1
|
Vazquez Bucheli JE, Lee Y, Kim B, Azevedo NF, Azevedo AS, Todorov SD, Ji Y, Kang H, Holzapfel WH. Use of FISH-FLOW as a Method for the Identification and Quantification of Bacterial Populations. Mol Nutr Food Res 2025; 69:e2400494. [PMID: 39363651 DOI: 10.1002/mnfr.202400494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 08/14/2024] [Indexed: 10/05/2024]
Abstract
The gastrointestinal tract (GIT) harbors the largest group of microbiotas among the microbial communities of the human host. The resident organisms typical of a healthy gut are well adapted to the gastrointestinal environment while alteration of these populations can trigger disorders that may affect the health and well-being of the host. Various investigations have applied different tools to study bacterial communities in the gut and their correlation with gastrointestinal disorders such as inflammatory bowel disease (IBD), obesity, and diabetes. This study proposes fluorescent in situ hybridization, combined with flow cytometry (FISH-FLOW), as an alternative approach for phylum level identification of Firmicutes, Bacteroidetes, Actinobacteria, and Proteobacteria and quantification of target bacteria from the GIT based on analysis of fecal samples, where results are validated by quantitative polymerase chain reaction (qPCR) and 16S ribosomal ribonucleic acid (16s rRNA) sequencing. The results obtained via FISH-FLOW experimental approach show high specificity for the developed probes for hybridization with the target bacteria. The study, therefore, suggests the FISH-FLOW as a reliable method for studying bacterial communities in the gut with results correlating well with those of metagenomic investigations of the same fecal samples.
Collapse
Affiliation(s)
- Jorge Enrique Vazquez Bucheli
- Human Effective Microbes, Department of Advanced Convergence, Handong Global University, Pohang, Gyeongbuk, 37554, Republic of Korea
- Facultad de medicina veterinaria y zootecnia, Universidad Autónoma de Chiapas, Chiapas, Calz. Emiliano Zapata Km. 8, Del Frigorífico, Chiapas, 29060, México
| | - Yuri Lee
- Human Effective Microbes, Department of Advanced Convergence, Handong Global University, Pohang, Gyeongbuk, 37554, Republic of Korea
| | - Bobae Kim
- HEMPharma Inc., Pohang, Gyeongbuk, 37554, Republic of Korea
| | - Nuno F Azevedo
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, rua Dr. Roberto Frias, Porto, 4200-465, Portugal
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, 4200-135, Portugal
- IPATIMUP - Instituto de Patologia e Imunologia Molecular, Universidade do Porto, Porto, 4200-135, Portugal
| | - Andreia S Azevedo
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, rua Dr. Roberto Frias, Porto, 4200-465, Portugal
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, 4200-135, Portugal
- IPATIMUP - Instituto de Patologia e Imunologia Molecular, Universidade do Porto, Porto, 4200-135, Portugal
| | - Svetoslav Dimitrov Todorov
- ProBacLab, Department of Advanced Convergence, Handong Global University, Pohang, Gyeongbuk, 37554, Republic of Korea
- ProBacLab, Laboratório de Microbiologia de Alimentos, Departamento de Alimentos e Nutrição, Experimental, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo (SP), 05501-000, Brazil
| | - Yosep Ji
- HEMPharma Inc., Pohang, Gyeongbuk, 37554, Republic of Korea
| | - Hyeji Kang
- Human Effective Microbes, Department of Advanced Convergence, Handong Global University, Pohang, Gyeongbuk, 37554, Republic of Korea
- HEMPharma Inc., Pohang, Gyeongbuk, 37554, Republic of Korea
| | - Wilhelm H Holzapfel
- Human Effective Microbes, Department of Advanced Convergence, Handong Global University, Pohang, Gyeongbuk, 37554, Republic of Korea
- HEMPharma Inc., Pohang, Gyeongbuk, 37554, Republic of Korea
| |
Collapse
|
2
|
Gustafson KL, McAdams ZL, Russell AL, Dorfmeyer RA, Turner GM, Ericsson AC. Effect size of delayed freezing, diurnal variation, and hindgut location on the mouse fecal microbiome. iScience 2024; 27:109090. [PMID: 38361608 PMCID: PMC10867441 DOI: 10.1016/j.isci.2024.109090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/09/2023] [Accepted: 01/29/2024] [Indexed: 02/17/2024] Open
Abstract
Practical considerations in fecal sample collection for microbiome research include time to sample storage, time of collection, and hindgut position during terminal collections. Here, parallel experiments were performed to investigate the relative effect of these factors on microbiome composition in mice colonized with two different vendor-origin microbiomes. 16S rRNA amplicon sequencing of immediately flash-frozen feces showed no difference in alpha or beta diversity compared to samples incubated up to 9 h at room temperature. Samples collected in the morning showed greater alpha diversity compared to samples collected in the afternoon. While a significant effect of time was detected in all hindgut regions, the effect increased from cecum to distal colon. This study highlights common scenarios in microbiome research that may affect outcome measures of microbial community analysis. However, we demonstrate a relatively low effect size of these technical factors when compared to a primary experimental factor with large intergroup variability.
Collapse
Affiliation(s)
- Kevin L. Gustafson
- University of Missouri (MU) Comparative Medicine Program, Columbia, MO 65201, USA
- Department of Veterinary Pathobiology, MU, Columbia, MO 65201, USA
| | - Zachary L. McAdams
- Molecular Pathogenesis and Therapeutics Program, MU, Columbia, MO 65201, USA
| | - Amber L. Russell
- Department of Veterinary Pathobiology, MU, Columbia, MO 65201, USA
| | - Rebecca A. Dorfmeyer
- MU Metagenomics Center (MUMC), Mutant Mouse Resource and Research Center at the University of Missouri (MU MMRRC), Columbia, MO 65201, USA
| | - Giedre M. Turner
- MU Metagenomics Center (MUMC), Mutant Mouse Resource and Research Center at the University of Missouri (MU MMRRC), Columbia, MO 65201, USA
| | - Aaron C. Ericsson
- University of Missouri (MU) Comparative Medicine Program, Columbia, MO 65201, USA
- Department of Veterinary Pathobiology, MU, Columbia, MO 65201, USA
- Molecular Pathogenesis and Therapeutics Program, MU, Columbia, MO 65201, USA
- MU Metagenomics Center (MUMC), Mutant Mouse Resource and Research Center at the University of Missouri (MU MMRRC), Columbia, MO 65201, USA
| |
Collapse
|
3
|
Jiang D, Goswami R, Dennis M, Heimsath H, Kozlowski PA, Ardeshir A, Van Rompay KKA, De Paris K, Permar SR, Surana NK. Sutterella and its metabolic pathways positively correlate with vaccine-elicited antibody responses in infant rhesus macaques. Front Immunol 2023; 14:1283343. [PMID: 38124733 PMCID: PMC10731017 DOI: 10.3389/fimmu.2023.1283343] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 11/21/2023] [Indexed: 12/23/2023] Open
Abstract
Introduction It is becoming clearer that the microbiota helps drive responses to vaccines; however, little is known about the underlying mechanism. In this study, we aimed to identify microbial features that are associated with vaccine immunogenicity in infant rhesus macaques. Methods We analyzed 16S rRNA gene sequencing data of 215 fecal samples collected at multiple timepoints from 64 nursery-reared infant macaques that received various HIV vaccine regimens. PERMANOVA tests were performed to determine factors affecting composition of the gut microbiota throughout the first eight months of life in these monkeys. We used DESeq2 to identify differentially abundant bacterial taxa, PICRUSt2 to impute metagenomic information, and mass spectrophotometry to determine levels of fecal short-chain fatty acids and bile acids. Results Composition of the early-life gut microbial communities in nursery-reared rhesus macaques from the same animal care facility was driven by age, birth year, and vaccination status. We identified a Sutterella and a Rodentibacter species that positively correlated with vaccine-elicited antibody responses, with the Sutterella species exhibiting more robust findings. Analysis of Sutterella-related metagenomic data revealed five metabolic pathways that significantly correlated with improved antibody responses following HIV vaccination. Given these pathways have been associated with short-chain fatty acids and bile acids, we quantified the fecal concentration of these metabolites and found several that correlated with higher levels of HIV immunogen-elicited plasma IgG. Discussion Our findings highlight an intricate bidirectional relationship between the microbiota and vaccines, where multiple aspects of the vaccination regimen modulate the microbiota and specific microbial features facilitate vaccine responses. An improved understanding of this microbiota-vaccine interplay will help develop more effective vaccines, particularly those that are tailored for early life.
Collapse
Affiliation(s)
- Danting Jiang
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, United States
- Program in Computational Biology and Bioinformatics, Duke University School of Medicine, Durham, NC, United States
| | - Ria Goswami
- Department of Pediatrics, Weill Cornell Medicine, New York, NY, United States
| | - Maria Dennis
- Department of Pediatrics, Weill Cornell Medicine, New York, NY, United States
| | - Holly Heimsath
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - Pamela A. Kozlowski
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Amir Ardeshir
- California National Primate Research Center, University of California, Davis, Davis, CA, United States
| | - Koen K. A. Van Rompay
- California National Primate Research Center, University of California, Davis, Davis, CA, United States
| | - Kristina De Paris
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, United States
| | - Sallie R. Permar
- Department of Pediatrics, Weill Cornell Medicine, New York, NY, United States
| | - Neeraj K. Surana
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, United States
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, United States
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, United States
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, United States
| |
Collapse
|
4
|
Muralitharan RR, Snelson M, Meric G, Coughlan MT, Marques FZ. Guidelines for microbiome studies in renal physiology. Am J Physiol Renal Physiol 2023; 325:F345-F362. [PMID: 37440367 DOI: 10.1152/ajprenal.00072.2023] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 06/28/2023] [Accepted: 07/07/2023] [Indexed: 07/15/2023] Open
Abstract
Gut microbiome research has increased dramatically in the last decade, including in renal health and disease. The field is moving from experiments showing mere association to causation using both forward and reverse microbiome approaches, leveraging tools such as germ-free animals, treatment with antibiotics, and fecal microbiota transplantations. However, we are still seeing a gap between discovery and translation that needs to be addressed, so that patients can benefit from microbiome-based therapies. In this guideline paper, we discuss the key considerations that affect the gut microbiome of animals and clinical studies assessing renal function, many of which are often overlooked, resulting in false-positive results. For animal studies, these include suppliers, acclimatization, baseline microbiota and its normalization, littermates and cohort/cage effects, diet, sex differences, age, circadian differences, antibiotics and sweeteners, and models used. Clinical studies have some unique considerations, which include sampling, gut transit time, dietary records, medication, and renal phenotypes. We provide best-practice guidance on sampling, storage, DNA extraction, and methods for microbial DNA sequencing (both 16S rRNA and shotgun metagenome). Finally, we discuss follow-up analyses, including tools available, metrics, and their interpretation, and the key challenges ahead in the microbiome field. By standardizing study designs, methods, and reporting, we will accelerate the findings from discovery to translation and result in new microbiome-based therapies that may improve renal health.
Collapse
Affiliation(s)
- Rikeish R Muralitharan
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash University, Melbourne, Victoria, Australia
- Institute for Medical Research, Ministry of Health Malaysia, Kuala Lumpur, Malaysia
| | - Matthew Snelson
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Guillaume Meric
- Cambridge-Baker Systems Genomics Initiative, Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia
- Department of Cardiometabolic Health, University of Melbourne, Melbourne, Victoria, Australia
- Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
- Department of Cardiovascular Research Translation and Implementation, La Trobe University, Melbourne, Victoria, Australia
| | - Melinda T Coughlan
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia
| | - Francine Z Marques
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash University, Melbourne, Victoria, Australia
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Victorian Heart Institute, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
5
|
Lampeter T, Love C, Tang TT, Marella AS, Lee HY, Oganyan A, Moffat D, Kareem A, Rusling M, Massmann A, Orr M, Bongiorno C, Yuan LL. Risk of bias assessment tool for systematic review and meta-analysis of the gut microbiome. GUT MICROBIOME (CAMBRIDGE, ENGLAND) 2023; 4:e13. [PMID: 39295908 PMCID: PMC11406368 DOI: 10.1017/gmb.2023.12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 04/10/2023] [Accepted: 07/01/2023] [Indexed: 09/21/2024]
Abstract
Risk of bias assessment is a critical step of any meta-analysis or systematic review. Given the low sample count of many microbiome studies, especially observational or cohort studies involving human subjects, many microbiome studies have low power. This increases the importance of performing meta-analysis and systematic review for microbiome research in order to enhance the relevance and applicability of microbiome results. This work proposes a method based on the ROBINS-I tool to systematically consider sources of bias in microbiome research seeking to perform meta-analysis or systematic review for microbiome studies.
Collapse
Affiliation(s)
- Thomas Lampeter
- New York Institute of Technology College of Osteopathic Medicine, Glen Head, NY, USA
| | - Charles Love
- Des Moines University College of Osteopathic Medicine, Des Moines, IA, USA
| | - Trien T Tang
- Des Moines University College of Osteopathic Medicine, Des Moines, IA, USA
| | - Aditi S Marella
- Des Moines University College of Osteopathic Medicine, Des Moines, IA, USA
| | - Hayden Y Lee
- Des Moines University College of Osteopathic Medicine, Des Moines, IA, USA
| | - Armani Oganyan
- Des Moines University College of Osteopathic Medicine, Des Moines, IA, USA
| | - Devin Moffat
- Des Moines University College of Osteopathic Medicine, Des Moines, IA, USA
| | - Anisha Kareem
- Des Moines University College of Osteopathic Medicine, Des Moines, IA, USA
| | - Matthew Rusling
- Des Moines University College of Osteopathic Medicine, Des Moines, IA, USA
| | - Aubrey Massmann
- Des Moines University College of Osteopathic Medicine, Des Moines, IA, USA
| | - Melanie Orr
- New York Institute of Technology College of Osteopathic Medicine, Glen Head, NY, USA
| | | | - Li-Lian Yuan
- Des Moines University College of Osteopathic Medicine, Des Moines, IA, USA
| |
Collapse
|
6
|
Kim YM, Choi JO, Cho YJ, Hong BK, Shon HJ, Kim BJ, Park JH, Kim WU, Kim D. Mycobacterium potentiates protection from colorectal cancer by gut microbial alterations. Immunology 2023; 168:493-510. [PMID: 36183156 DOI: 10.1111/imm.13586] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 09/26/2022] [Indexed: 11/29/2022] Open
Abstract
Not only are many Mycobacteria pathogens, but they can act as strong non-specific immunopotentiators, generating beneficial effects on the pathogenesis of some diseases. However, there has been no direct evidence of the effect of mycobacterial species on colorectal cancer (CRC). Herein, we showed that there may be a meaningful inverse correlation between the incidence of tuberculosis and CRC based on global statistics and that heat-killed Mycobacterial tuberculosis and live Mycobacterium bovis (Bacillus Calmette-Guérin strain) could ameliorate CRC development. In particular, using a faecal microbiota transplantation and a comparison between separate housing and cohousing, we demonstrated that the gut microbiota is involved in the protective effects. The microbial alterations can be elucidated by the modulation of antimicrobial activities including those of the Reg3 family genes. Furthermore, interleukin-22 production by T helper cells contributed to the anti-inflammatory activity of Mycobacteria. Our results revealed a novel role of Mycobacteria involving gut microbial alterations in dampening inflammation-associated CRC and an immunological mechanism underlying the interaction between microbes and host immunity.
