551
|
Goto Y. Epithelial Cells as a Transmitter of Signals From Commensal Bacteria and Host Immune Cells. Front Immunol 2019; 10:2057. [PMID: 31555282 PMCID: PMC6724641 DOI: 10.3389/fimmu.2019.02057] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 08/14/2019] [Indexed: 12/20/2022] Open
Abstract
Intestinal epithelial cells (IECs) are non-hematopoietic cells that form a physical barrier against external antigens. Recent studies indicate that IECs have pleiotropic functions in the regulation of luminal microbiota and the host immune system. IECs produce various immune modulatory cytokines and chemokines in response to commensal bacteria and contribute to developing the intestinal immune system. In contrast, IECs receive cytokine signals from immune cells and produce various immunological factors against luminal bacteria. This bidirectional function of IECs is critical to regulate homeostasis of microbiota and the host immune system. Disruption of the epithelial barrier leads to detrimental host diseases such as inflammatory bowel disease, colonic cancer, and pathogenic infection. This review provides an overview of the functions and physiology of IECs and highlights their bidirectional functions against luminal bacteria and immune cells, which contribute to maintaining gut homeostasis.
Collapse
Affiliation(s)
- Yoshiyuki Goto
- Division of Molecular Immunology, Medical Mycology Research Center, Chiba University, Chiba, Japan.,Division of Mucosal Symbiosis, International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
552
|
Cerf-Bensussan N. Microbiology and immunology: An ideal partnership for a tango at the gut surface-A tribute to Philippe Sansonetti. Cell Microbiol 2019; 21:e13097. [PMID: 31414516 PMCID: PMC7027583 DOI: 10.1111/cmi.13097] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 08/02/2019] [Accepted: 08/12/2019] [Indexed: 12/12/2022]
Abstract
Over the past 20 years, the highly dynamic interactions that take place between hosts and the gut microbiota have emerged as a major determinant in health and disease. The complexity of the gut microbiota represents, however, a considerable challenge, and reductionist approaches are indispensable to define the contribution of individual bacteria to host responses and to dissect molecular mechanisms. In this tribute to Philippe Sansonetti, I would like to show how rewarding collaborations with microbiologists have guided our team of immunologists in the study of host–microbiota interactions and, thanks to the use of controlled colonisation experiments in gnotobiotic mice, toward the demonstration that segmented filamentous bacteria (SFB) are indispensable to drive the post‐natal maturation of the gut immune barrier in mice. The work led with Philippe Sansonetti to set up in vitro culture conditions has been one important milestone that laid the ground for in‐depth characterization of the molecular attributes of this unusual symbiont. Recent suggestions that SFB may be present in the human microbiota encourage further cross‐fertilising interactions between microbiologists and immunologists to define whether results from mice can be translated to humans and, if so, how SFB may be used to promote human intestinal defences against enteropathogens. Nurturing the competences to pursue this inspiring project is one legacy of Philippe Sansonetti.
Collapse
Affiliation(s)
- Nadine Cerf-Bensussan
- Laboratory of Intestinal Immunity, INSERM UMR 1163, Institut Imagine, Paris, France.,Université de Paris, Paris, France
| |
Collapse
|
553
|
Oral Bacteria and Intestinal Dysbiosis in Colorectal Cancer. Int J Mol Sci 2019; 20:ijms20174146. [PMID: 31450675 PMCID: PMC6747549 DOI: 10.3390/ijms20174146] [Citation(s) in RCA: 169] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 08/19/2019] [Accepted: 08/23/2019] [Indexed: 02/07/2023] Open
Abstract
The human organism coexists with its microbiota in a symbiotic relationship. These polymicrobial communities are involved in many crucial functions, such as immunity, protection against pathogens, and metabolism of dietary compounds, thus maintaining homeostasis. The oral cavity and the colon, although distant anatomic regions, are both highly colonized by distinct microbiotas. However, studies indicate that oral bacteria are able to disseminate into the colon. This is mostly evident in conditions such as periodontitis, where specific bacteria, namely Fusobacterium nucrelatum and Porphyromonas gingivalis project a pathogenic profile. In the colon these bacteria can alter the composition of the residual microbiota, in the context of complex biofilms, resulting in intestinal dysbiosis. This orally-driven disruption promotes aberrant immune and inflammatory responses, eventually leading to colorectal cancer (CRC) tumorigenesis. Understanding the exact mechanisms of these interactions will yield future opportunities regarding prevention and treatment of CRC.
Collapse
|
554
|
van der Meulen TA, Harmsen HJM, Bootsma H, Liefers SC, Vich Vila A, Zhernakova A, Fu J, Wijmenga C, Spijkervet FKL, Kroese FGM, Vissink A. Dysbiosis of the buccal mucosa microbiome in primary Sjögren's syndrome patients. Rheumatology (Oxford) 2019; 57:2225-2234. [PMID: 30060225 DOI: 10.1093/rheumatology/key215] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Indexed: 12/14/2022] Open
Abstract
Objectives Environmental factors in the aetiology of primary Sjögren's syndrome (pSS) are largely unknown. Host-microbiome interaction at mucosal surfaces is presumed to be involved in the aetiopathogenesis of pSS. Here, we assessed whether the microbiome of the buccal mucosa is specific for pSS compared with symptom-controls. Methods The bacterial composition of buccal swab samples from 37 pSS patients, 86 non-SS sicca patients (with similar dryness symptoms to pSS patients, but not fulfilling the classification criteria) and 24 healthy controls (HCs) was determined with 16S rRNA sequencing. Multivariate Association with Linear Models was used to find associations between individual taxa and pSS, taking into account smoking and dental status. Associations were replicated in a general population cohort (n = 103). Results The buccal mucosa microbiome of pSS and non-SS sicca patients both differed from HCs. A higher Firmicutes/Proteobacteria ratio was characteristic for both pSS and non-SS sicca patients. Disease status (pSS, non-SS sicca, HCs) and salivary secretion rate contributed almost equally to the variation in bacterial composition between individuals (3.8 and 4.3%, respectively). Two taxa were associated with pSS compared with non-SS sicca patients and 19 compared with HCs. When salivary secretion rate was taken into account, no taxon was associated with pSS compared with non-SS sicca. Twelve of the 19 pSS-associated taxa were correlated with salivary secretion. Conclusion Dysbiosis of the buccal mucosa microbiome in pSS patients resembles that of symptom-controls. The buccal mucosa microbiome in pSS patients is determined by a combination of reduced salivary secretion and disease-specific factors.
