1
|
Meinen-Jochum J, Skow CJ, Mellata M. Layer segmented filamentous bacteria colonize and impact gut health of broiler chickens. mSphere 2024; 9:e0049224. [PMID: 39422489 PMCID: PMC11580430 DOI: 10.1128/msphere.00492-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 09/23/2024] [Indexed: 10/19/2024] Open
Abstract
In commercial poultry farms, chicks hatch away from their progenitors from which they acquire key host-specific microbiota, like segmented filamentous bacteria (SFB) involved in gut maturation in early life. This study investigated whether providing chicken SFB to newly hatched broilers would increase their gut maturation and resistance to bacteria relevant to broiler and human health. One-day-old Ross308 broilers were orally treated with either phosphate-buffered saline (CON) or layer-derived SFB (D-SFB). On days 5, 10, 17, and 24, feces were collected to detect and enumerate SFB and Enterobacteriaceae. On days 8, 15, 22, and 29, birds were euthanized, intestinal samples were collected to detect and enumerate SFB through quantitative PCR (qPCR) and microscopy and expression of genes associated with gut immune function through reverse transcription-qPCR. This study showed that, despite their host specificity, layer SFB can colonize their genetically distinct relative broilers. Ileal SFB colonization was accelerated by a week with the SFB treatment and covered the proximal, medial, and distal sections of the ileum. Colonization of the ileum by SFB in early life highly activated gene expression of intestinal barrier proteins and cytokines, e.g., IL-10 and IFNγ but not IL-17. SFB treatment reduced the level of Enterobacteriaceae in the gut and provided superior resistance to intestinal and extraintestinal pathogens as tested in vitro. Overall, early gut colonization of SFB is imperative for the maturation of the gut immune system and the establishment of a homeostatic gut environment. Improving our understanding of gut immune maturation in food-producing animals is crucial for both human and animal health.IMPORTANCEIn commercial farms, newly hatched chicks may lack host-specific microbiota that help mature their gut immune system for lifelong health benefits. Here, introducing an avian segmented filamentous bacteria (SFB) to commercially sourced chickens orally at hatch accelerated SFB colonization of the ileum. Remarkably, SFB from layers were able to colonize broilers and enhance gut immune maturation, and this immunomodulation impacted the ability to increase intestinal and extraintestinal resistance to bacteria relevant to poultry and human health. With the antibiotic restrictions in animal production, strategies that will help mitigate infections are urgently needed. In summary, we developed a live prophylactic for newly hatched chicks to improve animal health and food safety. Due to the host specificity of SFB, our data highlight the importance of investigating the molecular mechanism of SFB interaction in their own host.
Collapse
Affiliation(s)
- Jared Meinen-Jochum
- Interdepartmental Microbiology Graduate Program, Iowa State University, Ames, Iowa, USA
- Department of Food Science and Human Nutrition, Iowa State University, Ames, Iowa, USA
| | - Caleb J. Skow
- Interdepartmental Microbiology Graduate Program, Iowa State University, Ames, Iowa, USA
- Department of Food Science and Human Nutrition, Iowa State University, Ames, Iowa, USA
| | - Melha Mellata
- Interdepartmental Microbiology Graduate Program, Iowa State University, Ames, Iowa, USA
- Department of Food Science and Human Nutrition, Iowa State University, Ames, Iowa, USA
| |
Collapse
|
2
|
Zhu Z, Zhu Z, Shi Z, Wang C, Chen F. Kaempferol Remodels Liver Monocyte Populations and Treats Hepatic Fibrosis in Mice by Modulating Intestinal Flora and Metabolic Reprogramming. Inflammation 2024:10.1007/s10753-024-02184-2. [PMID: 39531210 DOI: 10.1007/s10753-024-02184-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 10/17/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
Changes in gut flora are associated with liver fibrosis. The interactions of host with intestinal flora are still unknown, with little research investigating such interactions with comprehensive multi-omics data. The present work analyzed and integrated large-scale multi-omics transcriptomics, microbiome, metabolome, and single-cell RNA-sequencing datasets from Kaempferol-treated and untreated control groups by advanced bioinformatics methods. This study concludes that kaempferol dose-dependently improved serum markers (like AST, ALT, TBil, Alb, and PT) and suppressed fibrosis markers (including HA, PC III, LN, α-SMA, and Collagen I), while kaempferol also increased body weight. Mechanistically, kaempferol improved the metabolic levels of intestinal flora dysbiosis and associated lipids. This was achieved by increasing the abundance of g__Robinsoniella, g__Erysipelotrichaceae_UCG-003, g__Coriobacteriaceae_UCG-002, and 5-Methylcytidine, all-trans-5,6- Epoxyretinoic acid, LPI (18:0), LPI (20:4), etc. to achieve this. Kaemferol exerts anti-inflammatory and immune-enhancing effects by down-regulating the Th17/IL-17 signaling pathway in PDGF-induced LX2 cells. In addition, kaempferol administration remarkably elevated CD4 + T and CD8 + T cellular proportions, thereby activating immune cells for protecting the body and controlling inflammatory conditions. The combined interaction of multiple data may explain how Kaempferol modulates the intestinal flora thereby remodeling the hepatocyte population and alleviating liver fibrosis.
Collapse
Affiliation(s)
- Zhiqin Zhu
- Department of Hepatology, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, 510315, China
| | - Zhiqi Zhu
- School of Materials Science and Engineering, Central South University, Changsha, 410083, China
| | - Zhenyi Shi
- Department of Biochemistry and Molecular Biology, School of Basic Medical & Sciences, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Single Cell Technology and Application, 10 Southern Medical University, Guangzhou, China
| | - Chen Wang
- The Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Fengsheng Chen
- Department of Hepatology, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, 510315, China.
| |
Collapse
|
3
|
Filho AMC, Gomes NS, Lós DB, Leite IB, Tremblay MÈ, Macêdo DS. Microglia and Microbiome-Gut-Brain Axis. ADVANCES IN NEUROBIOLOGY 2024; 37:303-331. [PMID: 39207699 DOI: 10.1007/978-3-031-55529-9_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
The mammalian gut contains a community of microorganisms called gut microbiome. The gut microbiome is integrated into mammalian physiology, contributing to metabolism, production of metabolites, and promoting immunomodulatory actions. Microglia, the brain's resident innate immune cells, play an essential role in homeostatic neurogenesis, synaptic remodeling, and glial maturation. Microglial dysfunction has been implicated in the pathogenesis of several neuropsychiatric disorders. Recent findings indicate that microglia are influenced by the gut microbiome and their derived metabolites throughout life. The pathways by which microbiota regulate microglia have only started to be understood, but this discovery has the potential to provide valuable insights into the pathogenesis of brain disorders associated with an altered microbiome. Here, we discuss the recent literature on the role of the gut microbiome in modulating microglia during development and adulthood and summarize the key findings on this bidirectional crosstalk in selected examples of neuropsychiatric and neurodegenerative disorders. We also highlight some current caveats and perspectives for the field.
Collapse
Affiliation(s)
- Adriano Maia Chaves Filho
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Department of Physiology and Pharmacology, Drug Research and Development Center, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Nayana Soares Gomes
- Department of Physiology and Pharmacology, Drug Research and Development Center, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Deniele Bezerra Lós
- Department of Physiology and Pharmacology, Drug Research and Development Center, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Isabel Bessa Leite
- Department of Physiology and Pharmacology, Drug Research and Development Center, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada.
- Department of Molecular Medicine, Université de Laval, Québec City, Canada.
- Department of Neurology and Neurosurgery, McGill University, Montréal, Canada.
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada.
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada.
| | - Danielle S Macêdo
- Department of Physiology and Pharmacology, Drug Research and Development Center, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil.
- National Institute for Translational Medicine (INCT-TM, CNPq), Ribeirão Preto, SP, Brazil.
| |
Collapse
|
4
|
Abstract
In this issue of Cell Host & Microbe, Woo et al. (2021) show that retinoic acid generated by symbiotic segmented filamentous bacteria in the intestine primes host intestinal epithelial defense and mediates early innate immune protection against Citrobacter rodentium infection.
Collapse
|
5
|
Woo V, Eshleman EM, Hashimoto-Hill S, Whitt J, Wu SE, Engleman L, Rice T, Karns R, Qualls JE, Haslam DB, Vallance BA, Alenghat T. Commensal segmented filamentous bacteria-derived retinoic acid primes host defense to intestinal infection. Cell Host Microbe 2021; 29:1744-1756.e5. [PMID: 34678170 DOI: 10.1016/j.chom.2021.09.010] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 07/14/2021] [Accepted: 09/21/2021] [Indexed: 12/30/2022]
Abstract
Interactions between the microbiota and mammalian host are essential for defense against infection, but the microbial-derived cues that mediate this relationship remain unclear. Here, we find that intestinal epithelial cell (IEC)-associated commensal bacteria, segmented filamentous bacteria (SFB), promote early protection against the pathogen Citrobacter rodentium, independent of CD4+ T cells. SFB induced histone modifications in IECs at sites enriched for retinoic acid receptor motifs, suggesting that SFB may enhance defense through retinoic acid (RA). Consistent with this, inhibiting RA signaling suppressed SFB-induced protection. Intestinal RA levels were elevated in SFB mice, despite the inhibition of mammalian RA production, indicating that SFB directly modulate RA. Interestingly, RA was produced by intestinal bacteria, and the loss of bacterial-intrinsic aldehyde dehydrogenase activity decreased the RA levels and increased infection. These data reveal RA as an unexpected microbiota-derived metabolite that primes innate defense and suggests that pre- and probiotic approaches to elevate RA could prevent or combat infections.
Collapse
Affiliation(s)
- Vivienne Woo
- Division of Immunobiology and Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Emily M Eshleman
- Division of Immunobiology and Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Seika Hashimoto-Hill
- Division of Immunobiology and Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Jordan Whitt
- Division of Immunobiology and Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Shu-En Wu
- Division of Immunobiology and Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Laura Engleman
- Division of Immunobiology and Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Taylor Rice
- Division of Immunobiology and Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Rebekah Karns
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Joseph E Qualls
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - David B Haslam
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Bruce A Vallance
- Department of Pediatrics, BC Children's Hospital Research Institute and the University of British Columbia, Vancouver, BC V5Z 4H4, Canada
| | - Theresa Alenghat
- Division of Immunobiology and Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA.
| |
Collapse
|
6
|
Chiaranunt P, Tai SL, Ngai L, Mortha A. Beyond Immunity: Underappreciated Functions of Intestinal Macrophages. Front Immunol 2021; 12:749708. [PMID: 34650568 PMCID: PMC8506163 DOI: 10.3389/fimmu.2021.749708] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 09/03/2021] [Indexed: 12/12/2022] Open
Abstract
The gastrointestinal tract hosts the largest compartment of macrophages in the body, where they serve as mediators of host defense and immunity. Seeded in the complex tissue-environment of the gut, an array of both hematopoietic and non-hematopoietic cells forms their immediate neighborhood. Emerging data demonstrate that the functional diversity of intestinal macrophages reaches beyond classical immunity and includes underappreciated non-immune functions. In this review, we discuss recent advances in research on intestinal macrophage heterogeneity, with a particular focus on how non-immune functions of macrophages impact tissue homeostasis and function. We delve into the strategic localization of distinct gut macrophage populations, describe the potential factors that regulate their identity and functional heterogeneity within these locations, and provide open questions that we hope will inspire research dedicated to elucidating a holistic view on macrophage-tissue cell interactions in the body's largest mucosal organ.
