1
|
Joos R, Boucher K, Lavelle A, Arumugam M, Blaser MJ, Claesson MJ, Clarke G, Cotter PD, De Sordi L, Dominguez-Bello MG, Dutilh BE, Ehrlich SD, Ghosh TS, Hill C, Junot C, Lahti L, Lawley TD, Licht TR, Maguin E, Makhalanyane TP, Marchesi JR, Matthijnssens J, Raes J, Ravel J, Salonen A, Scanlan PD, Shkoporov A, Stanton C, Thiele I, Tolstoy I, Walter J, Yang B, Yutin N, Zhernakova A, Zwart H, Doré J, Ross RP. Examining the healthy human microbiome concept. Nat Rev Microbiol 2025; 23:192-205. [PMID: 39443812 DOI: 10.1038/s41579-024-01107-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/04/2024] [Indexed: 10/25/2024]
Abstract
Human microbiomes are essential to health throughout the lifespan and are increasingly recognized and studied for their roles in metabolic, immunological and neurological processes. Although the full complexity of these microbial communities is not fully understood, their clinical and industrial exploitation is well advanced and expanding, needing greater oversight guided by a consensus from the research community. One of the most controversial issues in microbiome research is the definition of a 'healthy' human microbiome. This concept is complicated by the microbial variability over different spatial and temporal scales along with the challenge of applying a unified definition to the spectrum of healthy microbiome configurations. In this Perspective, we examine the progress made and the key gaps that remain to be addressed to fully harness the benefits of the human microbiome. We propose a road map to expand our knowledge of the microbiome-health relationship, incorporating epidemiological approaches informed by the unique ecological characteristics of these communities.
Collapse
Affiliation(s)
- Raphaela Joos
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Katy Boucher
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Aonghus Lavelle
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Manimozhiyan Arumugam
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Martin J Blaser
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ, USA
| | - Marcus J Claesson
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Gerard Clarke
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - Paul D Cotter
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Teagasc Food Research Centre and VistaMilk SFI Research Centre, Moorepark, Fermoy, Moorepark, Ireland
| | - Luisa De Sordi
- Centre de Recherche Saint Antoine, Sorbonne Université, INSERM, Paris, France
| | | | - Bas E Dutilh
- Institute of Biodiversity, Faculty of Biological Sciences, Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, Jena, Germany
- Theoretical Biology and Bioinformatics, Department of Biology, Science for Life, Utrecht University, Utrecht, The Netherlands
| | - Stanislav D Ehrlich
- Université Paris-Saclay, INRAE, MetaGenoPolis (MGP), Jouy-en-Josas, France
- Department of Clinical and Movement Neurosciences, University College London, London, UK
| | - Tarini Shankar Ghosh
- Department of Computational Biology, Indraprastha Institute of Information Technology Delhi (IIIT-Delhi), New Delhi, India
| | - Colin Hill
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Christophe Junot
- Département Médicaments et Technologies pour La Santé (DMTS), Université Paris-Saclay, CEA, INRAE, MetaboHUB, Gif-sur-Yvette, France
| | - Leo Lahti
- Department of Computing, University of Turku, Turku, Finland
| | - Trevor D Lawley
- Host-Microbiota Interactions Laboratory, Wellcome Sanger Institute, Hinxton, UK
| | - Tine R Licht
- National Food Institute, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Emmanuelle Maguin
- Université Paris-Saclay, INRAE, AgroParisTech, MICALIS, Jouy-en-Josas, France
| | - Thulani P Makhalanyane
- Department of Microbiology, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
| | - Julian R Marchesi
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Jelle Matthijnssens
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute, Leuven, Belgium
| | - Jeroen Raes
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute, Leuven, Belgium
- Vlaams Instituut voor Biotechnologie (VIB) Center for Microbiology, Leuven, Belgium
| | - Jacques Ravel
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Anne Salonen
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Pauline D Scanlan
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Andrey Shkoporov
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Catherine Stanton
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Teagasc Food Research Centre and VistaMilk SFI Research Centre, Moorepark, Fermoy, Moorepark, Ireland
| | - Ines Thiele
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Medicine, University of Ireland, Galway, Ireland
| | - Igor Tolstoy
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, USA
| | - Jens Walter
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
- Department of Medicine, University College Cork, Cork, Ireland
| | - Bo Yang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Natalia Yutin
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, USA
| | - Alexandra Zhernakova
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Hub Zwart
- Erasmus School of Philosophy, Erasmus University Rotterdam, Rotterdam, The Netherlands
| | - Joël Doré
- Université Paris-Saclay, INRAE, MetaGenoPolis (MGP), Jouy-en-Josas, France
- Université Paris-Saclay, INRAE, AgroParisTech, MICALIS, Jouy-en-Josas, France
| | - R Paul Ross
- APC Microbiome Ireland, University College Cork, Cork, Ireland.
- School of Microbiology, University College Cork, Cork, Ireland.
| |
Collapse
|
2
|
Su JW, Hou XW, Liu S, Chen Y, Zhao JX, Zhang XX, Ni HB, Ma H, Liu R. Dynamic characterization of the changes in intestinal fungi and fecal metabolites during the reproductive cycle of sows. Vet Res Commun 2025; 49:116. [PMID: 39992463 DOI: 10.1007/s11259-025-10665-4] [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: 12/19/2024] [Accepted: 01/30/2025] [Indexed: 02/25/2025]
Abstract
Pregnancy-induced changes in the intestinal microbiota have been widely demonstrated. However, the research on the alterations of intestinal fungi and fecal metabolism during the reproductive cycle of sows is limited. In this study, fresh fecal samples were collected from 12 sows during the second day before fertilization (SBF), early pregnancy period (EEP; including gestational days 28, 42, and 56), late pregnancy period (LPP; including gestational days 70, 84, and 98), farrowing day (FD; collected after delivery), and lactation period (LAC, including days 7, 14, and 21 postpartum) for Internal Transcribed Space amplicon sequencing and untargeted metabolomics sequencing. The results indicated that intestinal fungi and fecal metabolites underwent significant dynamic changes during EPP and LPP, stabilizing after FD. Correlation analysis revealed a significant association between intestinal fungi and fecal metabolites, suggesting a potential role of intestinal fungi in the regulation of host health and reproductive performance. These results indicated that pregnancy may play a key role in driving the dynamic changes in intestinal fungi and fecal metabolites observed throughout the reproductive cycle. This study explores the dynamic changes and correlations of intestinal fungi and fecal metabolites in sows, providing theoretical insights for sow breeding production, feed management, and the development of microecological agents.
Collapse
Affiliation(s)
- Jin-Wen Su
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong Province, PR China
| | - Xin-Wen Hou
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong Province, PR China
| | - Shuo Liu
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong Province, PR China
| | - Yu Chen
- Animal Microecology Institute, College of Veterinary, Sichuan Agricultural University, Chengdu, Sichuan Province, PR China
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang Province, PR China
| | - Ji-Xin Zhao
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong Province, PR China
| | - Xiao-Xuan Zhang
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong Province, PR China
| | - Hong-Bo Ni
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong Province, PR China
| | - He Ma
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong Province, PR China
| | - Rui Liu
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong Province, PR China.
| |
Collapse
|
3
|
Kriti M, Ojha R, Singh S, Sarma DK, Verma V, Yadav AK, Nagpal R, Kumar M. Implication of Gut Mycobiome and Virome in Type-2 Diabetes Mellitus: Uncovering the Hidden Players. PHENOMICS (CHAM, SWITZERLAND) 2025; 5:51-64. [PMID: 40313607 PMCID: PMC12040793 DOI: 10.1007/s43657-024-00199-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 08/12/2024] [Accepted: 08/15/2024] [Indexed: 05/03/2025]
Abstract
Type-2 diabetes mellitus (T2DM) is a global epidemic with significant societal costs. The gut microbiota, including its metabolites, plays a pivotal role in maintaining health, while gut dysbiosis is implicated in several metabolic disorders, including T2DM. Although data exists on the relationship between the gut bacteriome and metabolic disorders, further attention is needed for the mycobiome and virome. Recent advancements have begun to shed light on these connections, offering potential avenues for preventive measures. However, more comprehensive investigations are required to untangle the interrelations between different microbial kingdoms and their role in T2DM development or mitigation. This review presents a simplified overview of the alterations in the gut bacteriome in T2DM and delves into the current understanding of the mycobiome and virome's role in T2DM, along with their interactions with the cohabiting bacteriome. Subsequently, it explores into the age-related dynamics of the gut microbiome and the changes observed in the microbiome composition with the onset of T2DM. Further, we explore the basic workflow utilized in gut microbiome studies. Lastly, we discuss potential therapeutic interventions in gut microbiome research, which could contribute to the amelioration of the condition, serve as preventive measures, or pave the way towards personalized medicine.