Collapse
Affiliation(s)
- Taco A van der Meulen
- Department of Oral and Maxillofacial Surgery, University Medical Center Groningen, Groningen, The Netherlands
| | - Hermie J M Harmsen
- Department of Medical Microbiology, University Medical Center Groningen, Groningen, The Netherlands
| | - Hendrika Bootsma
- Department of Rheumatology & Clinical Immunology, University Medical Center Groningen, Groningen, The Netherlands
| | - Silvia C Liefers
- Department of Oral and Maxillofacial Surgery, University Medical Center Groningen, Groningen, The Netherlands
| | - Arnau Vich Vila
- Department of Gastroenterology, University Medical Center Groningen, Groningen, The Netherlands.,Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Alexandra Zhernakova
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jingyuan Fu
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Cisca Wijmenga
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Fred K L Spijkervet
- Department of Oral and Maxillofacial Surgery, University Medical Center Groningen, Groningen, The Netherlands
| | - Frans G M Kroese
- Department of Rheumatology & Clinical Immunology, University Medical Center Groningen, Groningen, The Netherlands
| | - Arjan Vissink
- Department of Oral and Maxillofacial Surgery, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
555
|
Tilg H, Zmora N, Adolph TE, Elinav E. The intestinal microbiota fuelling metabolic inflammation. Nat Rev Immunol 2019; 20:40-54. [DOI: 10.1038/s41577-019-0198-4] [Citation(s) in RCA: 377] [Impact Index Per Article: 62.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/05/2019] [Indexed: 02/06/2023]
|
556
|
Kanamori M, Nakatsukasa H, Ito M, Chikuma S, Yoshimura A. Reprogramming of Th1 cells into regulatory T cells through rewiring of the metabolic status. Int Immunol 2019; 30:357-373. [PMID: 29982622 DOI: 10.1093/intimm/dxy043] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 06/28/2018] [Indexed: 12/16/2022] Open
Abstract
T helper type 1 (Th1) cells form one of the most stable CD4 T-cell subsets, and direct conversion of fully differentiated Th1 to regulatory T (Treg) cells has been poorly investigated. Here, we established a culture method for inducing Foxp3 from Th1 cells of mice and humans. This is achieved simply by resting Th1 cells without T-cell receptor ligation before stimulation in the presence of transforming growth factor-beta (TGF-β). We named the resulting Th1-derived Foxp3+ cells Th1reg cells. Mouse Th1reg cells showed an inducible Treg-like phenotype and suppressive ability both in vitro and in vivo. Th1reg cells could also be induced from in vivo-developed mouse Th1 cells. Unexpectedly, the resting process enabled Foxp3 expression not through epigenetic changes at the locus, but through metabolic change resulting from reduced mammalian target of rapamycin complex 1 (mTORC1) activity. mTORC1 suppressed TGF-β-induced phosphorylation of Smad2/3 in Th1 cells, which was restored in rested cells. Our study warrants future research aiming at development of immunotherapy with Th1reg cells.
Collapse
Affiliation(s)
- Mitsuhiro Kanamori
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Hiroko Nakatsukasa
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Minako Ito
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Shunsuke Chikuma
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Akihiko Yoshimura
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| |
Collapse
|
557
|
Zhang HN, Zhou XD, Xu X, Wang Y. [Oral microbiota and inflammatory bowel disease]. HUA XI KOU QIANG YI XUE ZA ZHI = HUAXI KOUQIANG YIXUE ZAZHI = WEST CHINA JOURNAL OF STOMATOLOGY 2019; 37:443-449. [PMID: 31512842 DOI: 10.7518/hxkq.2019.04.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory disease of the gastrointestinal tract with a high incidence but a poor therapeutic outcome. However, IBD is generally caused by complicated interactions between environmental factors and gut microflora in genetically susceptible individuals. In view of a series of oral manifestations in patients with IBD and a high detection rate of oral bacteria among this population, oral microbiota may play an important role in the development of IBD. This article reviews the relationship between oral microbiota and IBD.
Collapse
Affiliation(s)
- Hao-Nan Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Xue-Dong Zhou
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Xin Xu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yan Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
558
|
Abstract
Pediatric ulcerative colitis incidence is rapidly rising, yet improved prognostic and therapeutic strategies are needed. In this issue of Cell Host & Microbe, Schirmer et al. (2018) reveal the dynamism of pediatric patient microbiomes through initial diagnosis and treatments, providing insights into microbial targets that predict therapeutic response and disease outcomes.