Collapse
Affiliation(s)
- Pailin Chiaranunt
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Siu Ling Tai
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Louis Ngai
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Arthur Mortha
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
7
|
Zhang C, Franklin CL, Ericsson AC. Consideration of Gut Microbiome in Murine Models of Diseases. Microorganisms 2021; 9:microorganisms9051062. [PMID: 34068994 PMCID: PMC8156714 DOI: 10.3390/microorganisms9051062] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 04/29/2021] [Accepted: 05/12/2021] [Indexed: 12/12/2022] Open
Abstract
The gut microbiome (GM), a complex community of bacteria, viruses, protozoa, and fungi located in the gut of humans and animals, plays significant roles in host health and disease. Animal models are widely used to investigate human diseases in biomedical research and the GM within animal models can change due to the impact of many factors, such as the vendor, husbandry, and environment. Notably, variations in GM can contribute to differences in disease model phenotypes, which can result in poor reproducibility in biomedical research. Variation in the gut microbiome can also impact the translatability of animal models. For example, standard lab mice have different pathogen exposure experiences when compared to wild or pet store mice. As humans have antigen experiences that are more similar to the latter, the use of lab mice with more simplified microbiomes may not yield optimally translatable data. Additionally, the literature describes many methods to manipulate the GM and differences between these methods can also result in differing interpretations of outcomes measures. In this review, we focus on the GM as a potential contributor to the poor reproducibility and translatability of mouse models of disease. First, we summarize the important role of GM in host disease and health through different gut–organ axes and the close association between GM and disease susceptibility through colonization resistance, immune response, and metabolic pathways. Then, we focus on the variation in the microbiome in mouse models of disease and address how this variation can potentially impact disease phenotypes and subsequently influence research reproducibility and translatability. We also discuss the variations between genetic substrains as potential factors that cause poor reproducibility via their effects on the microbiome. In addition, we discuss the utility of complex microbiomes in prospective studies and how manipulation of the GM through differing transfer methods can impact model phenotypes. Lastly, we emphasize the need to explore appropriate methods of GM characterization and manipulation.
Collapse
Affiliation(s)
- Chunye Zhang
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65201, USA;
| | - Craig L. Franklin
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65201, USA;
- Mutant Mouse Resource and Research Center, University of Missouri, Columbia, MO 65201, USA
- Metagenomics Center, University of Missouri, Columbia, MO 65201, USA
- Correspondence: (C.L.F.); (A.C.E.)
| | - Aaron C. Ericsson
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65201, USA;
- Mutant Mouse Resource and Research Center, University of Missouri, Columbia, MO 65201, USA
- Metagenomics Center, University of Missouri, Columbia, MO 65201, USA
- Correspondence: (C.L.F.); (A.C.E.)
| |
Collapse
|
8
|
Buchheister S, Bleich A. Health Monitoring of Laboratory Rodent Colonies-Talking about (R)evolution. Animals (Basel) 2021; 11:1410. [PMID: 34069175 PMCID: PMC8155880 DOI: 10.3390/ani11051410] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/02/2021] [Accepted: 05/10/2021] [Indexed: 01/15/2023] Open
Abstract
The health monitoring of laboratory rodents is essential for ensuring animal health and standardization in biomedical research. Progress in housing, gnotobiotic derivation, and hygienic monitoring programs led to enormous improvement of the microbiological quality of laboratory animals. While traditional health monitoring and pathogen detection methods still serve as powerful tools for the diagnostics of common animal diseases, molecular methods develop rapidly and not only improve test sensitivities but also allow high throughput analyses of various sample types. Concurrently, to the progress in pathogen detection and elimination, the research community becomes increasingly aware of the striking influence of microbiome compositions in laboratory animals, affecting disease phenotypes and the scientific value of research data. As repeated re-derivation cycles and strict barrier husbandry of laboratory rodents resulted in a limited diversity of the animals' gut microbiome, future monitoring approaches will have to reform-aiming at enhancing the validity of animal experiments. This review will recapitulate common health monitoring concepts and, moreover, outline strategies and measures on coping with microbiome variation in order to increase reproducibility, replicability and generalizability.
Collapse
Affiliation(s)
| | - André Bleich
- Institute for Laboratory Animal Science, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany;
| |
Collapse
|
9
|
Fekete E, Allain T, Siddiq A, Sosnowski O, Buret AG. Giardia spp. and the Gut Microbiota: Dangerous Liaisons. Front Microbiol 2021; 11:618106. [PMID: 33510729 PMCID: PMC7835142 DOI: 10.3389/fmicb.2020.618106] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 12/15/2020] [Indexed: 12/11/2022] Open
Abstract
Alteration of the intestinal microbiome by enteropathogens is commonly associated with gastrointestinal diseases and disorders and has far-reaching consequences for overall health. Significant advances have been made in understanding the role of microbial dysbiosis during intestinal infections, including infection with the protozoan parasite Giardia duodenalis, one of the most prevalent gut protozoa. Altered species composition and diversity, functional changes in the commensal microbiota, and changes to intestinal bacterial biofilm structure have all been demonstrated during the course of Giardia infection and have been implicated in Giardia pathogenesis. Conversely, the gut microbiota has been found to regulate parasite colonization and establishment and plays a critical role in immune modulation during mono and polymicrobial infections. These disruptions to the commensal microbiome may contribute to a number of acute, chronic, and post-infectious clinical manifestations of giardiasis and may account for variations in disease presentation within and between infected populations. This review discusses recent advances in characterizing Giardia-induced bacterial dysbiosis in the gut and the roles of dysbiosis in Giardia pathogenesis.
Collapse
Affiliation(s)
- Elena Fekete
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
- Inflammation Research Network, University of Calgary, Calgary, AB, Canada
- Host-Parasite Interactions, University of Calgary, Calgary, AB, Canada
| | - Thibault Allain
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
- Inflammation Research Network, University of Calgary, Calgary, AB, Canada
- Host-Parasite Interactions, University of Calgary, Calgary, AB, Canada
| | - Affan Siddiq
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
- Inflammation Research Network, University of Calgary, Calgary, AB, Canada
- Host-Parasite Interactions, University of Calgary, Calgary, AB, Canada
| | - Olivia Sosnowski
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
- Inflammation Research Network, University of Calgary, Calgary, AB, Canada
- Host-Parasite Interactions, University of Calgary, Calgary, AB, Canada
| | - Andre G. Buret
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
- Inflammation Research Network, University of Calgary, Calgary, AB, Canada
- Host-Parasite Interactions, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
10
|
Ericsson AC, Franklin CL. The gut microbiome of laboratory mice: considerations and best practices for translational research. Mamm Genome 2021; 32:239-250. [PMID: 33689000 PMCID: PMC8295156 DOI: 10.1007/s00335-021-09863-7] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 02/18/2021] [Indexed: 12/14/2022]
Abstract
Just as the gut microbiota (GM) is now recognized as an integral mediator of environmental influences on human physiology, susceptibility to disease, and response to pharmacological intervention, so too does the GM of laboratory mice affect the phenotype of research using mouse models. Multiple experimental factors have been shown to affect the composition of the GM in research mice, as well as the model phenotype, suggesting that the GM represents a major component in experimental reproducibility. Moreover, several recent studies suggest that manipulation of the GM of laboratory mice can substantially improve the predictive power or translatability of data generated in mouse models to the human conditions under investigation. This review provides readers with information related to these various factors and practices, and recommendations regarding methods by which issues with poor reproducibility or translatability can be transformed into discoveries.
Collapse
Affiliation(s)
- Aaron C Ericsson
- University of Missouri Metagenomics Center (MUMC), MU Mutant Mouse Resource and Research Center (MU MMRRC), Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA.
| | - Craig L Franklin
- University of Missouri Metagenomics Center (MUMC), MU Mutant Mouse Resource and Research Center (MU MMRRC), Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA
| |
Collapse
|
11
|
Daniel N, Lécuyer E, Chassaing B. Host/microbiota interactions in health and diseases-Time for mucosal microbiology! Mucosal Immunol 2021; 14:1006-1016. [PMID: 33772148 PMCID: PMC8379076 DOI: 10.1038/s41385-021-00383-w] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 01/25/2021] [Accepted: 01/26/2021] [Indexed: 02/04/2023]
Abstract
During the last 20 years, a new field of research delineating the importance of the microbiota in health and diseases has emerged. Inappropriate host-microbiota interactions have been shown to trigger a wide range of chronic inflammatory diseases, and defining the exact mechanisms behind perturbations of such relationship, as well as ways by which these disturbances can lead to disease states, both remain to be fully elucidated. The mucosa-associated microbiota constitutes a recently studied microbial population closely linked with the promotion of chronic intestinal inflammation and associated disease states. This review will highlight seminal works that have brought into light the importance of the mucosa-associated microbiota in health and diseases, emphasizing the challenges and promises of expending the mucosal microbiology field of research.
Collapse
Affiliation(s)
- Noëmie Daniel
- grid.508487.60000 0004 7885 7602INSERM U1016, team “Mucosal microbiota in chronic inflammatory diseases”, CNRS UMR 8104, Université de Paris, Paris, France
| | - Emelyne Lécuyer
- grid.428999.70000 0001 2353 6535Microenvironment & Immunity Unit, Pasteur Institute, INSERM U1224, Paris, France
| | - Benoit Chassaing
- grid.508487.60000 0004 7885 7602INSERM U1016, team “Mucosal microbiota in chronic inflammatory diseases”, CNRS UMR 8104, Université de Paris, Paris, France
| |
Collapse
|
12
|
Chen H, Wang L, Wang X, Wang X, Liu H, Yin Y. Distribution and Strain Diversity of Immunoregulating Segmented Filamentous Bacteria in Human Intestinal Lavage Samples. MICROBIAL ECOLOGY 2020; 79:1021-1033. [PMID: 31728601 DOI: 10.1007/s00248-019-01441-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 09/11/2019] [Indexed: 06/10/2023]
Abstract
Segmented filamentous bacteria (SFB) are well known for their functions in the immunoregulation of hosts including the promotion of Th17 cell differentiation, B cell maturation, and immune system development. However, most analyses of SFB have focused on animal models, and thus, investigation of SFB prevalence in humans and their roles in human immunoregulation and health is needed. Although little is known overall of SFB prevalence in humans, they are characteristically abundant in animals during weaning. In this study, SFB-like bacteria were detected in ileal lavage samples from human children that were aged between 1 to 17 years old by scanning electron microscopy (SEM) analysis, and their insertion into the mucosa was also observed. In addition, the expression of SFB flagellin at the human bacterial interface was observed by immunohistochemistry (IHC) and western blot. Moreover, two pairs of primers specific for SFB, but targeting different genes, were used to detect SFB in human intestinal lavage samples. These analyses indicated that SFB were present in over 50% of patient ileal samples independent of age. High-throughput gene sequencing indicated that different SFB strains were detected among samples. Between nine and 23 SFB flagellin gene operational taxonomic units were identified. In addition to evaluating the prevalence of SFB in human samples, correlations between SFB presence and chief complaints of clinical symptoms were evaluated, as well as the relationship between SFB and patient serum immunoglobulin concentrations. SFB prevalence was significantly higher in hematochezia patients (68%) than in abdominal pain (56.10%) and diarrhea (43.75%) patients. Furthermore, the concentrations of serum IgA, IgM, and IgE, were similar between SFB-positive and SFB-negative patient groups, although IgG concentrations were significantly higher in the SFB-negative group.
