1
|
de Brito CB, do Nascimento Arifa RD, de Oliveira Bezerra R, Dias Igídio CE, de Amorim-Santos BM, de Menezes Santos ACP, Barbosa LM, Barbosa JPP, Cassiano LMG, Kohlhoff M, Fagundes M, Batista RRÁ, Queiroz-Junior CM, Saliba AM, Raposo JDA, Braga FC, Coimbra RS, Teixeira MM, Fagundes CT, Souza DG. Antibiotic-Induced Dysbiosis of the Gut Microbiota Shifts Host Tryptophan Metabolism and Increases the Susceptibility of Mice to Pulmonary Infection With Pseudomonas aeruginosa. Immunology 2025. [PMID: 40387573 DOI: 10.1111/imm.13932] [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: 08/07/2024] [Revised: 04/04/2025] [Accepted: 04/07/2025] [Indexed: 05/20/2025] Open
Abstract
Pseudomonas aeruginosa is an opportunistic bacterium that mainly infects those who have previously been treated with antibiotics. We hypothesised that antibiotic treatment disrupts tryptophan metabolism, leading to increased susceptibility to P. aeruginosa infection. Our results showed that mice receiving antibiotics exhibited intestinal dysbiosis with alterations in host tryptophan metabolism, a higher mortality rate and a higher bacterial load compared to eubiotic mice. In the lungs of the dysbiotic mice, there was an increase in IDO1 (Indoleamine 2,3-dioxygenase 1) activity and an accumulation of kynurenine after infection, and IDO1-/- mice were resistant to infection after induction of dysbiosis. Importantly, dysbiosis led to increased expression and activation of AHR (Aryl Hydrocarbon Receptor) in an IDO1-dependent manner. Blocking AHR activation in dysbiotic mice resulted in a lower bacterial load. Our data showed that increased AHR activation by kynurenine was associated with decreased phagocytosis of P. aeruginosa by macrophages and neutrophils. In conclusion, our results indicate that dysbiosis resulting from prolonged antimicrobial treatment alters tryptophan metabolism, leading to activation of the IDO1-AHR axis and increasing susceptibility to P. aeruginosa infection. Furthermore, these data suggest that IDO1 or AHR are potential host targets for the prevention of opportunistic infections in patients undergoing antimicrobial therapy.
Collapse
Affiliation(s)
- Camila Bernardo de Brito
- Laboratório de Interação Microrganismo-Hospedeiro, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Raquel Duque do Nascimento Arifa
- Laboratório de Interação Microrganismo-Hospedeiro, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Rafael de Oliveira Bezerra
- Laboratório de Interação Microrganismo-Hospedeiro, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Carlos Eduardo Dias Igídio
- Laboratório de Interação Microrganismo-Hospedeiro, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
- Centro de Pesquisa e Desenvolvimento de Fármacos, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Bárbara Maria de Amorim-Santos
- Laboratório de Interação Microrganismo-Hospedeiro, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Anna Clara Paiva de Menezes Santos
- Laboratório de Interação Microrganismo-Hospedeiro, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Larissa Mendes Barbosa
- Laboratório de Interação Microrganismo-Hospedeiro, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - João Paulo Pezzini Barbosa
- Laboratório de Interação Microrganismo-Hospedeiro, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Markus Kohlhoff
- Instituto René Rachou (IRR) - FIOCRUZ. FIOCRUZ MG, Belo Horizonte, MG, Brazil
| | - Micheli Fagundes
- Laboratório de Interação Microrganismo-Hospedeiro, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
- Centro de Pesquisa e Desenvolvimento de Fármacos, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Rafaela Ribeiro Álvares Batista
- Laboratório de Interação Microrganismo-Hospedeiro, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
- Centro de Pesquisa e Desenvolvimento de Fármacos, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Celso Martins Queiroz-Junior
- Centro de Pesquisa e Desenvolvimento de Fármacos, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Alessandra M Saliba
- Departamento de Microbiologia e Imunologia, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Juliana Divina Almeida Raposo
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Fernão Castro Braga
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Mauro Martins Teixeira
- Centro de Pesquisa e Desenvolvimento de Fármacos, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Caio Tavares Fagundes
- Laboratório de Interação Microrganismo-Hospedeiro, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
- Centro de Pesquisa e Desenvolvimento de Fármacos, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Danielle G Souza
- Laboratório de Interação Microrganismo-Hospedeiro, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| |
Collapse
|
2
|
Zhou H, Tang L, Fenton KA, Song X. Exploring and evaluating microbiome resilience in the gut. FEMS Microbiol Ecol 2025; 101:fiaf046. [PMID: 40302016 PMCID: PMC12065411 DOI: 10.1093/femsec/fiaf046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 03/30/2025] [Accepted: 04/28/2025] [Indexed: 05/01/2025] Open
Abstract
The gut ecosystem is closely related to human gastrointestinal health and overall wellness. Microbiome resilience refers to the capability of a microbial community to resist or recover from perturbations to its original state of balance. So far, there is no consensus on the criteria for assessing microbiome resilience. This article provides new insights into the metrics and techniques for resilience assessment. We discussed several potential parameters, such as microbiome structure, keystone species, biomarkers, persistence degree, recovery rate, and various research techniques in microbiology, metagenomics, biochemistry, and dynamic modeling. The article further explores the factors that influence the gut microbiome resilience. The microbiome structure (i.e. abundance and diversity), keystone species, and microbe-microbe interplays determine microbiome resilience. Microorganisms employ a variety of mechanisms to achieve the microbiome resilience, including flexible metabolism, quorum sensing, functional redundancy, microbial cooperation, and competition. Host-microbe interactions play a crucial role in maintaining microbiome stability and functionality. Unlike other articles, we focus on the regulation of host immune system on microbiome resilience. The immune system facilitates bacterial preservation and colonization, community construction, probiotic protection, and pathogen elimination through the mechanisms of immunological tolerance, immune-driven microbial compartmentalization, and immune inclusion and exclusion. Microbial immunomodulation indirectly modulates microbiome resilience.
Collapse
Affiliation(s)
- Huimin Zhou
- Department of Microbiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Li Tang
- Department of Microecology, College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Kristin A Fenton
- Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, 9037, Norway
| | - Xiaobo Song
- Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, 9037, Norway
| |
Collapse
|
3
|
Bruno P, Schüler T, Rosshart SP. Born to be wild: utilizing natural microbiota for reliable biomedical research. Trends Immunol 2025; 46:17-28. [PMID: 39690004 DOI: 10.1016/j.it.2024.11.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 11/11/2024] [Accepted: 11/19/2024] [Indexed: 12/19/2024]
Abstract
Laboratory mice housed under specific pathogen-free (SPF) conditions are the standard model in biomedical research. However, experiments with a particular inbred mouse strain performed in different laboratories often yield inconsistent or conflicting data due to housing-specific variations in the composition and diversity of SPF microbiota. These variations affect immune and nonimmune cell functions, leading to systemic physiological changes. Consequently, microbiota-dependent inconsistencies have raised general doubts regarding the suitability of mice as model organisms. Since stability positively correlates with biological diversity, we postulate that increasing species diversity can improve microbiota stability and mouse physiology, enhancing robustness, reproducibility, and experimental validity. Similar to the generation of inbred mouse strains in the last century, we suggest a worldwide initiative to define a transplantable 'wild' microbiota that stably colonizes mice irrespective of housing conditions.
Collapse
Affiliation(s)
- Philipp Bruno
- Department of Microbiome Research, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany.
| | - Thomas Schüler
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto von Guericke University Magdeburg, Magdeburg, Germany.
| | - Stephan P Rosshart
- Department of Microbiome Research, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany; Department of Medicine II, Faculty of Medicine, Medical Center - University of Freiburg, Freiburg, Germany.
| |
Collapse
|
4
|
Inciuraite R, Gedgaudas R, Lukosevicius R, Tilinde D, Ramonaite R, Link A, Kasetiene N, Malakauskas M, Kiudelis G, Jonaitis LV, Kupcinskas J, Juzenas S, Skieceviciene J. Constituents of stable commensal microbiota imply diverse colonic epithelial cell reactivity in patients with ulcerative colitis. Gut Pathog 2024; 16:16. [PMID: 38521943 PMCID: PMC10960424 DOI: 10.1186/s13099-024-00612-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 03/15/2024] [Indexed: 03/25/2024] Open
Abstract
BACKGROUND Despite extensive research on microbiome alterations in ulcerative colitis (UC), the role of the constituent stable microbiota remains unclear. RESULTS This study, employing 16S rRNA-gene sequencing, uncovers a persistent microbial imbalance in both active and quiescent UC patients compared to healthy controls. Using co-occurrence and differential abundance analysis, the study highlights microbial constituents, featuring Phocaeicola, Collinsella, Roseburia, Holdemanella, and Bacteroides, that are not affected during the course of UC. Co-cultivation experiments, utilizing commensal Escherichia coli and Phocaeicola vulgatus, were conducted with intestinal epithelial organoids derived from active UC patients and controls. These experiments reveal a tendency for a differential response in tight junction formation and maintenance in colonic epithelial cells, without inducing pathogen recognition and stress responses, offering further insights into the roles of these microorganisms in UC pathogenesis. These experiments also uncover high variation in patients' response to the same bacteria, which indicate the need for more comprehensive, stratified analyses with an expanded sample size. CONCLUSION This study reveals that a substantial part of the gut microbiota remains stable throughout progression of UC. Functional experiments suggest that members of core microbiota - Escherichia coli and Phocaeicola vulgatus - potentially differentially regulate the expression of tight junction gene in the colonic epithelium of UC patients and healthy individuals.
Collapse
Affiliation(s)
- Ruta Inciuraite
- Institute for Digestive Research, Academy of Medicine, Lithuanian University of Health Sciences, Kaunas, Lithuania.
| | - Rolandas Gedgaudas
- Institute for Digestive Research, Academy of Medicine, Lithuanian University of Health Sciences, Kaunas, Lithuania
- Department of Gastroenterology, Academy of Medicine, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Rokas Lukosevicius
- Institute for Digestive Research, Academy of Medicine, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Deimante Tilinde
- Institute for Digestive Research, Academy of Medicine, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Rima Ramonaite
- Institute for Digestive Research, Academy of Medicine, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Alexander Link
- Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Hospital Magdeburg, Magdeburg, Germany
| | - Neringa Kasetiene
- Department of Food Safety and Quality, Academy of Veterinary, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Mindaugas Malakauskas
- Department of Food Safety and Quality, Academy of Veterinary, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Gediminas Kiudelis
- Institute for Digestive Research, Academy of Medicine, Lithuanian University of Health Sciences, Kaunas, Lithuania
- Department of Gastroenterology, Academy of Medicine, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Laimas Virginijus Jonaitis
- Institute for Digestive Research, Academy of Medicine, Lithuanian University of Health Sciences, Kaunas, Lithuania
- Department of Gastroenterology, Academy of Medicine, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Juozas Kupcinskas
- Institute for Digestive Research, Academy of Medicine, Lithuanian University of Health Sciences, Kaunas, Lithuania
- Department of Gastroenterology, Academy of Medicine, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Simonas Juzenas
- Institute for Digestive Research, Academy of Medicine, Lithuanian University of Health Sciences, Kaunas, Lithuania
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Jurgita Skieceviciene
- Institute for Digestive Research, Academy of Medicine, Lithuanian University of Health Sciences, Kaunas, Lithuania.
| |
Collapse
|
5
|
Wang J, Zhao X, Wan YY. Intricacies of TGF-β signaling in Treg and Th17 cell biology. Cell Mol Immunol 2023; 20:1002-1022. [PMID: 37217798 PMCID: PMC10468540 DOI: 10.1038/s41423-023-01036-7] [Citation(s) in RCA: 94] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 04/27/2023] [Indexed: 05/24/2023] Open
Abstract
Balanced immunity is pivotal for health and homeostasis. CD4+ helper T (Th) cells are central to the balance between immune tolerance and immune rejection. Th cells adopt distinct functions to maintain tolerance and clear pathogens. Dysregulation of Th cell function often leads to maladies, including autoimmunity, inflammatory disease, cancer, and infection. Regulatory T (Treg) and Th17 cells are critical Th cell types involved in immune tolerance, homeostasis, pathogenicity, and pathogen clearance. It is therefore critical to understand how Treg and Th17 cells are regulated in health and disease. Cytokines are instrumental in directing Treg and Th17 cell function. The evolutionarily conserved TGF-β (transforming growth factor-β) cytokine superfamily is of particular interest because it is central to the biology of both Treg cells that are predominantly immunosuppressive and Th17 cells that can be proinflammatory, pathogenic, and immune regulatory. How TGF-β superfamily members and their intricate signaling pathways regulate Treg and Th17 cell function is a question that has been intensely investigated for two decades. Here, we introduce the fundamental biology of TGF-β superfamily signaling, Treg cells, and Th17 cells and discuss in detail how the TGF-β superfamily contributes to Treg and Th17 cell biology through complex yet ordered and cooperative signaling networks.