Collapse
Affiliation(s)
- Mona Kriti
- ICMR- National Institute for Research in Environmental Health, Bhopal Bypass Road, Bhauri, Bhopal, 462030 Madhya Pradesh India
| | - Raj Ojha
- ICMR- National Institute for Research in Environmental Health, Bhopal Bypass Road, Bhauri, Bhopal, 462030 Madhya Pradesh India
| | - Samradhi Singh
- ICMR- National Institute for Research in Environmental Health, Bhopal Bypass Road, Bhauri, Bhopal, 462030 Madhya Pradesh India
| | - Devojit Kumar Sarma
- ICMR- National Institute for Research in Environmental Health, Bhopal Bypass Road, Bhauri, Bhopal, 462030 Madhya Pradesh India
| | - Vinod Verma
- Stem Cell Research Centre, Department of Hematology, Sanjay Gandhi Post-Graduate Institute of Medical Sciences, Lucknow, 226014 Uttar Pradesh India
| | - Ashok Kumar Yadav
- Department of Zoology, Central University of Jammu, 181143 Jammu, India
| | - Ravinder Nagpal
- Department of Nutrition & Integrative Physiology, College of Health & Human Sciences, Florida State University, Tallahassee, FL 32306 USA
| | - Manoj Kumar
- ICMR- National Institute for Research in Environmental Health, Bhopal Bypass Road, Bhauri, Bhopal, 462030 Madhya Pradesh India
| |
Collapse
|
4
|
Ding T, Liu C, Li Z. The mycobiome in human cancer: analytical challenges, molecular mechanisms, and therapeutic implications. Mol Cancer 2025; 24:18. [PMID: 39815314 PMCID: PMC11734361 DOI: 10.1186/s12943-025-02227-8] [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: 11/13/2024] [Accepted: 01/06/2025] [Indexed: 01/18/2025] Open
Abstract
The polymorphic microbiome is considered a new hallmark of cancer. Advances in High-Throughput Sequencing have fostered rapid developments in microbiome research. The interaction between cancer cells, immune cells, and microbiota is defined as the immuno-oncology microbiome (IOM) axis. Fungal microbes (the mycobiome), although representing only ∼ 0.1-1% of the microbiome, are a critical immunologically active component of the tumor microbiome. Accumulating evidence suggests a possible involvement of commensal and pathogenic fungi in cancer initiation, progression, and treatment responsiveness. The tumor-associated mycobiome mainly consists of the gut mycobiome, the oral mycobiome, and the intratumoral mycobiome. However, the role of fungi in cancer remains poorly understood, and the diversity and complexity of analytical methods make it challenging to access this field. This review aims to elucidate the causal and complicit roles of mycobiome in cancer development and progression while highlighting the issues that need to be addressed in executing such research. We systematically summarize the advantages and limitations of current fungal detection and analysis methods. We enumerate and integrate these recent findings into our current understanding of the tumor mycobiome, accompanied by the prospect of novel and exhilarating clinical implications.
Collapse
Affiliation(s)
- Ting Ding
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, No. 20, Section 3, Renmin South Road, Chengdu, Sichuan Province, 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| | - Chang Liu
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, No. 20, Section 3, Renmin South Road, Chengdu, Sichuan Province, 610041, China
| | - Zhengyu Li
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, No. 20, Section 3, Renmin South Road, Chengdu, Sichuan Province, 610041, China.
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China.
| |
Collapse
|
5
|
Kellogg TD, Ceglia S, Mortzfeld BM, Tanna TM, Zeamer AL, Mancini MR, Foley SE, Ward DV, Bhattarai SK, McCormick BA, Reboldi A, Bucci V. Succinate-producing microbiota drives tuft cell hyperplasia to protect against Clostridioides difficile. J Exp Med 2025; 222:e20232055. [PMID: 39589553 PMCID: PMC11602550 DOI: 10.1084/jem.20232055] [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: 11/08/2023] [Revised: 09/26/2024] [Accepted: 10/23/2024] [Indexed: 11/27/2024] Open
Abstract
The role of microbes and their metabolites in modulating tuft cell (TC) dynamics in the large intestine and the relevance of this pathway to infections is unknown. Here, we uncover that microbiome-driven colonic TC hyperplasia protects against Clostridioides difficile infection. Using selective antibiotics, we demonstrate increased type 2 cytokines and TC hyperplasia in the colon but not in the ileum. We demonstrate the causal role of the microbiome in modulating this phenotype using fecal matter transplantation and administration of consortia of succinate-producing bacteria. Administration of succinate production-deficient microbes shows a reduced response in a Pou2f3-dependent manner despite similar intestinal colonization. Finally, antibiotic-treated mice prophylactically administered with succinate-producing bacteria show increased protection against C. difficile-induced morbidity and mortality. This effect is nullified in Pou2f3-/- mice, confirming that the protection occurs via the TC pathway. We propose that activation of TCs by the microbiota in the colon is a mechanism evolved by the host to counterbalance microbiome-derived cues that facilitate invasion by pathogens.
Collapse
Affiliation(s)
- Tasia D. Kellogg
- Department of Microbiology, UMass Chan Medical School, Worcester, MA, USA
- Program in Microbiome Dynamics, UMass Chan Medical School, Worcester, MA, USA
- Immunology and Microbial Pathogenesis Program, UMass Chan Medical School, Worcester, MA, USA
| | - Simona Ceglia
- Immunology and Microbial Pathogenesis Program, UMass Chan Medical School, Worcester, MA, USA
- Department of Pathology, UMass Chan Medical School, Worcester, MA, USA
| | - Benedikt M. Mortzfeld
- Department of Microbiology, UMass Chan Medical School, Worcester, MA, USA
- Program in Microbiome Dynamics, UMass Chan Medical School, Worcester, MA, USA
- Immunology and Microbial Pathogenesis Program, UMass Chan Medical School, Worcester, MA, USA
| | - Tanvi M. Tanna
- Department of Pathology, UMass Chan Medical School, Worcester, MA, USA
| | - Abigail L. Zeamer
- Department of Microbiology, UMass Chan Medical School, Worcester, MA, USA
- Program in Microbiome Dynamics, UMass Chan Medical School, Worcester, MA, USA
| | - Matthew R. Mancini
- Department of Microbiology, UMass Chan Medical School, Worcester, MA, USA
- Program in Microbiome Dynamics, UMass Chan Medical School, Worcester, MA, USA
| | - Sage E. Foley
- Department of Microbiology, UMass Chan Medical School, Worcester, MA, USA
| | - Doyle V. Ward
- Department of Microbiology, UMass Chan Medical School, Worcester, MA, USA
- Program in Microbiome Dynamics, UMass Chan Medical School, Worcester, MA, USA
| | - Shakti K. Bhattarai
- Department of Microbiology, UMass Chan Medical School, Worcester, MA, USA
- Program in Microbiome Dynamics, UMass Chan Medical School, Worcester, MA, USA
- Immunology and Microbial Pathogenesis Program, UMass Chan Medical School, Worcester, MA, USA
| | - Beth A. McCormick
- Department of Microbiology, UMass Chan Medical School, Worcester, MA, USA
- Program in Microbiome Dynamics, UMass Chan Medical School, Worcester, MA, USA
- Immunology and Microbial Pathogenesis Program, UMass Chan Medical School, Worcester, MA, USA
| | - Andrea Reboldi
- Immunology and Microbial Pathogenesis Program, UMass Chan Medical School, Worcester, MA, USA
- Department of Pathology, UMass Chan Medical School, Worcester, MA, USA
| | - Vanni Bucci
- Department of Microbiology, UMass Chan Medical School, Worcester, MA, USA
- Program in Microbiome Dynamics, UMass Chan Medical School, Worcester, MA, USA
- Immunology and Microbial Pathogenesis Program, UMass Chan Medical School, Worcester, MA, USA
| |
Collapse
|
6
|
Liu HY, Li S, Ogamune KJ, Ahmed AA, Kim IH, Zhang Y, Cai D. Fungi in the Gut Microbiota: Interactions, Homeostasis, and Host Physiology. Microorganisms 2025; 13:70. [PMID: 39858841 PMCID: PMC11767893 DOI: 10.3390/microorganisms13010070] [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: 12/10/2024] [Revised: 12/25/2024] [Accepted: 12/30/2024] [Indexed: 01/27/2025] Open
Abstract
The mammalian gastrointestinal tract is a stage for dynamic inter-kingdom interactions among bacteria, fungi, viruses, and protozoa, which collectively shape the gut micro-ecology and influence host physiology. Despite being a modest fraction, the fungal community, also referred to as mycobiota, represents a critical component of the gut microbiota. Emerging evidence suggests that fungi act as early colonizers of the intestine, exerting a lasting influence on gut development. Meanwhile, the composition of the mycobiota is influenced by multiple factors, with diet, nutrition, drug use (e.g., antimicrobials), and physical condition standing as primary drivers. During its establishment, the mycobiota forms both antagonistic and synergistic relationships with bacterial communities within the host. For instance, intestinal fungi can inhibit bacterial colonization by producing alcohol, while certain bacterial pathogens exploit fungal iron carriers to enhance their growth. However, the regulatory mechanisms governing these complex interactions remain poorly understood. In this review, we first introduce the methodologies for studying the microbiota, then address the significance of the mycobiota in the mammalian intestine, especially during weaning when all 'primary drivers' change, and, finally, discuss interactions between fungi and bacteria under various influencing factors. Our review aims to shed light on the complex inter-kingdom dynamics between fungi and bacteria in gut homeostasis and provide insights into how they can be better understood and managed to improve host health and disease outcomes.