Collapse
Affiliation(s)
- Yu-Mi Kim
- Center for Integrative Rheumatoid Transcriptomics and Dynamics, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jin-Ouk Choi
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yong-Joon Cho
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
- Institute for Basic Science, Seoul, Republic of Korea
| | - Bong-Ki Hong
- Center for Integrative Rheumatoid Transcriptomics and Dynamics, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hoh-Jeong Shon
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Bum-Joon Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Institute of Endemic Diseases, Seoul National University Medical Research Center, Seoul, Republic of Korea
| | - Joo-Hong Park
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Wan-Uk Kim
- Center for Integrative Rheumatoid Transcriptomics and Dynamics, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Republic of Korea
- Division of Rheumatology, Department of Internal Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Donghyun Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Institute of Endemic Diseases, Seoul National University Medical Research Center, Seoul, Republic of Korea
| |
Collapse
|
7
|
High-fat diet and estrogen modulate the gut microbiota in a sex-dependent manner in mice. Commun Biol 2023; 6:20. [PMID: 36624306 PMCID: PMC9829864 DOI: 10.1038/s42003-022-04406-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 12/23/2022] [Indexed: 01/10/2023] Open
Abstract
A high-fat diet can lead to gut microbiota dysbiosis, chronic intestinal inflammation, and metabolic syndrome. Notably, resulting phenotypes, such as glucose and insulin levels, colonic crypt cell proliferation, and macrophage infiltration, exhibit sex differences, and females are less affected. This is, in part, attributed to sex hormones. To investigate if there are sex differences in the microbiota and if estrogenic ligands can attenuate high-fat diet-induced dysbiosis, we used whole-genome shotgun sequencing to characterize the impact of diet, sex, and estrogenic ligands on the microbial composition of the cecal content of mice. We here report clear host sex differences along with remarkably sex-dependent responses to high-fat diet. Females, specifically, exhibited increased abundance of Blautia hansenii, and its levels correlated negatively with insulin levels in both sexes. Estrogen treatment had a modest impact on the microbiota diversity but altered a few important species in males. This included Collinsella aerofaciens F, which we show correlated with colonic macrophage infiltration. In conclusion, male and female mice exhibit clear differences in their cecal microbial composition and in how diet and estrogens impact the composition. Further, specific microbial strains are significantly correlated with metabolic parameters.
Collapse
|
8
|
Prakash PK, Lakshmi A J. Effect of milk protein hydrolysate supplementation on protein energy malnutrition-induced gut dysbiosis. Food Funct 2022; 13:10305-10319. [PMID: 36125286 DOI: 10.1039/d2fo00714b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Dairy proteins in the diet are beneficial for the growth of probiotics; however, what is unknown is the gut-mediated immune responses under protein energy malnutrition (PEM) and if dairy protein hydrolysates can be effective as dietary interventions. This study compares the composition of the gut microbiota of rats with moderate protein deficiency (M.PEM) and severe protein deficiency (S.PEM) induced by feeding 5% and 1% hypoprotein diets, followed by replenishment with buffalo and whey protein hydrolysates. Fecal samples were collected, and the composition of the gut bacteria was analyzed by whole genome sequencing using long-read sequencing. Gene expression studies of the immunomodulatory cytokines involved and quantification of sIgA were carried out. IL-6 and IFN-γ were downregulated by about 0.17 ± 0.06 and 0.12 ± 0.10 fold when supplemented with whey protein hydrolysate in SP-RWC rats and by about 0.02 ± 0.06 and 0.35 ± 0.12 fold when using buffalo milk hydrolysate. The percentage of Firmicutes decreased in M.PEM and S.PEM rats (33.57%, 28.83 versus 47.73% of control at 3 weeks) but increased upon protein replenishment for all three protein sources at the end of nine weeks. The percentage of Bacteroidetes increased to 31.03% in S.PEM-induced rats as against 28.17% in control rats. The relative abundance of Lactobacillus sp. decreased in M.PEM and S.PEM rats while it showed the opposite effect upon protein replenishment. Gut microbiota modulated the pathogenesis of PEM differentially based on protein intervention along with a significant increase in the relative abundance of the keystone Lactobacillus genus.
Collapse
Affiliation(s)
- Pavan Kumar Prakash
- Protein Chemistry and Technology Department, CSIR-Central Food Technological Research Institute, Mysore-570020, India. .,Department of Bioscience, Mangalore University, Mangalagangotri, Mangaluru, India
| | - Jyothi Lakshmi A
- Protein Chemistry and Technology Department, CSIR-Central Food Technological Research Institute, Mysore-570020, India.
| |
Collapse
|
9
|
Lin Q, Kuypers M, Liu Z, Copeland JK, Chan D, Robertson SJ, Kontogiannis J, Guttman DS, Banks EK, Philpott DJ, Mallevaey T. Invariant natural killer T cells minimally influence gut microbiota composition in mice. Gut Microbes 2022; 14:2104087. [PMID: 35912530 PMCID: PMC9348128 DOI: 10.1080/19490976.2022.2104087] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Invariant Natural Killer T (iNKT) cells are unconventional T cells that respond to glycolipid antigens found in microbes in a CD1d-dependent manner. iNKT cells exert innate-like functions and produce copious amounts of cytokines, chemokines and cytotoxic molecules within only minutes of activation. As such, iNKT cells can fuel or dampen inflammation in a context-dependent manner. In addition, iNKT cells provide potent immunity against bacteria, viruses, parasites and fungi. Although microbiota-iNKT cell interactions are not well-characterized, mounting evidence suggests that microbiota colonization early in life impacts iNKT cell homeostasis and functions in disease. In this study, we showed that CD1d-/- and Vα14 Tg mice, which lack and have increased numbers of iNKT cells, respectively, had no significant alterations in gut microbiota composition compared to their littermate controls. Furthermore, specific iNKT cell activation by glycolipid antigens only resulted in a transient and minimal shift in microbiota composition when compared to the natural drift found in our colony. Our findings demonstrate that iNKT cells have little to no influence in regulating commensal bacteria at steady state.Abbreviations: iNKT: invariant Natural Killer T cell; αGC: α-galactosylceramide.
Collapse
Affiliation(s)
- Qiaochu Lin
- Department of Immunology, University of Toronto, Toronto, ON, Canada,CONTACT Thierry Mallevaey University of Toronto, Department of Immunology, Medical Sciences Building, Room 7334,1 King’s College Circle, Toronto, OntarioM5S 1A8, Canada
| | - Meggie Kuypers
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Zhewei Liu
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Julia K Copeland
- Centre for the Analysis of Genome Evolution & Function, University of Toronto, Toronto, Ontario, Canada
| | - Donny Chan
- Centre for the Analysis of Genome Evolution & Function, University of Toronto, Toronto, Ontario, Canada
| | - Susan J Robertson
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Jean Kontogiannis
- Division of Comparative Medicine, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - David S Guttman
- Centre for the Analysis of Genome Evolution & Function, University of Toronto, Toronto, Ontario, Canada,Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - E. Kate Banks
- Division of Comparative Medicine, Faculty of Medicine, University of Toronto, Toronto, ON, Canada,Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Dana J Philpott
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Thierry Mallevaey
- Department of Immunology, University of Toronto, Toronto, ON, Canada,Institute of Biomaterials & Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
10
|
Tanelian A, Nankova B, Miari M, Nahvi RJ, Sabban EL. Resilience or susceptibility to traumatic stress: Potential influence of the microbiome. Neurobiol Stress 2022; 19:100461. [PMID: 35789769 PMCID: PMC9250071 DOI: 10.1016/j.ynstr.2022.100461] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 05/13/2022] [Accepted: 05/15/2022] [Indexed: 11/24/2022] Open
Abstract
Exposure to traumatic stress is a major risk factor for development of neuropsychiatric disorders in a sub-population of individuals, while others remain resilient. The mechanisms and contributing factors differentiating between these phenotypes are still unclear. We hypothesize that inter-individual differences in the microbial composition and function contribute to host resilience or susceptibility to stress-induced psychopathologies. The current study aimed to characterize gut microbial community before and after exposure to traumatic stress in an animal model of PTSD. Sprague-Dawley male rats were randomly divided into unstressed controls and experimental group subjected to Single Prolonged Stress (SPS). After 14 days, behavioral analyses were performed using Open Field, Social Interaction and Elevated Plus Maze tests. Based on the anxiety measures, the SPS group was further subdivided into resilient (SPS-R) and susceptible (SPS–S) cohorts. The animals were sacrificed after the last behavioral test and cecum, colon, hippocampus, and medial prefrontal cortex were dissected. Prior to SPS and immediately after Open Field test, fecal samples were collected from each rat for 16S V3–V4 ribosomal DNA sequencing, whereas urine samples were collected before SPS, 90 min into immobilization and on the day of sacrifice to measure epinephrine and norepinephrine levels. Analyses of the fecal microbiota revealed significant differences in microbial communities and in their predictive functionality among the groups before and after SPS stressors. Before SPS, the SPS-S subgroup harbored microbiota with an overall pro-inflammatory phenotype, whereas SPS-R subgroup had microbiota with an overall anti-inflammatory phenotype, with predictive functional pathways enriched in carbohydrate and lipid metabolism and decreased in amino acid metabolism and neurodegenerative diseases. After SPS, the gut microbial communities and their predictive functionality shifted especially in SPS cohorts, with volatility at the genus level correlating inversely with Anxiety Index. In line with the alterations seen in the gut microbiota, the levels of cecal short chain fatty acids were also altered, with SPS-S subgroup having significantly lower levels of acetate, valerate and caproate. The levels of acetate inversely correlated with Anxiety Index. Interestingly, urinary epinephrine and norepinephrine levels were also higher in the SPS-S subgroup at baseline and during stress, indicative of an altered sympathoadrenal stress axis. Finally, shorter colon (marker of intestinal inflammation) and a lower claudin-5 protein expression (marker for increased blood brain barrier permeability) were observed in the SPS-S subgroup. Taken together, our results suggest microbiota is a potential factor in predisposing subjects either to stress susceptibility or resilience. Moreover, SPS triggered significant shifts in the gut microbiota, their metabolites and brain permeability. These findings could lead to new therapeutic directions for PTSD possibly through the controlled manipulation of gut microbiota. It may enable early identification of individuals more likely to develop prolonged anxiogenic symptoms following traumatic stress. Preexisting individual differences in microbiome relate to host's stress response. Shift in the microbial composition differs in SPS-R and SPS-S subgroups after SPS. Cecal levels of acetate in SPS subgroups correlate inversely with anxiety index. Basal and stress-induced urinary catecholamine levels are higher in SPS-S subgroup. SPS-S subgroup has shorter colon, less cecal SCFA and lower brain TJ protein.
Collapse
|
11
|
Hulme H, Meikle LM, Strittmatter N, Swales J, Hamm G, Brown SL, Milling S, MacDonald AS, Goodwin RJ, Burchmore R, Wall DM. Mapping the Influence of the Gut Microbiota on Small Molecules across the Microbiome Gut Brain Axis. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2022; 33:649-659. [PMID: 35262356 PMCID: PMC9047441 DOI: 10.1021/jasms.1c00298] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 02/28/2022] [Accepted: 02/28/2022] [Indexed: 06/14/2023]
Abstract
Microbes exert influence across the microbiome-gut-brain axis through neurotransmitter production, induction of host immunomodulators, or the release or induction of other microbial or host molecules. Here, we used mass spectrometry imaging (MSI), a label-free imaging tool, to map molecular changes in the gut and brain in germ-free, antibiotic-treated and control mice. We determined spatial distribution and relative quantification of neurotransmitters and their precursors in response to the microbiome. Using untargeted MSI, we detected a significant change in the levels of four identified small molecules in the brains of germ-free animals compared to controls. However, antibiotic treatment induced no significant changes in these same metabolites in the brain after 1 week of treatment. This work exemplifies the utility of MSI as a tool for the study of known and discovery of novel, mediators of microbiome-gut-brain axis communication.