Collapse
Affiliation(s)
- Junying Wang
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Xingqi Zhao
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Yisong Y Wan
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
6
|
Gweon TG. [Gut Microbiome and Colorectal Cancer]. THE KOREAN JOURNAL OF GASTROENTEROLOGY = TAEHAN SOHWAGI HAKHOE CHI 2023; 82:56-62. [PMID: 37621240 DOI: 10.4166/kjg.2023.089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 08/04/2023] [Accepted: 08/06/2023] [Indexed: 08/26/2023]
Abstract
Colorectal cancer (CRC) is one of the most common cancers in Korea. A majority of CRCs are caused by progressive genomic alterations referred to as the adenoma-carcinoma sequence. The factors that may increase the risk of CRC include obesity and consumption of a high-fat diet, red meat, processed meat, and alcohol. Recently, the role of gut microbiota in the formation, progression and treatment of CRCs has been investigated in depth. An altered gut microbiota can drive carcinogenesis and cause the development of CRC. Studies have also shown the role of gut microbiota in the prevention of CRC and the impact of therapies involving gut microbiota on CRC. Herein, we summarize the current understanding of the role of the gut microbiota in the development of CRC and its therapeutic potential, including the prevention of CRC and in enhancing efficacy of chemotherapy and immunotherapy.
Collapse
Affiliation(s)
- Tae-Geun Gweon
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
7
|
Zhang H, Xie R, Zhang H, Sun R, Li S, Xia C, Li Z, Zhang L, Guo Y, Huang J. Recombinant Hemagglutinin protein and DNA-RNA-combined nucleic acid vaccines harbored by Yeast elicit protective immunity against H9N2 Avian Influenza infection. Poult Sci 2023; 102:102662. [PMID: 37043959 PMCID: PMC10140169 DOI: 10.1016/j.psj.2023.102662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 03/13/2023] [Accepted: 03/13/2023] [Indexed: 03/22/2023] Open
Abstract
A safe, convenience, and effective vaccine for controlling avian influenza virus infection is crucial in scale poultry production. Yeasts are considered useful vaccine vehicles for the delivery of antigens, which has been used to protect human and animal health. We report here the development of H9N2 strain hemagglutinin (HA)-based recombinant protein vaccines (rH9HA) and DNA-RNA-combined vaccine (rH9-DNA-RNA) in Saccharomyces cerevisiae for the first time. The immunogenicity assay indicated that both rH9HA and rH9-DNA-RNA could induce robust production of serum IgG, mucosal sIgA, and cellular immune responses. The reshape and diversification of gut microbiota and an enriched Lactobacillus, Debaryomyces were observed after oral immunization with rH9HA or rH9-DNA-RNA yeast vaccine, which might contribute to modulate the intestinal mucosal immunity and antiviral process. Oral immunized birds with either rH9HA or rH9-DNA-RNA were effectively protected from H9N2 virus challenge. Our findings suggested that yeast-derived H9N2 HA-based recombinant protein vaccines and DNA-RNA-combined nucleic acid vaccines are feasible and efficacious, opening up a new avenue for rapid and cost-effective production of avian influenza vaccines to achieve good protection effect.
Collapse
|
8
|
Ou S, Wang H, Tao Y, Luo K, Ye J, Ran S, Guan Z, Wang Y, Hu H, Huang R. Fusobacterium nucleatum and colorectal cancer: From phenomenon to mechanism. Front Cell Infect Microbiol 2022; 12:1020583. [PMID: 36523635 PMCID: PMC9745098 DOI: 10.3389/fcimb.2022.1020583] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 11/09/2022] [Indexed: 11/30/2022] Open
Abstract
Colorectal cancer(CRC) is the third most frequent malignant tumor. The gut microbiome acts as a vital component of CRC etiology. Fusobacterium nucleatum(Fn) is a key member of colorectal cancer-associated bacteria. But we lack a systematic and in-depth understanding on its role in CRC evolution. In this article, We reviewed the abundance changes and distribution of Fn in CRC occurrence and development, potential effect of Fn in the initiation of CRC, the source of intratumoral Fn and the cause of its tropism to CRC. In addition, We described the mechanism by which Fn promotes the malignant biological behavior of CRC, affects CRC response to therapy, and shapes the tumor immune microenvironment in great detail. Based on the relationship between Fn and CRC, we proposed strategies for CRC prevention and treatment, and discussed the feasibility and limitations of specific cases, to gain insights into further basic and clinical research in the future.
Collapse
Affiliation(s)
- Suwen Ou
- Department of Colorectal Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Hufei Wang
- Department of Colorectal Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yangbao Tao
- Department of Colorectal Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Kangjia Luo
- Department of Colorectal Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China,Department of Gastrointestinal Surgery, The Affiliated Hospital of Medical School of Ningbo University, Ningbo, Zhejiang, China
| | - Jinhua Ye
- Department of Colorectal Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Songlin Ran
- Department of Colorectal Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Zilong Guan
- Department of Colorectal Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China,Department of General Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yuliuming Wang
- Department of Colorectal Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Hanqing Hu
- Department of Colorectal Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Rui Huang
- Department of Colorectal Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China,*Correspondence: Rui Huang,
| |
Collapse
|
9
|
Sanchez MM, Bagdasarian IA, Darch W, Morgan JT. Organotypic cultures as aging associated disease models. Aging (Albany NY) 2022; 14:9338-9383. [PMID: 36435511 PMCID: PMC9740367 DOI: 10.18632/aging.204361] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 10/21/2022] [Indexed: 11/24/2022]
Abstract
Aging remains a primary risk factor for a host of diseases, including leading causes of death. Aging and associated diseases are inherently multifactorial, with numerous contributing factors and phenotypes at the molecular, cellular, tissue, and organismal scales. Despite the complexity of aging phenomena, models currently used in aging research possess limitations. Frequently used in vivo models often have important physiological differences, age at different rates, or are genetically engineered to match late disease phenotypes rather than early causes. Conversely, routinely used in vitro models lack the complex tissue-scale and systemic cues that are disrupted in aging. To fill in gaps between in vivo and traditional in vitro models, researchers have increasingly been turning to organotypic models, which provide increased physiological relevance with the accessibility and control of in vitro context. While powerful tools, the development of these models is a field of its own, and many aging researchers may be unaware of recent progress in organotypic models, or hesitant to include these models in their own work. In this review, we describe recent progress in tissue engineering applied to organotypic models, highlighting examples explicitly linked to aging and associated disease, as well as examples of models that are relevant to aging. We specifically highlight progress made in skin, gut, and skeletal muscle, and describe how recently demonstrated models have been used for aging studies or similar phenotypes. Throughout, this review emphasizes the accessibility of these models and aims to provide a resource for researchers seeking to leverage these powerful tools.
Collapse
Affiliation(s)
- Martina M. Sanchez
- Department of Bioengineering, University of California, Riverside, CA 92521, USA
| | | | - William Darch
- Department of Bioengineering, University of California, Riverside, CA 92521, USA
| | - Joshua T. Morgan
- Department of Bioengineering, University of California, Riverside, CA 92521, USA
| |
Collapse
|
10
|
Consonni A, Miglietti M, De Luca CMG, Cazzaniga FA, Ciullini A, Dellarole IL, Bufano G, Di Fonzo A, Giaccone G, Baggi F, Moda F. Approaching the Gut and Nasal Microbiota in Parkinson's Disease in the Era of the Seed Amplification Assays. Brain Sci 2022; 12:1579. [PMID: 36421902 PMCID: PMC9688507 DOI: 10.3390/brainsci12111579] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 11/10/2022] [Accepted: 11/17/2022] [Indexed: 10/30/2023] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder often associated with pre-motor symptoms involving both gastrointestinal and olfactory tissues. PD patients frequently suffer from hyposmia, hyposalivation, dysphagia and gastrointestinal dysfunctions. During the last few years it has been speculated that microbial agents could play a crucial role in PD. In particular, alterations of the microbiota composition (dysbiosis) might contribute to the formation of misfolded α-synuclein, which is believed to be the leading cause of PD. However, while several findings confirmed that there might be an important link between intestinal microbiota alterations and PD onset, little is known about the potential contribution of the nasal microbiota. Here, we describe the latest findings on this topic by considering that more than 80% of patients with PD develop remarkable olfactory deficits in their prodromal disease stage. Therefore, the nasal microbiota might contribute to PD, eventually boosting the gut microbiota in promoting disease onset. Finally, we present the applications of the seed amplification assays to the study of the gut and olfactory mucosa of PD patients, and how they could be exploited to investigate whether pathogenic bacteria present in the gut and the nose might promote α-synuclein misfolding and aggregation.
Collapse
Affiliation(s)
- Alessandra Consonni
- Division of Neurology 4-Neuroimmunology and Neuromuscular Diseases, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy
| | - Martina Miglietti
- Division of Neurology 4-Neuroimmunology and Neuromuscular Diseases, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy
| | - Chiara Maria Giulia De Luca
- Division of Neurology 5-Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy
| | - Federico Angelo Cazzaniga
- Division of Neurology 5-Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy
| | - Arianna Ciullini
- Division of Neurology 5-Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy
| | - Ilaria Linda Dellarole
- Division of Neurology 5-Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy
| | - Giuseppe Bufano
- Division of Neurology 5-Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy
| | - Alessio Di Fonzo
- Division of Neurology, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Giorgio Giaccone
- Division of Neurology 5-Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy
| | - Fulvio Baggi
- Division of Neurology 4-Neuroimmunology and Neuromuscular Diseases, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy
| | - Fabio Moda
- Division of Neurology 5-Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy
| |
Collapse
|
11
|
Crosstalk between mucosal microbiota, host gene expression, and sociomedical factors in the progression of colorectal cancer. Sci Rep 2022; 12:13447. [PMID: 35927305 PMCID: PMC9352898 DOI: 10.1038/s41598-022-17823-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 08/01/2022] [Indexed: 11/24/2022] Open
Abstract
Various omics-based biomarkers related to the occurrence, progression, and prognosis of colorectal cancer (CRC) have been identified. In this study, we attempted to identify gut microbiome-based biomarkers and detect their association with host gene expression in the initiation and progression of CRC by integrating analysis of the gut mucosal metagenome, RNA sequencing, and sociomedical factors. We performed metagenome and RNA sequencing on colonic mucosa samples from 13 patients with advanced CRC (ACRC), 10 patients with high-risk adenoma (HRA), and 7 normal control (NC) individuals. All participants completed a questionnaire on sociomedical factors. The interaction and correlation between changes in the microbiome and gene expression were assessed using bioinformatic analysis. When comparing HRA and NC samples, which can be considered to represent the process of tumor initiation, 28 genes and five microbiome species were analyzed with correlation plots. When comparing ACRC and HRA samples, which can be considered to represent the progression of CRC, seven bacterial species and 21 genes were analyzed. When comparing ACRC and NC samples, 16 genes and five bacterial species were analyzed, and four correlation plots were generated. A network visualizing the relationship between bacterial and host gene expression in the initiation and progression of CRC indicated that Clostridium spiroforme and Tyzzerella nexilis were hub bacteria in the development and progression of CRC. Our study revealed the interactions of and correlation between the colonic mucosal microbiome and host gene expression to identify potential roles of the microbiome in the initiation and progression of CRC. Our results provide gut microbiome-based biomarkers that may be potential diagnostic markers and therapeutic targets in patients with CRC.
Collapse
|
12
|
Abstract
Newer 'omics approaches, such as metatranscriptomics and metabolomics, allow functional assessments of the interaction(s) between the gut microbiome and the human host. However, in order to generate meaningful data with these approaches, the method of sample collection is critical. Prior studies have relied on expensive and invasive means toward sample acquisition, such as intestinal biopsy, while other studies have relied on easier methods of collection, such as fecal samples that do not necessarily represent those microbes in contact with the host. In this pilot study, we attempt to characterize a novel, minimally invasive method toward sampling the human microbiome using mucosal cytology brush sampling compared to intestinal gut biopsy samples on 5 healthy participants undergoing routine screening colonoscopy. We compared metatranscriptomic analyses between the two collection methods and identified increased taxonomic evenness and beta diversity in the cytology brush samples and similar community transcriptional profiles between the two methods. Metabolomics assessment demonstrated striking differences between the two methods, implying a difference in bacterial-derived versus human-absorbed metabolites. Put together, this study supports the use of microbiome sampling with cytology brushes, but caution must be exercised when performing metabolomics assessment, as this represents differential metabolite production but not absorption by the host. IMPORTANCE In order to generate meaningful metabolomic and microbiome data, the method of sample collection is critical. This study utilizes and compares two methods for intestinal tissue collection for evaluation of metabolites and microbiomes, finding that using a brush to sample the microbiome provides valuable data. However, for metabolomics assessment, biopsy samples may still be required.
Collapse
|
13
|
Leonardi I, Gao IH, Lin WY, Allen M, Li XV, Fiers WD, De Celie MB, Putzel GG, Yantiss RK, Johncilla M, Colak D, Iliev ID. Mucosal fungi promote gut barrier function and social behavior via Type 17 immunity. Cell 2022; 185:831-846.e14. [PMID: 35176228 PMCID: PMC8897247 DOI: 10.1016/j.cell.2022.01.017] [Citation(s) in RCA: 190] [Impact Index Per Article: 63.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 11/30/2021] [Accepted: 01/20/2022] [Indexed: 02/07/2023]
Abstract
Fungal communities (the mycobiota) are an integral part of the gut microbiota, and the disruption of their integrity contributes to local and gut-distal pathologies. Yet, the mechanisms by which intestinal fungi promote homeostasis remain unclear. We characterized the mycobiota biogeography along the gastrointestinal tract and identified a subset of fungi associated with the intestinal mucosa of mice and humans. Mucosa-associated fungi (MAF) reinforced intestinal epithelial function and protected mice against intestinal injury and bacterial infection. Notably, intestinal colonization with a defined consortium of MAF promoted social behavior in mice. The gut-local effects on barrier function were dependent on IL-22 production by CD4+ T helper cells, whereas the effects on social behavior were mediated through IL-17R-dependent signaling in neurons. Thus, the spatial organization of the gut mycobiota is associated with host-protective immunity and epithelial barrier function and might be a driver of the neuroimmune modulation of mouse behavior through complementary Type 17 immune mechanisms.