Collapse
Affiliation(s)
- Hao-Yu Liu
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (H.-Y.L.); (S.L.); (K.J.O.)
- Jiangsu Key Laboratory of Animal Genetic Breeding and Molecular Design, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agricultural & Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Shicheng Li
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (H.-Y.L.); (S.L.); (K.J.O.)
- Jiangsu Key Laboratory of Animal Genetic Breeding and Molecular Design, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agricultural & Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Kennedy Jerry Ogamune
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (H.-Y.L.); (S.L.); (K.J.O.)
- Jiangsu Key Laboratory of Animal Genetic Breeding and Molecular Design, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agricultural & Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Abdelkareem A. Ahmed
- Department of Veterinary Science, Botswana University of Agriculture and Natural Resources, Private Bag 0027, Gaborone P.O. Box 100, Botswana;
| | - In Ho Kim
- Department of Animal Resource & Science, Dankook University, 119 Dandero, Donnamgu Cheonan, Cheonan-si 31116, Republic of Korea;
| | - Yunzeng Zhang
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China;
| | - Demin Cai
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (H.-Y.L.); (S.L.); (K.J.O.)
- Jiangsu Key Laboratory of Animal Genetic Breeding and Molecular Design, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agricultural & Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| |
Collapse
|
7
|
Mu C, Kesler M, Chen X, Shearer J, Teskey GC, Rho JM. Exogenous ketones exert antiseizure effects and modulate the gut microbiome and mycobiome in a clinically relevant murine model of epilepsy. Epilepsia 2024; 65:3676-3688. [PMID: 39412260 DOI: 10.1111/epi.18150] [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: 03/05/2024] [Revised: 10/01/2024] [Accepted: 10/01/2024] [Indexed: 12/17/2024]
Abstract
OBJECTIVE Despite growing interest in the potential use of exogenous ketones for the treatment of epilepsy, their impact on seizures and the gut microbiome and mycobiome remain unclear. METHODS Here, we examined the effects of both oral gavage and subcutaneous (SC) injection of a ketone ester (KE) in spontaneously epileptic Kcna1-null (KO) mice that model seminal aspects of human temporal lobe epilepsy. Electroencephalographic recordings and biochemical analyses were performed in KE-treated KO mice. Fecal microbial and fungal communities were profiled to determine whether the antiseizure activity of KE involves changes in the gut microbiome. RESULTS We found that exogenous KE administration by SC injection was more effective than oral gavage in terms of rendering antiseizure effects while generating similar degrees of ketonemia. However, reductions in mean daily seizure counts were accompanied by overall alterations in the fecal bacterial microbiome. Either oral or SC injection imposed a greater impact on the microbiome in male than female mice. In males, oral KE decreased Bacteroidota phylum and genera of Ligilactobacillus and Muribaculaceae, whereas SC injection decreased Bacteroides, Lactobacillus, and Lachnospiraceae. The fecal mycobiome was affected by KE injection to a greater degree than by oral gavage, and more in females than in males, as reflected by an increase in Ascomycota and Saccharomyces. Correlation analysis between microbiome and seizure counts revealed that in mice receiving KE injection, the seizure count was positively correlated with an amplicon sequencing variant of Lactobacillus (Spearman rho = .64, p = .03) and tended toward a negative correlation with Saccharomyces (Spearman rho = -.57, p = .057). SIGNIFICANCE Our findings demonstrate that exogenous ketone administration alone can induce antiseizure effects equally via different routes of administration, and that they induce differential shifts in both the bacterial microbiome and mycobiome.
Collapse
Affiliation(s)
- Chunlong Mu
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Mitchell Kesler
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Xingyu Chen
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Jane Shearer
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - G Campbell Teskey
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta, Canada
| | - Jong M Rho
- Departments of Neurosciences, Pediatrics, and Pharmacology, University of California, San Diego, Rady Children's Hospital, San Diego, California, USA
| |
Collapse
|
8
|
Cortese N, Procopio A, Merola A, Zaffino P, Cosentino C. Applications of genome-scale metabolic models to the study of human diseases: A systematic review. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2024; 256:108397. [PMID: 39232376 DOI: 10.1016/j.cmpb.2024.108397] [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: 05/09/2024] [Revised: 08/25/2024] [Accepted: 08/25/2024] [Indexed: 09/06/2024]
Abstract
BACKGROUND AND OBJECTIVES Genome-scale metabolic networks (GEMs) represent a valuable modeling and computational tool in the broad field of systems biology. Their ability to integrate constraints and high-throughput biological data enables the study of intricate metabolic aspects and processes of different cell types and conditions. The past decade has witnessed an increasing number and variety of applications of GEMs for the study of human diseases, along with a huge effort aimed at the reconstruction, integration and analysis of a high number of organisms. This paper presents a systematic review of the scientific literature, to pursue several important questions about the application of constraint-based modeling in the investigation of human diseases. Hopefully, this paper will provide a useful reference for researchers interested in the application of modeling and computational tools for the investigation of metabolic-related human diseases. METHODS This systematic review was conducted according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. Elsevier Scopus®, National Library of Medicine PubMed® and Clarivate Web of Science™ databases were enquired, resulting in 566 scientific articles. After applying exclusion and eligibility criteria, a total of 169 papers were selected and individually examined. RESULTS The reviewed papers offer a thorough and up-to-date picture of the latest modeling and computational approaches, based on genome-scale metabolic models, that can be leveraged for the investigation of a large variety of human diseases. The numerous studies have been categorized according to the clinical research area involved in the examined disease. Furthermore, the paper discusses the most typical approaches employed to derive clinically-relevant information using the computational models. CONCLUSIONS The number of scientific papers, utilizing GEM-based approaches for the investigation of human diseases, suggests an increasing interest in these types of approaches; hopefully, the present review will represent a useful reference for scientists interested in applying computational modeling approaches to investigate the aetiopathology of human diseases; we also hope that this work will foster the development of novel applications and methods for the discovery of clinically-relevant insights on metabolic-related diseases.