Collapse
|
559
|
Kitamura M, Mochizuki Y, Miyata Y, Obata Y, Mitsunari K, Matsuo T, Ohba K, Mukae H, Yoshimura A, Nishino T, Sakai H. Pathological Characteristics of Periodontal Disease in Patients with Chronic Kidney Disease and Kidney Transplantation. Int J Mol Sci 2019; 20:ijms20143413. [PMID: 31336777 PMCID: PMC6678374 DOI: 10.3390/ijms20143413] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Revised: 07/06/2019] [Accepted: 07/10/2019] [Indexed: 02/07/2023] Open
Abstract
Chronic kidney disease (CKD) is recognized as an irreversible reduction of functional nephrons and leads to an increased risk of various pathological conditions, including cardiovascular disease and neurological disorders, such as coronary artery calcification, hypertension, and stroke. In addition, CKD patients have impaired immunity against bacteria and viruses. Conversely, kidney transplantation (KT) is performed for patients with end-stage renal disease as a renal replacement therapy. Although kidney function is almost normalized by KT, immunosuppressive therapy is essential to maintain kidney allograft function and to prevent rejection. However, these patients are more susceptible to infection due to the immunosuppressive therapy required to maintain kidney allograft function. Thus, both CKD and KT present disadvantages in terms of suppression of immune function. Periodontal disease is defined as a chronic infection and inflammation of oral and periodontal tissues. Periodontal disease is characterized by the destruction of connective tissues of the periodontium and alveolar bone, which may lead to not only local symptoms but also systemic diseases, such as cardiovascular diseases, diabetes, liver disease, chronic obstructive pulmonary disease, and several types of cancer. In addition, the prevalence and severity of periodontal disease are significantly associated with mortality. Many researchers pay special attention to the pathological roles and clinical impact of periodontal disease in patients with CKD or KT. In this review, we provide information regarding important modulators of periodontal disease to better understand the relationship between periodontal disease and CKD and/or KT. Furthermore; we evaluate the impact of periodontal disease on various pathological conditions in patients with CKD and KT. Moreover, pathogens of periodontal disease common to CKD and KT are also discussed. Finally, we examine the importance of periodontal care in these patients. Thus, this review provides a comprehensive overview of the pathological roles and clinical significance of periodontal disease in patients with CKD and KT.
Collapse
Affiliation(s)
- Mineaki Kitamura
- Department of Nephrology, Nagasaki University Hospital, Nagasaki 852-8501, Japan
- Division of Blood Purification, Nagasaki University Hospital, Nagasaki 852-8501, Japan
| | - Yasushi Mochizuki
- Division of Blood Purification, Nagasaki University Hospital, Nagasaki 852-8501, Japan
- Department of Urology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Yasuyoshi Miyata
- Department of Urology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan.
| | - Yoko Obata
- Department of Nephrology, Nagasaki University Hospital, Nagasaki 852-8501, Japan
| | - Kensuke Mitsunari
- Department of Urology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Tomohiro Matsuo
- Department of Urology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Kojiro Ohba
- Department of Urology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Hiroshi Mukae
- Department of Respiratory Medicine, Unit of Basic Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Atsutoshi Yoshimura
- Department of Periodontology and Endodontology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Tomoya Nishino
- Department of Nephrology, Nagasaki University Hospital, Nagasaki 852-8501, Japan
| | - Hideki Sakai
- Division of Blood Purification, Nagasaki University Hospital, Nagasaki 852-8501, Japan
- Department of Urology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| |
Collapse
|
560
|
Wang L, Yin G, Guo Y, Zhao Y, Zhao M, Lai Y, Sui P, Shi T, Guo W, Huang Z. Variations in Oral Microbiota Composition Are Associated With a Risk of Throat Cancer. Front Cell Infect Microbiol 2019; 9:205. [PMID: 31334130 PMCID: PMC6618584 DOI: 10.3389/fcimb.2019.00205] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 05/27/2019] [Indexed: 12/23/2022] Open
Abstract
In this study, a next-generation sequencing strategy on 16S ribosomal RNA (16S rRNA) gene was employed to analyze 70 oral samples from 32 patients with throat cancer, nine patients with vocal cord polyp, and 29 healthy individuals (normal controls). Using this strategy, we demonstrated, for the first time, that the salivary microbiota of cancer patients were significantly different from those of patients with a polyp and healthy individuals. We observed that the beta diversity of the cancer group was divergent from both the normal and polyp groups, while alpha-diversity indices such as the Chao1 estimator (P = 8.1e-05), Simpson (P = 0.0045), and Shannon (P = 0.0071) were significantly reduced in cancer patients compared with patients containing a polyp and normal healthy individuals. Linear discriminant analysis (LDA) and Kruskal–Wallis test analyses and real-time quantitative polymerase chain reaction (qPCR) verification test revealed that the genera Aggregatibacter, Pseudomonas, Bacteroides, and Ruminiclostridium were significantly enriched in the throat cancer group compared with the vocal cord polyp and normal control groups (score value >2). Finally, diagnostic models based on putatively important constituent bacteria were constructed with 87.5% accuracy [area under the curve (AUC) = 0.875, 95% confidence interval (CI): 0.695–1]. In summary, in this study we characterized, for the first time, the oral microbiota of throat cancer patients without smoking history. We speculate that these results will help in the pathogenic mechanism and early diagnosis of throat cancer.
