1
|
Jing Z, Yinhang W, Jian C, Zhanbo Q, Xinyue W, Shuwen H. Interaction between gut microbiota and T cell immunity in colorectal cancer. Autoimmun Rev 2025; 24:103807. [PMID: 40139455 DOI: 10.1016/j.autrev.2025.103807] [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: 07/02/2024] [Revised: 02/26/2025] [Accepted: 03/22/2025] [Indexed: 03/29/2025]
Abstract
This review delves into the complex and multi-layered mechanisms that govern the interaction between gut microbiota and T cells in the context of colorectal cancer (CRC), revealing a novel "microbiota-immune regulatory landscape" within the tumor microenvironment. As CRC progresses, the gut microbiota experiences a significant transformation in both its composition and metabolic patterns. On one hand, specific microbial entities within the gut microbiota can directly engage with T cells, functioning as "immunological triggers" that shape T-cell behavior. Simultaneously, microbial metabolites, such as short-chain fatty acids and bile acids, serve as "molecular regulators" that intricately govern T-cell function and differentiation, fine-tuning the immune response. On the other hand, the quorum-sensing mechanism, a recently recognized communication network among bacteria, also plays a pivotal role in orchestrating T-cell immunity. Additionally, the gut microbiota forms an intriguing connection with the neuro-immune regulatory axis, a largely unexplored "territory" in CRC research. Regarding treatment strategies, a diverse array of intervention approaches-including dietary modifications, the utilization of probiotics, bacteriophages, and targeted antibiotic therapies-offer promising prospects for restoring the equilibrium of the gut microbiota, thereby acting as "ecosystem renovators" that impede tumor initiation and progression. Nevertheless, the current research landscape in this field is fraught with challenges. These include significant variations in microbial composition, dietary preferences, and tumor microenvironments among individuals, a lack of large-scale cohort studies, and insufficient research that integrates tumor mutation analysis, gut microbiota investigations, and immune microenvironment evaluations. This review emphasizes the necessity for future research efforts to seamlessly incorporate multiple factors and utilize bioinformatics analysis to construct a more comprehensive "interactive map" of the gut microbiota-T cell relationship in CRC. The aim is to establish a solid theoretical basis for the development of highly effective and personalized treatment regimens, ultimately transforming the therapeutic approach to CRC.
Collapse
Affiliation(s)
- Zhuang Jing
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China; Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China; Zhejiang-France United Laboratory of Integrated Traditional Chinese and Modern Medicine in Colorectal Cancer, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China
| | - Wu Yinhang
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China; Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China; Zhejiang-France United Laboratory of Integrated Traditional Chinese and Modern Medicine in Colorectal Cancer, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China
| | - Chu Jian
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China; Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China; Zhejiang-France United Laboratory of Integrated Traditional Chinese and Modern Medicine in Colorectal Cancer, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China
| | - Qu Zhanbo
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China; Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China; Zhejiang-France United Laboratory of Integrated Traditional Chinese and Modern Medicine in Colorectal Cancer, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China
| | - Wu Xinyue
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China; Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China; Zhejiang-France United Laboratory of Integrated Traditional Chinese and Modern Medicine in Colorectal Cancer, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China
| | - Han Shuwen
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China; Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China; Zhejiang-France United Laboratory of Integrated Traditional Chinese and Modern Medicine in Colorectal Cancer, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province 313000, People's Republic of China; ASIR (Institute - Association of intelligent systems and robotics), 14B rue Henri Sainte Claire Deville, 92500 Rueil-Malmaison, France.
| |
Collapse
|
2
|
Cornejo-Granados F, Gallardo-Becerra L, Romero-Hidalgo S, Lopez-Zavala AA, Cota-Huízar A, Cervantes-Echeverría M, Sotelo-Mundo RR, Ochoa-Leyva A. Host genome drives the microbiota enrichment of beneficial microbes in shrimp: exploring the hologenome perspective. Anim Microbiome 2025; 7:50. [PMID: 40405248 PMCID: PMC12100935 DOI: 10.1186/s42523-025-00414-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 04/18/2025] [Indexed: 05/24/2025] Open
Abstract
BACKGROUND Pacific Whiteleg shrimp (Litopenaeus vannamei) is an important model for breeding programs to improve global aquaculture productivity. However, the interaction between host genetics and microbiota in enhancing productivity remains poorly understood. We investigated the effect of two shrimp genetic lines, Fast-Growth (Gen1) and Disease-Resistant (Gen2), on the microbiota of L. vannamei. RESULTS Using genome-wide SNP microarray analysis, we confirmed that Gen1 and Gen2 represented distinct genetic populations. After confirming that the rearing pond did not significantly influence the microbiota composition, we determined that genetic differences explained 15.8% of the microbiota variability, with a stronger selective pressure in the hepatopancreas than in the intestine. Gen1, which exhibited better farm productivity, fostered a microbiota with greater richness, diversity, and resilience than Gen2, along with a higher abundance of beneficial microbes. Further, we demonstrated that a higher abundance of beneficial microbes was associated with healthier shrimp vs. diseased specimens, suggesting that Gen1 could improve shrimp's health and productivity by promoting beneficial microbes. Finally, we determined that the microbiota of both genetic lines was significantly different from their wild-type counterparts, suggesting farm environments and selective breeding programs strongly alter the natural microbiome. CONCLUSIONS This study highlights the importance of exploring the hologenome perspective, where integrating host genetics and microbiome composition can enhance breeding programs and farming practices.
Collapse
Affiliation(s)
- Fernanda Cornejo-Granados
- Departamento de Microbiología Molecular, Instituto de Biotecnología (IBT), Universidad Nacional Autónoma de México (UNAM), Av. Universidad 2001, Col. Chamilpa, 62210, Cuernavaca, Morelos, México.
| | - Luigui Gallardo-Becerra
- Departamento de Microbiología Molecular, Instituto de Biotecnología (IBT), Universidad Nacional Autónoma de México (UNAM), Av. Universidad 2001, Col. Chamilpa, 62210, Cuernavaca, Morelos, México
| | - Sandra Romero-Hidalgo
- Departamento de Genómica Computacional, Instituto Nacional de Medicina Genómica, Secretaría de Salud (INMEGEN), Periférico Sur No. 4809, 14610, México, DF, México
| | - Alonso A Lopez-Zavala
- Departamento de Ciencias Químico Biológicas, Universidad de Sonora (UNISON), Blvd., Rosales y Luis Encinas, 83000, Hermosillo, Sonora, México
| | - Andrés Cota-Huízar
- Camarones El Renacimiento SPR de RI, Justino Rubio No. 26, Col Ejidal, 81330, Higuera de Zaragoza, Sinaloa, México
| | - Melany Cervantes-Echeverría
- Departamento de Microbiología Molecular, Instituto de Biotecnología (IBT), Universidad Nacional Autónoma de México (UNAM), Av. Universidad 2001, Col. Chamilpa, 62210, Cuernavaca, Morelos, México
| | - Rogerio R Sotelo-Mundo
- Laboratorio de Estructura Biomolecular, Centro de Investigación en Alimentación y Desarrollo, A.C. (CIAD), Carretera Gustavo Enrique Astiazarán Rosas Num. 46. Col. La Victoria, 83304, Hermosillo, Sonora, México
| | - Adrian Ochoa-Leyva
- Departamento de Microbiología Molecular, Instituto de Biotecnología (IBT), Universidad Nacional Autónoma de México (UNAM), Av. Universidad 2001, Col. Chamilpa, 62210, Cuernavaca, Morelos, México.
| |
Collapse
|
3
|
Ismeurt-Walmsley C, Giannoni P, Servant F, Mekki LN, Baranger K, Rivera S, Marin P, Lelouvier B, Claeysen S. The same but different: impact of animal facility sanitary status on a transgenic mouse model of Alzheimer's disease. mBio 2025; 16:e0400124. [PMID: 40243365 PMCID: PMC12077201 DOI: 10.1128/mbio.04001-24] [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: 12/21/2024] [Accepted: 03/25/2025] [Indexed: 04/18/2025] Open
Abstract
The gut-brain axis has emerged as a key player in the regulation of brain function and cognitive health. Gut microbiota dysbiosis has been observed in preclinical models of Alzheimer's disease and patients. Manipulating the composition of the gut microbiota enhances or delays neuropathology and cognitive deficits in mouse models. Accordingly, the health status of the animal facility may strongly influence these outcomes. In the present study, we longitudinally analyzed the fecal microbiota composition and amyloid pathology of 5XFAD mice housed in a specific opportunistic pathogen-free (SOPF) and a conventional facility. The composition of the microbiota of 5XFAD mice after aging in conventional facility showed marked differences compared to WT littermates that were not observed when the mice were bred in SOPF facility. The development of amyloid pathology was also enhanced by conventional housing. We then transplanted fecal microbiota (FMT) from both sources into wild-type (WT) mice and measured memory performance, assessed in the novel object recognition test, in transplanted animals. Mice transplanted with microbiota from conventionally bred 5XFAD mice showed impaired memory performance, whereas FMT from mice housed in SOPF facility did not induce memory deficits in transplanted mice. Finally, 18 weeks of housing SOPF-born animals in a conventional facility resulted in the reappearance of specific microbiota compositions in 5XFAD vs WT mice. In conclusion, these results show a strong impact of housing conditions on microbiota-associated phenotypes and question the relevance of breeding preclinical models in specific pathogen-free (SPF) facilities. IMPORTANCE Housing conditions affect the composition of the gut microbiota. Gut microbiota of 6-month-old conventionally bred Alzheimer's mice is dysbiotic. Gut dysbiosis is absent in Alzheimer's mice housed in highly sanitized facilities. Transfer of fecal microbiota from conventionally bred mice affects cognition. Microbiota of mice housed in highly sanitized facilities has no effect on cognition.
Collapse
Affiliation(s)
| | - Patrizia Giannoni
- IGF, Univ. Montpellier, CNRS, INSERM, Montpellier, Occitanie, France
| | | | - Linda-Nora Mekki
- IGF, Univ. Montpellier, CNRS, INSERM, Montpellier, Occitanie, France
| | - Kevin Baranger
- Aix-Marseille Univ, CNRS, INP, Inst Neurophysiopathol, Marseille, Provence-Alpes-Côte d'Azur, France
| | - Santiago Rivera
- Aix-Marseille Univ, CNRS, INP, Inst Neurophysiopathol, Marseille, Provence-Alpes-Côte d'Azur, France
| | - Philippe Marin
- IGF, Univ. Montpellier, CNRS, INSERM, Montpellier, Occitanie, France
| | | | - Sylvie Claeysen
- IGF, Univ. Montpellier, CNRS, INSERM, Montpellier, Occitanie, France
| |
Collapse
|
4
|
Zhao JX, Elsheikha HM, Shang KM, Su JW, Wei YJ, Qin Y, Zhao ZY, Ma H, Zhang XX. Investigation of the genetic diversity of gut mycobiota of the wild and laboratory mice. Microbiol Spectr 2025; 13:e0284024. [PMID: 40162766 PMCID: PMC12054021 DOI: 10.1128/spectrum.02840-24] [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: 11/05/2024] [Accepted: 03/05/2025] [Indexed: 04/02/2025] Open
Abstract
Mice are colonized by diverse gut fungi, known as the mycobiota, which have received much less attention than bacterial microbiota. Here, we studied the diversities and structures of cecal fungal communities in wild (Lasiopodomys brandtii, Apodemus agrarius, and Microtus fortis) vs laboratory C57BL/6J mice to disentangle the contributions of gut fungi to the adaptation of mice to genetic diversity. Using ITS1 gene sequencing, we obtained 2,912 amplicon sequence variants (ASVs) and characterized the composition and diversity of cecal mycobiota in mice. There were significant differences in the composition of cecal fungal communities between wild and C57BL/6J mice, with more species diversity and richness of fungi in wild mice than C57BL/6J mice. We cultured 428 fungal strains from the cecal mycobiota, sequenced the whole genome of 48 selected strains, and identified 500,849 genes. Functional annotation analysis revealed multiple pathways related to energy metabolism, carbohydrate metabolism, fatty acid metabolism, and enzymes involved in the degradation of polysaccharides, lipids, and proteins, and secondary metabolite biosynthesis. The functions and abundance of Hypocreales and Pleosporales, which included the majority of the crucial metabolic pathways, were significantly higher in wild mice than in C57BL/6J mice. The results suggest that variations in the fungal community composition may relate to the adaptability of mice to their environmental habitats. IMPORTANCE In this study, we analyzed the fungal microbiota of three wild mouse species alongside laboratory mice using ITS1 amplicon sequencing. By integrating whole-genome sequencing with culturomics, we sequenced the genomes of 48 fungi isolated from cultured strains and investigated their biological functions to understand the role of intestinal fungi in the environmental adaptability of wild mice. This investigation has expanded the functional gene repository of gut fungi and shed new light on the intricate interplay between mice and their gut fungal communities. The data offer valuable insight into the ecological adaptation in wild mice, emphasizing the complex and dynamic relationship between the murine hosts and their mycobiota.
Collapse
Affiliation(s)
- Ji-Xin Zhao
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong, China
| | - Hany M. Elsheikha
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Science, University of Nottingham, Loughborough, United Kingdom
| | - Kai-Meng Shang
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong, China
| | - Jin-Wen Su
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong, China
| | - Yong-Jie Wei
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong, China
| | - Ya Qin
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong, China
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, Jilin, China
| | - Zi-Yu Zhao
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, Jilin, China
| | - He Ma
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong, China
| | - Xiao-Xuan Zhang
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong, China
| |
Collapse
|
5
|
Metwaly A, Kriaa A, Hassani Z, Carraturo F, Druart C, Arnauts K, Wilmes P, Walter J, Rosshart S, Desai MS, Dore J, Fasano A, Blottiere HM, Maguin E, Haller D. A Consensus Statement on establishing causality, therapeutic applications and the use of preclinical models in microbiome research. Nat Rev Gastroenterol Hepatol 2025; 22:343-356. [PMID: 40033063 DOI: 10.1038/s41575-025-01041-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/17/2025] [Indexed: 03/05/2025]
Abstract
The gut microbiome comprises trillions of microorganisms and profoundly influences human health by modulating metabolism, immune responses and neuronal functions. Disruption in gut microbiome composition is implicated in various inflammatory conditions, metabolic disorders and neurodegenerative diseases. However, determining the underlying mechanisms and establishing cause and effect is extremely difficult. Preclinical models offer crucial insights into the role of the gut microbiome in diseases and help identify potential therapeutic interventions. The Human Microbiome Action Consortium initiated a Delphi survey to assess the utility of preclinical models, including animal and cell-based models, in elucidating the causal role of the gut microbiome in these diseases. The Delphi survey aimed to address the complexity of selecting appropriate preclinical models to investigate disease causality and to study host-microbiome interactions effectively. We adopted a structured approach encompassing a literature review, expert workshops and the Delphi questionnaire to gather insights from a diverse range of stakeholders. Experts were requested to evaluate the strengths, limitations, and suitability of these models in addressing the causal relationship between the gut microbiome and disease pathogenesis. The resulting consensus statements and recommendations provide valuable insights for selecting preclinical models in future studies of gut microbiome-related diseases.
Collapse
Affiliation(s)
- Amira Metwaly
- Chair of Nutrition and Immunology, TUM School of Life Sciences, Technical University Munich, Freising, Germany
- ZIEL Institute for Food & Health, Technical University Munich, Freising, Germany
| | - Aicha Kriaa
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | | | - Federica Carraturo
- European Biomedical Research Institute of Salerno (EBRIS), Salerno, Italy
| | | | - Kaline Arnauts
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, KU Leuven, Leuven, Belgium
| | - 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
| | - Jens Walter
- APC Microbiome Ireland, School of Microbiology, and Department of Medicine, University College Cork, Cork, Ireland
| | - Stephan Rosshart
- Department of Microbiome Research, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Department of Medicine II, Medical Center - University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Mahesh S Desai
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Joel Dore
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
- Université Paris-Saclay, INRAE, MetaGenoPolis, Jouy-en-Josas, France
| | - Alessio Fasano
- European Biomedical Research Institute of Salerno (EBRIS), Salerno, Italy
- Department of Paediatric Gastroenterology and Nutrition, Mucosal Immunology and Biology Research Center,Massachusetts General Hospital Brigham, Harvard Medical School, Boston, MA, USA
| | - Hervé M Blottiere
- Université Paris-Saclay, INRAE, MetaGenoPolis, Jouy-en-Josas, France
- Nantes Université, INRAE, UMR1280, PhAN, Nantes, France
| | - Emmanuelle Maguin
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France.
| | - Dirk Haller
- Chair of Nutrition and Immunology, TUM School of Life Sciences, Technical University Munich, Freising, Germany.