Collapse
Affiliation(s)
- Nicola Cortese
- Department of Experimental and Clinical Medicine, Università degli Studi Magna Græcia, Catanzaro, 88100, Italy
| | - Anna Procopio
- Department of Experimental and Clinical Medicine, Università degli Studi Magna Græcia, Catanzaro, 88100, Italy
| | - Alessio Merola
- Department of Experimental and Clinical Medicine, Università degli Studi Magna Græcia, Catanzaro, 88100, Italy
| | - Paolo Zaffino
- Department of Experimental and Clinical Medicine, Università degli Studi Magna Græcia, Catanzaro, 88100, Italy
| | - Carlo Cosentino
- Department of Experimental and Clinical Medicine, Università degli Studi Magna Græcia, Catanzaro, 88100, Italy.
| |
Collapse
|
9
|
Spychała K, Piecuch A, Szleszkowski Ł, Kadej M, Ogórek R. Microscopic fungi on the corpse - Promising tool requiring further research. Forensic Sci Int 2024; 361:112129. [PMID: 38986228 DOI: 10.1016/j.forsciint.2024.112129] [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: 07/17/2023] [Revised: 01/21/2024] [Accepted: 06/29/2024] [Indexed: 07/12/2024]
Abstract
Forensic microbiology is a relatively new area of forensic sciences. It considers the potential of microorganisms to be used in criminal investigations. As most studies involve the role of bacteria in fields like post-mortem interval estimation, personal identification or geolocation, the data on the role of fungi is comparatively scarce. Forensic mycology involves the application of fungi and their structures in forensic cases. The aim of this review is the evaluation of the current state of knowledge on fungi associated with human cadavers and their possible role in estimating the time since death. In accordance with the available reports, we focused on the relation between microscopic fungi isolated from human corpses and the cadaver condition e.g., the stage of decomposition. We also emphasised the contrast between the reported methodologies and attempted to standardise research methods in forensic mycology from sample collection to its storage, mycological analysis and identification of the obtained fungal cultures. Moreover, the potential usage of microscopic fungi in criminal cases was discussed based on various case reports.
Collapse
Affiliation(s)
- Klaudyna Spychała
- Department of Mycology and Genetics, Faculty of Biological Sciences, University of Wrocław, Przybyszewskiego 63, Wroclaw PL-51-148, Poland
| | - Agata Piecuch
- Department of Mycology and Genetics, Faculty of Biological Sciences, University of Wrocław, Przybyszewskiego 63, Wroclaw PL-51-148, Poland
| | - Łukasz Szleszkowski
- Department of Forensic Medicine, Wroclaw Medical University, Mikulicza-Radeckiego 4, Wrocław PL-50-345, Poland
| | - Marcin Kadej
- Centre for Forensic Biology and Entomology, Department of Invertebrate Biology, Evolution and Conservation, Faculty of Biological Sciences, University of Wrocław, Przybyszewskiego 65, Wrocław PL-51-148, Poland.
| | - Rafał Ogórek
- Department of Mycology and Genetics, Faculty of Biological Sciences, University of Wrocław, Przybyszewskiego 63, Wroclaw PL-51-148, Poland
| |
Collapse
|
10
|
Niu X, Dang Z, Hong M, Shi H, Ding L. Effects of Freshwater Acidification on the Gut Microbial Community of Trachemys scripta elegans. Animals (Basel) 2024; 14:1898. [PMID: 38998010 PMCID: PMC11240511 DOI: 10.3390/ani14131898] [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: 05/29/2024] [Revised: 06/21/2024] [Accepted: 06/24/2024] [Indexed: 07/14/2024] Open
Abstract
Freshwater acidification (FA) has become a global environmental problem, posing a potential threat to freshwater ecosystems. The gut microbiota plays a crucial role in the host's response and adaptation to new environments. In this study, we investigated the changes in microbial communities in Red-eared slider (Trachemys scripta elegans) under acidic conditions to reveal the ecological impacts of acidification on freshwater turtles. The results showed that there were significant differences in β-diversity (p = 0.03), while there were no significant differences in the α-diversity of gut microbiota in T. s. elegans between the different levels of acidification (pH of 5.5, 6.5, 7.5). Both the Gut Microbiome Health Index (GMHI) and the Microbial Dysbiosis Index (MDI) exhibited significant differences when comparing environments with a pH of 5.5 to those with a pH of 6.5 (p < 0.01). A comparative analysis between pH levels of 5.5 and 6.5 also revealed substantial differences (p < 0.01). Likewise, a comparative analysis between pH levels of 6.5 and 7.5 also revealed substantial differences (p < 0.01). At the phylum level, Firmicutes, Fusobacteria, and Bacteroidota formed a major part of the gut microbial community, Fusobacteria showed significant differences in different acidity environments (p = 0.03). At the genus level, Cetobacterium, Turicibacter, unclassified Eubacteriaceae, and Anaerorhabdus_furcosa_group showed significant differences in different acidity environments. The pH reduced interactivity in the gut microbiota of T. s. elegans. In addition, LEfSe analysis and functional prediction revealed that the potentially_pathogenic and stress_tolerant functional characteristics also showed significant differences in different acidity environments. The findings underscore the pivotal role of the gut microbiota in T. s. elegans in response to freshwater acidification and provide a foundation for further exploration into the impacts of acidification on freshwater ecosystems.
Collapse
Affiliation(s)
| | | | - Meiling Hong
- Ministry of Education Key Laboratory for Ecology of Tropical Islands, Key Laboratory of Tropical Animal and Plant Ecology of Hainan Province, College of Life Sciences, Hainan Normal University, Haikou 571158, China; (X.N.); (Z.D.); (H.S.)
| | | | - Li Ding
- Ministry of Education Key Laboratory for Ecology of Tropical Islands, Key Laboratory of Tropical Animal and Plant Ecology of Hainan Province, College of Life Sciences, Hainan Normal University, Haikou 571158, China; (X.N.); (Z.D.); (H.S.)
| |
Collapse
|
11
|
Iyer P, Ojcius DM. Unveiling the mycobiota: The fungal frontier of human health. Biomed J 2024; 47:100751. [PMID: 38838983 PMCID: PMC11220527 DOI: 10.1016/j.bj.2024.100751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 05/24/2024] [Accepted: 06/03/2024] [Indexed: 06/07/2024] Open
Abstract
The microbiota and its effect on health has been extensively studied over the past decade. In many studies, the term microbiota has become synonymous with the bacterial component of the microbiota. Other microbes in the microbiota, such as viruses and fungi, have been neglected until recently. This special issue provides some background on the mycobiota and explores the role of gut fungi in human diseases such as cancer, metabolic diseases, and infection by Clostridiodes difficile, and describes the incidence of fungal infections in transplant patients. The mycobiota, once overlooked, now garners increasing attention.
Collapse
Affiliation(s)
- Parvati Iyer
- Department of Diagnostic Sciences, University of the Pacific, Arthur Dugoni School of Dentistry, San Francisco, CA, USA
| | - David M Ojcius
- Department of Biomedical Sciences, University of the Pacific, Arthur Dugoni School of Dentistry, San Francisco, CA, USA.
| |
Collapse
|
12
|
Chuandong Z, Hu J, Li J, Wu Y, Wu C, Lai G, Shen H, Wu F, Tao C, Liu S, Zhang W, Shao H. Distribution and roles of Ligilactobacillus murinus in hosts. Microbiol Res 2024; 282:127648. [PMID: 38367479 DOI: 10.1016/j.micres.2024.127648] [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: 08/30/2023] [Revised: 10/26/2023] [Accepted: 02/10/2024] [Indexed: 02/19/2024]
Abstract
Ligilactobacillus murinus, a member of the Ligilactobacillus genus, holds significant potential as a probiotic. While research on Ligilactobacillus murinus has been relatively limited compared to well-studied probiotic lactic acid bacteria such as Limosilactobacillus reuteri and Lactobacillus gasseri, a mounting body of evidence highlights its extensive involvement in host intestinal metabolism and immune activities. Moreover, its abundance exhibits a close correlation with intestinal health. Notably, beyond the intestinal context, Ligilactobacillus murinus is gaining recognition for its contributions to metabolism and regulation in the oral cavity, lungs, and vagina. As such, Ligilactobacillus murinus emerges as a potential probiotic candidate with a pivotal role in supporting host well-being. This review delves into studies elucidating the multifaceted roles of Ligilactobacillus murinus. It also examines its medicinal potential and associated challenges, underscoring the imperative to delve deeper into unraveling the mechanisms of its actions and exploring its health applications.