Collapse
Affiliation(s)
- Irina Leonardi
- Gastroenterology and Hepatology Division, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA.,The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| | - Iris H. Gao
- Gastroenterology and Hepatology Division, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA.,The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA.,Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, Cornell University, New York, NY 10065, USA
| | - Woan-Yu Lin
- Gastroenterology and Hepatology Division, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA.,The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA.,Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, Cornell University, New York, NY 10065, USA
| | - Megan Allen
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, Cornell University, New York City, NY, USA
| | - Xin V. Li
- Gastroenterology and Hepatology Division, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA.,The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| | - William D. Fiers
- Gastroenterology and Hepatology Division, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA.,The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| | - Meghan Bialt De Celie
- Gastroenterology and Hepatology Division, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA.,The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| | - Gregory G. Putzel
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| | - Rhonda K. Yantiss
- MJ Department of Pathology & Laboratory Medicine, Weill Cornell Medicine, Cornell University, New York, NY 10065, USA
| | - Melanie Johncilla
- MJ Department of Pathology & Laboratory Medicine, Weill Cornell Medicine, Cornell University, New York, NY 10065, USA
| | - Dilek Colak
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, Cornell University, New York City, NY, USA.,Gale and Ira Drukier Institute for Children’s Health, Weill Cornell Medical College, Cornell University, New York City, NY, USA
| | - Iliyan D. Iliev
- Gastroenterology and Hepatology Division, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA.,The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA.,Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY 10065, USA.,Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, Cornell University, New York, NY 10065, USA
| |
Collapse
|
14
|
Aprile F, Bruno G, Palma R, Mascellino MT, Panetta C, Scalese G, Oliva A, Severi C, Pontone S. Microbiota Alterations in Precancerous Colon Lesions: A Systematic Review. Cancers (Basel) 2021; 13:cancers13123061. [PMID: 34205378 PMCID: PMC8234190 DOI: 10.3390/cancers13123061] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 06/09/2021] [Accepted: 06/14/2021] [Indexed: 12/22/2022] Open
Abstract
Simple Summary Even with recent advances, gut microbiota is still one of the most demanding challenges that research needs to handle. In particular, given its deep impact on gastrointestinal health, microbiota could explain the development and progression of certain diseases. Moreover, it could be used as a potential predictive biomarker. Given this, the relationship between intestinal microbiota and colorectal adenoma, considered a premalignant lesion leading to carcinoma, has been deeply evaluated. This review highlights the historical and novel data on microbiota characteristics in adenoma patients to provide an updated summary of current knowledge and its limits. Abstract Gut microbiota plays an important role in human health. It may promote carcinogenesis and is related to several diseases of the gastrointestinal tract. This study of microbial dysbiosis in the etiology of colorectal adenoma aimed to investigate the possible causative role of microbiota in the adenoma–carcinoma sequence and its possible preventive role. A systematic, PRISMA-guided review was performed. The PubMed database was searched using “adenoma microbiota” and selecting original articles between January 2010 and May 2020 independently screened. A higher prevalence of Proteobacteria, Fusobacteria, and Bacteroidetes phyla was observed in the fecal luminal and mucosa-associated microbiota of patients with adenoma. However, other studies provided evidence of depletion of Clostridium, Faecalibacterium, Bacteroides and Romboutsia. Results on the relationship between adenoma endoscopic resection and microbiota were inconsistent. In conclusion, none of the analyzed studies developed a predictive model that could differentiate adenoma from non-adenoma patients, and therefore, to prevent cancer progression. The impact of adenoma’s endoscopic resection on microbiota was investigated, but the results were inconclusive. Further research in the field is required.
Collapse
Affiliation(s)
- Francesca Aprile
- Department of Translational and Precision Medicine, Gastroenterology Unit, Sapienza University of Rome, 00161 Rome, Italy; (F.A.); (G.B.); (G.S.); (C.S.)
| | - Giovanni Bruno
- Department of Translational and Precision Medicine, Gastroenterology Unit, Sapienza University of Rome, 00161 Rome, Italy; (F.A.); (G.B.); (G.S.); (C.S.)
| | - Rossella Palma
- Department of Surgical Sciences, Sapienza University of Rome, 00161 Rome, Italy; (R.P.); (C.P.)
| | - Maria Teresa Mascellino
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00161 Rome, Italy; (M.T.M.); (A.O.)
| | - Cristina Panetta
- Department of Surgical Sciences, Sapienza University of Rome, 00161 Rome, Italy; (R.P.); (C.P.)
| | - Giulia Scalese
- Department of Translational and Precision Medicine, Gastroenterology Unit, Sapienza University of Rome, 00161 Rome, Italy; (F.A.); (G.B.); (G.S.); (C.S.)
| | - Alessandra Oliva
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00161 Rome, Italy; (M.T.M.); (A.O.)
| | - Carola Severi
- Department of Translational and Precision Medicine, Gastroenterology Unit, Sapienza University of Rome, 00161 Rome, Italy; (F.A.); (G.B.); (G.S.); (C.S.)
| | - Stefano Pontone
- Department of Surgical Sciences, Sapienza University of Rome, 00161 Rome, Italy; (R.P.); (C.P.)
- Correspondence: ; Tel.: +39-06-49975568
| |
Collapse
|
15
|
Troyer EA, Kohn JN, Ecklu-Mensah G, Aleti G, Rosenberg DR, Hong S. Searching for host immune-microbiome mechanisms in obsessive-compulsive disorder: A narrative literature review and future directions. Neurosci Biobehav Rev 2021; 125:517-534. [PMID: 33639178 PMCID: PMC8106658 DOI: 10.1016/j.neubiorev.2021.02.034] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 02/09/2021] [Accepted: 02/22/2021] [Indexed: 12/20/2022]
Abstract
Obsessive-compulsive disorder (OCD) is disabling and often treatment-refractory. Host immunity and gut microbiota have bidirectional communication with each other and with the brain. Perturbations to this axis have been implicated in neuropsychiatric disorders, but immune-microbiome signaling in OCD is relatively underexplored. We review support for further pursuing such investigations in OCD, including: 1) gut microbiota has been associated with OCD, but causal pathogenic mechanisms remain unclear; 2) early environmental risk factors for OCD overlap with critical periods of immune-microbiome development; 3) OCD is associated with increased risk of immune-mediated disorders and changes in immune parameters, which are separately associated with the microbiome; and 4) gut microbiome manipulations in animal models are associated with changes in immunity and some obsessive-compulsive symptoms. Theoretical pathogenic mechanisms could include microbiota programming of cytokine production, promotion of expansion and trafficking of peripheral immune cells to the CNS, and regulation of microglial function. Immune-microbiome signaling in OCD requires further exploration, and may offer novel insights into pathogenic mechanisms and potential treatment targets for this disabling disorder.
Collapse
Affiliation(s)
- Emily A Troyer
- Department of Psychiatry, University of California San Diego, La Jolla, California, United States.
| | - Jordan N Kohn
- Department of Psychiatry, University of California San Diego, La Jolla, California, United States
| | - Gertrude Ecklu-Mensah
- Department of Medicine and Scripps Institution of Oceanography, University of California San Diego, La Jolla, California, United States
| | - Gajender Aleti
- Department of Psychiatry, University of California San Diego, La Jolla, California, United States
| | - David R Rosenberg
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University, Detroit, Michigan, United States
| | - Suzi Hong
- Department of Psychiatry, University of California San Diego, La Jolla, California, United States; Herbert Wertheim School of Public Health and Human Longevity Science, University of California San Diego, La Jolla, California, United States
| |
Collapse
|
16
|
Loftus M, Hassouneh SAD, Yooseph S. Bacterial associations in the healthy human gut microbiome across populations. Sci Rep 2021; 11:2828. [PMID: 33531651 PMCID: PMC7854710 DOI: 10.1038/s41598-021-82449-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 01/20/2021] [Indexed: 01/30/2023] Open
Abstract
In a microbial community, associations between constituent members play an important role in determining the overall structure and function of the community. The human gut microbiome is believed to play an integral role in host health and disease. To understand the nature of bacterial associations at the species level in healthy human gut microbiomes, we analyzed previously published collections of whole-genome shotgun sequence data, totaling over 1.6 Tbp, generated from 606 fecal samples obtained from four different healthy human populations. Using a Random Forest Classifier, we identified 202 signature bacterial species that were prevalent in these populations and whose relative abundances could be used to accurately distinguish between the populations. Bacterial association networks were constructed with these signature species using an approach based on the graphical lasso. Network analysis revealed conserved bacterial associations across populations and a dominance of positive associations over negative associations, with this dominance being driven by associations between species that are closely related either taxonomically or functionally. Bacterial species that form network modules, and species that constitute hubs and bottlenecks, were also identified. Functional analysis using protein families suggests that much of the taxonomic variation across human populations does not foment substantial functional or structural differences.
Collapse
Affiliation(s)
- Mark Loftus
- grid.170430.10000 0001 2159 2859Burnett School of Biomedical Sciences, Genomics and Bioinformatics Cluster, University of Central Florida, Orlando, 32787 USA
| | - Sayf Al-Deen Hassouneh
- grid.170430.10000 0001 2159 2859Burnett School of Biomedical Sciences, Genomics and Bioinformatics Cluster, University of Central Florida, Orlando, 32787 USA
| | - Shibu Yooseph
- grid.170430.10000 0001 2159 2859Department of Computer Science, Genomics and Bioinformatics Cluster, University of Central Florida, Orlando, FL 32816-2993 USA
| |
Collapse
|
17
|
Interactions of Symbiotic Partners Drive the Development of a Complex Biogeography in the Squid-Vibrio Symbiosis. mBio 2020; 11:mBio.00853-20. [PMID: 32457244 PMCID: PMC7251207 DOI: 10.1128/mbio.00853-20] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The complexity, inaccessibility, and time scales of initial colonization of most animal microbiomes present challenges for the characterization of how the bacterial symbionts influence the form and function of tissues in the minutes to hours following the initial interaction of the partners. Here, we use the naturally occurring binary squid-vibrio association to explore this phenomenon. Imaging of the spatiotemporal landscape of this symbiosis during its onset provides a window into the impact of differences in both host-tissue maturation and symbiont strain phenotypes on the establishment of a dynamically stable symbiotic system. These data provide evidence that the symbionts shape the host-tissue landscape and that tissue maturation impacts the influence of strain-level differences on the daily rhythms of the symbiosis, the competitiveness for colonization, and antibiotic sensitivity. Microbes live in complex microniches within host tissues, but how symbiotic partners communicate to create such niches during development remains largely unexplored. Using confocal microscopy and symbiont genetics, we characterized the shaping of host microenvironments during light organ colonization of the squid Euprymna scolopes by the bacterium Vibrio fischeri. During embryogenesis, three pairs of invaginations form sequentially on the organ’s surface, producing pores that lead to interior compressed tubules at different stages of development. After hatching, these areas expand, allowing V. fischeri cells to enter and migrate ∼120 μm through three anatomically distinct regions before reaching blind-ended crypt spaces. A dynamic gatekeeper, or bottleneck, connects these crypts with the migration path. Once V. fischeri cells have entered the crypts, the bottlenecks narrow, and colonization by the symbiont population becomes spatially restricted. The actual timing of constriction and restriction varies with crypt maturity and with different V. fischeri strains. Subsequently, starting with the first dawn following colonization, the bottleneck controls a lifelong cycle of dawn-triggered expulsions of most of the symbionts into the environment and a subsequent regrowth in the crypts. Unlike other developmental phenotypes, bottleneck constriction is not induced by known microbe-associated molecular patterns (MAMPs) or by V. fischeri-produced bioluminescence, but it does require metabolically active symbionts. Further, while symbionts in the most mature crypts have a higher proportion of live cells and a greater likelihood of expulsion at dawn, they have a lower resistance to antibiotics. The overall dynamics of these distinct microenvironments reflect the complexity of the host-symbiont dialogue.
