1
|
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
|
2
|
Seo SM, Kim NW, Yoo ES, Lee JH, Kang AR, Jeong HB, Shim WY, Kim DH, Park YJ, Bae K, Yoon KA, Choi YK. Development of a novel complex inflammatory bowel disease mouse model: Reproducing human inflammatory bowel disease etiologies in mice. PLoS One 2024; 19:e0311310. [PMID: 39570897 PMCID: PMC11581264 DOI: 10.1371/journal.pone.0311310] [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: 02/13/2024] [Accepted: 08/30/2024] [Indexed: 11/24/2024] Open
Abstract
Inflammatory bowel disease (IBD), caused by environmental factors associated with the host's genetic traits, is represented by Crohn's disease and ulcerative colitis. Despite the increasing number of patients with IBD, the current treatment is limited to symptomatic therapy. A complex IBD model mimicking the human IBD etiology is required to overcome this limitation. Herein, we developed novel complex IBD models using interleukin 2 receptor subunit gamma (Il2rg)-deficient mice, high-fat diet, dextran sodium sulfate, and Citrobacter rodentium. The more IBD factors applied complexly, colon length shortened and inflammation worsened. The levels of pro-inflammatory cytokines increased with increased IBD factors. Anti-inflammatory cytokine decreased in all factors application but increased in Il2rg deficiency and Westernized diet combination. Additionally, the pro-inflammatory transcription factors and leaky intestinal epithelial marker were upregulated by a combination of IBD factors. Species diversity decreased with IBD factors. Phylogenetic diversity decreased as IBD factors were applied but increased with combined Il2rg deficiency and Westernized diet. The more IBD factors applied complexly, the more severe the dysbiosis. Finally, we developed a novel complex IBD model using various IBD factors. This model more closely mimic human IBD based on colonic inflammation and dysbiosis than the previous models. Based on these results, our novel complex IBD model could be a valuable tool for further IBD research.
Collapse
Affiliation(s)
- Sun-Min Seo
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Na-Won Kim
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Eun-Seon Yoo
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Ji-Hun Lee
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Ah-Reum Kang
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Han-Bi Jeong
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Won-Yong Shim
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Dong-Hyun Kim
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Young-Jun Park
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Kieun Bae
- Department of Veterinary Biochemistry, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Kyong-Ah Yoon
- Department of Veterinary Biochemistry, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Yang-Kyu Choi
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| |
Collapse
|
3
|
Niccolai E, Di Gloria L, Trolese MC, Fabbrizio P, Baldi S, Nannini G, Margotta C, Nastasi C, Ramazzotti M, Bartolucci G, Bendotti C, Nardo G, Amedei A. Host genetics and gut microbiota influence lipid metabolism and inflammation: potential implications for ALS pathophysiology in SOD1 G93A mice. Acta Neuropathol Commun 2024; 12:174. [PMID: 39506789 PMCID: PMC11539544 DOI: 10.1186/s40478-024-01877-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: 08/01/2024] [Accepted: 10/16/2024] [Indexed: 11/08/2024] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a devastating neurodegenerative disorder characterized by the progressive loss of motor neurons, with genetic and environmental factors contributing to its complex pathogenesis. Dysregulated immune responses and altered energetic metabolism are key features, with emerging evidence implicating the gut microbiota (GM) in disease progression. We investigated the interplay among genetic background, GM composition, metabolism, and immune response in two distinct ALS mouse models: 129Sv_G93A and C57Ola_G93A, representing rapid and slow disease progression, respectively.Using 16 S rRNA sequencing and fecal metabolite analysis, we characterized the GM composition and metabolite profiles in non-transgenic (Ntg) and SOD1G93A mutant mice of both strains. Our results revealed strain-specific differences in GM composition and functions, particularly in the abundance of taxa belonging to Erysipelotrichaceae and the levels of short and medium-chain fatty acids in fecal samples. The SOD1 mutation induces significant shifts in GM colonization in both strains, with C57Ola_G93A mice showing changes resembling those in 129 Sv mice, potentially affecting disease pathogenesis. ALS symptom progression does not significantly alter microbiota composition, suggesting stability.Additionally, we assessed systemic immunity and inflammatory responses revealing strain-specific differences in immune cell populations and cytokine levels.Our findings underscore the substantial influence of genetic background on GM composition, metabolism, and immune response in ALS mouse models. These strain-specific variations may contribute to differences in disease susceptibility and progression rates. Further elucidating the mechanisms underlying these interactions could offer novel insights into ALS pathogenesis and potential therapeutic targets.
Collapse
Affiliation(s)
- Elena Niccolai
- Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, Florence, 50134, Italy
| | - Leandro Di Gloria
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale Morgagni 50, Florence, 50134, Italy
| | - Maria Chiara Trolese
- Laboratory of Molecular Neurobiology and Preclinical Therapy, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, Milano, 20156, Italy
| | - Paola Fabbrizio
- Laboratory of Molecular Neurobiology and Preclinical Therapy, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, Milano, 20156, Italy
| | - Simone Baldi
- Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, Florence, 50134, Italy
| | - Giulia Nannini
- Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, Florence, 50134, Italy
| | - Cassandra Margotta
- Laboratory of Molecular Neurobiology and Preclinical Therapy, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, Milano, 20156, Italy
| | - Claudia Nastasi
- Unit of Immunopharmacology, Department of Experimental Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, Milano, 20156, Italy
| | - Matteo Ramazzotti
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale Morgagni 50, Florence, 50134, Italy
| | - Gianluca Bartolucci
- Department of Neurosciences, Psychology, Drug Research and Child Health Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Viale Pieraccini 6, Florence, 50139, Italy
| | - Caterina Bendotti
- Laboratory of Molecular Neurobiology and Preclinical Therapy, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, Milano, 20156, Italy
| | - Giovanni Nardo
- Laboratory of Molecular Neurobiology and Preclinical Therapy, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, Milano, 20156, Italy.
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, Florence, 50134, Italy.
| |
Collapse
|
4
|
Rook GAW. Evolution and the critical role of the microbiota in the reduced mental and physical health associated with low socioeconomic status (SES). Neurosci Biobehav Rev 2024; 161:105653. [PMID: 38582194 DOI: 10.1016/j.neubiorev.2024.105653] [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: 11/28/2023] [Accepted: 04/03/2024] [Indexed: 04/08/2024]
Abstract
The evolution of the gut-microbiota-brain axis in animals reveals that microbial inputs influence metabolism, the regulation of inflammation and the development of organs, including the brain. Inflammatory, neurodegenerative and psychiatric disorders are more prevalent in people of low socioeconomic status (SES). Many aspects of low SES reduce exposure to the microbial inputs on which we are in a state of evolved dependence, whereas the lifestyle of wealthy citizens maintains these exposures. This partially explains the health deficit of low SES, so focussing on our evolutionary history and on environmental and lifestyle factors that distort microbial exposures might help to mitigate that deficit. But the human microbiota is complex and we have poor understanding of its functions at the microbial and mechanistic levels, and in the brain. Perhaps its composition is more flexible than the microbiota of animals that have restricted habitats and less diverse diets? These uncertainties are discussed in relation to the encouraging but frustrating results of attempts to treat psychiatric disorders by modulating the microbiota.
Collapse
Affiliation(s)
- Graham A W Rook
- Centre for Clinical Microbiology, Department of infection, UCL (University College London), London, UK.
| |
Collapse
|
5
|
Zou Y, Wang S, Zhang H, Gu Y, Chen H, Huang Z, Yang F, Li W, Chen C, Men L, Tian Q, Xie T. The triangular relationship between traditional Chinese medicines, intestinal flora, and colorectal cancer. Med Res Rev 2024; 44:539-567. [PMID: 37661373 DOI: 10.1002/med.21989] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 07/05/2023] [Accepted: 08/05/2023] [Indexed: 09/05/2023]
Abstract
Over the past decade, colorectal cancer has reported a higher incidence in younger adults and a lower mortality rate. Recently, the influence of the intestinal flora in the initiation, progression, and treatment of colorectal cancer has been extensively studied, as well as their positive therapeutic impact on inflammation and the cancer microenvironment. Historically, traditional Chinese medicine (TCM) has been widely used in the treatment of colorectal cancer via promoted cancer cell apoptosis, inhibited cancer metastasis, and reduced drug resistance and side effects. The present research is more on the effect of either herbal medicine or intestinal flora on colorectal cancer. The interactions between TCM and intestinal flora are bidirectional and the combined impacts of TCM and gut microbiota in the treatment of colon cancer should not be neglected. Therefore, this review discusses the role of intestinal bacteria in the progression and treatment of colorectal cancer by inhibiting carcinogenesis, participating in therapy, and assisting in healing. Then the complex anticolon cancer effects of different kinds of TCM monomers, TCM drug pairs, and traditional Chinese prescriptions embodied in apoptosis, metastasis, immune suppression, and drug resistance are summarized separately. In addition, the interaction between TCM and intestinal flora and the combined effect on cancer treatment were analyzed. This review provides a mechanistic reference for the application of TCM and intestinal flora in the clinical treatment of colorectal cancer and paves the way for the combined development and application of microbiome and TCM.
Collapse
Affiliation(s)
- Yuqing Zou
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Shuling Wang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Honghua Zhang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Yuxin Gu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Huijuan Chen
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Zhihua Huang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Feifei Yang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Wenqi Li
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Cheng Chen
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Lianhui Men
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Qingchang Tian
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Tian Xie
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| |
Collapse
|
6
|
Peled S, Freilich S, Hanani H, Kashi Y, Livney YD. Next-generation prebiotics: Maillard-conjugates of 2'-fucosyllactose and lactoferrin hydrolysates beneficially modulate gut microbiome composition and health promoting activity in a murine model. Food Res Int 2024; 177:113830. [PMID: 38225111 DOI: 10.1016/j.foodres.2023.113830] [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: 10/19/2023] [Revised: 12/01/2023] [Accepted: 12/05/2023] [Indexed: 01/17/2024]
Abstract
Current prebiotics are predominantly carbohydrates. However, great competition exists among gut microbes for the scarce protein in the colon, as most consumed protein is digested and absorbed in the small intestine. Herein we evaluated in-vivo novel next-generation prebiotics: protein-containing-prebiotics, for selectively-targeted delivery of protein to colonic probiotics, to boost their growth. This system is based on micellar-particles, composed of Maillard-glycoconjugates of 2'-Fucosyllactose (2'-FL, human-milk-oligosaccharide) shell, engulfing lactoferrin peptic-then-tryptic hydrolysate (LFH) core. This core-shell structure lowers protein-core digestibility, while the prebiotic glycans are hypothesized to serve as molecular-recognition ligands for selectively targeting probiotics. To study the efficacy of this novel prebiotic, we fed C57BL/6JRccHsd mice with either 2'-FL-LFH Maillard-glycoconjugates, unconjugated components (control), or saline (blank). Administration of 2'-FL-LFH significantly increased the levels of short-chain-fatty-acids (SCFAs)-producing bacterial families (Ruminococcaceae, Lachnospiraceae) and genus (Odoribacter) and the production of the health-related metabolites, SCFAs, compared to the unconjugated components and to saline. The SCFAs-producing genus Prevotella significantly increased upon 2'-FL-LFH consumption, compared to only moderate increase in the unconjugated components. Interestingly, the plasma-levels of inflammation-inducing lipopolysaccharides (LPS), which indicate increased gut-permeability, were significantly lower in the 2'-FL-LFH group compared to the unconjugated-components and the saline groups. We found that Maillard-glycoconjugates of 2'-FL-LFH can serve as novel protein-containing prebiotics, beneficially modulating gut microbial composition and its metabolic activity, thereby contributing to host health more effectively than the conventional carbohydrate-only prebiotics.