- ZIEL Institute for Food & Health, Technical University Munich, Freising, Germany.
| |
Collapse
|
6
|
Zhang R, Zhang X, Lau HCH, Yu J. Gut microbiota in cancer initiation, development and therapy. SCIENCE CHINA. LIFE SCIENCES 2025; 68:1283-1308. [PMID: 39821827 DOI: 10.1007/s11427-024-2831-x] [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: 12/03/2024] [Accepted: 12/12/2024] [Indexed: 01/19/2025]
Abstract
Cancer has long been associated with genetic and environmental factors, but recent studies reveal the important role of gut microbiota in its initiation and progression. Around 13% of cancers are linked to infectious agents, highlighting the need to identify the specific microorganisms involved. Gut microbiota can either promote or inhibit cancer growth by influencing oncogenic signaling pathways and altering immune responses. Dysbiosis can lead to cancer, while certain probiotics and their metabolites may help reestablish micro-ecological balance and improve anti-tumor immune responses. Research into targeted approaches that enhance therapy with probiotics is promising. However, the effects of probiotics in humans are complex and not yet fully understood. Additionally, methods to counteract harmful bacteria are still in development. Early clinical trials also indicate that modifying gut microbiota may help manage side effects of cancer treatments. Ongoing research is crucial to understand better how gut microbiota can be used to improve cancer prevention and treatment outcomes.
Collapse
Affiliation(s)
- Ruyi Zhang
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Xiang Zhang
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Harry Cheuk Hay Lau
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Jun Yu
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
7
|
Rehermann B, Graham AL, Masopust D, Hamilton SE. Integrating natural commensals and pathogens into preclinical mouse models. Nat Rev Immunol 2025; 25:385-397. [PMID: 39562646 DOI: 10.1038/s41577-024-01108-3] [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: 10/18/2024] [Indexed: 11/21/2024]
Abstract
Fundamental discoveries in many aspects of mammalian physiology have been made using laboratory mice as research models. These studies have been facilitated by the genetic tractability and inbreeding of such mice, the large set of immunological reagents that are available, and the establishment of environmentally controlled, high-throughput facilities. Such facilities typically include barriers to keep the mouse colonies free of pathogens and the frequent re-derivation of the mice severely limits their commensal flora. Because humans have co-evolved with microorganisms and are exposed to a variety of pathogens, a growing community of researchers posits that preclinical disease research can be improved by studying mice in the context of the microbiota and pathogens that they would encounter in the natural world. Here, we provide a perspective of how these different approaches can be combined and integrated to improve existing mouse models to enhance our understanding of disease mechanisms and develop new therapies for humans. We also propose that the term 'mice with natural microbiota' is more appropriate for describing these models than existing terms such as 'dirty mice'.
Collapse
Affiliation(s)
- Barbara Rehermann
- Immunology Section, Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA.
| | - Andrea L Graham
- Department of Ecology & Evolutionary Biology, Princeton University, Princeton, NJ, USA
| | - David Masopust
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Sara E Hamilton
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
8
|
Weitzman CL, Day K, Brown GP, Gibb K, Christian K. Differential Temporal Shifts in Skin Bacteria on Wild and Captive Toads. MICROBIAL ECOLOGY 2025; 88:35. [PMID: 40301143 PMCID: PMC12040999 DOI: 10.1007/s00248-025-02537-w] [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: 01/21/2025] [Accepted: 04/17/2025] [Indexed: 05/01/2025]
Abstract
Skin bacteria on amphibian hosts play an important role in host health, but those communities are also constantly shifting based on environmental and host-related feedback. On some hosts, stability of skin communities depends on relatively abundant taxa, with less abundant taxa more readily entering and exiting the system. Cane toads (Rhinella marina) have invaded widespread, diverse tropical ecosystems, with varying ecology, physiology, and behaviour in different environments. In this study, we described temporal patterns of skin bacterial communities on cane toads at a site in northern Australia through the wet and dry seasons over two years. Toads in the wild population were paired with a captive-held population, housed in a semi-natural environment, to detect effects of time and season on wild toads, explore bacterial transience and volatility in skin taxa, and determine the extent to which skin communities on captive toads represent those on the wild population. We found community differences by captivity status, sampling timepoint, and season, with increased richness in the wet season on wild toads. Bacterial communities also became more similar among individuals (lower dispersion) in the wet season. Captive toads harboured more stable communities over time, likely owing to the reduced bacterial reservoirs experienced while in captivity. We propose that cane toads, with varied movement patterns among their diverse invaded habitats, provide an interesting direction for future work understanding the influences of habitat and movement on skin microbes, and the flexibility of microbial symbiotic interactions in invasive hosts.
Collapse
Affiliation(s)
- Chava L Weitzman
- Research Institute for the Environment and Livelihoods, Charles Darwin University, Casuarina, NT, Australia.
| | - Kimberley Day
- Research Institute for the Environment and Livelihoods, Charles Darwin University, Casuarina, NT, Australia
| | - Gregory P Brown
- School of Natural Sciences, Macquarie University, Sydney, NSW, Australia
| | - Karen Gibb
- Research Institute for the Environment and Livelihoods, Charles Darwin University, Casuarina, NT, Australia
| | - Keith Christian
- Research Institute for the Environment and Livelihoods, Charles Darwin University, Casuarina, NT, Australia
| |
Collapse
|
9
|
Brandstetter J, Hoffmann L, Koopmann I, Schreiber T, Schulz B, Rosshart SP, Zechner D, Vollmar B, Kumstel S. Burrowing Behavior as Robust Parameter for Early Humane Endpoint Determination in Murine Models for Pancreatic Cancer. Animals (Basel) 2025; 15:1241. [PMID: 40362056 PMCID: PMC12071103 DOI: 10.3390/ani15091241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 04/25/2025] [Accepted: 04/26/2025] [Indexed: 05/15/2025] Open
Abstract
Due to late-stage diagnoses and limited treatment options, pancreatic cancer is predicted to be the second leading cause of cancer deaths by 2030. Many different murine models were developed over the past decades to test new therapies for this tumor entity. The laws and regulations demand the continuous refinement of animal experiments in order to improve animal welfare sustainably. A key aspect here is the definition of early humane endpoint criteria to avoid severe and lasting suffering of the animals through timely euthanasia. The present study analyzed the welfare of mice in different pancreatic cancer models, various mouse strains, and under different therapeutic interventions in preclinical testing. Their welfare was monitored before any intervention, during tumor progression, and on the days before the individual humane endpoint for each mouse by assessing body weight change, distress score, perianal temperature, burrowing behavior, nesting activity, and mouse grimace scale. The data was retrospectively analyzed via receiver operating characteristic curve analysis to quantify the predictability of each parameter for humane endpoint determination. Burrowing behavior proved to be a robust predictor of the humane endpoint two days in advance under various conditions, including diverse pancreatic cancer models and different therapeutic approaches.
Collapse
Affiliation(s)
- Jakob Brandstetter
- Rudolf-Zenker-Institute of Experimental Surgery, University Medical Center Rostock, 18057 Rostock, Germany; (J.B.); (L.H.); (I.K.); (T.S.); (D.Z.); (B.V.)
| | - Lisa Hoffmann
- Rudolf-Zenker-Institute of Experimental Surgery, University Medical Center Rostock, 18057 Rostock, Germany; (J.B.); (L.H.); (I.K.); (T.S.); (D.Z.); (B.V.)
| | - Ingo Koopmann
- Rudolf-Zenker-Institute of Experimental Surgery, University Medical Center Rostock, 18057 Rostock, Germany; (J.B.); (L.H.); (I.K.); (T.S.); (D.Z.); (B.V.)
| | - Tim Schreiber
- Rudolf-Zenker-Institute of Experimental Surgery, University Medical Center Rostock, 18057 Rostock, Germany; (J.B.); (L.H.); (I.K.); (T.S.); (D.Z.); (B.V.)
| | - Benjamin Schulz
- Rudolf-Zenker-Institute of Experimental Surgery, University Medical Center Rostock, 18057 Rostock, Germany; (J.B.); (L.H.); (I.K.); (T.S.); (D.Z.); (B.V.)
| | - Stephan Patrick Rosshart
- Department of Microbiome Research, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany;
- Department of Medicine 1, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Dietmar Zechner
- Rudolf-Zenker-Institute of Experimental Surgery, University Medical Center Rostock, 18057 Rostock, Germany; (J.B.); (L.H.); (I.K.); (T.S.); (D.Z.); (B.V.)
| | - Brigitte Vollmar
- Rudolf-Zenker-Institute of Experimental Surgery, University Medical Center Rostock, 18057 Rostock, Germany; (J.B.); (L.H.); (I.K.); (T.S.); (D.Z.); (B.V.)
| | - Simone Kumstel
- Rudolf-Zenker-Institute of Experimental Surgery, University Medical Center Rostock, 18057 Rostock, Germany; (J.B.); (L.H.); (I.K.); (T.S.); (D.Z.); (B.V.)
| |
Collapse
|
10
|
Kolland D, Kuhlmann M, de Almeida GP, Köhler A, Arifovic A, von Strempel A, Pourjam M, Bolsega S, Wurmser C, Steiger K, Basic M, Neuhaus K, Schmidt-Weber CB, Stecher B, Zehn D, Ohnmacht C. A specific microbial consortium enhances Th1 immunity, improves LCMV viral clearance but aggravates LCMV disease pathology in mice. Nat Commun 2025; 16:3902. [PMID: 40274773 PMCID: PMC12022176 DOI: 10.1038/s41467-025-59073-x] [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: 02/07/2024] [Accepted: 04/10/2025] [Indexed: 04/26/2025] Open
Abstract
Anti-viral immunity can vary tremendously from individual to individual but mechanistic understanding is still scarce. Here, we show that a defined, low complex bacterial community (OMM12) but not the general absence of microbes in germ-free mice leads to a more potent immune response compared to the microbiome of specific-pathogen-free (SPF) mice after a systemic viral infection with LCMV Clone-13. Consequently, gnotobiotic mice colonized with OMM12 have more severe LCMV-induced disease pathology but also enhance viral clearance in the intestinal tract. Mechanistically, single-cell RNA sequencing analysis of adoptively transferred virus-specific T helper cells and endogenous T helper cells in the intestinal tract reveal a stronger pro-inflammatory Th1 profile and a more vigorous expansion in OMM12 than SPF mice. Altogether, our work highlights the causative function of the intestinal microbiome for shaping adaptive anti-viral immunity with implications for vaccination strategies and anti-cancer treatment regimens.
Collapse
Affiliation(s)
- Daphne Kolland
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center, Munich, Germany
| | - Miriam Kuhlmann
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Gustavo P de Almeida
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
- Center for Infection Prevention (ZIP), School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Amelie Köhler
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center, Munich, Germany
| | - Anela Arifovic
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center, Munich, Germany
| | - Alexandra von Strempel
- Max von Pettenkofer Institute of Hygiene and Medical Microbiology, Faculty of Medicine, LMU, Munich, Germany
| | - Mohsen Pourjam
- Core Facility Microbiome ZIEL - Institute for Food & Health, Technical University of Munich, Freising, Germany
| | - Silvia Bolsega
- Institute for Laboratory Animal Science and Central Animal Facility, Hannover Medical School, Hannover, Germany
| | - Christine Wurmser
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
- Center for Infection Prevention (ZIP), School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Katja Steiger
- Institute of Pathology, School of Medicine and Health, Technical University Munich, Munich, Germany
| | - Marijana Basic
- Institute for Laboratory Animal Science and Central Animal Facility, Hannover Medical School, Hannover, Germany
| | - Klaus Neuhaus
- Core Facility Microbiome ZIEL - Institute for Food & Health, Technical University of Munich, Freising, Germany
| | - Carsten B Schmidt-Weber
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center, Munich, Germany
- Member of the German Center of Lung Research (DZL), Partner Site Munich, Munich, Germany
| | - Bärbel Stecher
- Max von Pettenkofer Institute of Hygiene and Medical Microbiology, Faculty of Medicine, LMU, Munich, Germany
- German Center for Infection Research (DZIF), partner site LMU, Munich, Germany
| | - Dietmar Zehn
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany.
- Center for Infection Prevention (ZIP), School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany.
| | - Caspar Ohnmacht
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center, Munich, Germany.
| |
Collapse
|
11
|
Alegre ML, Atkinson C, Issa F, Valujskikh A, Zhang ZJ. Best practices of heart transplantation in mice. Am J Transplant 2025:S1600-6135(25)00217-5. [PMID: 40252924 DOI: 10.1016/j.ajt.2025.04.012] [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: 01/09/2025] [Revised: 03/21/2025] [Accepted: 04/12/2025] [Indexed: 04/21/2025]
Abstract
Heart transplantation in mice has served as a reliable in vivo model in transplant research worldwide for more than half a century. It is not only useful for addressing cardiac graft-specific questions but also provides mechanistic insights and therapeutic strategies that have a broad impact across all solid organ transplants. Compared to other mouse models of solid organ transplantation, such as kidney, lung, or small intestine transplants, the surgical techniques to perform mouse heart transplantation (mHT) are relatively easy to master, and the graft heartbeat offers a simple means to evaluate transplant viability. However, as with other in vivo mouse models, mHT has distinct strengths and limitations. Multiple factors can influence the accuracy and reproducibility of the results, including microsurgical techniques and microsurgeons' skills, postoperative monitoring methodologies, mouse strain combinations, and sex/age. As innovative biotechnologies continue to emerge, the future holds many opportunities for preclinical research utilizing the mHT model. It is therefore imperative to provide the field with optimized mHT protocols and maintain standard reporting requirements. This minireview provided a concise summary and recommendations for standardized practices to ensure the accuracy, reproducibility, and translational value of findings generated from the mHT model.
Collapse
Affiliation(s)
- Maria-Luisa Alegre
- Department of Medicine, Section of Rheumatology, The University of Chicago, Chicago, IL, USA
| | - Carl Atkinson
- Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine; Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Fadi Issa
- Nuffield Department of Surgical Sciences, University of Oxford, United Kingdom
| | - Anna Valujskikh
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland, Ohio, USA; Cleveland Clinic Lerner College of Medicine, Cleveland, Ohio, USA
| | - Zheng J Zhang
- Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine; Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
| |
Collapse
|
12
|
Ioannou P, Katsoulieris E, Afratis NA. Matrix Dynamics and Microbiome Crosstalk: Matrix Metalloproteinases as Key Players in Disease and Therapy. Int J Mol Sci 2025; 26:3621. [PMID: 40332093 PMCID: PMC12027064 DOI: 10.3390/ijms26083621] [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: 02/28/2025] [Revised: 04/02/2025] [Accepted: 04/09/2025] [Indexed: 05/08/2025] Open
Abstract
Matrix metalloproteinases (MMPs) are key enzymes involved in extracellular matrix (ECM) remodeling, regulating a wide range of cellular and immune processes in both homeostatic and pathological conditions. Host-microbiota interactions play a critical role in maintaining ECM balance; however, during dysbiosis, this regulation is disrupted, leading to compromised barrier integrity, pathogen translocation into circulation, and the development of systemic diseases and cancer. This review highlights the bidirectional relationship between MMP expression/activity and microbiota dysbiosis, emphasizing tissue-specific alterations in MMP activity that contribute to disease progression. In addition, it integrates interdisciplinary evidence to illustrate the MMP-dependent mechanisms underlying various pathologies associated with oral and gut microbiome dysbiosis, including long-range effects through the gut-skin and gut-brain axes. Thus, this review introduces the emerging field of MatrixBiome, which explores the complex interactions between the ECM, microbiota, and host tissues. Finally, it also outlines therapeutic strategies to modulate MMP levels, either indirectly through microbiome-targeted approaches (e.g., prebiotics, probiotics, and postbiotics) or directly using MMP inhibitors, offering promising avenues for future clinical interventions.