Collapse
Affiliation(s)
- Heather Hulme
- Institute
of Infection, Immunity and Inflammation, College of Medical, Veterinary
and Life Sciences, Sir Graeme Davies Building, University of Glasgow, Glasgow G12 8TA, United Kingdom
| | - Lynsey M. Meikle
- Institute
of Infection, Immunity and Inflammation, College of Medical, Veterinary
and Life Sciences, Sir Graeme Davies Building, University of Glasgow, Glasgow G12 8TA, United Kingdom
| | - Nicole Strittmatter
- Imaging
and Data Analytics, Clinical Pharmacology and Safety Sciences, Biopharmaceuticals R&D, AstraZeneca, Cambridge CB4 0WG, U.K.
| | - John Swales
- Imaging
and Data Analytics, Clinical Pharmacology and Safety Sciences, Biopharmaceuticals R&D, AstraZeneca, Cambridge CB4 0WG, U.K.
| | - Gregory Hamm
- Imaging
and Data Analytics, Clinical Pharmacology and Safety Sciences, Biopharmaceuticals R&D, AstraZeneca, Cambridge CB4 0WG, U.K.
| | - Sheila L. Brown
- Lydia
Becker Institute of Immunology and Inflammation, Faculty of Biology,
Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9NT, U.K.
| | - Simon Milling
- Institute
of Infection, Immunity and Inflammation, College of Medical, Veterinary
and Life Sciences, Sir Graeme Davies Building, University of Glasgow, Glasgow G12 8TA, United Kingdom
| | - Andrew S. MacDonald
- Lydia
Becker Institute of Immunology and Inflammation, Faculty of Biology,
Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9NT, U.K.
| | - Richard J.A. Goodwin
- Imaging
and Data Analytics, Clinical Pharmacology and Safety Sciences, Biopharmaceuticals R&D, AstraZeneca, Cambridge CB4 0WG, U.K.
| | - Richard Burchmore
- Institute
of Infection, Immunity and Inflammation, College of Medical, Veterinary
and Life Sciences, Sir Graeme Davies Building, University of Glasgow, Glasgow G12 8TA, United Kingdom
| | - Daniel M. Wall
- Institute
of Infection, Immunity and Inflammation, College of Medical, Veterinary
and Life Sciences, Sir Graeme Davies Building, University of Glasgow, Glasgow G12 8TA, United Kingdom
| |
Collapse
|
12
|
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
|
13
|
Moossavi S, Arrieta MC, Sanati-Nezhad A, Bishehsari F. Gut-on-chip for ecological and causal human gut microbiome research. Trends Microbiol 2022; 30:710-721. [DOI: 10.1016/j.tim.2022.01.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 01/16/2022] [Accepted: 01/20/2022] [Indexed: 10/19/2022]
|
14
|
Kandel Gambarte PC, Wolansky MJ. The gut microbiota as a biomarker for realistic exposures to pesticides: A critical consideration. Neurotoxicol Teratol 2022; 91:107074. [DOI: 10.1016/j.ntt.2022.107074] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 12/24/2021] [Accepted: 01/10/2022] [Indexed: 10/19/2022]
|
15
|
Decoding gut microbiota by imaging analysis of fecal samples. iScience 2021; 24:103481. [PMID: 34927025 PMCID: PMC8652011 DOI: 10.1016/j.isci.2021.103481] [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: 11/23/2019] [Revised: 09/21/2021] [Accepted: 11/19/2021] [Indexed: 01/09/2023] Open
Abstract
The gut microbiota plays a crucial role in maintaining health. Monitoring the complex dynamics of its microbial population is, therefore, important. Here, we present a deep convolution network that can characterize the dynamic changes in the gut microbiota using low-resolution images of fecal samples. Further, we demonstrate that the microbial relative abundances, quantified via 16S rRNA amplicon sequencing, can be quantitatively predicted by the neural network. Our approach provides a simple and inexpensive method of gut microbiota analysis. A deep convolution network classifies gut microbiota based on fecal sample images Image-based quantitative prediction of gut microbiota composition is demonstrated This result provides a simple and inexpensive method of gut microbiota analysis
Collapse
|
16
|
Cage Environment Regulates Gut Microbiota Independent of Toll-Like Receptors. Infect Immun 2021; 89:e0018721. [PMID: 33941577 DOI: 10.1128/iai.00187-21] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The gut microbiome orchestrates epithelial homeostasis and both local and remote immunological responses. Critical to these regulatory interactions are innate immune receptors termed Toll-like receptors (TLRs). Studies to date have implicated innate immunity and Toll-like receptors in shaping key features of the gut microbiome. However, a variety of biological and environmental variables are also implicated in determining gut microbiota composition. In this report, we hypothesized that cohousing and environment dominated the regulation of the gut microbiota in animal models independent of innate immunity. To determine the importance of these variables, innate immunity, or environment in shaping gut microbiota, we used a randomized cohousing strategy and transgenic TLR-deficient mice. We have found that mice cohoused together by genotype exhibited limited changes over time in the composition of the gut microbiota. However, for mice randomized to cage, we report extensive changes in the gut microbiota, independent of TLR function, whereby the fecal microbiota of TLR-deficient mice converges with that of wild-type mice. TLR5-deficient mice in these experiments exhibit greater susceptibility to comparative changes in the microbiota than other TLR-deficient mice and wild-type mice. Our work has broad implications for the study of innate immunity and host-microbiota interactions. Given the profound impact that gut dysbiosis may have on immunity, this report highlights the potential impact of cohousing on the gut microbiota and indices of inflammation as outcomes in biological models of infectious or inflammatory disease.
Collapse
|
17
|
Long LL, Svenson KL, Mourino AJ, Michaud M, Fahey JR, Waterman L, Vandegrift KL, Adams MD. Shared and distinctive features of the gut microbiome of C57BL/6 mice from different vendors and production sites, and in response to a new vivarium. Lab Anim (NY) 2021; 50:185-195. [PMID: 34127866 DOI: 10.1038/s41684-021-00777-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Accepted: 05/07/2021] [Indexed: 02/08/2023]
Abstract
Animal models play a critical role in establishing causal relationships between gut microbiota and disease. The laboratory mouse is widely used to study the role of microbes in various disorders; however, differences between mouse vendors, genetic lineages and husbandry protocols have been shown to contribute to variation in phenotypes and to non-reproducibility of experimental results. We sought to understand how gut microbiome profiles of mice vary by vendor, vendor production facility and health status upon receipt into an academic facility and how they change over 12 weeks in the new environment. C57BL/6 mice were sourced from two different production sites for each of three different vendors. Mice were shipped to an academic research vivarium, and fresh-catch stool samples were collected from mice immediately from the shipping box upon receipt, and again after 2, 6 and 12 weeks in the new facility. Substantial variation in bacterial proportional abundance was observed among mice from each vendor at the time of receipt, but shared microbes accounted for most sequence reads. Vendor-specific microbes were generally of low abundance. Microbial profiles of mice from all vendors exhibited shifts over time, highlighting the importance of environmental conditions on microbial dynamics. Our results emphasize the need for continued efforts to account for sources of variation in animal models and understand how they contribute to experimental reproducibility.
Collapse
Affiliation(s)
- Lauren L Long
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Karen L Svenson
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME, USA
| | | | - Michael Michaud
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - James R Fahey
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME, USA
| | - Linda Waterman
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | | | - Mark D Adams
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA.
| |
Collapse
|
18
|
Low A, Soh M, Miyake S, Aw VZJ, Feng J, Wong A, Seedorf H. Longitudinal Changes in Diet Cause Repeatable and Largely Reversible Shifts in Gut Microbial Communities of Laboratory Mice and Are Observed across Segments of the Entire Intestinal Tract. Int J Mol Sci 2021; 22:5981. [PMID: 34205981 PMCID: PMC8198505 DOI: 10.3390/ijms22115981] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 05/27/2021] [Accepted: 05/27/2021] [Indexed: 01/04/2023] Open
Abstract
Dietary changes are known to alter the composition of the gut microbiome. However, it is less understood how repeatable and reversible these changes are and how diet switches affect the microbiota in the various segments of the gastrointestinal tract. Here, a treatment group of conventionally raised laboratory mice is subjected to two periods of western diet (WD) interrupted by a period of standard diet (SD) of the same duration. Beta-diversity analyses show that diet-induced microbiota changes are largely reversible (q = 0.1501; PERMANOVA, weighted-UniFrac comparison of the treatment-SD group to the control-SD group) and repeatable (q = 0.032; PERMANOVA, weighted-UniFrac comparison of both WD treatments). Furthermore, we report that diet switches alter the gut microbiota composition along the length of the intestinal tract in a segment-specific manner, leading to gut segment-specific Firmicutes/Bacteroidota ratios. We identified prevalent and distinct Amplicon Sequencing Variants (ASVs), particularly in genera of the recently described Muribaculaceae, along the gut as well as ASVs that are differentially abundant between segments of treatment and control groups. Overall, this study provides insights into the reversibility of diet-induced microbiota changes and highlights the importance of expanding sampling efforts beyond the collections of fecal samples to characterize diet-dependent and segment-specific microbiome differences.
Collapse
Affiliation(s)
- Adrian Low
- Temasek Life Sciences Laboratory, 1 Research Link, Singapore 117604, Singapore; (A.L.); (M.S.); (S.M.); (V.Z.J.A.); (J.F.); (A.W.)
| | - Melissa Soh
- Temasek Life Sciences Laboratory, 1 Research Link, Singapore 117604, Singapore; (A.L.); (M.S.); (S.M.); (V.Z.J.A.); (J.F.); (A.W.)
| | - Sou Miyake
- Temasek Life Sciences Laboratory, 1 Research Link, Singapore 117604, Singapore; (A.L.); (M.S.); (S.M.); (V.Z.J.A.); (J.F.); (A.W.)
| | - Vanessa Zhi Jie Aw
- Temasek Life Sciences Laboratory, 1 Research Link, Singapore 117604, Singapore; (A.L.); (M.S.); (S.M.); (V.Z.J.A.); (J.F.); (A.W.)
| | - Jian Feng
- Temasek Life Sciences Laboratory, 1 Research Link, Singapore 117604, Singapore; (A.L.); (M.S.); (S.M.); (V.Z.J.A.); (J.F.); (A.W.)
| | - Adeline Wong
- Temasek Life Sciences Laboratory, 1 Research Link, Singapore 117604, Singapore; (A.L.); (M.S.); (S.M.); (V.Z.J.A.); (J.F.); (A.W.)
| | - Henning Seedorf
- Temasek Life Sciences Laboratory, 1 Research Link, Singapore 117604, Singapore; (A.L.); (M.S.); (S.M.); (V.Z.J.A.); (J.F.); (A.W.)
- Department of Biological Sciences, National University of Singapore, Singapore 117604, Singapore
| |
Collapse
|
19
|
Cage and maternal effects on the bacterial communities of the murine gut. Sci Rep 2021; 11:9841. [PMID: 33972615 PMCID: PMC8110963 DOI: 10.1038/s41598-021-89185-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 02/05/2021] [Indexed: 01/04/2023] Open
Abstract
Findings from gut microbiome studies are strongly influenced by both experimental and analytical factors that can unintentionally bias their interpretation. Environment is also critical. Both co-housing and maternal effects are expected to affect microbiomes and have the potential to confound other manipulated factors, such as genetics. We therefore analysed microbiome data from a mouse experiment using littermate controls and tested differences among genotypes (wildtype versus colitis prone-mdr1a−/−), gut niches (stool versus mucus), host ages (6 versus 18 weeks), social groups (co-housed siblings of different genotypes) and maternal influence. We constructed a 16S phylogenetic tree from bacterial communities, fitting random forest models using all 428,234 clades identified. Models discriminated all criteria except host genotype, where no community differences were found. Host social groups differed in abundant, low-level, taxa whereas intermediate phylogenetic and abundance scales distinguished ages and niches. Thus, a carefully controlled experiment treating evolutionary clades of microbes equivalently without reference to taxonomy, clearly identifies whether and how gut microbial communities are distinct across ecologically important factors (niche and host age) and other experimental factors, notably cage effects and maternal influence. These findings highlight the importance of considering such environmental factors in future microbiome studies.
Collapse
|
20
|
Lee S, Knotts TA, Goodson ML, Barboza M, Wudeck E, England G, Raybould HE. Metabolic Responses to Butyrate Supplementation in LF- and HF-Fed Mice Are Cohort-Dependent and Associated with Changes in Composition and Function of the Gut Microbiota. Nutrients 2020; 12:nu12113524. [PMID: 33207675 PMCID: PMC7696936 DOI: 10.3390/nu12113524] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/04/2020] [Accepted: 11/11/2020] [Indexed: 12/12/2022] Open
Abstract
The gut microbiota and associated metabolites have emerged as potential modulators of pathophysiological changes in obesity and related metabolic disorders. Butyrate, a product of bacterial fermentation, has been shown to have beneficial effects in obesity and rodent models of diet-induced obesity. Here, we aimed to determine the beneficial effects of butyrate (as glycerol ester of butyrate monobutyrin, MB) supplementation on metabolic phenotype, intestinal permeability and inflammation, feeding behavior, and the gut microbiota in low-fat (LF)- and high-fat (HF)-fed mice. Two cohorts (separated by 2 weeks) of male C57BL/6J mice (n = 24 in each cohort, 6/group/cohort; 6 weeks old) were separated into four weight-matched groups and fed either a LF (10 % fat/kcal) or HF (45% fat/kcal) with or without supplementation of MB (LF/MB or HF/MB) at 0.25% (w/v) in drinking water for 6 weeks. Metabolic phenotypes (body weight and adiposity), intestinal inflammation, feeding behavior, and fecal microbiome and metabolites were measured. Despite identical genetic and experimental conditions, we found marked differences between cohorts in the response (body weight gain, adiposity, and intestinal permeability) to HF-diet and MB. Notably, the composition of the gut microbiota was significantly different between cohorts, characterized by lower species richness and differential abundance of a large number of taxa, including subtaxa from five phyla, including increased abundance of the genera Bacteroides, Proteobacteria, and Parasutterella in cohort 2 compared to cohort 1. These differences may have contributed to the differential response in intestinal permeability to the HF diet in cohort 2. MB supplementation had no significant effect on metabolic phenotype, but there was a trend to protect from HF-induced impairments in intestinal barrier function in cohort 1 and in sensitivity to cholecystokinin (CCK) in both cohorts. These data support the concept that microbiota composition may have a crucial effect on metabolic responses of a host to dietary interventions and highlight the importance of taking steps to ensure reproducibility in rodent studies.
Collapse
Affiliation(s)
- Sunhye Lee
- Department of Anatomy, Physiology, and Cell Biology, University of California Davis School of Veterinary Medicine, Davis, CA 95616, USA; (S.L.); (M.L.G.); (M.B.); (E.W.); (G.E.)
| | - Trina A. Knotts
- Department of Molecular Biosciences, University of California Davis School of Veterinary Medicine, Davis, CA 95616, USA;
| | - Michael L. Goodson
- Department of Anatomy, Physiology, and Cell Biology, University of California Davis School of Veterinary Medicine, Davis, CA 95616, USA; (S.L.); (M.L.G.); (M.B.); (E.W.); (G.E.)
| | - Mariana Barboza
- Department of Anatomy, Physiology, and Cell Biology, University of California Davis School of Veterinary Medicine, Davis, CA 95616, USA; (S.L.); (M.L.G.); (M.B.); (E.W.); (G.E.)
- Department of Chemistry, University of California Davis, Davis, CA 95616, USA
| | - Elyse Wudeck
- Department of Anatomy, Physiology, and Cell Biology, University of California Davis School of Veterinary Medicine, Davis, CA 95616, USA; (S.L.); (M.L.G.); (M.B.); (E.W.); (G.E.)
| | - Grace England
- Department of Anatomy, Physiology, and Cell Biology, University of California Davis School of Veterinary Medicine, Davis, CA 95616, USA; (S.L.); (M.L.G.); (M.B.); (E.W.); (G.E.)
| | - Helen E. Raybould
- Department of Anatomy, Physiology, and Cell Biology, University of California Davis School of Veterinary Medicine, Davis, CA 95616, USA; (S.L.); (M.L.G.); (M.B.); (E.W.); (G.E.)
