involved in dopamine metabolism. This streamlined, scalable approach unlocks the potential for selective enrichment, advancing microbiome research by understanding the impact of different cultivation parameters on gut microbes.
Collapse
Affiliation(s)
- Jeremy Armetta
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark Kgs., Lyngby, Denmark
- Novonesis A/S, Hørsholm, Denmark
| | - Simone S Li
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark Kgs., Lyngby, Denmark
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia
- Department of Microbiology, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
- Centre to Impact Antimicrobial Resistance, Monash University, Melbourne, VIC, Australia
| | - Troels Holger Vaaben
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark Kgs., Lyngby, Denmark
| | - Ruben Vazquez-Uribe
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark Kgs., Lyngby, Denmark
- Center for Microbiology, VIB, Leuven, Belgium
| | - Morten O A Sommer
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark Kgs., Lyngby, Denmark.
| |
Collapse
|
22
|
Dera N, Kosińska-Kaczyńska K, Żeber-Lubecka N, Brawura-Biskupski-Samaha R, Massalska D, Szymusik I, Dera K, Ciebiera M. Impact of Early-Life Microbiota on Immune System Development and Allergic Disorders. Biomedicines 2025; 13:121. [PMID: 39857705 PMCID: PMC11762082 DOI: 10.3390/biomedicines13010121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 12/26/2024] [Accepted: 01/02/2025] [Indexed: 01/27/2025] Open
Abstract
Introduction: The shaping of the human intestinal microbiota starts during the intrauterine period and continues through the subsequent stages of extrauterine life. The microbiota plays a significant role in the predisposition and development of immune diseases, as well as various inflammatory processes. Importantly, the proper colonization of the fetal digestive system is influenced by maternal microbiota, the method of pregnancy completion and the further formation of the microbiota. In the subsequent stages of a child's life, breastfeeding, diet and the use of antibiotics influence the state of eubiosis, which determines proper growth and development from the neonatal period to adulthood. The literature data suggest that there is evidence to confirm that the intestinal microbiota of the infant plays an important role in regulating the immune response associated with the development of allergic diseases. However, the identification of specific bacterial species in relation to specific types of reactions in allergic diseases is the basic problem. Background: The main aim of the review was to demonstrate the influence of the microbiota of the mother, fetus and newborn on the functioning of the immune system in the context of allergies and asthma. Methods: We reviewed and thoroughly analyzed the content of over 1000 articles and abstracts between the beginning of June and the end of August 2024. Over 150 articles were selected for the detailed study. Results: The selection was based on the PubMed National Library of Medicine search engine, using selected keywords: "the impact of intestinal microbiota on the development of immune diseases and asthma", "intestinal microbiota and allergic diseases", "the impact of intrauterine microbiota on the development of asthma", "intrauterine microbiota and immune diseases", "intrauterine microbiota and atopic dermatitis", "intrauterine microbiota and food allergies", "maternal microbiota", "fetal microbiota" and "neonatal microbiota". The above relationships constituted the main criteria for including articles in the analysis. Conclusions: In the present review, we showed a relationship between the proper maternal microbiota and the normal functioning of the fetal and neonatal immune system. The state of eubiosis with an adequate amount and diversity of microbiota is essential in preventing the development of immune and allergic diseases. The way the microbiota is shaped, resulting from the health-promoting behavior of pregnant women, the rational conduct of the medical staff and the proper performance of the diagnostic and therapeutic process, is necessary to maintain the health of the mother and the child. Therefore, an appropriate lifestyle, rational antibiotic therapy as well as the way of completing the pregnancy are indispensable in the prevention of the above conditions. At the same time, considering the intestinal microbiota of the newborn in relation to the genera and phyla of bacteria that have a potentially protective effect, it is worth noting that the use of suitable probiotics and prebiotics seems to contribute to the protective effect.
Collapse
Affiliation(s)
- Norbert Dera
- Department of Obstetrics, Perinatology and Neonatology, Center of Postgraduate Medical Education, 01-809 Warsaw, Poland; (N.D.); (K.K.-K.); (R.B.-B.-S.); (I.S.)
- Warsaw Institute of Women’s Health, 00-189 Warsaw, Poland; (D.M.); (M.C.)
| | - Katarzyna Kosińska-Kaczyńska
- Department of Obstetrics, Perinatology and Neonatology, Center of Postgraduate Medical Education, 01-809 Warsaw, Poland; (N.D.); (K.K.-K.); (R.B.-B.-S.); (I.S.)
| | - Natalia Żeber-Lubecka
- Department of Gastroenterology, Hepatology and Clinical Oncology, Center of Postgraduate Medical Education, 02-781 Warsaw, Poland;
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Robert Brawura-Biskupski-Samaha
- Department of Obstetrics, Perinatology and Neonatology, Center of Postgraduate Medical Education, 01-809 Warsaw, Poland; (N.D.); (K.K.-K.); (R.B.-B.-S.); (I.S.)
| | - Diana Massalska
- Warsaw Institute of Women’s Health, 00-189 Warsaw, Poland; (D.M.); (M.C.)
- Second Department of Obstetrics and Gynecology, Center of Postgraduate Medical Education, 00-189 Warsaw, Poland
| | - Iwona Szymusik
- Department of Obstetrics, Perinatology and Neonatology, Center of Postgraduate Medical Education, 01-809 Warsaw, Poland; (N.D.); (K.K.-K.); (R.B.-B.-S.); (I.S.)
| | - Kacper Dera
- Pediatric Ward, Department of Pediatrics, Center of Postgraduate Medical Education, Bielański Hospital, 01-809 Warsaw, Poland
| | - Michał Ciebiera
- Warsaw Institute of Women’s Health, 00-189 Warsaw, Poland; (D.M.); (M.C.)
- Second Department of Obstetrics and Gynecology, Center of Postgraduate Medical Education, 00-189 Warsaw, Poland
| |
Collapse
|
23
|
Wang J, Wang X, Zhuo E, Chen B, Chan S. Gut‑liver axis in liver disease: From basic science to clinical treatment (Review). Mol Med Rep 2025; 31:10. [PMID: 39450549 PMCID: PMC11541166 DOI: 10.3892/mmr.2024.13375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 06/14/2024] [Indexed: 10/26/2024] Open
Abstract
Incidence of a number of liver diseases has increased. Gut microbiota serves a role in the pathogenesis of hepatitis, cirrhosis and liver cancer. Gut microbiota is considered 'a new virtual metabolic organ'. The interaction between the gut microbiota and liver is termed the gut‑liver axis. The gut‑liver axis provides a novel research direction for mechanism of liver disease development. The present review discusses the role of the gut‑liver axis and how this can be targeted by novel treatments for common liver diseases.
Collapse
Affiliation(s)
- Jianpeng Wang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032, P.R. China
- Department of Clinical Medicine, The First Clinical Medical College, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Xinyi Wang
- Department of Radiation Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Enba Zhuo
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Bangjie Chen
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Shixin Chan
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032, P.R. China
| |
Collapse
|
24
|
Gilaberte Y, Piquero‐Casals J, Schalka S, Leone G, Brown A, Trullàs C, Jourdan E, Lim HW, Krutmann J, Passeron T. Exploring the impact of solar radiation on skin microbiome to develop improved photoprotection strategies. Photochem Photobiol 2025; 101:38-52. [PMID: 38767119 PMCID: PMC11737011 DOI: 10.1111/php.13962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 03/29/2024] [Accepted: 04/25/2024] [Indexed: 05/22/2024]
Abstract
The skin microbiome undergoes constant exposure to solar radiation (SR), with its effects on health well-documented. However, understanding SR's influence on host-associated skin commensals remains nascent. This review surveys existing knowledge on SR's impact on the skin microbiome and proposes innovative sun protection methods that safeguard both skin integrity and microbiome balance. A team of skin photodamage specialists conducted a comprehensive review of 122 articles sourced from PubMed and Research Gateway. Key terms included skin microbiome, photoprotection, photodamage, skin cancer, ultraviolet radiation, solar radiation, skin commensals, skin protection, and pre/probiotics. Experts offered insights into novel sun protection products designed not only to shield the skin but also to mitigate SR's effects on the skin microbiome. Existing literature on SR's influence on the skin microbiome is limited. SR exposure can alter microbiome composition, potentially leading to dysbiosis, compromised skin barrier function, and immune system activation. Current sun protection methods generally overlook microbiome considerations. Tailored sun protection products that prioritize both skin and microbiome health may offer enhanced defense against SR-induced skin conditions. By safeguarding both skin and microbiota, these specialized products could mitigate dysbiosis risks associated with SR exposure, bolstering skin defense mechanisms and reducing the likelihood of SR-mediated skin issues.