Collapse
Affiliation(s)
- Zhou Chuandong
- School of Life Science and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, Guangdong, PR China
| | - Jicong Hu
- School of Life Science and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, Guangdong, PR China
| | - Jiawen Li
- School of Life Science and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, Guangdong, PR China
| | - Yuting Wu
- School of Clinical Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong, PR China
| | - Chan Wu
- School of Clinical Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong, PR China
| | - Guanxi Lai
- School of Clinical Pharmacy, Guangdong Pharmaceutical University, Guangzhou, Guangdong, PR China
| | - Han Shen
- School of Life Science and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, Guangdong, PR China
| | - Fenglin Wu
- School of Life Science and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, Guangdong, PR China
| | - Changli Tao
- School of Life Science and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, Guangdong, PR China
| | - Song Liu
- School of Clinical Pharmacy, Guangdong Pharmaceutical University, Guangzhou, Guangdong, PR China
| | - Wenfeng Zhang
- School of Life Science and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, Guangdong, PR China.
| | - Hongwei Shao
- School of Life Science and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, Guangdong, PR China.
| |
Collapse
|
13
|
Nenciarini S, Renzi S, di Paola M, Meriggi N, Cavalieri D. Ascomycetes yeasts: The hidden part of human microbiome. WIREs Mech Dis 2024; 16:e1641. [PMID: 38228159 DOI: 10.1002/wsbm.1641] [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: 05/17/2023] [Revised: 12/17/2023] [Accepted: 12/19/2023] [Indexed: 01/18/2024]
Abstract
The fungal component of the microbiota, the mycobiota, has been neglected for a long time due to its poor richness compared to bacteria. Limitations in fungal detection and taxonomic identification arise from using metagenomic approaches, often borrowed from bacteriome analyses. However, the relatively recent discoveries of the ability of fungi to modulate the host immune response and their involvement in human diseases have made mycobiota a fundamental component of the microbial communities inhabiting the human host, deserving some consideration in host-microbe interaction studies and in metagenomics. Here, we reviewed recent data on the identification of yeasts of the Ascomycota phylum across human body districts, focusing on the most representative genera, that is, Saccharomyces and Candida. Then, we explored the key factors involved in shaping the human mycobiota across the lifespan, ranging from host genetics to environment, diet, and lifestyle habits. Finally, we discussed the strengths and weaknesses of culture-dependent and independent methods for mycobiota characterization. Overall, there is still room for some improvements, especially regarding fungal-specific methodological approaches and bioinformatics challenges, which are still critical steps in mycobiota analysis, and to advance our knowledge on the role of the gut mycobiota in human health and disease. This article is categorized under: Immune System Diseases > Genetics/Genomics/Epigenetics Immune System Diseases > Environmental Factors Infectious Diseases > Environmental Factors.
Collapse
Affiliation(s)
| | - Sonia Renzi
- Department of Biology, University of Florence, Florence, Italy
| | - Monica di Paola
- Department of Biology, University of Florence, Florence, Italy
| | - Niccolò Meriggi
- Department of Biology, University of Florence, Florence, Italy
| | | |
Collapse
|
14
|
Mok K, Poolsawat T, Somnuk S, Wanikorn B, Patumcharoenpol P, Nitisinprasert S, Vongsangnak W, Nakphaichit M. Preliminary characterization of gut mycobiome enterotypes reveals the correlation trends between host metabolic parameter and diet: a case study in the Thai Cohort. Sci Rep 2024; 14:5805. [PMID: 38461361 PMCID: PMC10924899 DOI: 10.1038/s41598-024-56585-2] [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: 10/08/2023] [Accepted: 03/08/2024] [Indexed: 03/11/2024] Open
Abstract
The association between the gut mycobiome and its potential influence on host metabolism in the Thai Cohort was assessed. Two distinct predominant enterotypes, Saccharomyces (Sa) and Aspergillus/Penicillium (Ap/Pe) showed differences in gut mycobiota diversity and composition. Notably, the Sa enterotype exhibited lower evenness and richness, likely due to the prevalence of Saccharomyces, while both enterotypes displayed unique metabolic behaviors related to nutrient metabolism and body composition. Fiber consumption was positively correlated with adverse body composition and fasting glucose levels in individuals with the Sa enterotype, whereas in the Ap/Pe enterotype it was positively correlated with fat and protein intake. The metabolic functional analysis revealed the Sa enterotype associated with carbohydrate metabolism, while the Ap/Pe enterotype involved in lipid metabolism. Very interestingly, the genes involved in the pentose and glucuronate interconversion pathway, such as polygalacturonase and L-arabinose-isomerase, were enriched in the Sa enterotype signifying a metabolic capacity for complex carbohydrate degradation and utilization of less common sugars as energy sources. These findings highlight the interplay between gut mycobiome composition, dietary habits, and metabolic outcomes within the Thai cohort studies.
Collapse
Affiliation(s)
- Kevin Mok
- Department of Biotechnology, Faculty of Agro-Industry, Kasetsart University, Bangkok, 10900, Thailand
- Specialized Research Unit: Probiotics and Prebiotics for Health, Faculty of Agro-Industry, Kasetsart University, Bangkok, 10900, Thailand
| | - Thitirat Poolsawat
- Department of Biotechnology, Faculty of Agro-Industry, Kasetsart University, Bangkok, 10900, Thailand
- Specialized Research Unit: Functional Food and Human Health Laboratory, Faculty of Agro-Industry, Kasetsart University, Bangkok, 10900, Thailand
| | - Surasawadee Somnuk
- Department of Sports and Health Sciences, Faculty of Sport Science, Kasetsart University, Kamphaeng Saen Campus, Nakhon Pathom, 73140, Thailand
| | - Bandhita Wanikorn
- Department of Biotechnology, Faculty of Agro-Industry, Kasetsart University, Bangkok, 10900, Thailand
- Specialized Research Unit: Functional Food and Human Health Laboratory, Faculty of Agro-Industry, Kasetsart University, Bangkok, 10900, Thailand
| | - Preecha Patumcharoenpol
- Department of Zoology, Faculty of Science, Kasetsart University, Bangkok, 10900, Thailand
- Omics Center for Agriculture, Bioresources, Food, and Health, Kasetsart University (OmiKU), Bangkok, 10900, Thailand
| | - Sunee Nitisinprasert
- Department of Biotechnology, Faculty of Agro-Industry, Kasetsart University, Bangkok, 10900, Thailand
- Specialized Research Unit: Probiotics and Prebiotics for Health, Faculty of Agro-Industry, Kasetsart University, Bangkok, 10900, Thailand
| | - Wanwipa Vongsangnak
- Department of Zoology, Faculty of Science, Kasetsart University, Bangkok, 10900, Thailand
- Omics Center for Agriculture, Bioresources, Food, and Health, Kasetsart University (OmiKU), Bangkok, 10900, Thailand
| | - Massalin Nakphaichit
- Department of Biotechnology, Faculty of Agro-Industry, Kasetsart University, Bangkok, 10900, Thailand.