Collapse
Affiliation(s)
- Huahai Chen
- Key Laboratory of Comprehensive Utilization of Advantage Plants Resources in Hunan South, College of Chemistry and Bioengineering, Hunan University of Science and Engineering, Yongzhou, Hunan, China
- State Key Laboratory of Breeding Base for Zhejiang Sustainable Pest and Disease Control, Institute of Plant Protection and Microbiology, Zhejiang Academy of Agricultural Sciences, Zhejiang, Hangzhou, China
| | - Ling Wang
- Children's Hospital of Shanghai, Children's Hospital of Shanghai Jiaotong University, Shanghai, China
| | - Xing Wang
- Children's Hospital of Shanghai, Children's Hospital of Shanghai Jiaotong University, Shanghai, China
| | - Xin Wang
- State Key Laboratory of Breeding Base for Zhejiang Sustainable Pest and Disease Control, Institute of Plant Protection and Microbiology, Zhejiang Academy of Agricultural Sciences, Zhejiang, Hangzhou, China.
| | - Haifeng Liu
- Children's Hospital of Shanghai, Children's Hospital of Shanghai Jiaotong University, Shanghai, China.
| | - Yeshi Yin
- Key Laboratory of Comprehensive Utilization of Advantage Plants Resources in Hunan South, College of Chemistry and Bioengineering, Hunan University of Science and Engineering, Yongzhou, Hunan, China.
- State Key Laboratory of Breeding Base for Zhejiang Sustainable Pest and Disease Control, Institute of Plant Protection and Microbiology, Zhejiang Academy of Agricultural Sciences, Zhejiang, Hangzhou, China.
| |
Collapse
|
13
|
|
14
|
Rahimi S, Kathariou S, Fletcher O, Grimes JL. The effectiveness of a dietary direct-fed microbial and mannan oligosaccharide on ultrastructural changes of intestinal mucosa of turkey poults infected with Salmonella and Campylobacter. Poult Sci 2020; 99:1135-1149. [PMID: 32036965 PMCID: PMC7587720 DOI: 10.1016/j.psj.2019.09.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 09/26/2019] [Accepted: 09/27/2019] [Indexed: 01/03/2023] Open
Abstract
Salmonella and Campylobacter are considered major public health burdens worldwide, and poultry are known to be one of the main reservoirs for these zoonotic pathogens. This study was conducted to evaluate the effect of a commercial probiotic or direct-fed microbial (DFM) Calsporin (CSP), and prebiotic or mannan oligosaccharide (MOS) (IMW50) on ultrastructural changes and the villous integrity of intestinal mucosa in turkey poults challenged with Salmonella and Campylobacter. A 21-day battery cage study was conducted using 4 dietary treatments including a basal diet (corn and soybean-based) nonsupplemented and uninfected as a negative control (NC); basal diet supplemented with 0.05% DFM (CSP); basal diet supplemented with 0.05% MOS (IMW50); and basal diet supplemented with 0.05% mixture of DFM and MOS at equal proportions. Female large white turkey poults aged 336 days were obtained from a local commercial hatchery and randomly distributed in electrically heated battery cages with 12 treatments of 4 replicates per treatment containing 7 poults per pen. The first 16 pens were not infected with bacteria, poults in pens 17-32 were orally challenged at day 7 with 105 cfu Salmonella Heidelberg, and the poults in pens 33-48 were orally challenged at day 7 with 105 cfu Campylobacter jejuni. Feed and water were provided ad libitum throughout the study. At day 21, ileal tissue samples from 1 bird per cage were collected for intestinal integrity and ultrastructural examination by scanning and electron microscopy. DFM and MOS supplementation was effective in both challenged and nonchallenged (not infected with Salmonella and Campylobacter) birds. Goblet cells and mucus were increased, with the presence of large numbers of segmented filamentous bacteria in DFM- and MOS-supplemented groups compared with birds in control treatments. The number and size of villi were reduced in poults exposed to Salmonella and Campylobacter. Results show that CSP and IMW50 provide protection of ileal mucosal integrity in poults exposed to Salmonella or Campylobacter.
Collapse
Affiliation(s)
- Shaban Rahimi
- Department of Poultry Science, Faculty of Agriculture, Tarbiat Modares University, 14115-336 Tehran, Iran
| | - Sophia Kathariou
- Department of Food Bioprocessing and Nutrition Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27695-7608
| | - Oscar Fletcher
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27695-7608
| | - Jesse L Grimes
- Prestage Department of Poultry Science, College of Agriculture and Life Sciences, North Carolina State University, Raleigh, NC 27695-7608.
| |
Collapse
|
15
|
Lai NY, Musser MA, Pinho-Ribeiro FA, Baral P, Jacobson A, Ma P, Potts DE, Chen Z, Paik D, Soualhi S, Yan Y, Misra A, Goldstein K, Lagomarsino VN, Nordstrom A, Sivanathan KN, Wallrapp A, Kuchroo VK, Nowarski R, Starnbach MN, Shi H, Surana NK, An D, Wu C, Huh JR, Rao M, Chiu IM. Gut-Innervating Nociceptor Neurons Regulate Peyer's Patch Microfold Cells and SFB Levels to Mediate Salmonella Host Defense. Cell 2020; 180:33-49.e22. [PMID: 31813624 PMCID: PMC6954329 DOI: 10.1016/j.cell.2019.11.014] [Citation(s) in RCA: 242] [Impact Index Per Article: 48.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Revised: 09/08/2019] [Accepted: 11/12/2019] [Indexed: 12/30/2022]
Abstract
Gut-innervating nociceptor sensory neurons respond to noxious stimuli by initiating protective responses including pain and inflammation; however, their role in enteric infections is unclear. Here, we find that nociceptor neurons critically mediate host defense against the bacterial pathogen Salmonella enterica serovar Typhimurium (STm). Dorsal root ganglia nociceptors protect against STm colonization, invasion, and dissemination from the gut. Nociceptors regulate the density of microfold (M) cells in ileum Peyer's patch (PP) follicle-associated epithelia (FAE) to limit entry points for STm invasion. Downstream of M cells, nociceptors maintain levels of segmentous filamentous bacteria (SFB), a gut microbe residing on ileum villi and PP FAE that mediates resistance to STm infection. TRPV1+ nociceptors directly respond to STm by releasing calcitonin gene-related peptide (CGRP), a neuropeptide that modulates M cells and SFB levels to protect against Salmonella infection. These findings reveal a major role for nociceptor neurons in sensing and defending against enteric pathogens.
Collapse
Affiliation(s)
- Nicole Y Lai
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Melissa A Musser
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA; Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
| | | | - Pankaj Baral
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Amanda Jacobson
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Pingchuan Ma
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - David E Potts
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Zuojia Chen
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA; Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Donggi Paik
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Salima Soualhi
- Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
| | - Yiqing Yan
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Aditya Misra
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Kaitlin Goldstein
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | | | - Anja Nordstrom
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Kisha N Sivanathan
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Antonia Wallrapp
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Vijay K Kuchroo
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Roni Nowarski
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | | | - Hailian Shi
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA; Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Neeraj K Surana
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA; Department of Pediatrics, Duke University, Durham, NC 27710, USA; Department of Molecular Genetics and Microbiology, Duke University, Durham, NC 27710, USA; Department of Immunology, Duke University, Durham, NC 27710, USA
| | - Dingding An
- Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
| | - Chuan Wu
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA; Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jun R Huh
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Meenakshi Rao
- Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
| | - Isaac M Chiu
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
16
|
|
17
|
Chen B, Chen H, Shu X, Yin Y, Li J, Qin J, Chen L, Peng K, Xu F, Gu W, Zhao H, Jiang L, Li L, Song J, Elitsur Y, Yu HD, Jiang M, Wang X, Xiang C. Presence of Segmented Filamentous Bacteria in Human Children and Its Potential Role in the Modulation of Human Gut Immunity. Front Microbiol 2018; 9:1403. [PMID: 30008704 PMCID: PMC6034559 DOI: 10.3389/fmicb.2018.01403] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 06/07/2018] [Indexed: 01/19/2023] Open
Abstract
Segmented filamentous bacteria (SFB) are commensal organisms that grow by anchoring a specialized holdfast structure to the intestinal walls of a variety of animals. Interaction of SFB with Peyer's patches in mice promotes the post-natal maturation of the immune system. We previously reported that the colonization of SFB in humans mainly occurs by 36 months of age, and is difficult to be detected afterward. In this study, we measured the level of SFB in intestinal fluids of human children. SFB were found via qPCR to represent a small fraction of the whole SFB-positive microbiota (105 SFB in 1011 total bacteria). Bacteria with filamentous segmented morphology were observed in intestinal fluids via fluorescent in situ hybridization, and from gut biopsies via scanning electron microscopy. SFB-specific DNA and peptide fragments were also identified via multiple displacement amplification PCR and mass spectrometry. There was an overall positive correlation between the presence of SFB and the titer of total secretory immunoglobulin A (sIgA), which is more apparent in intestinal fluids of the age group of 8-36 months. Afterward there was a decline of SFB in numbers correlated with a reduction of total sIgA. RT-qPCR analysis of the terminal ileal biopsies revealed that the expression of Th17 pathway genes were induced in SFB-positive samples, while the markers of T and B cell receptor signaling pathways were also upregulated. Collectively, these data suggest that SFB is a rare member of microbiota, and may play an important role in the development of human gut immunity.
Collapse
Affiliation(s)
- Bo Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases–Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Gastroenterology, Children’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Huahai Chen
- State Key Laboratory of Breeding Base for Zhejiang Sustainable Pest–Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Xiaoli Shu
- Department of Gastroenterology, Children’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yeshi Yin
- State Key Laboratory of Breeding Base for Zhejiang Sustainable Pest–Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Jia Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases–Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | | | - Lijun Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases–Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kerong Peng
- Department of Gastroenterology, Children’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Fei Xu
- State Key Laboratory of Breeding Base for Zhejiang Sustainable Pest–Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Weizhong Gu
- Department of Gastroenterology, Children’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hong Zhao
- Department of Gastroenterology, Children’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Liqin Jiang
- Department of Gastroenterology, Children’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases–Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jian Song
- Advanced Information Systems, Exelis, Herndon, VA, United States
| | - Yoram Elitsur
- Department of Pediatrics, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, United States
| | - Hongwei D. Yu
- Department of Pediatrics, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, United States
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, United States
| | - Mizu Jiang
- Department of Gastroenterology, Children’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xin Wang
- State Key Laboratory of Breeding Base for Zhejiang Sustainable Pest–Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Charlie Xiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases–Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
18
|
Flannigan KL, Denning TL. Segmented filamentous bacteria-induced immune responses: a balancing act between host protection and autoimmunity. Immunology 2018; 154:537-546. [PMID: 29771448 PMCID: PMC6050222 DOI: 10.1111/imm.12950] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 04/15/2018] [Accepted: 04/18/2018] [Indexed: 02/07/2023] Open
Abstract
Segmented filamentous bacteria (SFB) are Gram-positive, spore-forming, bacteria that primarily colonize the ileum of the small intestine. Upon direct adherence to intestinal epithelial cells, SFB actively stimulate innate and adaptive immune cell activation. The cardinal features of SFB-induced gut immunity - T helper type 17 (Th17) cell differentiation, IgA production and barrier protection - lead to the containment of SFB and further afford protection against invading pathogens. Th17 cells and interleukin-17A, however, can also reach peripheral sites and exacerbate autoimmunity. In this review, we highlight salient characteristics of SFB-host interactions and detail the cellular and molecular immune mechanisms involved in coordinating these responses.