- Correspondence: ; Tel.: +1-530-754-6555
| |
Collapse
|
21
|
Dynamic and Asymmetric Changes of the Microbial Communities after Cohousing in Laboratory Mice. Cell Rep 2020; 27:3401-3412.e3. [PMID: 31189120 DOI: 10.1016/j.celrep.2019.05.042] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 10/08/2018] [Accepted: 05/10/2019] [Indexed: 12/17/2022] Open
Abstract
Horizontal transmission of the microbiota between different individuals is widely used to normalize the microbiota in laboratory mice. However, little is known about the dynamics of microbial communities and the level of microbiota transmission after cohousing. We extensively analyzed the fecal microbiota in Jackson and Taconic C57BL/6 mice to study horizontal transmission after weaning. Changes in the microbiota were clearly detected on day 3, almost plateaued on day 7, and resulted in near-comparable composition by day 28 after cohousing. Notably, the transmission of bacterial species was asymmetric in kinetics and abundance, resulting in a microbiota that is more similar to that of Jackson mice than Taconic. Several operational taxonomic units (OTUs) increased their abundance rapidly after cohousing in Taconic mice whereas several OTUs including two mucus-associated bacteria increased their abundance with delayed kinetics in Jackson mice. These studies provide insight into the dynamics and normalization of the microbiota during horizontal transmission.
Collapse
|
22
|
Mandal RK, Denny JE, Waide ML, Li Q, Bhutiani N, Anderson CD, Baby BV, Jala VR, Egilmez NK, Schmidt NW. Temporospatial shifts within commercial laboratory mouse gut microbiota impact experimental reproducibility. BMC Biol 2020; 18:83. [PMID: 32620114 PMCID: PMC7334859 DOI: 10.1186/s12915-020-00810-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 06/16/2020] [Indexed: 12/05/2022] Open
Abstract
BACKGROUND Experimental reproducibility in mouse models is impacted by both genetics and environment. The generation of reproducible data is critical for the biomedical enterprise and has become a major concern for the scientific community and funding agencies alike. Among the factors that impact reproducibility in experimental mouse models is the variable composition of the microbiota in mice supplied by different commercial vendors. Less attention has been paid to how the microbiota of mice supplied by a particular vendor might change over time. RESULTS In the course of conducting a series of experiments in a mouse model of malaria, we observed a profound and lasting change in the severity of malaria in mice infected with Plasmodium yoelii; while for several years mice obtained from a specific production suite of a specific commercial vendor were able to clear the parasites effectively in a relatively short time, mice subsequently shipped from the same unit suffered much more severe disease. Gut microbiota analysis of frozen cecal samples identified a distinct and lasting shift in bacteria populations that coincided with the altered response of the later shipments of mice to infection with malaria parasites. Germ-free mice colonized with cecal microbiota from mice within the same production suite before and after this change followed by Plasmodium infection provided a direct demonstration that the change in gut microbiota profoundly impacted the severity of malaria. Moreover, spatial changes in gut microbiota composition were also shown to alter the acute bacterial burden following Salmonella infection, and tumor burden in a lung tumorigenesis model. CONCLUSION These changes in gut bacteria may have impacted the experimental reproducibility of diverse research groups and highlight the need for both laboratory animal providers and researchers to collaborate in determining the methods and criteria needed to stabilize the gut microbiota of animal breeding colonies and research cohorts, and to develop a microbiota solution to increase experimental rigor and reproducibility.
Collapse
Affiliation(s)
- Rabindra K Mandal
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, 40202, USA
- Present Address: Ryan White Center for Pediatric Infectious Diseases and Global Health, Department of Pediatrics, Indiana University School of Medicine, 1044 W. Walnut St., Indianapolis, IN, 46202, USA
| | - Joshua E Denny
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, 40202, USA
| | - Morgan L Waide
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, 40202, USA
| | - Qingsheng Li
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, 40202, USA
| | - Neal Bhutiani
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, 40202, USA
| | - Charles D Anderson
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, 40202, USA
| | - Becca V Baby
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, 40202, USA
| | - Venkatakrishna R Jala
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, 40202, USA
| | - Nejat K Egilmez
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, 40202, USA
| | - Nathan W Schmidt
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, 40202, USA.
- Present Address: Ryan White Center for Pediatric Infectious Diseases and Global Health, Department of Pediatrics, Indiana University School of Medicine, 1044 W. Walnut St., Indianapolis, IN, 46202, USA.
| |
Collapse
|
23
|
Scheelings TF, Moore RJ, Van TTH, Klaassen M, Reina RD. The gut bacterial microbiota of sea turtles differs between geographically distinct populations. ENDANGER SPECIES RES 2020. [DOI: 10.3354/esr01042] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The microbiota of metazoans can be influenced by a variety of factors including diet, environment and genetics. In this study we sampled multiple populations from 2 host species that do not overlap in distribution, in order to test whether their bacterial microbiotas are species-specific or more variable. Intestinal swabs were collected from loggerhead turtles originating from Florida, USA, and Queensland, Australia, as well as from flatback turtles from Crab Island, Queensland, and Port Hedland, Western Australia. We then manually extracted bacterial DNA and used 16S rRNA sequencing to explore bacterial microbial community composition and structure. Our investigation showed that the bacterial microbiota of sea turtles is heavily influenced by geography, with loggerhead turtles originating from the USA and Australia harbouring significantly different bacterial microbial populations in terms of composition. Similarly, we also found that flatback turtles from Crab Island had significantly less diverse microbiotas, with a predominance of the bacterial phylum Firmicutes, in comparison to their genetically similar counterparts from Port Hedland. Factors that may explain these observed differences between populations include host genetics, differences in foraging habitat quality and differences in migratory distance (and thus durations of inappetence) between foraging and breeding grounds. The mechanisms by which these factors may influence bacterial microbial composition of sea turtle gastrointestinal tracts warrants further investigation. The results of this study highlight the importance of interpreting microbiota data of wild animals in the context of geography.
Collapse
Affiliation(s)
- TF Scheelings
- School of Biological Sciences, Monash University, Wellington Rd, Clayton, Victoria 3800, Australia
| | - RJ Moore
- School of Science, RMIT University, Bundoora West Campus, Plenty Rd, Bundoora, Victoria 3083, Australia
| | - TTH Van
- School of Science, RMIT University, Bundoora West Campus, Plenty Rd, Bundoora, Victoria 3083, Australia
| | - M Klaassen
- Centre for Integrative Ecology, Deakin University, Waurn Ponds, Victoria 3216, Australia
| | - RD Reina
- School of Biological Sciences, Monash University, Wellington Rd, Clayton, Victoria 3800, Australia
| |
Collapse
|
24
|
Chen CY, Hsu KC, Yeh HY, Ho HC. Visualizing the effects of antibiotics on the mouse colonic mucus layer. Tzu Chi Med J 2020; 32:145-153. [PMID: 32269946 PMCID: PMC7137371 DOI: 10.4103/tcmj.tcmj_70_19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 04/30/2019] [Accepted: 05/27/2019] [Indexed: 01/18/2023] Open
Abstract
Objective Mucus provides a protective barrier separating sensitive epithelial surfaces from the outside world. The mouse colonic mucus is organized as a bacteria-free inner layer and a bacteria-colonized outer layer. Antibiotic treatments are known to disturb gut microbiota, but their effect on the mucosal barrier is rarely discussed. The aim was to evaluate and visualize the impact of antibiotics on the colonic mucus and the microbial community. Materials and Methods Two sets of experiments were conducted. In the antibiotic experiment, mice orally ingested both streptomycin and bacitracin for 7 days. In the recovery experiment, mice were allowed to recover for 7 days without antibiotics after having received the 7-day antibiotic treatment. Mouse colons were isolated and divided into proximal, middle, and distal parts. Specimens were examined under a transmission electron microscope to identify morphological changes. The gut microbial community was evaluated by analyzing 16S rDNA sequences isolated from the different parts of the mouse colon. Results The antibiotic-treated mice were physiologically normal. However, a significantly increased inner mucus layer in the proximal and middle colon and a dramatic decrease in bacterial numbers in the outer mucus layers were observed. The 16S rDNA compositions showed a similarity in the dominant taxa among different colon sections. While control mice had a diverse microbiota, antibiotic treatments effectively eliminated most of the bacteria, such that the community was dominated by only one genus (Turicibacter or Staphylococcus). Furthermore, following antibiotic withdrawal in treated mice, the thickness of the inner mucus layer returned to control levels, and the microbial community regained a more complex structure, dominated by Firmicutes, Bacteroidetes, and Proteobacteria. Conclusions Our results indicated that antibiotic treatments not only disturbed the microbiota but also altered the structure of the mucus layer. After the withdrawal of antibiotics, the mucus layer was quickly regenerated within days, probably in response to microbial growth. The recolonization by gut inhabitants with diverse ecological roles, such as mucin-degraders and fermenters indicate that the gut ecosystem is functionally sound and highly resilient.
Collapse
Affiliation(s)
- Chun-Yao Chen
- Department of Life Sciences, Tzu Chi University, Hualien, Taiwan
| | - Kai-Chieh Hsu
- Master Program in Medical Physiology, School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Hsuan-Yu Yeh
- Master Program in Medical Physiology, School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Han-Chen Ho
- Department of Anatomy, Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
25
|
Barbee RW, Turner PV. Incorporating Laboratory Animal Science into Responsible Biomedical Research. ILAR J 2019; 60:9-16. [DOI: 10.1093/ilar/ilz017] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 07/20/2019] [Accepted: 08/05/2019] [Indexed: 12/22/2022] Open
Abstract
Abstract
Biomedical research has made great strides in the past century leading to rapid advances in human life expectancy, all derived from improved understanding, prevention, and treatment of many diseases and conditions. Research involving laboratory animals has played a significant role in this medical progress. However, there continues to be controversy surrounding the use of animals in research, and animal models have been questioned regarding their relevance to human conditions. While research fraud and questionable research practices could potentially contribute to this problem, we argue that a relative ignorance of laboratory animal science has contributed to the “uncontrolled vivarium experiment” that runs parallel to the more controlled scientific experiment. Several variables are discussed, including husbandry, animal environment, social housing, and more, that can contribute to this uncontrolled experiment, and that can simultaneously decrease quality of life for rodent test subjects when ignored. An argument is put forward that laboratory animal veterinarians and scientists can and should play an important role in better controlling such variables. Similarly, the laboratory animal veterinarian and scientist should play an important role in responsible science by addressing complex interdisciplinary challenges.
Collapse
Affiliation(s)
- R Wayne Barbee
- Virginia Commonwealth University, Office of Research and Innovation
| | - Patricia V Turner
- Charles River Laboratories Inc., Global Animal Welfare & Training, University of Guelph Pathobiology
| |
Collapse
|
26
|
Chang AM, Liu Q, Hajjar AM, Greer A, McLean JS, Darveau RP. Toll-like receptor-2 and -4 responses regulate neutrophil infiltration into the junctional epithelium and significantly contribute to the composition of the oral microbiota. J Periodontol 2019; 90:1202-1212. [PMID: 31111967 PMCID: PMC6791728 DOI: 10.1002/jper.18-0719] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 01/27/2019] [Accepted: 02/20/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND Oral gingival tissue, especially the junctional epithelium (JE), is constantly exposed to sub-gingival plaque. A key component of gingival health is the regulation of the number of neutrophils that migrate into the gingival crevice to counteract its harmful effects. This report investigates the contribution of innate defense receptors, Toll-like receptor (TLR)2, TLR4, and both (TLR2/4) to the maintenance of neutrophil homeostasis in the JE. METHODS Bacterial composition was analyzed from whole oral swabs collected from 12- to 14-week-old TLR2, TLR4, TLR2/4 double knock-out (KO) mice using a MiSeq platform targeting the V3-V4 region of the 16S ribosomal RNA gene. Mandibles were histologically examined for quantification of neutrophils in the JE and bone loss. Lastly, total bacterial load was quantitated using quantitative real-time PCR. RESULTS Compared with wild-type, all TLR KO mice displayed significantly increased recruitment of neutrophils (P = 0.0079) into the JE. In addition, TLR4 and TLR2/4 KO mice demonstrated a significant increase in the number of bacteria (P = 0.0022 and P = 0.0152, respectively). Lastly, comparative compositional analyses of the oral microbiome revealed that each KO strain harbored unique microbial communities that are distinct from each other but maintained similar levels of alveolar bone. CONCLUSIONS Neutrophil migration into healthy mouse JE does not require TLR2 or TLR4. However, a significant increase in the number of neutrophils as well as a significant change in the oral microbial composition in both TLR2 and TLR4 KO mice demonstrate that these TLRs contribute to the homeostatic relationship between bacteria and the host in healthy mice periodontal tissue.
Collapse
Affiliation(s)
- Ana M. Chang
- Department of Oral Health Sciences, University of Washington School of Dentistry, Seattle, WA 98195
| | - Quanhui Liu
- Department of Periodontics, University of Washington School of Dentistry, Seattle, WA 98195
| | - Adeline M. Hajjar
- Department of Comparative Medicine, University of Washington School of Medicine, Seattle, WA 98195
| | - Ara Greer
- Department of Oral Health Sciences, University of Washington School of Dentistry, Seattle, WA 98195
| | - Jeffrey S. McLean
- Department of Periodontics, University of Washington School of Dentistry, Seattle, WA 98195
| | - Richard P. Darveau
- Department of Periodontics, University of Washington School of Dentistry, Seattle, WA 98195
| |
Collapse
|
27
|
Berry ASF, Kelly BJ, Barnhart D, Kelly DJ, Beiting DP, Baldassano RN, Redding LE. Gut microbiota features associated with Clostridioides difficile colonization in puppies. PLoS One 2019; 14:e0215497. [PMID: 31469837 PMCID: PMC6716646 DOI: 10.1371/journal.pone.0215497] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 08/05/2019] [Indexed: 02/06/2023] Open
Abstract
In people, colonization with Clostridioides difficile, the leading cause of antibiotic-associated diarrhea, has been shown to be associated with distinct gut microbial features, including reduced bacterial community diversity and depletion of key taxa. In dogs, the gut microbiota features that define C. difficile colonization are less well understood. We sought to define the gut microbiota features associated with C. difficile colonization in puppies, a population where the prevalence of C. difficile has been shown to be elevated, and to define the effect of puppy age and litter upon these features and C. difficile risk. We collected fecal samples from weaned (n = 27) and unweaned (n = 74) puppies from 13 litters and analyzed the effects of colonization status, age and litter on microbial diversity using linear mixed effects models. Colonization with C. difficile was significantly associated with younger age, and colonized puppies had significantly decreased bacterial community diversity and differentially abundant taxa compared to non-colonized puppies, even when adjusting for age. C. difficile colonization remained associated with decreased bacterial community diversity, but the association did not reach statistical significance in a mixed effects model incorporating litter as a random effect. Even though litter explained a greater proportion (67%) of the variability in microbial diversity than colonization status, we nevertheless observed heterogeneity in gut microbial community diversity and colonization status within more than half of the litters, suggesting that the gut microbiota contributes to colonization resistance against C. difficile. The colonization of puppies with C. difficile has important implications for the potential zoonotic transfer of this organism to people. The identified associations point to mechanisms by which C. difficile colonization may be reduced.