Collapse
Affiliation(s)
- Lili Wang
- Beijing Cheer Land Biotechnology Co., Ltd., CL Investment Group, Beijing, China
| | - Gaofei Yin
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Ying Guo
- Beijing Cheer Land Biotechnology Co., Ltd., CL Investment Group, Beijing, China
| | - Yaqi Zhao
- Beijing Cheer Land Biotechnology Co., Ltd., CL Investment Group, Beijing, China
| | - Meng Zhao
- Beijing Cheer Land Biotechnology Co., Ltd., CL Investment Group, Beijing, China
| | - Yunyun Lai
- Beijing Cheer Land Biotechnology Co., Ltd., CL Investment Group, Beijing, China
| | - Pengcheng Sui
- Beijing Cheer Land Biotechnology Co., Ltd., CL Investment Group, Beijing, China
| | - Taiping Shi
- Beijing Cheer Land Biotechnology Co., Ltd., CL Investment Group, Beijing, China
| | - Wei Guo
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Zhigang Huang
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
561
|
Yamashita T, Kawada-Matsuo M, Katsumata T, Watanabe A, Oogai Y, Nishitani Y, Miyawaki S, Komatsuzawa H. Antibacterial activity of disodium succinoyl glycyrrhetinate, a derivative of glycyrrhetinic acid against Streptococcus mutans. Microbiol Immunol 2019; 63:251-260. [PMID: 31166029 DOI: 10.1111/1348-0421.12717] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 05/09/2019] [Accepted: 05/23/2019] [Indexed: 11/28/2022]
Abstract
Streptococcus mutans is a cariogenic bacterium that localizes in the oral cavity. Glycyrrhetinic acid (GRA) is a major component of licorice extract. GRA and several derivatives, including disodium succinoyl glycyrrhetinate (GR-SU), are known to have anti-inflammatory effects in humans. In this study, the antimicrobial effect of GRA and its derivatives against the S. mutans UA159 strain were investigated. Minimum inhibitory concentrations (MICs) of GRA and GR-SU showed antibacterial activity against the S. mutans strain, whereas other tested derivatives did not. Because GR-SU is more soluble than GRA, GR-SU was used for further experiments. The antibacterial activity of GR-SU against 100 S. mutans strains was evaluated and it was found that all strains are susceptible to GR-SU, with MIC values below 256 µg/mL. A cell viability assay showed that GR-SU has a bacteriostatic effect on S. mutans cells. As to growth kinetics, sub-MICs of GR-SU inhibited growth. The effect of GR-SU on S. mutans virulence was then investigated. GR-SU at sub-MICs suppresses biofilm formation. Additionally, GR-SU greatly suppresses the pH drop caused by the addition of glucose and glucose-induced expression of the genes responsible for acid production (ldh and pykF) and tolerance (aguD and atpD). Additionally, expression of enolase, which is responsible for the carbohydrate phosphotransferase system, was not increased in the presence of GR-SU, indicating that GR-SU suppresses incorporation of sugars into S. mutans. In conclusion, GR-SU has antibacterial activity against S. mutans and also decreases S. mutans virulence.
Collapse
Affiliation(s)
- Takahito Yamashita
- Department of Oral Microbiology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Miki Kawada-Matsuo
- Department of Oral Microbiology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Tamaki Katsumata
- Department of Restorative Dentistry and Endodontology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Atsuko Watanabe
- Department of Orthodontics, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Yuichi Oogai
- Department of Oral Microbiology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Yoshihiro Nishitani
- Department of Restorative Dentistry and Endodontology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Shouichi Miyawaki
- Department of Orthodontics, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Hitoshi Komatsuzawa
- Department of Bacteriology, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| |
Collapse
|
562
|
Konkel JE, O'Boyle C, Krishnan S. Distal Consequences of Oral Inflammation. Front Immunol 2019; 10:1403. [PMID: 31293577 PMCID: PMC6603141 DOI: 10.3389/fimmu.2019.01403] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Accepted: 06/03/2019] [Indexed: 12/11/2022] Open
Abstract
Periodontitis is an incredibly prevalent chronic inflammatory disease, which results in the destruction of tooth supporting structures. However, in addition to causing tooth and alveolar bone loss, this oral inflammatory disease has been shown to contribute to disease states and inflammatory pathology at sites distant from the oral cavity. Epidemiological and experimental studies have linked periodontitis to the development and/or exacerbation of a plethora of other chronic diseases ranging from rheumatoid arthritis to Alzheimer's disease. Such studies highlight how the inflammatory status of the oral cavity can have a profound impact on systemic health. In this review we discuss the disease states impacted by periodontitis and explore potential mechanisms whereby oral inflammation could promote loss of homeostasis at distant sites.
Collapse
Affiliation(s)
- Joanne E. Konkel
- Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
- Manchester Collaborative Centre for Inflammation Research (MCCIR), University of Manchester, Manchester, United Kingdom
| | - Conor O'Boyle
- Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Siddharth Krishnan
- Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
- Manchester Collaborative Centre for Inflammation Research (MCCIR), University of Manchester, Manchester, United Kingdom
| |
Collapse
|
563
|
Jiménez-Hernández N, Serrano-Villar S, Domingo A, Pons X, Artacho A, Estrada V, Moya A, Gosalbes MJ. Modulation of Saliva Microbiota through Prebiotic Intervention in HIV-Infected Individuals. Nutrients 2019; 11:1346. [PMID: 31208015 PMCID: PMC6627446 DOI: 10.3390/nu11061346] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 06/07/2019] [Accepted: 06/12/2019] [Indexed: 02/07/2023] Open
Abstract
Human immunodeficiency virus (HIV) infection is characterized by an early depletion of the mucosal associated T helper (CD4+) cells that impair the host immunity and impact the oral and gut microbiomes. Although, the HIV-associated gut microbiota was studied in depth, few works addressed the dysbiosis of oral microbiota in HIV infection and, to our knowledge, no studies on intervention with prebiotics were performed. We studied the effect of a six-week-long prebiotic administration on the salivary microbiota in HIV patients and healthy subjects. Also, the co-occurrence of saliva microorganisms in the fecal bacteria community was explored. We assessed salivary and feces microbiota composition using deep 16S ribosomal RNA (rRNA) gene sequencing with Illumina methodology. At baseline, the different groups shared the same most abundant genera, but the HIV status had an impact on the saliva microbiota composition and diversity parameters. After the intervention with prebiotics, we found a drastic decrease in alpha diversity parameters, as well as a change of beta diversity, without a clear directionality toward a healthy microbiota. Interestingly, we found a differential response to the prebiotics, depending on the initial microbiota. On the basis of 100% identity clustering, we detected saliva sequences in the feces datasets, suggesting a drag of microorganisms from the upper to the lower gastrointestinal tract.