Collapse
|
18
|
Shen W, Sun J, Yao F, Lin K, Yuan Y, Chen Y, Han H, Li Z, Zou J, Jiao X. Microbiome in Intestinal Lavage Fluid May Be A Better Indicator in Evaluating The Risk of Developing Colorectal Cancer Compared with Fecal Samples. Transl Oncol 2020; 13:100772. [PMID: 32298987 PMCID: PMC7160452 DOI: 10.1016/j.tranon.2020.100772] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 03/23/2020] [Accepted: 03/24/2020] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVE Intestinal microbiota plays a vital role in the pathogenesis of colorectal cancer (CRC), which is crucial for assessing the risk and prognosis of CRC. Most studies regarding human gut microbiota mainly based on the feces, but the exact composition of microbiota vary significantly due to fecal composition is easily affected by many factors. We aim to evaluate whether intestinal lavage fluid (IVF) is a better substitution mirroring the gut microbiota. METHODS We performed 16S rRNA gene analysis on fecal and IVF samples from 30 CRC patients and 25 healthy individuals, comparison in luminal (feces) / mucosal (IVF) adherent bacterial community profiles were analyzed. RESULTS The difference between feces and IVF were observed, including the diversity and abundance of pathogenic bacteria (either in single strain or in co-occurrence pattern). IVF group shared 605 OTUs with the fecal group, but there was 94 OTUs only observed in fecal samples, while 247 OTUs were mainly existing in the IVF group. Among them, 27 vital bacterial species detected in IVF, while 10 critical species detected in fecal samples. The co-occurrence bacteria Fusobacteria Cluster and Proteobacteria Cluster 2 significantly increased in IVF than in control (P < .01), while Firmicutes Cluster 1, Firmicutes Cluster 2 and Proteobacteria Cluster 1 were markedly lower in IVF than in control (P < .001). In CRC feces, Fusobacteria Cluster was higher than in control (P < .05), but Firmicutes Cluster 1 was of substantially less abundance than in control (P < .001). Proteobacteria Cluster 2 was increased dramatically in IVF than in feces (P < .05), Firmicutes Cluster 1 were of substantially less abundance than in feces (P < .05). CONCLUSION Pathogenic microbiota is more abundant in IVF than in feces. Microbiota of IVF may closely be related to the mucosal-associated microbial communities, which benefit from elucidating the relationship of the intestinal microbiota and CRC carcinogenesis.
Collapse
Affiliation(s)
- Weitao Shen
- The second affiliated hospital of Shantou University Medical College, Shantou, Guangdong, China 515041.
| | - Jiayu Sun
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong, China 515041.
| | - Fen Yao
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong, China 515041.
| | - Kaihuang Lin
- The second affiliated hospital of Shantou University Medical College, Shantou, Guangdong, China 515041.
| | - Yumeng Yuan
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong, China 515041.
| | - Yexi Chen
- The second affiliated hospital of Shantou University Medical College, Shantou, Guangdong, China 515041.
| | - Hui Han
- The second affiliated hospital of Shantou University Medical College, Shantou, Guangdong, China 515041.
| | - Zhiyang Li
- The second affiliated hospital of Shantou University Medical College, Shantou, Guangdong, China 515041.
| | - Juan Zou
- The second affiliated hospital of Shantou University Medical College, Shantou, Guangdong, China 515041.
| | - Xiaoyang Jiao
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong, China 515041.
| |
Collapse
|
19
|
Evasion of Innate Lymphoid Cell-Regulated Gamma Interferon Responses by Chlamydia muridarum To Achieve Long-Lasting Colonization in Mouse Colon. Infect Immun 2020; 88:IAI.00798-19. [PMID: 31818961 DOI: 10.1128/iai.00798-19] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 12/05/2019] [Indexed: 12/14/2022] Open
Abstract
Revealing the mechanisms by which bacteria establish long-lasting colonization in the gastrointestinal tract is an area of intensive investigation. The obligate intracellular bacterium Chlamydia is known to colonize mouse colon for long periods. A colonization-deficient mutant strain of this intracellular bacterium is able to regain long-lasting colonization in gamma interferon (IFN-γ) knockout mice following intracolon inoculation. We now report that mice deficient in conventional T lymphocytes or recombination-activating gene (Rag) failed to show rescue of mutant colonization. Nevertheless, antibody depletion of IFN-γ or genetic deletion of interleukin 2 (IL-2) receptor common gamma chain in Rag-deficient mice did rescue mutant colonization. These observations suggest that colonic IFN-γ, responsible for inhibiting the intracellular bacterial mutant, is produced by innate lymphoid cells (ILCs). Consistently, depletion of NK1.1+ cells in Rag-deficient mice both prevented IFN-γ production and rescued mutant colonization. Furthermore, mice deficient in transcriptional factor RORγt, but not chemokine receptor CCR6, showed full rescue of the long-lasting colonization of the mutant, indicating a role for group 3-like ILCs. However, the inhibitory function of the responsible group 3-like ILCs was not dependent on the natural killer cell receptor (NCR1), since NCR1-deficient mice still inhibited mutant colonization. Consistently, mice deficient in the transcriptional factor T-bet only delayed the clearance of the bacterial mutant without fully rescuing the long-lasting colonization of the mutant. Thus, we have demonstrated that the obligate intracellular bacterium Chlamydia maintains its long-lasting colonization in the colon by evading IFN-γ from group 3-like ILCs.
Collapse
|
20
|
Tarantino G, Citro V, Capone D. Nonalcoholic Fatty Liver Disease: A Challenge from Mechanisms to Therapy. J Clin Med 2019; 9:15. [PMID: 31861591 PMCID: PMC7019297 DOI: 10.3390/jcm9010015] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 12/05/2019] [Accepted: 12/16/2019] [Indexed: 12/11/2022] Open
Abstract
Focusing on previously published mechanisms of non-alcoholic fatty liver disease (NAFLD), their uncertainty does not always permit a clear elucidation of the grassroot alterations that are at the basis of the wide-spread illness, and thus curing it is still a challenge. There is somehow exceptional progress, but many controversies persist in NAFLD research and clinical investigation. It is likely that hidden mechanisms will be brought to light in the near future. Hereby, the authors present, with some criticism, classical mechanisms that stand at the basis of NAFLD, and consider contextually different emerging processes. Without ascertaining these complex interactions, investigators have a long way left ahead before finding an effective therapy for NAFLD beyond diet and exercise.
Collapse
Affiliation(s)
- Giovanni Tarantino
- Department of Clinical Medicine and Surgery, “Federico II” University Medical School of Naples, 80131 Naples, Italy
| | - Vincenzo Citro
- Department of General Medicine, “Umberto I” Hospital, 84014 Nocera Inferiore (Sa), Italy;
| | - Domenico Capone
- Care Department of Public Health and Drug-Use, Section of Medical Pharmacology and Toxicology, “Federico II” University, 80131 Naples, Italy;
| |
Collapse
|
21
|
Coates M, Lee MJ, Norton D, MacLeod AS. The Skin and Intestinal Microbiota and Their Specific Innate Immune Systems. Front Immunol 2019; 10:2950. [PMID: 31921196 PMCID: PMC6928192 DOI: 10.3389/fimmu.2019.02950] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Accepted: 12/02/2019] [Indexed: 12/22/2022] Open
Abstract
The skin and intestine are active organs of the immune system that are constantly exposed to the outside environment. They support diverse microbiota, both commensal and pathogenic, which encompass bacteria, viruses, fungi, and parasites. The skin and intestine must maintain homeostasis with the diversity of commensal organisms present on epithelial surfaces. Here we review the current literature pertaining to epithelial barrier formation, microbial composition, and the complex regulatory mechanisms governing the interaction between the innate immune system and microbiota in the skin and intestine. We also compare and contrast the skin and intestine—two different organ systems responsible creating a protective barrier against the external environment, each of which has unique mechanisms for interaction with commensal populations and host repair.
Collapse
Affiliation(s)
- Margaret Coates
- Department of Dermatology, Duke University, Durham, NC, United States
| | - Min Jin Lee
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, United States
| | - Diana Norton
- Department of Dermatology, Duke University, Durham, NC, United States
| | - Amanda S MacLeod
- Department of Dermatology, Duke University, Durham, NC, United States.,Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, United States.,Department of Immunology, Duke University, Durham, NC, United States.,Pinnell Center for Investigative Dermatology, Duke University, Durham, NC, United States
| |
Collapse
|
22
|
Kumar A, Smith C, Jobin C, Trinchieri G, Howcroft TK, Seifried H, Espey MG, Flores R, Kim YS, Daschner PJ. Workshop Report: Modulation of Antitumor Immune Responses by Dietary and Microbial Metabolites. J Natl Cancer Inst 2019; 109:3806188. [PMID: 30053241 DOI: 10.1093/jnci/djx040] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 02/22/2017] [Indexed: 12/13/2022] Open
Abstract
The human microbiota maintains an enormous and diverse capacity to produce a diet-dependent metabolome that impacts both host tissue and microbial community homeostasis. Recent discoveries support a growing appreciation that microbial metabolites derived from bioactive foods are also important regulators of host immune and metabolic functions. To gain a better understanding of the current evidence for the roles of dietary and microbial metabolites in tumor immunity, the Division of Cancer Biology and the Division of Cancer Prevention, National Cancer Institute, cosponsored a workshop on August 31 and September 1, 2016, in Bethesda, Maryland. Workshop participants examined several lines of converging science that link nutrition, microbiology, and tumor immunology and identified key concepts and research opportunities that will accelerate our understanding of these interactions. In addition, the participants identified some of the critical gaps and research challenges that could be addressed through interdisciplinary collaborations, including future opportunities for translating new information into novel cancer prevention and treatment strategies based on targeting host immune functions that are altered by metabolite sensing pathways.
Collapse
Affiliation(s)
- Amit Kumar
- Affiliations of authors: Division of Cancer Prevention, National Cancer Institute, Bethesda, MD (AK, HS, RF, YSK); Center for Cancer Research (CS, GT) and Division of Cancer Biology (TKH, MGE, PJD), National Cancer Institute, Bethesda, MD (CS, GT); Department of Infectious Diseases and Pathology, University of Florida, Gainesville, FL (CJ)
| | - Carolyne Smith
- Affiliations of authors: Division of Cancer Prevention, National Cancer Institute, Bethesda, MD (AK, HS, RF, YSK); Center for Cancer Research (CS, GT) and Division of Cancer Biology (TKH, MGE, PJD), National Cancer Institute, Bethesda, MD (CS, GT); Department of Infectious Diseases and Pathology, University of Florida, Gainesville, FL (CJ)
| | - Christian Jobin
- Affiliations of authors: Division of Cancer Prevention, National Cancer Institute, Bethesda, MD (AK, HS, RF, YSK); Center for Cancer Research (CS, GT) and Division of Cancer Biology (TKH, MGE, PJD), National Cancer Institute, Bethesda, MD (CS, GT); Department of Infectious Diseases and Pathology, University of Florida, Gainesville, FL (CJ)
| | - Giorgio Trinchieri
- Affiliations of authors: Division of Cancer Prevention, National Cancer Institute, Bethesda, MD (AK, HS, RF, YSK); Center for Cancer Research (CS, GT) and Division of Cancer Biology (TKH, MGE, PJD), National Cancer Institute, Bethesda, MD (CS, GT); Department of Infectious Diseases and Pathology, University of Florida, Gainesville, FL (CJ)
| | - T Kevin Howcroft
- Affiliations of authors: Division of Cancer Prevention, National Cancer Institute, Bethesda, MD (AK, HS, RF, YSK); Center for Cancer Research (CS, GT) and Division of Cancer Biology (TKH, MGE, PJD), National Cancer Institute, Bethesda, MD (CS, GT); Department of Infectious Diseases and Pathology, University of Florida, Gainesville, FL (CJ)
| | - Harold Seifried
- Affiliations of authors: Division of Cancer Prevention, National Cancer Institute, Bethesda, MD (AK, HS, RF, YSK); Center for Cancer Research (CS, GT) and Division of Cancer Biology (TKH, MGE, PJD), National Cancer Institute, Bethesda, MD (CS, GT); Department of Infectious Diseases and Pathology, University of Florida, Gainesville, FL (CJ)
| | - Michael Graham Espey
- Affiliations of authors: Division of Cancer Prevention, National Cancer Institute, Bethesda, MD (AK, HS, RF, YSK); Center for Cancer Research (CS, GT) and Division of Cancer Biology (TKH, MGE, PJD), National Cancer Institute, Bethesda, MD (CS, GT); Department of Infectious Diseases and Pathology, University of Florida, Gainesville, FL (CJ)
| | - Roberto Flores
- Affiliations of authors: Division of Cancer Prevention, National Cancer Institute, Bethesda, MD (AK, HS, RF, YSK); Center for Cancer Research (CS, GT) and Division of Cancer Biology (TKH, MGE, PJD), National Cancer Institute, Bethesda, MD (CS, GT); Department of Infectious Diseases and Pathology, University of Florida, Gainesville, FL (CJ)
| | - Young S Kim
- Affiliations of authors: Division of Cancer Prevention, National Cancer Institute, Bethesda, MD (AK, HS, RF, YSK); Center for Cancer Research (CS, GT) and Division of Cancer Biology (TKH, MGE, PJD), National Cancer Institute, Bethesda, MD (CS, GT); Department of Infectious Diseases and Pathology, University of Florida, Gainesville, FL (CJ)
| | - Phillip J Daschner
- Affiliations of authors: Division of Cancer Prevention, National Cancer Institute, Bethesda, MD (AK, HS, RF, YSK); Center for Cancer Research (CS, GT) and Division of Cancer Biology (TKH, MGE, PJD), National Cancer Institute, Bethesda, MD (CS, GT); Department of Infectious Diseases and Pathology, University of Florida, Gainesville, FL (CJ)
| |
Collapse
|
23
|
Abstract
Alterations in the human gut microbiota play an important role in disease pathogenesis. Although next-generation sequencing has provided observational evidence linking shifts in gut microbiota composition to alterations in the human host, underlying mechanisms remain elusive. Metabolites generated within complex microbial communities and at the crossroads with host cells may be able to explain the impact of the gut microbiome on human homeostasis. Emerging technologies including novel culturing protocols, microfluidic systems, engineered organoids, and single-cell imaging approaches are providing new perspectives from which the gut microbiome can be studied paving the way to new diagnostic markers and personalized therapeutic interventions.