Collapse
Affiliation(s)
- Stav Peled
- Laboratory of Biopolymers for Food and Health, Department of Biotechnology and Food Engineering, Technion - Israel Institute of Technology, Haifa 3200003, Israel
| | - Shay Freilich
- Laboratory of Applied Genomics, Department of Biotechnology and Food Engineering, Technion - Israel Institute of Technology, Haifa 3200003, Israel
| | - Hila Hanani
- Laboratory of Applied Genomics, Department of Biotechnology and Food Engineering, Technion - Israel Institute of Technology, Haifa 3200003, Israel
| | - Yechezkel Kashi
- Laboratory of Applied Genomics, Department of Biotechnology and Food Engineering, Technion - Israel Institute of Technology, Haifa 3200003, Israel
| | - Yoav D Livney
- Laboratory of Biopolymers for Food and Health, Department of Biotechnology and Food Engineering, Technion - Israel Institute of Technology, Haifa 3200003, Israel.
| |
Collapse
|
7
|
Duffy EP, Bachtell RK, Ehringer MA. Opioid trail: Tracking contributions to opioid use disorder from host genetics to the gut microbiome. Neurosci Biobehav Rev 2024; 156:105487. [PMID: 38040073 PMCID: PMC10836641 DOI: 10.1016/j.neubiorev.2023.105487] [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: 08/29/2023] [Revised: 11/22/2023] [Accepted: 11/27/2023] [Indexed: 12/03/2023]
Abstract
Opioid use disorder (OUD) is a worldwide public health crisis with few effective treatment options. Traditional genetics and neuroscience approaches have provided knowledge about biological mechanisms that contribute to OUD-related phenotypes, but the complexity and magnitude of effects in the brain and body remain poorly understood. The gut-brain axis has emerged as a promising target for future therapeutics for several psychiatric conditions, so characterizing the relationship between host genetics and the gut microbiome in the context of OUD will be essential for development of novel treatments. In this review, we describe evidence that interactions between host genetics, the gut microbiome, and immune signaling likely play a key role in mediating opioid-related phenotypes. Studies in humans and model organisms consistently demonstrated that genetic background is a major determinant of gut microbiome composition. Furthermore, the gut microbiome is susceptible to environmental influences such as opioid exposure. Additional work focused on gene by microbiome interactions will be necessary to gain improved understanding of their effects on OUD-related behaviors.
Collapse
Affiliation(s)
- Eamonn P Duffy
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA; Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, USA.
| | - Ryan K Bachtell
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, USA; Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, USA
| | - Marissa A Ehringer
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA; Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, USA
| |
Collapse
|
8
|
Cui W, Hull L, Zizzo A, Wang L, Lin B, Zhai M, Xiao M. The gut microbiome changes in wild type and IL-18 knockout mice after 9.0 Gy total body irradiation. Anim Microbiome 2023; 5:42. [PMID: 37679818 PMCID: PMC10485964 DOI: 10.1186/s42523-023-00262-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 08/24/2023] [Indexed: 09/09/2023] Open
Abstract
BACKGROUND Recent studies have shown that gut microbiome plays important roles in response to radiation exposure. IL-18, an inflammatory cytokine, is highly elevated in mice, mini-pigs and nonhuman primates after radiation exposure. Blocking IL-18 using its endogenous binding protein (IL-18BP) increases mice survival after radiation exposure by decreasing bone marrow interferon-gamma levels. METHODS To further characterize the roles of IL-18 in response to radiation, both wild type and IL-18 knockout (IL-18 KO) mice were exposed to 9.0 Gy total body irradiation (TBI). The 30-day survival result demonstrated that IL-18 KO mice were significantly more resistant to radiation compared to the wild type mice (p < 0.0001). Mouse faecal samples were collected at pre-radiation (d0), d1, d3, d7, d14, d21 and d29 after radiation exposure. Microbiome profiling was performed on the faecal samples using 16S and ITS sequencing technology. RESULTS Data analysis showed that there was significant difference in the bacterial microbiome between wild type and IL-18 KO mice. Cohousing of wild type and IL-18 KO mice decreased the bacterial microbiome difference between the two genotypes. Much fewer bacterial genera were significantly changed in wild type mice than the IL-18 KO mice after radiation exposure. The different composition of the IL-18 KO mice and wild type mice persisted even after radiation exposure. Bacterial genera that significantly correlated with other genera were identified in the IL-18 KO and wild type mice. The metabolic pathways that differentially expressed in both genotypes were identified. The animal bacterial microbiome data could be used to predict the animal's radiation status. The fungal microbiome had no significant difference regarding genotype or time after radiation exposure. CONCLUSION The current study helps understand the gut microbiome in different genetic backgrounds and its temporal changes after radiation exposure. Our data provide insight into the mechanisms underlying radiation-induced toxicity and help identify bacteria important in response to radiation.
Collapse
Affiliation(s)
- Wanchang Cui
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, 4555 South Palmer Road, Bethesda, MD, 20889-5648, USA.
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, 20817, USA.
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA.
| | - Lisa Hull
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, 4555 South Palmer Road, Bethesda, MD, 20889-5648, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, 20817, USA
| | - Alex Zizzo
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, 4555 South Palmer Road, Bethesda, MD, 20889-5648, USA
| | - Li Wang
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, 4555 South Palmer Road, Bethesda, MD, 20889-5648, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, 20817, USA
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
| | - Bin Lin
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, 4555 South Palmer Road, Bethesda, MD, 20889-5648, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, 20817, USA
| | - Min Zhai
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, 4555 South Palmer Road, Bethesda, MD, 20889-5648, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, 20817, USA
| | - Mang Xiao
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, 4555 South Palmer Road, Bethesda, MD, 20889-5648, USA.
| |
Collapse
|
9
|
Vallée N, Dugrenot E, Desruelle AV, Richard S, Coupé S, Ramdani C, Guieu R, Risso JJ, Gaillard S, Guerrero F. Highlighting of the interactions of MYD88 and NFKB1 SNPs in rats resistant to decompression sickness: toward an autoimmune response. Front Physiol 2023; 14:1253856. [PMID: 37664439 PMCID: PMC10470123 DOI: 10.3389/fphys.2023.1253856] [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: 07/06/2023] [Accepted: 08/04/2023] [Indexed: 09/05/2023] Open
Abstract
Decompression sickness (DCS) with neurological disorders includes an inappropriate inflammatory response which degenerates slowly, even after the disappearance of the bubbles. There is high inter-individual variability in terms of the occurrence of DCS that could have been mastered by the selection and then the breeding of DCS-resistant rats. We hypothesized the selection of single-nucleotide polymorphisms (SNPs) linked to autoimmunity operated upon a generation of a DCS-resistant strain of rats. We used the candidate gene approach and targeted SNPs linked to the signaling cascade that directly regulates inflammation of innate immunity transiting by the Toll-like receptors. Twenty candidate SNPs were investigated in 36 standard rats and 33 DCS-resistant rats. For the first time, we identify a diplotype (i.e., with matched haplotypes)-when coinherited-that strengthens protection against DCS, which is not strictly homozygous and suggests that a certain tolerance may be considered. We deduced an ideal haplotype of six variants from it (MyD88_50-T, _49-A, _97-C coupled to NFKB_85-T, _69-T, _45-T) linked to the resistant phenotype. Four among the six identified variants are located in pre- and/or post-transcriptional areas regulating MyD88 or NFKB1 expression. Because of missense mutations, the other two variants induce a structural change in the NFKB1 protein complex including one damage alteration according to the Missense3D algorithm. In addition to the MyD88/NFKB1 haplotype providing rats with a strong resistance to DCS, this also highlights the importance that the immune response, here linked to the genetic heritage, can have in the development of DCS and offer a new perspective for therapeutic strategies.
Collapse
Affiliation(s)
- Nicolas Vallée
- Institut de Recherche Biomédicale des Armées, Equipe de Recherche Subaquatique Opérationnelle, Toulon, France
| | | | - Anne-Virginie Desruelle
- Institut de Recherche Biomédicale des Armées, Equipe de Recherche Subaquatique Opérationnelle, Toulon, France
| | | | | | - Céline Ramdani
- Institut de Recherche Biomédicale des Armées, Equipe de Recherche Subaquatique Opérationnelle, Toulon, France
| | - Régis Guieu
- Université d’Aix-Marseille, Marseille, France
| | - Jean-Jacques Risso
- Institut de Recherche Biomédicale des Armées, Equipe de Recherche Subaquatique Opérationnelle, Toulon, France
| | | | | |
Collapse
|
10
|
AIZAWA S, UEBANSO T, SHIMOHATA T, MAWATARI K, TAKAHASHI A. Effects of the loss of maternal gut microbiota before pregnancy on gut microbiota, food allergy susceptibility, and epigenetic modification on subsequent generations. BIOSCIENCE OF MICROBIOTA, FOOD AND HEALTH 2023; 42:203-212. [PMID: 37404565 PMCID: PMC10315195 DOI: 10.12938/bmfh.2022-093] [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: 12/21/2022] [Accepted: 03/17/2023] [Indexed: 07/06/2023]
Abstract
Maternal environments affect the health of offspring in later life. Changes in epigenetic modifications may partially explain this phenomenon. The gut microbiota is a critical environmental factor that influences epigenetic modifications of host immune cells and the development of food allergies. However, whether changes in the maternal gut microbiota affect the development of food allergies and related epigenetic modifications in subsequent generations remains unclear. Here, we investigated the effects of antibiotic treatment before pregnancy on the development of the gut microbiota, food allergies, and epigenetic modifications in F1 and F2 mice. We found that pre-conception antibiotic treatment affected the gut microbiota composition in F1 but not F2 offspring. F1 mice born to antibiotic-treated mothers had a lower proportion of butyric acid-producing bacteria and, consequently, a lower butyric acid concentration in their cecal contents. The methylation level in the DNA of intestinal lamina propria lymphocytes, food allergy susceptibility, and production of antigen-specific IgE in the F1 and F2 mice were not different between those born to control and antibiotic-treated mothers. In addition, F1 mice born to antibiotic-treated mothers showed increased fecal excretion related to the stress response in a novel environment. These results suggest that the maternal gut microbiota is effectively passed onto F1 offspring but has little effect on food allergy susceptibility or DNA methylation levels in offspring.
Collapse
Affiliation(s)
- Shinta AIZAWA
- Department of Preventive Environment and Nutrition, Institute
of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho,
Tokushima-shi, Tokushima 770-8503, Japan
| | - Takashi UEBANSO
- Department of Preventive Environment and Nutrition, Institute
of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho,
Tokushima-shi, Tokushima 770-8503, Japan
- Department of Microbial Control, Institute of Biomedical
Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima-shi,
Tokushima 770-8503, Japan
| | - Takaaki SHIMOHATA
- Faculty of Marine Biosciences, Fukui Prefectural University,
1-1 Gakuen-cho, Obama-shi, Fukui 917-0003, Japan
| | - Kazuaki MAWATARI
- Department of Preventive Environment and Nutrition, Institute
of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho,
Tokushima-shi, Tokushima 770-8503, Japan
- Department of Microbial Control, Institute of Biomedical
Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima-shi,
Tokushima 770-8503, Japan
| | - Akira TAKAHASHI
- Department of Preventive Environment and Nutrition, Institute
of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho,
Tokushima-shi, Tokushima 770-8503, Japan
- Department of Microbial Control, Institute of Biomedical
Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima-shi,
Tokushima 770-8503, Japan
| |
Collapse
|
11
|
Hazlett LD, Xu S, Somayajulu M, McClellan SA. Host-microbe interactions in cornea. Ocul Surf 2023; 28:413-423. [PMID: 34619389 PMCID: PMC8977393 DOI: 10.1016/j.jtos.2021.09.008] [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: 08/20/2021] [Revised: 09/23/2021] [Accepted: 09/26/2021] [Indexed: 11/23/2022]
Abstract
Corneal infections result through interaction between microbes and host innate immune receptors. Damage to the cornea occurs as a result of microbial virulence factors and is often exacerbated by lack of a controlled host immune response; the latter contributing to bystander damage to corneal structure. Understanding mechanisms involved in host microbial interactions is critical to development of novel therapeutic targets, ultimate control of microbial pathogenesis, and restoration of tissue homeostasis. Studies on these interactions continue to provide exciting findings directly related to this ultimate goal.