Collapse
Affiliation(s)
- Alexander S. F. Berry
- Division of Gastroenterology, Hepatology, and Nutrition, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Brendan J. Kelly
- Divisions of Infectious Diseases and Epidemiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Denise Barnhart
- Department of Pathobiology, University of Pennsylvania, School of Veterinary Medicine, Kennett Square, Pennsylvania, United States of America
| | - Donna J. Kelly
- Department of Pathobiology, University of Pennsylvania, School of Veterinary Medicine, Kennett Square, Pennsylvania, United States of America
| | - Daniel P. Beiting
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Robert N. Baldassano
- Division of Gastroenterology, Hepatology, and Nutrition, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
| | - Laurel E. Redding
- Department of Clinical Sciences, University of Pennsylvania, School of Veterinary Medicine, Kennett Square, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
28
|
Murine Genetic Background Has a Stronger Impact on the Composition of the Gut Microbiota than Maternal Inoculation or Exposure to Unlike Exogenous Microbiota. Appl Environ Microbiol 2019; 85:AEM.00826-19. [PMID: 31350316 DOI: 10.1128/aem.00826-19] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 06/29/2019] [Indexed: 01/07/2023] Open
Abstract
The gut microbiota is a complex ecosystem, affected by both environmental factors and host genetics. Here, we aim at uncovering the bacterial taxa whose gut persistence is controlled by host genetic variation. We used a murine model based on inbred lines BALB/c and C57BL/6J and their F1 reciprocal hybrids (♀C57BL/6J × ♂BALB/c; ♀BALB/c × ♂C57BL/6J). To guarantee genetic similarity of F1 offspring, including the sex chromosomes, we used only female mice. Based on 16S rRNA gene sequencing, we found that the genetically different inbred lines present different microbiota, whereas their genetically identical F1 reciprocal hybrids presented similar microbiota. Moreover, the F1 microbial composition differed from that of both parental lines. Twelve taxa were shown to have genetically controlled gut persistence, while none were found to show maternal effects. Nine of these taxa were dominantly inherited by the C57BL/6J line. Cohousing of the parental inbred lines resulted in a temporary and minor shift in microbiota composition, which returned back to the former microbial composition following separation, indicating that each line tends to maintain a unique bacterial signature reflecting the line. Taken together, our findings indicate that mouse genetics has an effect on the microbial composition in the gut, which is greater than maternal effect and continuous exposure to different microbiota of the alternative line. Uncovering the bacterial taxa associated with host genetics and understanding their role in the gut ecosystem could lead to the development of genetically oriented probiotic products, as part of the personalized medicine approach.IMPORTANCE The gut microbiota play important roles for their host. The link between host genetics and their microbial composition has received increasing interest. Using a unique reciprocal cross model, generating genetically similar F1 hybrids with different maternal inoculation, we demonstrate the inheritance of gut persistence of 12 bacterial taxa. No taxa identified as maternally transmitted. Moreover, cohabitation of two genetically different inbred lines did not dramatically affect the microbiota composition. Taken together, our results demonstrate the importance of the genetic effect over maternal inoculation or effect of exposure to unlike exogenous microbiota. These findings may lead to the development of personalized probiotic products, specifically designed according to the genetic makeup.
Collapse
|
29
|
Gonzalez CG, Tankou SK, Cox LM, Casavant EP, Weiner HL, Elias JE. Latent-period stool proteomic assay of multiple sclerosis model indicates protective capacity of host-expressed protease inhibitors. Sci Rep 2019; 9:12460. [PMID: 31462662 PMCID: PMC6713765 DOI: 10.1038/s41598-019-48495-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 08/02/2019] [Indexed: 01/20/2023] Open
Abstract
Diseases are often diagnosed once overt symptoms arise, ignoring the prior latent period when effective prevention may be possible. Experimental autoimmune encephalomyelitis (EAE), a model for multiple sclerosis, exhibits such disease latency, but the molecular processes underlying this asymptomatic period remain poorly characterized. Gut microbes also influence EAE severity, yet their impact on the latent period remains unknown. Here, we show the latent period between immunization and EAE's overt symptom onset is characterized by distinct host responses as measured by stool proteomics. In particular, we found a transient increase in protease inhibitors that inversely correlated with disease severity. Vancomycin administration attenuated both EAE symptoms and protease inhibitor induction potentially by decreasing immune system reactivity, supporting a subset of the microbiota's role in modulating the host's latent period response. These results strengthen previous evidence of proteases and their inhibitors in EAE and highlight the utility stool-omics for revealing complex, dynamic biology.
Collapse
Affiliation(s)
- Carlos G Gonzalez
- Chemical and Systems Biology Department, Stanford University School of Medicine, Stanford, USA
| | - Stephanie K Tankou
- Ann Romney Center for Neurological Diseases, Brigham and Women's Hospital, Harvard School of Medicine, Boston, MA, USA
- Department Of Neurology, Icahn School Of Medicine at Mount Sinai, New York, NY, USA
- Precision Immunology Institute, Icahn School Of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School Of Medicine at Mount Sinai, New York, NY, USA
| | - Laura M Cox
- Ann Romney Center for Neurological Diseases, Brigham and Women's Hospital, Harvard School of Medicine, Boston, MA, USA
| | - Ellen P Casavant
- Chemical and Systems Biology Department, Stanford University School of Medicine, Stanford, USA
| | - Howard L Weiner
- Ann Romney Center for Neurological Diseases, Brigham and Women's Hospital, Harvard School of Medicine, Boston, MA, USA
| | - Joshua E Elias
- Chemical and Systems Biology Department, Stanford University School of Medicine, Stanford, USA.
- Chan Zuckerberg Biohub, San Francisco, California, USA.
| |
Collapse
|
30
|
Bello-Gil D, Audebert C, Olivera-Ardid S, Pérez-Cruz M, Even G, Khasbiullina N, Gantois N, Shilova N, Merlin S, Costa C, Bovin N, Mañez R. The Formation of Glycan-Specific Natural Antibodies Repertoire in GalT-KO Mice Is Determined by Gut Microbiota. Front Immunol 2019; 10:342. [PMID: 30891034 PMCID: PMC6411795 DOI: 10.3389/fimmu.2019.00342] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 02/11/2019] [Indexed: 12/19/2022] Open
Abstract
Gut commensal bacteria are known to have a significant role in regulating the innate and adaptive immune homeostasis. Alterations in the intestinal microbial composition have been associated with several disease states, including autoimmune and inflammatory conditions. However, it is not entirely clear how commensal gut microbiota modulate and contribute to the systemic immunity, and whether circulating elements of the host immune system could regulate the microbiome. Thus, we have studied the diversity and abundance of specific taxons in the gut microbiota of inbred GalT-KO mice during 7 months of animal life by metagenetic high-throughput sequencing (16S rRNA gene, variable regions V3-V5). The repertoire of glycan-specific natural antibodies, obtained by printed glycan array technology, was then associated with the microbial diversity for each animal by metagenome-wide association studies (MWAS). Our data show that the orders clostridiales (most abundant), bacteriodales, lactobacillales, and deferribacterales may be associated with the development of the final repertoire of natural anti-glycan antibodies in GalT-KO mice. The main changes in microbiota diversity (month-2 and month-3) were related to important changes in levels and repertoire of natural anti-glycan antibodies in these mice. Additionally, significant positive and negative associations were found between the gut microbiota and the pattern of specific anti-glycan antibodies. Regarding individual features, the gut microbiota and the corresponding repertoire of natural anti-glycan antibodies showed differences among the examined animals. We also found redundancy in different taxa associated with the development of specific anti-glycan antibodies. Differences in microbial diversity did not, therefore, necessarily influence the overall functional output of the gut microbiome of GalT-KO mice. In summary, the repertoire of natural anti-carbohydrate antibodies may be partially determined by the continuous antigenic stimulation produced by the gut bacterial population of each GalT-KO mouse. Small differences in gut microbiota diversity could determine different repertoire and levels of natural anti-glycan antibodies and consequently might induce different immune responses to pathogens or other potential threats.
Collapse
Affiliation(s)
- Daniel Bello-Gil
- Infectious Pathology and Transplantation Division, Institut d'Investigació Biomèdica de Bellvitge, Barcelona, Spain
| | - Christophe Audebert
- Genes Diffusion, Douai, France.,PEGASE-Biosciences, Institut Pasteur de Lille, Lille, France
| | - Sara Olivera-Ardid
- Infectious Pathology and Transplantation Division, Institut d'Investigació Biomèdica de Bellvitge, Barcelona, Spain
| | - Magdiel Pérez-Cruz
- CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR 8204-CIIL-Centre d'Infection et d'Immunité de Lille, Université de Lille, Lille, France
| | - Gaël Even
- Genes Diffusion, Douai, France.,PEGASE-Biosciences, Institut Pasteur de Lille, Lille, France
| | | | - Nausicaa Gantois
- Lille University, CNRS, Inserm, Lille University Hospital, Pasteur Institute of Lille, U1019 -UMR 8204 -CIIL-Centre d'Infection et d'Immunité de Lille, Lille, France
| | - Nadezhda Shilova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Sophie Merlin
- Genes Diffusion, Douai, France.,PEGASE-Biosciences, Institut Pasteur de Lille, Lille, France
| | - Cristina Costa
- Infectious Pathology and Transplantation Division, Institut d'Investigació Biomèdica de Bellvitge, Barcelona, Spain
| | - Nicolai Bovin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Rafael Mañez
- Infectious Pathology and Transplantation Division, Institut d'Investigació Biomèdica de Bellvitge, Barcelona, Spain.,Intensive Care Department, Bellvitge University Hospital, Barcelona, Spain
| |
Collapse
|
31
|
Maki KA, Diallo AF, Lockwood MB, Franks AT, Green SJ, Joseph PV. Considerations When Designing a Microbiome Study: Implications for Nursing Science. Biol Res Nurs 2019; 21:125-141. [PMID: 30409024 PMCID: PMC6700895 DOI: 10.1177/1099800418811639] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Nurse scientists play an important role in studying complex relationships among human genetics, environmental factors, and the microbiome, all of which can contribute to human health and disease. Therefore, it is essential that they have the tools necessary to execute a successful microbiome research study. The purpose of this article is to highlight important methodological factors for nurse scientists to consider when designing a microbiome study. In addition to considering factors that influence host-associated microbiomes (i.e., microorganisms associated with organisms such as humans, mice, and rats), this manuscript highlights study designs and methods for microbiome analysis. Exemplars are presented from nurse scientists who have incorporated microbiome methods into their program of research. This review is intended to be a resource to guide nursing-focused microbiome research and highlights how study of the microbiome can be incorporated to answer research questions.
Collapse
Affiliation(s)
- Katherine A. Maki
- Department of Biobehavioral Health Science, College of Nursing,
University of Illinois at Chicago, Chicago, IL, USA
| | - Ana F. Diallo
- Institute of Inclusion, Inquiry and Innovation, Richmond Health and
Wellness Clinics, Virginia Commonwealth University School of Nursing, Richmond, VA,
USA
| | - Mark B. Lockwood
- Department of Biobehavioral Health Science, College of Nursing,
University of Illinois at Chicago, Chicago, IL, USA
| | - Alexis T. Franks
- Sensory Science and Metabolism Unit, Biobehavioral Branch, Division
of Intramural Research, National Institute of Nursing Research, National Institutes
of Health, Department of Health and Human Services, Bethesda, MD, USA
| | - Stefan J. Green
- Research Resources Center, University of Illinois at Chicago,
Chicago, IL, USA
| | - Paule V. Joseph
- Sensory Science and Metabolism Unit, Biobehavioral Branch, Division
of Intramural Research, National Institute of Nursing Research, National Institutes
of Health, Department of Health and Human Services, Bethesda, MD, USA
| |
Collapse
|
32
|
Ibrahim A, Hugerth LW, Hases L, Saxena A, Seifert M, Thomas Q, Gustafsson JÅ, Engstrand L, Williams C. Colitis-induced colorectal cancer and intestinal epithelial estrogen receptor beta impact gut microbiota diversity. Int J Cancer 2019; 144:3086-3098. [PMID: 30515752 PMCID: PMC6519213 DOI: 10.1002/ijc.32037] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 11/14/2018] [Accepted: 11/15/2018] [Indexed: 12/18/2022]
Abstract
Chronic inflammation of the colon (colitis) is a risk factor for colorectal cancer (CRC). Hormone‐replacement therapy reduces CRC incidences, and the estrogen receptor beta (ERβ/ESR2) has been implicated in this protection. Gut microbiota is altered in both colitis and CRC and may influence the severity of both. Here we test the hypothesis that intestinal ERβ impacts the gut microbiota. Mice with and without intestine‐specific deletion of ERβ (ERβKOVil) were generated using the Cre‐LoxP system. Colitis and CRC were induced with a single intraperitoneal injection of azoxymethane (AOM) followed by administration of three cycles of dextran sulfate sodium (DSS) in drinking water. The microbiota population were characterized by high‐throughput 16S rRNA gene sequencing of DNA extracted from fecal samples (N = 39). Differences in the microbiota due to AOM/DSS and absence of ERβ were identified through bioinformatic analyses of the 16S‐Seq data, and the distribution of bacterial species was corroborated using qPCR. We demonstrate that colitis‐induced CRC reduced the gut microbiota diversity and that loss of ERβ enhanced this process. Further, the Bacteroidetes genus Prevotellaceae_UCG_001 was overrepresented in AOM/DSS mice compared to untreated controls (3.5‐fold, p = 0.004), and this was enhanced in females and in ERβKOVil mice. Overall, AOM/DSS enriched for microbiota impacting immune system diseases and metabolic functions, and lack of ERβ in combination with AOM/DSS enriched for microbiota impacting carbohydrate metabolism and cell motility, while reducing those impacting the endocrine system. Our data support that intestinal ERβ contributes to a more favorable microbiome that could attenuate CRC development. What's new? Chronic inflammation of the colon is a risk factor for colorectal cancer (CRC). Hormone‐replacement therapy reduces CRC incidence, and the estrogen receptor beta (ERβ/ESR2) has been implicated in this protection. The microbiota of the gut is altered in both colitis and CRC, but whether intestinal ERβ affects gut microbiota remains to be investigated. Here, the authors demonstrate, in a mouse model, that colitis‐induced CRC reduces the gut microbiota diversity and that loss of ERβ enhances this process. The findings could enable novel therapeutic or preventive approaches toward a more favorable microbiome in inflammatory bowel disease and/or colon cancer development.