Collapse
Affiliation(s)
- Yolanda Gilaberte
- Department of DermatologyMiguel Servet University Hospital, IIS AragónZaragozaSpain
| | - Jaime Piquero‐Casals
- Department of DermatologyDermik Multidisciplinary Dermatology ClinicBarcelonaSpain
| | - Sergio Schalka
- Medcin Skin Research Center and Biochemistry DepartmentChemistry Institute of São Paulo UniversitySão PauloBrazil
| | - Giovanni Leone
- Photodermatology and Vitiligo Treatment UnitIsraelite HospitalRomeItaly
| | | | | | | | - Henry W. Lim
- The Henry W. Lim Division of Photobiology and Photomedicine, Department of DermatologyHenry Ford HealthDetroitMichiganUSA
| | - Jean Krutmann
- IUF – Leibniz‐Institut für umweltmedizinische ForschungDüsseldorfGermany
| | - Thierry Passeron
- Department of DermatologyCentre Hospitalier Universitaire de Nice, Université Côte d'AzurNiceFrance
- Centre Méditerranéen de Médecine Moléculaire, INSERM U1065Université Côte d'AzurNiceFrance
| |
Collapse
|
25
|
Shaaban R, Busi SB, Wilmes P, Guéant JL, Heinken A. Personalized modeling of gut microbiome metabolism throughout the first year of life. COMMUNICATIONS MEDICINE 2024; 4:281. [PMID: 39739091 DOI: 10.1038/s43856-024-00715-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 12/17/2024] [Indexed: 01/02/2025] Open
Abstract
BACKGROUND Early-life exposures including diet, and the gut microbiome have been proposed to predispose infants towards multifactorial diseases later in life. Delivery via Cesarian section disrupts the establishment of the gut microbiome and has been associated with negative long-term outcomes. Here, we hypothesize that Cesarian section delivery alters not only the composition of the developing infant gut microbiome but also its metabolic capabilities. To test this, we developed a metabolic modeling workflow targeting the infant gut microbiome. METHODS The AGORA2 resource of human microbial genome-scale reconstructions was expanded with a human milk oligosaccharide degradation module. Personalized metabolic modeling of the gut microbiome was performed for a cohort of 20 infants at four time points during the first year of life as well as for 13 maternal gut microbiome samples. RESULTS Here we show that at the earliest stages, the gut microbiomes of infants delivered through Cesarian section are depleted in their metabolic capabilities compared with vaginal delivery. Various metabolites such as fermentation products, human milk oligosaccharide degradation products, and amino acids are depleted in Cesarian section delivery gut microbiomes. Compared with maternal gut microbiomes, infant gut microbiomes produce less butyrate but more L-lactate and are enriched in the potential to synthesize B-vitamins. CONCLUSIONS Our simulations elucidate the metabolic capabilities of the infant gut microbiome demonstrating they are altered in Cesarian section delivery at the earliest time points. Our workflow can be readily applied to other cohorts to evaluate the effect of feeding type, or maternal factors such as diet on host-gut microbiome inactions in early life.
Collapse
Affiliation(s)
- Rola Shaaban
- Inserm UMRS 1256 NGERE, University of Lorraine, Nancy, France
- Nantes University, Nantes, France
| | - Susheel Bhanu Busi
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
- UK Centre for Ecology and Hydrology, Wallingford, Oxfordshire, UK
| | - Paul Wilmes
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Jean-Louis Guéant
- Inserm UMRS 1256 NGERE, University of Lorraine, Nancy, France
- National Center of Inborn Errors of Metabolism, University Regional Hospital Center of Nancy, Nancy, France
| | - Almut Heinken
- Inserm UMRS 1256 NGERE, University of Lorraine, Nancy, France.
| |
Collapse
|
26
|
Turner M, Van Hulzen L, Guse K, Agany D, Pietri JE. The gut microbiota confers resistance against Salmonella Typhimurium in cockroaches by modulating innate immunity. iScience 2024; 27:111293. [PMID: 39628558 PMCID: PMC11612784 DOI: 10.1016/j.isci.2024.111293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 10/17/2024] [Accepted: 10/28/2024] [Indexed: 12/06/2024] Open
Abstract
Cockroaches exhibit unexplained intra- and interpopulation variation in susceptibility to Salmonella enterica serovar Typhimurium (S. Typhimurium) infection. Here, we show that the gut microbiota has a protective effect against colonization by ingested S. Typhimurium in cockroaches. We further examine two potential mechanisms for this effect, showing that commensal bacteria present in the gut do not compete with S. Typhimurium during growth in cockroach feces, but rather prime expression of host antimicrobial peptide genes that suppress S. Typhimurium infection. Lastly, we determine that neither absolute abundance of the microbiota nor its overall diversity is linked to infection susceptibility. Instead, we identify several minority bacterial taxa that exhibit interindividual variation in abundance as key indicators of infection susceptibility among genetically similar individuals. These findings illuminate the potential of cockroaches as an invertebrate model for interspecies microbial interactions and provide insight into vector-borne Salmonella transmission, suggesting that the microbiota of cockroaches could be targeted to reduce pathogen transmission.
Collapse
Affiliation(s)
- Matthew Turner
- University of South Dakota, Sanford School of Medicine, Division of Basic Biomedical Sciences, Vermillion, SD, USA
| | - Landen Van Hulzen
- University of South Dakota, Sanford School of Medicine, Division of Basic Biomedical Sciences, Vermillion, SD, USA
| | - Kylene Guse
- University of South Dakota, Sanford School of Medicine, Division of Basic Biomedical Sciences, Vermillion, SD, USA
| | - Diing Agany
- University of South Dakota, Sanford School of Medicine, Division of Basic Biomedical Sciences, Vermillion, SD, USA
| | - Jose E. Pietri
- University of South Dakota, Sanford School of Medicine, Division of Basic Biomedical Sciences, Vermillion, SD, USA
- Purdue University, Department of Entomology, Center for Urban and Industrial Pest Management, West Lafayette, IN, USA
- Purdue University, Institute of Inflammation, Immunology and Infectious Disease, West Lafayette, IN, USA
- Purdue University, Department of Biological Sciences, West Lafayette, IN, USA
| |
Collapse
|
27
|
Tsuchida S, Umemura H, Iizuka K, Yamamoto H, Shimazaki I, Shikata E, Nakayama T. Recent findings on metabolomics and the microbiome of oral bacteria involved in dental caries and periodontal disease. World J Microbiol Biotechnol 2024; 41:11. [PMID: 39690257 DOI: 10.1007/s11274-024-04224-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 12/06/2024] [Indexed: 12/19/2024]
Abstract
Periodontal disease is characterized by bacterial toxins within the oral biofilm surrounding the teeth, leading to gingivitis and the gradual dissolution of the alveolar bone, which supports the teeth. Notably, symptoms in the early stages of the disease are often absent. Similarly, dental caries occurs when oral bacteria metabolize dietary sugars, producing acids that dissolve tooth enamel and dentin. These bacteria are commonly present in the oral cavity of most individuals. Metabolomics, a relatively recent addition to the "omics" research landscape, involves the comprehensive analysis of metabolites in vivo to elucidate pathological mechanisms and accelerate drug discovery. Meanwhile, the term "microbiome" refers to the collection of microorganisms within a specific environmental niche or their collective genomes. The human microbiome plays a critical role in health and disease, influencing a wide array of physiological and pathological processes. Recent advances in microbiome research have identified numerous bacteria implicated in dental caries and periodontal disease. Additionally, studies have uncovered various pathogenic factors associated with these microorganisms. This review focuses on recent findings in metabolomics and the microbiome, specifically targeting oral bacteria linked to dental caries and periodontal disease. We acknowledge the limitation of relying exclusively on the MEDLINE database via PubMed, while excluding other sources such as gray literature, conference proceedings, and clinical practice guidelines.
Collapse
Affiliation(s)
- Sachio Tsuchida
- Divisions of Laboratory Medicine, Department of Pathology and Microbiology, Nihon University School of Medicine, 30-1 Oyaguchikamicho, Itabashi-ku, Tokyo, Japan
| | - Hiroshi Umemura
- Divisions of Laboratory Medicine, Department of Pathology and Microbiology, Nihon University School of Medicine, 30-1 Oyaguchikamicho, Itabashi-ku, Tokyo, Japan
| | - Kazuhide Iizuka
- Divisions of Laboratory Medicine, Department of Pathology and Microbiology, Nihon University School of Medicine, 30-1 Oyaguchikamicho, Itabashi-ku, Tokyo, Japan
| | - Haruka Yamamoto
- Divisions of Laboratory Medicine, Department of Pathology and Microbiology, Nihon University School of Medicine, 30-1 Oyaguchikamicho, Itabashi-ku, Tokyo, Japan
| | - Isamu Shimazaki
- Divisions of Laboratory Medicine, Department of Pathology and Microbiology, Nihon University School of Medicine, 30-1 Oyaguchikamicho, Itabashi-ku, Tokyo, Japan
| | - Elisa Shikata
- Divisions of Laboratory Medicine, Department of Pathology and Microbiology, Nihon University School of Medicine, 30-1 Oyaguchikamicho, Itabashi-ku, Tokyo, Japan
| | - Tomohiro Nakayama
- Divisions of Laboratory Medicine, Department of Pathology and Microbiology, Nihon University School of Medicine, 30-1 Oyaguchikamicho, Itabashi-ku, Tokyo, Japan.
| |
Collapse
|
28
|
Huang J, Lu Y, Tian F, Ni Y. Association of body index with fecal microbiome in children cohorts with ethnic-geographic factor interaction: accurately using a Bayesian zero-inflated negative binomial regression model. mSystems 2024; 9:e0134524. [PMID: 39570024 DOI: 10.1128/msystems.01345-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 10/24/2024] [Indexed: 11/22/2024] Open
Abstract
The exponential growth of high-throughput sequencing (HTS) data on the microbial communities presents researchers with an unparalleled opportunity to delve deeper into the association of microorganisms with host phenotype. However, this growth also poses a challenge, as microbial data are complex, sparse, discrete, and prone to zero inflation. Herein, by utilizing 10 distinct counting models for analyzing simulated data, we proposed an innovative Bayesian zero-inflated negative binomial (ZINB) regression model that is capable of identifying differentially abundant taxa associated with distinctive host phenotypes and quantifying the effects of covariates on these taxa. Our proposed model exhibits excellent accuracy compared with conventional Hurdle and INLA models, especially in scenarios characterized by inflation and overdispersion. Moreover, we confirm that dispersion parameters significantly affect the accuracy of model results, with defects gradually alleviating as the number of analyzed samples increases. Subsequently applying our model to amplicon data in real multi-ethnic children cohort, we found that only a subset of taxa were identified as having zero inflation in real data, suggesting that the prevailing understanding and processing of microbial count data in most previous microbiome studies were overly dogmatic. In practice, our pipeline of integrating bacterial differential abundance in microbiome data and relevant covariates is effective and feasible. Taken together, our method is expected to be extended to the microbiota studies of various multi-cohort populations. IMPORTANCE The microbiome is closely associated with physical indicators of the body, such as height, weight, age and BMI, which can be used as measures of human health. Accurately identifying which taxa in the microbiome are closely related to indicators of physical development is valuable as microbial markers of regional child growth trajectory. Zero-inflated negative binomial (ZINB) model, a type of Bayesian generalized linear model, can be effectively modeled in complex biological systems. We present an innovative ZINB regression model that is capable of identifying differentially abundant taxa associated with distinctive host phenotypes and quantifying the effects of covariates on these taxa, and demonstrate that its accuracy is superior to traditional Hurdle and INLA models. Our pipeline of integrating bacterial differential abundance in microbiome data and relevant covariates is effective and feasible.