- Specialized Research Unit: Probiotics and Prebiotics for Health, Faculty of Agro-Industry, Kasetsart University, Bangkok, 10900, Thailand.
| |
Collapse
|
15
|
Kellogg TD, Ceglia S, Mortzfeld BM, Zeamer AL, Foley SE, Ward DV, Bhattarai SK, McCormick BA, Reboldi A, Bucci V. Microbiota encoded fatty-acid metabolism expands tuft cells to protect tissues homeostasis during Clostridioides difficile infection in the large intestine. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.29.574039. [PMID: 38352546 PMCID: PMC10862725 DOI: 10.1101/2024.01.29.574039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
Metabolic byproducts of the intestinal microbiota are crucial in maintaining host immune tone and shaping inter-species ecological dynamics. Among these metabolites, succinate is a driver of tuft cell (TC) differentiation and consequent type 2 immunity-dependent protection against invading parasites in the small intestine. Succinate is also a growth enhancer of the nosocomial pathogen Clostridioides difficile in the large intestine. To date, no research has shown the role of succinate in modulating TC dynamics in the large intestine, or the relevance of this immune pathway to C. difficile pathophysiology. Here we reveal the existence of a three-way circuit between commensal microbes, C. difficile and host epithelial cells which centers around succinate. Through selective microbiota depletion experiments we demonstrate higher levels of type 2 cytokines leading to expansion of TCs in the colon. We then demonstrate the causal role of the microbiome in modulating colonic TC abundance and subsequent type 2 cytokine induction using rational supplementation experiments with fecal transplants and microbial consortia of succinate-producing bacteria. We show that administration of a succinate-deficient Bacteroides thetaiotaomicron knockout (Δfrd) significantly reduces the enhanced type 2 immunity in mono-colonized mice. Finally, we demonstrate that mice prophylactically administered with the consortium of succinate-producing bacteria show reduced C. difficile-induced morbidity and mortality compared to mice administered with heat-killed bacteria or the vehicle. This effect is reduced in a partial tuft cell knockout mouse, Pou2f3+/-, and nullified in the tuft cell knockout mouse, Pou2f3-/-, confirming that the observed protection occurs via the TC pathway. Succinate is an intermediary metabolite of the production of short-chain fatty acids, and its concentration often increases during dysbiosis. The first barrier to enteric pathogens alike is the intestinal epithelial barrier, and host maintenance and strengthening of barrier integrity is vital to homeostasis. Considering our data, we propose that activation of TC by the microbiota-produced succinate in the colon is a mechanism evolved by the host to counterbalance microbiome-derived cues that facilitate invasion by intestinal pathogens.
Collapse
Affiliation(s)
- Tasia D. Kellogg
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, MA, USA
- Program in Microbiome Dynamics, UMass Chan Medical School, Worcester, MA, USA
- Immunology and Microbial Pathogenesis Program, UMass Chan Medical School, Worcester, MA, USA
| | - Simona Ceglia
- Immunology and Microbial Pathogenesis Program, UMass Chan Medical School, Worcester, MA, USA
- Department of Pathology, UMass Chan Medical School, Worcester, MA, USA
| | - Benedikt M. Mortzfeld
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, MA, USA
- Program in Microbiome Dynamics, UMass Chan Medical School, Worcester, MA, USA
- Immunology and Microbial Pathogenesis Program, UMass Chan Medical School, Worcester, MA, USA
| | - Abigail L. Zeamer
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, MA, USA
- Program in Microbiome Dynamics, UMass Chan Medical School, Worcester, MA, USA
| | - Sage E. Foley
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, MA, USA
- Current address: Transformational and Translational Immunology Discovery Department, AbbVie, Cambridge, MA, USA
| | - Doyle V. Ward
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, MA, USA
- Program in Microbiome Dynamics, UMass Chan Medical School, Worcester, MA, USA
| | - Shakti K. Bhattarai
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, MA, USA
- Program in Microbiome Dynamics, UMass Chan Medical School, Worcester, MA, USA
- Immunology and Microbial Pathogenesis Program, UMass Chan Medical School, Worcester, MA, USA
| | - Beth A. McCormick
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, MA, USA
- Program in Microbiome Dynamics, UMass Chan Medical School, Worcester, MA, USA
- Immunology and Microbial Pathogenesis Program, UMass Chan Medical School, Worcester, MA, USA
| | - Andrea Reboldi
- Immunology and Microbial Pathogenesis Program, UMass Chan Medical School, Worcester, MA, USA
- Department of Pathology, UMass Chan Medical School, Worcester, MA, USA
| | - Vanni Bucci
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, MA, USA
- Program in Microbiome Dynamics, UMass Chan Medical School, Worcester, MA, USA
- Immunology and Microbial Pathogenesis Program, UMass Chan Medical School, Worcester, MA, USA
| |
Collapse
|
16
|
Wang Y, Chen J, Ni Y, Liu Y, Gao X, Tse MA, Panagiotou G, Xu A. Exercise-changed gut mycobiome as a potential contributor to metabolic benefits in diabetes prevention: an integrative multi-omics study. Gut Microbes 2024; 16:2416928. [PMID: 39473051 PMCID: PMC11533799 DOI: 10.1080/19490976.2024.2416928] [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: 07/22/2024] [Revised: 10/05/2024] [Accepted: 10/10/2024] [Indexed: 11/06/2024] Open
Abstract
BACKGROUND The importance of gut microbes in mediating the benefits of lifestyle intervention is increasingly recognized. However, compared to the bacterial microbiome, the role of intestinal fungi in exercise remains elusive. With our established randomized controlled trial of exercise intervention in Chinese males with prediabetes (n = 39, ClinicalTrials.gov:NCT03240978), we investigated the dynamics of human gut mycobiome and further interrogated their associations with exercise-elicited outcomes using multi-omics approaches. METHODS Clinical variations and biological samples were collected before and after training. Fecal fungal composition was analyzed using the internal transcribed spacer 2 (ITS2) sequencing and integrated with paired shotgun metagenomics, untargeted metabolomics, and Olink proteomics. RESULTS Twelve weeks of exercise training profoundly promoted fungal ecological diversity and intrakingdom connection. We further identified exercise-responsive genera with potential metabolic benefits, including Verticillium, Sarocladium, and Ceratocystis. Using multi-omics approaches, we elucidated comprehensive associations between changes in gut mycobiome and exercise-shaped metabolic phenotypes, bacterial microbiome, and circulating metabolomics and proteomics profiles. Furthermore, a machine-learning algorithm built using baseline microbial signatures and clinical characteristics predicted exercise responsiveness in improvements of insulin sensitivity, with an area under the receiver operating characteristic (AUROC) of 0.91 (95% CI: 0.85-0.97) in the discovery cohort and of 0.79 (95% CI: 0.74-0.86) in the independent validation cohort (n = 30). CONCLUSIONS Our findings suggest that intense exercise training significantly remodels the human fungal microbiome composition. Changes in gut fungal composition are associated with the metabolic benefits of exercise, indicating gut mycobiome is a possible molecular transducer of exercise. Moreover, baseline gut fungal signatures predict exercise responsiveness for diabetes prevention, highlighting that targeting the gut mycobiome emerges as a prospective strategy in tailoring personalized training for diabetes prevention.