Collapse
Affiliation(s)
- Ahmed Ibrahim
- Department of Biosciences and Nutrition, Karolinska Institute, Stockholm, Sweden.,Department of Protein Science, KTH Royal Institute of Technology, Science for Life Laboratory, Solna, Sweden
| | - Luisa W Hugerth
- Department of Microbiology, Tumor & Cell Biology, Karolinska Institute, Science for Life Laboratory, Solna, Sweden
| | - Linnea Hases
- Department of Biosciences and Nutrition, Karolinska Institute, Stockholm, Sweden.,Department of Protein Science, KTH Royal Institute of Technology, Science for Life Laboratory, Solna, Sweden
| | - Ashish Saxena
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, TX, USA
| | - Maike Seifert
- Department of Microbiology, Tumor & Cell Biology, Karolinska Institute, Science for Life Laboratory, Solna, Sweden
| | - Quentin Thomas
- Department of Protein Science, KTH Royal Institute of Technology, Science for Life Laboratory, Solna, Sweden
| | - Jan-Åke Gustafsson
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, TX, USA
| | - Lars Engstrand
- Department of Microbiology, Tumor & Cell Biology, Karolinska Institute, Science for Life Laboratory, Solna, Sweden
| | - Cecilia Williams
- Department of Biosciences and Nutrition, Karolinska Institute, Stockholm, Sweden.,Department of Protein Science, KTH Royal Institute of Technology, Science for Life Laboratory, Solna, Sweden
| |
Collapse
|
33
|
Microbial embryonal colonization during pipefish male pregnancy. Sci Rep 2019; 9:3. [PMID: 30626884 PMCID: PMC6327025 DOI: 10.1038/s41598-018-37026-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 11/08/2018] [Indexed: 02/06/2023] Open
Abstract
While originally acquired from the environment, a fraction of the microbiota is transferred from parents to offspring. The immune system shapes the microbial colonization, while commensal microbes may boost host immune defences. Parental transfer of microbes in viviparous animals remains ambiguous, as the two transfer routes (transovarial vs. pregnancy) are intermingled within the maternal body. Pipefishes and seahorses (syngnathids) are ideally suited to disentangle transovarial microbial transfer from a contribution during pregnancy due to their maternal egg production and their unique male pregnancy. We assessed the persistency and the changes in the microbial communities of the maternal and paternal reproductive tracts over proceeding male pregnancy by sequencing microbial 16S rRNA genes of swabs from maternal gonads and brood pouches of non-pregnant and pregnant fathers. Applying parental immunological activation with heat-killed bacteria, we evaluated the impact of parental immunological status on microbial development. Our data indicate that maternal gonads and paternal brood pouches harbor distinct microbial communities, which could affect embryonal development in a sex-specific manner. Upon activation of the immune system, a shift of the microbial community was observed. The activation of the immune system induced the expansion of microbiota richness during late pregnancy, which corresponds to the time point of larval mouth opening, when initial microbial colonization must take place.
Collapse
|
34
|
Abstract
RATIONALE Researchers in psychiatry and neuroscience are increasingly recognizing the importance of gut-brain communication in mental health. Both genetics and environmental factors influence gut microbiota composition and function. This study examines host-microbe signaling at the gastrointestinal barrier to identify bottom-up mechanisms of microbiota-brain communication. OBJECTIVES We examined differences in gut microbiota composition and fecal miRNA profiles in BALB/c and C57BL/6 mice, in relation to gastrointestinal homeostasis and evaluated the response to perturbation of the gut microbiota by broad-spectrum antibiotic treatment. METHODS AND RESULTS Differences in the gut microbiota composition between BALB/c and C57BL/6 mice, evaluated by fecal 16S rRNA gene sequencing, included significant differences in genera Prevotella, Alistipes, Akkermansia, and Ruminococcus. Significant differences in fecal miRNA profiles were determined using the nCounter NanoString platform. A BLASTn analysis identified conserved fecal miRNA target regions in bacterial metagenomes with 14 significant correlations found between fecal miRNA and predicted taxa relative abundance in our dataset. Treatment with broad-spectrum antibiotics for 2 weeks resulted in a host-specific physiological response at the gastrointestinal barrier including a decrease in barrier permeability in BALB/c mice and alterations in the expression of barrier regulating genes in both strains. Genera Parabacteroides and Bacteroides were associated with changes in barrier function. CONCLUSIONS The results of this study provide insight into how specific taxa influence gut barrier integrity and function. More generally, these data in the context of recent published studies makes a significant contribution to our understanding of host-microbe interactions providing new knowledge that can be harnessed by us and others in future mechanistic studies.
Collapse
|
35
|
Lindheim L, Manti M, Fornes R, Bashir M, Czarnewski P, Diaz OE, Seifert M, Engstrand L, Villablanca EJ, Obermayer-Pietsch B, Stener-Victorin E. Reproductive and Behavior Dysfunction Induced by Maternal Androgen Exposure and Obesity Is Likely Not Gut Microbiome-Mediated. J Endocr Soc 2018; 2:1363-1380. [PMID: 30534630 PMCID: PMC6280317 DOI: 10.1210/js.2018-00266] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 09/21/2018] [Indexed: 02/07/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is a common endocrine and metabolic disorder of unclear etiology in women and is characterized by androgen excess, insulin resistance, and mood disorders. The gut microbiome is known to influence conditions closely related with PCOS, and several recent studies have observed changes in the stool microbiome of women with PCOS. The mechanism by which the gut microbiome interacts with PCOS is still unknown. We used a mouse model to investigate if diet-induced maternal obesity and maternal DHT exposure, mimicking the lean and obese PCOS women, cause lasting changes in the gut microbiome of offspring. Fecal microbiome profiles were assessed using Illumina paired-end sequencing of 16S rRNA gene V4 amplicons. We found sex-specific effects of maternal and offspring diet, and maternal DHT exposure on fecal bacterial richness and taxonomic composition. Female offspring exposed to maternal obesity and DHT displayed reproductive dysfunction and anxietylike behavior. Fecal microbiota transplantation from DHT and diet-induced obesity exposed female offspring to wild-type mice did not transfer reproductive dysfunction and did not cause the expected increase in anxietylike behavior in recipients. Maternal obesity and androgen exposure affect the gut microbiome of offspring, but the disrupted estrous cycles and anxietylike behavior are likely not microbiome-mediated.
Collapse
Affiliation(s)
- Lisa Lindheim
- Division of Endocrinology and Diabetology, Medical University Graz, Graz, Austria
| | - Maria Manti
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Romina Fornes
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Mina Bashir
- Division of Endocrinology and Diabetology, Medical University Graz, Graz, Austria
| | - Paulo Czarnewski
- Immunology and Allergy Unit, Department of Medicine, Solna, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - Oscar E Diaz
- Immunology and Allergy Unit, Department of Medicine, Solna, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - Maike Seifert
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
- Clinical Genomics Facility, Science for Life Laboratory, Solna, Sweden
| | - Lars Engstrand
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
- Clinical Genomics Facility, Science for Life Laboratory, Solna, Sweden
| | - Eduardo J Villablanca
- Immunology and Allergy Unit, Department of Medicine, Solna, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | | | | |
Collapse
|
36
|
Wong SY, Coffre M, Ramanan D, Hines MJ, Gomez LE, Peters LA, Schadt EE, Koralov SB, Cadwell K. B Cell Defects Observed in Nod2 Knockout Mice Are a Consequence of a Dock2 Mutation Frequently Found in Inbred Strains. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 201:1442-1451. [PMID: 30012848 PMCID: PMC6103850 DOI: 10.4049/jimmunol.1800014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 06/22/2018] [Indexed: 02/06/2023]
Abstract
Phenotypic differences among substrains of laboratory mice due to spontaneous mutations or pre-existing genetic variation confound the interpretation of targeted mutagenesis experiments and contribute to challenges with reproducibility across institutions. Notably, C57BL/6 Hsd mice and gene-targeted mice that have been backcrossed to this substrain have been reported to harbor a duplication in exons 28 and 29 of Dock2 In this study, we demonstrate the presence of this Dock2 variant in the widely used Nod2-/- mice. Nucleotide-binding oligomerization domain-containing protein 2 (NOD2) is a cytosolic innate immune receptor associated with inflammatory bowel disease susceptibility. Consistent with a role of NOD2 in an immunological disorder, Nod2-/- mice bred at our institution displayed multiple B cell defects including deficiencies in recirculating B cells, marginal zone B cells, and B1a cells in vivo, as well as defects in class switch recombination in vitro. However, we found that these effects are due to the Dock2 variant and are independent of Nod2 deletion. Despite originating from the same gene-targeted founder mice, Nod2-/- mice from another source did not harbor the Dock2 variant or B cell defects. Finally, we show that Dock2-/- mice display the same B cell defects as mice harboring the Dock2 variant, confirming that the variant is a loss-of-function mutation and is sufficient to explain the alterations to the B cell compartment observed in Nod2-/- mice. Our findings highlight the effects of confounding mutations from widely used inbred strains on gene-targeted mice and reveal new functions of DOCK2 in B cells.
Collapse
Affiliation(s)
- Serre-Yu Wong
- Kimmel Center for Biology and Medicine, Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, NY 10016
- Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Maryaline Coffre
- Department of Pathology, New York University School of Medicine, New York, NY 10016
| | - Deepshika Ramanan
- Kimmel Center for Biology and Medicine, Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, NY 10016
| | - Marcus J Hines
- Department of Pathology, New York University School of Medicine, New York, NY 10016
| | - Luis E Gomez
- Kimmel Center for Biology and Medicine, Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, NY 10016
| | - Lauren A Peters
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Sema4, a Mount Sinai Venture, Stamford, CT 06902; and
| | - Eric E Schadt
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Sema4, a Mount Sinai Venture, Stamford, CT 06902; and
| | - Sergei B Koralov
- Department of Pathology, New York University School of Medicine, New York, NY 10016
| | - Ken Cadwell
- Kimmel Center for Biology and Medicine, Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, NY 10016;
- Department of Microbiology, New York University School of Medicine, New York, NY 10016
| |
Collapse
|
37
|
Spacova I, Ceuppens JL, Seys SF, Petrova MI, Lebeer S. Probiotics against airway allergy: host factors to consider. Dis Model Mech 2018; 11:11/7/dmm034314. [PMID: 30037806 PMCID: PMC6078401 DOI: 10.1242/dmm.034314] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The worldwide prevalence of allergic diseases has drastically increased in the past decades. Recent studies underline the importance of microbial exposure for the development of a balanced immune system. Consequently, probiotic bacteria are emerging as a safe and natural strategy for allergy prevention and treatment. However, clinical probiotic intervention studies have so far yielded conflicting results. There is increasing awareness about the importance of host-associated factors that determine whether an individual will respond to a specific probiotic treatment, and it is therefore crucial to promote a knowledge-based instead of an empirical selection of promising probiotic strains and their administration regimen.In this Review, we summarize the insights from animal model studies of allergic disease, which reveal how host-related factors - such as genetic makeup, sex, age and microbiological status - can impact the outcomes of preventive or curative probiotic treatment. We explore why and how these factors can influence the results of probiotic studies and negatively impact the reproducibility in animal experiments. These same factors might profoundly influence the outcomes of human clinical trials, and can potentially explain the conflicting results from probiotic intervention studies. Therefore, we also link these host-related factors to human probiotic study outcomes in the context of airway allergies.
Collapse
Affiliation(s)
- Irina Spacova
- Research Group Environmental Ecology and Applied Microbiology, Department of Bioscience Engineering, University of Antwerp, 2020 Antwerp, Belgium.,Centre of Microbial and Plant Genetics, Department of Microbial and Molecular Systems (M²S), KU Leuven, Belgium
| | - Jan L Ceuppens
- Laboratory of Clinical Immunology, Department of Microbiology and Immunology, KU Leuven, 3000 Leuven, Belgium
| | - Sven F Seys
- Laboratory of Clinical Immunology, Department of Microbiology and Immunology, KU Leuven, 3000 Leuven, Belgium
| | - Mariya I Petrova
- Centre of Microbial and Plant Genetics, Department of Microbial and Molecular Systems (M²S), KU Leuven, Belgium
| | - Sarah Lebeer
- Research Group Environmental Ecology and Applied Microbiology, Department of Bioscience Engineering, University of Antwerp, 2020 Antwerp, Belgium .,Centre of Microbial and Plant Genetics, Department of Microbial and Molecular Systems (M²S), KU Leuven, Belgium
| |
Collapse
|
38
|
Miyoshi J, Leone V, Nobutani K, Musch MW, Martinez-Guryn K, Wang Y, Miyoshi S, Bobe AM, Eren AM, Chang EB. Minimizing confounders and increasing data quality in murine models for studies of the gut microbiome. PeerJ 2018; 6:e5166. [PMID: 30013837 PMCID: PMC6046200 DOI: 10.7717/peerj.5166] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 06/15/2018] [Indexed: 12/28/2022] Open
Abstract
Murine models are widely used to explore host-microbe interactions because of the challenges and limitations inherent to human studies. However, microbiome studies in murine models are not without their nuances. Inter-individual variations in gut microbiota are frequent even in animals housed within the same room. We therefore sought to find an efficient and effective standard operating procedure (SOP) to minimize these effects to improve consistency and reproducibility in murine microbiota studies. Mice were housed in a single room under specific-pathogen free conditions. Soiled cage bedding was routinely mixed weekly and distributed among all cages from weaning (three weeks old) until the onset of the study. Females and males were separated by sex and group-housed (up to five mice/cage) at weaning. 16S rRNA gene analyses of fecal samples showed that this protocol significantly reduced pre-study variability of gut microbiota amongst animals compared to other conventional measures used to normalize microbiota when large experimental cohorts have been required. A significant and consistent effect size was observed in gut microbiota when mice were switched from regular chow to purified diet in both sexes. However, sex and aging appeared to be independent drivers of gut microbial assemblage and should be taken into account in studies of this nature. In summary, we report a practical and effective pre-study SOP for normalizing the gut microbiome of murine cohorts that minimizes inter-individual variability and resolves co-housing problems inherent to male mice. This SOP may increase quality, rigor, and reproducibility of data acquisition and analysis.