Collapse
Affiliation(s)
- Paraskevi Ioannou
- Laboratory of Biotechnology and Molecular Analysis, Department of Agricultural Development, Agri-Food & Management of Natural Resources, National and Kapodistrian University of Athens, Evripos Campus, 34400 Psachna, Evia, Greece (E.K.)
| | - Elias Katsoulieris
- Laboratory of Biotechnology and Molecular Analysis, Department of Agricultural Development, Agri-Food & Management of Natural Resources, National and Kapodistrian University of Athens, Evripos Campus, 34400 Psachna, Evia, Greece (E.K.)
| | - Nikolaos A. Afratis
- Laboratory of Biotechnology and Molecular Analysis, Department of Agricultural Development, Agri-Food & Management of Natural Resources, National and Kapodistrian University of Athens, Evripos Campus, 34400 Psachna, Evia, Greece (E.K.)
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, 234 Herzl Street, Rehovot 7610001, Israel
| |
Collapse
|
13
|
Achasova KM, Snytnikova OA, Chanushkina KE, Morozova MV, Tsentalovich YP, Kozhevnikova EN. Baseline abundance of Akkermansia muciniphila and Bacteroides acidifaciens in a healthy state predicts inflammation associated tumorigenesis in the AOM/DSS mouse model. Sci Rep 2025; 15:12241. [PMID: 40210644 PMCID: PMC11985942 DOI: 10.1038/s41598-025-96514-5] [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: 01/09/2025] [Accepted: 03/28/2025] [Indexed: 04/12/2025] Open
Abstract
Numerous studies demonstrate that intestinal microbiota contribute to colorectal cancer (CRC), which is often associated with dysbiosis. Most of the data were obtained from studies on CRC patients, making it challenging to determine whether alterations in microbiota are a consequence of the pathology or whether they actively drive its progression. Several studies using laboratory animals suggest that gut microbiota may be involved in both the onset and progression of CRC. In the present study we utilized the azoxymethane-dextran sulfate sodium (AOM/DSS) mouse model of CRC to investigate the contribution of healthy-state microbiota to inflammation-associated tumorigenesis. Two cohorts of C57BL/6 mice harboring different intestinal microbiota demonstrated different susceptibility to AOM/DSS treatment. Sequencing of 16S rRNA bacterial DNA from fecal samples revealed Akkermansia muciniphila and Bacteroides acidifaciens as marker features in the healthy-state microbiota (before AOM/DSS administration), which showed a strong positive correlation with tumor incidence. Moreover, the healthy-state abundance of these markers, considered beneficial bacteria, was strongly positively correlated with the sulfate-reducing bacteria Desulfovibrio fairfieldensis identified as a marker of chronic colitis-associated microbiota. Furthermore, the abundances of these marker features, associated with CRC outcome, correlated with the expression of interferon gamma and nitric oxide synthase 2 genes in colon tissue during the early stage of DSS-induced intestinal inflammation. In contrast to multiple studies demonstrating the anti-inflammatory properties of A. muciniphila and B. acidifaciens, our results point out their potential adverse effect under specific conditions of genotoxicity and inflammation in the intestine. Taken together, our findings suggest a complex, context-dependent role of commensal microbiota in inflammation-associated dysbiosis and CRC.
Collapse
Affiliation(s)
- Kseniya M Achasova
- Scientific Research Institute of Neurosciences and Medicine, Novosibirsk, Russia, 630117
- Institute of Molecular and Cellular Biology SB RAS, Novosibirsk, Russia, 630090
| | | | | | - Maryana V Morozova
- Institute of Molecular and Cellular Biology SB RAS, Novosibirsk, Russia, 630090
| | | | - Elena N Kozhevnikova
- Institute of Molecular and Cellular Biology SB RAS, Novosibirsk, Russia, 630090.
- Laboratory of Bioengineering, Novosibirsk State Agrarian University, Novosibirsk, Russia, 630039.
| |
Collapse
|
14
|
Hanski E, Joseph S, Curtis MA, Swann JW, Vallier M, Linnenbrink M, Baines JF, Jensen JK, Wolfenden A, Mair I, Else KJ, Bradley JE, Holthuijzen W, Plissner JH, Raulo A, Quicray M, Knowles SCL. Wild house mice have a more dynamic and aerotolerant gut microbiota than laboratory mice. BMC Microbiol 2025; 25:204. [PMID: 40205328 PMCID: PMC11983872 DOI: 10.1186/s12866-025-03937-1] [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: 10/15/2024] [Accepted: 03/26/2025] [Indexed: 04/11/2025] Open
Abstract
The mammalian gut microbiota is a complex microbial community with diverse impacts on host biology. House mice (Mus musculus) are the major model organism for research on mammals, but laboratory domestication has altered their gut microbiota from that of their wild counterparts. Knowledge about how and why the gut microbiota of this species varies between lab and wild settings and among natural populations could improve its utility as a model organism. Here, we use a large dataset comprising over 800 house mouse samples from multiple laboratory facilities and strains and wild mice from mainland and island populations to investigate gut microbiota variation in this species across contrasting genetic and environmental settings. Across geographically disparate populations, we find that wild mice possess a gut microbiota that is compositionally distinct, displays a higher relative abundance and richness of aerotolerant taxa, and is taxonomically and functionally more diverse than that of lab mice. Longitudinally sampled wild mice also display markedly higher temporal turnover in microbiota composition than lab mice. Wild mice from oceanic islands harboured microbiotas that differed subtly from those of mainland wild mice and were more divergent from lab mouse microbiotas. These findings highlight much greater spatial and temporal turnover of gut microbes in wild compared to laboratory mice.
Collapse
Affiliation(s)
- Eveliina Hanski
- Department of Biology, University of Oxford, Oxford, UK.
- Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| | - Susan Joseph
- Centre for Host-Microbiome Interactions, King's College London, London, UK
| | - Michael A Curtis
- Centre for Host-Microbiome Interactions, King's College London, London, UK
| | - James W Swann
- Columbia Stem Cell Initiative, Columbia University, New York, USA
| | - Marie Vallier
- Max Planck Institute for Evolutionary Biology, Plön, Germany
| | | | - John F Baines
- Max Planck Institute for Evolutionary Biology, Plön, Germany
- Institute of Experimental Medicine, Kiel University, Kiel, Germany
| | | | - Andrew Wolfenden
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Iris Mair
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
- Institute of Ecology and Evolution, University of Edinburgh, Edinburgh, UK
| | - Kathryn J Else
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| | | | - Wieteke Holthuijzen
- Department of Ecology & Evolutionary Biology, University of Tennessee, Knoxville, USA
| | | | - Aura Raulo
- Department of Biology, University of Oxford, Oxford, UK
- Department of Computing, University of Turku, Turku, Finland
| | - Maude Quicray
- Department of Functional and Evolutionary Entomology, University of Liège, Liège, Belgium
| | | |
Collapse
|
15
|
Falkenberg C, Sørensen DB, Hansen CH, Toft MF, Hansen AK. Pre-immunization of diet-induced obese male mice with inactivated pathogens increases power in a liraglutide intervention study. Lab Anim 2025; 59:203-214. [PMID: 39696895 DOI: 10.1177/00236772241279058] [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] [Indexed: 12/20/2024]
Abstract
Pre-immunization with inactivated antigens has been developed as an alternative to the use of 'dirty' mice, which in contrast to specific pathogen free (SPF) mice, harbour a range of pathogens. Within certain research areas, such mice are considered better models for humans than SPF mice, as they have an immune system that better mirrors human immunity. We inactivated murine adenovirus type 1 (FL), minute virus of mice, mouse hepatitis virus (A59), respirovirus muris (Sendai), Theiler's encephalomyelitis virus (GD7) and Mycoplasma pulmonis by ultraviolet irradiation. We show that pre-immunization with these inactivated pathogens combined with adjuvant prior to the dietary induction of obesity in C57BL/6NTac mice substantially reduced the group sizes needed for showing an effect of the GLP-1 receptor analogue, liraglutide. Nesting, open field and novel object behaviours of the mice were unaffected. We conclude that pre-immunization with inactivated pathogens may be a simple tool to increase power in this type of intervention study on the DIO mouse model.
Collapse
Affiliation(s)
- Caroline Falkenberg
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Dorte B Sørensen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Camilla Hf Hansen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | | | - Axel K Hansen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| |
Collapse
|
16
|
Elmore J, Sahler J, Solouki S, Koylass N, Wang A, Nelissen S, Redko A, Huang W, August A. Diverse microbial exposure exacerbates the development of allergic airway inflammation in adult mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.21.644556. [PMID: 40196567 PMCID: PMC11974674 DOI: 10.1101/2025.03.21.644556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Background Exposure to a diversity of microbes has been implicated in playing a major role in susceptibility to the development of allergic lung type diseases. The hygiene hypothesis suggests that those exposed to a broad diversity of microbes are more likely to be protected against developing allergic type diseases. However, changes in exposure to microbial diversity can occur in both younger individuals, as well as in adults, and the effects are not always understood. Objective We investigated the effect of exposure to broad microbial diversity on the airway T cell response in house dust mite (HDM) induced allergic airway disease (AAD, a model of allergic asthma). Methods We increased exposure to broad microbial diversity by co-housing specific pathogen free (SPF) adult or newborn mice with pet store mice (PSE or BiPSE, respectively). Mice were then exposed to HDM to induce AAD. Results We found that the effect of increased microbial exposure on the development of allergic airway inflammation differs by age. Increasing exposure to diverse microbes as adults exacerbates the development of allergic airway inflammation, whereas this was not observed when exposure occurred at birth. Conclusion We suggest that experimental evaluation of the hygiene hypothesis in inflammation, particularly those using mouse models, may need to consider age of the host and time of microbial exposure. Capsule Summary Mouse models of increased exposure to diverse microbial environment shown to differentially affect the development of allergic airway inflammation, depending on the age of microbial exposure.
Collapse
|
17
|
Roach SN, Phillips W, Pross LM, Sanders AE, Pierson MJ, Hunter RC, Langlois RA. Virus-induced perturbations in the mouse microbiome are impacted by microbial experience. mSphere 2025; 10:e0056324. [PMID: 39945519 PMCID: PMC11934326 DOI: 10.1128/msphere.00563-24] [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: 07/09/2024] [Accepted: 01/21/2025] [Indexed: 03/01/2025] Open
Abstract
The bacterial microbiome has a major impact on health and can shape metabolism, host tolerance, immune responses, and the outcome of future infections. The bacterial microbiome is highly variable between individuals. Specific pathogen-free animals have reduced microbiome diversity, making it difficult to evaluate the impact of infection-induced microbiome disruption that would be observed in free-living animals, including people. Mice are commonly used as a preclinical model but unfortunately often fail to predict translation success or failure, particularly for immune and infectious disease-targeting therapies. Here, we utilize pet store mouse cohoused "dirty" mice with diverse microbial experience to explore how host variability and infection may be interacting to drive unique microbiome changes. We found that cohoused animals had significantly increased bacterial diversity in the small intestine and cecum but not in the large intestine. There were differentially abundant taxa between clean and dirty animals in all three tissues. After infection with influenza A virus, samples clustered by both housing condition and infection status in the cecum and large intestine, while small intestine samples clustered predominantly by infection. Altogether, these results highlight the differential impact of housing, infection, and interaction between the two in dictating community composition across the gastrointestinal microbiome.IMPORTANCETraditionally housed pathogen-free mouse models do not fully capture the natural variability observed among human microbiomes, which may underlie their poor translationally predictive value. Understanding the difference between pathogen-induced shifts in the bacterial microbiome and natural microbiome variance is a major hurdle to determining bacterial biomarkers of disease. It is also critical to understand how diverse baseline microbiomes may be differentially impacted by infection and contribute to disease. Pet store cohoused "dirty" mice have diverse microbial experiences and microbiomes, allowing us to evaluate how baseline variation, infection, and interaction between the two impact the microbiome.
Collapse
Affiliation(s)
- Shanley N. Roach
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Wendy Phillips
- Minnesota Supercomputing Institute, University of Minnesota, Minneapolis, Minnesota, USA
| | - Lauren M. Pross
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Autumn E. Sanders
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Mark J. Pierson
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Ryan C. Hunter
- Department of Microbiology and Immunology, University at Buffalo, Getzville, New York, USA
| | - Ryan A. Langlois
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
18
|
Rahman R, Fouhse JM, Ju T, Fan Y, Bhardwaj T, Brook RK, Nosach R, Harding J, Willing BP. The impact of wild-boar-derived microbiota transplantation on piglet microbiota, metabolite profile, and gut proinflammatory cytokine production differs from sow-derived microbiota. Appl Environ Microbiol 2025; 91:e0226524. [PMID: 39902926 PMCID: PMC11921332 DOI: 10.1128/aem.02265-24] [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: 12/19/2024] [Indexed: 02/06/2025] Open
Abstract
Colonization of co-evolved, species-specific microbes in early life plays a crucial role in gastrointestinal development and immune function. This study hypothesized that modern pig production practices have resulted in the loss of co-evolved species and critical symbiotic host-microbe interactions. To test this, we reintroduced microbes from wild boars (WB) into conventional piglets to explore their colonization dynamics and effects on gut microbial communities, metabolite profiles, and immune responses. At postnatal day (PND) 21, 48 piglets were assigned to four treatment groups: (i) WB-derived mixed microbial community (MMC), (ii) sow-derived MMC, (iii) a combination of WB and sow MMC (Mix), or (iv) Control (PBS). Post-transplantation analyses at PND 48 revealed distinct microbial communities in WB-inoculated piglets compared with Controls, with trends toward differentiation from Sow but not Mix groups. WB-derived microbes were more successful in colonizing piglets, particularly in the Mix group, where they competed with Sow-derived microbes. WB group cecal digesta enriched with Lactobacillus helveticus, Lactobacillus mucosae, and Lactobacillus pontis. Cecal metabolite analysis showed that WB piglets were enriched in histamine, acetyl-ornithine, ornithine, citrulline, and other metabolites, with higher histamine levels linked to Lactobacillus abundance. WB piglets exhibited lower cecal IL-1β and IL-6 levels compared with Control and Sow groups, whereas the Mix group showed reduced IFN-γ, IL-2, and IL-6 compared with the Sow group. No differences in weight gain, fecal scores, or plasma cytokines were observed, indicating no adverse effects. These findings support that missing WB microbes effectively colonize domestic piglets and may positively impact metabolite production and immune responses.IMPORTANCEThis study addresses the growing concern over losing co-evolved, species-specific microbes in modern agricultural practices, particularly in pig production. The implementation of strict biosecurity measures and widespread antibiotic use in conventional farming systems may disrupt crucial host-microbe interactions that are essential for gastrointestinal development and immune function. Our research demonstrates that by reintroducing wild boar-derived microbes into domestic piglets, these microbes can successfully colonize the gut, influence microbial community composition, and alter metabolite profiles and immune responses without causing adverse effects. These findings also suggest that these native microbes can fill an intestinal niche, positively impacting immune activation. This research lays the groundwork for future strategies to enhance livestock health and performance by restoring natural microbial populations that produce immune-modulating metabolites.
Collapse
Affiliation(s)
- Rajibur Rahman
- Department of Agricultural Food & Nutritional Science, Faculty of Agricultural, Life & Environmental Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Janelle M. Fouhse
- Department of Agricultural Food & Nutritional Science, Faculty of Agricultural, Life & Environmental Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Tingting Ju
- Department of Agricultural Food & Nutritional Science, Faculty of Agricultural, Life & Environmental Sciences, University of Alberta, Edmonton, Alberta, Canada
- Department of Animal Sciences, Purdue University, West Lafayette, Indiana, USA
| | - Yi Fan
- Department of Agricultural Food & Nutritional Science, Faculty of Agricultural, Life & Environmental Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Tulika Bhardwaj
- Department of Agricultural Food & Nutritional Science, Faculty of Agricultural, Life & Environmental Sciences, University of Alberta, Edmonton, Alberta, Canada
- University of Calgary, Calgary, Alberta, Canada
| | - Ryan K. Brook
- College of Agriculture and Bioresources, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Roman Nosach
- Department of Large Animal Clinical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - John Harding
- Department of Large Animal Clinical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Benjamin P. Willing
- Department of Agricultural Food & Nutritional Science, Faculty of Agricultural, Life & Environmental Sciences, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
19
|
Chen YH, Zaldana K, Yeung F, Vujkovic-Cvijin I, Downie AE, Lin JD, Yang Y, Herrmann C, Oyesola O, Rozenberg F, Schwartz RE, Kim D, Tio K, Belkaid Y, Loke P, Graham AL, Koralov SB, Cadwell K. Rewilding catalyzes maturation of the humoral immune system. SCIENCE ADVANCES 2025; 11:eads2364. [PMID: 40053586 PMCID: PMC11887799 DOI: 10.1126/sciadv.ads2364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 01/31/2025] [Indexed: 03/09/2025]
Abstract
Inbred mice used for biomedical research display an underdeveloped immune system compared with adult humans, which is attributed in part to the artificial laboratory environment. Despite representing a central component of adaptive immunity, the impact of the laboratory environment on the B cell compartment has not been investigated in detail. Here, we performed an in-depth examination of B cells following rewilding, the controlled release of inbred laboratory mice into an outdoor enclosure. In rewilded mice, we observed B cells in circulation with increased signs of maturation, alongside heightened germinal center responses within secondary lymphoid organs. Rewilding also expanded B cells in the gut, which was accompanied by elevated systemic levels of immunoglobulin G (IgG) and IgM antibodies reactive to the microbiota. Our findings indicate that exposing laboratory mice to a more natural environment enhances B cell development to better reflect the immune system of free-living mammals.