Collapse
Affiliation(s)
- Yao Wang
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
- Department of Medicine, The University of Hong Kong, Hong Kong, China
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong, China
| | - Jiarui Chen
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
- Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yueqiong Ni
- Department of Microbiome Dynamics, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knoll Institute, Jena, Germany
| | - Yan Liu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
- Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Xiang Gao
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
| | - Michael Andrew Tse
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
- Centre for Sports and Exercise, The University of Hong Kong, Hong Kong, China
| | - Gianni Panagiotou
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
- Department of Medicine, The University of Hong Kong, Hong Kong, China
- Department of Microbiome Dynamics, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knoll Institute, Jena, Germany
- Faculty of Biological Sciences, Friedrich Schiller University, Jena, Germany
- Jena University Hospital, Friedrich Schiller University, Jena, Germany
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
- Department of Medicine, The University of Hong Kong, Hong Kong, China
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
17
|
Ho SM, Tsai WH, Lai CH, Chiang MH, Lee WP, Wu HY, Bai PY, Wu T, Wu CL. Probiotic Lactobacillus spp. improves Drosophila memory by increasing lactate dehydrogenase levels in the brain mushroom body neurons. Gut Microbes 2024; 16:2316533. [PMID: 38372783 PMCID: PMC10877976 DOI: 10.1080/19490976.2024.2316533] [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: 09/21/2023] [Accepted: 02/05/2024] [Indexed: 02/20/2024] Open
Abstract
Probiotics are live microorganisms that offer potential benefits to their hosts and can occasionally influence behavioral responses. However, the detailed mechanisms by which probiotics affect the behavior of their hosts and the underlying biogenic effects remain unclear. Lactic acid bacteria, specifically Lactobacillus spp. are known probiotics. Drosophila melanogaster, commonly known as the fruit fly, is a well-established model organism for investigating the interaction between the host and gut microbiota in translational research. Herein, we showed that 5-day administration of Lactobacillus acidophilus (termed GMNL-185) or Lacticaseibacillus rhamnosus (termed GMNL-680) enhances olfactory-associative memory in Drosophila. Moreover, a combined diet of GMNL-185 and GMNL-680 demonstrated synergistic effects on memory functions. Live brain imaging revealed a significant increase in calcium responses to the training odor in the mushroom body β and γ lobes of flies that underwent mixed feeding with GMNL-185 and GMNL-680. Quantitative reverse transcription polymerase chain reaction (qRT-PCR) and whole-mount brain immunohistochemistry revealed significant upregulation of lactate dehydrogenase (LDH) expression in the fly brain following the mixed feeding. Notably, the genetic knockdown of Ldh in neurons, specifically in mushroom body, ameliorated the beneficial effects of mixed feeding with GMNL-185 and GMNL-680 on memory improvement. Altogether, our results demonstrate that supplementation with L. acidophilus and L. rhamnosus enhances memory functions in flies by increasing brain LDH levels.
Collapse
Affiliation(s)
- Shuk-Man Ho
- Department of Biochemistry, Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, Department of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Wan-Hua Tsai
- Research and Development Department, GenMont Biotech Incorporation, Tainan, Taiwan
| | - Chih-Ho Lai
- Department of Biochemistry, Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, Department of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Molecular Infectious Disease Research Center, Department of Pediatrics, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Department of Medical Research, School of Medicine, China Medical University and Hospital, Taichung, Taiwan
- Department of Nursing, Asia University, Taichung, Taiwan
- Research Center for Emerging Viral Infections, Chang Gung University, Taoyuan, Taiwan
| | - Meng-Hsuan Chiang
- Department of Biochemistry, Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, Department of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Wang-Po Lee
- Department of Biochemistry, Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, Department of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Hui-Yu Wu
- Department of Biochemistry, Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, Department of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Pei-Yi Bai
- Department of Biochemistry, Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, Department of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Tony Wu
- Department of Neurology, New Taipei Municipal TuCheng Hospital, Chang Gung Memorial Hospital, New Taipei City, Taiwan
| | - Chia-Lin Wu
- Department of Biochemistry, Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, Department of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Neurology, New Taipei Municipal TuCheng Hospital, Chang Gung Memorial Hospital, New Taipei City, Taiwan
- Brain Research Center, National Tsing Hua University, Hsinchu, Taiwan
| |
Collapse
|
18
|
Kumbhare SV, Pedroso I, Ugalde JA, Márquez-Miranda V, Sinha R, Almonacid DE. Drug and gut microbe relationships: Moving beyond antibiotics. Drug Discov Today 2023; 28:103797. [PMID: 37806386 DOI: 10.1016/j.drudis.2023.103797] [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: 05/17/2023] [Revised: 09/23/2023] [Accepted: 10/04/2023] [Indexed: 10/10/2023]
Abstract
Our understanding of drug-microbe relationships has evolved from viewing microbes as mere drug producers to a dynamic, modifiable system where they can serve as drugs or targets of precision pharmacology. This review highlights recent findings on the gut microbiome, particularly focusing on four aspects of research: (i) drugs for bugs, covering recent strategies for targeting gut pathogens; (ii) bugs as drugs, including probiotics; (iii) drugs from bugs, including postbiotics; and (iv) bugs and drugs, discussing additional types of drug-microbe interactions. This review provides a perspective on future translational research, including efficient companion diagnostics in pharmaceutical interventions.
Collapse
Affiliation(s)
| | | | - Juan A Ugalde
- Center for Bioinformatics and Integrative Biology, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
| | - Valeria Márquez-Miranda
- Center for Bioinformatics and Integrative Biology, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
| | | | | |
Collapse
|
19
|
Liu Q, Zhang W, Pei Y, Tao H, Ma J, Li R, Zhang F, Wang L, Shen L, Liu Y, Jia X, Hu Y. Gut mycobiome as a potential non-invasive tool in early detection of lung adenocarcinoma: a cross-sectional study. BMC Med 2023; 21:409. [PMID: 37904139 PMCID: PMC10617124 DOI: 10.1186/s12916-023-03095-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 09/26/2023] [Indexed: 11/01/2023] Open
Abstract
BACKGROUND The gut mycobiome of patients with lung adenocarcinoma (LUAD) remains unexplored. This study aimed to characterize the gut mycobiome in patients with LUAD and evaluate the potential of gut fungi as non-invasive biomarkers for early diagnosis. METHODS In total, 299 fecal samples from Beijing, Suzhou, and Hainan were collected prospectively. Using internal transcribed spacer 2 sequencing, we profiled the gut mycobiome. Five supervised machine learning algorithms were trained on fungal signatures to build an optimized prediction model for LUAD in a discovery cohort comprising 105 patients with LUAD and 61 healthy controls (HCs) from Beijing. Validation cohorts from Beijing, Suzhou, and Hainan comprising 44, 17, and 15 patients with LUAD and 26, 19, and 12 HCs, respectively, were used to evaluate efficacy. RESULTS Fungal biodiversity and richness increased in patients with LUAD. At the phylum level, the abundance of Ascomycota decreased, while that of Basidiomycota increased in patients with LUAD. Candida and Saccharomyces were the dominant genera, with a reduction in Candida and an increase in Saccharomyces, Aspergillus, and Apiotrichum in patients with LUAD. Nineteen operational taxonomic unit markers were selected, and excellent performance in predicting LUAD was achieved (area under the curve (AUC) = 0.9350) using a random forest model with outcomes superior to those of four other algorithms. The AUCs of the Beijing, Suzhou, and Hainan validation cohorts were 0.9538, 0.9628, and 0.8833, respectively. CONCLUSIONS For the first time, the gut fungal profiles of patients with LUAD were shown to represent potential non-invasive biomarkers for early-stage diagnosis.
Collapse
Affiliation(s)
- Qingyan Liu
- Graduate School, Chinese People's Liberation Army Medical School, Beijing, China
- Department of Oncology, Fifth Medical Center of the Chinese People's Liberation Army General Hospital, 28 Fuxing Road, Haidian Distrist, Beijing, 100000, China
| | - Weidong Zhang
- Graduate School, Chinese People's Liberation Army Medical School, Beijing, China
- Department of Thoracic Surgery, First Medical Center of the Chinese People's Liberation Army General Hospital, 28 Fuxing Road, Haidian District, Beijing, 100000, China
| | - Yanbin Pei
- Graduate School, Chinese People's Liberation Army Medical School, Beijing, China
| | - Haitao Tao
- Department of Oncology, Fifth Medical Center of the Chinese People's Liberation Army General Hospital, 28 Fuxing Road, Haidian Distrist, Beijing, 100000, China
| | - Junxun Ma
- Department of Oncology, Fifth Medical Center of the Chinese People's Liberation Army General Hospital, 28 Fuxing Road, Haidian Distrist, Beijing, 100000, China
| | - Rong Li
- Department of Health Medicine, Second Medical Center of the Chinese People's Liberation Army General Hospital, Beijing, China
| | - Fan Zhang
- Department of Oncology, Fifth Medical Center of the Chinese People's Liberation Army General Hospital, 28 Fuxing Road, Haidian Distrist, Beijing, 100000, China
| | - Lijie Wang
- Department of Oncology, Fifth Medical Center of the Chinese People's Liberation Army General Hospital, 28 Fuxing Road, Haidian Distrist, Beijing, 100000, China
| | - Leilei Shen
- Department of Thoracic Surgery, Hainan Medical Center of the Chinese People's Liberation Army General Hospital, Hainan, China
| | - Yang Liu
- Department of Thoracic Surgery, First Medical Center of the Chinese People's Liberation Army General Hospital, 28 Fuxing Road, Haidian District, Beijing, 100000, China.