Collapse
Affiliation(s)
- Kyle L. Flannigan
- Department of Physiology and PharmacologyCumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
- Snyder Institute for Chronic DiseasesUniversity of CalgaryCalgaryAlbertaCanada
| | | |
Collapse
|
19
|
Finotti A, Gasparello J, Lampronti I, Cosenza LC, Maconi G, Matarese V, Gentili V, Di Luca D, Gambari R, Caselli M. PCR detection of segmented filamentous bacteria in the terminal ileum of patients with ulcerative colitis. BMJ Open Gastroenterol 2017; 4:e000172. [PMID: 29259792 PMCID: PMC5728270 DOI: 10.1136/bmjgast-2017-000172] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 10/31/2017] [Accepted: 11/17/2017] [Indexed: 02/07/2023] Open
Abstract
Objectives Segmented filamentous bacteria (SFB) have been detected in a wide range of different animal. Recently, the presence of SFB-like bacteria was shown in biopsies of the terminal ileum and ileocecal valve of both patients with ulcerative colitis and control subjects. The aim of this study was to verify whether PCR methods could be used for the detection of SFB in biopsy of patients with ulcerative colitis and its relationships with the disease stage. Methods PCR methods were used to identify SFB in biopsies from the terminal ileum of patients with ulcerative colitis, showing that this approach represents a useful tool for the detection of SFB presence and analysis of the bacterial load. Results Our analysis detected SFB in all faecal samples of children at the time of weaning, and also show that putative SFB sequences are present in both patients with ulcerative colitis and control subjects. Results obtained using real-time quantitative PCR analysis confirm the presence of putative SFB sequences in samples from the terminal ileum of patients with ulcerative colitis and in control subjects. Conclusions The presence of putative SFB sequence in both patients with ulcerative colitis and control subject suggests that SFB cannot be considered as being uniquely associated with the disease. The second conclusion is that among the patients with ulcerative colitis, a tendency does exist for active disease samples to show higher SFB load, opening new perspectives about possible identification and pharmacological manipulation of SFB-mediated processes for new therapeutic strategy.
Collapse
Affiliation(s)
- Alessia Finotti
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Jessica Gasparello
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Ilaria Lampronti
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Lucia Carmela Cosenza
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Giovanni Maconi
- Department of Gastroenterology, Sacco Hospital, University of Milan, Milan, Italy
| | | | - Valentina Gentili
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Dario Di Luca
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Roberto Gambari
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Michele Caselli
- School of Gastroenterology, University of Ferrara, Ferrara, Italy
| |
Collapse
|
20
|
Ericsson AC, Personett AR, Turner G, Dorfmeyer RA, Franklin CL. Variable Colonization after Reciprocal Fecal Microbiota Transfer between Mice with Low and High Richness Microbiota. Front Microbiol 2017; 8:196. [PMID: 28280484 PMCID: PMC5322181 DOI: 10.3389/fmicb.2017.00196] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 01/26/2017] [Indexed: 12/29/2022] Open
Abstract
Several associations have been made between characteristics of the resident gut microbiota and human health and disease susceptibility. Animal models provide the means to test these correlations prospectively and evaluate causality. Experimental fecal microbiota transfer (FMT), or the intentional transplantation of gut microbes into recipient mice depleted of their autochthonous microbes with antibiotics, is a commonly used method of testing these relationships. The true completeness of microbial transfer through such procedures is poorly documented in the literature, particularly in the context of reciprocal transfer of microbes between recipient and donor mice harboring microbial populations of differing richness and diversity. Moreover, it is unclear whether the use of frozen fecal contents or cecal contents would confer any difference in the outcomes of transfer. Herein, groups of mice colonized with distinct gut microbiota of differing richness and composition were used in a reciprocal FMT study, with different groups receiving transfer of material prepared from fresh cecal contents, fresh feces, or frozen feces. Targeted 16S rRNA gene amplicon sequencing was used at intervals throughout the study to characterize the microbiota. Notably, despite comparable depletion of the microbiota in recipient mice prior to transfer, donor-specific taxa reliably colonized recipients only when relatively rich donor material was transferred to mice originally colonized with a simpler microbiota. It is unclear whether these differences were due to differences in the endogenous recipient microbiota or host factors induced in early life by microbial factors. These findings are of practical import for researchers using FMT to prospectively assess the influence of the gut microbiota in mouse models, and to those studying host-microbial interactions and their influence on gut barrier function.
Collapse
Affiliation(s)
- Aaron C Ericsson
- University of Missouri Metagenomics Center, University of MissouriColumbia, MO, USA; University of Missouri Mutant Mouse Resource and Research Center, University of MissouriColumbia, MO, USA; Comparative Metagenomics Laboratory, Department of Veterinary Pathobiology, University of MissouriColumbia, MO, USA
| | - Alexa R Personett
- Comparative Metagenomics Laboratory, Department of Veterinary Pathobiology, University of Missouri Columbia, MO, USA
| | - Giedre Turner
- University of Missouri Mutant Mouse Resource and Research Center, University of MissouriColumbia, MO, USA; Comparative Metagenomics Laboratory, Department of Veterinary Pathobiology, University of MissouriColumbia, MO, USA
| | - Rebecca A Dorfmeyer
- University of Missouri Mutant Mouse Resource and Research Center, University of MissouriColumbia, MO, USA; Comparative Metagenomics Laboratory, Department of Veterinary Pathobiology, University of MissouriColumbia, MO, USA
| | - Craig L Franklin
- University of Missouri Metagenomics Center, University of MissouriColumbia, MO, USA; University of Missouri Mutant Mouse Resource and Research Center, University of MissouriColumbia, MO, USA; Comparative Metagenomics Laboratory, Department of Veterinary Pathobiology, University of MissouriColumbia, MO, USA
| |
Collapse
|
21
|
Hansen AK, Krych Ł, Nielsen DS, Hansen CHF. A Review of Applied Aspects of Dealing with Gut Microbiota Impact on Rodent Models. ILAR J 2016; 56:250-64. [PMID: 26323634 DOI: 10.1093/ilar/ilv010] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The gut microbiota (GM) affects numerous human diseases, as well as rodent models for these. We will review this impact and summarize ways to handle this challenge in animal research. The GM is complex, with the largest fractions being the gram-positive phylum Firmicutes and the gram-negative phylum Bacteroidetes. Other important phyla are the gram-negative phyla Proteobacteria and Verrucomicrobia, and the gram-positive phylum Actinobacteria. GM members influence models for diseases, such as inflammatory bowel diseases, allergies, autoimmunity, cancer, and neuropsychiatric diseases. GM characterization of all individual animals and incorporation of their GM composition in data evaluation may therefore be considered in future protocols. Germfree isolator-housed rodents or rodents made virtually germ free by antibiotic cocktails can be used to study diverse microbial influences on disease expression. Through subsequent inoculation with selected strains or cocktails of microbes, new "defined flora" models can yield valuable knowledge on the impact of the GM, and of specific GM members and their interactions, on important disease phenotypes and mechanisms. Rodent husbandry and microbial quality assurance practices will be important to ensure and confirm appropriate and research relevant GM.
Collapse
Affiliation(s)
- Axel Kornerup Hansen
- Axel Kornerup Hansen, DVM, DVsc, DipECLAM, Professor, Section of Experimental Animal Models, Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Thorvaldsensvej 57, 1871 Frederiksberg C, Denmark. Łukasz Krych, MSc, PhD, Postdoc, Department of Food Science, Faculty of Science, University of Copenhagen, Rolighedsvej 26, 1958 Frederiksberg C, Denmark. Dennis Sandris Nielsen, MSc, PhD, Associate Professor, Department of Food Science, Faculty of Science, University of Copenhagen, Rolighedsvej 26, 1958 Frederiksberg C, Denmark. Camilla Hartmann Friis Hansen, DVM, PhD, Assistant Professor, Section of Experimental Animal Models, Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Thorvaldsensvej 57, 1871 Frederiksberg C, Denmark
| | - Łukasz Krych
- Axel Kornerup Hansen, DVM, DVsc, DipECLAM, Professor, Section of Experimental Animal Models, Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Thorvaldsensvej 57, 1871 Frederiksberg C, Denmark. Łukasz Krych, MSc, PhD, Postdoc, Department of Food Science, Faculty of Science, University of Copenhagen, Rolighedsvej 26, 1958 Frederiksberg C, Denmark. Dennis Sandris Nielsen, MSc, PhD, Associate Professor, Department of Food Science, Faculty of Science, University of Copenhagen, Rolighedsvej 26, 1958 Frederiksberg C, Denmark. Camilla Hartmann Friis Hansen, DVM, PhD, Assistant Professor, Section of Experimental Animal Models, Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Thorvaldsensvej 57, 1871 Frederiksberg C, Denmark
| | - Dennis Sandris Nielsen
- Axel Kornerup Hansen, DVM, DVsc, DipECLAM, Professor, Section of Experimental Animal Models, Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Thorvaldsensvej 57, 1871 Frederiksberg C, Denmark. Łukasz Krych, MSc, PhD, Postdoc, Department of Food Science, Faculty of Science, University of Copenhagen, Rolighedsvej 26, 1958 Frederiksberg C, Denmark. Dennis Sandris Nielsen, MSc, PhD, Associate Professor, Department of Food Science, Faculty of Science, University of Copenhagen, Rolighedsvej 26, 1958 Frederiksberg C, Denmark. Camilla Hartmann Friis Hansen, DVM, PhD, Assistant Professor, Section of Experimental Animal Models, Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Thorvaldsensvej 57, 1871 Frederiksberg C, Denmark
| | - Camilla Hartmann Friis Hansen
- Axel Kornerup Hansen, DVM, DVsc, DipECLAM, Professor, Section of Experimental Animal Models, Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Thorvaldsensvej 57, 1871 Frederiksberg C, Denmark. Łukasz Krych, MSc, PhD, Postdoc, Department of Food Science, Faculty of Science, University of Copenhagen, Rolighedsvej 26, 1958 Frederiksberg C, Denmark. Dennis Sandris Nielsen, MSc, PhD, Associate Professor, Department of Food Science, Faculty of Science, University of Copenhagen, Rolighedsvej 26, 1958 Frederiksberg C, Denmark. Camilla Hartmann Friis Hansen, DVM, PhD, Assistant Professor, Section of Experimental Animal Models, Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Thorvaldsensvej 57, 1871 Frederiksberg C, Denmark
| |
Collapse
|
22
|
Immunometabolism of obesity and diabetes: microbiota link compartmentalized immunity in the gut to metabolic tissue inflammation. Clin Sci (Lond) 2015; 129:1083-96. [PMID: 26464517 DOI: 10.1042/cs20150431] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The bacteria that inhabit us have emerged as factors linking immunity and metabolism. Changes in our microbiota can modify obesity and the immune underpinnings of metabolic diseases such as Type 2 diabetes. Obesity coincides with a low-level systemic inflammation, which also manifests within metabolic tissues such as adipose tissue and liver. This metabolic inflammation can promote insulin resistance and dysglycaemia. However, the obesity and metabolic disease-related immune responses that are compartmentalized in the intestinal environment do not necessarily parallel the inflammatory status of metabolic tissues that control blood glucose. In fact, a permissive immune environment in the gut can exacerbate metabolic tissue inflammation. Unravelling these discordant immune responses in different parts of the body and establishing a connection between nutrients, immunity and the microbiota in the gut is a complex challenge. Recent evidence positions the relationship between host gut barrier function, intestinal T cell responses and specific microbes at the crossroads of obesity and inflammation in metabolic disease. A key problem to be addressed is understanding how metabolite, immune or bacterial signals from the gut are relayed and transferred into systemic or metabolic tissue inflammation that can impair insulin action preceding Type 2 diabetes.