Collapse
Affiliation(s)
- Linda D Hazlett
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, 48201, USA.
| | - Shunbin Xu
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Mallika Somayajulu
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Sharon A McClellan
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| |
Collapse
|
12
|
Maraci Ö, Antonatou-Papaioannou A, Jünemann S, Engel K, Castillo-Gutiérrez O, Busche T, Kalinowski J, Caspers BA. Timing matters: age-dependent impacts of the social environment and host selection on the avian gut microbiota. MICROBIOME 2022; 10:202. [PMID: 36434663 PMCID: PMC9700942 DOI: 10.1186/s40168-022-01401-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 10/25/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND The establishment of the gut microbiota in early life is a critical process that influences the development and fitness of vertebrates. However, the relative influence of transmission from the early social environment and host selection throughout host ontogeny remains understudied, particularly in avian species. We conducted conspecific and heterospecific cross-fostering experiments in zebra finches (Taeniopygia guttata) and Bengalese finches (Lonchura striata domestica) under controlled conditions and repeatedly sampled the faecal microbiota of these birds over the first 3 months of life. We thus documented the development of the gut microbiota and characterised the relative impacts of the early social environment and host selection due to species-specific characteristics and individual genetic backgrounds across ontogeny by using 16S ribosomal RNA gene sequencing. RESULTS The taxonomic composition and community structure of the gut microbiota changed across ontogenetic stages; juvenile zebra finches exhibited higher alpha diversity than adults at the post-breeding stage. Furthermore, in early development, the microbial communities of juveniles raised by conspecific and heterospecific foster parents resembled those of their foster family, emphasising the importance of the social environment. In later stages, the social environment continued to influence the gut microbiota, but host selection increased in importance. CONCLUSIONS We provided a baseline description of the developmental succession of gut microbiota in zebra finches and Bengalese finches, which is a necessary first step for understanding the impact of the early gut microbiota on host fitness. Furthermore, for the first time in avian species, we showed that the relative strengths of the two forces that shape the establishment and maintenance of the gut microbiota (i.e. host selection and dispersal from the social environment) change during development, with host selection increasing in importance. This finding should be considered when experimentally manipulating the early-life gut microbiota. Our findings also provide new insights into the mechanisms of host selection. Video Abstract.
Collapse
Affiliation(s)
- Öncü Maraci
- Department of Behavioural Ecology, Bielefeld University, Bielefeld, Germany.
| | - Anna Antonatou-Papaioannou
- Evolutionary Biology, Bielefeld University, Bielefeld, Germany
- Institute of Biology-Zoology, Freie Universität Berlin, Berlin, Germany
| | - Sebastian Jünemann
- Institute for Bio- and Geosciences, Research Center Jülich, Jülich, Germany
- Faculty of Technology, Bielefeld University, Bielefeld, Germany
| | - Kathrin Engel
- Department of Behavioural Ecology, Bielefeld University, Bielefeld, Germany
| | - Omar Castillo-Gutiérrez
- Faculty of Technology, Bielefeld University, Bielefeld, Germany
- Center for Biotechnology (CeBiTec), Bielefeld University, Bielefeld, Germany
| | - Tobias Busche
- Center for Biotechnology (CeBiTec), Bielefeld University, Bielefeld, Germany
| | - Jörn Kalinowski
- Center for Biotechnology (CeBiTec), Bielefeld University, Bielefeld, Germany
| | - Barbara A Caspers
- Department of Behavioural Ecology, Bielefeld University, Bielefeld, Germany
| |
Collapse
|
13
|
Kleber KT, Iranpur KR, Perry LM, Cruz SM, Razmara AM, Culp WTN, Kent MS, Eisen JA, Rebhun RB, Canter RJ. Using the canine microbiome to bridge translation of cancer immunotherapy from pre-clinical murine models to human clinical trials. Front Immunol 2022; 13:983344. [PMID: 36032113 PMCID: PMC9412231 DOI: 10.3389/fimmu.2022.983344] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 07/26/2022] [Indexed: 11/27/2022] Open
Abstract
The microbiome has clearly been established as a cutting-edge field in tumor immunology and immunotherapy. Growing evidence supports the role of the microbiome in immune surveillance, self-tolerance, and response to immune checkpoint inhibitors such as anti PD-L1 and CTLA-4 blockade (1-6). Moreover, recent studies including those using fecal microbial transplantation (FMT) have demonstrated that response to checkpoint immunotherapies may be conferred or eliminated through gut microbiome modulation (7, 8). Consequently, studies evaluating microbiota-host immune and metabolic interactions remain an area of high impact research. While observations in murine models have highlighted the importance of the microbiome in response to therapy, we lack sufficient understanding of the exact mechanisms underlying these interactions. Furthermore, mouse and human gut microbiome composition may be too dissimilar for discovery of all relevant gut microbial biomarkers. Multiple cancers in dogs, including lymphoma, high grade gliomas, melanomas and osteosarcoma (OSA) closely resemble their human analogues, particularly in regard to metastasis, disease recurrence and response to treatment. Importantly, dogs with these spontaneous cancers also have intact immune systems, suggesting that microbiome analyses in these subjects may provide high yield information, especially in the setting of novel immunotherapy regimens which are currently expanding rapidly in canine comparative oncology (9, 10). Additionally, as onco-microbiotic therapies are developed to modify gut microbiomes for maximal responsiveness, large animal models with intact immune systems will be useful for trialing interventions and monitoring adverse events. Together, pre-clinical mechanistic studies and large animal trials can help fully unlock the potential of the microbiome as a diagnostic and therapeutic target in cancer.
Collapse
Affiliation(s)
- Kara T. Kleber
- Division of Surgical Oncology, Department of Surgery, University of California Davis Medical Center, Sacramento, CA, United States
| | - Khurshid R. Iranpur
- Division of Surgical Oncology, Department of Surgery, University of California Davis Medical Center, Sacramento, CA, United States
| | - Lauren M. Perry
- Division of Surgical Oncology, Department of Surgery, University of California Davis Medical Center, Sacramento, CA, United States
| | - Sylvia M. Cruz
- Division of Surgical Oncology, Department of Surgery, University of California Davis Medical Center, Sacramento, CA, United States
| | - Aryana M. Razmara
- School of Veterinary Medicine, University of California Davis, Sacramento, CA, United States
| | - William T. N. Culp
- Center for Companion Animal Health Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California Davis, Davis, CA, United States
| | - Michael S. Kent
- Center for Companion Animal Health Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California Davis, Davis, CA, United States
| | - Jonathan A. Eisen
- Department of Medical Microbiology and Immunology, University of California Davis, Davis, CA, United States
| | - Robert B. Rebhun
- Center for Companion Animal Health Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California Davis, Davis, CA, United States
| | - Robert J. Canter
- Division of Surgical Oncology, Department of Surgery, University of California Davis Medical Center, Sacramento, CA, United States
| |
Collapse
|
14
|
Desruelle AV, de Maistre S, Gaillard S, Richard S, Tardivel C, Martin JC, Blatteau JE, Boussuges A, Rives S, Risso JJ, Vallee N. Cecal Metabolomic Fingerprint of Unscathed Rats: Does It Reflect the Good Response to a Provocative Decompression? Front Physiol 2022; 13:882944. [PMID: 35655958 PMCID: PMC9152359 DOI: 10.3389/fphys.2022.882944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 04/27/2022] [Indexed: 11/23/2022] Open
Abstract
On one side, decompression sickness (DCS) with neurological disorders lead to a reshuffle of the cecal metabolome of rats. On the other side, there is also a specific and different metabolomic signature in the cecum of a strain of DCS-resistant rats, that are not exposed to hyperbaric protocol. We decide to study a conventional strain of rats that resist to an accident-provoking hyperbaric exposure, and we hypothesize that the metabolomic signature put forward may correspond to a physiological response adapted to the stress induced by diving. The aim is to verify and characterize whether the cecal compounds of rats resistant to the provocative dive have a cecal metabolomic signature different from those who do not dive. 35 asymptomatic diver rats are selected to be compared to 21 rats non-exposed to the hyperbaric protocol. Because our aim is essentially to study the differences in the cecal metabolome associated with the hyperbaric exposure, about half of the rats are fed soy and the other half of maize in order to better rule out the effect of the diet itself. Lower levels of IL-1β and glutathione peroxidase (GPX) activity are registered in blood of diving rats. No blood cell mobilization is noted. Conventional and ChemRICH approaches help the metabolomic interpretation of the 185 chemical compounds analyzed in the cecal content. Statistical analysis show a panel of 102 compounds diet related. 19 are in common with the hyperbaric protocol effect. Expression of 25 compounds has changed in the cecal metabolome of rats resistant to the provocative dive suggesting an alteration of biliary acids metabolism, most likely through actions on gut microbiota. There seem to be also weak changes in allocations dedicated to various energy pathways, including hormonal reshuffle. Some of the metabolites may also have a role in regulating inflammation, while some may be consumed for the benefit of oxidative stress management.
Collapse
Affiliation(s)
- Anne-Virginie Desruelle
- Institut de Recherche Biomédicale des Armées, Equipe de Recherche Subaquatique Opérationnelle, Toulon Cedex, France
| | - Sébastien de Maistre
- Service de Médecine Hyperbare Expertise Plongée, Hôpital d'Instruction des Armées Sainte-Anne, Toulon Cedex, France
| | | | | | - Catherine Tardivel
- C2VN, INRAE, INSERM, BIOMET, Aix Marseille University, Faculté de Médecine La Timone, Marseille, France
| | - Jean-Charles Martin
- C2VN, INRAE, INSERM, BIOMET, Aix Marseille University, Faculté de Médecine La Timone, Marseille, France
| | - Jean-Eric Blatteau
- Service de Médecine Hyperbare Expertise Plongée, Hôpital d'Instruction des Armées Sainte-Anne, Toulon Cedex, France
| | - Alain Boussuges
- Institut de Recherche Biomédicale des Armées, Equipe de Recherche Subaquatique Opérationnelle, Toulon Cedex, France
| | - Sarah Rives
- Institut de Recherche Biomédicale des Armées, Equipe de Recherche Subaquatique Opérationnelle, Toulon Cedex, France
| | - Jean-Jacques Risso
- Institut de Recherche Biomédicale des Armées, Equipe de Recherche Subaquatique Opérationnelle, Toulon Cedex, France
| | - Nicolas Vallee
- Institut de Recherche Biomédicale des Armées, Equipe de Recherche Subaquatique Opérationnelle, Toulon Cedex, France
- *Correspondence: Nicolas Vallee,
| |
Collapse
|
15
|
Ryu EP, Davenport ER. Host Genetic Determinants of the Microbiome Across Animals: From Caenorhabditis elegans to Cattle. Annu Rev Anim Biosci 2022; 10:203-226. [PMID: 35167316 PMCID: PMC11000414 DOI: 10.1146/annurev-animal-020420-032054] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Animals harbor diverse communities of microbes within their gastrointestinal tracts. Phylogenetic relationship, diet, gut morphology, host physiology, and ecology all influence microbiome composition within and between animal clades. Emerging evidence points to host genetics as also playing a role in determining gut microbial composition within species. Here, we discuss recent advances in the study of microbiome heritability across a variety of animal species. Candidate gene and discovery-based studies in humans, mice, Drosophila, Caenorhabditis elegans, cattle, swine, poultry, and baboons reveal trends in the types of microbes that are heritable and the host genes and pathways involved in shaping the microbiome. Heritable gut microbes within a host species tend to be phylogenetically restricted. Host genetic variation in immune- and growth-related genes drives the abundances of these heritable bacteria within the gut. With only a small slice of the metazoan branch of the tree of life explored to date, this is an area rife with opportunities to shed light into the mechanisms governing host-microbe relationships.