Collapse
Affiliation(s)
- Ying-Han Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Kimberly Zaldana
- Vilcek Institute of Graduate Biomedical Sciences, New York University Grossman School of Medicine, New York, NY 10016, USA
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Frank Yeung
- Vilcek Institute of Graduate Biomedical Sciences, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Ivan Vujkovic-Cvijin
- Department of Biomedical Sciences & F. Widjaja Inflammatory Bowel Disease Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Alexander E. Downie
- Department of Primate Behavior and Evolution, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Jian-Da Lin
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei City, Taiwan
| | - Yi Yang
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Christin Herrmann
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Oyebola Oyesola
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Felix Rozenberg
- SUNY Downstate Health Sciences University, New York, NY 11203, USA
| | - Robert E. Schwartz
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - David Kim
- Vilcek Institute of Graduate Biomedical Sciences, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Kurt Tio
- Vilcek Institute of Graduate Biomedical Sciences, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Yasmine Belkaid
- Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
- Metaorganism Laboratory, Department of Immunology, Pasteur Institute, Paris, France
| | - P’ng Loke
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Andrea L. Graham
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, NJ 08544, USA
- Santa Fe Institute, Santa Fe, NM 87501, USA
| | - Sergei B. Koralov
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Ken Cadwell
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA
| |
Collapse
|
20
|
Sprockett DD, Dillard BA, Landers AA, Sanders JG, Moeller AH. Recent genetic drift in the co-diversified gut bacterial symbionts of laboratory mice. Nat Commun 2025; 16:2218. [PMID: 40044678 PMCID: PMC11883045 DOI: 10.1038/s41467-025-57435-z] [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: 08/07/2024] [Accepted: 02/19/2025] [Indexed: 03/09/2025] Open
Abstract
Laboratory mice (Mus musculus domesticus) harbor gut bacterial strains that are distinct from those of wild mice but whose evolutionary histories are unclear. Here, we show that laboratory mice have retained gut bacterial lineages that diversified in parallel (co-diversified) with rodent species for > 25 million years, but that laboratory-mouse gut microbiota (LGM) strains of these ancestral symbionts have experienced accelerated accumulation of genetic load during the past ~ 120 years of captivity. Compared to closely related wild-mouse gut microbiota (WGM) strains, co-diversified LGM strains displayed significantly faster genome-wide rates of nonsynonymous substitutions, indicating elevated genetic drift-a difference that was absent in non-co-diversified symbiont clades. Competition experiments in germ-free mice further indicated that LGM strains within co-diversified clades displayed significantly reduced fitness in vivo compared to WGM relatives to an extent not observed within non-co-diversified clades. Thus, stochastic processes (e.g., bottlenecks), not natural selection in the laboratory, have been the predominant evolutionary forces underlying divergence of co-diversified symbiont strains between laboratory and wild house mice. Our results show that gut bacterial lineages conserved in diverse rodent species have acquired novel mutational burdens in laboratory mice, providing an evolutionary rationale for restoring laboratory mice with wild gut bacterial strain diversity.
Collapse
Affiliation(s)
- Daniel D Sprockett
- Department of Ecology and Evolutionary Biology, Cornell University, Ithaca, NY, 14853, USA
| | - Brian A Dillard
- Department of Ecology and Evolutionary Biology, Cornell University, Ithaca, NY, 14853, USA
| | - Abigail A Landers
- Department of Microbiology, Cornell University, Ithaca, NY, 14853, USA
| | - Jon G Sanders
- Department of Ecology and Evolutionary Biology, Cornell University, Ithaca, NY, 14853, USA
| | - Andrew H Moeller
- Department of Ecology and Evolutionary Biology, Cornell University, Ithaca, NY, 14853, USA.
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, NJ, 08540, USA.
| |
Collapse
|
21
|
Thiran A, Vereecke L. New thoughts on the gut-immune axis of arthritis. Trends Immunol 2025; 46:206-218. [PMID: 40069048 DOI: 10.1016/j.it.2025.01.006] [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: 11/20/2024] [Revised: 12/23/2024] [Accepted: 01/27/2025] [Indexed: 03/21/2025]
Abstract
Arthritis is associated with varying degrees of intestinal inflammation and microbiota dysbiosis, leading to the 'gut-joint axis hypothesis' in which intestinal and joint inflammation are suggested to be interconnected through immune-microbiota interactions. While clinical observations support this, causality remains uncertain. Rodent models have provided insights into potential mechanisms by uncovering microbial influences and immune pathways that either connect or uncouple gut and joint inflammation. Based on recent findings, we propose the 'immune hypersensitivity hypothesis' whereby central immune hyper-reactivity can independently drive joint inflammation via local sterile triggers, and gut inflammation via microbial triggers. We argue that this suggests a more nuanced role of the microbiota in arthritis pathogenesis that varies according to the predominant immune mechanisms in disease subtypes. We explore gut-immune interactions in arthritis, highlight ongoing challenges, and propose future research directions.
Collapse
Affiliation(s)
- Alexandra Thiran
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium; Vlaams Instituut voor Biotechnologie (VIB)-University of Ghent (UGent) Center for Inflammation Research, Ghent, Belgium; Ghent Gut Inflammation Group (GGIG), Ghent, Belgium
| | - Lars Vereecke
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium; Vlaams Instituut voor Biotechnologie (VIB)-University of Ghent (UGent) Center for Inflammation Research, Ghent, Belgium; Ghent Gut Inflammation Group (GGIG), Ghent, Belgium.
| |
Collapse
|
22
|
Zhou H, Huang W, Li J, Chen P, Shen L, Huang W, Mai K, Zou H, Shi X, Weng Y, Liu Y, Yang Z, Ou C. Oral probiotic extracellular vesicle therapy mitigates Influenza A Virus infection via blunting IL-17 signaling. Bioact Mater 2025; 45:401-416. [PMID: 39697241 PMCID: PMC11652895 DOI: 10.1016/j.bioactmat.2024.11.016] [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: 07/30/2024] [Revised: 11/12/2024] [Accepted: 11/12/2024] [Indexed: 12/20/2024] Open
Abstract
The influenza A virus (IAV) damages intestinal mucosal tissues beyond the respiratory tract. Probiotics play a crucial role in maintaining the balance and stability of the intestinal microecosystem. Extracellular vesicles (EVs) derived from probiotics have emerged as potential mediators of host immune response and anti-inflammatory effect. However, the specific anti-inflammatory effects and underlying mechanisms of probiotics-derived EVs on IAV remain unclear. In the present study, we investigated the therapeutic efficacy of Lactobacillus reuteri EHA2-derived EVs (LrEVs) in a mouse model of IAV infection. Oral LrEVs were distributed in the liver, lungs, and gastrointestinal tract. In mice infected with IAV, oral LrEVs administration alleviated IAV-induced damages in the lungs and intestines, modified the microbiota compositions, and increased the levels of short-chain fatty acids in those organs. Mechanistically, LrEVs exerted their protective effects against IAV infection by blunting the pro-inflammatory IL-17 signaling. Furthermore, FISH analysis detected miR-4239, one of the most abundant miRNAs in LrEVs, in both lung and intestinal tissues. We confirmed that miR-4239 directly targets IL-17a. Our findings paved the ground for future application of LrEVs in influenza treatment and offered new mechanistic insights regarding the anti-inflammatory role of miR-4239.
Collapse
Affiliation(s)
- Hongxia Zhou
- Dongguan Institute of Respiratory and Critical Care Medicine, The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan, 523018, China
| | - Wenbo Huang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Jieting Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Peier Chen
- The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan, 523018, China
| | - Lihan Shen
- Dongguan Institute of Respiratory and Critical Care Medicine, The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan, 523018, China
| | - Wenjing Huang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Kailin Mai
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Heyan Zou
- Dongguan Institute of Respiratory and Critical Care Medicine, The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan, 523018, China
| | - Xueqin Shi
- The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan, 523018, China
| | - Yunceng Weng
- Becton Dickinson Medical Devices (Shanghai) Co., Ltd., Guangzhou, 510180, China
| | - Yuhua Liu
- Department of General Practice, The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan, 523018, China
| | - Zifeng Yang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
- Guangzhou National Laboratory, Guangzhou, 510000, China
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau SAR, 519020, China
| | - Caiwen Ou
- The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan, 523018, China
| |
Collapse
|
23
|
Zalila-Kolsi I, Dhieb D, Osman HA, Mekideche H. The Gut Microbiota and Colorectal Cancer: Understanding the Link and Exploring Therapeutic Interventions. BIOLOGY 2025; 14:251. [PMID: 40136508 PMCID: PMC11939563 DOI: 10.3390/biology14030251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 02/23/2025] [Accepted: 02/26/2025] [Indexed: 03/27/2025]
Abstract
CRC remains a significant public health challenge due to its high prevalence and mortality rates. Emerging evidence highlights the critical role of the gut microbiota in both the pathogenesis of CRC and the efficacy of treatment strategies, including chemotherapy and immunotherapy. Dysbiosis, characterized by imbalances in microbial communities, has been implicated in CRC progression and therapeutic outcomes. This review examines the intricate relationship between gut microbiota composition and CRC, emphasizing the potential for microbial profiles to serve as biomarkers for early detection and prognosis. Various interventions, such as prebiotics, probiotics, postbiotics, fecal microbiota transplantation, and dietary modifications, aim to restore microbiota balance and shift dysbiosis toward eubiosis, thereby improving health outcomes. Additionally, the integration of microbial profiling into clinical practice could enhance diagnostic capabilities and personalize treatment strategies, advancing the field of oncology. The study of intratumoral microbiota offers new diagnostic and prognostic tools that, combined with artificial intelligence algorithms, could predict treatment responses and assess the risk of adverse effects. Given the growing understanding of the gut microbiome-cancer axis, developing microbiota-oriented strategies for CRC prevention and treatment holds promise for improving patient care and clinical outcomes.
Collapse
Affiliation(s)
- Imen Zalila-Kolsi
- Faculty of Medical and Health Sciences, Liwa College, Abu Dhabi P.O. Box 41009, United Arab Emirates; (H.A.O.); (H.M.)
| | - Dhoha Dhieb
- College of Pharmacy, QU Health, Qatar University, Doha P.O. Box 2713, Qatar;
| | - Hussam A. Osman
- Faculty of Medical and Health Sciences, Liwa College, Abu Dhabi P.O. Box 41009, United Arab Emirates; (H.A.O.); (H.M.)
| | - Hadjer Mekideche
- Faculty of Medical and Health Sciences, Liwa College, Abu Dhabi P.O. Box 41009, United Arab Emirates; (H.A.O.); (H.M.)
| |
Collapse
|
24
|
Luo S, Huang X, Chen S, Li J, Wu H, He Y, Zhou L, Liu B, Feng J. The Gut Microbiota of the Greater Horseshoe Bat Confers Rapidly Corresponding Immune Cells in Mice. Animals (Basel) 2025; 15:685. [PMID: 40075967 PMCID: PMC11899282 DOI: 10.3390/ani15050685] [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: 01/26/2025] [Revised: 02/19/2025] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Emerging infectious diseases threaten human and animal health, with most pathogens originating from wildlife. Bats are natural hosts for many infectious agents. Previous studies have demonstrated that changes in some specific genes in bats may contribute to resistance to viral infections, but they have mostly overlooked the immune function of the bat gut microbiota. AIMS In this study, we used fecal transplants to transfer the gut microbiota from the Greater Horseshoe Bat (Rhinolophus ferrumequinum) into mice treated with antibiotics. The gut microbiota changes in mice were detected using 16S rRNA high-throughput sequencing technology. Flow cytometry was used to detect changes in associated immune cells in the spleen and mesenteric lymph nodes of the mice. RESULTS The results showed that the gut microbiota of mice showed characteristics of some bat gut microbiota. The Greater Horseshoe Bat's gut microbiota changed some immune cells' composition in the spleen and mesenteric lymph nodes of mice and also conferred a faster and higher proportion of natural killer cell activation. CONCLUSION This result provides new evidence for the regulatory immune function of bat gut microbiota and contributes to a deeper insight into the unique immune system of bats.
Collapse
Affiliation(s)
- Shan Luo
- College of Animal Science and Technology, Jilin Agricultural Science and Technology University, Jilin 132109, China
| | - Xinlei Huang
- College of Life Science, Jilin Agricultural University, Changchun 130117, China
| | - Siyu Chen
- College of Animal Science and Technology, Jilin Agricultural Science and Technology University, Jilin 132109, China
| | - Junyi Li
- College of Veterinary Medicine, Jilin Agricultural University, Changchun 130117, China
| | - Hui Wu
- College of Life Science, Jilin Agricultural University, Changchun 130117, China
| | - Yuhua He
- College of Animal Science and Technology, Jilin Agricultural Science and Technology University, Jilin 132109, China
| | - Lei Zhou
- College of Animal Science and Technology, Jilin Agricultural Science and Technology University, Jilin 132109, China
| | - Boyu Liu
- College of Animal Science and Technology, Jilin Agricultural Science and Technology University, Jilin 132109, China
| | - Jiang Feng
- College of Life Science, Jilin Agricultural University, Changchun 130117, China
- Jilin Provincial Key Laboratory of Animal Resource Conservation and Utilization, Northeast Normal University, Changchun 130024, China
| |
Collapse
|
25
|
Dickey JR, Mercer NM, Kuijpers MCM, Props R, Jackrel SL. Biodiversity within phytoplankton-associated microbiomes regulates host physiology, host community ecology, and nutrient cycling. mSystems 2025; 10:e0146224. [PMID: 39873522 PMCID: PMC11834400 DOI: 10.1128/msystems.01462-24] [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] [Accepted: 01/03/2025] [Indexed: 01/30/2025] Open
Abstract
Biological diversity is declining across the tree of life, including among prokaryotes. With the increasing awareness of host-associated microbes as potential regulators of eukaryotic host physiology, behavior, and ecology, it is important to understand the implications of declining diversity within host microbiomes on host fitness, ecology, and ecosystem function. We used phytoplankton and their associated environmental microbiomes as model systems to test the independent and interactive effects of declining microbiome diversity with and without other stressors often caused by human activity-elevated temperature and altered nutrient availability. We found effects of low microbiome diversity on host physiology, phytoplankton community dynamics, and nutrient cycling. Low microbiome diversity caused greater host cellular stress, as indicated by elevated δ13C and δ15N. Microbiome diversity also significantly affected host cell morphological metrics, likely as a consequence of this effect on cell stress. Despite causing greater host cellular stress, the effects of low microbiome diversity on host community ecology included elevated phytoplankton community diversity and biomass. The diversity of these host-associated microbes also had cascading implications on ecosystem nutrient cycling, where lower microbiome diversity caused a depletion of total dissolved N and P in the environment. The magnitude of these effects, caused by microbiome diversity, was greatest among nutrient-depleted environments and at elevated temperatures. Our results emphasize the widespread implications of declining host-associated microbial diversity from host cellular physiology to ecosystem nutrient cycling. These demonstrated effects of declining microbiome diversity are likely to be amplified in ecosystems experiencing multiple stressors caused by anthropogenic activities. IMPORTANCE As evidence is emerging of the key roles that host-associated microbiomes often play in regulating the physiology, fitness, and ecology of their eukaryotic hosts, human activities are causing declines in biological diversity, including within the microbial world. Here, we use a multifactorial manipulative experiment to test the effects of declining diversity within host microbiomes both alone and in tandem with the effects of emerging global changes, including climate warming and shifts in nutrient bioavailability, which are inflicting increasing abiotic stress on host organisms. Using single-celled eukaryotic phytoplankton that harbor an external microbiome as a model system, we demonstrate that diversity within host-associated microbiomes impacts multiple tiers of biological organization, including host physiology, the host population and community ecology, and ecosystem nutrient cycling. Notably, these microbiome diversity-driven effects became magnified in abiotically stressful environments, suggesting that the importance of microbiome diversity may have increased over time during the Anthropocene.