Collapse
Affiliation(s)
- Jian Huang
- School of Food Science and Technology, Shihezi University, Shihezi, Xinjiang, China
- Key Laboratory of Xinjiang Special Probiotics and Dairy Technology, Shihezi University, Shihezi, Xinjiang, China
| | - Yanzhuan Lu
- School of Food Science and Technology, Shihezi University, Shihezi, Xinjiang, China
- Key Laboratory of Xinjiang Special Probiotics and Dairy Technology, Shihezi University, Shihezi, Xinjiang, China
| | - Fengwei Tian
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Yongqing Ni
- School of Food Science and Technology, Shihezi University, Shihezi, Xinjiang, China
- Key Laboratory of Xinjiang Special Probiotics and Dairy Technology, Shihezi University, Shihezi, Xinjiang, China
| |
Collapse
|
29
|
Wang Z, Shang P, Song X, Wu M, Zhang T, Zhao Q, Zhu S, Qiao Y, Zhao F, Zhang R, Wang J, Yu Y, Han H, Dong H. Alterations in Ileal Microbiota and Fecal Metabolite Profiles of Chickens with Immunity to Eimeria mitis. Animals (Basel) 2024; 14:3515. [PMID: 39682480 DOI: 10.3390/ani14233515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 11/19/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
Coccidiosis, caused by different species of Eimeria parasites, is an economically important disease in poultry and livestock worldwide. This study aimed to investigate the changes in the ileal microbiota and fecal metabolites in chickens after repeated infections with low-dose E. mitis. The chickens developed solid immunity against a high dose of E. mitis infection after repeated infections with low-dose E. mitis. The composition of the ileal microbiota and the metabonomics of the Eimeria-immunized group and the control group were detected using 16S rRNA sequencing and liquid chromatography-mass spectrometry (LC-MS). The relative abundance of Neisseria, Erysipelotrichaceae, Incertae sedis, Coprobacter, Capnocytophaga, Bifidobacterium, and the Ruminococcus torques group declined in the Eimeria-immunized chickens, whereas Alloprevotella, Staphylococcus, Haemophilus, and Streptococcus increased. Furthermore, 286 differential metabolites (including N-undecylbenzenesulfonic acid, 1,25-dihydroxyvitamin D3, gluconic acid, isoleucylproline, proline, and 1-kestose) and 19 significantly altered metabolic pathways (including galactose metabolism, ABC transporters, starch and sucrose metabolism, the ErbB signaling pathway, and the MAPK signaling pathway) were identified between the Eimeria-immunized group and the control group. These discoveries will help us learn more about the composition and dynamics of the gut microbiota as well as the metabolic changes in chickens infected with Eimeria spp.
Collapse
Affiliation(s)
- Zhongchuang Wang
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Minhang, Shanghai 200241, China
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Peiyao Shang
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Minhang, Shanghai 200241, China
| | - Xingju Song
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Minghui Wu
- Beijing Yuanda Spark Medical Technology Co., Ltd., Beijing 102615, China
| | - Tong Zhang
- Beijing Yuanda Spark Medical Technology Co., Ltd., Beijing 102615, China
| | - Qiping Zhao
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Minhang, Shanghai 200241, China
| | - Shunhai Zhu
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Minhang, Shanghai 200241, China
| | - Yu Qiao
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Minhang, Shanghai 200241, China
| | - Fanghe Zhao
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Minhang, Shanghai 200241, China
| | - Ruiting Zhang
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Minhang, Shanghai 200241, China
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Jinwen Wang
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Minhang, Shanghai 200241, China
| | - Yu Yu
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Minhang, Shanghai 200241, China
| | - Hongyu Han
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Minhang, Shanghai 200241, China
| | - Hui Dong
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Minhang, Shanghai 200241, China
| |
Collapse
|
30
|
Wang R, Song J, Cai M, Xue Y, Liu J, Zuo N, De Felici M, Wang J, Shen W, Sun X. Gut microbiota modulation by L-Fucose as a strategy to alleviate Ochratoxin A toxicity on primordial follicle formation. JOURNAL OF HAZARDOUS MATERIALS 2024; 480:136469. [PMID: 39536347 DOI: 10.1016/j.jhazmat.2024.136469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/16/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
In this study, we investigated the potential benefits of L-Fucose administration to pregnant mice exposed to Ochratoxin A (OTA), a widespread mycotoxin, producing ovarian damage in offspring. The results showed that administration of 3.5 μg/d OTA induced alterations in intestinal tissues and gut microbiota of pregnant mice, leading to heightened local and systemic inflammation. This inflammatory affected the ovaries of their 3 dpp offspring, in which elevated levels of LPS and ROS were found associated to significant decreased oocyte count and impaired primordial follicle assembly. Moreover, mRNA-Seq analysis showed significant changes in ovarian transcriptomes linked to various GO terms and KEGG pathways, notably ferroptosis, a recognized form of cell death observed. Interestingly, administration of 0.3 g/kg b. w. L-Fucose following OTA exposure mitigated these effects on intestinal tissues and gut microbiota in mothers and on the offspring's ovaries. Similar benefits were obtained by gut microbiota transplantation from L-Fucose-treated pregnant females into OTA-exposed mothers. These findings suggest that inflammatory impact of OTA on maternal intestine/gut can pass to the fetus causing offspring ovary defects and support the use of L-Fucose as adjuvant to counteract the adverse effects of mycotoxins on the gut microbiota, particularly reference to those affecting reproductive organs.
Collapse
Affiliation(s)
- Ruiting Wang
- College of Life Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao 266109, China
| | - Jie Song
- College of Life Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao 266109, China
| | - Muyu Cai
- College of Life Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao 266109, China
| | - Yuan Xue
- College of Life Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao 266109, China
| | - Jing Liu
- Central laboratory of Qingdao Agricultural University, Qingdao 266109, China
| | - Ning Zuo
- College of Life Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao 266109, China
| | - Massimo De Felici
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome 00133, Italy
| | - Junjie Wang
- College of Life Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao 266109, China
| | - Wei Shen
- College of Life Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao 266109, China
| | - Xiaofeng Sun
- College of Life Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao 266109, China.
| |
Collapse
|
31
|
Kapoor B, Biswas P, Gulati M, Rani P, Gupta R. Gut microbiome and Alzheimer's disease: What we know and what remains to be explored. Ageing Res Rev 2024; 102:102570. [PMID: 39486524 DOI: 10.1016/j.arr.2024.102570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 10/26/2024] [Accepted: 10/26/2024] [Indexed: 11/04/2024]
Abstract
With advancement in human microbiome research, an increasing number of scientific evidences have endorsed the key role of gut microbiota in the pathogenesis of Alzheimer disease. Microbiome dysbiosis, characterized by altered diversity and composition, as well as rise of pathobionts influence not only various gut disorder but also central nervous system disorders such as AD. On the basis of accumulated evidences of past few years now it is quite clear that the gut microbiota can control the functions of the central nervous system (CNS) through the gut-brain axis, which provides a new prospective into the interactions between the gut and brain. The main focus of this review is on the molecular mechanism of the crosstalk between the gut microbiota and the brain through the gut-brain axis, and on the onset and development of neurological disorders triggered by the dysbiosis of gut microbiota. Due to microbiota dysbiosis the permeability of the gut and blood brain barrier is increased which may mediate or affect AD. Along with this, bacterial population of the gut microbiota can secrete amyloid proteins and lipopolysaccharides in a large quantity which may create a disturbance in the signaling pathways and the formation of proinflammatory cytokines associated with the pathogenesis of AD. These topics are followed by a critical analysis of potential intervention strategies targeting gut microbiota dysbiosis, including the use of probiotics, prebiotics, metabolites, diets and fecal microbiota transplantation. The main purpose of this review includes the summarization and discussion on the recent finding that may explain the role of the gut microbiota in the development of AD. Understanding of these fundamental mechanisms may provide a new insight into the novel therapeutic strategies for AD.
Collapse
Affiliation(s)
- Bhupinder Kapoor
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India.
| | - Pratim Biswas
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, NSW 2007, Australia
| | - Pooja Rani
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Reena Gupta
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| |
Collapse
|
32
|
Olasunkanmi OI, Aremu J, Wong ML, Licinio J, Zheng P. Maternal gut-microbiota impacts the influence of intrauterine environmental stressors on the modulation of human cognitive development and behavior. J Psychiatr Res 2024; 180:307-326. [PMID: 39488009 DOI: 10.1016/j.jpsychires.2024.10.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 11/01/2023] [Accepted: 10/23/2024] [Indexed: 11/04/2024]
Abstract
This review examines the longstanding debate of nature and intrauterine environmental challenges that shapes human development and behavior, with a special focus on the influence of maternal prenatal gut microbes. Recent research has revealed the critical role of the gut microbiome in human neurodevelopment, and evidence suggest that maternal microbiota can impact fetal gene and microenvironment composition, as well as immunophysiology and neurochemical responses. Furthermore, intrauterine neuroepigenetic regulation may be influenced by maternal microbiota, capable of having long-lasting effects on offspring behavior and cognition. By examining the complex relationship between maternal prenatal gut microbes and human development, this review highlights the importance of early-life environmental factors in shaping neurodevelopment and cognition.