Collapse
Affiliation(s)
- Erica P Ryu
- Department of Biology, Pennsylvania State University, University Park, Pennsylvania, USA; ,
| | - Emily R Davenport
- Department of Biology, Pennsylvania State University, University Park, Pennsylvania, USA; ,
- Huck Institutes of the Life Sciences and Institute for Computational and Data Sciences, Pennsylvania State University, University Park, Pennsylvania, USA
| |
Collapse
|
16
|
Comparative analysis of gut microbial composition and potential functions in captive forest and alpine musk deer. Appl Microbiol Biotechnol 2022; 106:1325-1339. [PMID: 35037997 PMCID: PMC8816758 DOI: 10.1007/s00253-022-11775-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 12/28/2021] [Accepted: 01/09/2022] [Indexed: 12/12/2022]
Abstract
Gut microbiota forms a unique microecosystem and performs various irreplaceable metabolic functions for ruminants. The gut microbiota is important for host health and provides new insight into endangered species conservation. Forest musk deer (FMD) and alpine musk deer (AMD) are typical small ruminants, globally endangered due to excessive hunting and habitat loss. Although nearly 60 years of captive musk deer breeding has reduced the hunting pressure in the wild, fatal gastrointestinal diseases restrict the growth of captive populations. In this study, 16S rRNA high-throughput sequencing revealed the differences in gut microbiota between FMD and AMD based on 166 fecal samples. The alpha diversity was higher in FMD than in AMD, probably helping FMD adapt to different and wider habitats. The ß-diversity was higher between adult FMD and AMD than juveniles and in winter than late spring. The phylum Firmicutes and the genera Christensenellaceae R7 group, Ruminococcus, Prevotellaceae UCG-004, and Monoglobus were significantly higher in abundance in FMD than in AMD. However, the phylum Bacteroidetes and genera Bacteroides, UCG-005, Rikenellaceae RC9 gut group, and Alistipes were significantly higher in AMD than FMD. The expression of metabolic functions was higher in AMD than in FMD, a beneficial pattern for AMD to maintain higher energy and substance metabolism. Captive AMD may be at higher risk of intestinal diseases than FMD, with higher relative abundances of most opportunistic pathogens and the expression of disease-related functions. These results provide valuable data for breeding healthy captive musk deer and assessing their adaptability in the wild. KEY POINTS: • Alpha diversity of gut microbiota was higher in FMD than that in AMD • Expression of metabolic and disease-related functions was higher in AMD than in FMD.
Collapse
|
17
|
Yang J, Zhong Y, Xu L, Zeng B, Lai K, Yang M, Li D, Zhao Y, Zhang M, Li D. The Dominating Role of Genetic Background in Shaping Gut Microbiota of Honeybee Queen Over Environmental Factors. Front Microbiol 2021; 12:722901. [PMID: 34803942 PMCID: PMC8603915 DOI: 10.3389/fmicb.2021.722901] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 09/29/2021] [Indexed: 11/13/2022] Open
Abstract
A balanced, diverse gut microbiota is vital for animal health. The microbial population is shaped by multiple factors including genetic background and environment, but other determinants remain controversial. Numerous studies suggest that the dominant factor is genetic background while others emphasize the environmental factors. Here, we bred asexual hybridization queens (AHQs) of honeybees through nutritional crossbreeding (laid in Apis mellifera colony but bred in Apis cerana colony), sequenced their gut microbiome, and compared it with normally bred sister queens to determine the primary factor shaping the gut microbiota. Our results showed that the dominant genera in the gut microbiota of AHQs were Brevundimonas, Bombella, and Lactobacillus, and its microbial community was more related to A. mellifera queens. The AHQs had a moderate number of different bacterial species and diversity, but total bacterial numbers were low. There were more significant taxa identified in the comparison between AHQ and A. cerana queen according to LEfSe analysis results. The only genetic-specific taxon we figured out was Brevundimonas. The growth of core bacterial abundance showed different characteristics among different queen groups in the first week after emerging. Collectively, this study suggested that the genetic background played a more dominant role than environmental factors in shaping the gut microbiota of honeybee queen and the microbiota of midgut was more sensitive than that of rectum to this impact.
Collapse
Affiliation(s)
- Jiandong Yang
- College of Animal Sciences and Technology, Sichuan Agricultural University, Chengdu, China
| | - Yun Zhong
- College of Animal Sciences and Technology, Sichuan Agricultural University, Chengdu, China
| | - Liqun Xu
- College of Animal Sciences and Technology, Sichuan Agricultural University, Chengdu, China
| | - Bo Zeng
- College of Animal Sciences and Technology, Sichuan Agricultural University, Chengdu, China
| | - Kang Lai
- Sichuan Province Apiculture Management Station, Chengdu, China
| | - Mingxian Yang
- College of Animal Sciences and Technology, Sichuan Agricultural University, Chengdu, China
| | - Diyan Li
- College of Animal Sciences and Technology, Sichuan Agricultural University, Chengdu, China
| | - Ye Zhao
- College of Animal Sciences and Technology, Sichuan Agricultural University, Chengdu, China
| | - Mingwang Zhang
- College of Animal Sciences and Technology, Sichuan Agricultural University, Chengdu, China
| | - Debing Li
- College of Animal Sciences and Technology, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
18
|
Li S, Hu J, Yao H, Geng F, Nie S. Interaction between four galactans with different structural characteristics and gut microbiota. Crit Rev Food Sci Nutr 2021:1-11. [PMID: 34669541 DOI: 10.1080/10408398.2021.1992605] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Human gut microbiota played a key role in maintaining and regulating host health. Gut microbiota composition could be altered by daily diet and related nutrients. Diet polysaccharide, an important dietary nutrient, was one kind of biological macromolecules linked by the glycosidic bonds. Galactans were widely used in foods due to their gelling, thickening and stabilizing properties. Recently, effects of different galactans on gut microbiota have attracted much attention. This review described the structural characteristics of 4 kinds of galactans, including porphyran, agarose, carrageenan, and arabinogalactan, along with the effects of different galactans on gut microbiota and production of short-chain fatty acids. The ability of gut microbiota to utilize galactans with different structural characteristics and related degradation mechanism were also summarized. All these four galactans could be used by gut Bacteroides. Besides, the porphyran could be utilized by Lactobacillus and Bifidobacterium, while the arabinogalactan could be utilized by Lactobacillus, Bifidobacterium and Roseburia. Four galactans with significant difference in molecular weight/degree of polymerization, glycosidic linkage, esterification, branching and monosaccharide composition required gut microbes which could utilize them have corresponding genes encoding the corresponding enzymes for decomposition. This review could help to understand the relationship between galactans with different structural characteristics and gut microbiota, and provide information for potential use of galactans as functional foods.
Collapse
Affiliation(s)
- Song Li
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Nanchang University, Nanchang China
| | - Jielun Hu
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Nanchang University, Nanchang China
| | - Haoyingye Yao
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Nanchang University, Nanchang China
| | - Fang Geng
- Key Laboratory of Coarse Cereal Processing (Ministry of Agriculture and Rural Affairs), School of Food and Biological Engineering, Chengdu University, Chengdu, China
| | - Shaoping Nie
- State Key Laboratory of Food Science and Technology, China-Canada Joint Lab of Food Science and Technology (Nanchang), Nanchang University, Nanchang China
| |
Collapse
|
19
|
Ücker M, Ansorge R, Sato Y, Sayavedra L, Breusing C, Dubilier N. Deep-sea mussels from a hybrid zone on the Mid-Atlantic Ridge host genetically indistinguishable symbionts. THE ISME JOURNAL 2021; 15:3076-3083. [PMID: 33972724 PMCID: PMC8443746 DOI: 10.1038/s41396-021-00927-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 01/26/2021] [Accepted: 02/03/2021] [Indexed: 02/04/2023]
Abstract
The composition and diversity of animal microbiomes is shaped by a variety of factors, many of them interacting, such as host traits, the environment, and biogeography. Hybrid zones, in which the ranges of two host species meet and hybrids are found, provide natural experiments for determining the drivers of microbiome communities, but have not been well studied in marine environments. Here, we analysed the composition of the symbiont community in two deep-sea, Bathymodiolus mussel species along their known distribution range at hydrothermal vents on the Mid-Atlantic Ridge, with a focus on the hybrid zone where they interbreed. In-depth metagenomic analyses of the sulphur-oxidising symbionts of 30 mussels from the hybrid zone, at a resolution of single nucleotide polymorphism analyses of ~2500 orthologous genes, revealed that parental and hybrid mussels (F2-F4 generation) have genetically indistinguishable symbionts. While host genetics does not appear to affect symbiont composition in these mussels, redundancy analyses showed that geographic location of the mussels on the Mid-Atlantic Ridge explained most of the symbiont genetic variability compared to the other factors. We hypothesise that geographic structuring of the free-living symbiont population plays a major role in driving the composition of the microbiome in these deep-sea mussels.
Collapse
Affiliation(s)
- Merle Ücker
- grid.419529.20000 0004 0491 3210Max Planck Institute for Marine Microbiology, Bremen, Germany ,grid.7704.40000 0001 2297 4381MARUM—Center for Marine Environmental Sciences of the University of Bremen, Bremen, Germany
| | - Rebecca Ansorge
- grid.419529.20000 0004 0491 3210Max Planck Institute for Marine Microbiology, Bremen, Germany ,grid.40368.390000 0000 9347 0159Quadram Institute Bioscience, Norwich, Norfolk UK
| | - Yui Sato
- grid.419529.20000 0004 0491 3210Max Planck Institute for Marine Microbiology, Bremen, Germany
| | - Lizbeth Sayavedra
- grid.419529.20000 0004 0491 3210Max Planck Institute for Marine Microbiology, Bremen, Germany ,grid.40368.390000 0000 9347 0159Quadram Institute Bioscience, Norwich, Norfolk UK
| | - Corinna Breusing
- grid.20431.340000 0004 0416 2242University of Rhode Island, Graduate School of Oceanography, Narragansett, RI USA
| | - Nicole Dubilier
- grid.419529.20000 0004 0491 3210Max Planck Institute for Marine Microbiology, Bremen, Germany ,grid.7704.40000 0001 2297 4381MARUM—Center for Marine Environmental Sciences of the University of Bremen, Bremen, Germany
| |
Collapse
|
20
|
Russell JT, Zhou Y, Weinstock GM, Bubier JA. The Gut Microbiome and Substance Use Disorder. Front Neurosci 2021; 15:725500. [PMID: 34531718 PMCID: PMC8439419 DOI: 10.3389/fnins.2021.725500] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 08/12/2021] [Indexed: 01/15/2023] Open
Abstract
Substance use disorders (SUDs) remain a significant public health challenge, affecting tens of millions of individuals worldwide each year. Often comorbid with other psychiatric disorders, SUD can be poly-drug and involve several different substances including cocaine, opiates, nicotine, and alcohol. SUD has a strong genetic component. Much of SUD research has focused on the neurologic and genetic facets of consumption behavior. There is now interest in the role of the gut microbiome in the pathogenesis of SUD. In this review, we summarize current animal and clinical evidence that the gut microbiome is involved in SUD, then address the underlying mechanisms by which the gut microbiome interacts with SUD through metabolomic, immune, neurological, and epigenetic mechanisms. Lastly, we discuss methods using various inbred and outbred mice models to gain an integrative understanding of the microbiome and host genetic controls in SUD.