Collapse
Affiliation(s)
- Jonathan R. Dickey
- Department of Ecology, Behavior and Evolution, University of California San Diego School of Biological Sciences, La Jolla, California, USA
| | - Nikki M. Mercer
- Department of Ecology, Behavior and Evolution, University of California San Diego School of Biological Sciences, La Jolla, California, USA
| | - Mirte C. M. Kuijpers
- Department of Ecology, Behavior and Evolution, University of California San Diego School of Biological Sciences, La Jolla, California, USA
| | | | - Sara L. Jackrel
- Department of Ecology, Behavior and Evolution, University of California San Diego School of Biological Sciences, La Jolla, California, USA
| |
Collapse
|
26
|
SHIMOKAWA H, SAKAKIBARA H, AMI Y, HIRANO R, KURIHARA S. The effect of culturing temperature on the growth of the most dominant bacterial species of human gut microbiota and harmful bacterial species. BIOSCIENCE OF MICROBIOTA, FOOD AND HEALTH 2025; 44:182-195. [PMID: 40171389 PMCID: PMC11957755 DOI: 10.12938/bmfh.2024-087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 01/22/2025] [Indexed: 04/03/2025]
Abstract
In recent years, the gut microbiota has attracted attention due to reported associations with various diseases and health conditions. Gut bacteria have been constantly cultured at 37°C, potentially limiting the understanding of the interaction between them and the host. However, the most dominant human gut microbial species have not been extensively cultured at temperatures other than 37°C. In this study, we analyzed the effects of various culturing temperatures on the growth of the 51 most dominant commensal species as well as 3 harmful bacteria, including Clostridium perfringens, a food poisoning bacterium, in the human intestine. The results showed that the growth of predominant gut microbes varied minimally at body temperatures conducive to human survival but that the growth of several bacteria involved in butyrate production in the intestinal lumen was repressed at temperatures other than 37°C. When cultured at 50°C, the growth of C. perfringens was less inhibited than that of other bacterial species. In addition, the growth of some gut bacteria was unaffected by a body temperature range that was not suitable for human survival.
Collapse
Affiliation(s)
- Hiromi SHIMOKAWA
- Faculty of Biology-Oriented Science and Technology, Kindai
University, Kinokawa, Wakayama 649-6493, Japan
| | - Hikaru SAKAKIBARA
- Faculty of Biology-Oriented Science and Technology, Kindai
University, Kinokawa, Wakayama 649-6493, Japan
| | - Yuta AMI
- Faculty of Biology-Oriented Science and Technology, Kindai
University, Kinokawa, Wakayama 649-6493, Japan
| | - Rika HIRANO
- Faculty of Biology-Oriented Science and Technology, Kindai
University, Kinokawa, Wakayama 649-6493, Japan
- Present address: Division of Animal Genetics, Laboratory
Animal Research Center, Institute of Medical Science, The University of Tokyo, 4-6-1
Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Shin KURIHARA
- Faculty of Biology-Oriented Science and Technology, Kindai
University, Kinokawa, Wakayama 649-6493, Japan
| |
Collapse
|
27
|
Vanhove MPM, Koblmüller S, Fernandes JMO, Hahn C, Plusquin M, Kmentová N. Cichlid fishes are promising underutilized models to investigate helminth-host-microbiome interactions. Front Immunol 2025; 16:1527184. [PMID: 40018030 PMCID: PMC11864961 DOI: 10.3389/fimmu.2025.1527184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 01/27/2025] [Indexed: 03/01/2025] Open
Abstract
The "Old Friends Hypothesis" suggests insufficient exposure to symbionts hinders immune development, contributing to increased immune-related diseases in the Global North. The microbiome is often the focus; helminths, potentially also offering health benefits, lack attention. Infection and effect of helminths are influenced and perhaps determined by micro-organisms. Mechanisms behind parasite-microbiome interactions are poorly understood, despite implications on host health. These interactions are typically studied for single helminth species in laboratory animal models, overlooking helminth diversity. Reviewing research on relationships between helminth and microbial diversity yielded 27 publications; most focused on human or other mammalian hosts, relying on natural exposure rather than experimental helminth inoculation. Only about half investigated host health outcomes. Remaining knowledge gaps warrant considering additional candidate model systems. Given the high helminthiasis burden and species diversity of helminths, we propose seeking models in the Global South, where a considerable proportion of research on diversity aspects of helminth-microbiome interactions took place. Low availability of genomic resources for helminths in the Global South, however, necessitates more integrative helminthological research efforts. Given substantial similarities in immune systems, several fishes are models for human health/disease. More effort could be done to establish this for cichlids, whose representatives in the African Great Lakes provide a well-delineated, closed natural system relevant to human health in view of fish-borne zoonoses and other water-borne parasites. A good baseline exists for these cichlids' genomics, parasitology, and microbiology. We suggest exploring African Great Lake cichlids as model hosts for interactions between microbial diversity, helminth diversity, and host health.
Collapse
Affiliation(s)
- Maarten P. M. Vanhove
- Research Group Zoology: Biodiversity and Toxicology, Centre for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
- International Union for Conservation of Nature (IUCN) Species Survival Commission (SSC) Parasite Specialist Group, Diepenbeek, Belgium
| | | | - Jorge M. O. Fernandes
- Renewable Marine Resources Department, Institut de Ciències del Mar, Spanish National Research Council, Barcelona, Spain
| | | | - Michelle Plusquin
- Research Group Environmental Biology, Center for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
| | - Nikol Kmentová
- Research Group Zoology: Biodiversity and Toxicology, Centre for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
- International Union for Conservation of Nature (IUCN) Species Survival Commission (SSC) Parasite Specialist Group, Diepenbeek, Belgium
- Freshwater Biology, Operational Directorate Natural Environment, Royal Belgian Institute of Natural Sciences, Brussels, Belgium
| |
Collapse
|
28
|
Verheijden RJ, van Eijs MJM, Paganelli FL, Viveen MC, Rogers MRC, Top J, May AM, van de Wijgert JHHM, Suijkerbuijk KPM. Gut microbiome and immune checkpoint inhibitor toxicity. Eur J Cancer 2025; 216:115221. [PMID: 39793444 DOI: 10.1016/j.ejca.2025.115221] [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: 12/13/2024] [Accepted: 01/02/2025] [Indexed: 01/13/2025]
Abstract
BACKGROUND Multiple studies have suggested that gut microbiome may influence immune checkpoint inhibitor (ICI) efficacy, but its association with immune-related adverse events (irAEs) is less well studied. In this prospective cohort study, we assessed whether gut microbiome composition at start, or changes during ICI, are associated with severe irAEs. METHODS Stool samples of cancer patients treated with anti-PD-1 ± anti-CTLA-4 were analyzed using 16S rRNA gene sequencing and metagenomic shotgun sequencing. Differences in alpha and beta diversity between patients with and without severe irAE were assessed, as well as differential relative abundance (RA) of taxa, MetaCyc pathways, and seven prespecified literature-based bacterial groups including pathobionts and Ruminococcaceae. FINDINGS We analyzed 497 samples of 195 patients before and soon after starting ICI, at severe irAE onset and after starting immunosuppression. Mean RA of the pathobionts group was significantly higher in patients who developed a severe irAE (8.2 %) compared to those who did not (4.8 %; odds ratio 1.40; 95 %CI 1.07-1.87) at baseline, and also early during ICI treatment and at severe irAE onset. A significantly stronger decrease in RA of Ruminococcaceae after starting ICI was observed in patients who developed a severe irAE compared to those who did not. RAs of Ruminococcaceae, the genus Ruminococcus, and the species R. bromii and R. callidus were significantly lower at severe irAE onset compared to other time points. INTERPRETATION Gut microbiome dysbiosis signaled by higher RA of pathobionts and decrease in RA of Ruminococcaceae may predispose to severe irAEs.
Collapse
Affiliation(s)
- Rik J Verheijden
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, PO box 85500, Utrecht 3584 CX, the Netherlands; Department of Epidemiology and Health Economics, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, PO box 85500, Utrecht 3584 CX, the Netherlands; Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, PO box 85500, Utrecht 3584 CX, the Netherlands
| | - Mick J M van Eijs
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, PO box 85500, Utrecht 3584 CX, the Netherlands; Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, PO box 85500, Utrecht 3584 CX, the Netherlands
| | - Fernanda L Paganelli
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, PO box 85500, Utrecht 3584 CX, the Netherlands; Winclove Probiotics, Amsterdam, 1033JS, the Netherlands
| | - Marco C Viveen
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, PO box 85500, Utrecht 3584 CX, the Netherlands
| | - Malbert R C Rogers
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, PO box 85500, Utrecht 3584 CX, the Netherlands
| | - Janetta Top
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, PO box 85500, Utrecht 3584 CX, the Netherlands
| | - Anne M May
- Department of Epidemiology and Health Economics, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, PO box 85500, Utrecht 3584 CX, the Netherlands
| | - Janneke H H M van de Wijgert
- Department of Epidemiology and Health Economics, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, PO box 85500, Utrecht 3584 CX, the Netherlands
| | - Karijn P M Suijkerbuijk
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, PO box 85500, Utrecht 3584 CX, the Netherlands.
| |
Collapse
|
29
|
Weitzman CL, Brown GP, Day K, Shilton CM, Gibb K, Christian K. Protection against anuran lungworm infection may be mediated by innate defenses rather than their microbiome. Int J Parasitol 2025:S0020-7519(25)00021-9. [PMID: 39909191 DOI: 10.1016/j.ijpara.2025.01.010] [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: 03/18/2024] [Revised: 12/22/2024] [Accepted: 01/31/2025] [Indexed: 02/07/2025]
Abstract
Host-associated microbiomes provide protection against disease in diverse systems, through both direct and indirect interactions with invaders, although these interactions are less understood in the context of non-gut helminth infections in wildlife. Here, we used a widespread, invasive host-parasite system to better understand helminth-amphibian-microbiome dynamics. We focus on cane toads and their lungworm parasites, which invade the host through the skin, to study the interactions between lungworm infection abundance and skin and gut (colon) bacterial microbiomes. Through two experiments, first reducing skin bacterial loads, and second reducing bacterial diversity, we found no evidence of protection by skin bacteria against infection. We also did not find divergent gut communities dependent on lungworm infection, signifying little to no immune modulation from infection causing changes to gut communities, at least in the first month after initial parasite exposure. In light of previous work in the system, these results underscore the contribution of toads' innate susceptibility (including possible protection provided by skin secretions) rather than skin microbes in determining the chance of infection by these macroparasites.
Collapse
Affiliation(s)
- Chava L Weitzman
- Research Institute for the Environment and Livelihoods, Charles Darwin University, Brinkin, NT 0909, Australia.
| | - Gregory P Brown
- Department of Biological Sciences, Macquarie University, New South Wales 2109, Australia
| | - Kimberley Day
- Research Institute for the Environment and Livelihoods, Charles Darwin University, Brinkin, NT 0909, Australia
| | - Catherine M Shilton
- Berrimah Veterinary Laboratories, Northern Territory Department of Primary Industries and Fisheries, Berrimah, NT 0828, Australia
| | - Karen Gibb
- Research Institute for the Environment and Livelihoods, Charles Darwin University, Brinkin, NT 0909, Australia
| | - Keith Christian
- Research Institute for the Environment and Livelihoods, Charles Darwin University, Brinkin, NT 0909, Australia
| |
Collapse
|
30
|
Yuan Z, Hong J, Khan J, Lu J, Sanogo B, Wu Z, Sun X, Lin D. Stochastic processes govern gut bacterial community assembly in a Schistosoma mansoni-transmitting snail, Biomphalaria straminea. PLoS Negl Trop Dis 2025; 19:e0012828. [PMID: 39908240 PMCID: PMC11798439 DOI: 10.1371/journal.pntd.0012828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 01/07/2025] [Indexed: 02/07/2025] Open
Abstract
BACKGROUND Studies have revealed extensive taxonomic classifications and patterns of gut microbial diversity in snails, with limited focus on community assembly processes. To better understand the balance between stochastic and deterministic processes in the snail gut microbial assembly and their associations with snail fitness, we used the freshwater snail Biomphalaria straminea as a model and analyzed the gut bacterial communities from 118 samples via high-throughput sequencing of the 16S rRNA gene. RESULTS This study reveals that Proteobacteria and Bacteroidota dominate the gut microbiota of B. straminea. Snails from different laboratory habitats exhibit similar gut bacterial diversity but significantly different community structures. The assembly of gut bacterial communities in both laboratory and wild samples is predominantly influenced by stochastic processes rather than deterministic processes, as evidenced by the neutral community model (NCM). Furthermore, during the snail invasion and adaptation to a new environment, stochastic processes are more crucial than deterministic ones in shaping the snail gut microbiota. This indicates that the interplay between stochastic and deterministic processes in the snail gut microbial assembly is associated with host fitness during snail adaptation to a new environment. Based on the null model analysis, we also found that stochastic processes (based on dispersal limitation, homogenizing dispersal, and undominated processes) play a larger role than deterministic (based on homogeneous selection and variable selection) in driving the snail gut bacterial community assembly. Furthermore, the significant difference in the proportions of dispersal limitation and undominated processes is linked to both adaptive and non-adaptive snails. CONCLUSIONS This study demonstrates that stochastic processes govern the assembly of the gut microbiota in B. straminea. Furthermore, snail adaptation is associated with the interplay between stochastic and deterministic processes in gut microbial composition. This study provides a better understanding of the dynamic patterns of the gut microbial community in freshwater gastropods and may contribute to the development of strategies for controlling intermediate hosts and schistosomiasis.
Collapse
Affiliation(s)
- Zhanhong Yuan
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Tropical Disease Control, Ministry of Education, Guangzhou, China
- Provincial Engineering Technology Research Center for Diseases-vectors Control, Chinese Atomic Energy Agency Center of Excellence on Nuclear Technology Applications for Insect Control, Guangzhou, China
| | - Jinni Hong
- Department of Traditional Chinese Medicine, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jehangir Khan
- Department of Zoology, Abdul Wali Khan University, Mardan, Pakistan
- Hainan General Hospital, Hainan Medical University, Haikou, China
| | - Jinghuang Lu
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Tropical Disease Control, Ministry of Education, Guangzhou, China
- Provincial Engineering Technology Research Center for Diseases-vectors Control, Chinese Atomic Energy Agency Center of Excellence on Nuclear Technology Applications for Insect Control, Guangzhou, China
| | - Benjamin Sanogo
- Laboratory of Parasitology, Institut National de Recherche en Sante Publique, Bamako, Mali
| | - Zhongdao Wu
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Tropical Disease Control, Ministry of Education, Guangzhou, China
- Provincial Engineering Technology Research Center for Diseases-vectors Control, Chinese Atomic Energy Agency Center of Excellence on Nuclear Technology Applications for Insect Control, Guangzhou, China
| | - Xi Sun
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Tropical Disease Control, Ministry of Education, Guangzhou, China
| | - Datao Lin
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Tropical Disease Control, Ministry of Education, Guangzhou, China
- Provincial Engineering Technology Research Center for Diseases-vectors Control, Chinese Atomic Energy Agency Center of Excellence on Nuclear Technology Applications for Insect Control, Guangzhou, China
| |
Collapse
|
31
|
Buday T, Brozmanova M, Jakusova J, Owesie AA, Ertl LS, Mokra D, Hanusrichterova J, Burjanivova T, Biringerova Z, Plevkova J. Impact of microbial diversity on inflammatory cytokines and respiratory pattern measured in whole-body plethysmography in guinea pig models. Respir Physiol Neurobiol 2025; 332:104384. [PMID: 39647679 DOI: 10.1016/j.resp.2024.104384] [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: 08/27/2024] [Revised: 12/05/2024] [Accepted: 12/05/2024] [Indexed: 12/10/2024]
Abstract
OBJECTIVE This study investigates the breathing patterns and immune status of guinea pigs raised under specific pathogen-free (SPF) conditions compared to conventionally bred (CON). METHODS Breathing pattern parameters were assessed using whole-body plethysmography (WBP) during quiet breathing and saline nebulisation. Blood and bronchoalveolar lavage fluid (BALF) were analysed for white blood cell, neutrophil and eosinophil counts, and cytokine levels (TNF-α, IL-1β, IL-4). RESULTS SPF guinea pigs exhibited higher tidal volume, expired volume, minute volume, and airflow parameters than CON guinea pigs. The immune analysis revealed lower white blood cell counts and IL-4 levels in SPF guinea pigs. These findings indicate that SPF guinea pigs have different respiratory and immune responses than CON guinea pigs. CONCLUSION The study highlights that the maturation processes affecting breathing pattern parameters in SPF guinea pigs differ significantly from those in CON guinea pigs. This suggests potential limitations of SPF animals in respiratory physiology research due to their different immune and respiratory responses.