Collapse
Affiliation(s)
- Nuria Jiménez-Hernández
- Área de Genómica y Salud, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valenciana (FISABIO), 46020 Valencia, Spain.
- CIBER en Epidemiología y Salud Pública, 28029 Madrid, Spain.
| | - Sergio Serrano-Villar
- Departamento de Enfermedades Infecciosas, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain.
| | - Alba Domingo
- Área de Genómica y Salud, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valenciana (FISABIO), 46020 Valencia, Spain.
| | - Xavier Pons
- Área de Genómica y Salud, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valenciana (FISABIO), 46020 Valencia, Spain.
| | - Alejandro Artacho
- Área de Genómica y Salud, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valenciana (FISABIO), 46020 Valencia, Spain.
| | - Vicente Estrada
- Unidad de Enfermedades Infecciosas/Medicina Interna, Hospital Clínico San Carlos-IdiSSC, Universidad Complutense, 28040 Madrid, Spain.
| | - Andrés Moya
- Área de Genómica y Salud, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valenciana (FISABIO), 46020 Valencia, Spain.
- CIBER en Epidemiología y Salud Pública, 28029 Madrid, Spain.
- Instituto de Biología Integrativa de Sistemas, Universidad de Valencia y CSIC, 46980 Paterna, Valencia, Spain.
| | - María José Gosalbes
- Área de Genómica y Salud, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valenciana (FISABIO), 46020 Valencia, Spain.
- CIBER en Epidemiología y Salud Pública, 28029 Madrid, Spain.
| |
Collapse
|
564
|
Klebsiella and Providencia emerge as lone survivors following long-term starvation of oral microbiota. Proc Natl Acad Sci U S A 2019; 116:8499-8504. [PMID: 30975748 PMCID: PMC6486781 DOI: 10.1073/pnas.1820594116] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
It is well-understood that many bacteria have evolved to survive catastrophic events using a variety of mechanisms, which include expression of stress-response genes, quiescence, necrotrophy, and metabolic advantages obtained through mutation. However, the dynamics of individuals leveraging these abilities to gain a competitive advantage in an ecologically complex setting remain unstudied. In this study, we observed the saliva microbiome throughout the ecological perturbation of long-term starvation, allowing only the species best equipped to access and use the limited resources to survive. During the first several days, the community underwent a death phase that resulted in a ∼50-100-fold reduction in the number of viable cells. Interestingly, after this death phase, only three species, Klebsiella pneumoniae, Klebsiella oxytoca, and Providencia alcalifaciens, all members of the family Enterobacteriaceae, appeared to be transcriptionally active and recoverable. Klebsiella are significant human pathogens, frequently resistant to multiple antibiotics, and recently, ectopic colonization of the gut by oral Klebsiella was documented to induce dysbiosis and inflammation. MetaOmics analyses provided several leads for further investigation regarding the ecological success of the Enterobacteriaceae. The isolates accumulated single nucleotide polymorphisms in known growth advantage in stationary phase alleles and produced natural products closely resembling antimicrobial cyclic depsipeptides. The results presented in this study suggest that pathogenic Enterobacteriaceae persist much longer than their more benign neighbors in the salivary microbiome when faced with starvation. This is particularly significant, given that hospital surfaces contaminated with oral fluids, especially sinks and drains, are well-established sources of outbreaks of drug-resistant Enterobacteriaceae.
Collapse
|
565
|
Li J, Wu T, Li N, Wang X, Chen G, Lyu X. Bilberry anthocyanin extract promotes intestinal barrier function and inhibits digestive enzyme activity by regulating the gut microbiota in aging rats. Food Funct 2019; 10:333-343. [PMID: 30575836 DOI: 10.1039/c8fo01962b] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
This study was aimed at understanding potential mechanisms regarding bilberry anthocyanin extract consumption and healthy aging and the effects on intestinal barrier function and digestive enzyme activity, through regulating the gut microbiota in aging rats. Medium-dose bilberry anthocyanin extract consumption (20 mg per kg bw per day) was the optimum amount to regulate the intestinal function of aging rats. After consumption, bacteria beneficial to the intestine (Aspergillus oryzae, Lactobacillus, Bacteroides, Clostridiaceae-1, the Bacteroidales-S24-7-group and the Lachnospiraceae_NK4A136_group) were induced to grow, and harmful bacteria (Verrucomicrobia and Euryarchaeota) were inhibited. However, high-dose bilberry anthocyanin extract consumption altered some intestinally beneficial bacteria in an adverse way. There was a correlation between changes in bacterial composition and changes in short-chain fatty acids and the intestinal mucosal barrier. Bilberry anthocyanin extract consumption also decreased the activity of digestive enzymes. Our results suggest that bilberry anthocyanin extract consumption is a potential approach for assisting healthy aging.