Collapse
Affiliation(s)
- Paola Brun
- Department of Molecular Medicine, University of Padova, Padova, Italy
| |
Collapse
|
24
|
Martínez-López M, Iborra S, Conde-Garrosa R, Mastrangelo A, Danne C, Mann ER, Reid DM, Gaboriau-Routhiau V, Chaparro M, Lorenzo MP, Minnerup L, Saz-Leal P, Slack E, Kemp B, Gisbert JP, Dzionek A, Robinson MJ, Rupérez FJ, Cerf-Bensussan N, Brown GD, Bernardo D, LeibundGut-Landmann S, Sancho D. Microbiota Sensing by Mincle-Syk Axis in Dendritic Cells Regulates Interleukin-17 and -22 Production and Promotes Intestinal Barrier Integrity. Immunity 2019; 50:446-461.e9. [PMID: 30709742 PMCID: PMC6382412 DOI: 10.1016/j.immuni.2018.12.020] [Citation(s) in RCA: 163] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 07/30/2018] [Accepted: 12/17/2018] [Indexed: 12/15/2022]
Abstract
Production of interleukin-17 (IL-17) and IL-22 by T helper 17 (Th17) cells and group 3 innate lymphoid cells (ILC3s) in response to the gut microbiota ensures maintenance of intestinal barrier function. Here, we examined the mechanisms whereby the immune system detects microbiota in the steady state. A Syk-kinase-coupled signaling pathway in dendritic cells (DCs) was critical for commensal-dependent production of IL-17 and IL-22 by CD4+ T cells. The Syk-coupled C-type lectin receptor Mincle detected mucosal-resident commensals in the Peyer's patches (PPs), triggered IL-6 and IL-23p19 expression, and thereby regulated function of intestinal Th17- and IL-17-secreting ILCs. Mice deficient in Mincle or with selective depletion of Syk in CD11c+ cells had impaired production of intestinal RegIIIγ and IgA and increased systemic translocation of gut microbiota. Consequently, Mincle deficiency led to liver inflammation and deregulated lipid metabolism. Thus, sensing of commensals by Mincle and Syk signaling in CD11c+ cells reinforces intestinal immune barrier and promotes host-microbiota mutualism, preventing systemic inflammation.
Collapse
Affiliation(s)
- María Martínez-López
- Immunobiology Laboratory, Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro 3, Madrid 28029, Spain
| | - Salvador Iborra
- Immunobiology Laboratory, Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro 3, Madrid 28029, Spain; Department of Immunology, School of Medicine, Universidad Complutense de Madrid, 12 de Octubre Health Research Institute (imas12), Madrid, Spain.
| | - Ruth Conde-Garrosa
- Immunobiology Laboratory, Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro 3, Madrid 28029, Spain
| | - Annalaura Mastrangelo
- Immunobiology Laboratory, Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro 3, Madrid 28029, Spain
| | - Camille Danne
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Elizabeth R Mann
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK; Manchester Collaborative Centre for Inflammation Research, Faculty of Biology, Medicine, and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Delyth M Reid
- Medical Research Council Centre for Medical Mycology at the University of Aberdeen, Aberdeen Fungal Group, Institute of Medical Sciences, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Valérie Gaboriau-Routhiau
- INRA Micalis Institut, UMR1319, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France; INSERM UMR1163, Institut Imagine, Laboratory of Intestinal Immunity, 75015 Paris, France; Université Paris Descartes-Sorbonne Paris Cité, 75006 Paris, France
| | - Maria Chaparro
- Gastroenterology Unit, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Diego de León 62, Madrid 28006, Spain
| | - María P Lorenzo
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, Urbanización Montepríncipe, km 0, M501, Alcorcón 28925, Spain
| | | | - Paula Saz-Leal
- Immunobiology Laboratory, Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro 3, Madrid 28029, Spain
| | - Emma Slack
- Institute of Food, Nutrition, and Health, ETH Zurich, Vladimir-Prelog-Weg 4, Zürich 8093, Switzerland
| | | | - Javier P Gisbert
- Gastroenterology Unit, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Diego de León 62, Madrid 28006, Spain
| | | | | | - Francisco J Rupérez
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, Urbanización Montepríncipe, km 0, M501, Alcorcón 28925, Spain
| | - Nadine Cerf-Bensussan
- INSERM UMR1163, Institut Imagine, Laboratory of Intestinal Immunity, 75015 Paris, France; Université Paris Descartes-Sorbonne Paris Cité, 75006 Paris, France
| | - Gordon D Brown
- Medical Research Council Centre for Medical Mycology at the University of Aberdeen, Aberdeen Fungal Group, Institute of Medical Sciences, Foresterhill, Aberdeen AB25 2ZD, UK
| | - David Bernardo
- Gastroenterology Unit, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Diego de León 62, Madrid 28006, Spain
| | - Salomé LeibundGut-Landmann
- Section of Immunology, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 266a Zurich 8057, Switzerland
| | - David Sancho
- Immunobiology Laboratory, Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro 3, Madrid 28029, Spain.
| |
Collapse
|
25
|
McKeen S, Young W, Mullaney J, Fraser K, McNabb WC, Roy NC. Infant Complementary Feeding of Prebiotics for theMicrobiome and Immunity. Nutrients 2019; 11:nu11020364. [PMID: 30744134 PMCID: PMC6412789 DOI: 10.3390/nu11020364] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 02/01/2019] [Accepted: 02/06/2019] [Indexed: 02/06/2023] Open
Abstract
Complementary feeding transitions infants from a milk-based diet to solid foods, providing essential nutrients to the infant and the developing gut microbiome while influencing immune development. Some of the earliest microbial colonisers readily ferment select oligosaccharides, influencing the ongoing establishment of the microbiome. Non-digestible oligosaccharides in prebiotic-supplemented formula and human milk oligosaccharides promote commensal immune-modulating bacteria such as Bifidobacterium, which decrease in abundance during weaning. Incorporating complex, bifidogenic, non-digestible carbohydrates during the transition to solid foods may present an opportunity to feed commensal bacteria and promote balanced concentrations of beneficial short chain fatty acid concentrations and vitamins that support gut barrier maturation and immunity throughout the complementary feeding window.
Collapse
Affiliation(s)
- Starin McKeen
- AgResearch, Food Nutrition & Health, Grasslands Research Centre, Private Bag 11008, Palmerston north4442, New Zealand.
- Riddet Institute, Massey University, Private Bag 11222, Palmerston North 4442, New Zealand.
- High-Value Nutrition National Science Challenge, Auckland, New Zealand.
| | - Wayne Young
- AgResearch, Food Nutrition & Health, Grasslands Research Centre, Private Bag 11008, Palmerston north4442, New Zealand.
- Riddet Institute, Massey University, Private Bag 11222, Palmerston North 4442, New Zealand.
- High-Value Nutrition National Science Challenge, Auckland, New Zealand.
| | - Jane Mullaney
- AgResearch, Food Nutrition & Health, Grasslands Research Centre, Private Bag 11008, Palmerston north4442, New Zealand.
- Riddet Institute, Massey University, Private Bag 11222, Palmerston North 4442, New Zealand.
- High-Value Nutrition National Science Challenge, Auckland, New Zealand.
| | - Karl Fraser
- AgResearch, Food Nutrition & Health, Grasslands Research Centre, Private Bag 11008, Palmerston north4442, New Zealand.
- Riddet Institute, Massey University, Private Bag 11222, Palmerston North 4442, New Zealand.
- High-Value Nutrition National Science Challenge, Auckland, New Zealand.
| | - Warren C McNabb
- Riddet Institute, Massey University, Private Bag 11222, Palmerston North 4442, New Zealand.
- High-Value Nutrition National Science Challenge, Auckland, New Zealand.
| | - Nicole C Roy
- AgResearch, Food Nutrition & Health, Grasslands Research Centre, Private Bag 11008, Palmerston north4442, New Zealand.
- Riddet Institute, Massey University, Private Bag 11222, Palmerston North 4442, New Zealand.
- High-Value Nutrition National Science Challenge, Auckland, New Zealand.
| |
Collapse
|
26
|
Queirós J, Villar M, Hernández-Jarguín A, López V, Fernández de Mera I, Vicente J, Alves PC, Gortazar C, Fuente JDL. A metaproteomics approach reveals changes in mandibular lymph node microbiota of wild boar naturally exposed to an increasing trend of Mycobacterium tuberculosis complex infection. Tuberculosis (Edinb) 2018; 114:103-112. [PMID: 30711148 DOI: 10.1016/j.tube.2018.12.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 11/14/2018] [Accepted: 12/16/2018] [Indexed: 12/19/2022]
Abstract
Constraints in the characterization of microbiota community that circulates in the host have limited the extent of co-infection studies in natural populations. In this study, we used a metaproteomics approach to characterize the mandibular lymph nodes microbiota of wild boar (Sus scrofa) naturally exposed to an increasing trend of Mycobacterium tuberculosis complex (MTC) infection. Our results showed a reduction in microbiota diversity and changes in the composition, structure and functionality of the microbiota community associated with an increase in tuberculosis prevalence, from 45% in 2002/06 to 83% in 2009/12. These temporal changes were accompanied by an increase in the relative abundance of Babesia, Theileria and Pestivirus genera and a decrease in the Ascogregarina and Chlorella. A positive association was also evidenced between the prevalence of tuberculosis and the presence of microbial proteins responsible for carbohydrate transport and metabolism. Our findings suggest MTC-host-microbiota interactions at the population level, which may occur in order to ensure sufficient metabolic resources for MTC survival, growth and transmission. We strongly recommend the use of metaproteomics when studying microbiota communities in wildlife populations, for which traditional diagnostic techniques are limited and in which new organisms with a pathogenic potential for domestic animals and humans may appear.
Collapse
Affiliation(s)
- João Queirós
- Centro de Investigacão em Biodiversidade e Recursos Genéticos (CIBIO)/InBio Laboratório Associado, Universidade do Porto, Campus de Vairão, R. Monte-Crasto, 4485-661, Vairão, Portugal; Departamento de Biologia, Faculdade de Ciências da Universidade do Porto (FCUP), Rua do Campo Alegre s⁄n, 4169-007, Porto, Portugal; SaBio, Instituto de Investigación en Recursos Cinegéticos IREC (CSIC-UCLM-JCCM), Ronda de Toledo s/n, 13071, Ciudad Real, Spain.
| | - Margarita Villar
- SaBio, Instituto de Investigación en Recursos Cinegéticos IREC (CSIC-UCLM-JCCM), Ronda de Toledo s/n, 13071, Ciudad Real, Spain.
| | - Angélica Hernández-Jarguín
- SaBio, Instituto de Investigación en Recursos Cinegéticos IREC (CSIC-UCLM-JCCM), Ronda de Toledo s/n, 13071, Ciudad Real, Spain.
| | - Vladimir López
- SaBio, Instituto de Investigación en Recursos Cinegéticos IREC (CSIC-UCLM-JCCM), Ronda de Toledo s/n, 13071, Ciudad Real, Spain.
| | - Isabel Fernández de Mera
- SaBio, Instituto de Investigación en Recursos Cinegéticos IREC (CSIC-UCLM-JCCM), Ronda de Toledo s/n, 13071, Ciudad Real, Spain.
| | - Joaquín Vicente
- SaBio, Instituto de Investigación en Recursos Cinegéticos IREC (CSIC-UCLM-JCCM), Ronda de Toledo s/n, 13071, Ciudad Real, Spain.
| | - Paulo C Alves
- Centro de Investigacão em Biodiversidade e Recursos Genéticos (CIBIO)/InBio Laboratório Associado, Universidade do Porto, Campus de Vairão, R. Monte-Crasto, 4485-661, Vairão, Portugal; Departamento de Biologia, Faculdade de Ciências da Universidade do Porto (FCUP), Rua do Campo Alegre s⁄n, 4169-007, Porto, Portugal; Wildlife Biology Program, University of Montana, Missoula, MT, 59812, USA.
| | - Christian Gortazar
- SaBio, Instituto de Investigación en Recursos Cinegéticos IREC (CSIC-UCLM-JCCM), Ronda de Toledo s/n, 13071, Ciudad Real, Spain.
| | - José de la Fuente
- SaBio, Instituto de Investigación en Recursos Cinegéticos IREC (CSIC-UCLM-JCCM), Ronda de Toledo s/n, 13071, Ciudad Real, Spain; Department of Veterinary Pathobiology, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK, 74078, USA.
| |
Collapse
|
27
|
Invited Review: From nose to gut – the role of the microbiome in neurological disease. Neuropathol Appl Neurobiol 2018; 45:195-215. [DOI: 10.1111/nan.12520] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Accepted: 09/29/2018] [Indexed: 12/14/2022]
|
28
|
Ceftriaxone Administration Disrupts Intestinal Homeostasis, Mediating Noninflammatory Proliferation and Dissemination of Commensal Enterococci. Infect Immun 2018; 86:IAI.00674-18. [PMID: 30224553 DOI: 10.1128/iai.00674-18] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 09/06/2018] [Indexed: 12/21/2022] Open
Abstract
Enterococci are Gram-positive commensals of the mammalian intestinal tract and harbor intrinsic resistance to broad-spectrum cephalosporins. Disruption of colonization resistance in humans by antibiotics allows enterococci to proliferate in the gut and cause disseminated infections. In this study, we used Enterococcus faecalis (EF)-colonized mice to study the dynamics of enterococci, commensal microbiota, and the host in response to systemic ceftriaxone administration. We found that the mouse model recapitulates intestinal proliferation and dissemination of enterococci seen in humans. Employing a ceftriaxone-sensitive strain of enterococci (E. faecalis JL308), we showed that increased intestinal abundance is critical for the systemic dissemination of enterococci. Investigation of the impact of ceftriaxone on the mucosal barrier defenses and integrity suggested that translocation of enterococci across the intestinal mucosa was not associated with intestinal pathology or increased permeability. Ceftriaxone-induced alteration of intestinal microbial composition was associated with transient increase in the abundance of multiple bacterial operational taxonomic units (OTUs) in addition to enterococci, for example, lactobacilli, which also disseminated to the extraintestinal organs. Collectively, these results emphasize that ceftriaxone-induced disruption of colonization resistance and alteration of mucosal homeostasis facilitate increased intestinal abundance of a limited number of commensals along with enterococci, allowing their translocation and systemic dissemination in a healthy host.