Collapse
|
23
|
Paramasiva I, Shouche Y, Kulkarni GJ, Krishnayya PV, Akbar SM, Sharma HC. Diversity in gut microflora of Helicoverpa armigera populations from different regions in relation to biological activity of Bacillus thuringiensis δ-endotoxin Cry1Ac. ARCHIVES OF INSECT BIOCHEMISTRY AND PHYSIOLOGY 2014; 87:201-213. [PMID: 25195523 DOI: 10.1002/arch.21190] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Transgenic crops expressing toxin proteins from Bacillus thuringiensis (Bt) have been deployed on a large scale for management of Helicoverpa armigera. Resistance to Bt toxins has been documented in several papers, and therefore, we examined the role of midgut microflora of H. armigera in its susceptibility to Bt toxins. The susceptibility of H. armigera to Bt toxin Cry1Ac was assessed using Log-dose-Probit analysis, and the microbial communities were identified by 16S rRNA sequencing. The H. armigera populations from nine locations harbored diverse microbial communities, and had some unique bacteria, suggesting a wide geographical variation in microbial community in the midgut of the pod borer larvae. Phylotypes belonging to 32 genera were identified in the H. armigera midgut in field populations from nine locations. Bacteria belonging to Enterobacteriaceae (Order Bacillales) were present in all the populations, and these may be the common members of the H. armigera larval midgut microflora. Presence and/or absence of certain species were linked to H. armigera susceptibility to Bt toxins, but there were no clear trends across locations. Variation in susceptibility of F1 neonates of H. armigera from different locations to the Bt toxin Cry1Ac was found to be 3.4-fold. These findings support the idea that insect migut microflora may influence the biological activity of Bt toxins.
Collapse
Affiliation(s)
- Inakarla Paramasiva
- Department of Entomology, Agricultural College, Acharya N.G. Ranga Agriculture University, Bapatla, Andhra Pradesh, India; Entomology, International Crops Research Institute for the Semi-Arid Tropics (ICRISAT), Patancheru, Telangana, India
| | | | | | | | | | | |
Collapse
|
24
|
Yoon MY, Yoon MY, Lee K, Yoon SS. Protective role of gut commensal microbes against intestinal infections. J Microbiol 2014; 52:983-9. [PMID: 25467115 DOI: 10.1007/s12275-014-4655-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 11/19/2014] [Accepted: 11/19/2014] [Indexed: 01/06/2023]
Abstract
The human gastrointestinal tract is colonized by multitudes of microorganisms that exert beneficial effects on human health. Mounting evidence suggests that intestinal microbiota contributes to host resistance against enteropathogenic bacterial infection. However, molecular details that account for such an important role has just begun to be understood. The commensal microbes in the intestine regulate gut homeostasis through activating the development of host innate immunity and producing molecules with antimicrobial activities that directly inhibit propagation of pathogenic bacteria. Understanding the protective roles of gut microbiota will provide a better insight into the molecular basis that underlies complicated interaction among host-pathogen-symbiont. In this review, we highlighted recent findings that help us broaden our knowledge of the intestinal ecosystem and thereby come up with a better strategy for combating enteropathogenic infection.
Collapse
Affiliation(s)
- Mi Young Yoon
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul, 120-752, Republic of Korea
| | - My Young Yoon
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul, 120-752, Republic of Korea
| | | | | |
Collapse
|
25
|
Dzutsev A, Goldszmid RS, Viaud S, Zitvogel L, Trinchieri G. The role of the microbiota in inflammation, carcinogenesis, and cancer therapy. Eur J Immunol 2014; 45:17-31. [PMID: 25328099 DOI: 10.1002/eji.201444972] [Citation(s) in RCA: 202] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Revised: 08/19/2014] [Accepted: 10/13/2014] [Indexed: 12/11/2022]
Abstract
Commensal microorganisms colonize barrier surfaces of all multicellular organisms, including those of humans. For more than 500 million years, commensal microorganisms and their hosts have coevolved and adapted to each other. As a result, the commensal microbiota affects many immune and nonimmune functions of their hosts, and de facto the two together comprise one metaorganism. The commensal microbiota communicates with the host via biologically active molecules. Recently, it has been reported that microbial imbalance may play a critical role in the development of multiple diseases, such as cancer, autoimmune conditions, and increased susceptibility to infection. In this review, we focus on the role of the commensal microbiota in the development, progression, and immune evasion of cancer, as well as some modulatory effects on the treatment of cancer. In particular, we discuss the mechanisms of microbiota-mediated regulation of innate and adaptive immune responses to tumors, and the consequences on cancer progression and whether tumors subsequently become resistant or susceptible to different anticancer therapeutic regiments.
Collapse
Affiliation(s)
- Amiran Dzutsev
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA; Leidos Biomedical Research, Inc, Frederick, MD, USA
| | | | | | | | | |
Collapse
|
26
|
Paramasiva I, Sharma HC, Krishnayya PV. Antibiotics influence the toxicity of the delta endotoxins of Bacillus thuringiensis towards the cotton bollworm, Helicoverpa armigera. BMC Microbiol 2014; 14:200. [PMID: 25059716 PMCID: PMC4222728 DOI: 10.1186/1471-2180-14-200] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Accepted: 07/12/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The cotton bollworm, Helicoverpa armigera is one of the most important crop pests worldwide. It has developed high levels of resistance to synthetic insecticides, and hence, Bacillus thuringiensis (Bt) formulations are used as a safer pesticide and the Bt genes have been deployed in transgenic crops for controlling this pest. There is an apprehension that H. armigera might develop resistance to transgenic crops in future. Therefore, we studied the role of gut microbes by eliminating them with antibiotics in H. armigera larvae on the toxicity of Bt toxins against this pest. RESULTS Commercial formulation of Bt (Biolep®) and the pure Cry1Ab and Cry1Ac toxin proteins were evaluated at ED50, LC50, and LC90 dosages against the H. armigera larvae with and without antibiotics (which removed the gut microbes). Lowest H. armigera larval mortality due to Bt formulation and the Bt toxins Cry1Ab and Cry1Ac was recorded in insects reared on diets with 250 and 500 μg ml-1 diet of each of the four antibiotics (gentamicin, penicillin, rifampicin, and streptomycin), while the highest larval mortality was recorded in insects reared on diets without the antibiotics. Mortality of H. armigera larvae fed on diets with Bt formulation and the δ-endotoxins Cry1Ab and Cry1Ac was inversely proportional to the concentration of antibiotics in the artificial diet. Nearly 30% reduction in larval mortality was observed in H. armigera larvae from F1 to F3 generation when the larvae were reared on diets without antibiotics (with gut microbes) and fed on 0.15% Bt or 12 μg Cry1Ab or Cry1Ac ml-1 diet, indicating development of resistance to Bt in the presence of gut microflora. However, there were no differences in larval mortality due to Bt, Cry1Ab or Cry1Ac across generations in insects when they were reared on diets with 250 μg of each antibiotic ml-1 diet (without gut microflora). CONCLUSIONS The results suggested that antibiotics which eliminated gut microflora influenced the toxicity of Bt towards H. armigera, and any variation in diversity and abundance of gut microflora will have a major bearing on development of resistance to Bt toxins applied as foliar sprays or deployed in transgenic crops for pest management.
Collapse
Affiliation(s)
| | - Hari C Sharma
- International Crops Research Institute for the Semi-Arid Tropics (ICRISAT), Patancheru 502 324 Andhra Pradesh, India.
| | | |
Collapse
|
27
|
Schnupf P, Gaboriau-Routhiau V, Cerf-Bensussan N. Host interactions with Segmented Filamentous Bacteria: an unusual trade-off that drives the post-natal maturation of the gut immune system. Semin Immunol 2013; 25:342-51. [PMID: 24184014 DOI: 10.1016/j.smim.2013.09.001] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Segmented Filamentous Bacteria (SFB) are present in the gut microbiota of a large number of vertebrate species where they are found intimately attached to the intestinal epithelium. SFB has recently attracted considerable attention due to its outstanding capacity to stimulate innate and adaptive host immune responses without causing pathology. Recent genomic analysis placed SFB between obligate and facultative symbionts, unraveled its highly auxotrophic needs, and provided a rationale for the complex SFB life-style in close contact with the epithelium. Herein, we examine how the SFB life-style may underlie its potent immunostimulatory properties and discuss how the trade-off set up between SFB and its hosts can simultaneously help to establish and maintain the ecological niche of SFB in the intestine and drive the post-natal maturation of the host gut immune barrier.
Collapse
Affiliation(s)
- Pamela Schnupf
- INSERM, U989, 75014 Paris, France; Université Paris Descartes, Sorbonne Paris Cité, and Institut IMAGINE, 75015 Paris, France; Institut Pasteur, Unité de Pathogénie Microbienne Moleculaire, 25-28 rue du Dr. Roux, 75015 Paris, France
| | | | | |
Collapse
|
28
|
Abstract
Commensal bacteria are necessary for the development and maintenance of a healthy immune system. Harnessing the ability of microbiota to affect host immunity is considered an important therapeutic strategy for many mucosal and nonmucosal immune-related conditions, such as inflammatory bowel diseases (IBDs), celiac disease, metabolic syndrome, diabetes, and microbial infections. In addition to well-established immunostimulatory effects of the microbiota, the presence of individual mutualistic commensal bacteria with immunomodulatory effects has been described. These organisms are permanent members of the commensal microbiota and affect host immune homeostasis in specific ways. Identification of individual examples of such immunomodulatory commensals and understanding their mechanisms of interaction with the host will be invaluable in designing therapeutic strategies to reverse intestinal dysbiosis and recover immunological homeostasis.