Collapse
Affiliation(s)
- Jordan T Russell
- School of Medicine, University of Connecticut Health Center, Farmington, CT, United States
| | - Yanjiao Zhou
- School of Medicine, University of Connecticut Health Center, Farmington, CT, United States
| | - George M Weinstock
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, United States
| | | |
Collapse
|
21
|
Maglione A, Zuccalà M, Tosi M, Clerico M, Rolla S. Host Genetics and Gut Microbiome: Perspectives for Multiple Sclerosis. Genes (Basel) 2021; 12:1181. [PMID: 34440354 PMCID: PMC8394267 DOI: 10.3390/genes12081181] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/28/2021] [Accepted: 07/28/2021] [Indexed: 12/12/2022] Open
Abstract
As a complex disease, Multiple Sclerosis (MS)'s etiology is determined by both genetic and environmental factors. In the last decade, the gut microbiome has emerged as an important environmental factor, but its interaction with host genetics is still unknown. In this review, we focus on these dual aspects of MS pathogenesis: we describe the current knowledge on genetic factors related to MS, based on genome-wide association studies, and then illustrate the interactions between the immune system, gut microbiome and central nervous system in MS, summarizing the evidence available from Experimental Autoimmune Encephalomyelitis mouse models and studies in patients. Finally, as the understanding of influence of host genetics on the gut microbiome composition in MS is in its infancy, we explore this issue based on the evidence currently available from other autoimmune diseases that share with MS the interplay of genetic with environmental factors (Inflammatory Bowel Disease, Rheumatoid Arthritis and Systemic Lupus Erythematosus), and discuss avenues for future research.
Collapse
Affiliation(s)
- Alessandro Maglione
- Department of Clinical and Biological Sciences, University of Torino, 10100 Torino, Italy; (A.M.); (M.C.)
| | - Miriam Zuccalà
- Department of Health Sciences, Center on Autoimmune and Allergic Diseases (CAAD), Università del Piemonte Orientale, 28100 Novara, Italy; (M.Z.); (M.T.)
| | - Martina Tosi
- Department of Health Sciences, Center on Autoimmune and Allergic Diseases (CAAD), Università del Piemonte Orientale, 28100 Novara, Italy; (M.Z.); (M.T.)
| | - Marinella Clerico
- Department of Clinical and Biological Sciences, University of Torino, 10100 Torino, Italy; (A.M.); (M.C.)
| | - Simona Rolla
- Department of Clinical and Biological Sciences, University of Torino, 10100 Torino, Italy; (A.M.); (M.C.)
| |
Collapse
|
22
|
Grant C, Loman B, Bailey M, Pyter L. Manipulations of the gut microbiome alter chemotherapy-induced inflammation and behavioral side effects in female mice. Brain Behav Immun 2021; 95:401-412. [PMID: 33895287 PMCID: PMC8461613 DOI: 10.1016/j.bbi.2021.04.014] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 04/06/2021] [Accepted: 04/13/2021] [Indexed: 12/27/2022] Open
Abstract
Chemotherapy treatment is associated with acute behavioral side effects (fatigue, anorexia) that significantly reduce patient quality of life and are dose-limiting, thereby increasing mortality (Kidwell et al., 2014). Disruptions to gut homeostasis (diarrhea, constipation, microbial dysbiosis) are also observed in patients receiving chemotherapy. In non-oncological patients, facets of mental health (fatigue, anxiety, depression) correlate with alterations in the gut microbiome, suggestive of a contribution of the gut in CNS disease etiology. The potential gut-to-brain pathway is poorly understood in patients receiving chemotherapy. Our prior studies have demonstrated a correlation between chemotherapy treatment, gut changes, peripheral and central inflammation, and behavioral symptoms in mice. Here we aimed to determine the extent to which chemotherapy-associated gut manipulations modulate the behavioral and biological consequences of chemotherapy. We measured sickness behaviors, peripheral and central inflammatory mediators, and anxiety in conventional or germ-free female mice: 1) cohabitating with mice of the opposite treatment group, 2) pre-treated with broad-spectrum antibiotics, or 3) given an intra-gastric gavage of gut content from chemotherapy-treated mice. In cohabitation studies, presumed coprophagia promoted body mass recovery, however strong associations with inflammation and behavior were not observed. Reduction of gut microbial alpha diversity via antibiotics did not prevent chemotherapy-associated side effects, however the relative abundances of the genera Tyzzerella, Romboutsia, and Turicibacter correlated with circulating inflammatory (IL-1β) and behavioral outcomes (lethargy, anxiety-like behavior). A gut microbiota transplant from chemotherapy-treated mice decreased central locomotion in open field testing, increased circulating CXCL1, and increased hippocampal Il6 and Tnfa in germ-free mice compared to germ-free mice that received a transplant from vehicle-treated mice. Taken together, these data provide further evidence that the gut microbiota likely contributes to the development of chemotherapy-associated side effects. This work has significant implications in the future treatment of anxiety in patients, and warrants future studies using microbe-based treatment options.
Collapse
Affiliation(s)
- C.V. Grant
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, Ohio, USA
| | - B.R. Loman
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children’s Hospital, Columbus, Ohio, USA
| | - M.T. Bailey
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, Ohio, USA.,Center for Microbial Pathogenesis, The Research Institute at Nationwide Children’s Hospital, Columbus, Ohio, USA.,Department of Pediatrics, The Ohio State University, Columbus, Ohio, USA
| | - L.M. Pyter
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, Ohio, USA.,Department of Psychiatry and Behavioral Health, Ohio State University, Columbus, Ohio, USA.,Department of Neuroscience, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
23
|
Badger R, Aho K, Serve K. Short-term exposure to synthetic flaxseed lignan LGM2605 alters gut microbiota in mice. Microbiologyopen 2021; 10:e1185. [PMID: 33970540 PMCID: PMC8087944 DOI: 10.1002/mbo3.1185] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 03/24/2021] [Accepted: 03/24/2021] [Indexed: 12/11/2022] Open
Abstract
LGM2605 is a synthetic version of the naturally occurring flaxseed lignan secoisolariciresinol diglucoside (SDG), with known anti‐inflammatory and antioxidant properties; however, its effects on gut microbial composition have not previously been evaluated. In the present study, we sought to determine how the 10‐day oral administration of LGM2605 alters the gut microbiota of mice. Eight‐week‐old female C57BL/6 mice were treated with either LGM2605 or saline, administered daily via oral gavage over a 10‐day treatment period. Upon termination of treatment, mouse cecums (n = 31) were collected, and cecal DNA was isolated. 16S rRNA genes were sequenced and analyzed in Mothur to identify changes in gut microbial composition induced by LGM2605 treatment (v. saline control). We then assessed community composition, performed indicator taxa analysis, and measured alpha and beta diversity. Overall, LGM2605 significantly altered the gut microbiota of mice; we reported alterations in 3 bacterial phyla and 22 genera as a result of treatment. The study here identifies for the first time significant alterations in the gut microbiota of mice following oral administration of LGM2605, in general shifting toward a more anti‐inflammatory composition. These findings lay the foundation for future investigations utilizing LGM2605 to control gut dysbiosis and, by extension, systemic inflammation.
Collapse
Affiliation(s)
- Reagan Badger
- Department of Biological Sciences, Idaho State University, Pocatello, ID, USA
| | - Ken Aho
- Department of Biological Sciences, Idaho State University, Pocatello, ID, USA
| | - Kinta Serve
- Department of Biological Sciences, Idaho State University, Pocatello, ID, USA
| |
Collapse
|
24
|
Strickland BA, Patel MC, Shilts MH, Boone HH, Kamali A, Zhang W, Stylos D, Boukhvalova MS, Rosas-Salazar C, Yooseph S, Rajagopala SV, Blanco JCG, Das SR. Microbial community structure and composition is associated with host species and sex in Sigmodon cotton rats. Anim Microbiome 2021; 3:29. [PMID: 33863395 PMCID: PMC8051552 DOI: 10.1186/s42523-021-00090-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 03/20/2021] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The cotton rat (genus Sigmodon) is an essential small animal model for the study of human infectious disease and viral therapeutic development. However, the impact of the host microbiome on infection outcomes has not been explored in this model, partly due to the lack of a comprehensive characterization of microbial communities across different cotton rat species. Understanding the dynamics of their microbiome could significantly help to better understand its role when modeling viral infections in this animal model. RESULTS We examined the bacterial communities of the gut and three external sites (skin, ear, and nose) of two inbred species of cotton rats commonly used in research (S. hispidus and S. fulviventer) by using 16S rRNA gene sequencing, constituting the first comprehensive characterization of the cotton rat microbiome. We showed that S. fulviventer maintained higher alpha diversity and richness than S. hispidus at external sites (skin, ear, nose), but there were no differentially abundant genera. However, S. fulviventer and S. hispidus had distinct fecal microbiomes composed of several significantly differentially abundant genera. Whole metagenomic shotgun sequencing of fecal samples identified species-level differences between S. hispidus and S. fulviventer, as well as different metabolic pathway functions as a result of differential host microbiome contributions. Furthermore, the microbiome composition of the external sites showed significant sex-based differences while fecal communities were not largely different. CONCLUSIONS Our study shows that host genetic background potentially exerts homeostatic pressures, resulting in distinct microbiomes for two different inbred cotton rat species. Because of the numerous studies that have uncovered strong relationships between host microbiome, viral infection outcomes, and immune responses, our findings represent a strong contribution for understanding the impact of different microbial communities on viral pathogenesis. Furthermore, we provide novel cotton rat microbiome data as a springboard to uncover the full therapeutic potential of the microbiome against viral infections.
Collapse
Affiliation(s)
- Britton A Strickland
- Pathology Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Mira C Patel
- Sigmovir Biosystems Inc., 9610 Medical Center Drive, Suite 100, Rockville, MD, 20850, USA
- Present Address: Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Meghan H Shilts
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Helen H Boone
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Arash Kamali
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Wei Zhang
- Sigmovir Biosystems Inc., 9610 Medical Center Drive, Suite 100, Rockville, MD, 20850, USA
| | - Daniel Stylos
- Sigmovir Biosystems Inc., 9610 Medical Center Drive, Suite 100, Rockville, MD, 20850, USA
| | - Marina S Boukhvalova
- Sigmovir Biosystems Inc., 9610 Medical Center Drive, Suite 100, Rockville, MD, 20850, USA
| | | | - Shibu Yooseph
- Department of Computer Science, Genomics and Bioinformatics Cluster, University of Central Florida, Orlando, FL, USA
| | | | - Jorge C G Blanco
- Sigmovir Biosystems Inc., 9610 Medical Center Drive, Suite 100, Rockville, MD, 20850, USA.
| | - Suman R Das
- Pathology Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department of Otolaryngology, Vanderbilt University Medical Center, Nashville, TN, USA.