Collapse
Affiliation(s)
- Tomas Buday
- Department of Pathophysiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Slovakia
| | - Mariana Brozmanova
- Department of Pathophysiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Slovakia
| | - Janka Jakusova
- Department of Pathophysiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Slovakia
| | - Abdullah Al Owesie
- Department of Pathophysiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Slovakia
| | - Laura Sophie Ertl
- Department of Pathophysiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Slovakia
| | - Daniela Mokra
- Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Slovakia
| | - Juliana Hanusrichterova
- Biomedical Centre Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Slovakia
| | - Tatiana Burjanivova
- Department of Molecular Biology and Genetics, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Slovakia
| | - Zuzana Biringerova
- Centre for Medical Education Support, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Slovakia
| | - Jana Plevkova
- Department of Pathophysiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Slovakia; Centre for Medical Education Support, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Slovakia.
| |
Collapse
|
32
|
McDaniel Mims B, Furr KL, Enriquez J, Grisham MB. Improving bench-to-bedside translation for acute graft-versus-host disease models. Dis Model Mech 2025; 18:DMM052084. [PMID: 40019007 PMCID: PMC11892683 DOI: 10.1242/dmm.052084] [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] [Indexed: 03/01/2025] Open
Abstract
The transplantation of allogeneic hematopoietic stem cells is a potentially curative treatment for hematological malignancies, inherited blood disorders and immune deficiencies. Unfortunately, 30-50% of patients receiving allogeneic hematopoietic stem cells will develop a potentially life-threatening inflammatory disease called acute graft-versus-host disease (aGVHD). In patients with aGVHD, graft-associated T cells, which typically target the skin, intestinal tract and liver, can also damage the lungs and lymphoid tissue. Damage to lymphoid tissue creates prolonged immunodeficiency that markedly increases the risk of infections and bleeding, resulting in considerable morbidity and mortality. Although mouse models of aGVHD have been instrumental to our understanding of this condition's pathogenesis, translation of preclinical data into new and more effective treatments for human disease has been limited for reasons that remain to be fully understood. However, evidence suggests that factors associated with mouse models of aGVHD likely contribute to these unsatisfactory results. In this Review, we identify and discuss the specific factors inherent to mouse models of aGVHD that may limit the translation of preclinical data to patient treatment, and suggest how to improve the translatability of these models.
Collapse
Affiliation(s)
- Brianyell McDaniel Mims
- Department of Oral Health Sciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Kathryn L. Furr
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX 79423, USA
| | - Josue Enriquez
- Department of Microbiology and Immunology, University of Gothenburg, Gothenburg 405 30, Sweden
| | - Matthew B. Grisham
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX 79423, USA
| |
Collapse
|
33
|
White PS, Broe TY, Kuijpers MCM, Dickey JR, Jackrel SL. Host identity drives the assembly of phytoplankton microbiomes across a continental-scale environmental gradient. THE ISME JOURNAL 2025; 19:wraf083. [PMID: 40302044 PMCID: PMC12118458 DOI: 10.1093/ismejo/wraf083] [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] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 04/03/2025] [Accepted: 04/28/2025] [Indexed: 05/01/2025]
Abstract
Host-associated microbiomes often promote host health, yet the key drivers of microbiome assembly and its consequences for host fitness remain unclear. We aimed to determine the relative roles of host identity versus the environment in driving host-microbiome assembly and the consequences of this variation in assembly for host fitness, which may help predict the resilience of host-associated microbiomes and host health amidst fluctuating environmental conditions. Here, we tracked microbiome assembly in association with initially axenic phytoplankton when incubated in seawater originating from four nearshore locations along a continental-scale environmental gradient of North America. Microbiome assembly was highly deterministic. Unexpectedly, host species identity was the overwhelming driver of microbiome community assembly despite continental-scale variation in the environment. Although secondary to host identity, the environment was a significant driver of microbiome assembly for each species evaluated, which, in turn, conferred cascading effects on host fitness as shown by thermal tolerance growth assays. We also found that host-specific microbiomes had host-specific fitness effects, particularly under thermally stressful conditions. Overall, our results advance our understanding of microbiome assembly by empirically demonstrating that although variation among host microbiomes imparted by the local environment has significant implications for host health, the host species is the overwhelming driver of microbiome assembly regardless of wide-scale variation in the environment.
Collapse
Affiliation(s)
- Patricia Signe White
- Department of Ecology, Behavior, and Evolution, University of California San Diego, La Jolla, CA 92093-0116, United States
- Department of Integrative Biology, University of California Berkeley, Berkeley, CA 94720-2284, United States
| | - Taryn Y Broe
- Department of Ecology, Behavior, and Evolution, University of California San Diego, La Jolla, CA 92093-0116, United States
| | - Mirte C M Kuijpers
- Department of Ecology, Behavior, and Evolution, University of California San Diego, La Jolla, CA 92093-0116, United States
| | - Jonathan R Dickey
- Department of Ecology, Behavior, and Evolution, University of California San Diego, La Jolla, CA 92093-0116, United States
| | - Sara L Jackrel
- Department of Ecology, Behavior, and Evolution, University of California San Diego, La Jolla, CA 92093-0116, United States
| |
Collapse
|
34
|
Bruno P, Schüler T, Rosshart SP. Born to be wild: utilizing natural microbiota for reliable biomedical research. Trends Immunol 2025; 46:17-28. [PMID: 39690004 DOI: 10.1016/j.it.2024.11.013] [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: 10/11/2024] [Revised: 11/11/2024] [Accepted: 11/19/2024] [Indexed: 12/19/2024]
Abstract
Laboratory mice housed under specific pathogen-free (SPF) conditions are the standard model in biomedical research. However, experiments with a particular inbred mouse strain performed in different laboratories often yield inconsistent or conflicting data due to housing-specific variations in the composition and diversity of SPF microbiota. These variations affect immune and nonimmune cell functions, leading to systemic physiological changes. Consequently, microbiota-dependent inconsistencies have raised general doubts regarding the suitability of mice as model organisms. Since stability positively correlates with biological diversity, we postulate that increasing species diversity can improve microbiota stability and mouse physiology, enhancing robustness, reproducibility, and experimental validity. Similar to the generation of inbred mouse strains in the last century, we suggest a worldwide initiative to define a transplantable 'wild' microbiota that stably colonizes mice irrespective of housing conditions.
Collapse
Affiliation(s)
- Philipp Bruno
- Department of Microbiome Research, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany.
| | - Thomas Schüler
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto von Guericke University Magdeburg, Magdeburg, Germany.
| | - Stephan P Rosshart
- Department of Microbiome Research, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany; Department of Medicine II, Faculty of Medicine, Medical Center - University of Freiburg, Freiburg, Germany.
| |
Collapse
|
35
|
Mann J, Runge S, Schell C, Gräwe K, Thoulass G, Lao J, Ammann S, Grün S, König C, Berger SA, Hild B, Aichele P, Rosshart SP, Ehl S. The Microbiome Modifies Manifestations of Hemophagocytic Lymphohistiocytosis in Perforin-Deficient Mice. Eur J Immunol 2025; 55:e202451061. [PMID: 39548906 PMCID: PMC11739664 DOI: 10.1002/eji.202451061] [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: 02/09/2024] [Revised: 10/30/2024] [Accepted: 11/01/2024] [Indexed: 11/18/2024]
Abstract
Primary hemophagocytic lymphohistiocytosis (HLH) is a life-threatening hyperinflammatory syndrome caused by inborn errors of cytotoxicity. Patients with biallelic PRF1 null mutations (encoding perforin) usually develop excessive immune cell activation, hypercytokinemia, and life-threatening immunopathology in the first 6 months of life, often without an apparent infectious trigger. In contrast, perforin-deficient (PKO) mice only develop HLH after systemic infection with lymphocytic choriomeningitis virus (LCMV). We hypothesized that restricted microbe-immune cell interactions due to specific pathogen-free (SPF) housing might explain the need for this specific viral trigger in PKO mice. To investigate the influence of a "wild" microbiome in PKO mice, we fostered PKO newborns with Wildling microbiota ('PKO-Wildlings') and monitored them for signs of HLH. PKO-Wildlings survived long-term without spontaneous disease. Also, systemic infection with vaccinia virus did not reach the threshold of immune activation required to trigger HLH in PKO-Wildlings. Interestingly, after infection with LCMV, PKO-Wildlings developed an altered HLH pattern. This included lower IFN-γ serum levels along with improved IFN-γ-driven anemia, but more elevated levels of IL-17 and increased liver inflammation compared with PKO-SPF mice. Thus, wild microbiota alone is not sufficient to trigger HLH in PKO mice, but host-microbe interactions shape inflammatory cytokine patterns, thereby influencing manifestations of HLH immunopathology.
Collapse
Affiliation(s)
- Jasmin Mann
- Institute for ImmunodeficiencyCenter for Chronic Immunodeficiency (CCI), Medical Center‐ University of FreiburgFaculty of MedicineUniversity of FreiburgFreiburgGermany
- Faculty of BiologyUniversity of FreiburgFreiburgGermany
| | - Solveig Runge
- Faculty of BiologyUniversity of FreiburgFreiburgGermany
- Department of Medicine II, Medical Center‐ University of Freiburg, Faculty of MedicineUniversity of FreiburgFreiburgGermany
- Department of Microbiome Research, University Hospital ErlangenFriedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU)ErlangenGermany
| | - Christoph Schell
- Institute for Surgical Pathology, Medical Center‐ University of Freiburg, Faculty of MedicineUniversity of FreiburgFreiburgGermany
| | - Katja Gräwe
- Institute for Surgical Pathology, Medical Center‐ University of Freiburg, Faculty of MedicineUniversity of FreiburgFreiburgGermany
| | - Gudrun Thoulass
- Institute for ImmunodeficiencyCenter for Chronic Immunodeficiency (CCI), Medical Center‐ University of FreiburgFaculty of MedicineUniversity of FreiburgFreiburgGermany
- Faculty of BiologyUniversity of FreiburgFreiburgGermany
| | - Jessica Lao
- Institute for ImmunodeficiencyCenter for Chronic Immunodeficiency (CCI), Medical Center‐ University of FreiburgFaculty of MedicineUniversity of FreiburgFreiburgGermany
- Faculty of BiologyUniversity of FreiburgFreiburgGermany
| | - Sandra Ammann
- Institute for ImmunodeficiencyCenter for Chronic Immunodeficiency (CCI), Medical Center‐ University of FreiburgFaculty of MedicineUniversity of FreiburgFreiburgGermany
| | - Sarah Grün
- Institute for ImmunodeficiencyCenter for Chronic Immunodeficiency (CCI), Medical Center‐ University of FreiburgFaculty of MedicineUniversity of FreiburgFreiburgGermany
- Faculty of BiologyUniversity of FreiburgFreiburgGermany
| | - Christoph König
- Institute for ImmunodeficiencyCenter for Chronic Immunodeficiency (CCI), Medical Center‐ University of FreiburgFaculty of MedicineUniversity of FreiburgFreiburgGermany
- Faculty of BiologyUniversity of FreiburgFreiburgGermany
| | - Sarah A. Berger
- Institute for ImmunodeficiencyCenter for Chronic Immunodeficiency (CCI), Medical Center‐ University of FreiburgFaculty of MedicineUniversity of FreiburgFreiburgGermany
- Faculty of BiologyUniversity of FreiburgFreiburgGermany
| | - Benedikt Hild
- Department of Gastroenterology, Hepatology and Transplantation MedicineMedical Faculty University of Duisburg‐EssenEssenGermany
| | - Peter Aichele
- Institute for ImmunodeficiencyCenter for Chronic Immunodeficiency (CCI), Medical Center‐ University of FreiburgFaculty of MedicineUniversity of FreiburgFreiburgGermany
| | - Stephan P. Rosshart
- Department of Medicine II, Medical Center‐ University of Freiburg, Faculty of MedicineUniversity of FreiburgFreiburgGermany
- Department of Microbiome Research, University Hospital ErlangenFriedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU)ErlangenGermany
| | - Stephan Ehl
- Institute for ImmunodeficiencyCenter for Chronic Immunodeficiency (CCI), Medical Center‐ University of FreiburgFaculty of MedicineUniversity of FreiburgFreiburgGermany
| |
Collapse
|
36
|
Dobson GP, Morris JL, Letson HL. Traumatic brain injury: Symptoms to systems in the 21st century. Brain Res 2024; 1845:149271. [PMID: 39395646 DOI: 10.1016/j.brainres.2024.149271] [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: 10/01/2024] [Accepted: 10/07/2024] [Indexed: 10/14/2024]
Abstract
Severe traumatic brain injury (TBI) is a devastating injury with a mortality of ∼ 25-30 %. Despite decades of high-quality research, no drug therapy has reduced mortality. Why is this so? We argue two contributing factors for the lack of effective drug therapies include the use of specific-pathogen free (SPF) animals for translational research and the flawed practice of single-nodal targeting for drug design. A revolution is required to better understand how the whole body responds to TBI, identify new markers of its progression, and discover new system-acting drugs to treat it. In this review, we present a brief history of TBI, discuss its system's pathophysiology and propose a new research strategy for the 21st century. TBI progression develops from injury signals radiating from the primary impact, which can cause local ischemia, hemorrhage, excitotoxicity, cellular depolarization, immune dysfunction, sympathetic hyperactivity, blood-brain barrier breach, coagulopathy and whole-body dysfunction. Metabolic reprograming of immune cells drives neuroinflammation and secondary injury processes. We propose if sympathetic hyperactivity and immune cell activation can be corrected early, cardiovascular function and endothelial-glycocalyx-mitochondrial coupling can be restored, and secondary injury minimized with improved patient outcomes. The therapeutic goal is to switch the injury phenotype to a healing phenotype by restoring homeostasis and maintaining sufficient tissue O2 delivery. We have been developing a small-volume fluid therapy comprising adenosine, lidocaine and magnesium (ALM) to treat TBI and have shown that it blunts the CNS-stress response, supports cardiovascular function and reduces secondary injury. Future research will investigate its suitability for human translation.
Collapse
Affiliation(s)
- Geoffrey P Dobson
- Heart, Sepsis and Trauma Research Laboratory, College of Medicine and Dentistry, James Cook University, Queensland 4811, Australia.
| | - Jodie L Morris
- Heart, Sepsis and Trauma Research Laboratory, College of Medicine and Dentistry, James Cook University, Queensland 4811, Australia.
| | - Hayley L Letson
- Heart, Sepsis and Trauma Research Laboratory, College of Medicine and Dentistry, James Cook University, Queensland 4811, Australia.
| |
Collapse
|
37
|
Benga L, Rehm A, Gougoula C, Westhoff P, Wachtmeister T, Benten WPM, Engelhardt E, Weber APM, Köhrer K, Sager M, Janssen S. The host genotype actively shapes its microbiome across generations in laboratory mice. MICROBIOME 2024; 12:256. [PMID: 39639355 PMCID: PMC11619136 DOI: 10.1186/s40168-024-01954-2] [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: 12/01/2023] [Accepted: 10/18/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND The microbiome greatly affects health and wellbeing. Evolutionarily, it is doubtful that a host would rely on chance alone to pass on microbial colonization to its offspring. However, the literature currently offers only limited evidence regarding two alternative hypotheses: active microbial shaping by host genetic factors or transmission of a microbial maternal legacy. RESULTS To further dissect the influence of host genetics and maternal inheritance, we collected two-cell stage embryos from two representative wild types, C57BL6/J and BALB/c, and transferred a mixture of both genotype embryos into hybrid recipient mice to be inoculated by an identical microbiome at birth. CONCLUSIONS Observing the offspring for six generations unequivocally emphasizes the impact of host genetic factors over maternal legacy in constant environments, akin to murine laboratory experiments. Interestingly, maternal legacy solely controlled the microbiome in the first offspring generation. However, current evidence supporting maternal legacy has not extended beyond this initial generation, resolving the aforementioned debate. Video Abstract.