Collapse
Affiliation(s)
- Ivaylo I Ivanov
- Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY 10032, USA.
| | | |
Collapse
|
29
|
Intestinal epithelial cells as mediators of the commensal-host immune crosstalk. Immunol Cell Biol 2013; 91:204-14. [PMID: 23318659 DOI: 10.1038/icb.2012.80] [Citation(s) in RCA: 113] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Commensal bacteria regulate the homeostasis of host effector immune cell subsets. The mechanisms involved in this commensal-host crosstalk are not well understood. Intestinal epithelial cells (IECs) not only create a physical barrier between the commensals and immune cells in host tissues, but also facilitate interactions between them. Perturbations of epithelial homeostasis or function lead to the development of intestinal disorders such as inflammatory bowel diseases (IBD) and intestinal cancer. IECs receive signals from commensals and produce effector immune molecules. IECs also affect the function of immune cells in the lamina propria. Here we discuss some of these properties of IECs that define them as innate immune cells. We focus on how IECs may integrate and transmit signals from individual commensal bacteria to mucosal innate and adaptive immune cells for the establishment of the unique mucosal immunological equilibrium.
Collapse
|
30
|
Liao N, Yin Y, Sun G, Xiang C, Liu D, Yu HD, Wang X. Colonization and distribution of segmented filamentous bacteria (SFB) in chicken gastrointestinal tract and their relationship with host immunity. FEMS Microbiol Ecol 2012; 81:395-406. [PMID: 22429007 DOI: 10.1111/j.1574-6941.2012.01362.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2011] [Revised: 03/03/2012] [Accepted: 03/06/2012] [Indexed: 11/30/2022] Open
Abstract
Uncultivable segmented filamentous bacteria (SFB) reside in the gastrointestinal (GI) tract of mammals and can boost the host immunity. Immunoglobulin A (IgA) from mother's milk has been previously shown to be a key factor in regulating SFB colonization. Because neonatal chicken cannot acquire IgA from maternal milk, they are a good model to examine the role of IgA in SFB colonization. Here, we used the fluorescent in situ hybridization (FISH) and quantitative PCR (qPCR) to monitor the colonization and distribution of SFB in chickens aged from 2-day-old to 6-week-old. Early SFB colonization, which primarily occurred in the ileal mucosa (< 13 days old), was IgA independent. From the age of 17-42 days, there was an increase in IgA in the gut mucosa, which was correlated with a decrease in SFB. To examine the effect of probiotics and immunosuppression on SFB colonization, we treated the chickens by feeding them Lactobacillus delbrueckii or giving them a subcutaneous injection of cyclophosphamide (CTX). Feeding lactobacilli at birth rendered SFB colonization occurring 4 days earlier, while CTX treatment increases the SFB colonization through reducing the other non-SFB bacteria. Altogether, our data suggest that early colonization of SFB in chicken occurs independently of IgA and the population of SFB in the GI tract of chicken may be manipulated from birth via probiotic or CTX treatment.
Collapse
Affiliation(s)
- Ningbo Liao
- State Key Laboratory Breeding Base for Zhejiang Sustainable Pest and Disease Control, Institute of Plant Protection and Microbiology, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | | | | | | | | | | | | |
Collapse
|
31
|
Pamp SJ, Harrington ED, Quake SR, Relman DA, Blainey PC. Single-cell sequencing provides clues about the host interactions of segmented filamentous bacteria (SFB). Genome Res 2012; 22:1107-19. [PMID: 22434425 PMCID: PMC3371716 DOI: 10.1101/gr.131482.111] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Segmented filamentous bacteria (SFB) are host-specific intestinal symbionts that comprise a distinct clade within the Clostridiaceae, designated Candidatus Arthromitus. SFB display a unique life cycle within the host, involving differentiation into multiple cell types. The latter include filaments that attach intimately to intestinal epithelial cells, and from which "holdfasts" and spores develop. SFB induce a multifaceted immune response, leading to host protection from intestinal pathogens. Cultivation resistance has hindered characterization of these enigmatic bacteria. In the present study, we isolated five SFB filaments from a mouse using a microfluidic device equipped with laser tweezers, generated genome sequences from each, and compared these sequences with each other, as well as to recently published SFB genome sequences. Based on the resulting analyses, SFB appear to be dependent on the host for a variety of essential nutrients. SFB have a relatively high abundance of predicted proteins devoted to cell cycle control and to envelope biogenesis, and have a group of SFB-specific autolysins and a dynamin-like protein. Among the five filament genomes, an average of 8.6% of predicted proteins were novel, including a family of secreted SFB-specific proteins. Four ADP-ribosyltransferase (ADPRT) sequence types, and a myosin-cross-reactive antigen (MCRA) protein were discovered; we hypothesize that they are involved in modulation of host responses. The presence of polymorphisms among mouse SFB genomes suggests the evolution of distinct SFB lineages. Overall, our results reveal several aspects of SFB adaptation to the mammalian intestinal tract.
Collapse
Affiliation(s)
- Sünje J Pamp
- Department of Microbiology and Immunology, The Howard Hughes Medical Institute
| | | | | | | | | |
Collapse
|
32
|
Complete genome sequences of rat and mouse segmented filamentous bacteria, a potent inducer of th17 cell differentiation. Cell Host Microbe 2012; 10:273-84. [PMID: 21925114 DOI: 10.1016/j.chom.2011.08.007] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2011] [Revised: 08/04/2011] [Accepted: 08/17/2011] [Indexed: 12/12/2022]
Abstract
Segmented filamentous bacteria (SFB) are noncultivable commensals inhabiting the gut of various vertebrate species and have been shown to induce Th17 cells in mice. We present the complete genome sequences of both rat and mouse SFB isolated from SFB-monocolonized hosts. The rat and mouse SFB genomes each harbor a single circular chromosome of 1.52 and 1.59 Mb encoding 1346 and 1420 protein-coding genes, respectively. The overall nucleotide identity between the two genomes is 86%, and the substitution rate was estimated to be similar to that of the free-living E. coli. SFB genomes encode typical genes for anaerobic fermentation and spore and flagella formation, but lack most of the amino acid biosynthesis enzymes, reminiscent of pathogenic Clostridia, exhibiting large dependency on the host. However, SFB lack most of the clostridial virulence-related genes. Comparative analysis with clostridial genomes suggested possible mechanisms for host responses and specific adaptations in the intestine.
Collapse
|
33
|
Bleich A, Hansen AK. Time to include the gut microbiota in the hygienic standardisation of laboratory rodents. Comp Immunol Microbiol Infect Dis 2012; 35:81-92. [PMID: 22257867 DOI: 10.1016/j.cimid.2011.12.006] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2011] [Revised: 11/17/2011] [Accepted: 12/19/2011] [Indexed: 02/06/2023]
Abstract
The gut microbiota (GM) composition and its impact on animal experiments has become currently dramatically relevant in our days: (1) recent progress in metagenomic technologies, (2) the availability of large scale quantitative analyses to characterize even subtle phenotypes, (3) the limited diversity of laboratory rodent GM due to strict barriers at laboratory animal vendors, and (4) the availability of up to 300.000 different transgenic mouse strains from different sources displaying a huge variety in their GM composition. In this review the GM is described as a variable in animal experiments which need to be reduced for scientific as well as ethical reasons, and strategies how to implement this in routine diagnostic procedures are proposed. We conclude that we have both enough information available to state that the GM has an essential impact on animal models, as well as the methods available to start dealing with these impacts.
Collapse
Affiliation(s)
- André Bleich
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany, Hannover, Germany.
| | | |
Collapse
|
34
|
Reading NC, Kasper DL. The starting lineup: key microbial players in intestinal immunity and homeostasis. Front Microbiol 2011; 2:148. [PMID: 21779278 PMCID: PMC3133820 DOI: 10.3389/fmicb.2011.00148] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2011] [Accepted: 06/26/2011] [Indexed: 12/29/2022] Open
Abstract
The complexity of microbiota inhabiting the intestine is increasingly apparent. Delicate balance of numerous bacterial species can affect development of the immune system, how susceptible a host is to pathogenic organisms, and the auto-inflammatory state of the host. In the last decade, with the increased use of germ-free mice, gnotobiotic mice, and animal models in which a germ-free animal has been colonized with a foreign microbiota such as humanized mice, it has been possible to delineate relationships that specific bacteria have with the host immune system and to show what role they may play in overall host health. These models have not only allowed us to tease out the roles of individual species, but have also allowed the discovery and characterization of functionally unknown organisms. For example, segmented filamentous bacteria (SFB) have been shown to play a vital role in expansion of IL-17 producing cells. Prior to linking their key role in immune system development, little was known about these organisms. Bacteroides fragilis can rescue some of the immune defects of gnotobiotic mice after mono-colonization and have anti-inflammatory properties that can alleviate colitis and experimental allergic encephalitis in murine models. Additionally, Clostridium species have most recently been shown to expand regulatory T-cell populations leading to anti-inflammatory conditions. This review will highlight and summarize some of the major findings within the last decade concerning the role of select groups of bacteria including SFB, Clostridium, Bacteroides, Bifidobacterium, and Lactobacillus, and their impact on host mucosal immune systems.
Collapse
Affiliation(s)
- Nicola C Reading
- Channing Laboratory, Department of Medicine, Brigham and Women's Hospital Boston, MA, USA
| | | |
Collapse
|
35
|
Abstract
Gut microbiota is an assortment of microorganisms inhabiting the length and width of the mammalian gastrointestinal tract. The composition of this microbial community is host specific, evolving throughout an individual's lifetime and susceptible to both exogenous and endogenous modifications. Recent renewed interest in the structure and function of this “organ” has illuminated its central position in health and disease. The microbiota is intimately involved in numerous aspects of normal host physiology, from nutritional status to behavior and stress response. Additionally, they can be a central or a contributing cause of many diseases, affecting both near and far organ systems. The overall balance in the composition of the gut microbial community, as well as the presence or absence of key species capable of effecting specific responses, is important in ensuring homeostasis or lack thereof at the intestinal mucosa and beyond. The mechanisms through which microbiota exerts its beneficial or detrimental influences remain largely undefined, but include elaboration of signaling molecules and recognition of bacterial epitopes by both intestinal epithelial and mucosal immune cells. The advances in modeling and analysis of gut microbiota will further our knowledge of their role in health and disease, allowing customization of existing and future therapeutic and prophylactic modalities.
Collapse
Affiliation(s)
- Inna Sekirov
- Michael Smith Laboratories, Department of Microbiology and Immunology, and Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Shannon L. Russell
- Michael Smith Laboratories, Department of Microbiology and Immunology, and Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, British Columbia, Canada
| | - L. Caetano M. Antunes
- Michael Smith Laboratories, Department of Microbiology and Immunology, and Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, British Columbia, Canada
| | - B. Brett Finlay
- Michael Smith Laboratories, Department of Microbiology and Immunology, and Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
36
|
Wu HJ, Ivanov II, Darce J, Hattori K, Shima T, Umesaki Y, Littman DR, Benoist C, Mathis D. Gut-residing segmented filamentous bacteria drive autoimmune arthritis via T helper 17 cells. Immunity 2010; 32:815-27. [PMID: 20620945 PMCID: PMC2904693 DOI: 10.1016/j.immuni.2010.06.001] [Citation(s) in RCA: 1233] [Impact Index Per Article: 82.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2009] [Revised: 03/30/2010] [Accepted: 05/12/2010] [Indexed: 02/07/2023]
Abstract
Commensal microbes can have a substantial impact on autoimmune disorders, but the underlying molecular and cellular mechanisms remain largely unexplored. We report that autoimmune arthritis was strongly attenuated in the K/BxN mouse model under germ-free (GF) conditions, accompanied by reductions in serum autoantibody titers, splenic autoantibody-secreting cells, germinal centers, and the splenic T helper 17 (Th17) cell population. Neutralization of interleukin-17 prevented arthritis development in specific-pathogen-free K/BxN mice resulting from a direct effect of this cytokine on B cells to inhibit germinal center formation. The systemic deficiencies of the GF animals reflected a loss of Th17 cells from the small intestinal lamina propria. Introduction of a single gut-residing species, segmented filamentous bacteria, into GF animals reinstated the lamina propria Th17 cell compartment and production of autoantibodies, and arthritis rapidly ensued. Thus, a single commensal microbe, via its ability to promote a specific Th cell subset, can drive an autoimmune disease.