- Division of Infectious Diseases, Vanderbilt University Medical Center, 1211 21st Avenue South, S2108 Medical Center North, Nashville, TN, 37232, USA.
| |
Collapse
|
25
|
Gut microbiota, determined by dietary nutrients, drive modification of the plasma lipid profile and insulin resistance. iScience 2021; 24:102445. [PMID: 33997711 PMCID: PMC8105675 DOI: 10.1016/j.isci.2021.102445] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 03/13/2021] [Accepted: 04/14/2021] [Indexed: 12/12/2022] Open
Abstract
The gut microbiota metabolizes the nutrients to produce various metabolites that play crucial roles in host metabolism. However, the links between the microbiota established by different nutrients and the microbiota-influenced changes in the plasma lipids remain unclear. Diets rich in cornstarch, fructose, branched chain amino acids, soybean oil (SO), or lard established a unique microbiota and had influence on glucose metabolism, which was partially reproduced by transferring the microbiota. Comparison of plasma lipidomic analysis between germ-free and colonized mice revealed significant impacts of the microbiota on various lipid classes, and of note, the microbiota established by the SO diet, which was associated with the greatest degree of glucose intolerance, caused the maximum alteration of the plasma lipid profile. Thus, the gut microbiota composed of dietary nutrients was associated with dynamic changes in the lipids potentially having differential effects on glucose metabolism. Diets with different nutrient compositions differentially affect glucose metabolism Gut microbiota established by soybean oil-rich (SO) diet impairs glucose metabolism Gut microbiota established by diets has dynamic effects on the plasma lipid profile SO diet has the greatest impact on the plasma lipid profile through gut microbiota
Collapse
|
26
|
Evidence of a hormonal reshuffle in the cecal metabolome fingerprint of a strain of rats resistant to decompression sickness. Sci Rep 2021; 11:8317. [PMID: 33859311 PMCID: PMC8050073 DOI: 10.1038/s41598-021-87952-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 04/07/2021] [Indexed: 02/02/2023] Open
Abstract
On one side, decompression sickness (DCS) with neurological disorders lead to a reshuffle of the fecal metabolome from rat caecum. On the other side, there is high inter-individual variability in terms of occurrence of DCS. One could wonder whether the fecal metabolome could be linked to the DCS-susceptibility. We decided to study male and female rats selected for their resistance to decompression sickness, and we hypothesize a strong impregnation concerning the fecal metabolome. The aim is to verify whether the rats resistant to the accident have a fecal metabolomic signature different from the stem generations sensitive to DCS. 39 DCS-resistant animals (21 females and 18 males), aged 14 weeks, were compared to 18 age-matched standard Wistar rats (10 females and 8 males), i.e., the same as those we used for the founding stock. Conventional and ChemRICH approaches helped the metabolomic interpretation of the 226 chemical compounds analyzed in the cecal content. Statistical analysis shows a panel of 81 compounds whose expression had changed following the selection of rats based on their resistance to DCS. 63 compounds are sex related. 39 are in common. This study shows the spectral fingerprint of the fecal metabolome from the caecum of a strain of rats resistant to decompression sickness. This study also confirms a difference linked to sex in the metabolome of non-selected rats, which disappear with selective breeding. Results suggest hormonal and energetic reshuffle, including steroids sugars or antibiotic compounds, whether in the host or in the microbial community.
Collapse
|
27
|
The acidified drinking water-induced changes in the behavior and gut microbiota of wild-type mice depend on the acidification mode. Sci Rep 2021; 11:2877. [PMID: 33536529 PMCID: PMC7858586 DOI: 10.1038/s41598-021-82570-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 01/18/2021] [Indexed: 02/06/2023] Open
Abstract
Acidification of drinking water to a pH between 2.5 and 3.0 is widely used to prevent the spread of bacterial diseases in animal colonies. Besides hydrochloric acid (HCl), sulfuric acid (H2SO4) is also used to acidify drinking water. Here we examined the effects of H2SO4-acidified drinking water (pH = 2.8) received from weaning (postnatal day 21) on the behavior and gut microflora of 129S6/SvEv mice, a mouse strain commonly used in transgenic studies. In contrast to HCl-acidified water, H2SO4-acidified water only temporarily impaired the pole-descending ability of mice (at 3 months of age), and did not change the performance in an accelerating rotarod test. As compared to 129S6/SvEv mice receiving non-acidified or HCl-acidified drinking water, the gut microbiota of 129S6/SvEv mice on H2SO4-acidified water displayed significant alterations at every taxonomic level especially at 6 months of age. Our results demonstrate that the effects of acidified drinking water on the behavior and gut microbiota of 129S6/SvEv mice depends on the acid used for acidification. To shed some light on how acidified drinking water affects the physiology of 129S6/SvEv mice, we analyzed the serum and fecal metabolomes and found remarkable, acidified water-induced alterations.
Collapse
|
28
|
Establishment of murine gut microbiota in gnotobiotic mice. iScience 2021; 24:102049. [PMID: 33537656 PMCID: PMC7840467 DOI: 10.1016/j.isci.2021.102049] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/19/2020] [Accepted: 01/06/2021] [Indexed: 12/11/2022] Open
Abstract
Determining whether associations between gut microbiota characteristics and host physiology represent causal relationships is a fundamental challenge for microbiome research. We report a detailed investigation of microbiome assembly in C57BL/6 germ-free mice across a period of 70 days and compare the effects of single and multiple rounds of gavage, using both native and antibiotic-disrupted murine donor material. Recipients of the native microbiota did not achieve compositional stability until day 28 and persistent differences to donor microbiota remained until day 70. Performing multiple rounds of gavage significantly increased the cumulative number of detected taxa (mean increase: 10.4%) and compositional similarity to donor, and significantly reduced within-group variance (p < 0.05). Multiple rounds of gavage with antibiotic-disrupted microbiota provided no substantial benefit in relation to compositional similarity to donor or within-group variance. The process of donor microbiota establishment in recipient animals is necessary before experimentation commences and is considerably influenced by donor microbiota characteristics. Gut microbiota in germ-free mice stabilize 28 days after native microbiota gavage Repeated gavage of native microbiota increases similarity to donor microbiota Establishment of antibiotic-disrupted microbiota does not improve with repeated gavage Donor microbiota characteristics influence fidelity of microbiota re-establishment
Collapse
|
29
|
Gheorghe CE, Ritz NL, Martin JA, Wardill HR, Cryan JF, Clarke G. Investigating causality with fecal microbiota transplantation in rodents: applications, recommendations and pitfalls. Gut Microbes 2021; 13:1941711. [PMID: 34328058 PMCID: PMC8331043 DOI: 10.1080/19490976.2021.1941711] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 06/02/2021] [Accepted: 06/04/2021] [Indexed: 02/04/2023] Open
Abstract
In recent years, studies investigating the role of the gut microbiota in health and diseases have increased enormously - making it essential to deepen and question the research methodology employed. Fecal microbiota transplantation (FMT) in rodent studies (either from human or animal donors) allows us to better understand the causal role of the intestinal microbiota across multiple fields. However, this technique lacks standardization and requires careful experimental design in order to obtain optimal results. By comparing several studies in which rodents are the final recipients of FMT, we summarize the common practices employed. In this review, we document the limitations of this method and highlight different parameters to be considered while designing FMT Studies. Standardizing this method is challenging, as it differs according to the research topic, but avoiding common pitfalls is feasible. Several methodological questions remain unanswered to this day and we offer a discussion on issues to be explored in future studies.
Collapse
Affiliation(s)
- Cassandra E. Gheorghe
- Department of Psychiatry and Neurobehavioral Science, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Nathaniel L. Ritz
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Jason A. Martin
- Department of Psychiatry and Neurobehavioral Science, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Hannah R. Wardill
- Precision Medicine, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, Australia
- Adelaide Medical School, the University of Adelaide, Adelaide, Australia
| | - John F. Cryan
- Department of Psychiatry and Neurobehavioral Science, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Gerard Clarke
- Department of Psychiatry and Neurobehavioral Science, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- INFANT Research Centre, University College Cork, Cork, Ireland
| |
Collapse
|
30
|
Subramanian M, Wojtusciszyn A, Favre L, Boughorbel S, Shan J, Letaief KB, Pitteloud N, Chouchane L. Precision medicine in the era of artificial intelligence: implications in chronic disease management. J Transl Med 2020; 18:472. [PMID: 33298113 PMCID: PMC7725219 DOI: 10.1186/s12967-020-02658-5] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 12/02/2020] [Indexed: 02/07/2023] Open
Abstract
Aberrant metabolism is the root cause of several serious health issues, creating a huge burden to health and leading to diminished life expectancy. A dysregulated metabolism induces the secretion of several molecules which in turn trigger the inflammatory pathway. Inflammation is the natural reaction of the immune system to a variety of stimuli, such as pathogens, damaged cells, and harmful substances. Metabolically triggered inflammation, also called metaflammation or low-grade chronic inflammation, is the consequence of a synergic interaction between the host and the exposome-a combination of environmental drivers, including diet, lifestyle, pollutants and other factors throughout the life span of an individual. Various levels of chronic inflammation are associated with several lifestyle-related diseases such as diabetes, obesity, metabolic associated fatty liver disease (MAFLD), cancers, cardiovascular disorders (CVDs), autoimmune diseases, and chronic lung diseases. Chronic diseases are a growing concern worldwide, placing a heavy burden on individuals, families, governments, and health-care systems. New strategies are needed to empower communities worldwide to prevent and treat these diseases. Precision medicine provides a model for the next generation of lifestyle modification. This will capitalize on the dynamic interaction between an individual's biology, lifestyle, behavior, and environment. The aim of precision medicine is to design and improve diagnosis, therapeutics and prognostication through the use of large complex datasets that incorporate individual gene, function, and environmental variations. The implementation of high-performance computing (HPC) and artificial intelligence (AI) can predict risks with greater accuracy based on available multidimensional clinical and biological datasets. AI-powered precision medicine provides clinicians with an opportunity to specifically tailor early interventions to each individual. In this article, we discuss the strengths and limitations of existing and evolving recent, data-driven technologies, such as AI, in preventing, treating and reversing lifestyle-related diseases.
Collapse
Affiliation(s)
- Murugan Subramanian
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, USA.,Genetic Intelligence Laboratory, Weill Cornell Medicine-Qatar, Qatar Foundation, Doha, Qatar
| | - Anne Wojtusciszyn
- Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital, Lausanne, Switzerland
| | - Lucie Favre
- Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital, Lausanne, Switzerland
| | - Sabri Boughorbel
- Clinical Bioinformatics Section, Research Division, Sidra Medicine, Doha, Qatar
| | - Jingxuan Shan
- Genetic Intelligence Laboratory, Weill Cornell Medicine-Qatar, Qatar Foundation, Doha, Qatar.,Department of Genetic Medicine, Weill Cornell Medicine, 45 E 69th Street, Suite 432, New York, NY, 10021, USA
| | - Khaled B Letaief
- Department of Electronic and Computer Engineering, Hong Kong University of Science and Technology, Kowloon, Hong Kong
| | - Nelly Pitteloud
- Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital, Lausanne, Switzerland.
| | - Lotfi Chouchane
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, USA. .,Genetic Intelligence Laboratory, Weill Cornell Medicine-Qatar, Qatar Foundation, Doha, Qatar. .,Department of Genetic Medicine, Weill Cornell Medicine, 45 E 69th Street, Suite 432, New York, NY, 10021, USA.
| |
Collapse
|
31
|
Kiyan Y, Tkachuk S, Rong S, Gorrasi A, Ragno P, Dumler I, Haller H, Shushakova N. TLR4 Response to LPS Is Reinforced by Urokinase Receptor. Front Immunol 2020; 11:573550. [PMID: 33362762 PMCID: PMC7757075 DOI: 10.3389/fimmu.2020.573550] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 10/26/2020] [Indexed: 12/18/2022] Open
Abstract
GPI-anchored uPAR is the receptor for the extracellular serine protease urokinase-type plasminogen activator (uPA). Though uPAR role in inflammatory processes is documented, underlying mechanisms are not fully understood. In this study we demonstrate that uPAR is a part of Toll-like receptor 4 (TLR4) interactome. Downregulation of uPAR expression resulted in diminished LPS-induced TLR4 signaling, less activation of NFκB, and decreased secretion of inflammatory mediators in myeloid and non-myeloid cells in vitro. In vivo uPAR−/− mice demonstrated better survival, strongly diminished inflammatory response and better organ functions in cecal ligation and puncture mouse polymicrobial sepsis model. Mechanistically, GPI-uPAR and soluble uPAR colocalized with TLR4 on the cell membrane and interacted with scavenger receptor CD36. Our data show that uPAR can interfere with innate immunity response via TLR4 and this mechanism represents a potentially important target in inflammation and sepsis therapy.