Collapse
Affiliation(s)
- Laurentiu Benga
- Central Unit for Animal Research and Animal Welfare Affairs, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
| | - Anna Rehm
- Algorithmic Bioinformatics, Justus Liebig University Giessen, Giessen, Germany
| | - Christina Gougoula
- Central Unit for Animal Research and Animal Welfare Affairs, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Philipp Westhoff
- Cluster of Excellence on Plant Science, Institute of Plant Biochemistry, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Thorsten Wachtmeister
- Genomics and Transcriptomics Laboratory, Biological and Medical Research Center, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - W Peter M Benten
- Central Unit for Animal Research and Animal Welfare Affairs, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Eva Engelhardt
- Central Unit for Animal Research and Animal Welfare Affairs, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Andreas P M Weber
- Cluster of Excellence on Plant Science, Institute of Plant Biochemistry, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Karl Köhrer
- Genomics and Transcriptomics Laboratory, Biological and Medical Research Center, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Martin Sager
- Central Unit for Animal Research and Animal Welfare Affairs, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Stefan Janssen
- Algorithmic Bioinformatics, Justus Liebig University Giessen, Giessen, Germany.
| |
Collapse
|
38
|
Islam MZ, Jozipovic D, Lopez PA, Krych L, Correia BSB, Bertram HC, Hansen AK, Hansen CHF. Wild-Mouse-Derived Gut Microbiome Transplantation in Laboratory Mice Partly Alleviates House-Dust-Mite-Induced Allergic Airway Inflammation. Microorganisms 2024; 12:2499. [PMID: 39770703 PMCID: PMC11728220 DOI: 10.3390/microorganisms12122499] [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: 11/20/2024] [Revised: 11/26/2024] [Accepted: 12/03/2024] [Indexed: 01/16/2025] Open
Abstract
Laboratory mice are instrumental for preclinical research but there are serious concerns that the use of a clean standardized environment for specific-pathogen-free (SPF) mice results in poor bench-to-bedside translation due to their immature immune system. The aim of the present study was to test the importance of the gut microbiota in wild vs. SPF mice for evaluating host immune responses in a house-dust-mite-induced allergic airway inflammation model without the influence of pathogens. The wild mouse microbiome reduced histopathological changes and TNF-α in the lungs and serum when transplanted to microbiota-depleted mice compared to mice transplanted with the microbiome from SPF mice. Moreover, the colonic gene expression of Gata3 was significantly lower in the wild microbiome-associated mice, whereas Muc1 was more highly expressed in both the ileum and colon. Intestinal microbiome and metabolomic analyses revealed distinct profiles associated with the wild-derived microbiome. The wild-mouse microbiome thus partly reduced sensitivity to house-dust-mite-induced allergic airway inflammation compared to the SPF mouse microbiome, and preclinical studies using this model should consider using both 'dirty' rewilded and SPF mice for testing new therapeutic compounds due to the significant effects of their respective microbiomes and derived metabolites on host immune responses.
Collapse
Affiliation(s)
- Md Zohorul Islam
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 1870 Frederiksberg, Denmark (A.K.H.)
- Section on Pathophysiology and Molecular Pharmacology, Joslin Diabetes Center, Boston, MA 02215, USA
- Department of Microbiology, Harvard Medical School, Boston, MA 02115, USA
- CSIRO Health & Biosecurity, Australian Centre for Disease Preparedness, Geelong, VIC 3220, Australia
| | - Danica Jozipovic
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 1870 Frederiksberg, Denmark (A.K.H.)
| | - Pablo Atienza Lopez
- Department of Food Science, Faculty of Science, University of Copenhagen, 1958 Frederiksberg, Denmark
| | - Lukasz Krych
- Department of Food Science, Faculty of Science, University of Copenhagen, 1958 Frederiksberg, Denmark
| | | | | | - Axel Kornerup Hansen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 1870 Frederiksberg, Denmark (A.K.H.)
| | - Camilla Hartmann Friis Hansen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 1870 Frederiksberg, Denmark (A.K.H.)
| |
Collapse
|
39
|
Drude N, Nagel-Riedasch S, Rosshart SP, Diefenbach A, Jordan S. A facility for laboratory mice with a natural microbiome at Charité - Universitätsmedizin Berlin. Lab Anim (NY) 2024; 53:351-354. [PMID: 39533119 PMCID: PMC11599040 DOI: 10.1038/s41684-024-01474-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Affiliation(s)
- Natascha Drude
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, BIH QUEST Center for Responsible Research, Berlin, Germany
| | - Stefan Nagel-Riedasch
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Forschungseinrichtungen für Experimentelle Medizin/FEM, Berlin, Germany
| | - Stephan P Rosshart
- Department of Microbiome Research, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Department of Medicine II, Medical Center - University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Andreas Diefenbach
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Microbiology, Infectious Diseases and Immunology, Berlin, Germany
| | - Stefan Jordan
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Microbiology, Infectious Diseases and Immunology, Berlin, Germany.
- Department of Infectious Diseases, Medical Microbiology and Hygiene, Heidelberg University, Heidelberg, Germany.
| |
Collapse
|
40
|
Zhang J, Tan S, Lyu B, Yu M, Lan Y, Tang R, Fan Z, Guo P, Shi L. Differences in Gut Microbial Composition and Characteristics Among Three Populations of the Bamboo Pitviper ( Viridovipera stejnegeri). Ecol Evol 2024; 14:e70742. [PMID: 39691431 PMCID: PMC11651729 DOI: 10.1002/ece3.70742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 09/13/2024] [Accepted: 12/02/2024] [Indexed: 12/19/2024] Open
Abstract
The gut microbiota contributes to host health by facilitating nutrient uptake, digestion, energy metabolism, intestinal development, vitamin synthesis, and immunomodulation, and plays an important role in the growth and reproduction of the animal itself. Considering the paucity of research on the gut microbiota of wild snakes, this study focused on bamboo pitviper (Viridovipera stejnegeri) populations from Anhui, Guizhou, and Hunan, with multiple fecal samples collected from each population (six, five, and three, respectively). Total microbial DNA was extracted from the fecal samples using metagenomic next-generation sequencing and differences in gut microbial composition, abundance, and carbohydrate-active enzymes (CAZymes) were analyzed and compared among the three populations. Results showed no significant variance in the α-diversity of the gut microbes across the three populations, while principal coordinate analysis revealed significant differences in gut microbe composition. The four most abundant phyla in the gut microbiota of V. stejnegeri were Pseudomonadota, Bacteroidota, Actinomycetota, and Bacillota, while the four most abundant genera were Salmonella, Citrobacter, Bacteroides, and Yokenella. Linear discriminant analysis effect size demonstrated notable differences in gut microbial abundance among the three populations. Marked differences in CAZyme abundance were also observed across the microbial communities. Future studies should incorporate diverse ecological factors to evaluate their influence on the composition and function of gut microbiota.
Collapse
Affiliation(s)
- Jiaqi Zhang
- Xinjiang Key Laboratory for Ecological Adaptation and Evolution of Extreme Environment Biology, College of Life SciencesXinjiang Agricultural UniversityUrumqiChina
- Faculty of Agriculture, Forestry and Food EngineeringYibin UniversityYibinChina
| | - Songwen Tan
- Faculty of Agriculture, Forestry and Food EngineeringYibin UniversityYibinChina
| | - Bing Lyu
- Faculty of Agriculture, Forestry and Food EngineeringYibin UniversityYibinChina
| | - Min Yu
- Faculty of Agriculture, Forestry and Food EngineeringYibin UniversityYibinChina
| | - Yue Lan
- Key Laboratory of Bioresources and Ecoenvironment (Ministry of Education), College of Life SciencesSichuan UniversityChengduChina
| | - Ruixiang Tang
- Key Laboratory of Bioresources and Ecoenvironment (Ministry of Education), College of Life SciencesSichuan UniversityChengduChina
| | - Zhenxin Fan
- Key Laboratory of Bioresources and Ecoenvironment (Ministry of Education), College of Life SciencesSichuan UniversityChengduChina
| | - Peng Guo
- Faculty of Agriculture, Forestry and Food EngineeringYibin UniversityYibinChina
| | - Lei Shi
- Xinjiang Key Laboratory for Ecological Adaptation and Evolution of Extreme Environment Biology, College of Life SciencesXinjiang Agricultural UniversityUrumqiChina
| |
Collapse
|
41
|
Sun X, Shukla M, Wang W, Li S. Unlocking gut-liver-brain axis communication metabolites: energy metabolism, immunity and barriers. NPJ Biofilms Microbiomes 2024; 10:136. [PMID: 39587086 PMCID: PMC11589602 DOI: 10.1038/s41522-024-00610-9] [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: 08/07/2024] [Accepted: 11/14/2024] [Indexed: 11/27/2024] Open
Abstract
The interaction between the gut-microbiota-derived metabolites and brain has long been recognized in both health and disease. The liver, as the primary metabolic organ for nutrients in animals or humans, plays an indispensable role in signal transduction. Therefore, in recent years, Researcher have proposed the Gut-Liver-Brain Axis (GLBA) as a supplement to the Gut-Brain Axis. The GLBA plays a crucial role in numerous physiological and pathological mechanisms through a complex interplay of signaling pathways. However, gaps remain in our knowledge regarding the developmental and functional influences of the GLBA communication pathway. The gut microbial metabolites serve as communication agents between these three distant organs, functioning prominently within the GLBA. In this review, we provide a comprehensive overview of the current understanding of the GLBA, focusing on signaling molecules role in animal and human health and disease. In this review paper elucidate its mechanisms of communication, explore its implications for immune, and energy metabolism in animal and human, and highlight future research directions. Understanding the intricate communication pathways of the GLBA holds promise for creating innovative treatment approaches for a wide range of immune and metabolic conditions.
Collapse
Affiliation(s)
- Xiaoge Sun
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, P. R. China
- Department of Neurosurgery, College of Medicine, The Pennsylvania State University, Hershey, PA, 17033, USA
| | - Manish Shukla
- Department of Neurosurgery, College of Medicine, The Pennsylvania State University, Hershey, PA, 17033, USA
| | - Wei Wang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, P. R. China.
| | - Shengli Li
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, P. R. China.
| |
Collapse
|
42
|
Kauer L, Imholt C, Jacob J, Berens C, Kühn R. Seasonal shifts and land-use impact: unveiling the gut microbiomes of bank voles (Myodes glareolus) and common voles (Microtus arvalis). FEMS Microbiol Ecol 2024; 100:fiae159. [PMID: 39611357 DOI: 10.1093/femsec/fiae159] [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: 06/26/2024] [Revised: 10/28/2024] [Accepted: 11/27/2024] [Indexed: 11/30/2024] Open
Abstract
Gut microbial diversity influences the health and vitality of the host, yet it is itself affected by internal and external factors, including land-use. The impact of land-use practices on wild rodents' gut microbiomes remains understudied, despite their abundance and potential as reservoirs for zoonotic pathogens. We examined the bacterial and fungal gut microbiomes of bank voles (Myodes glareolus) and common voles (Microtus arvalis) across grassland and forest habitats with varying land-use intensities and types. We collected rodents seasonally and used 16S rRNA and ITS amplicon sequencing for microbe identification. We found significant differences in alpha and beta diversities between the species, with M. arvalis exhibiting higher diversity. Seasonality emerged as a prominent factor influencing microbial diversity, with significant variations between sampling months. While land-use affects the gut microbiome, its impact is subordinate to seasonal variations. Differential abundance analysis underscores the dynamic nature of microbial composition, with seasonal changes playing a predominant role. Overall, our findings highlight the significant influence of seasonality on gut microbiome diversity and composition in wild rodents, reflecting dietary shifts associated with seasonal changes. Understanding the interplay between environmental factors and microbial communities in wild rodents enahnces our knowledge of ecosystem health and resilience, warranting further investigation.
Collapse
Affiliation(s)
- Lea Kauer
- Molecular Zoology, Department of Zoology, TUM School of Life Sciences, Technical University of Munich, 85354 Freising, Germany
| | - Christian Imholt
- Julius Kühn-Institute, Federal Research Centre for Cultivated Plants, Institute for Epidemiology and Pathogen Diagnostics, Rodent Research, 48161 Münster, Germany
| | - Jens Jacob
- Julius Kühn-Institute, Federal Research Centre for Cultivated Plants, Institute for Epidemiology and Pathogen Diagnostics, Rodent Research, 48161 Münster, Germany
| | - Christian Berens
- Friedrich-Loeffler-Institut, Institute of Molecular Pathogenesis, 07743 Jena, Germany
| | - Ralph Kühn
- Molecular Zoology, Department of Zoology, TUM School of Life Sciences, Technical University of Munich, 85354 Freising, Germany
- Department of Fish, Wildlife and Conservation Ecology, New Mexico State University, 8803 Las Cruces, NM, United States
| |
Collapse
|
43
|
Su Y, Fan X, Cai X, Ning J, Shen M. Effects of fecal microbiota transplantation combined with selenium on intestinal microbiota in mice with colorectal cancer. Biochem Biophys Res Commun 2024; 733:150580. [PMID: 39213702 DOI: 10.1016/j.bbrc.2024.150580] [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: 06/02/2024] [Revised: 08/01/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024]
Abstract
Colorectal cancer (CRC) is the third most common cancer in the world. With the development of high-throughput gene sequencing technology, homeostasis imbalance of the intestinal microbiota has been proven to play a key role in the pathogenesis of CRC. Furthermore, fecal bacteria transplantation (FMT) has been shown to alter the intestinal microecology, and is potentially an effective treatment for CRC. Sodium selenite plays an important role in anticancer adjuvant therapy due to its high pro-oxidation characteristics. In this study, a murine CRC tumor model was induced by AOM/DSS, and CRC mice were treated by FMT, sodium selenite, and FMT combined with sodium selenite. The results showed that FMT, sodium selenite, and FMT combined with sodium selenite inhibited the occurrence of CRC in mice, increased the abundance of beneficial intestinal bacteria, produced different microorganisms, and changed the metabolic pathways of the intestinal microbiota. In summary, FMT, sodium selenite, and FMT combined with sodium selenite can inhibit the occurrence of CRC by increasing the abundance of beneficial bacteria and regulating phenotypes and metabolic pathways. Notably, the effect of FMT combined with sodium selenite in reducing the number of tumors, protecting intestinal tissues, and restoring the diversity and richness of the intestinal microbiota is superior to that of FMT alone or sodium selenite alone. The results of this study provide new ideas for the application of FMT and selenium in the treatment of CRC.
Collapse
Affiliation(s)
- Yintong Su
- Department of Hygiene Inspection & Quarantine Science, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Xingxing Fan
- Department of Hygiene Inspection & Quarantine Science, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Xiaohua Cai
- Department of Hygiene Inspection & Quarantine Science, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Jiayu Ning
- Department of Hygiene Inspection & Quarantine Science, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Mei Shen
- Department of Hygiene Inspection & Quarantine Science, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
44
|
Araujo David B, Atif J, Vargas E Silva Castanheira F, Yasmin T, Guillot A, Ait Ahmed Y, Peiseler M, Hommes JW, Salm L, Brundler MA, Surewaard BGJ, Elhenawy W, MacParland S, Ginhoux F, McCoy K, Kubes P. Kupffer cell reverse migration into the liver sinusoids mitigates neonatal sepsis and meningitis. Sci Immunol 2024; 9:eadq9704. [PMID: 39485859 DOI: 10.1126/sciimmunol.adq9704] [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: 06/08/2024] [Accepted: 09/27/2024] [Indexed: 11/03/2024]
Abstract
In adults, liver-resident macrophages, or Kupffer cells (KCs), reside in the sinusoids and sterilize circulating blood by capturing rapidly flowing microbes. We developed quantitative intravital imaging of 1-day-old mice combined with transcriptomics, genetic manipulation, and in vivo infection assays to interrogate increased susceptibility of newborns to bloodstream infections. Whereas 1-day-old KCs were better at catching Escherichia coli in vitro, we uncovered a critical 1-week window postpartum when KCs have limited access to blood and must translocate from liver parenchyma into the sinusoids. KC migration was independent of the microbiome but depended on macrophage migration inhibitory factor, its receptor CD74, and the adhesion molecule CD44. On the basis of our findings, we propose a model of progenitor macrophage seeding of the liver sinusoids via a reverse transmigration process from liver parenchyma. These results also illustrate the importance of developing newborn mouse models to understand newborn immunity and disease.