Collapse
|
29
|
Jitsumura M, Cunningham AL, Hitchings MD, Islam S, Davies AP, Row PE, Riddell AD, Kinross J, Wilkinson TS, Jenkins GJ, Williams JG, Harris DA. Protocol for faecal microbiota transplantation in ulcerative colitis (FMTUC): a randomised feasibility study. BMJ Open 2018; 8:e021987. [PMID: 30341117 PMCID: PMC6196852 DOI: 10.1136/bmjopen-2018-021987] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The interaction of the gut microbiota with the human host is implicated in the pathogenesis of inflammatory and immunological diseases including ulcerative colitis (UC). Faecal microbiota transplantation (FMT) as a method of restoring gut microbial diversity is of increasing interest as a therapeutic approach in the management of UC. The current literature lacks consensus about the dose of FMT, route of administration and duration of response. METHODS AND ANALYSIS This single-blinded randomised trial will explore the feasibility of FMT in 30 treatment-naïve patients with histologically confirmed distal UC limited to the recto-sigmoid region (up to 40 cm from the anal verge). This study aims to estimate the magnitude of treatment response to FMT under controlled conditions. The intervention (FMT) will be administered by rectal retention enema. It will test the feasibility of randomising patients to: (i) single FMT dose, (ii) five daily FMT doses or (iii) control (no FMT dose). All groups will receive standard antibiotic gut decontamination and bowel preparation before FMT. Recruitment will take place over a 24-month period with a 12-week patient follow-up. Trial objectives include evaluation of the magnitude of treatment response to FMT, investigation of the clinical value of metabolic phenotyping for predicting the clinical response to FMT and testing the recruitment rate of donors and patients for a study in FMT. This feasibility trial will enable an estimate of number of patients needed, help determine optimal study conditions and inform the choice of endpoints for a future definitive phase III study. ETHICS AND DISSEMINATION The trial is approved by the regional ethics committee and is sponsored by Abertawe Bro Morgannwg University's Health Board. Written informed consent from all patients will be obtained. Serious adverse events will be reported to the sponsor. Trial results will be disseminated via peer review publication and shared with trial participants. TRIAL REGISTRATION NUMBER ISRCTN 58082603; Pre-results.
Collapse
Affiliation(s)
- Maki Jitsumura
- Department of Colorectal Surgery, Singleton Hospital, Swansea, UK
| | | | - Matthew David Hitchings
- Medical Microbiology and Infectious Diseases, Swansea University Medical School, Swansea, UK
| | - Saiful Islam
- Swansea Trial Unit, Swansea University, Swansea, UK
| | - Angharad P Davies
- Public Health Wales Microbiology, Singleton Hospital, Swansea University Medical School, Swansea, UK
| | - Paula E Row
- Biochemistry Group, Swansea University Medical School, Swansea, UK
| | - Andrew D Riddell
- Department of Colorectal Surgery, Redcliffe Hospital, Brisbane, Queensland, Australia
| | - James Kinross
- Department of Surgery and Cancer, St. Mary's Hospital, Imperial College London, London, UK
| | - Tom S Wilkinson
- Medical Microbiology and Infectious Diseases, Swansea University Medical School, Swansea, UK
| | - G J Jenkins
- Molecular Carcinogenesis, Institute of Life Science, Swansea University Medical School, Swansea, UK
| | - John G Williams
- Institute of Life Science 2, Swansea University Medical School, Swansea, UK
| | | |
Collapse
|
30
|
Park CH, Eun CS, Han DS. Intestinal microbiota, chronic inflammation, and colorectal cancer. Intest Res 2018; 16:338-345. [PMID: 30090032 PMCID: PMC6077304 DOI: 10.5217/ir.2018.16.3.338] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Revised: 05/03/2018] [Accepted: 05/03/2018] [Indexed: 12/22/2022] Open
Abstract
In addition to genetic and epigenetic factors, various environmental factors, including diet, play important roles in the development of colorectal cancer (CRC). Recently, there is increasing interest in the intestinal microbiota as an environmental risk factor for CRC, because diet also influences the composition of the intestinal microbiota. The human intestinal microbiota comprises about 100 trillion microbes. This microbiome thrives on undigested dietary residues in the intestinal lumen and produces various metabolites. It is well known that the dietary risk factors for CRC are mediated by dysbiosis of the intestinal microbiota and their metabolites. In this review, we describe the bacterial taxa associated with CRC, including Fusobacterium nucleatum, enterotoxigenic Bacteroides fragilis, Escherichia coli, and butyrate-producing bacteria. We also discuss the host-diet interaction in colorectal carcinogenesis.
Collapse
Affiliation(s)
- Chan Hyuk Park
- Department of Internal Medicine, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri, Korea
| | - Chang Soo Eun
- Department of Internal Medicine, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri, Korea
| | - Dong Soo Han
- Department of Internal Medicine, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri, Korea
| |
Collapse
|
31
|
Zhang P, Minardi LM, Kuenstner JT, Zekan SM, Kruzelock R. Anti-microbial Antibodies, Host Immunity, and Autoimmune Disease. Front Med (Lausanne) 2018; 5:153. [PMID: 29876352 PMCID: PMC5974924 DOI: 10.3389/fmed.2018.00153] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Accepted: 05/03/2018] [Indexed: 01/05/2023] Open
Abstract
Autoimmune diseases are a spectrum of clinical inflammatory syndromes with circulating autoantibodies. Autoimmune diseases affect millions of patients worldwide with enormous costs to patients and society. The diagnosis of autoimmune diseases relies on the presence of autoantibodies and the treatment strategy is to suppress the immune system using specific or non-specific immunosuppressive agents. The discovery of anti-microbial antibodies in the blood of patients with Crohn's disease and Sjogren's syndrome and cross-reactivity of anti-microbial antibodies to human tissue suggests a new molecular mechanism of pathogenesis, raising the possibility of designing a new therapeutic strategy for these patients. The presence of anti-microbial antibodies indicates the failure of the innate immunity system to clear the microbial agents from the blood and activation of adaptive immunity through B-lymphocytes/plasma cells. More importantly, the specific antibodies against the microbial proteins are directed toward the commensal microbes commonly present on the surface of the human host, and these commensal microbes are important in shaping the development of the immune system and in maintaining the interaction between the human host and the environment. Persistence of these anti-microbial antibodies in patients but not in normal healthy individuals suggests abnormal interaction between the human host and the commensal microbes in the body. Elimination of the organism/organisms that elicits the antibody response would be a new avenue of therapy to investigate in patients with autoimmune diseases.
Collapse
Affiliation(s)
- Peilin Zhang
- PZM Diagnostics, LLC, Charleston, WV, United States
| | | | | | | | - Rusty Kruzelock
- WV Regional Technology Park, South Charleston, WV, United States
| |
Collapse
|
32
|
Reiss-Mandel A, Rubin C, Zayoud M, Rahav G, Regev-Yochay G. Staphylococcus aureus Colonization Induces Strain-Specific Suppression of Interleukin-17. Infect Immun 2018; 86:e00834-17. [PMID: 29311230 PMCID: PMC5820966 DOI: 10.1128/iai.00834-17] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Accepted: 12/14/2017] [Indexed: 01/02/2023] Open
Abstract
Staphylococcus aureus is a pathogen that causes significant morbidity and mortality. Nasal carriage is a major source of transmission and of endogenous infection. Persistent carriage is detected in ∼30% of healthy individuals. While Th17 cells have been shown to play a role in S. aureus infection and clearance, the immune response to carriage is not well understood. Here, we evaluate the Th17 response and its potential inhibitors during S. aureus carriage. We recruited 25 volunteers, of whom 11 were persistent carriers. Volunteers' peripheral blood mononuclear cells (PBMCs) were stimulated with either their endogenous strain (a strain isolated from that carrier) or exogenous ones (strains not carried by that volunteer). Changes in Th17 cell frequency and numbers, interleukin-17 (IL-17) mRNA expression, and IL-17 protein abundance were measured by fluorescence-activated cell sorting, real-time PCR, and enzyme-linked immunosorbent assay. Similarly, responses of IL-17 suppressors (regulatory T cells [FOXP3], IL-10, IL-27, and IL-19) were measured. Th17 and IL-17 levels in response to stimulation with endogenous strains were significantly lower than those in response to stimulation with exogenous ones. Of the suppressive cytokines tested, only IL-19 exhibited a stronger response to endogenous than to exogenous strains. Addition of recombinant IL-19 to exogenous-strain-stimulated PBMCs caused decreased IL-17 expression, whereas addition of IL-19 antibodies to endogenous-strain-stimulated cells resulted in an increased IL-17 response. Together, our results suggest that S. aureus carriage induced a tolerogenic response to a carried strain that could be reproduced through the addition of recombinant IL-19 or prevented by the addition of IL-19 antibodies. This differential immune response may play a role in the determination of S. aureus carriage patterns.
Collapse
Affiliation(s)
- Aylana Reiss-Mandel
- Infectious Disease Unit, Sheba Medical Center, Ramat Gan, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Carmit Rubin
- Infectious Disease Unit, Sheba Medical Center, Ramat Gan, Israel
| | - Morad Zayoud
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Sheba Cancer Research Center, Sheba Medical Center, Ramat Gan, Israel
| | - Galia Rahav
- Infectious Disease Unit, Sheba Medical Center, Ramat Gan, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Gili Regev-Yochay
- Infectious Disease Unit, Sheba Medical Center, Ramat Gan, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
33
|
Hernandez CJ. Bone Mechanical Function and the Gut Microbiota. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1033:249-270. [DOI: 10.1007/978-3-319-66653-2_12] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
34
|
Abstract
Inflammatory bowel disease (IBD), including Crohn disease and ulcerative colitis, is characterized by chronic intestinal inflammation due to a complex interaction of genetic determinants, disruption of mucosal barriers, aberrant inflammatory signals, loss of tolerance, and environmental triggers. Importantly, the incidence of pediatric IBD is rising, particularly in children younger than 10 years. In this review, we discuss the clinical presentation of these patients and highlight environmental exposures that may affect disease risk, particularly among people with a background genetic risk. With regard to both children and adults, we review advancements in understanding the intestinal epithelium, the mucosal immune system, and the resident microbiota, describing how dysfunction at any level can lead to diseases like IBD. We conclude with future directions for applying advances in IBD genetics to better understand pathogenesis and develop therapeutics targeting key pathogenic nodes.
Collapse
Affiliation(s)
- Joanna M Peloquin
- Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease and.,Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, Massachusetts 02114.,Harvard Medical School, Boston, Massachusetts 02115; , , ,
| | - Gautam Goel
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, Massachusetts 02114.,Harvard Medical School, Boston, Massachusetts 02115; , , ,
| | - Eduardo J Villablanca
- Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease and.,Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, Massachusetts 02114.,Harvard Medical School, Boston, Massachusetts 02115; , , ,
| | - Ramnik J Xavier
- Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease and.,Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, Massachusetts 02114.,Harvard Medical School, Boston, Massachusetts 02115; , , , .,Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts 02142.,Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| |
Collapse
|
35
|
Grigg JB, Sonnenberg GF. Host-Microbiota Interactions Shape Local and Systemic Inflammatory Diseases. THE JOURNAL OF IMMUNOLOGY 2017; 198:564-571. [PMID: 28069751 DOI: 10.4049/jimmunol.1601621] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 10/31/2016] [Indexed: 02/06/2023]
Abstract
Recent advances in understanding how the mammalian immune system and intestinal microbiota functionally interact have yielded novel insights for human health and disease. Modern technologies to quantitatively measure specific members and functional characteristics of the microbiota in the gastrointestinal tract, along with fundamental and emerging concepts in the field of immunology, have revealed numerous ways in which host-microbiota interactions proceed beneficially, neutrally, or detrimentally for mammalian hosts. It is clear that the gut microbiota has a strong influence on the shape and quality of the immune system; correspondingly, the immune system guides the composition and localization of the microbiota. In the following review, we examine the evidence that these interactions encompass homeostasis and inflammation in the intestine and, in certain cases, extraintestinal tissues. Lastly, we discuss translational therapies stemming from research on host-microbiota interactions that could be used for the treatment of chronic inflammatory diseases.