Collapse
Affiliation(s)
- Yulia Kiyan
- Nephrology Department, Hannover Medical School, Hannover, Germany
| | - Sergey Tkachuk
- Nephrology Department, Hannover Medical School, Hannover, Germany
| | | | | | - Pia Ragno
- Department of Chemistry and Biology, University of Salerno, Salerno, Italy
| | - Inna Dumler
- Nephrology Department, Hannover Medical School, Hannover, Germany
| | - Hermann Haller
- Nephrology Department, Hannover Medical School, Hannover, Germany
| | - Nelli Shushakova
- Nephrology Department, Hannover Medical School, Hannover, Germany.,Phenos GmbH, Hannover, Germany
| |
Collapse
|
32
|
Mateos-Hernández L, Obregón D, Maye J, Borneres J, Versille N, de la Fuente J, Estrada-Peña A, Hodžić A, Šimo L, Cabezas-Cruz A. Anti-Tick Microbiota Vaccine Impacts Ixodes ricinus Performance during Feeding. Vaccines (Basel) 2020; 8:E702. [PMID: 33233316 PMCID: PMC7711837 DOI: 10.3390/vaccines8040702] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 11/16/2020] [Accepted: 11/18/2020] [Indexed: 01/04/2023] Open
Abstract
The tick microbiota is a highly complex ensemble of interacting microorganisms. Keystone taxa, with a central role in the microbial networks, support the stability and fitness of the microbial communities. The keystoneness of taxa in the tick microbiota can be inferred from microbial co-occurrence networks. Microbes with high centrality indexes are highly connected with other taxa of the microbiota and are expected to provide important resources to the microbial community and/or the tick. We reasoned that disturbance of vector microbiota by removal of ubiquitous and abundant keystone bacteria may disrupt the tick-microbiota homeostasis causing harm to the tick host. These observations and reasoning prompted us to test the hypothesis that antibodies targeting keystone bacteria may harm the ticks during feeding on immunized hosts. To this aim, in silico analyses were conducted to identify keystone bacteria in the microbiota of Ixodes nymphs. The family Enterobacteriaceae was among the top keystone taxa identified in Ixodes microbiota. Immunization of α-1,3-galactosyltransferase-deficient-C57BL/6 (α1,3GT KO) mice with a live vaccine containing the Enterobacteriaceae bacterium Escherichia coli strain BL21 revealed that the production of anti-E. coli and anti-α-Gal IgM and IgG was associated with high mortality of I. ricinus nymphs during feeding. However, this effect was absent in two different strains of wild type mice, BALB/c and C57BL/6. This result concurred with a wide distribution of α-1,3-galactosyltransferase genes, and possibly α-Gal, in Enterobacteriaceae and other bacteria of tick microbiota. Interestingly, the weight of I. ricinus nymphs that fed on E. coli-immunized C57BL/6 was significantly higher than the weight of ticks that fed on C57BL/6 immunized with a mock vaccine. Our results suggest that anti-tick microbiota vaccines are a promising tool for the experimental manipulation of vector microbiota, and potentially the control of ticks and tick-borne pathogens.
Collapse
Affiliation(s)
- Lourdes Mateos-Hernández
- UMR BIPAR, INRAE, ANSES, Ecole Nationale Vétérinaire d’Alfort, Université Paris-Est, Marie Curie, 94706 Maisons-Alfort, France;
| | - Dasiel Obregón
- School of Environmental Sciences, University of Guelph, Guelph, ON N1G 2W1, Canada;
- Center for Nuclear Energy in Agriculture, University of São Paulo, Piracicaba 13400-970, Brazil
| | - Jennifer Maye
- SEPPIC Paris La Défense, 92250 La Garenne Colombes, France; (J.M.); (J.B.); (N.V.)
| | - Jeremie Borneres
- SEPPIC Paris La Défense, 92250 La Garenne Colombes, France; (J.M.); (J.B.); (N.V.)
| | - Nicolas Versille
- SEPPIC Paris La Défense, 92250 La Garenne Colombes, France; (J.M.); (J.B.); (N.V.)
| | - José de la Fuente
- SaBio, Instituto de Investigación en Recursos Cinegéticos (IREC-CSIC-UCLM-JCCM), 13005 Ciudad Real, Spain;
- Department of Veterinary Pathobiology, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK 74078, USA
| | | | - Adnan Hodžić
- Institute of Parasitology, Department of Pathobiology, University of Veterinary Medicine Vienna, Vienna 1210, Austria;
| | - Ladislav Šimo
- UMR BIPAR, INRAE, ANSES, Ecole Nationale Vétérinaire d’Alfort, Université Paris-Est, Marie Curie, 94706 Maisons-Alfort, France;
| | - Alejandro Cabezas-Cruz
- UMR BIPAR, INRAE, ANSES, Ecole Nationale Vétérinaire d’Alfort, Université Paris-Est, Marie Curie, 94706 Maisons-Alfort, France;
| |
Collapse
|
33
|
Marietta E, Mangalam AK, Taneja V, Murray JA. Intestinal Dysbiosis in, and Enteral Bacterial Therapies for, Systemic Autoimmune Diseases. Front Immunol 2020; 11:573079. [PMID: 33193357 PMCID: PMC7655733 DOI: 10.3389/fimmu.2020.573079] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 10/05/2020] [Indexed: 12/19/2022] Open
Abstract
Recent studies have shown that a number of common autoimmune diseases have perturbations of their intestinal microbiome (dysbiosis). These include: Celiac Disease (CeD), Multiple Sclerosis (MS), Rheumatoid Arthritis (RA), Sjogren’s Syndrome (SS), and Type 1 diabetes (T1D). All of these have intestinal microbiomes that are different from healthy controls. There have been numerous studies using animal models of single probiotics (monoclonal) or mixtures of probiotics (polyclonal) and even complete microbiota transfer (fecal microbial transfer-FMT) to inhibit or delay the onset of autoimmune diseases such as the aforementioned common ones. However, proportionally, fewer clinical trials have utilized monoclonal therapies or FMT than polyclonal therapies for treating autoimmune diseases, even though bacterial mono-therapies do inhibit the development of autoimmune diseases and/or delay the onset of autoimmune diseases in rodent models of those autoimmune diseases. In this review then, we review the previously completed and currently ongoing clinical trials that are testing bacterial therapies (FMT, monoclonal, and polyclonal) to treat common autoimmune dseases and discuss the successes in using bacterial monotherapies to treat rodent models of these common autoimmune diseases.
Collapse
Affiliation(s)
- Eric Marietta
- Department of Gastroenterology and Hepatology, Mayo Clinic Rochester, Rochester, MN, United States.,Department of Immunology, Mayo Clinic Rochester, Rochester, MN, United States.,Department of Dermatology, Mayo Clinic Rochester, Rochester, MN, United States
| | | | - Veena Taneja
- Department of Immunology, Mayo Clinic Rochester, Rochester, MN, United States
| | - Joseph A Murray
- Department of Gastroenterology and Hepatology, Mayo Clinic Rochester, Rochester, MN, United States.,Department of Immunology, Mayo Clinic Rochester, Rochester, MN, United States
| |
Collapse
|
34
|
Cahana I, Iraqi FA. Impact of host genetics on gut microbiome: Take-home lessons from human and mouse studies. Animal Model Exp Med 2020; 3:229-236. [PMID: 33024944 PMCID: PMC7529332 DOI: 10.1002/ame2.12134] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 08/23/2020] [Accepted: 08/23/2020] [Indexed: 12/19/2022] Open
Abstract
The intestinal microbiome has emerged as an important component involved in various diseases. Therefore, the interest in understanding the factors shaping its composition is growing. The gut microbiome, often defined as a complex trait, contains diverse components and its properties are determined by a combination of external and internal effects. Although much effort has been invested so far, it is still difficult to evaluate the extent to which human genetics shape the composition of the gut microbiota. However, in mouse studies, where the environmental factors are better controlled, the effect of the genetic background was significant. The purpose of this paper is to provide a current assessment of the role of human host genetics in shaping the gut microbiome composition. Despite the inconsistency of the reported results, it can be estimated that the genetic factor affects a portion of the microbiome. However, this effect is currently lower than the initial estimates, and it is difficult to separate the genetic influence from the environmental effect. Additionally, despite the differences between the microbial composition of humans and mice, results from mouse models can strengthen our knowledge of host genetics underlying the human gut microbial variation.
Collapse
Affiliation(s)
- Inbal Cahana
- Department of Human Microbiology and ImmunologySackler Faculty of MedicineTel‐Aviv UniversityTel‐AvivIsrael
| | - Fuad A. Iraqi
- Department of Human Microbiology and ImmunologySackler Faculty of MedicineTel‐Aviv UniversityTel‐AvivIsrael
| |
Collapse
|
35
|
Park JC, Im SH. Of men in mice: the development and application of a humanized gnotobiotic mouse model for microbiome therapeutics. Exp Mol Med 2020; 52:1383-1396. [PMID: 32908211 PMCID: PMC8080820 DOI: 10.1038/s12276-020-0473-2] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 06/08/2020] [Accepted: 06/16/2020] [Indexed: 12/11/2022] Open
Abstract
Considerable evidence points to the critical role of the gut microbiota in physiology and disease. The administration of live microbes as a therapeutic modality is increasingly being considered. However, key questions such as how to identify candidate microorganisms and which preclinical models are relevant to recapitulate human microbiota remain largely unanswered. The establishment of a humanized gnotobiotic mouse model through the fecal microbiota transplantation of human feces into germ-free mice provides an innovative and powerful tool to mimic the human microbial system. However, numerous considerations are required in designing such a model, as various elements, ranging from the factors pertaining to human donors to the mouse genetic background, affect how microbes colonize the gut. Thus, it is critical to match the murine context to that of human donors to provide a continuous and faithful progression of human flora in mice. This is of even greater importance when the need for accuracy and reproducibility across global research groups are taken into account. Here, we review the key factors that affect the formulation of a humanized mouse model representative of the human gut flora and propose several approaches as to how researchers can effectively design such models for clinical relevance.
Collapse
Affiliation(s)
- John Chulhoon Park
- Department of Life Sciences, Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang, Gyeongbuk, 37673, Republic of Korea
| | - Sin-Hyeog Im
- Department of Life Sciences, Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang, Gyeongbuk, 37673, Republic of Korea.