Collapse
Affiliation(s)
- Oluwatayo Israel Olasunkanmi
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Institute for Brain Science and Disease, Chongqing Medical University, Chongqing, China; Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education) Chongqing Medical University, Chongqing, China.
| | - John Aremu
- Department of Neuroscience, Chongqing Medical University, Chongqing, China
| | - Ma-Li Wong
- Department of Psychiatry, College of Medicine, Upstate Medical University, Syracuse, NY, USA
| | - Julio Licinio
- Department of Psychiatry, College of Medicine, Upstate Medical University, Syracuse, NY, USA.
| | - Peng Zheng
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Institute for Brain Science and Disease, Chongqing Medical University, Chongqing, China; Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education) Chongqing Medical University, Chongqing, China.
| |
Collapse
|
33
|
Tang H, Du S, Niu Z, Zhang D, Tang Z, Chen H, Chen Z, Zhang M, Xu Y, Sun Y, Fu X, Norback D, Shao J, Zhao Z. Nasal, dermal, oral and indoor dust microbe and their interrelationship in children with allergic rhinitis. BMC Microbiol 2024; 24:505. [PMID: 39614169 DOI: 10.1186/s12866-024-03668-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 11/19/2024] [Indexed: 12/01/2024] Open
Abstract
BACKGROUND Allergic rhinitis (AR) subjects might have their microenvironment changed due to pathogenesis and living environment. Whether the nasal microbe in AR children differs from healthy subjects and how it interplays with dermal, oral and indoor dust microbe needs to be elucidated. METHODS In this case-control study, we analyzed and compared the bacterial characterization and associations in nasal, dermal, oral swab samples and dust samples in 62 children with physician-diagnosed AR(cases) and 51 age- and gender-matched healthy ones with no history of allergic diseases(controls). Full-length 16S rRNA sequencing(swabs) and shotgun metagenomics(dust) were applied. Bacterial diversity, composition, abundance difference characteristics and fast expectation-maximization for microbial source tracking(FEAST) analysis were performed and compared between cases and controls. RESULTS The α-diversity of dust microorganisms in AR was lower than that in control group (P = 0.034), and the β-diversity indices of microorganisms in nasal cavity (P = 0.020), skin (P = 0.001) and dust (P = 0.004) were significantly different from those in control group. At species levels, a total of 10, 15, 12, and 15 bacterial species were differentially enriched in either cases or controls in nasal, dermal, oral, and dust samples, respectively(Linear Discriminant Analysis(LDA) score > 2, P < 0.05). Staphylococcus epidermidis was the single species simultaneously more abundant in nasal, dermal and dust samples in AR children. By FEAST analysis, 8.85% and 10.11% of S. epidermidis in AR dermal and dust samples came from nasal cavity. These proportions were significantly higher than those in controls (2.70% and 3.86%) (P < 0.05). The same significantly higher transfer proportions(P < 0.05) were observed for Staphylococcus aureus enriched in the nasal cavity in AR children. Classification models by random forest regression at species levels showed, bacterial species enriched in indoor dust, nasal and dermal samples had substantial power in distinguishing AR children from healthy ones, with the highest power in the dust samples (AUC = 0.88) followed by nasal(AUC = 0.81) and dermal ones(AUC = 0.80). CONCLUSIONS Our study presented the microbial enrichment characteristics in AR children both in the living environment(dust) and body sites exposed to environment through inhalation(nasal cavity), contact(skin) and ingestion(oral cavity) pathways, respectively. Nasal S.epidermidis and S.aureus had dominant influences on dust and other body sites in AR children.
Collapse
Affiliation(s)
- Hao Tang
- Department of Environmental Health, School of Public Health, Fudan University, NHC Key Laboratory of Health Technology Assessment (Fudan University), Shanghai, 200032, China
| | - Shuang Du
- Department of Environmental Health, School of Public Health, Fudan University, NHC Key Laboratory of Health Technology Assessment (Fudan University), Shanghai, 200032, China
| | - Zhiping Niu
- Department of Environmental Health, School of Public Health, Fudan University, NHC Key Laboratory of Health Technology Assessment (Fudan University), Shanghai, 200032, China
| | - Dongjun Zhang
- Department of Allergy, Wu Xi Branch of Ruijin Hospital Shang Hai Jiao Tong University School of Medicine, Jiangsu, 214028, China
| | - Zhiwei Tang
- Department of Pediatrics, Ruijin Hospital Affiliated to Shang Hai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Han Chen
- Department of Environmental Health, School of Public Health, Fudan University, NHC Key Laboratory of Health Technology Assessment (Fudan University), Shanghai, 200032, China
| | - Zhuoru Chen
- Department of Environmental Health, School of Public Health, Fudan University, NHC Key Laboratory of Health Technology Assessment (Fudan University), Shanghai, 200032, China
| | - Mei Zhang
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou, 510642, China
| | - Yanyi Xu
- Department of Environmental Health, School of Public Health, Fudan University, NHC Key Laboratory of Health Technology Assessment (Fudan University), Shanghai, 200032, China
| | - Yu Sun
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou, 510642, China
| | - Xi Fu
- Guangdong Provincial Engineering Research Center of Public Health Detection and Assessment, School of Public Health, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Dan Norback
- Department of Medical Sciences, Uppsala University, Uppsala, SE-751, Sweden
| | - Jie Shao
- Department of Allergy, Wu Xi Branch of Ruijin Hospital Shang Hai Jiao Tong University School of Medicine, Jiangsu, 214028, China.
| | - Zhuohui Zhao
- Department of Environmental Health, School of Public Health, Fudan University, NHC Key Laboratory of Health Technology Assessment (Fudan University), Shanghai, 200032, China.
- Typhoon Institute/CMA, Shanghai Key Laboratory of Meteorology and Health, IRDR International Center of Excellence on Risk Interconnectivity and Governance on Weather/Climate Extremes Impact and Public Health WMO/IGAC MAP-AQ Asian Office Shanghai, Fudan University, Shanghai, China.
| |
Collapse
|
34
|
Feng M, Zou Z, Shou P, Peng W, Liu M, Li X. Gut microbiota and Parkinson's disease: potential links and the role of fecal microbiota transplantation. Front Aging Neurosci 2024; 16:1479343. [PMID: 39679259 PMCID: PMC11638248 DOI: 10.3389/fnagi.2024.1479343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 11/14/2024] [Indexed: 12/17/2024] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease worldwide and seriously affects the quality of life of elderly patients. PD is characterized by the loss of dopaminergic neurons in the substantia nigra as well as abnormal accumulation of α-synuclein in neurons. Recent research has deepened our understanding of the gut microbiota, revealing that it participates in the pathological process of PD through the gut-brain axis, suggesting that the gut may be the source of PD. Therefore, studying the relationship between gut microbiota and PD is crucial for improving our understanding of the disease's prevention, diagnosis, and treatment. In this review, we first describe the bidirectional regulation of the gut-brain axis by the gut microbiota and the mechanisms underlying the involvement of gut microbiota and their metabolites in PD. We then summarize the different species of gut microbiota found in patients with PD and their correlations with clinical symptoms. Finally, we review the most comprehensive animal and human studies on treating PD through fecal microbiota transplantation (FMT), discussing the challenges and considerations associated with this treatment approach.
Collapse
Affiliation(s)
- Maosen Feng
- NHC Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
- Department of Gastroenterology, National Clinical Key Specialty, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
- School of Life Sciences and Engineering, Southwest University of Science and Technology, Mianyang, China
| | - Zhiyan Zou
- NHC Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| | - Pingping Shou
- School of Life Sciences and Engineering, Southwest University of Science and Technology, Mianyang, China
| | - Wei Peng
- Department of Gastroenterology, National Clinical Key Specialty, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| | - Mingxue Liu
- School of Life Sciences and Engineering, Southwest University of Science and Technology, Mianyang, China
| | - Xiaoan Li
- NHC Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
- Department of Gastroenterology, National Clinical Key Specialty, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| |
Collapse
|
35
|
Bai B, Luo L, Yao F, Sun Q, Chen X, Zheng W, Jiang L, Wang X, Su G. The causal relationship between the human gut microbiota and pyogenic arthritis: a Mendelian randomization study. Front Cell Infect Microbiol 2024; 14:1452480. [PMID: 39660282 PMCID: PMC11629706 DOI: 10.3389/fcimb.2024.1452480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 11/07/2024] [Indexed: 12/12/2024] Open
Abstract
Background Recent studies have indicated the role of the gut microbiota in the progression of osteoarticular diseases, however, the causal relationship between the gut microbiota and pyogenic arthritis remains unclear. There is also a lack of theoretical basis for the application of the gut microbiota in the treatment of pyogenic arthritis. Methods In our study, we utilized the largest genome-wide association study (GWAS) data from the MiBioGen Consortium involving 13,400 participants and extracted summary statistical data of the microbiota metabolic pathways of 7,738 participants of European descent from the Dutch Microbiome Project (DMP) The data of pyogenic arthritis were derived from the FinnGen R10 database, including 1,086 patients and 147,221 controls. We employed the two-sample Mendelian randomization approach to investigate the causal association between the gut microbiota and pyogenic arthritis. Our methods comprised inverse variance weighting, Mendelian Randomization Egger regression, weighted median, and weighted modal methods. Subsequently, polygenic and heterogeneity analyses were conducted. Results At the class level, β-proteobacteria is positively correlated with the risk of pyogenic arthritis. At the order level, Burkholderia is positively associated with the disease. At the genus level, the unclassified genus of Sutterellaceae is positively correlated with the disease, while the unnamed genus of Lachnospiraceae, Rothia, and the unnamed genus of Erysipelotrichaceae are negatively correlated with the disease. In addition, Faecalibacterium and Finegoldia are also negatively correlated with the disease. Sensitivity analysis did not show any abnormal evidence. Conclusion This study indicates that β-proteobacteria, Burkholderiales, and the unclassified genus of Sutterellaceae are associated with an increased risk of the disease, while the unnamed genus of Lachnospiraceae, Rothia, the unnamed genus of Erysipelotrichaceae, Faecalibacterium, and Finegoldia are related to a reduced risk. Future studies are needed to elucidate the specific mechanisms by which these specific bacterial groups affect pyogenic arthritis.