| | - Xiaodong Jia
- Department of Oncology, Fifth Medical Center of the Chinese People's Liberation Army General Hospital, 28 Fuxing Road, Haidian Distrist, Beijing, 100000, China.
| | - Yi Hu
- Department of Oncology, Fifth Medical Center of the Chinese People's Liberation Army General Hospital, 28 Fuxing Road, Haidian Distrist, Beijing, 100000, China.
| |
Collapse
|
20
|
Li W, Shu Y, Zhang J, Wu M, Zhu GH, Huang WY, Shen L, Kang Y. Long-term prednisone treatment causes fungal microbiota dysbiosis and alters the ecological interaction between gut mycobiome and bacteriome in rats. Front Microbiol 2023; 14:1112767. [PMID: 37342562 PMCID: PMC10277626 DOI: 10.3389/fmicb.2023.1112767] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 05/16/2023] [Indexed: 06/23/2023] Open
Abstract
Glucocorticoids (GCs) are widely used in the treatment of immune-mediated diseases due to their anti-inflammatory and immunosuppressive effects. Prednisone is one of the most commonly used GCs. However, it is still unknown whether prednisone affects gut fungi in rats. Herein we investigated whether prednisone changed the composition of gut fungi and the interactions between gut mycobiome and bacteriome/fecal metabolome in rats. Twelve male Sprague-Dawley rats were randomly assigned to a control group and a prednisone group which received prednisone daily by gavage for 6 weeks. ITS2 rRNA gene sequencing of fecal samples was performed to identify differentially abundant gut fungi. The associations between gut mycobiome and bacterial genera/fecal metabolites obtained from our previously published study were explored by using Spearman correlation analysis. Our data showed that there were no changes in the richness of gut mycobiome in rats after prednisone treatment, but the diversity increased significantly. The relative abundance of genera Triangularia and Ciliophora decreased significantly. At the species level, the relative abundance of Aspergillus glabripes increased significantly, while Triangularia mangenotii and Ciliophora sp. decreased. In addition, prednisone altered the gut fungi-bacteria interkingdom interactions in rats after prednisone treatment. Additionally, the genus Triangularia was negatively correlated with m-aminobenzoic acid, but positively correlated with hydrocinnamic acid and valeric acid. Ciliophora was negatively correlated with phenylalanine and homovanillic acid, but positively correlated with 2-Phenylpropionate, hydrocinnamic acid, propionic acid, valeric acid, isobutyric acid, and isovaleric acid. In conclusion, long-term prednisone treatment caused fungal microbiota dysbiosis and might alter the ecological interaction between gut mycobiome and bacteriome in rats.
Collapse
Affiliation(s)
- Wenyan Li
- Department of Nephrology and Rheumatology, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yun Shu
- Department of Nephrology and Rheumatology, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jing Zhang
- Department of Nephrology and Rheumatology, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Mengmeng Wu
- Department of Nephrology and Rheumatology, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Guang-hua Zhu
- Department of Nephrology and Rheumatology, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wen-yan Huang
- Department of Nephrology and Rheumatology, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Li Shen
- Department of Cardiothoracic Surgery, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yulin Kang
- Department of Nephrology and Rheumatology, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
21
|
Wang L, Zhang K, Zeng Y, Luo Y, Peng J, Zhang J, Kuang T, Fan G. Gut mycobiome and metabolic diseases: The known, the unknown, and the future. Pharmacol Res 2023; 193:106807. [PMID: 37244385 DOI: 10.1016/j.phrs.2023.106807] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/23/2023] [Accepted: 05/24/2023] [Indexed: 05/29/2023]
Abstract
Metabolic diseases, such as type 2 diabetes mellitus (T2DM), non-alcoholic fatty liver disease (NAFLD) and obesity, have become a major public health problem worldwide. In recent years, most research on the role of gut microbes in metabolic diseases has focused on bacteria, whereas fungal microbes have been neglected. This review aims to provide a comprehensive overview of gut fungal alterations in T2DM, obesity, and NAFLD, and to discuss the mechanisms associated with disease development. In addition, several novel strategies targeting gut mycobiome and/or their metabolites to improve T2DM, obesity and NAFLD, including fungal probiotics, antifungal drugs, dietary intervention, and fecal microbiota transplantation, are critically discussed. The accumulated evidence suggests that gut mycobiome plays an important role in the occurrence and development of metabolic diseases. The possible mechanisms by which the gut mycobiome affects metabolic diseases include fungal-induced immune responses, fungal-bacterial interactions, and fungal-derived metabolites. Candida albicans, Aspergillus and Meyerozyma may be potential pathogens of metabolic diseases because they can activate the immune system and/or produce harmful metabolites. Moreover, Saccharomyces boulardii, S. cerevisiae, Alternaria, and Cochliobolus fungi may have the potential to improve metabolic diseases. The information may provide an important reference for the development of new therapeutics for metabolic diseases based on gut mycobiome.
Collapse
Affiliation(s)
- Lijie Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy and School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Kun Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy and School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yujiao Zeng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy and School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yuting Luo
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy and School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jiayan Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy and School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jing Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy and School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Tingting Kuang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy and School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Gang Fan
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy and School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; Meishan Hospital of Chengdu University of Traditional Chinese Medicine, Meishan, 620010, China.
| |
Collapse
|
22
|
Hartmann P, Schnabl B. Fungal infections and the fungal microbiome in hepatobiliary disorders. J Hepatol 2023; 78:836-851. [PMID: 36565724 PMCID: PMC10033447 DOI: 10.1016/j.jhep.2022.12.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 12/05/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022]
Abstract
Liver and biliary diseases affect more than a billion people worldwide, with high associated morbidity and mortality. The impact of the intestinal bacterial microbiome on liver diseases has been well established. However, the fungal microbiome, or mycobiome, has been overlooked for a long time. Recently, several studies have shed light on the role of the mycobiome in the development and progression of hepatobiliary diseases. In particular, the fungal genus Candida has been found to be involved in the pathogenesis of multiple hepatobiliary conditions. Herein, we compare colonisation and infection, describe mycobiome findings in the healthy state and across the various hepatobiliary conditions, and point toward communalities. We detail how quantitation of immune responses to fungal antigens can be employed to predict disease severity, e.g. using antibodies to Saccharomyces cerevisiae or specific anti-Candida albicans antibodies. We also show how fungal products (e.g. beta-glucans, candidalysin) activate the host's immune system to exacerbate liver and biliary diseases. Finally, we describe how the gut mycobiome can be modulated to ameliorate hepatobiliary conditions.
Collapse
Affiliation(s)
- Phillipp Hartmann
- Department of Medicine, University of California San Diego, La Jolla, CA, USA; Department of Pediatrics, University of California San Diego, La Jolla, CA, USA; Division of Gastroenterology, Hepatology & Nutrition, Rady Children's Hospital San Diego, San Diego, CA, USA
| | - Bernd Schnabl
- Department of Medicine, University of California San Diego, La Jolla, CA, USA; Department of Medicine, VA San Diego Healthcare System, San Diego, CA, USA.
| |
Collapse
|
23
|
The Role of the Mycobiome in Women’s Health. J Fungi (Basel) 2023; 9:jof9030348. [PMID: 36983516 PMCID: PMC10051763 DOI: 10.3390/jof9030348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 02/19/2023] [Accepted: 03/09/2023] [Indexed: 03/14/2023] Open
Abstract
Although the human bacteriome and virome have gained a great deal of attention over the years, the human mycobiome has been far more neglected despite having significant value and implications in human health. In women, mycobiome profiles in breastmilk, vaginal regions, the gut, skin, and the oral cavity can provide insight into women’s health, diseases, and microbiome dysbiosis. Analyses of mycobiome composition under factors, such as health, age, diet, weight, and drug exposure (including antibiotic therapies), help to elucidate the various roles of women’s mycobiome in homeostasis, microbiome interactions (synergistic and antagonistic), and health. This review summarizes the most recent updates to mycobiome knowledge in these critical areas.
Collapse
|