Collapse
Affiliation(s)
- Jing Li
- State Key Laboratory of Food Nutrition and Safety (Tianjin University of Science and Technology), Tianjin 300457, China.
| | | | | | | | | | | |
Collapse
|
566
|
Paramsothy S, Nielsen S, Kamm MA, Deshpande NP, Faith JJ, Clemente JC, Paramsothy R, Walsh AJ, van den Bogaerde J, Samuel D, Leong RWL, Connor S, Ng W, Lin E, Borody TJ, Wilkins MR, Colombel JF, Mitchell HM, Kaakoush NO. Specific Bacteria and Metabolites Associated With Response to Fecal Microbiota Transplantation in Patients With Ulcerative Colitis. Gastroenterology 2019; 156:1440-1454.e2. [PMID: 30529583 DOI: 10.1053/j.gastro.2018.12.001] [Citation(s) in RCA: 338] [Impact Index Per Article: 56.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 11/30/2018] [Accepted: 12/03/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Fecal microbiota transplantation (FMT) can induce remission in patients with ulcerative colitis (UC). In a randomized controlled trial of FMT in patients with active UC, we aimed to identify bacterial taxonomic and functional factors associated with response to therapy. METHODS We performed a double-blind trial of 81 patients with active UC randomly assigned to groups that received an initial colonoscopic infusion and then intensive multidonor FMT or placebo enemas, 5 d/wk for 8 weeks. Patients in the FMT group received blended homogenized stool from 3-7 unrelated donors. Patients in the placebo group were eligible to receive open-label FMT after the double-blind study period. We collected 314 fecal samples from the patients at screening, every 4 weeks during treatment, and 8 weeks after the blinded or open-label FMT therapy. We also collected 160 large-bowel biopsy samples from the patients at study entry, at completion of 8 weeks of blinded therapy, and at the end of open-label FMT, if applicable. We analyzed 105 fecal samples from the 14 individual donors (n = 55), who in turn contributed to 21 multidonor batches (n = 50). Bacteria in colonic and fecal samples were analyzed by both 16S ribosomal RNA gene and transcript amplicon sequencing; 285 fecal samples were analyzed by shotgun metagenomics, and 60 fecal samples were analyzed for metabolome features. RESULTS FMT increased microbial diversity and altered composition, based on analyses of colon and fecal samples collected before vs after FMT. Diversity was greater in fecal and colon samples collected before and after FMT treatment from patients who achieved remission compared with patients who did not. Patients in remission after FMT had enrichment of Eubacterium hallii and Roseburia inulivorans compared with patients who did not achieve remission after FMT and had increased levels of short-chain fatty acid biosynthesis and secondary bile acids. Patients who did not achieve remission had enrichment of Fusobacterium gonidiaformans, Sutterella wadsworthensis, and Escherichia species and increased levels of heme and lipopolysaccharide biosynthesis. Bacteroides in donor stool were associated with remission in patients receiving FMT, and Streptococcus species in donor stool was associated with no response to FMT. CONCLUSIONS In an analysis of fecal and colonic mucosa samples from patients receiving FMT for active UC and stool samples from donors, we associated specific bacteria and metabolic pathways with induction of remission. These findings may be of value in the design of microbe-based therapies for UC. ClinicalTrials.gov, Number NCT01896635.
Collapse
Affiliation(s)
- Sudarshan Paramsothy
- University of New South Wales, Sydney, Australia; Icahn School of Medicine at Mount Sinai, New York, New York; St Vincent's Hospital, Sydney, Australia; Nambour General Hospital, Nambour, Australia; Bankstown-Lidcombe Hospital, Sydney, Australia
| | | | - Michael A Kamm
- St Vincent's Hospital and University of Melbourne, Melbourne, Australia
| | | | | | | | | | | | | | | | | | | | - Watson Ng
- Liverpool Hospital, Sydney, Australia
| | - Enmoore Lin
- Centre for Digestive Diseases, Sydney, Australia
| | | | | | | | | | | |
Collapse
|
567
|
Affiliation(s)
- Liam Shaw
- From the UCL Genetics Institute, UCL
| | - Nigel Klein
- UCL Great Ormond Street Institute for Child Health, UCL, London, United Kingdom
| |
Collapse
|
568
|
Olsen I, Yamazaki K. Can oral bacteria affect the microbiome of the gut? J Oral Microbiol 2019; 11:1586422. [PMID: 30911359 PMCID: PMC6427756 DOI: 10.1080/20002297.2019.1586422] [Citation(s) in RCA: 212] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 02/04/2019] [Accepted: 02/19/2019] [Indexed: 12/13/2022] Open
Abstract
Oral bacteria spreading through the body have been associated with a number of systemic diseases. The gut is no exception. Studies in animals and man have indicated that oral bacteria can translocate to the gut and change its microbiota and possibly immune defense. The ectopic displacement of oral bacteria particularly occurs in severe systemic diseases, but also in patients with “chronic” periodontitis. Thus, Porphyromonas gingivalis, which creates dysbiosis in the subgingival microbiota and immune defense, may also cause dysregulation in the gut. A dysbiotic gut microbiota may cause diseases elsewhere in the body. The fact that “chronic” periodontitis may affect the gut microbiota could imply that consideration might in the future be given to a coordinated approach to the treatment of periodontitis and gastrointestinal disease. This area of investigation, which is in its infancy, may represent another pathway for oral bacteria to cause systemic diseases and deserves more research.