Collapse
Affiliation(s)
- Jun Miyoshi
- Department of Medicine, The University of Chicago, Chicago, IL, United States of America
| | - Vanessa Leone
- Department of Medicine, The University of Chicago, Chicago, IL, United States of America
| | - Kentaro Nobutani
- Department of Medicine, The University of Chicago, Chicago, IL, United States of America
| | - Mark W Musch
- Department of Medicine, The University of Chicago, Chicago, IL, United States of America
| | - Kristina Martinez-Guryn
- Department of Medicine, The University of Chicago, Chicago, IL, United States of America.,Biomedical Sciences Program, Midwestern University, Downers Grove, IL, United States of America
| | - Yunwei Wang
- Department of Medicine, The University of Chicago, Chicago, IL, United States of America
| | - Sawako Miyoshi
- Department of Medicine, The University of Chicago, Chicago, IL, United States of America
| | - Alexandria M Bobe
- Department of Medicine, The University of Chicago, Chicago, IL, United States of America
| | - A Murat Eren
- Department of Medicine, The University of Chicago, Chicago, IL, United States of America
| | - Eugene B Chang
- Department of Medicine, The University of Chicago, Chicago, IL, United States of America
| |
Collapse
|
39
|
Turner PV. The role of the gut microbiota on animal model reproducibility. Animal Model Exp Med 2018; 1:109-115. [PMID: 30891555 PMCID: PMC6388061 DOI: 10.1002/ame2.12022] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Accepted: 06/08/2018] [Indexed: 12/24/2022] Open
Abstract
The gut microbiota is composed of approximately 1010-1014 cells, including fungi, bacteria, archaea, protozoa, viruses, and bacteriophages; their genes and their various metabolites were found throughout the gastrointestinal tract. It has co-evolved with each species to assist with day to day bodily functions, such as digestion, metabolism of xenobiotics, development of mucosal immunity and immunomodulation, and protection against invading pathogens. Because of the significant beneficial impact that gut microbiota may have, there is interest in learning more about it and translating these findings into clinical therapies. Results from recent studies characterizing the gut microbiota of various species have demonstrated the range of influences that may affect gut microbiota diversity, including animal strain, obesity, types of enrichment used, bedding and housing methods, treatment with antimicrobials, vendor source, specific animal housing, diet, and intercurrent disease. Relatively little is known about the functional consequences of alterations of the gut microbiota and exactly how changes in richness and diversity of the microbiota translate into changes in health and susceptibility to disease. Furthermore, questions have been raised as to whether germ-free or even ultraclean, barrier-raised mice are relevant models of human disease, given their significantly reduced gut microbiota diversity and complexity compared with conventionally housed mice. In addition, evidence suggests that the specific anatomical location selected for assessing the gut microbiota has a highly significant effect on study outcomes, in that bacterial phyla change significantly along the gastrointestinal tract. This paper will explore animal model reproducibility in light of this information about the gut microbiota.
Collapse
|
40
|
Montonye DR, Ericsson AC, Busi SB, Lutz C, Wardwell K, Franklin CL. Acclimation and Institutionalization of the Mouse Microbiota Following Transportation. Front Microbiol 2018; 9:1085. [PMID: 29892276 PMCID: PMC5985407 DOI: 10.3389/fmicb.2018.01085] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Accepted: 05/07/2018] [Indexed: 01/02/2023] Open
Abstract
Using animal models, the gut microbiota has been shown to play a critical role in the health and disease of many organ systems. Unfortunately, animal model studies often lack reproducibility when performed at different institutions. Previous studies in our laboratory have shown that the gut microbiota of mice can vary with a number of husbandry factors leading us to speculate that differing environments may alter gut microbiota, which in turn may influence animal model phenotypes. As an extension of these studies, we hypothesized that the shipping of mice from a mouse producer to an institution will result in changes in the type, relative abundance, and functional composition of the gut microbiota. Furthermore, we hypothesized that mice will develop a microbiota unique to the institution and facility in which they are housed. To test these hypotheses, mice of two strains (C57BL/6J and BALB/cJ), two age groups (4 week and 8 week old), and originating from two types of housing (research animal facility under conventional housing and production facilities under maximum barrier housing) were obtained from The Jackson Laboratory. Fecal samples were collected the day prior to shipping, immediately upon arrival, and then on days 2, 5, 7, and weeks 2, 4, and 9 post-arrival. Following the first post-arrival fecal collection, mice were separated into 2 groups and housed at different facilities at our institution while keeping their caging, diet, and husbandry practices the same. DNA was extracted from the collected fecal pellets and 16S rRNA amplicons were sequenced in order to characterize the type and relative abundance of gut bacteria. Principal component analysis (PCA) and permutational multivariate analysis of variance (PERMANOVA) demonstrated that both the shipping and the institution and facility in which mice were housed altered the gut microbiota. Phylogenetic investigation of communities by reconstruction of unobserved states (PICRUSt) predicted differences in functional composition in the gut microbiota of mice based on time of acclimation.
Collapse
Affiliation(s)
- Dan R Montonye
- Comparative Medicine Program, Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, United States
| | - Aaron C Ericsson
- Comparative Medicine Program, Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, United States.,University of Missouri Metagenomics Center, University of Missouri, Columbia, MO, United States.,University of Missouri Mutant Mouse Resource and Research Center, University of Missouri, Columbia, MO, United States
| | - Susheel B Busi
- Comparative Medicine Program, Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, United States
| | - Cathleen Lutz
- The Jackson Laboratory, Bar Harbor, ME, United States
| | | | - Craig L Franklin
- Comparative Medicine Program, Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, United States.,University of Missouri Metagenomics Center, University of Missouri, Columbia, MO, United States.,University of Missouri Mutant Mouse Resource and Research Center, University of Missouri, Columbia, MO, United States
| |
Collapse
|
41
|
Llop P, Latorre A, Moya A. Experimental Epidemiology of Antibiotic Resistance: Looking for an Appropriate Animal Model System. Microbiol Spectr 2018; 6:10.1128/microbiolspec.mtbp-0007-2016. [PMID: 29637886 PMCID: PMC11633557 DOI: 10.1128/microbiolspec.mtbp-0007-2016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Indexed: 02/07/2023] Open
Abstract
Antibiotic resistance is recognized as one of the major challenges in public health. The global spread of antibiotic resistance is the consequence of a constant flow of information across multi-hierarchical interactions, involving cellular (clones), subcellular (resistance genes located in plasmids, transposons, and integrons), and supracellular (clonal complexes, genetic exchange communities, and microbiotic ensembles) levels. In order to study such multilevel complexity, we propose to establish an experimental epidemiology model for the transmission of antibiotic resistance with the cockroach Blatella germanica. This paper reports the results of five types of preliminary experiments with B. germanica populations that allow us to conclude that this animal is an appropriate model for experimental epidemiology: (i) the composition, transmission, and acquisition of gut microbiota and endosymbionts; (ii) the effect of different diets on gut microbiota; (iii) the effect of antibiotics on host fitness; (iv) the evaluation of the presence of antibiotic resistance genes in natural- and lab-reared populations; and (v) the preparation of plasmids harboring specific antibiotic resistance genes. The basic idea is to have populations with higher and lower antibiotic exposure, simulating the hospital and the community, respectively, and with a certain migration rate of insects between populations. In parallel, we present a computational model based on P-membrane computing that will mimic the experimental system of antibiotic resistance transmission. The proposal serves as a proof of concept for the development of more-complex population dynamics of antibiotic resistance transmission that are of interest in public health, which can help us evaluate procedures and design appropriate interventions in epidemiology.
Collapse
Affiliation(s)
- Pablo Llop
- Foundation for the Promotion of Sanitary and Biomedical Research in the Valencian Region (FISABIO), València, Spain
| | - Amparo Latorre
- Foundation for the Promotion of Sanitary and Biomedical Research in the Valencian Region (FISABIO), València, Spain
- Integrative Systems Biology Institute, Universitat de València, València, Spain
- Network Research Center for Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Andrés Moya
- Foundation for the Promotion of Sanitary and Biomedical Research in the Valencian Region (FISABIO), València, Spain
- Integrative Systems Biology Institute, Universitat de València, València, Spain
- Network Research Center for Epidemiology and Public Health (CIBERESP), Madrid, Spain
| |
Collapse
|
42
|
Inamine H, Ellner SP, Newell PD, Luo Y, Buchon N, Douglas AE. Spatiotemporally Heterogeneous Population Dynamics of Gut Bacteria Inferred from Fecal Time Series Data. mBio 2018; 9:e01453-17. [PMID: 29317508 PMCID: PMC5760738 DOI: 10.1128/mbio.01453-17] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 11/27/2017] [Indexed: 02/02/2023] Open
Abstract
A priority in gut microbiome research is to develop methods to investigate ecological processes shaping microbial populations in the host from readily accessible data, such as fecal samples. Here, we demonstrate that these processes can be inferred from the proportion of ingested microorganisms that is egested and their egestion time distribution, by using general mathematical models that link within-host processes to statistics from fecal time series. We apply this framework to Drosophila melanogaster and its gut bacterium Acetobacter tropicalis Specifically, we investigate changes in their interactions following ingestion of a food bolus containing bacteria in a set of treatments varying the following key parameters: the density of exogenous bacteria ingested by the flies (low/high) and the association status of the host (axenic or monoassociated with A. tropicalis). At 5 h post-ingestion, ~35% of the intact bacterial cells have transited through the gut with the food bolus and ~10% are retained in a viable and culturable state, leaving ~55% that have likely been lysed in the gut. Our models imply that lysis and retention occur over a short spatial range within the gut when the bacteria are ingested from a low density, but more broadly in the host gut when ingested from a high density, by both gnotobiotic and axenic hosts. Our study illustrates how time series data complement the analysis of static abundance patterns to infer ecological processes as bacteria traverse the host. Our approach can be extended to investigate how different bacterial species interact within the host to understand the processes shaping microbial community assembly.IMPORTANCE A major challenge to our understanding of the gut microbiome in animals is that it is profoundly difficult to investigate the fate of ingested microbial cells as they travel through the gut. Here, we created mathematical tools to analyze microbial dynamics in the gut from the temporal pattern of their abundance in fecal samples, i.e., without direct observation of the dynamics, and validated them with Drosophila fruit flies. Our analyses revealed that over 5 h after ingestion, most bacteria have likely died in the host or have been egested as intact cells, while some living cells have been retained in the host. Bacterial lysis or retention occurred across a larger area of the gut when flies ingest bacteria from high densities than when flies ingest bacteria from low densities. Our mathematical tools can be applied to other systems, including the dynamics of gut microbial populations and communities in humans.
Collapse
Affiliation(s)
- Hidetoshi Inamine
- Department of Ecology and Evolutionary Biology, Cornell University, Ithaca, New York, USA
| | - Stephen P Ellner
- Department of Ecology and Evolutionary Biology, Cornell University, Ithaca, New York, USA
| | - Peter D Newell
- Department of Entomology, Cornell University, Ithaca, New York, USA
| | - Yuan Luo
- Department of Entomology, Cornell University, Ithaca, New York, USA
| | - Nicolas Buchon
- Department of Entomology, Cornell University, Ithaca, New York, USA
| | - Angela E Douglas
- Department of Entomology, Cornell University, Ithaca, New York, USA
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, USA
| |
Collapse
|
43
|
Su Y, Chen C, Guo L, Du J, Li X, Liu Y. Ecological Balance of Oral Microbiota Is Required to Maintain Oral Mesenchymal Stem Cell Homeostasis. Stem Cells 2018; 36:551-561. [PMID: 29266799 DOI: 10.1002/stem.2762] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 12/07/2017] [Accepted: 12/09/2017] [Indexed: 12/11/2022]
Abstract
Oral microbiome is essential for maintenance of oral cavity health. Imbalanced oral microbiome causes periodontal and other diseases. It is unknown whether oral microbiome affect oral stem cells function. This study used a common clinical antibiotic treatment approach to alter oral microbiome ecology and examine whether oral mesenchymal stem cells (MSCs) are affected. We found that altered oral microbiome resulted gingival MSCs deficiency, leading to a delayed wound healing in male mice. Mechanistically, oral microbiome release lipopolysaccharide (LPS) that stimulates the expression of microRNA-21 (miR-21) and then impair the normal function of gingival MSCs and wound healing process through miR-21/Sp1/telomerase reverse transcriptase pathway. This is the first study indicate that interplay between oral microbiome and MSCs homeostasis in male mice. Stem Cells 2018;36:551-561.
Collapse
Affiliation(s)
- Yingying Su
- Department of Stomatology, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Chider Chen
- Department of Anatomy and Cell Biology, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Lijia Guo
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, People's Republic of China
| | - Juan Du
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, People's Republic of China
| | - Xiaoyan Li
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, People's Republic of China
| | - Yi Liu
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, People's Republic of China
| |
Collapse
|
44
|
Bello-Gil D, Khasbiullina N, Shilova N, Bovin N, Mañez R. Repertoire of BALB/c Mice Natural Anti-Carbohydrate Antibodies: Mice vs. Humans Difference, and Otherness of Individual Animals. Front Immunol 2017; 8:1449. [PMID: 29163519 PMCID: PMC5681490 DOI: 10.3389/fimmu.2017.01449] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 10/17/2017] [Indexed: 01/12/2023] Open
Abstract
One of the most common genetic backgrounds for mice used as a model to investigate human diseases is the inbred BALB/c strain. This work is aimed to characterize the pattern of natural anti-carbohydrate antibodies present in the serum of 20 BALB/c mice by printed glycan array technology and to compare their binding specificities with that of human natural anti-carbohydrate antibodies. Natural antibodies (NAbs) from the serum of BALB/c mice interacted with 71 glycans from a library of 419 different carbohydrate structures. However, only seven of these glycans were recognized by the serum of all the animals studied, and other five glycans by at least 80% of mice. The pattern of the 12 glycans mostly recognized by the circulating antibodies of BALB/c mice differed significantly from that observed with natural anti-carbohydrate antibodies in humans. This lack of identical repertoires of natural anti-carbohydrate antibodies between individual inbred mice, and between mice and humans, should be taken into consideration when mouse models are intended to be used for investigation of NAbs in biomedical research.