Collapse
Affiliation(s)
- Bruna Araujo David
- Calvin, Phoebe, and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 1N4, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 1N4, Canada
| | - Jawairia Atif
- Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A1, Canada
- Ajmera Transplant Centre, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario M5G 2C4, Canada
| | - Fernanda Vargas E Silva Castanheira
- Calvin, Phoebe, and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 1N4, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 1N4, Canada
| | - Tamanna Yasmin
- Department of Biomedical and Molecular Science, Queen's University, Kingston, Ontario K7L 3N6, Canada
| | - Adrien Guillot
- Department of Hepatology and Gastroenterology, Charité-Universitätsmedizin Berlin, Berlin 13353, Germany
| | - Yeni Ait Ahmed
- Department of Hepatology and Gastroenterology, Charité-Universitätsmedizin Berlin, Berlin 13353, Germany
| | - Moritz Peiseler
- Department of Hepatology and Gastroenterology, Charité-Universitätsmedizin Berlin, Berlin 13353, Germany
- Berlin Institute of Health (BIH), Berlin 10178, Germany
| | - Josefien W Hommes
- Calvin, Phoebe, and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 1N4, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 1N4, Canada
| | - Lilian Salm
- Calvin, Phoebe, and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 1N4, Canada
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern 3010, Switzerland
| | - Marie-Anne Brundler
- Department of Pathology and Laboratory Medicine and Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 1N4, Canada
| | - Bas G J Surewaard
- Calvin, Phoebe, and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 1N4, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 1N4, Canada
| | - Wael Elhenawy
- Department of Medical Microbiology & Immunology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta T6G 2E1, Canada
- Women and Children's Health Research Institute, University of Alberta, Edmonton, Alberta T6G 1C9, Canada
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Alberta T6G 2E1, Canada
- Antimicrobial Resistance, One Health Consortium, University of Calgary, Calgary, Alberta T2N 1N4, Canada
- Striving for Pandemic Preparedness, Alberta Research Consortium, University of Alberta, Edmonton, Alberta T6G 2E1, Canada
| | - Sonya MacParland
- Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A1, Canada
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), Agency for Science, Technology, and Research (A(∗)STAR), Singapore 138648, Singapore
- Gustave Roussy Cancer Campus, Villejuif 94800, France
| | - Kathy McCoy
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 1N4, Canada
| | - Paul Kubes
- Calvin, Phoebe, and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 1N4, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 1N4, Canada
- Department of Biomedical and Molecular Science, Queen's University, Kingston, Ontario K7L 3N6, Canada
| |
Collapse
|
45
|
Shang KM, Elsheikha HM, Ma H, Wei YJ, Zhao JX, Qin Y, Li JM, Zhao ZY, Zhang XX. Metagenomic profiling of cecal microbiota and antibiotic resistome in rodents. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 286:117186. [PMID: 39426111 DOI: 10.1016/j.ecoenv.2024.117186] [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: 06/14/2024] [Revised: 09/18/2024] [Accepted: 10/10/2024] [Indexed: 10/21/2024]
Abstract
The rodent gut microbiota is a known reservoir of antimicrobial resistance, yet the distribution of antibiotic resistance genes (ARGs) within rodent cecal microbial communities and the specific bacterial species harboring these ARGs remain largely underexplored. This study employed high-throughput sequencing of 122 samples from five distinct rodent species to comprehensively profile the diversity and distribution of ARGs and to identify the bacterial hosts of these genes. A gene catalog of the rodent cecal microbiome was constructed, comprising 22,757,369 non-redundant genes. Analysis of the microbial composition and diversity revealed that Bacillota and Bacteroidota were the dominant bacterial phyla across different rodent species, with significant variations in species composition among the rodents. In total, 3703 putative antimicrobial resistance protein-coding genes were identified, corresponding to 392 unique ARG types classified into 32 resistance classes. The most enriched ARGs in the rodent cecal microbiome were associated with multidrug resistance, followed by glycopeptide and elfamycin antibiotics. Procrustes analysis demonstrated a correlation between the structure of the microbial community and the resistome. Metagenomic assembly-based host tracking indicated that most ARG-carrying contigs originated from the bacterial family Oscillospiraceae. Additionally, 130 ARGs showed significant correlations with mobile genetic elements. These findings provide new insights into the cecal microbiota and the prevalence of ARGs across five rodent species. Future research on a wider range of wild rodent species carrying ARGs will further elucidate the mechanisms underlying the transmission of antimicrobial resistance.
Collapse
Affiliation(s)
- Kai-Meng Shang
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong Province, PR China
| | - Hany M Elsheikha
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington Campus, Loughborough, United Kingdom
| | - He Ma
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong Province, PR China
| | - Yong-Jie Wei
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong Province, PR China
| | - Ji-Xin Zhao
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong Province, PR China
| | - Ya Qin
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong Province, PR China; College of Veterinary Medicine, Jilin Agricultural University, Changchun, Jilin Province, PR China
| | - Jian-Ming Li
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, Jilin Province, PR China; Jilin Provincial Engineering Research Center for Efficient Breeding and Product Development of Sika Deer, Changchun, Jilin Province, PR China; Key Laboratory of Animal Production and Product Quality and Security, Ministry of Education, Ministry of National Education, Changchun, Jilin Province, PR China
| | - Zi-Yu Zhao
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, Jilin Province, PR China
| | - Xiao-Xuan Zhang
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong Province, PR China.
| |
Collapse
|
46
|
van Leeuwen PML, Mastromonaco GF, Mykytczuk N, Schulte-Hostedde AI. Captivity conditions matter for the gut microbiota of an endangered obligate hibernator. CONSERVATION PHYSIOLOGY 2024; 12:coae072. [PMID: 39464172 PMCID: PMC11503477 DOI: 10.1093/conphys/coae072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 09/27/2024] [Accepted: 09/30/2024] [Indexed: 10/29/2024]
Abstract
Conservation breeding programmes include translocations of animals across breeding facilities, both in and ex situ, and to/from their natural habitat. Newly reintroduced Vancouver Island marmots (VIMs) originating from the captive breeding programme are known to experience high winter mortality once reintroduced. Whilst high winter mortality rates amongst reintroduced VIM populations remain a concern of unknown causes, this health issue could potentially be linked to changes in gut microbiota prior to hibernation. Furthermore, captivity is known to impact the gut microbiota of mammals that could be crucial for hibernation. In this study, we explored the diversity of bacterial communities in the gut of captive marmots during the entire active season, both kept in captivity at in situ and ex situ facilities, as well as free-ranging marmots during the summer period. Gut microbial diversity was higher in marmots held in ex situ facilities, outside of their habitat range, compared to captive marmots held within their habitat range, and in the wild, and differences in composition were also observed. In the entire active season, animals kept in the ex situ facility had increased abundance in taxa known to be mucin degraders, sulphate producers and possible cross-feeders, whilst an increase in fibre degraders of in situ and free-ranging marmots is potentially linked to diet variation between facilities. These results confirm the interest to transfer animals held at zoos to an in situ facility before relocation and expand our understanding of microbiota variation according to hibernation cycles in the context of conservation biology.
Collapse
Affiliation(s)
- Pauline M L van Leeuwen
- Department of Biology, Laurentian University, Sudbury, ON, Canada
- Conservation Genetics Laboratory, University of Liège, Liège, Belgium
| | | | - Nadia Mykytczuk
- Vale Living with Lakes Centre, Laurentian University, Sudbury, ON, Canada
| | | |
Collapse
|
47
|
Zhang X, Liu Z, Liu G, Wei Z, Qin Z, Li R, Liu Y, Jiang Z, Min Y, Peng X. Causal effect of gut microbiota on occurrence of herpes zoster and postherpetic neuralgia, and role of Tyzzerella 3. Eur J Med Res 2024; 29:511. [PMID: 39438941 PMCID: PMC11515789 DOI: 10.1186/s40001-024-02106-w] [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: 10/05/2023] [Accepted: 10/12/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Previous research has established connections between gut microbiota, immune modulation, and several virus-related diseases. However, no study has explored the relationships between gut microbiota and herpes zoster and postherpetic neuralgia (PHN). METHODS A total of 205 taxa of gut microbiota were regarded as exposures. The occurrences of herpes zoster and PHN were selected as outcomes. The causal effects of gut microbiota on herpes zoster and PHN were estimated with multiple methods for two-sample Mendelian randomization, such as inverse variance weighted (IVW), MR-Egger, and weighted median. All results were subjected to FDR correction to prevent from possibility of multiple comparison. RESULTS Among the significant findings, four taxa and one genus were identified as facilitators of herpes zoster and PHN, respectively. Conversely, six genera and eleven taxa were found to inhibit herpes zoster and PHN, respectively. The causal effect of the Tyzzerella 3 was confirmed through FDR correction, making it a key focus in this study. Specifically, it was found to causally facilitate herpes zoster primarily with IVW (OR 1.420, 95% CI 1.174-1.718, p < 0.001, q = 0.039), as there is no heterogeneity or horizontal pleiotropy found. CONCLUSIONS With investigation of the causal association between gut microbiota, and herpes zoster/PHN, significant findings were identified in 22 different taxa. Among them, Tyzzerella 3 keeps significant after multiple comparison correction, and displays potential to facilitate the occurrence of herpes zoster.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Zheran Liu
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Guihong Liu
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Zhigong Wei
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Zijian Qin
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Ruidan Li
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yingtong Liu
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Zheng Jiang
- Department of Otolaryngology-Head and Neck Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yu Min
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xingchen Peng
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
48
|
Somers SE, Davidson GL, Mbandlwa P, McKeon CM, Stanton C, Ross RP, Quinn JL. Manipulating a host-native microbial strain compensates for low microbial diversity by increasing weight gain in a wild bird population. Proc Natl Acad Sci U S A 2024; 121:e2402352121. [PMID: 39401350 PMCID: PMC11513901 DOI: 10.1073/pnas.2402352121] [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: 02/02/2024] [Accepted: 08/25/2024] [Indexed: 10/30/2024] Open
Abstract
Empirical studies from laboratory systems and humans show that the gut microbiota is linked to host health. Similar evidence for effects on traits linked to fitness in nature is rare, not least because experimentally manipulating the gut microbiota is challenging. We isolated, characterized, and cultured a bacterial strain, Lactobacillus kimchicus APC4233, directly from a wild bird (the great tit Parus major) and provided it as a self-administered dietary supplement. We assessed the impact of the treatment on the host microbiota community, on weight, and tested whether the treatment affected a previous result linking microbiota alpha diversity to weight in nestlings. The treatment dramatically increased L. kimchicus abundance in the gut microbiota and increased alpha diversity. This effect was strongest in the youngest birds, validating earlier findings pointing to a brief developmental window when the gut microbiota are most sensitive. In time-lagged models, nestling weight was higher in the treatment birds suggesting L. kimchicus may have probiotic potential. There was also a positive time-lagged relationship between diversity and weight in control birds but not in the treatment birds, suggesting L. kimchicus helped birds compensate for low alpha diversity. We discuss why ecological context is likely key when predicting impacts of the microbiome. The manipulation of the gut microbiota with a host native strain in this wild population provides direct evidence for the role of the microbiota in the ecology and evolution of natural populations.
Collapse
Affiliation(s)
- Shane E. Somers
- School of Biological, Earth and Environmental Sciences, Distillery Fields, University College Cork, CorkT23 TK30, Ireland
- APC Microbiome Ireland, University College Cork, CorkT12 YT20, Ireland
| | - Gabrielle L. Davidson
- School of Biological Sciences, University of East Anglia, NorwichNR4 7TU, United Kingdom
| | - Philiswa Mbandlwa
- APC Microbiome Ireland, University College Cork, CorkT12 YT20, Ireland
- Biosciences Department, Teagasc Food Research Centre, Moorepark, Fermoy, CorkP61 CK84, Ireland
| | - Caroline M. McKeon
- Environment and Marine Sciences, Agri-Food and Biosciences Institute, Northern IrelandBT9 5PX, United Kingdom
- Zoology Department, School of Natural Sciences, Trinity College Dublin, DublinD02 PN40, Ireland
| | - Catherine Stanton
- APC Microbiome Ireland, University College Cork, CorkT12 YT20, Ireland
- Biosciences Department, Teagasc Food Research Centre, Moorepark, Fermoy, CorkP61 CK84, Ireland
| | - R. Paul Ross
- APC Microbiome Ireland, University College Cork, CorkT12 YT20, Ireland
- Biosciences Department, Teagasc Food Research Centre, Moorepark, Fermoy, CorkP61 CK84, Ireland
| | - John L. Quinn
- School of Biological, Earth and Environmental Sciences, Distillery Fields, University College Cork, CorkT23 TK30, Ireland
- Environmental Research Institute, University College Cork, CorkT23 XE10, Ireland
| |
Collapse
|
49
|
Yu B, Wang KY, Wang NR, Zhang L, Zhang JP. Effect of probiotics and paraprobiotics on patients with sleep disorders and sub-healthy sleep conditions: a meta-analysis of randomized controlled trials. Front Neurol 2024; 15:1477533. [PMID: 39479010 PMCID: PMC11521871 DOI: 10.3389/fneur.2024.1477533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 10/02/2024] [Indexed: 11/02/2024] Open
Abstract
Background The microbial-gut-brain axis has received much attention in recent years, and regulating intestinal flora can effectively improve sleep disorders, which hints the potential effects of probiotics on sleep disorders, but lack of research evidence for meta-analysis. Therefore, this study aims to quantitatively evaluate the influence of probiotics on sleep disorders and sub-healthy sleep conditions. Methods Up to 2023, online databases including Pubmed, Embase, Cochrane library, Web of science have been searched for studies involving adults who consume probiotics or paraprobiotics in controlled trials, during which, changes in subjective and/or objective sleep parameters and contributing factors in sleeping quality are examined. We conduct a meta-analysis of 11 clinical randomized controlled studies. Results Probiotic supplementation improves sleep states to some extent in adults with sleep disorders and healthy adults with condition-induced sleep disorders (-0.34 [-0.56 to -0.13]; I 2 = 42.6%; p = 0.001). Meanwhile, subgroup analysis shows that the effect of probiotics on improving sleep disorders is influenced by other factors such as the health states of the subjects, the duration of the intervention, the type of strain, and the test criteria. Conclusion Probiotics and paraprobiotics have a significant positive effect on the sleep quality of adults with sleep disorders or sub-healthy sleep conditions. However, the therapeutic effects of probiotics on sleep problems need future additional trials. Systematic review registration https://inplasy.com/inplasy-2022-12-0066/, identifier 2022120066.
Collapse
Affiliation(s)
- Bei Yu
- The Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ke-Yi Wang
- The Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ning-Rui Wang
- The Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lu Zhang
- The Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jian-Ping Zhang
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
50
|
Huang Y, Wang Y, Huang X, Yu X. Unveiling the overlooked fungi: the vital of gut fungi in inflammatory bowel disease and colorectal cancer. Gut Pathog 2024; 16:59. [PMID: 39407244 PMCID: PMC11481806 DOI: 10.1186/s13099-024-00651-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 09/27/2024] [Indexed: 10/19/2024] Open
Abstract
The fungi of the human microbiota play important roles in the nutritional metabolism and immunological balance of the host. Recently, research has increasingly emphasised the role of fungi in modulating inflammation in intestinal diseases and maintaining health in this environment. It is therefore necessary to understand more clearly the interactions and mechanisms of the microbiota/pathogen/host relationship and the resulting inflammatory processes, as well as to offer new insights into the prevention, diagnosis and treatment of inflammatory bowel disease (IBD), colorectal cancer (CRC) and other intestinal pathologies. In this review, we comprehensively elucidate the fungal-associated pathogenic mechanisms of intestinal inflammation in IBD and related CRC, with an emphasis on three main aspects: the direct effects of fungi and their metabolites on the host, the indirect effects mediated by interactions with other intestinal microorganisms and the immune regulation of the host. Understanding these mechanisms will enable the development of innovative approaches based on the use of fungi from the resident human microbiota such as dietary interventions, fungal probiotics and faecal microbiota transplantation in the prevention, diagnosis and treatment of intestinal diseases.
Collapse
Affiliation(s)
- Yilin Huang
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
- Huankui Academy, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
| | - Yang Wang
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Xiaotian Huang
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China.
| | - Xiaomin Yu
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China.
| |
Collapse
|