Collapse
Affiliation(s)
- Ingar Olsen
- Department of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - Kazuhisa Yamazaki
- Research Unit for Oral-Systemic Connection, Division of Oral Science for Health Promotion, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| |
Collapse
|
569
|
Rhoades N, Mendoza N, Jankeel A, Sureshchandra S, Alvarez AD, Doratt B, Heidari O, Hagan R, Brown B, Scheibel S, Marbley T, Taylor J, Messaoudi I. Altered Immunity and Microbial Dysbiosis in Aged Individuals With Long-Term Controlled HIV Infection. Front Immunol 2019; 10:463. [PMID: 30915086 PMCID: PMC6423162 DOI: 10.3389/fimmu.2019.00463] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 02/20/2019] [Indexed: 01/22/2023] Open
Abstract
The introduction of highly active antiretroviral therapy (HAART) resulted in a significant increase in life expectancy for HIV patients. Indeed, in 2015, 45% of the HIV+ individuals in the United States were ≥55 years of age. Despite improvements in diagnosis and treatment of HIV infection, geriatric HIV+ patients suffer from higher incidence of comorbidities compared to age-matched HIV- individuals. Both chronic inflammation and dysbiosis of the gut microbiome are believed to be major contributors to this phenomenon, however carefully controlled studies investigating the impact of long-term (>10 years) controlled HIV (LTC-HIV) infection are lacking. To address this question, we profiled circulating immune cells, immune mediators, and the gut microbiome from elderly (≥55 years old) LTC-HIV+ and HIV- gay men living in the Palm Springs area. LTC-HIV+ individuals had lower frequency of circulating monocytes and CD4+ T-cells, and increased frequency CD8+ T-cells. Moreover, levels of systemic INFγ and several growth factors were increased while levels of IL-2 and several chemokines were reduced. Upon stimulation, immune cells from LTC-HIV+ individuals produced higher levels of pro-inflammatory cytokines. Last but not least, the gut microbiome of LTC-HIV+ individuals was enriched in bacterial taxa typically found in the oral cavity suggestive of loss of compartmentalization, while levels of beneficial butyrate producing taxa were reduced. Additionally, prevalence of Prevotella negatively correlated with CD4+ T-cells numbers in LTC-HIV+ individuals. These results indicate that despite long-term adherence and undetectable viral loads, LTC-HIV infection results in significant shifts in immune cell frequencies and gut microbial communities.
Collapse
Affiliation(s)
- Nicholas Rhoades
- Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA, United States
| | - Norma Mendoza
- Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA, United States
| | - Allen Jankeel
- Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA, United States
| | - Suhas Sureshchandra
- Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA, United States
| | - Alexander D Alvarez
- Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA, United States
| | - Brianna Doratt
- Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA, United States
| | - Omeid Heidari
- School of Nursing, John Hopkins University, Baltimore, MD, United States
| | - Rod Hagan
- Stonewall Medical Center, Borrego Health, Cathedral City, CA, United States
| | - Brandon Brown
- School of Medicine, University of California, Riverside, Riverside, CA, United States
| | - Steven Scheibel
- Stonewall Medical Center, Borrego Health, Cathedral City, CA, United States
| | - Theodore Marbley
- Stonewall Medical Center, Borrego Health, Cathedral City, CA, United States
| | - Jeff Taylor
- HIV+ Aging-Palm Springs, Palm Springs, CA, United States
| | - Ilhem Messaoudi
- Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA, United States
| |
Collapse
|
570
|
|
571
|
Pandiyan P, Bhaskaran N, Zou M, Schneider E, Jayaraman S, Huehn J. Microbiome Dependent Regulation of T regs and Th17 Cells in Mucosa. Front Immunol 2019; 10:426. [PMID: 30906299 PMCID: PMC6419713 DOI: 10.3389/fimmu.2019.00426] [Citation(s) in RCA: 170] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Accepted: 02/18/2019] [Indexed: 12/19/2022] Open
Abstract
Mammals co-exist with resident microbial ecosystem that is composed of an incredible number and diversity of bacteria, viruses and fungi. Owing to direct contact between resident microbes and mucosal surfaces, both parties are in continuous and complex interactions resulting in important functional consequences. These interactions govern immune homeostasis, host response to infection, vaccination and cancer, as well as predisposition to metabolic, inflammatory and neurological disorders. Here, we discuss recent studies on direct and indirect effects of resident microbiota on regulatory T cells (Tregs) and Th17 cells at the cellular and molecular level. We review mechanisms by which commensal microbes influence mucosa in the context of bioactive molecules derived from resident bacteria, immune senescence, chronic inflammation and cancer. Lastly, we discuss potential therapeutic applications of microbiota alterations and microbial derivatives, for improving resilience of mucosal immunity and combating immunopathology.
Collapse
Affiliation(s)
- Pushpa Pandiyan
- Department of Biological Sciences, School of Dental Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Natarajan Bhaskaran
- Department of Biological Sciences, School of Dental Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Mangge Zou
- Experimental Immunology, Helmholtz Centre for Infection Research, Hamburg, Germany.,Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Elizabeth Schneider
- Department of Biological Sciences, School of Dental Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Sangeetha Jayaraman
- Department of Biological Sciences, School of Dental Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Jochen Huehn
- Experimental Immunology, Helmholtz Centre for Infection Research, Hamburg, Germany.,Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| |
Collapse
|
572
|
Heath JE, Scholz GM, Veith PD, Reynolds EC. IL-36γ regulates mediators of tissue homeostasis in epithelial cells. Cytokine 2019; 119:24-31. [PMID: 30856602 DOI: 10.1016/j.cyto.2019.02.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 01/31/2019] [Accepted: 02/12/2019] [Indexed: 02/06/2023]
Abstract
IL-36 cytokines are critical regulators of mucosal inflammation and homeostasis. IL-36γ regulates the expression of inflammatory cytokines and antimicrobial proteins by gingival epithelial cells (e.g. TIGK cells). Here, we show that IL-36γ also regulates the expression of matrix metalloproteinase 9 (MMP9) and neutrophil gelatinase-associated lipocalin (NGAL), important mediators of antimicrobial immunity and tissue homeostasis in mucosal epithelia. MMP9 and NGAL were not similarly induced by IL-17 or IL-22, thus indicating the importance of IL-36γ in the regulation of MMP9 and NGAL. Mechanistically, MMP9 and NGAL expression was demonstrated to be induced in an IRAK1- and NF-κB-dependent manner. Furthermore, signaling by p38 MAP kinase may enable their expression to be independently regulated by IL-36γ. The stronger IL-36γ-inducible expression of MMP9 and NGAL in terminally differentiating TIGK cells suggests that control of their expression is associated with the maturation of the gingival epithelium. Although MMP9 and NGAL expression in epithelial cells can also be induced by bacteria, their expression in TIGK cells was not induced by the periodontal pathogen Porphyromonas gingivalis, most likely due to antagonism by the gingipain proteinase virulence factors. This study advances our understanding of how IL-36γ may promote oral mucosal immunity and tissue homeostasis, and how this may be dysregulated by bacterial pathogens.