Collapse
Affiliation(s)
- Hsin-Jung Wu
- Department of Pathology, Harvard Medical School, Boston, MA 02115
- Section on Immunology and Immunogenetics, Joslin Diabetes Center, Boston, MA 02215
| | - Ivaylo I. Ivanov
- Molecular Pathogenesis Program, The Kimmel Center for Biology and Medicine of the Skirball Institute, New York University School of Medicine, New York, NY 10016
| | - Jaime Darce
- Department of Pathology, Harvard Medical School, Boston, MA 02115
- Section on Immunology and Immunogenetics, Joslin Diabetes Center, Boston, MA 02215
| | - Kimie Hattori
- Department of Pathology, Harvard Medical School, Boston, MA 02115
- Section on Immunology and Immunogenetics, Joslin Diabetes Center, Boston, MA 02215
| | - Tatsuichiro Shima
- Yakult Central Institute for Microbiological Research, Yaho 1796, Kunitachi, Tokyo 186-8650, Japan
| | - Yoshinori Umesaki
- Yakult Central Institute for Microbiological Research, Yaho 1796, Kunitachi, Tokyo 186-8650, Japan
| | - Dan R. Littman
- Molecular Pathogenesis Program, The Kimmel Center for Biology and Medicine of the Skirball Institute, New York University School of Medicine, New York, NY 10016
- Howard Hughes Medical Institute, The Kimmel Center for Biology and Medicine of the Skirball Institute, New York University School of Medicine, New York, NY 10016
| | - Christophe Benoist
- Department of Pathology, Harvard Medical School, Boston, MA 02115
- Section on Immunology and Immunogenetics, Joslin Diabetes Center, Boston, MA 02215
- The Broad Institute, Cambridge, MA 02142
| | - Diane Mathis
- Department of Pathology, Harvard Medical School, Boston, MA 02115
- Section on Immunology and Immunogenetics, Joslin Diabetes Center, Boston, MA 02215
- The Broad Institute, Cambridge, MA 02142
| |
Collapse
|
37
|
Del-Pozo J, Turnbull J, Ferguson H, Crumlish M. A comparative molecular study of the presence of "Candidatus arthromitus" in the digestive system of rainbow trout, Oncorhynchus mykiss (Walbaum), healthy and affected with rainbow trout gastroenteritis. JOURNAL OF FISH DISEASES 2010; 33:241-250. [PMID: 19912454 DOI: 10.1111/j.1365-2761.2009.01117.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Observations were made using histopathological techniques in conjunction with a nested polymerase chain reaction (PCR) protocol for the specific detection of "Candidatus arthromitus" on DNA extracted from wax-embedded tissues and fresh digestive contents of rainbow trout. Samples positive for "Candidatus arthromitus" DNA included fish with rainbow trout gastroenteritis (RTGE), clinically normal cohabiting fish, and apparently healthy controls from RTGE positive and RTGE negative sites. The results obtained from the PCR were confirmed by nucleotide sequencing. "Candidatus arthromitus" DNA was found in distal intestine as well as in sections of pyloric caeca, suggesting that both these locations are appropriate for molecular detection of "Candidatus arthromitus" DNA in trout. Furthermore, rainbow trout fry distal intestinal samples from two different hatcheries where RTGE had not been reported were also positive. Differences in "Candidatus arthromitus" DNA detection between paraffin wax-embedded and fresh digestive content samples from the same fish suggested that it may be predominantly epithelium-associated in healthy trout. Parallel histopathological observations indicated that pyloric caeca are the preferred site for visualizing segmented filamentous bacteria (SFB) in trout with RTGE. The results of this study showed that the presence of SFB was not invariably associated with clinical disease and that more information is required to understand the role of these organisms.
Collapse
Affiliation(s)
- J Del-Pozo
- Department of veterinary pathology, Easter Bush Veterinary Centre, Roslin, Midlothian, UK.
| | | | | | | |
Collapse
|
38
|
Caselli M, Holton J, Boldrini P, Vaira D, Calò G. Morphology of segmented filamentous bacteria and their patterns of contact with the follicle-associated epithelium of the mouse terminal ileum: implications for the relationship with the immune system. Gut Microbes 2010; 1:367-72. [PMID: 21468217 PMCID: PMC3056100 DOI: 10.4161/gmic.1.6.14390] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2010] [Revised: 11/04/2010] [Accepted: 12/06/2010] [Indexed: 02/03/2023] Open
Abstract
Recent evidence indicates that segmented filamentous bacteria (SFB), "Candidatus Arthromitus", play a unique role in different aspects of the maturation of the immune system, including T cell responses. Thus, it seems particularly relevant in this moment to shortly review the information on these bacteria and their relationship with the immune system, and to actively investigate their morphological aspects. We distinguished a developmental form from a vegetative form of these organisms. These different forms have distinct roles in the life cycle: the developmental form permits a rapid growth of the organisms while the vegetative form permits the attachment of SFB to the follicular epithelium. We have also given special attention to the modes of contact between SFB and the epithelial cells of the terminal ileum to better understand the unique relationship between these bacteria and the immune system.
Collapse
Affiliation(s)
- Michele Caselli
- School of Gastroenterology; Department of Experimental and Clinical Medicine; University of Ferrara; Ferrara, Italy
| | - John Holton
- University of Middlesex; Windeyer Insitiute of Medical Sciences; London, UK
| | - Paola Boldrini
- Center of Electron Microscopy; University of Ferrara; Ferrara, Italy
| | - Dino Vaira
- University College Hospital Trust; Windeyer Insitiute of Medical Sciences; London, UK
| | - Girolamo Calò
- Section of Pharmacology; University of Ferrara; Ferrara, Italy
| |
Collapse
|
39
|
Ivanov II, Atarashi K, Manel N, Brodie EL, Shima T, Karaoz U, Wei D, Goldfarb KC, Santee CA, Lynch SV, Tanoue T, Imaoka A, Itoh K, Takeda K, Umesaki Y, Honda K, Littman DR. Induction of intestinal Th17 cells by segmented filamentous bacteria. Cell 2009; 139:485-98. [PMID: 19836068 PMCID: PMC2796826 DOI: 10.1016/j.cell.2009.09.033] [Citation(s) in RCA: 3497] [Impact Index Per Article: 218.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2009] [Revised: 09/02/2009] [Accepted: 09/30/2009] [Indexed: 11/21/2022]
Abstract
The gastrointestinal tract of mammals is inhabited by hundreds of distinct species of commensal microorganisms that exist in a mutualistic relationship with the host. How commensal microbiota influence the host immune system is poorly understood. We show here that colonization of the small intestine of mice with a single commensal microbe, segmented filamentous bacterium (SFB), is sufficient to induce the appearance of CD4(+) T helper cells that produce IL-17 and IL-22 (Th17 cells) in the lamina propria. SFB adhere tightly to the surface of epithelial cells in the terminal ileum of mice with Th17 cells but are absent from mice that have few Th17 cells. Colonization with SFB was correlated with increased expression of genes associated with inflammation and antimicrobial defenses and resulted in enhanced resistance to the intestinal pathogen Citrobacter rodentium. Thus, manipulation of this commensal-regulated pathway may provide new opportunities for enhancing mucosal immunity and treating autoimmune disease.