Collapse
Affiliation(s)
- Boliang Bai
- Department of Orthopedics, Affiliated Children’s Hospital of Soochow University, Suzhou, China
| | - Longfei Luo
- Department of Bioinformatics, Center for Systems Biology, School of Biology and Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, China
| | - Feng Yao
- Department of Orthopedics, Affiliated Children’s Hospital of Soochow University, Suzhou, China
| | - Qian Sun
- Department of Orthopedics, Affiliated Children’s Hospital of Soochow University, Suzhou, China
| | - Xingguang Chen
- Department of Orthopedics, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Wen Zheng
- Department of Orthopedics, Affiliated Children’s Hospital of Soochow University, Suzhou, China
| | - Lang Jiang
- Department of Orthopedics, Affiliated Children’s Hospital of Soochow University, Suzhou, China
| | - Xiaodong Wang
- Department of Orthopedics, Affiliated Children’s Hospital of Soochow University, Suzhou, China
| | - Guanghao Su
- Department of Orthopedics, Affiliated Children’s Hospital of Soochow University, Suzhou, China
| |
Collapse
|
36
|
Kim S, Thapa I, Ali H. A novel computational approach for the mining of signature pathways using species co-occurrence networks in gut microbiomes. BMC Microbiol 2024; 24:490. [PMID: 39574009 PMCID: PMC11580338 DOI: 10.1186/s12866-024-03633-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 11/05/2024] [Indexed: 11/25/2024] Open
Abstract
BACKGROUND Advances in metagenome sequencing data continue to enable new methods for analyzing biological systems. When handling microbial profile data, metagenome sequencing has proven to be far more comprehensive than traditional methods such as 16s rRNA data, which rely on partial sequences. Microbial community profiling can be used to obtain key biological insights that pave the way for more accurate understanding of complex systems that are critical for advancing biomedical research and healthcare. However, such attempts have mostly used partial or incomplete data to accurately capture those associations. METHODS This study introduces a novel computational approach for the identification of co-occurring microbial communities using the abundance and functional roles of species-level microbiome data. The proposed approach is then used to identify signature pathways associated with inflammatory bowel disease (IBD). Furthermore, we developed a computational pipeline to identify microbial species co-occurrences from metagenome data at various granularity levels. RESULTS When comparing the IBD group to a control group, we show that certain co-occurring communities of species are enriched for potential pathways. We also show that the identified co-occurring microbial species operate as a community to facilitate pathway enrichment. CONCLUSIONS The obtained findings suggest that the proposed network model, along with the computational pipeline, provide a valuable analytical tool to analyze complex biological systems and extract pathway signatures that can be used to diagnose certain health conditions.
Collapse
Affiliation(s)
- Suyeon Kim
- College of Information Science and Technology, University of Nebraska at Omaha, Omaha, NE, 68182, USA
| | - Ishwor Thapa
- College of Information Science and Technology, University of Nebraska at Omaha, Omaha, NE, 68182, USA
| | - Hesham Ali
- College of Information Science and Technology, University of Nebraska at Omaha, Omaha, NE, 68182, USA.
| |
Collapse
|
37
|
Dai Y, Qian Y, Qu Y, Guan W, Xie J, Wang D, Butler C, Dashper S, Carroll I, Divaris K, Liu Y, Wu D. Longitudinal Microbiome-based Interpretable Machine Learning for Identification of Time-Varying Biomarkers in Early Prediction of Disease Outcomes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.18.619118. [PMID: 39605360 PMCID: PMC11601495 DOI: 10.1101/2024.10.18.619118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Information generated from longitudinally-sampled microbial data has the potential to illuminate important aspects of development and progression for many human conditions and diseases. Identifying microbial biomarkers and their time-varying effects can not only advance our understanding of pathogenetic mechanisms, but also facilitate early diagnosis and guide optimal timing of interventions. However, longitudinal predictive modeling of highly noisy and dynamic microbial data (e.g., metagenomics) poses analytical challenges. To overcome these challenges, we introduce a robust and interpretable machine-learning-based longitudinal microbiome analysis framework, LP-Micro, that encompasses: (i) longitudinal microbial feature screening via a polynomial group lasso, (ii) disease outcome prediction implemented via machine learning methods (e.g., XGBoost, deep neural networks), and (iii) interpretable association testing between time points, microbial features, and disease outcomes via permutation feature importance. We demonstrate in simulations that LP-Micro can not only identify incident disease-related microbiome taxa but also offers improved prediction accuracy compared to existing approaches. Applications of LP-Micro in two longitudinal microbiome studies with clinical outcomes of childhood dental disease and weight loss following bariatric surgery yield consistently high prediction accuracy. The identified critical early predictive time points are informative and aligned with clinical expectations.
Collapse
Affiliation(s)
- Yifan Dai
- Department of Biostatistics, Gillings School of Global Public Health at University of North Carolina at Chapel Hill
| | - Yunzhi Qian
- Department of Nutrition, Gillings School of Global Public Health at University of North Carolina at Chapel Hill
| | - Yixiang Qu
- Department of Biostatistics, Gillings School of Global Public Health at University of North Carolina at Chapel Hill
| | - Wyliena Guan
- Department of Biostatistics, Gillings School of Global Public Health at University of North Carolina at Chapel Hill
| | - Jialiu Xie
- Department of Biostatistics, Gillings School of Global Public Health at University of North Carolina at Chapel Hill
| | - Duan Wang
- North Carolina School of Science and Mathematics
| | | | | | - Ian Carroll
- Department of Nutrition, Gillings School of Global Public Health at University of North Carolina at Chapel Hill
| | - Kimon Divaris
- Department of Pediatric Dentistry and Dental Public Health, Adams School of Dentistry, University of North Carolina at Chapel Hill
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill
| | - Yufeng Liu
- Department of Biostatistics, Gillings School of Global Public Health at University of North Carolina at Chapel Hill
- Department of Statistics and Operations Research, University of North Carolina at Chapel Hill
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill
| | - Di Wu
- Department of Biostatistics, Gillings School of Global Public Health at University of North Carolina at Chapel Hill
- Department of Biomedical Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill
| |
Collapse
|
38
|
Zhang Q, Song J, Wu H, Wang L, Zhuo G, Li H, He S, Pan Y, Liu G. Intratumoral microbiota associates with systemic immune inflammation state in nasopharyngeal carcinoma. Int Immunopharmacol 2024; 141:112984. [PMID: 39173404 DOI: 10.1016/j.intimp.2024.112984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 07/07/2024] [Accepted: 08/16/2024] [Indexed: 08/24/2024]
Abstract
BACKGROUND The nasopharynx serves as a crucial niche for the microbiome of the upper respiratory tract. However, the association between the intratumoral microbiota and host systemic inflammation and immune status in nasopharyngeal carcinoma (NPC) remain uncertain. METHODS We performed 5R 16S rDNA sequencing on NPC tissue samples, followed by diversity analysis, LEfSe differential analysis, and KEGG functional prediction. The analyses were based on indices such as AISI, SIRI, PAR, PLR, and NAR. Correlation analyses between microbes and these indices were performed to identify microbes associated with inflammation and immune status. Additionally, regression analysis based on tumor TNM stage was performed to identify key microbes linked to tumor progression. The head and neck squamous cell carcinoma (HNSC) transcriptome and the paired HNSC microbiome data from TCGA were utilized to validate the analyses. RESULTS The Proteobacteria, Actinobacteria, Firmicutes, and Bacteroidetes were the most enriched phyla in NPC tissues. Microbes within these phyla demonstrated high sensitivity to changes in host systemic inflammation and immune status. Proteobacteria and Firmicutes showed significant differences between inflammation groups. Actinobacteria varied specifically with platelet-related inflammatory indices, and Bacteroidetes genera exhibited significant differences between NAR groups. Corynebacterium and Brevundimonas significantly impacted the T stage of tumors, with a high load of Corynebacterium within tumors associated with a better prognosis CONCLUSION: Our analysis indicates that Proteobacteria play a crucial role in the inflammatory state of NPC, while Bacteroidetes are more sensitive to the tumor immune status.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei 430071, China
| | - Jiangqin Song
- Department of Laboratory Medicine, The First People's Hospital of Tianmen City, Tianmen, Hubei 431700, China
| | - Huiqing Wu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei 430071, China
| | - Liping Wang
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei 430071, China
| | - Guangzheng Zhuo
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei 430071, China
| | - Huashun Li
- Department of Pathology, The First People's Hospital of Tianmen City, Tianmen, Hubei 431700, China
| | - Siyu He
- Department of Laboratory Medicine, The First People's Hospital of Tianmen City, Tianmen, Hubei 431700, China
| | - Yunbao Pan
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei 430071, China; Hubei Engineering Center for Infectious Disease Prevention, Control and Treatment, Wuhan, China.
| | - Guohong Liu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei 430071, China.
| |
Collapse
|
39
|
Xiao S, Zhou W, Caldwell R, Decker S, Oh J, Milstone AM. Association of Neonatal and Maternal Nasal Microbiome Among Neonates in the Intensive Care Unit. Open Forum Infect Dis 2024; 11:ofae644. [PMID: 39544492 PMCID: PMC11561572 DOI: 10.1093/ofid/ofae644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 10/25/2024] [Indexed: 11/17/2024] Open
Abstract
The neonatal nasal microbiota may help protect neonates in the neonatal intensive care unit from pathogen colonization and infection. This preliminary study characterized the biodiversity of nasal microbiota comparing neonates in the neonatal intensive care unit and their mothers, highlighting the potential of strain sharing between mother-neonate pairs.