- ImmunoBiome Inc. POSTECH Biotech Center, Pohang, 37673, Republic of Korea.
| |
Collapse
|
36
|
Abstract
The gut microbiota plays important roles in the host’s normal function and health, including protection against colonization by pathogenic bacteria. Alterations in the gut microbial profile can potentially serve as an early diagnostic tool, as well as a therapeutic strategy against colonization by and carriage of harmful bacteria, including antibiotic-resistant pathogens. Here, we show that the microbiota of hospitalized patients demonstrated specific taxa which differed between carriers of carbapenem-resistant Enterobacteriaceae (CRE) and noncarriers. The difference in the microbiota also dictates alterations in microbiome-specific metabolic capabilities, in association with increased prevalence of systemic infection. Reintroducing specific strains and/or correction of dysbiosis with probiotics or fecal transplantation may potentially lead to colonization by bacterial taxa responsible for protection against or depletion of antibiotic-resistant pathogens. Infection with carbapenem-resistant Enterobacteriaceae (CRE) has become an important challenge in health care settings and a growing concern worldwide. Since infection is preceded by colonization, an understanding of the latter may reduce CRE infections. We aimed to characterize the gut microbiota in CRE carriers, assuming that microbiota alterations precede CRE colonization. We evaluated the gut microbiota using 16S rRNA gene sequencing extracted of fecal samples collected from hospitalized CRE carriers and two control groups, hospitalized noncarriers and healthy adults. The microbiota diversity and composition in CRE-colonized patients differed from those of the control group participants. These CRE carriers displayed lower phylogenetic diversity and dysbiotic microbiota, enriched with members of the family Enterobacteriaceae. Concurrent with the enrichment in Enterobacteriaceae, a depletion of anaerobic commensals was observed. Additionally, changes in several predicted metabolic pathways were observed for the CRE carriers. Concomitantly, we found higher prevalence of bacteremia in the CRE carriers. Several clinical factors that might induce changes in the microbiota were examined and found to be insignificant between the groups. The compositional and functional changes in the microbiota of CRE-colonized patients are associated with increased risk for systemic infection. Our study results provide justification for attempts to restore the dysbiotic microbiota with probiotics or fecal transplantation. IMPORTANCE The gut microbiota plays important roles in the host’s normal function and health, including protection against colonization by pathogenic bacteria. Alterations in the gut microbial profile can potentially serve as an early diagnostic tool, as well as a therapeutic strategy against colonization by and carriage of harmful bacteria, including antibiotic-resistant pathogens. Here, we show that the microbiota of hospitalized patients demonstrated specific taxa which differed between carriers of carbapenem-resistant Enterobacteriaceae (CRE) and noncarriers. The difference in the microbiota also dictates alterations in microbiome-specific metabolic capabilities, in association with increased prevalence of systemic infection. Reintroducing specific strains and/or correction of dysbiosis with probiotics or fecal transplantation may potentially lead to colonization by bacterial taxa responsible for protection against or depletion of antibiotic-resistant pathogens.
Collapse
|
37
|
Walsh J, Olavarria-Ramirez L, Lach G, Boehme M, Dinan TG, Cryan JF, Griffin BT, Hyland NP, Clarke G. Impact of host and environmental factors on β-glucuronidase enzymatic activity: implications for gastrointestinal serotonin. Am J Physiol Gastrointest Liver Physiol 2020; 318:G816-G826. [PMID: 32146834 DOI: 10.1152/ajpgi.00026.2020] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The gastrointestinal tract houses a reservoir of bacterial-derived enzymes that can directly catalyze the metabolism of drugs, dietary elements and endogenous molecules. Both host and environmental factors may influence this enzymatic activity, with the potential to dictate the availability of the biologically-active form of endogenous molecules in the gut and influence inter-individual variation in drug metabolism. We aimed to investigate the influence of the microbiota, and the modulation of its composition, on fecal enzymatic activity. Intrinsic factors related to the host, including age, sex and genetic background, were also explored. Fecalase, a cell-free extract of feces, was prepared and used in a colorimetric-based assay to quantify enzymatic activity. To demonstrate the functional effects of fecal enzymatic activity, we examined β-glucuronidase-mediated cleavage of serotonin β-d-glucuronide (5-HT-GLU) and the resultant production of free 5-HT by HPLC. As expected, β-glucuronidase and β-glucosidase activity were absent in germ-free mice. Enzymatic activity was significantly influenced by mouse strain and animal species. Sex and age significantly altered metabolic activity with implications for free 5-HT. β-Glucuronidase and β-glucosidase activity remained at reduced levels for nearly two weeks after cessation of antibiotic administration. This effect on fecalase corresponded to significantly lower 5-HT levels as compared with incubation with pre-antibiotic fecalase from the same mice. Dietary targeting of the microbiota using prebiotics did not alter β-glucuronidase or β-glucosidase activity. Our data demonstrate that multiple factors influence the activity of bacterial-derived enzymes which may have potential clinical implications for drug metabolism and the deconjugation of host-produced glucuronides in the gut.NEW & NOTEWORTHY This article explores a comprehensive range of host and environmental factors that introduce variability in the expression of bacterial-derived metabolic enzymes. Our results demonstrate that altered β-glucuronidase activity has implications for the bioavailability of luminal serotonin. The experimental approach employed, fecalase, provides a mechanistic basis and translational platform to further delineate the functional outputs of altered metabolic activity, and the associated physiological effects of microbiota-targeted interventions on host response to drugs and host-produced glucuronides.
Collapse
Affiliation(s)
- Jacinta Walsh
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,School of Pharmacy, University College Cork, Cork, Ireland
| | - Loreto Olavarria-Ramirez
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.,Department of Psychiatry and Neurobehavioral Science, University College Cork, Cork, Ireland
| | - Gilliard Lach
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Marcus Boehme
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Timothy G Dinan
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Psychiatry and Neurobehavioral Science, University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Brendan T Griffin
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,School of Pharmacy, University College Cork, Cork, Ireland
| | - Niall P Hyland
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Physiology, University College Cork, Cork, Ireland
| | - Gerard Clarke
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Psychiatry and Neurobehavioral Science, University College Cork, Cork, Ireland.,Irish Centre for Maternal and Child Health Research (INFANT), University College Cork, Cork, Ireland
| |
Collapse
|
38
|
Soybean Oil Modulates the Gut Microbiota Associated with Atherogenic Biomarkers. Microorganisms 2020; 8:microorganisms8040486. [PMID: 32235412 PMCID: PMC7232217 DOI: 10.3390/microorganisms8040486] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 03/12/2020] [Accepted: 03/25/2020] [Indexed: 12/11/2022] Open
Abstract
During the last few decades there has been a staggering rise in human consumption of soybean-oil (SO). The microbiome and specific taxa composing it are dramatically affected by diet; specifically, by high-fat diets. Increasing evidence indicates the association between dysbiosis and health or disease state, including cardiovascular diseases (CVD) and atherosclerosis pathogenesis in human and animal models. To investigate the effects of high SO intake, C57BL/6 mice were orally supplemented with SO-based emulsion (SOE) for one month, followed by analyses of atherosclerosis-related biomarkers and microbiota profiling by 16S rRNA gene sequencing of fecal DNA. SOE-supplementation caused compositional changes to 64 taxa, including enrichment in Bacteroidetes, Mucispirillum, Prevotella and Ruminococcus, and decreased Firmicutes. These changes were previously associated with atherosclerosis in numerous studies. Among the shifted taxa, 40 significantly correlated with at least one atherosclerosis-related biomarker (FDR < 0.05), while 13 taxa positively correlated with the average of all biomarkers. These microbial alterations also caused a microbial-derived metabolic-pathways shift, including enrichment in different amino-acid metabolic-pathways known to be implicated in CVD. In conclusion, our results demonstrate dysbiosis following SOE supplementation associated with atherosclerosis-related biomarkers. These findings point to the microbiome as a possible mediator to CVD, and it may be implemented into non-invasive diagnostic tools or as potential therapeutic strategies.
Collapse
|
39
|
Sorkin BC, Kuszak AJ, Bloss G, Fukagawa NK, Hoffman FA, Jafari M, Barrett B, Brown PN, Bushman FD, Casper S, Chilton FH, Coffey CS, Ferruzzi MG, Hopp DC, Kiely M, Lakens D, MacMillan JB, Meltzer DO, Pahor M, Paul J, Pritchett-Corning K, Quinney SK, Rehermann B, Setchell KD, Sipes NS, Stephens JM, Taylor DL, Tiriac H, Walters MA, Xi D, Zappalá G, Pauli GF. Improving natural product research translation: From source to clinical trial. FASEB J 2020; 34:41-65. [PMID: 31914647 PMCID: PMC7470648 DOI: 10.1096/fj.201902143r] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 10/12/2019] [Accepted: 10/21/2019] [Indexed: 12/28/2022]
Abstract
While great interest in health effects of natural product (NP) including dietary supplements and foods persists, promising preclinical NP research is not consistently translating into actionable clinical trial (CT) outcomes. Generally considered the gold standard for assessing safety and efficacy, CTs, especially phase III CTs, are costly and require rigorous planning to optimize the value of the information obtained. More effective bridging from NP research to CT was the goal of a September, 2018 transdisciplinary workshop. Participants emphasized that replicability and likelihood of successful translation depend on rigor in experimental design, interpretation, and reporting across the continuum of NP research. Discussions spanned good practices for NP characterization and quality control; use and interpretation of models (computational through in vivo) with strong clinical predictive validity; controls for experimental artefacts, especially for in vitro interrogation of bioactivity and mechanisms of action; rigorous assessment and interpretation of prior research; transparency in all reporting; and prioritization of research questions. Natural product clinical trials prioritized based on rigorous, convergent supporting data and current public health needs are most likely to be informative and ultimately affect public health. Thoughtful, coordinated implementation of these practices should enhance the knowledge gained from future NP research.
Collapse
Affiliation(s)
- Barbara C. Sorkin
- Office of Dietary Supplements, National Institutes of Health (NIH), Bethesda, MD, US
| | - Adam J. Kuszak
- Office of Dietary Supplements, National Institutes of Health (NIH), Bethesda, MD, US
| | - Gregory Bloss
- National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD, US
| | | | | | | | | | - Paula N. Brown
- British Columbia Institute of Technology, Burnaby, British Columbia, Canada
| | | | - Steven Casper
- Office of Dietary Supplement Programs, Center for Food Safety and Applied Nutrition, Food and Drug Administration (FDA), Hyattsville, MD, US
| | - Floyd H. Chilton
- Department of Nutritional Sciences and the BIO5 Institute, University of Arizona, Tucson, AZ, US
| | | | - Mario G. Ferruzzi
- Plants for Human Health Institute, North Carolina State University, Kannapolis, NC, US
| | - D. Craig Hopp
- National Center for Complementary and Integrative Health, NIH, Bethesda, MD, US
| | - Mairead Kiely
- Cork Centre for Vitamin D and Nutrition Research, School of Food and Nutritional Sciences, University College Cork, Ireland
| | - Daniel Lakens
- Eindhoven University of Technology, Eindhoven, Netherlands
| | | | | | | | - Jeffrey Paul
- Drexel Graduate College of Biomedical Sciences, College of Medicine, Evanston, IL, US
| | | | | | - Barbara Rehermann
- National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, US
| | | | - Nisha S. Sipes
- National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC, US
| | | | | | - Hervé Tiriac
- University of California, San Diego, La Jolla, CA, US]
| | - Michael A. Walters
- Institute for Therapeutics Discovery and Development, University of Minnesota, Minneapolis, MN, US
| | - Dan Xi
- Office of Cancer Complementary and Alternative Medicine, National Cancer Institute, NIH, Shady Grove, MD, US
| | | | - Guido F. Pauli
- CENAPT and PCRPS, University of Illinois at Chicago College of Pharmacy, Chicago, IL, US
| |
Collapse
|