Collapse
Affiliation(s)
- John B Grigg
- Gastroenterology and Hepatology Division, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, NY 10021; Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY 10065; and The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, New York, NY 10021
| | - Gregory F Sonnenberg
- Gastroenterology and Hepatology Division, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, NY 10021; Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY 10065; and The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, New York, NY 10021
| |
Collapse
|
36
|
Yoon H, Kim N, Park JH, Kim YS, Lee J, Kim HW, Choi YJ, Shin CM, Park YS, Lee DH, Jung HC. Comparisons of Gut Microbiota Among Healthy Control, Patients With Conventional Adenoma, Sessile Serrated Adenoma, and Colorectal Cancer. J Cancer Prev 2017; 22:108-114. [PMID: 28698865 PMCID: PMC5503223 DOI: 10.15430/jcp.2017.22.2.108] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 06/07/2017] [Indexed: 12/14/2022] Open
Abstract
Background Studies on gut microbiota regarding colorectal carcinogenesis, including sessile serrated adenoma (SSA), have been scarce. The aim of this study is to investigate the role of mucosa-associated gut microbiota in the colorectal carcinogenesis. Methods We collected biopsy samples of normal rectal mucosa during colonoscopy from healthy control and patients with conventional adenoma, SSA, and colorectal cancer (CRC), respectively (n = 6). Pyrosequencing for 16S rRNA gene of bacteria was performed to compare gut microbiota. Results The most abundant phylum in total samples was Proteobacteria (55.6%), followed by Firmicutes (27.4%) and Bacteroidetes (11.6%). There was no significant difference in relative abundance of the phylum level among the four groups. Fusobacterium nucleatum, known to be frequently detected during colorectal carcinogenesis, was found in only one sample of patient with SSA. The rarefaction curves showed that the diversity of mucosal communities of patients with CRC is the lowest among the four groups and the diversity of mucosal communities of patients with SSA is higher than that of healthy control. Among the four groups, Shannon’s and Simpson’s index for diversity was the lowest and the highest in the patients with CRC, respectively; it did not reach statistical significance. The proportion of genus Pseudomonas was very high in the samples of patients with stage II–IV CRC compared with those with stage I CRC (59.3% vs. 0.3%, P = 0.064). Conclusions Our study suggests no significant role of mucosa-associated gut microbiota in the colorectal carcinogenesis. Further study for many samples or using fecal material could be helpful.
Collapse
Affiliation(s)
- Hyuk Yoon
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Nayoung Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea.,Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Ji Hyun Park
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Yong Sung Kim
- Gastroenterology and Digestive Disease Research Institute, Wonkwang University Sanbon Hospital, Gunpo, Korea
| | - Jongchan Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Hyoung Woo Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Yoon Jin Choi
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Cheol Min Shin
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Young Soo Park
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Dong Ho Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea.,Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Hyun Chae Jung
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
37
|
Tropini C, Earle KA, Huang KC, Sonnenburg JL. The Gut Microbiome: Connecting Spatial Organization to Function. Cell Host Microbe 2017; 21:433-442. [PMID: 28407481 DOI: 10.1016/j.chom.2017.03.010] [Citation(s) in RCA: 417] [Impact Index Per Article: 52.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The first rudimentary evidence that the human body harbors a microbiota hinted at the complexity of host-associated microbial ecosystems. Now, almost 400 years later, a renaissance in the study of microbiota spatial organization, driven by coincident revolutions in imaging and sequencing technologies, is revealing functional relationships between biogeography and health, particularly in the vertebrate gut. In this Review, we present our current understanding of principles governing the localization of intestinal bacteria, and spatial relationships between bacteria and their hosts. We further discuss important emerging directions that will enable progressing from the inherently descriptive nature of localization and -omics technologies to provide functional, quantitative, and mechanistic insight into this complex ecosystem.
Collapse
Affiliation(s)
- Carolina Tropini
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kristen A Earle
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kerwyn Casey Huang
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Bioengineering, Stanford University, Stanford, CA 94305, USA.
| | | |
Collapse
|
38
|
Guo CJ, Chang FY, Wyche TP, Backus KM, Acker TM, Funabashi M, Taketani M, Donia MS, Nayfach S, Pollard KS, Craik CS, Cravatt BF, Clardy J, Voigt CA, Fischbach MA. Discovery of Reactive Microbiota-Derived Metabolites that Inhibit Host Proteases. Cell 2017; 168:517-526.e18. [PMID: 28111075 DOI: 10.1016/j.cell.2016.12.021] [Citation(s) in RCA: 176] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 10/28/2016] [Accepted: 12/14/2016] [Indexed: 11/19/2022]
Abstract
The gut microbiota modulate host biology in numerous ways, but little is known about the molecular mediators of these interactions. Previously, we found a widely distributed family of nonribosomal peptide synthetase gene clusters in gut bacteria. Here, by expressing a subset of these clusters in Escherichia coli or Bacillus subtilis, we show that they encode pyrazinones and dihydropyrazinones. At least one of the 47 clusters is present in 88% of the National Institutes of Health Human Microbiome Project (NIH HMP) stool samples, and they are transcribed under conditions of host colonization. We present evidence that the active form of these molecules is the initially released peptide aldehyde, which bears potent protease inhibitory activity and selectively targets a subset of cathepsins in human cell proteomes. Our findings show that an approach combining bioinformatics, synthetic biology, and heterologous gene cluster expression can rapidly expand our knowledge of the metabolic potential of the microbiota while avoiding the challenges of cultivating fastidious commensals.
Collapse
Affiliation(s)
- Chun-Jun Guo
- Department of Bioengineering and Therapeutic Sciences and California Institute for Quantitative Biosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Fang-Yuan Chang
- Department of Biological Engineering and Synthetic Biology Center, Massachusetts Institute of Technology, Cambridge, MA 02115, USA
| | - Thomas P Wyche
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Keriann M Backus
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA 92307, USA
| | - Timothy M Acker
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Masanori Funabashi
- Department of Bioengineering and Therapeutic Sciences and California Institute for Quantitative Biosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Mao Taketani
- Department of Bioengineering and Therapeutic Sciences and California Institute for Quantitative Biosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Mohamed S Donia
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Stephen Nayfach
- Integrative Program in Quantitative Biology, Gladstone Institutes, and Division of Biostatistics, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Katherine S Pollard
- Integrative Program in Quantitative Biology, Gladstone Institutes, and Division of Biostatistics, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Charles S Craik
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Benjamin F Cravatt
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA 92307, USA
| | - Jon Clardy
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Christopher A Voigt
- Department of Biological Engineering and Synthetic Biology Center, Massachusetts Institute of Technology, Cambridge, MA 02115, USA
| | - Michael A Fischbach
- Department of Bioengineering and Therapeutic Sciences and California Institute for Quantitative Biosciences, University of California, San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
39
|
Brown RL, Clarke TB. The regulation of host defences to infection by the microbiota. Immunology 2017; 150:1-6. [PMID: 27311879 PMCID: PMC5221693 DOI: 10.1111/imm.12634] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 05/27/2016] [Accepted: 06/02/2016] [Indexed: 12/18/2022] Open
Abstract
The skin and mucosal epithelia of humans and other mammals are permanently colonized by large microbial communities (the microbiota). Due to this life-long association with the microbiota, these microbes have an extensive influence over the physiology of their host organism. It is now becoming apparent that nearly all tissues and organ systems, whether in direct contact with the microbiota or in deeper host sites, are under microbial influence. The immune system is perhaps the most profoundly affected, with the microbiota programming both its innate and adaptive arms. The regulation of immunity by the microbiota helps to protect the host against intestinal and extra-intestinal infection by many classes of pathogen. In this review, we will discuss the experimental evidence supporting a role for the microbiota in regulating host defences to extra-intestinal infection, draw together common mechanistic themes, including the central role of pattern recognition receptors, and outline outstanding questions that need to be answered.
Collapse
Affiliation(s)
- Rebecca L. Brown
- MRC Centre for Molecular Bacteriology and InfectionDepartment of MedicineImperial College LondonLondonUK
| | - Thomas B. Clarke
- MRC Centre for Molecular Bacteriology and InfectionDepartment of MedicineImperial College LondonLondonUK
| |
Collapse
|
40
|
García-Castillo V, Sanhueza E, McNerney E, Onate SA, García A. Microbiota dysbiosis: a new piece in the understanding of the carcinogenesis puzzle. J Med Microbiol 2016; 65:1347-1362. [PMID: 27902422 DOI: 10.1099/jmm.0.000371] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Cancer is defined as an uncontrolled proliferation of malignant cells in a host and it is one of the main causes of death worldwide. Genetic and environmental factors play an important role in its development, and the involvement of microbial communities has also recently been recognized. The close relationship that characterizes the colonization by human commensal communities involves health risks, particularly when the homeostasis is disturbed. It has been hypothesized that this process may lead to cancer by modulating the inflammatory response of the host, by the production of carcinogenic metabolic products or by the production of toxins, which disrupt the cell cycle. The metabolic effects of the intestinal microbiota have been studied in greater detail in the gastrointestinal tract, and it has been recognized that microbial communities of other body surfaces can cause effects either locally or at a distance. In vitro and in vivo studies have allowed the characterization of the microbiota and the establishment of a cause and effect relationship with some types of cancer. Nevertheless, despite the results, representative studies are necessary to validate the findings and definitively establish the role of microbiota in cancer development in order to open the possibility of promising therapeutic and diagnostic applications. Thus, the aims of this review are to briefly examine the available evidence, and to analyse the mechanisms described for pancreatic, lung, colorectal cancer , oral squamous cell carcinoma and hepatocellular carcinoma and the impact of the current knowledge about the effects of the microbiota on carcinogenesis.
Collapse
Affiliation(s)
- Valeria García-Castillo
- Department of Microbiology, School of Biological Sciences, Bacterial Pathogenicity Laboratory, University of Concepción, Concepción, Biobío, Chile
| | - Enrique Sanhueza
- Department of Microbiology, School of Biological Sciences, Bacterial Pathogenicity Laboratory, University of Concepción, Concepción, Biobío, Chile
| | - Eileen McNerney
- Molecular Endocrinology and Oncology Laboratory, School of Medicine, University of Concepción, Concepción, Biobío, Chile
| | - Sergio A Onate
- Molecular Endocrinology and Oncology Laboratory, School of Medicine, University of Concepción, Concepción, Biobío, Chile
| | - Apolinaria García
- Department of Microbiology, School of Biological Sciences, Bacterial Pathogenicity Laboratory, University of Concepción, Concepción, Biobío, Chile
| |
Collapse
|
41
|
Berthelot JM, Claudepierre P. Trafficking of antigens from gut to sacroiliac joints and spine in reactive arthritis and spondyloarthropathies: Mainly through lymphatics? Joint Bone Spine 2016; 83:485-90. [DOI: 10.1016/j.jbspin.2015.10.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 10/28/2015] [Indexed: 12/19/2022]
|
42
|
Effects of ceftriaxone induced intestinal dysbacteriosis on lymphocytes in different tissues in mice. Immunobiology 2016; 221:994-1000. [DOI: 10.1016/j.imbio.2016.04.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 04/13/2016] [Indexed: 12/21/2022]
|
43
|
Blander JM. Death in the intestinal epithelium-basic biology and implications for inflammatory bowel disease. FEBS J 2016; 283:2720-30. [PMID: 27250564 PMCID: PMC4956528 DOI: 10.1111/febs.13771] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 05/06/2016] [Accepted: 06/01/2016] [Indexed: 12/11/2022]
Abstract
Every 4-5 days, intestinal epithelial cells (IEC) are terminated as they reach the end of their life. This process ensures that the epithelium is comprised of the fittest cells that maintain an impermeable barrier to luminal contents and the gut microbiota, as well as the most metabolically able cells that conduct functions in nutrient absorption, digestion, and secretion of antimicrobial peptides. IEC are terminated by apical extrusion-or shedding-from the intestinal epithelial monolayer into the gut lumen. Whether death by apoptosis signals extrusion or death follows expulsion by younger IEC has been a matter of debate. Seemingly a minor detail, IEC death before or after apical extrusion bears weight on the potential contribution of apoptotic IEC to intestinal homeostasis as a consequence of their recognition by intestinal lamina propria phagocytes. In inflammatory bowel disease (IBD), excessive death is observed in the ileal and colonic epithelium. The precise mode of IEC death in IBD is not defined. A highly inflammatory milieu within the intestinal lamina propria, rich in the proinflammatory cytokine, TNF-α, increases IEC shedding and compromises barrier integrity fueling more inflammation. A milestone in the treatment of IBD, anti-TNF-α therapy, may promote mucosal healing by reversing increased and inflammation-associated IEC death. Understanding the biology and consequences of cell death in the intestinal epithelium is critical to the design of new avenues for IBD therapy.