Collapse
Affiliation(s)
- Shaoming Xiao
- Division of Pediatric Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Wei Zhou
- The Jackson Laboratory, Farmington, Connecticut, USA
| | - Ryan Caldwell
- The Jackson Laboratory, Farmington, Connecticut, USA
| | - Slade Decker
- Division of Pediatric Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Julia Oh
- The Jackson Laboratory, Farmington, Connecticut, USA
| | - Aaron M Milstone
- Division of Pediatric Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
40
|
Eyal N, Williams B, Esvelt KM, Bambauer J. Metagenomic Sequencing for Early Detection of Future Engineered Pandemics: Foreshadowing the Privacy Challenge. Health Secur 2024; 22:466-475. [PMID: 39632779 DOI: 10.1089/hs.2023.0140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024] Open
Affiliation(s)
- Nir Eyal
- Nir Eyal, DPhil, is the Henry Rutgers and Dr. and Mrs. Stanley S. Bergen Professor of Bioethics, Institute for Health, School of Public Health, and Philosophy Department, Rutgers University, New Brunswick, NJ. Bridget Williams, MBBS, MPH, MSt, is a DPhil Candidate, Oxford Uehiro Centre for Practical Ethics, University of Oxford, Oxford, United Kingdom. Kevin M. Esvelt, PhD, is an Associate Professor, Media Laboratory, Massachusetts Institute of Technology, Cambridge, MA. Jane Bambauer, JD, Brechner Eminent Scholar and Professor of Law, University of Florida Levin College of Law, Gainesville, FL
| | - Bridget Williams
- Nir Eyal, DPhil, is the Henry Rutgers and Dr. and Mrs. Stanley S. Bergen Professor of Bioethics, Institute for Health, School of Public Health, and Philosophy Department, Rutgers University, New Brunswick, NJ. Bridget Williams, MBBS, MPH, MSt, is a DPhil Candidate, Oxford Uehiro Centre for Practical Ethics, University of Oxford, Oxford, United Kingdom. Kevin M. Esvelt, PhD, is an Associate Professor, Media Laboratory, Massachusetts Institute of Technology, Cambridge, MA. Jane Bambauer, JD, Brechner Eminent Scholar and Professor of Law, University of Florida Levin College of Law, Gainesville, FL
| | - Kevin M Esvelt
- Nir Eyal, DPhil, is the Henry Rutgers and Dr. and Mrs. Stanley S. Bergen Professor of Bioethics, Institute for Health, School of Public Health, and Philosophy Department, Rutgers University, New Brunswick, NJ. Bridget Williams, MBBS, MPH, MSt, is a DPhil Candidate, Oxford Uehiro Centre for Practical Ethics, University of Oxford, Oxford, United Kingdom. Kevin M. Esvelt, PhD, is an Associate Professor, Media Laboratory, Massachusetts Institute of Technology, Cambridge, MA. Jane Bambauer, JD, Brechner Eminent Scholar and Professor of Law, University of Florida Levin College of Law, Gainesville, FL
| | - Jane Bambauer
- Nir Eyal, DPhil, is the Henry Rutgers and Dr. and Mrs. Stanley S. Bergen Professor of Bioethics, Institute for Health, School of Public Health, and Philosophy Department, Rutgers University, New Brunswick, NJ. Bridget Williams, MBBS, MPH, MSt, is a DPhil Candidate, Oxford Uehiro Centre for Practical Ethics, University of Oxford, Oxford, United Kingdom. Kevin M. Esvelt, PhD, is an Associate Professor, Media Laboratory, Massachusetts Institute of Technology, Cambridge, MA. Jane Bambauer, JD, Brechner Eminent Scholar and Professor of Law, University of Florida Levin College of Law, Gainesville, FL
| |
Collapse
|
41
|
Wang J, Huang H, Yang H, Wang S, Li M, Zhu Z, Trumble JT, Di N, Zang L. Heavy metal exposure reduces larval gut microbiota diversity of the rice striped stem borer, Chilo suppressalis. ECOTOXICOLOGY (LONDON, ENGLAND) 2024; 33:973-983. [PMID: 39066991 DOI: 10.1007/s10646-024-02792-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 07/12/2024] [Indexed: 07/30/2024]
Abstract
Cadmium (Cd), a widely distributed environmental pollutant in agroecosystems, causes negative effects on crops and herbivores through bottom-up processes. The gut microbial community of an insect can play a critical role in response to metal stress. To understand how microbiota affect the stress responses of organisms to heavy metals in agroecosystems, we initially used 16S rRNA sequencing to characterize the larval gut microbiota of Chilo suppressalis, an important agricultural pest, exposed to a diet containing Cd. The species richness, diversity, and composition of the gut microbial community was then analyzed. Results revealed that while the richness (Chao1 and ACE) of gut microbiota in larvae exposed to Cd was not significantly affected, diversity (Shannon and Simpson) was reduced due to changes in species distribution and relative abundance. Overall, the most abundant genus was Enterococcus, while the abundance of the genera Micrococcaceae and Faecalibaculum in the control significantly superior to that in Cd-exposed pests. Phylogenetic investigation of microbial communities by the reconstruction of unobserved states (PICRUSt) showed that the intestinal microorganisms appear to participate in 34 pathways, especially those used in environmental information processing and the metabolism of the organism. This study suggests that the gut microbiota of C. suppressalis are significantly impacted by Cd exposure and highlights the importance of the gut microbiome in host stress responses and negative effects of Cd pollution in agroecosystems.
Collapse
Affiliation(s)
- Jie Wang
- Key Laboratory of Natural Enemies Insects, Ministry of Agriculture and Rural Affairs; Key Laboratory of Environment Friendly Management on Fruit and Vegetable Pests in North China (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs; Institute of Plant Protection, Beijing Academy of Agriculture and Forestry Sciences, 100097, Beijing, China
- College of Plant Protection, Nanjing Agricultural University, Nanjing, 210095, China
| | - Hexi Huang
- Key Laboratory of Natural Enemies Insects, Ministry of Agriculture and Rural Affairs; Key Laboratory of Environment Friendly Management on Fruit and Vegetable Pests in North China (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs; Institute of Plant Protection, Beijing Academy of Agriculture and Forestry Sciences, 100097, Beijing, China
- State Key Laboratory of Green Pesticide; Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang, 550025, China
| | - Hailin Yang
- Yunnan Tobacco Company, Yuxi Branch, Yuxi, 653100, China
| | - Su Wang
- Key Laboratory of Natural Enemies Insects, Ministry of Agriculture and Rural Affairs; Key Laboratory of Environment Friendly Management on Fruit and Vegetable Pests in North China (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs; Institute of Plant Protection, Beijing Academy of Agriculture and Forestry Sciences, 100097, Beijing, China
| | - Mengnan Li
- Key Laboratory of Natural Enemies Insects, Ministry of Agriculture and Rural Affairs; Key Laboratory of Environment Friendly Management on Fruit and Vegetable Pests in North China (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs; Institute of Plant Protection, Beijing Academy of Agriculture and Forestry Sciences, 100097, Beijing, China
| | - Zhengyang Zhu
- Key Laboratory of Natural Enemies Insects, Ministry of Agriculture and Rural Affairs; Key Laboratory of Environment Friendly Management on Fruit and Vegetable Pests in North China (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs; Institute of Plant Protection, Beijing Academy of Agriculture and Forestry Sciences, 100097, Beijing, China
| | - John T Trumble
- Department of Entomology, University of California, Riverside, CA, 92521, USA
| | - Ning Di
- Key Laboratory of Natural Enemies Insects, Ministry of Agriculture and Rural Affairs; Key Laboratory of Environment Friendly Management on Fruit and Vegetable Pests in North China (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs; Institute of Plant Protection, Beijing Academy of Agriculture and Forestry Sciences, 100097, Beijing, China.
| | - Liansheng Zang
- State Key Laboratory of Green Pesticide; Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang, 550025, China.
| |
Collapse
|
42
|
Adler I, Kotta J, Robal M, Humayun S, Vene K, Tuvikene R. Valorization of Baltic Sea farmed blue mussels: Chemical profiling and prebiotic potential for nutraceutical and functional food development. Food Chem X 2024; 23:101736. [PMID: 39253010 PMCID: PMC11381619 DOI: 10.1016/j.fochx.2024.101736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 08/08/2024] [Accepted: 08/12/2024] [Indexed: 09/11/2024] Open
Abstract
The severe eutrophication of the Baltic Sea requires mussel (Mytilus spp.) farming to remove nutrients, but farming in a low salinity environment results in smaller mussels that require value enhancement to be economically viable. This study evaluates the biomass valorisation of smaller Baltic mussels, focusing on the extraction of oil, protein and glycogen. It analyses the amino acid profiles, oil and fatty acid contents and glycogen levels of the mussels, as well as their prebiotic properties on beneficial gut bacteria. In addition, the study improves the extraction of bioactive compounds through enzymatic hydrolysis. Results indicate significant seasonal differences, with summer mussels having higher meat and lower ash content, and a rich content of essential fatty acids, particularly omega-3, and amino acids, underscoring the mussels' sustainability as a food source. The enzymatically treated biomass exhibited notable prebiotic activity, proposing health-promoting benefits. The study underscores the valorization of Baltic mussel biomass, highlighting its role in health, nutrition, and environmental sustainability.