Collapse
Affiliation(s)
- Ivaylo I. Ivanov
- Molecular Pathogenesis Program, The Kimmel Center for Biology and Medicine of the Skirball Institute, New York University School of Medicine, New York, NY 10016, USA
| | - Koji Atarashi
- Laboratory of Immune Regulation, Graduate School of Medicine, WPI Immunology Frontier Research Center, Osaka University, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Nicolas Manel
- Molecular Pathogenesis Program, The Kimmel Center for Biology and Medicine of the Skirball Institute, New York University School of Medicine, New York, NY 10016, USA
| | - Eoin L. Brodie
- Center for Environmental Biotechnology, Earth Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Tatsuichiro Shima
- Yakult Central Institute for Microbiological Research, Yaho 1796, Kunitachi, Tokyo 186-8650, Japan
| | - Ulas Karaoz
- Center for Environmental Biotechnology, Earth Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Dongguang Wei
- Carl Zeiss SMT, Inc., Nanotechnology Systems Div. One Corporation Way, Peabody, MA 01960, USA
| | - Katherine C. Goldfarb
- Center for Environmental Biotechnology, Earth Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Clark A. Santee
- Center for Environmental Biotechnology, Earth Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Susan V. Lynch
- Division of Gastroenterology, Department of Medicine, University of California San Francisco, 513 Parnassus Ave., San Francisco, CA 94143, USA
| | - Takeshi Tanoue
- Laboratory of Immune Regulation, Graduate School of Medicine, WPI Immunology Frontier Research Center, Osaka University, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Akemi Imaoka
- Yakult Central Institute for Microbiological Research, Yaho 1796, Kunitachi, Tokyo 186-8650, Japan
| | - Kikuji Itoh
- Department of Veterinary Public Health, University of Tokyo, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Kiyoshi Takeda
- Laboratory of Immune Regulation, Graduate School of Medicine, WPI Immunology Frontier Research Center, Osaka University, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Yoshinori Umesaki
- Yakult Central Institute for Microbiological Research, Yaho 1796, Kunitachi, Tokyo 186-8650, Japan
| | - Kenya Honda
- Laboratory of Immune Regulation, Graduate School of Medicine, WPI Immunology Frontier Research Center, Osaka University, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan
- Precursory Research for Embryonic Science and Technology (PREST), Japan Science and Technology Agency, 4-1-8 Honcho Kawaguchi, Saitama, 332-0012 Japan
| | - Dan R. Littman
- Molecular Pathogenesis Program, The Kimmel Center for Biology and Medicine of the Skirball Institute, New York University School of Medicine, New York, NY 10016, USA
- Howard Hughes Medical Institute, The Kimmel Center for Biology and Medicine of the Skirball Institute, New York University School of Medicine, New York, NY 10016, USA
| |
Collapse
|
40
|
Incharoen T, Yamauchi K. Production Performance, Egg Quality and Intestinal Histology in Laying Hens Fed Dietary Dried Fermented Ginger. ACTA ACUST UNITED AC 2009. [DOI: 10.3923/ijps.2009.1078.1085] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
41
|
Klaasen HLBM, Koopman JP, Vollaard EJ, Theeuwes AGM, Van Den Brink ME, Scholten PM, Bakker MH, Beynen AC. Influence of Antimicrobial Drugs on Segmented Filamentous Bacteria in the Ileum of Mice. MICROBIAL ECOLOGY IN HEALTH AND DISEASE 2009. [DOI: 10.3109/08910609109140155] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
| | - J. P. Koopman
- Central Animal Laboratory, Catholic University of Nijmegen, Nijmegen
| | - E. J. Vollaard
- Department of Pharmacy, Canisius-Wilhelmina Hospital, Nijmegen
| | - A. G. M. Theeuwes
- Department of Medical Statistics, Catholic University of Nijmegen, The Netherlands
| | | | - P. M. Scholten
- Central Animal Laboratory, Catholic University of Nijmegen, Nijmegen
| | - M. H. Bakker
- Central Animal Laboratory, Catholic University of Nijmegen, Nijmegen
| | - A. C. Beynen
- Department of Human Nutrition, Wageningen Agricultural University, The Netherlands
- Department of Laboratory Animal Science, State University of Utrecht, The Netherlands
| |
Collapse
|
42
|
Klaasen HLBM, Koopman JP, Scholten PM, Van Den Brink ME, Theeuwes AGM. Effect of Preventing Coprophagy on Colonisation by Segmented Filamentous Bacteria in the Small Bowel of Mice. MICROBIAL ECOLOGY IN HEALTH AND DISEASE 2009. [DOI: 10.3109/08910609009140123] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
| | - J. P. Koopman
- Central Animal Laboratory, Catholic University of Nijmegen, The Netherlands
| | - P. M. Scholten
- Central Animal Laboratory, Catholic University of Nijmegen, The Netherlands
| | | | - A. G. M. Theeuwes
- Department of Medical Statistics, Catholic University of Nijmegen, The Netherlands
| |
Collapse
|
43
|
Klaasen HLBM, Koopman JP, Van Den Brink ME, Scholten PM, Beynen AC. Influence of Macronutrients on Segmented Filamentous Bacteria in the Small Intestine of Mice. MICROBIAL ECOLOGY IN HEALTH AND DISEASE 2009. [DOI: 10.3109/08910609109140263] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
| | - J. P. Koopman
- Central Animal Laboratory, Catholic University of Nijmegen, The Netherlands
| | | | - P. M. Scholten
- Central Animal Laboratory, Catholic University of Nijmegen, The Netherlands
| | - A. C. Beynen
- Department of Human Nutrition, Wageningen Agricultural University, The Netherlands
- Department of Laboratory Animal Science, State University of Utrecht, The Netherlands
| |
Collapse
|
44
|
Klaasen HLBM, Koopman JP, Poelma FGJ, Van Den Brink ME, Barker MH, Beynen AC. Intestinal, Segmented, Filamentous Bacteria and Colonisation Resistance of Mice to Pathogenic Bacteria. MICROBIAL ECOLOGY IN HEALTH AND DISEASE 2009. [DOI: 10.3109/08910609209141551] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
| | - J. P. Koopman
- Central Animal Laboratory, Catholic University, Nijmegen, The Netherlands
| | - F. G. J. Poelma
- Central Animal Laboratory, Catholic University, Nijmegen, The Netherlands
| | | | - M. H. Barker
- Central Animal Laboratory, Catholic University, Nijmegen, The Netherlands
| | - A. C. Beynen
- Central Animal Laboratory, Catholic University, Nijmegen, The Netherlands
- Department of Human Nutrition, Agricultural University, Wageningen, The Netherlands
- Department of Laboratory Animal Science, State University, Utrecht, The Netherlands
| |
Collapse
|
45
|
Klaasen HLBM, Koopman JP, Van Den Brink ME, Van Wezel HPN, Scholten PM, Beynen AC. Colonisation of Germ-free Mice by Segmented Filamentous Bacteria after Oral Administration of Various Murine Intestinal Wall Preparations. MICROBIAL ECOLOGY IN HEALTH AND DISEASE 2009. [DOI: 10.3109/08910609009140247] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- H. L. B. M. Klaasen
- Central Animal Laboratory, Catholic University, PO Box 9101, 6500, HB Nijmegen
| | - J. P. Koopman
- Central Animal Laboratory, Catholic University, PO Box 9101, 6500, HB Nijmegen
| | - M. E. Van Den Brink
- Central Animal Laboratory, Catholic University, PO Box 9101, 6500, HB Nijmegen
| | - H. P. N. Van Wezel
- Central Animal Laboratory, Catholic University, PO Box 9101, 6500, HB Nijmegen
| | - P. M. Scholten
- Central Animal Laboratory, Catholic University, PO Box 9101, 6500, HB Nijmegen
| | - A. C. Beynen
- Department of Laboratory Animal Science, State University, PO Box 80.166, 3508, TD Utrecht
- Department of Human Nutrition, Agricultural University, PO Box 8129, 6700, EV Wageningen, The Netherlands
| |
Collapse
|
46
|
Garner CD, Antonopoulos DA, Wagner B, Duhamel GE, Keresztes I, Ross DA, Young VB, Altier C. Perturbation of the small intestine microbial ecology by streptomycin alters pathology in a Salmonella enterica serovar typhimurium murine model of infection. Infect Immun 2009; 77:2691-702. [PMID: 19433544 PMCID: PMC2708583 DOI: 10.1128/iai.01570-08] [Citation(s) in RCA: 107] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2008] [Revised: 02/13/2009] [Accepted: 04/30/2009] [Indexed: 12/11/2022] Open
Abstract
The small intestine is an important site of infection for many enteric bacterial pathogens, and murine models, including the streptomycin-treated mouse model of infection, are frequently used to study these infections. The environment of the mouse small intestine and the microbiota with which enteric pathogens are likely to interact, however, have not been well described. Therefore, we compared the microbiota and the concentrations of short-chain fatty acids (SCFAs) present in the ileum and cecum of streptomycin-treated mice and untreated controls. We found that the microbiota in the ileum of untreated mice differed greatly from that of the cecum of the same mice, primarily among families of the phylum Firmicutes. Upon treatment with streptomycin, substantial changes in the microbial composition occurred, with a marked loss of population complexity. Characterization of the metabolic products of the microbiota, the SCFAs, showed that formate was present in the ileum but low or not detectable in the cecum while butyrate was present in the cecum but not the ileum. Treatment with streptomycin altered the SCFAs in the cecum, significantly decreasing the concentration of acetate, propionate, and butyrate. In this work, we also characterized the pathology of Salmonella infection in the ileum. Infection of streptomycin-treated mice with Salmonella was characterized by a significant increase in the relative and absolute levels of the pathogen and was associated with more severe ileal inflammation and pathology. Together these results provide a better understanding of the ileal environment in the mouse and the changes that occur upon streptomycin treatment.
Collapse
MESH Headings
- Animals
- Anti-Bacterial Agents/therapeutic use
- Bacteria/drug effects
- Bacterial Translocation/drug effects
- Biodiversity
- Cecum/chemistry
- Cecum/microbiology
- Cluster Analysis
- DNA, Bacterial/chemistry
- DNA, Bacterial/genetics
- DNA, Ribosomal/chemistry
- DNA, Ribosomal/genetics
- Fatty Acids, Volatile/analysis
- Female
- Humans
- Intestine, Small/chemistry
- Intestine, Small/microbiology
- Mice
- Molecular Sequence Data
- Phylogeny
- RNA, Ribosomal, 16S/genetics
- Salmonella Infections, Animal/microbiology
- Salmonella Infections, Animal/pathology
- Salmonella typhimurium/drug effects
- Sequence Analysis, DNA
- Streptomycin/therapeutic use
Collapse
Affiliation(s)
- Cherilyn D Garner
- Department of Population Medicine and Diagnostic Sciences, Cornell University, Ithaca, NY 14853, USA
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Burns-Guydish SM, Olomu IN, Zhao H, Wong RJ, Stevenson DK, Contag CH. Monitoring age-related susceptibility of young mice to oral Salmonella enterica serovar Typhimurium infection using an in vivo murine model. Pediatr Res 2005; 58:153-8. [PMID: 15774831 DOI: 10.1203/01.pdr.0000157725.44213.c4] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Neonates and young children are acutely susceptible to infections by gastrointestinal bacterial pathogens, such as Salmonella enterica serovar Typhimurium (S. typhimurium). To reveal age-related differences in susceptibility to this pathogen, we used in vivo bioluminescence imaging (BLI) to monitor the progression of infection in neonatal (1-wk-old), suckling (2-wk-old), juvenile (4-wk-old), and adult (6-wk-old) BALB/c mice. Mice were orally infected with various doses of a bioluminescent-labeled wild-type or mutant S. typhimurium strain, and progression of infection was monitored by BLI for 2 wks. We found that neonatal and suckling mice were more susceptible to the wild-type strain at inoculum sizes 4 and 2 log(10)'s lower for neonatal and suckling mice, respectively, than those for adult mice. At the lower inocula, newborn mice showed disseminated systemic infection as indicated by the pattern of photon emission assessed by BLI, whereas no bioluminescent signals were detectable in adult mice. In addition, an orgA(-) mutant strain of S. typhimurium with reduced virulence in adult mice produced systemic infection in newborn, suckling, and juvenile mice. Furthermore, as low as 3 log(10) CFU could be detected by BLI in tissue. The present study demonstrates that susceptibility to S. typhimurium infection decreases with age. Also, we established that BLI can be used to monitor the progression of infection in mice. Thus, this model of age-related susceptibility to S. typhimurium using BLI can be used to advance our understanding of the mechanisms involved in newborn susceptibility to infection.
Collapse
Affiliation(s)
- Stacy M Burns-Guydish
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305, USA.
| | | | | | | | | | | |
Collapse
|
48
|
Abstract
M cells are located in the epithelia overlying mucosa-associated lymphoid tissues such as Peyer's patches where they function as the antigen sampling cells of the mucosal immune system. Paradoxically, some pathogens exploit M cells as a route of invasion. Here we review our current knowledge of intestinal M cells with particular emphasis on the mechanisms underlying bacterial infection of these atypical epithelial cells.
Collapse
Affiliation(s)
- M Ann Clark
- Department of Physiological Sciences, University of Newcastle, Newcastle upon Tyne NE2 4HH, UK
| | | |
Collapse
|
49
|
Meyerholz DK, Stabel TJ, Cheville NF. Segmented filamentous bacteria interact with intraepithelial mononuclear cells. Infect Immun 2002; 70:3277-80. [PMID: 12011024 PMCID: PMC128008 DOI: 10.1128/iai.70.6.3277-3280.2002] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Segmented filamentous bacteria (SFB) are found in multiple species and play an important role in the development of mucosal immunity. The mechanism by which the bacteria interact with the immune system has not been well defined. We provide morphologic evidence of direct interaction between SFB and intraepithelial mononuclear cells.
Collapse
Affiliation(s)
- David K Meyerholz
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, Iowa 50010, USA
| | | | | |
Collapse
|
50
|
Morphological Demonstration of the Stimulative Effects of Charcoal Powder Including Wood Vinegar Compound Solution on Growth Performance and Intestinal Villus Histology in Chickens. J Poult Sci 2002. [DOI: 10.2141/jpsa.39.42] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|