Collapse
Affiliation(s)
- Daniel Bello-Gil
- Infectious Pathology and Transplantation Division, Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Spain
| | - Nailya Khasbiullina
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Nadezhda Shilova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Nicolai Bovin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Rafael Mañez
- Infectious Pathology and Transplantation Division, Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Spain
- Intensive Care Department, Bellvitge University Hospital, Hospitalet de Llobregat, Spain
| |
Collapse
|
45
|
Zacarías MF, Souza TC, Zaburlín N, Carmona Cara D, Reinheimer J, Nicoli J, Vinderola G. Influence of Technological Treatments on the Functionality ofBifidobacterium lactisINL1, a Breast Milk-Derived Probiotic. J Food Sci 2017; 82:2462-2470. [DOI: 10.1111/1750-3841.13852] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 07/03/2017] [Accepted: 07/25/2017] [Indexed: 12/31/2022]
Affiliation(s)
- María Florencia Zacarías
- Inst. de Lactología Industrial (INLAIN); Univ. Nacional del Litoral; Santiago del Estero 2829 3000 Santa Fe Argentina
| | - Tassia Costa Souza
- Inst. de Lactología Industrial (INLAIN); Univ. Nacional del Litoral; Santiago del Estero 2829 3000 Santa Fe Argentina
| | - Natalia Zaburlín
- Inst. de Lactología Industrial (INLAIN); Univ. Nacional del Litoral; Santiago del Estero 2829 3000 Santa Fe Argentina
| | - Denise Carmona Cara
- the Dept. de Morfologia, Inst. de Ciências Biológicas (ICB); Univ. Federal de Minas Gerais; Belo Horizonte MG Brazil
| | - Jorge Reinheimer
- Inst. de Lactología Industrial (INLAIN); Univ. Nacional del Litoral; Santiago del Estero 2829 3000 Santa Fe Argentina
| | - Jacques Nicoli
- the Dept. de Microbiologia; Inst. de Ciências Biológicas (ICB); Univ. Federal de Minas Gerais; Belo Horizonte MG Brazil
| | - Gabriel Vinderola
- Inst. de Lactología Industrial (INLAIN); Univ. Nacional del Litoral; Santiago del Estero 2829 3000 Santa Fe Argentina
| |
Collapse
|
46
|
Ehret T, Torelli F, Klotz C, Pedersen AB, Seeber F. Translational Rodent Models for Research on Parasitic Protozoa-A Review of Confounders and Possibilities. Front Cell Infect Microbiol 2017. [PMID: 28638807 PMCID: PMC5461347 DOI: 10.3389/fcimb.2017.00238] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Rodents, in particular Mus musculus, have a long and invaluable history as models for human diseases in biomedical research, although their translational value has been challenged in a number of cases. We provide some examples in which rodents have been suboptimal as models for human biology and discuss confounders which influence experiments and may explain some of the misleading results. Infections of rodents with protozoan parasites are no exception in requiring close consideration upon model choice. We focus on the significant differences between inbred, outbred and wild animals, and the importance of factors such as microbiota, which are gaining attention as crucial variables in infection experiments. Frequently, mouse or rat models are chosen for convenience, e.g., availability in the institution rather than on an unbiased evaluation of whether they provide the answer to a given question. Apart from a general discussion on translational success or failure, we provide examples where infections with single-celled parasites in a chosen lab rodent gave contradictory or misleading results, and when possible discuss the reason for this. We present emerging alternatives to traditional rodent models, such as humanized mice and organoid primary cell cultures. So-called recombinant inbred strains such as the Collaborative Cross collection are also a potential solution for certain challenges. In addition, we emphasize the advantages of using wild rodents for certain immunological, ecological, and/or behavioral questions. The experimental challenges (e.g., availability of species-specific reagents) that come with the use of such non-model systems are also discussed. Our intention is to foster critical judgment of both traditional and newly available translational rodent models for research on parasitic protozoa that can complement the existing mouse and rat models.
Collapse
Affiliation(s)
- Totta Ehret
- FG16 - Mycotic and Parasitic Agents and Mycobacteria, Robert Koch InstituteBerlin, Germany.,Department of Molecular Parasitology, Humboldt-Universität zu BerlinBerlin, Germany
| | - Francesca Torelli
- FG16 - Mycotic and Parasitic Agents and Mycobacteria, Robert Koch InstituteBerlin, Germany
| | - Christian Klotz
- FG16 - Mycotic and Parasitic Agents and Mycobacteria, Robert Koch InstituteBerlin, Germany
| | - Amy B Pedersen
- School of Biological Sciences, University of EdinburghEdinburgh, United Kingdom
| | - Frank Seeber
- FG16 - Mycotic and Parasitic Agents and Mycobacteria, Robert Koch InstituteBerlin, Germany
| |
Collapse
|
47
|
Choo JM, Trim PJ, Leong LEX, Abell GCJ, Brune C, Jeffries N, Wesselingh S, Dear TN, Snel MF, Rogers GB. Inbred Mouse Populations Exhibit Intergenerational Changes in Intestinal Microbiota Composition and Function Following Introduction to a Facility. Front Microbiol 2017; 8:608. [PMID: 28443082 PMCID: PMC5387074 DOI: 10.3389/fmicb.2017.00608] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 03/24/2017] [Indexed: 12/31/2022] Open
Abstract
Inbred mice are used to investigate many aspects of human physiology, including susceptibility to disease and response to therapies. Despite increasing evidence that the composition and function of the murine intestinal microbiota can substantially influence a broad range of experimental outcomes, relatively little is known about microbiome dynamics within experimental mouse populations. We investigated changes in the intestinal microbiome between C57BL/6J mice spanning six generations (assessed at generations 1, 2, 3, and 6), following their introduction to a stringently controlled facility. Fecal microbiota composition and function were assessed by 16S rRNA gene amplicon sequencing and liquid chromatography mass spectrometry, respectively. Significant divergence of the intestinal microbiota between founder and second generation mice, as well as continuing inter-generational variance, was observed. Bacterial taxa whose relative abundance changed significantly through time included Akkermansia, Turicibacter, and Bifidobacterium (p < 0.05), all of which are recognized as having the potential to substantially influence host physiology. Shifts in microbiota composition were mirrored by corresponding differences in the fecal metabolome (r = 0.57, p = 0.0001), with notable differences in levels of tryptophan pathway metabolites and amino acids, including glutamine, glutamate and aspartate. We related the magnitude of changes in the intestinal microbiota and metabolome characteristics during acclimation to those observed between populations housed in separate facilities, which differed in regards to husbandry, barrier conditions and dietary intake. The microbiome variance reported here has implications for experimental reproducibility, and as a consequence, experimental design and the interpretation of research outcomes across wide range of contexts.
Collapse
Affiliation(s)
- Jocelyn M Choo
- Infection and Immunity Theme, South Australian Health and Medical Research Institute, AdelaideSA, Australia
| | - Paul J Trim
- Lysosomal Diseases Research Unit, Nutrition and Metabolism Theme, South Australia Health and Medical Research Institute, AdelaideSA, Australia
| | - Lex E X Leong
- Infection and Immunity Theme, South Australian Health and Medical Research Institute, AdelaideSA, Australia
| | - Guy C J Abell
- School of Medicine, Flinders University, AdelaideSA, Australia
| | - Carly Brune
- Bioresources facility, South Australia Health and Medical Research Institute, AdelaideSA, Australia
| | - Nicole Jeffries
- Bioresources facility, South Australia Health and Medical Research Institute, AdelaideSA, Australia
| | - Steve Wesselingh
- Infection and Immunity Theme, South Australian Health and Medical Research Institute, AdelaideSA, Australia
| | - T N Dear
- Infection and Immunity Theme, South Australian Health and Medical Research Institute, AdelaideSA, Australia
| | - Marten F Snel
- Lysosomal Diseases Research Unit, Nutrition and Metabolism Theme, South Australia Health and Medical Research Institute, AdelaideSA, Australia
| | - Geraint B Rogers
- Infection and Immunity Theme, South Australian Health and Medical Research Institute, AdelaideSA, Australia.,School of Medicine, Flinders University, AdelaideSA, Australia
| |
Collapse
|
48
|
Microbiota alteration is associated with the development of stress-induced despair behavior. Sci Rep 2017; 7:43859. [PMID: 28266612 PMCID: PMC5339726 DOI: 10.1038/srep43859] [Citation(s) in RCA: 253] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 01/31/2017] [Indexed: 12/29/2022] Open
Abstract
Depressive disorders often run in families, which, in addition to the genetic component, may point to the microbiome as a causative agent. Here, we employed a combination of behavioral, molecular and computational techniques to test the role of the microbiota in mediating despair behavior. In chronically stressed mice displaying despair behavior, we found that the microbiota composition and the metabolic signature dramatically change. Specifically, we observed reduced Lactobacillus and increased circulating kynurenine levels as the most prominent changes in stressed mice. Restoring intestinal Lactobacillus levels was sufficient to improve the metabolic alterations and behavioral abnormalities. Mechanistically, we identified that Lactobacillus-derived reactive oxygen species may suppress host kynurenine metabolism, by inhibiting the expression of the metabolizing enzyme, IDO1, in the intestine. Moreover, maintaining elevated kynurenine levels during Lactobacillus supplementation diminished the treatment benefits. Collectively, our data provide a mechanistic scenario for how a microbiota player (Lactobacillus) may contribute to regulating metabolism and resilience during stress.
Collapse
|
49
|
Lundberg R, Bahl MI, Licht TR, Toft MF, Hansen AK. Microbiota composition of simultaneously colonized mice housed under either a gnotobiotic isolator or individually ventilated cage regime. Sci Rep 2017; 7:42245. [PMID: 28169374 PMCID: PMC5294411 DOI: 10.1038/srep42245] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 01/05/2017] [Indexed: 12/26/2022] Open
Abstract
Germ-free rodents colonized with microbiotas of interest are used for host-microbiota investigations and for testing microbiota-targeted therapeutic candidates. Traditionally, isolators are used for housing such gnotobiotic rodents due to optimal protection from the environment, but research groups focused on the microbiome are increasingly combining or substituting isolator housing with individually ventilated cage (IVC) systems. We compared the effect of housing systems on the gut microbiota composition of germ-free mice colonized with a complex microbiota and housed in either multiple IVC cages in an IVC facility or in multiple open-top cages in an isolator during three generations and five months. No increase in bacterial diversity as assessed by 16S rRNA gene sequencing was observed in the IVC cages, despite not applying completely aseptic cage changes. The donor bacterial community was equally represented in both housing systems. Time-dependent clustering between generations was observed in both systems, but was strongest in the IVC cages. Different relative abundance of a Rikenellaceae genus contributed to separate clustering of the isolator and IVC communities. Our data suggest that complex microbiotas are protected in IVC systems, but challenges related to temporal dynamics should be addressed.
Collapse
Affiliation(s)
- Randi Lundberg
- Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, 1871 Frederiksberg C, Denmark.,Internal Research and Development, Taconic Biosciences, 4623 Lille Skensved, Denmark
| | - Martin I Bahl
- National Food Institute, Technical University of Denmark, 2860 Søborg, Denmark
| | - Tine R Licht
- National Food Institute, Technical University of Denmark, 2860 Søborg, Denmark
| | - Martin F Toft
- Internal Research and Development, Taconic Biosciences, 4623 Lille Skensved, Denmark
| | - Axel K Hansen
- Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, 1871 Frederiksberg C, Denmark
| |
Collapse
|
50
|
Kreisinger J, Kropáčková L, Petrželková A, Adámková M, Tomášek O, Martin JF, Michálková R, Albrecht T. Temporal Stability and the Effect of Transgenerational Transfer on Fecal Microbiota Structure in a Long Distance Migratory Bird. Front Microbiol 2017; 8:50. [PMID: 28220109 PMCID: PMC5292904 DOI: 10.3389/fmicb.2017.00050] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 01/06/2017] [Indexed: 12/20/2022] Open
Abstract
Animal bodies are inhabited by a taxonomically and functionally diverse community of symbiotic and commensal microorganisms. From an ecological and evolutionary perspective, inter-individual variation in host-associated microbiota contributes to physiological and immune system variation. As such, host-associated microbiota may be considered an integral part of the host’s phenotype, serving as a substrate for natural selection. This assumes that host-associated microbiota exhibits high temporal stability, however, and that its composition is shaped by trans-generational transfer or heritable host-associated microbiota modulators encoded by the host genome. Although this concept is widely accepted, its crucial assumptions have rarely been tested in wild vertebrate populations. We performed 16S rRNA metabarcoding on an extensive set of fecal microbiota (FM) samples from an insectivorous, long-distance migratory bird, the barn swallow (Hirundo rustica). Our data revealed clear differences in FM among juveniles and adults as regards taxonomic and functional composition, diversity and co-occurrence network complexity. Multiple FM samples from the same juvenile or adult collected within single breeding seasons exhibited higher similarity than expected by chance, as did adult FM samples over two consecutive years. Despite low effect sizes for FM stability over time at the community level, we identified an adult FM subset with relative abundances exhibiting significant temporal consistency, possibly inducing long-term effects on the host phenotype. Our data also indicate a slight maternal (but not paternal) effect on FM composition in social offspring, though this is unlikely to persist into adulthood. We discuss our findings in the context of both evolution and ecology of microbiota vs. host interactions and barn swallow biology.
Collapse
Affiliation(s)
- Jakub Kreisinger
- Department of Zoology, Faculty of Science, Charles University Prague, Czechia
| | - Lucie Kropáčková
- Department of Zoology, Faculty of Science, Charles University Prague, Czechia
| | - Adéla Petrželková
- Department of Ecology, Faculty of Science, Charles University Prague, Czechia
| | - Marie Adámková
- Institute of Vertebrate Biology, Czech Academy of Sciences, Studenec Czechia
| | - Oldřich Tomášek
- Department of Zoology, Faculty of Science, Charles UniversityPrague, Czechia; Institute of Vertebrate Biology, Czech Academy of Sciences, StudenecCzechia
| | - Jean-François Martin
- Montpellier-SupAgro, UMR Centre de Biologie pour la Gestion des Populations Montferrier-sur-Lez, France
| | - Romana Michálková
- Department of Zoology, Faculty of Science, Charles University Prague, Czechia
| | - Tomáš Albrecht
- Department of Zoology, Faculty of Science, Charles UniversityPrague, Czechia; Institute of Vertebrate Biology, Czech Academy of Sciences, StudenecCzechia
| |
Collapse
|