Collapse
Affiliation(s)
- Indrek Adler
- Estonian Maritime Academy, Tallinn University of Technology, Kopli 101, 11712 Tallinn, Estonia
| | - Jonne Kotta
- Estonian Maritime Academy, Tallinn University of Technology, Kopli 101, 11712 Tallinn, Estonia
- Estonian Marine Institute, University of Tartu, Mäealuse 14, 12618 Tallinn, Estonia
| | - Marju Robal
- School of Natural Sciences and Health, Tallinn University, Narva mnt 25, 10120 Tallinn, Estonia
| | - Sanjida Humayun
- School of Natural Sciences and Health, Tallinn University, Narva mnt 25, 10120 Tallinn, Estonia
| | - Kristel Vene
- Tallinn University of Technology, School of Science, Department of Chemistry and Biotechnology, Akadeemia tee 15, 12618 Tallinn, Estonia
| | - Rando Tuvikene
- School of Natural Sciences and Health, Tallinn University, Narva mnt 25, 10120 Tallinn, Estonia
| |
Collapse
|
43
|
Feng C, Jia H, Wang H, Wang J, Lin M, Hu X, Yu C, Song H, Wang L. MicroNet-MIMRF: a microbial network inference approach based on mutual information and Markov random fields. BIOINFORMATICS ADVANCES 2024; 4:vbae167. [PMID: 39526038 PMCID: PMC11549015 DOI: 10.1093/bioadv/vbae167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 10/19/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024]
Abstract
Motivation The human microbiome, comprises complex associations and communication networks among microbial communities, which are crucial for maintaining health. The construction of microbial networks is vital for elucidating these associations. However, existing microbial networks inference methods cannot solve the issues of zero-inflation and non-linear associations. Therefore, necessitating novel methods to improve the accuracy of microbial networks inference. Results In this study, we introduce the Microbial Network based on Mutual Information and Markov Random Fields (MicroNet-MIMRF) as a novel approach for inferring microbial networks. Abundance data of microbes are modeled through the zero-inflated Poisson distribution, and the discrete matrix is estimated for further calculation. Markov random fields based on mutual information are used to construct accurate microbial networks. MicroNet-MIMRF excels at estimating pairwise associations between microbes, effectively addressing zero-inflation and non-linear associations in microbial abundance data. It outperforms commonly used techniques in simulation experiments, achieving area under the curve values exceeding 0.75 for all parameters. A case study on inflammatory bowel disease data further demonstrates the method's ability to identify insightful associations. Conclusively, MicroNet-MIMRF is a powerful tool for microbial network inference that handles the biases caused by zero-inflation and overestimation of associations. Availability and implementation The MicroNet-MIMRF is provided at https://github.com/Fionabiostats/MicroNet-MIMRF.
Collapse
Affiliation(s)
- Chenqionglu Feng
- Department of Epidemiology and Health Statistics, School of Public Health, China Medical University, Shenyang 110122, China
- Department of Infectious Disease Prevention and Control, Chinese PLA Center for Disease Control and Prevention, Beijing 100071, China
| | - Huiqun Jia
- Department of Infectious Disease Prevention and Control, Chinese PLA Center for Disease Control and Prevention, Beijing 100071, China
| | - Hui Wang
- Department of Infectious Disease Prevention and Control, Chinese PLA Center for Disease Control and Prevention, Beijing 100071, China
| | - Jiaojiao Wang
- The State Key Laboratory of Multimodal Artificial Intelligence Systems, Institute of Automation Chinese Academy of Sciences, Beijing 100190, China
| | - Mengxuan Lin
- The Academy of Military Medical Sciences, Academy of Military Science of Chinese People’s Liberation Army, Beijing 100071, China
| | - Xiaoyan Hu
- Department of Infectious Disease Prevention and Control, Chinese PLA Center for Disease Control and Prevention, Beijing 100071, China
| | - Chenjing Yu
- Department of Infectious Disease Prevention and Control, Chinese PLA Center for Disease Control and Prevention, Beijing 100071, China
| | - Hongbin Song
- Department of Infectious Disease Prevention and Control, Chinese PLA Center for Disease Control and Prevention, Beijing 100071, China
| | - Ligui Wang
- Department of Infectious Disease Prevention and Control, Chinese PLA Center for Disease Control and Prevention, Beijing 100071, China
| |
Collapse
|
44
|
Liu H, Zhang J, Rao Y, Jin S, Zhang C, Bai D. Intratumoral microbiota: an emerging force in diagnosing and treating hepatocellular carcinoma. Med Oncol 2024; 41:300. [PMID: 39453562 DOI: 10.1007/s12032-024-02545-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 10/17/2024] [Indexed: 10/26/2024]
Abstract
Hepatocellular carcinoma (HCC) ranks among the most prevalent types of cancer in the world and its incidence and mortality are increasing year by year, frequently diagnosed at an advanced stage. Traditional treatments such as surgery, chemotherapy, and radiotherapy have limited efficacy, so new diagnostic and treatment strategies are urgently needed. Recent research has discovered that intratumoral microbiota significantly influences the development, progression, and metastasis of HCC by modulating inflammation, immune responses, and cellular signaling pathways. Intratumoral microbiota contributes to the pathologic process of HCC by influencing the tumor microenvironment and altering the function of immune system. This article reviews the mechanism of intratumoral microbiota in HCC and anticipates the future possibilities of intratumoral microbiota-based therapeutic strategies for HCC management. This emerging field provides fresh insights into early diagnosis and personalized approaches for HCC while holding substantial clinical application potential to improve patient outcomes and tailor interventions to individual tumor profiles.
Collapse
Affiliation(s)
- Huanxiang Liu
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225001, China
| | - Jiahao Zhang
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225001, China
| | - Yuye Rao
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225001, China
| | - Shengjie Jin
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225001, China
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital, Yangzhou, 225001, China
| | - Chi Zhang
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225001, China
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital, Yangzhou, 225001, China
| | - Dousheng Bai
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225001, China.
- Department of Hepatobiliary Surgery, Northern Jiangsu People's Hospital, Yangzhou, 225001, China.
| |
Collapse
|
45
|
Dai DLY, Petersen C, Turvey SE. Reduce, reinforce, and replenish: safeguarding the early-life microbiota to reduce intergenerational health disparities. Front Public Health 2024; 12:1455503. [PMID: 39507672 PMCID: PMC11537995 DOI: 10.3389/fpubh.2024.1455503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 10/02/2024] [Indexed: 11/08/2024] Open
Abstract
Socioeconomic (SE) disparity and health inequity are closely intertwined and associated with cross-generational increases in the rates of multiple chronic non-communicable diseases (NCDs) in North America and beyond. Coinciding with this social trend is an observed loss of biodiversity within the community of colonizing microbes that live in and on our bodies. Researchers have rightfully pointed to the microbiota as a key modifiable factor with the potential to ease existing health inequities. Although a number of studies have connected the adult microbiome to socioeconomic determinants and health outcomes, few studies have investigated the role of the infant microbiome in perpetuating these outcomes across generations. It is an essential and important question as the infant microbiota is highly sensitive to external forces, and observed shifts during this critical window often portend long-term outcomes of health and disease. While this is often studied in the context of direct modulators, such as delivery mode, family size, antibiotic exposure, and breastfeeding, many of these factors are tied to underlying socioeconomic and/or cross-generational factors. Exploring cross-generational socioeconomic and health inequities through the lens of the infant microbiome may provide valuable avenues to break these intergenerational cycles. In this review, we will focus on the impact of social inequality in infant microbiome development and discuss the benefits of prioritizing and restoring early-life microbiota maturation for reducing intergenerational health disparities.
Collapse
Affiliation(s)
| | | | - Stuart E. Turvey
- Department of Pediatrics, BC Children’s Hospital, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
46
|
Liu J, Chen Y, Laurent I, Yang P, Xiao X, Li X. Gestational diabetes exacerbates intrauterine microbial exposure induced intestinal microbiota change in offspring contributing to increased immune response. Nutr Diabetes 2024; 14:87. [PMID: 39424815 PMCID: PMC11489853 DOI: 10.1038/s41387-024-00346-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 09/20/2024] [Accepted: 10/09/2024] [Indexed: 10/21/2024] Open
Abstract
BACKGROUND maternal health during pregnancy can affect the intestinal microbial community of offspring, but currently the impact of intrauterine environmental changes resulting from gestational diabetes mellitus (GDM) on the microbiota of offspring as well as its interaction with the immune system remains unclear. AIMS to explore the impact of intrauterine microbial exposure during pregnancy of gestational diabetes mellitus on the development of neonate's intestinal microbiota and activation of immune responses. METHODS Levels of lipopolysaccharides in cord blood from GDM and expression of microbial recognition-related proteins in the placenta were measured. To evaluate embryonic intestinal colonization, pregnant mice with GDM were administered with labeled Escherichia coli or Lactobacillus. The intestinal colonization of pups was analyzed through 16S rRNA gene sequencing and labeled microbial culture. Additionally, memory T lymphocyte and dendritic cell co-culture experiments were conducted to elucidate the immune memory of intestinal microbes during the embryonic stages. RESULT Gestational diabetes mellitus led to elevated umbilical cord blood LPS level and increased GFP labeled Escherichia coli in the offspring's intestine after gestational microbial exposure. The mouse model of GDM exhibited increased immune markers including TLR4, TLR5, IL-22 and IL-23 in the placenta and a recall response from memory T cells in offspring's intestines, with similar observations found in human experiments. Furthermore, reduced intestinal microbiome diversity and an increased ratio of Firmicutes/Bacteroidetes was found in GDM progeny, with the stability of bacterial colonization been interfered. CONCLUSIONS Our investigation has revealed a noteworthy correlation between gestational diabetes and intrauterine microbial exposure, as well as alterations in the neonatal microbiota and activation of immune responses. These findings highlight the gestational diabetes's role on offspring's gut microbiota and immune system interactions with early-life pathogen exposure.