Collapse
Affiliation(s)
- J. Magarian Blander
- Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| |
Collapse
|
44
|
Hohlfeld R, Wekerle H. [Multiple sclerosis and microbiota. From genome to metagenome?]. DER NERVENARZT 2016; 86:925-33. [PMID: 26099498 DOI: 10.1007/s00115-014-4248-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The individual risk of contracting multiple sclerosis (MS) is determined by genetic predisposition as well as environmental factors. In monozygotic twins the concordance rate for MS is approximately 30 % indicating that environmental factors are even more important than genetic factors. Observations in a T-cell receptor-transgenic, spontaneous mouse model strongly point to an important contribution of the individual gut microbiome (microbiota). Mice maintained in a germ-free environment are completely protected from experimental autoimmune encephalomyelitis (EAE) in this model, whereas mice that are kept under normal conditions spontaneously develop a relapsing-remitting central nervous system (CNS) disease which is astoundingly similar to human MS. It appears that the autoimmune reaction against CNS tissue is "remotely controlled" by the gut microbiota. This may be explained by the facts that the microbiota influences the gut-associated lymphoid tissue (GALT) and, vice versa, the GALT regulates systemic immunity. The precise role of the microbiota in MS remains to be clarified. New methods of DNA sequencing and bioinformatics allow the analysis of very complex bacterial metagenomes. If individual microbial risk profiles can be identified this would provide completely new perspectives for the prophylaxis and therapy of MS.
Collapse
Affiliation(s)
- R Hohlfeld
- Institut für Klinische Neuroimmunologie, Klinikum der LMU, Campus Großhadern, Marchioninistr. 15, 81377, München, Deutschland,
| | | |
Collapse
|
45
|
Shimizu J, Kubota T, Takada E, Takai K, Fujiwara N, Arimitsu N, Ueda Y, Wakisaka S, Suzuki T, Suzuki N. Bifidobacteria Abundance-Featured Gut Microbiota Compositional Change in Patients with Behcet's Disease. PLoS One 2016; 11:e0153746. [PMID: 27105322 PMCID: PMC4841557 DOI: 10.1371/journal.pone.0153746] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 04/04/2016] [Indexed: 11/21/2022] Open
Abstract
Gut microbiota compositional alteration may have an association with immune dysfunction in patients with Behcet’s disease (BD). We conducted a fecal metagenomic analysis of BD patients. We analyzed fecal microbiota obtained from 12 patients with BD and 12 normal individuals by sequencing of 16S ribosomal RNA gene. We compared the relative abundance of bacterial taxa. Direct comparison of the relative abundance of bacterial taxa demonstrated that the genera Bifidobacterium and Eggerthella increased significantly and the genera Megamonas and Prevotella decreased significantly in BD patients compared with normal individuals. A linear discriminant analysis of bacterial taxa showed that the phylum Actinobacteria, including Bifidobacterium, and the family Lactobacillaceae exhibited larger positive effect sizes than other bacteria in patients with BD. The phylum Firmicutes and the class Clostridia had large effect sizes in normal individuals. There was no significant difference in annotated species numbers (as numbers of operational taxonomic unit; OTU) and bacterial diversity of each sample (alpha diversity) between BD patients and normal individuals. We next assigned each sample to a position using three axes by principal coordinates analysis of the OTU table. The two groups had a significant distance as beta diversity in the 3-axis space. Fecal sIgA concentrations increased significantly in BD patients but did not correlate with any bacterial taxonomic abundance. These data suggest that the compositional changes of gut microbes may be one type of dysbiosis (unfavorable microbiota alteration) in patients with BD. The dysbiosis may have an association with the pathophysiology of BD.
Collapse
Affiliation(s)
- Jun Shimizu
- Department of Immunology and Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Takao Kubota
- Department of Medicine, the Japan Self Defense Forces Central Hospital, Tokyo, Japan
| | - Erika Takada
- Department of Immunology and Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Kenji Takai
- Department of Immunology and Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Naruyoshi Fujiwara
- Department of Immunology and Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Nagisa Arimitsu
- Department of Immunology and Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Yuji Ueda
- Department of Immunology and Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Sueshige Wakisaka
- Department of Immunology and Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Tomoko Suzuki
- Department of Immunology and Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Noboru Suzuki
- Department of Immunology and Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
- * E-mail:
| |
Collapse
|
46
|
Wang P, Li Y, Xiao H, Shi Y, Le GW, Sun J. Isolation of lactobacillus reuteri from Peyer's patches and their effects on sIgA production and gut microbiota diversity. Mol Nutr Food Res 2016; 60:2020-30. [PMID: 27030202 DOI: 10.1002/mnfr.201501065] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2015] [Revised: 02/27/2016] [Accepted: 03/16/2016] [Indexed: 01/04/2023]
Abstract
SCOPE We previously reported that specific Lactobacillus reuteri colonized within mouse Peyer's patches (PP) effectively prevented high fat diet induced obesity and low-grade chronic inflammation. We further investigated the role of PP Lactobacillus reuteri on sIgA production in rats in this study. METHODS AND RESULTS Lactobacilli were isolated from rat PP. All isolates were L. reuteri and belonged to three phenotypes according to amplified fragment length polymorphism analysis. Typical strains of two main clusters, PP1 and PP2, were used to treat control and vitamin A deficient (VAD) rats, respectively. The feeding of PP1 and PP2 affected sIgA and Lactobacillus diversity by strain-specific manner. Free sIgA was significantly increased by PP1 (p = 0.069) and PP2 (p < 0.05) in the control rats but not in the VAD rats. Only PP1 significantly changed PP Lactobacillus diversity in the control rats (p < 0.05). However, PP2 specifically changed ileal Lactobacillus diversity in both control and VAD rats. Fecal sIgA was correlated with PP Lactobacillus diversity (R(2) = 0.7958, p = 0.011). CONCLUSION Modulation of sIgA production by PP L. reuteri of rat is dependent on vitamin A and change of Lactobacillus diversity in PP.
Collapse
Affiliation(s)
- Panpan Wang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, PR China.,Food Nutrition and Functional Factors Research Center, School of Food Science and Technology, Jiangnan University, Wuxi, PR China
| | - Ya Li
- Food Nutrition and Functional Factors Research Center, School of Food Science and Technology, Jiangnan University, Wuxi, PR China
| | - Hang Xiao
- Department of Food Science, University of Massachusetts, Amherst, MA, USA
| | - Yonghui Shi
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, PR China.,Food Nutrition and Functional Factors Research Center, School of Food Science and Technology, Jiangnan University, Wuxi, PR China
| | - Guo-Wei Le
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, PR China.,Food Nutrition and Functional Factors Research Center, School of Food Science and Technology, Jiangnan University, Wuxi, PR China
| | - Jin Sun
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, PR China. .,Food Nutrition and Functional Factors Research Center, School of Food Science and Technology, Jiangnan University, Wuxi, PR China.
| |
Collapse
|
47
|
Leszczyszyn JJ, Radomski M, Leszczyszyn AM. Intestinal microbiota transplant - current state of knowledge. Reumatologia 2016; 54:24-8. [PMID: 27407273 PMCID: PMC4847330 DOI: 10.5114/reum.2016.58758] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 02/26/2016] [Indexed: 12/21/2022] Open
Abstract
Faecal microbiota transplantation (FMT) has induced a lot scientific interest and hopes for the last couple of years. FMT has been approved as a treatment of recurrent Clostridium difficile colitis. Highly sophisticated molecular DNA identification methods have been used to assess the healthy human microbiome as well as its disturbances in several diseases. The metabolic and immunologic functions of the microbiome have become more clear and understandable. A lot of pathological changes, such as production of short-chain fatty acids or components of the inflammatory cascade, caused by changes in microbiome diversity, variability and richness have been observed among patients suffering from inflammatory bowel diseases, irritable bowel syndrome, type 2 diabetes or rheumatoid arthritis. The published clinical results are encouraging, but still there is huge demand for FMT controlled clinical trials.
Collapse
Affiliation(s)
- Jarosław Jerzy Leszczyszyn
- National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
- Biotransplant Research, Wroclaw, Poland
| | - Marek Radomski
- National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
- School of Pharmacy and Pharmaceutical Sciences, University of Dublin, Trinity College, Dublin, Ireland
| | | |
Collapse
|
48
|
Fung TC, Bessman NJ, Hepworth MR, Kumar N, Shibata N, Kobuley D, Wang K, Ziegler CGK, Goc J, Shima T, Umesaki Y, Sartor RB, Sullivan KV, Lawley TD, Kunisawa J, Kiyono H, Sonnenberg GF. Lymphoid-Tissue-Resident Commensal Bacteria Promote Members of the IL-10 Cytokine Family to Establish Mutualism. Immunity 2016; 44:634-646. [PMID: 26982365 PMCID: PMC4845739 DOI: 10.1016/j.immuni.2016.02.019] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2014] [Revised: 09/16/2015] [Accepted: 12/29/2015] [Indexed: 12/19/2022]
Abstract
Physical separation between the mammalian immune system and commensal bacteria is necessary to limit chronic inflammation. However, selective species of commensal bacteria can reside within intestinal lymphoid tissues of healthy mammals. Here, we demonstrate that lymphoid-tissue-resident commensal bacteria (LRC) colonized murine dendritic cells and modulated their cytokine production. In germ-free and antibiotic-treated mice, LRCs colonized intestinal lymphoid tissues and induced multiple members of the IL-10 cytokine family, including dendritic-cell-derived IL-10 and group 3 innate lymphoid cell (ILC3)-derived IL-22. Notably, IL-10 limited the development of pro-inflammatory Th17 cell responses, and IL-22 production enhanced LRC colonization in the steady state. Furthermore, LRC colonization protected mice from lethal intestinal damage in an IL-10-IL-10R-dependent manner. Collectively, our data reveal a unique host-commensal-bacteria dialog whereby selective subsets of commensal bacteria interact with dendritic cells to facilitate tissue-specific responses that are mutually beneficial for both the host and the microbe.
Collapse
Affiliation(s)
- Thomas C Fung
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Joan and Sanford I. Weill Department of Medicine, Division of Gastroenterology, Weill Cornell Medicine, New York, NY 10021 USA; Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY 10021 USA; Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, New York, NY 10021, USA
| | - Nicholas J Bessman
- Joan and Sanford I. Weill Department of Medicine, Division of Gastroenterology, Weill Cornell Medicine, New York, NY 10021 USA; Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY 10021 USA; Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, New York, NY 10021, USA
| | - Matthew R Hepworth
- Joan and Sanford I. Weill Department of Medicine, Division of Gastroenterology, Weill Cornell Medicine, New York, NY 10021 USA; Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY 10021 USA; Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, New York, NY 10021, USA
| | - Nitin Kumar
- Host Microbiota Interactions Laboratory, Wellcome Trust Sanger Institute, Hinxton CB10 1SA, UK
| | - Naoko Shibata
- Department of Microbiology and Immunology, The Institute of Medical Science, The University of Toyko, Toyko 108-8639, Japan; Japan Science and Technology Agency, Core Research for Evolutional Science and Technology, Tokyo 102-0076, Japan
| | - Dmytro Kobuley
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kelvin Wang
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Carly G K Ziegler
- Department of Computational Biology and Immunology, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA
| | - Jeremy Goc
- Joan and Sanford I. Weill Department of Medicine, Division of Gastroenterology, Weill Cornell Medicine, New York, NY 10021 USA; Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY 10021 USA; Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, New York, NY 10021, USA
| | | | | | - R Balfour Sartor
- Department of Microbiology and Immunology, Center for Gastrointestinal Biology and Disease, University of North Carolina, Chapel Hill, NC 27599-7032, USA
| | - Kaede V Sullivan
- The Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Trevor D Lawley
- Host Microbiota Interactions Laboratory, Wellcome Trust Sanger Institute, Hinxton CB10 1SA, UK
| | - Jun Kunisawa
- Laboratory of Vaccine Materials, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan
| | - Hiroshi Kiyono
- Department of Microbiology and Immunology, The Institute of Medical Science, The University of Toyko, Toyko 108-8639, Japan; Japan Science and Technology Agency, Core Research for Evolutional Science and Technology, Tokyo 102-0076, Japan
| | - Gregory F Sonnenberg
- Joan and Sanford I. Weill Department of Medicine, Division of Gastroenterology, Weill Cornell Medicine, New York, NY 10021 USA; Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY 10021 USA; Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, New York, NY 10021, USA.
| |
Collapse
|
49
|
Abstract
Detailed analyses of the gut microbiome and its effect on human physiology and disease are emerging, thanks to advances in high-throughput DNA-sequencing technology and the burgeoning field of metagenomics. Metagenomics examines the structure and functional potential of microbial communities in their native habitats through the direct isolation and analysis of community DNA. In inflammatory bowel disease, gut microbiome studies have shown an association with perturbations in community composition and, especially, function. In this review, we discuss the application of next-generation sequencing to microbiome research and highlight the importance of modeling microbiome structure and function to the future of inflammatory bowel disease research and treatment.
Collapse
|
50
|
Deng K, Chen T, Wu Q, Xin H, Wei Q, Hu P, Wang X, Wang X, Wei H, Shah NP. In vitro and in vivo examination of anticolonization of pathogens by Lactobacillus paracasei FJ861111.1. J Dairy Sci 2015; 98:6759-66. [DOI: 10.3168/jds.2015-9761] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 06/20/2015] [Indexed: 12/24/2022]
|