Collapse
Affiliation(s)
- Juncheng Liu
- Department of Endocrinology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Gastroenterology, Chongqing University Cancer Hospital, School of Medicine, Chongqing University, Chongqing, China
| | - Yan Chen
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Endocrinology and Nephrology, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing, China
| | - Irakoze Laurent
- Department of Endocrinology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ping Yang
- Yongchuan Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Xiaoqiu Xiao
- Department of Endocrinology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Xinyu Li
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
- Department of Pharmacy, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
47
|
Ma X, Li J, Li Z, Chen B, Ling Z, Feng S, Zhong Z, Peng G, Wang Y, Jiang Y, Gu Y. Analysis of fungal diversity in the feces of Arborophila rufipectus. Front Vet Sci 2024; 11:1430518. [PMID: 39469585 PMCID: PMC11514364 DOI: 10.3389/fvets.2024.1430518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 09/26/2024] [Indexed: 10/30/2024] Open
Abstract
Background Intestinal fungal composition plays a crucial role in modulating host health, and thus is of great significance in the conservation of endangered bird species. However, research on gut fungal composition in birds is limited. Therefore, in this study, we aimed to examine gut fungal community and potential fecal pathogen composition in wild Arborophila rufipectus. Methods Fecal samples were collected from the habitats of wild A. rufipectus and Lophura nycthemera (a widely distributed species belonging to the same family as A. rufipectus) in summer and autumn. Thereafter, RNA was collected and the internal transcribed spacer rRNA gene was sequenced via high-throughput sequencing to investigate seasonal variations in intestinal core fungi, microbial fungi, and potential pathogenic fungi. Results The gut microbiota of A. rufipectus and L. nycthemera were highly similar and mainly consisted of three phyla, Ascomycota (58.46%), Basidiomycota (28.80%), and Zygomycota (3.56%), which accounted for 90.82% of the fungal community in all the samples. Further, the predominant genera were Ascomycota_unclassified (12.24%), Fungi_unclassified (8.37%), Davidiella (5.18%), Helotiales_unclassified (2.76%), Wickerhamomyces (1.84%), and Pleosporales_unclassified (1.14%), and the potential fecal pathogens identified included Candida, Cryptococcus, Trichosporon, and Malassezia. Conclusion Our results provide evidence that the diversity of intestinal fungi in the endangered species, A. rufipectus, is similar to that in the common species, L. nycthemera, and may serve as a basis for monitoring the status of A. rufipectus and for developing conservation measures.
Collapse
Affiliation(s)
- Xiaoping Ma
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Junshu Li
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Zhiguo Li
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Benping Chen
- Sichuan Laojunshan National Nature Reserve, Yibin, China
| | - Zhenwen Ling
- Sichuan Laojunshan National Nature Reserve, Yibin, China
| | - Shenglin Feng
- Sichuan Laojunshan National Nature Reserve, Yibin, China
| | - Zhijun Zhong
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Guangneng Peng
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Ya Wang
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yaozhang Jiang
- Bioengineering Department, Sichuan Water Conservancy Vocational College, Chengdu, China
| | - Yu Gu
- College of Life Sciences, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
48
|
Pepoyan A. Gut Akkermansia muciniphila, Prevotellaceae, and Enterobacteriaceae spp. as Possible Markers in Women-Related Nutritional and Clinical Trials: Familial Mediterranean Fever Disease. WOMEN'S HEALTH REPORTS (NEW ROCHELLE, N.Y.) 2024; 5:785-793. [PMID: 39439767 PMCID: PMC11491588 DOI: 10.1089/whr.2024.0076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 08/28/2024] [Indexed: 10/25/2024]
Abstract
Background Studies have shown that the gut microbiota of healthy men and men with familial Mediterranean fever (FMF) disease respond differently to placebo. Given the fact that the composition of the gut microbiota is different in men and women, this study aimed to describe in detail the placebo response of the gut microbiota in healthy and FMF women. Materials and Methods The bacterial response to placebo was fully evaluated on a previous PhyloChip™ DNA microarray-based assay (GEO Series; accession number GSE111835). Results The change in the total number of operational taxonomic units in healthy women exposed to placebo is more than that of healthy men, in contrast to FMF people (704 vs. 140 and 409 vs. 7560, respectively [p < 0.05]). Gut Firmicutes diversities are more sensitive to placebo, whereas Akkermansia muciniphila remained unchanged after the placebo administration for both healthy and FMF people. Gut Prevotellaceae and Enterobacteriaceae diversities of healthy subjects and FMF women are also almost unchanged from placebo. Meanwhile, only 56.35% of gut Enterobacteriaceae diversities in FMF men were placebo resistant. Conclusion The response to a placebo varies depending on a person's gender and health status. Healthy and FMF women's placebo study groups could be avoided by excluding placebo-sensitive 704 of 18,725 and 409 of 18,725 bacterial diversities, respectively. Because the placebo causes changes in all gut bacterial phyla in healthy and FMF women, and only the representatives of Enterobacteriaceae and Prevotellaceae families and A. muciniphila spp. are not affected by placebo, these bacteria can be considered as possible markers in women-related nutritional/clinical trials. Data on the response of the gut microbiota in healthy women to placebo might be used in studies of diseases other than FMF. The response of gut bacteria from different taxonomic affiliations to placebo may provide a basis for uncovering the role of these bacteria in the gut-brain axis.
Collapse
Affiliation(s)
- Astghik Pepoyan
- Food Safety and Biotechnology Department, Scientific Research Institute of Food Science and Biotechnology, Armenian National Agrarian University, Yerevan, Armenia
- International Association for Human and Animals Health Improvement, Yerevan, Armenia
| |
Collapse
|
49
|
Hsu CY, Khachatryan LG, Younis NK, Mustafa MA, Ahmad N, Athab ZH, Polyanskaya AV, Kasanave EV, Mirzaei R, Karampoor S. Microbiota-derived short chain fatty acids in pediatric health and diseases: from gut development to neuroprotection. Front Microbiol 2024; 15:1456793. [PMID: 39439941 PMCID: PMC11493746 DOI: 10.3389/fmicb.2024.1456793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 09/12/2024] [Indexed: 10/25/2024] Open
Abstract
The infant gut microbiota undergoes significant changes during early life, which are essential for immune system maturation, nutrient absorption, and metabolic programming. Among the various microbial metabolites, short-chain fatty acids (SCFAs), primarily acetate, propionate, and butyrate, produced through the fermentation of dietary fibers by gut bacteria, have emerged as critical modulators of host-microbiota interactions. SCFAs serve as energy sources for colonic cells and play pivotal roles in regulating immune responses, maintaining gut barrier integrity, and influencing systemic metabolic pathways. Recent research highlights the potential neuroprotective effects of SCFAs in pediatric populations. Disruptions in gut microbiota composition and SCFA production are increasingly associated with a range of pediatric health issues, including obesity, allergic disorders, inflammatory bowel disease (IBD), and neurodevelopmental disorders. This review synthesizes current knowledge on the role of microbiota-derived SCFAs in pediatric health, emphasizing their contributions from gut development to neuroprotection. It also underscores the need for further research to unravel the precise mechanisms by which SCFAs influence pediatric health and to develop targeted interventions that leverage SCFAs for therapeutic benefits.
Collapse
Affiliation(s)
- Chou-Yi Hsu
- Thunderbird School of Global Management, Arizona State University Tempe Campus, Phoenix, AZ, United States
| | - Lusine G. Khachatryan
- Department of Pediatric Diseases, N. F. Filatov Clinical Institute of Children’s Health, I. M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | | | - Mohammed Ahmed Mustafa
- Department of Medical Laboratory Techniques, University of Imam Jafar Al-Sadiq, College of Technology, Baghdad, Iraq
| | - Nabeel Ahmad
- Department of Biotechnology and Genetics, Jain (Deemed-to-be) University, Bengaluru, Karnataka, India
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan, India
- Department of Biotechnology, School of Allied Sciences, Dev Bhoomi Uttarakhand University Dehradun, Uttarakhand, India
| | - Zainab H. Athab
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq
| | - Angelina V. Polyanskaya
- Department of Pediatric Diseases, N. F. Filatov Clinical Institute of Children’s Health, I. M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Elena Victorovna Kasanave
- Department of Pediatric Diseases, N. F. Filatov Clinical Institute of Children’s Health, I. M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Rasoul Mirzaei
- Venom and Biotherapeutics Molecules Lab, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Sajad Karampoor
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
50
|
Tanegashima T, Shiota M, Tsukahara S, Mutaguch J, Goto S, Kobayashi S, Matsumoto T, Eto M. Impact of proton pump inhibitors on the efficacy of androgen receptor signaling inhibitors in metastatic castration-resistant prostate cancer patients. Prostate 2024; 84:1329-1335. [PMID: 38988005 DOI: 10.1002/pros.24769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 06/29/2024] [Accepted: 07/03/2024] [Indexed: 07/12/2024]
Abstract
BACKGROUND Proton pump inhibitors (PPIs) are widely used due to their affordability and minimal severe side effects. However, their influence on the efficacy of cancer treatments, particularly androgen receptor signaling inhibitors (ARSIs), remains unclear. This study investigates the impact of PPI usage on the treatment outcomes in patients with metastatic castration-resistant prostate cancer (mCRPC). METHODS A total of 117 mCRPC patients were retrospectively analyzed and divided into two groups based on the concomitant use of PPI at the initiation of ARSI treatment: PPI+ (n = 38) and PPI- (n = 79). Patient characteristics, including age at ARSI treatment administered, prostate-specific antigen (PSA) value at ARSI treatment administered, International Society of Urological Pathology grade group at prostate biopsy, metastatic site at ARSI treatment administered, prior docetaxel (DTX) treatment, and type of ARSI (abiraterone acetate or enzalutamide) were recorded. Progression-free survival (PFS), overall survival (OS), and PSA response rates were compared between the two groups. Patients were further stratified by clinical background to compare PFS and OS between the two groups. RESULTS The PPI- group exhibited significantly extended PFS and a trend toward improved OS. For PSA response (reduction of 50% or more from baseline), the rates were 62.3% and 45.9% in the PPI- group and the PPI+ group, respectively. For deep PSA response (reductions of 90% or more from baseline), the rates were 36.4% and 24.3% in the PPI- group and the PPI+ group, respectively. The effects were consistent across subgroups divided by prior DTX treatment and type of ARSI administered. CONCLUSIONS The administration of PPIs appears to diminish the therapeutic efficacy of ARSIs in mCRPC patients. Further prospective studies are needed to confirm these findings and explore the biological mechanisms involved.
Collapse
Affiliation(s)
- Tokiyoshi Tanegashima
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masaki Shiota
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shigehiro Tsukahara
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Jun Mutaguch
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shunsuke Goto
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Satoshi Kobayashi
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takashi Matsumoto
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masatoshi Eto
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|