Collapse
Affiliation(s)
- Jacqueline E Heath
- Oral Health Cooperative Research Centre, Melbourne Dental School, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Victoria, Australia
| | - Glen M Scholz
- Oral Health Cooperative Research Centre, Melbourne Dental School, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Victoria, Australia.
| | - Paul D Veith
- Oral Health Cooperative Research Centre, Melbourne Dental School, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Victoria, Australia
| | - Eric C Reynolds
- Oral Health Cooperative Research Centre, Melbourne Dental School, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Victoria, Australia.
| |
Collapse
|
573
|
Ma H, Li Y, Liang H, Chen S, Pan S, Chang L, Li S, Zhang Y, Liu X, Xu Y, Shao Y, Yang Y, Guo J. Sleep deprivation and a non–24‐h working schedule lead to extensive alterations in physiology and behavior. FASEB J 2019; 33:6969-6979. [DOI: 10.1096/fj.201802727r] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Huan Ma
- Ministry of Education (MOE) Key Laboratory of Gene Function and RegulationSchool of Life SciencesState Key Laboratory of BiocontrolSun Yat-sen UniversityGuangzhouChina
| | - Yunzhen Li
- Ministry of Education (MOE) Key Laboratory of Gene Function and RegulationSchool of Life SciencesState Key Laboratory of BiocontrolSun Yat-sen UniversityGuangzhouChina
| | - Haojian Liang
- Ministry of Education (MOE) Key Laboratory of Gene Function and RegulationSchool of Life SciencesState Key Laboratory of BiocontrolSun Yat-sen UniversityGuangzhouChina
| | - Shijunyin Chen
- Ministry of Education (MOE) Key Laboratory of Gene Function and RegulationSchool of Life SciencesState Key Laboratory of BiocontrolSun Yat-sen UniversityGuangzhouChina
| | - Siyu Pan
- Ministry of Education (MOE) Key Laboratory of Gene Function and RegulationSchool of Life SciencesState Key Laboratory of BiocontrolSun Yat-sen UniversityGuangzhouChina
| | - Lulin Chang
- Ministry of Education (MOE) Key Laboratory of Gene Function and RegulationSchool of Life SciencesState Key Laboratory of BiocontrolSun Yat-sen UniversityGuangzhouChina
- Guangdong Jiangmen Chinese Traditional Medicine CollegeJiangmenChina
| | - Silin Li
- Ministry of Education (MOE) Key Laboratory of Gene Function and RegulationSchool of Life SciencesState Key Laboratory of BiocontrolSun Yat-sen UniversityGuangzhouChina
| | - Ying Zhang
- Eighth Medical CenterGeneral Hospital of Chinese People's Liberation ArmyBeijingChina
| | - Xinyan Liu
- Reproductive Medicine CenterFirst Affiliated Hospital of Sun Yat-sen UniversitySun Yat-sen UniversityGuangzhouChina
| | - Yanwen Xu
- Reproductive Medicine CenterFirst Affiliated Hospital of Sun Yat-sen UniversitySun Yat-sen UniversityGuangzhouChina
| | - Yongcong Shao
- Eighth Medical CenterGeneral Hospital of Chinese People's Liberation ArmyBeijingChina
| | - Yebing Yang
- Sixth Medical CenterGeneral Hospital of Chinese People's Liberation ArmyBeijingChina
| | - Jinhu Guo
- Ministry of Education (MOE) Key Laboratory of Gene Function and RegulationSchool of Life SciencesState Key Laboratory of BiocontrolSun Yat-sen UniversityGuangzhouChina
| |
Collapse
|
574
|
Pope JL, Yang Y, Newsome RC, Sun W, Sun X, Ukhanova M, Neu J, Issa JP, Mai V, Jobin C. Microbial Colonization Coordinates the Pathogenesis of a Klebsiella pneumoniae Infant Isolate. Sci Rep 2019; 9:3380. [PMID: 30833613 PMCID: PMC6399262 DOI: 10.1038/s41598-019-39887-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 01/28/2019] [Indexed: 12/22/2022] Open
Abstract
Enterobacteriaceae are among the first colonizers of neonate intestine. Members of this family, such as Escherichia and Klebsiella, are considered pathobionts and as such are capable of inducing local and systemic disease under specific colonization circumstances. Interplay between developing microbiota and pathogenic function of pathobionts are poorly understood. In this study, we investigate the functional interaction between various colonization patterns on an early colonizer, K. pneumoniae. K. pneumoniae 51-5 was isolated from stool of a healthy, premature infant, and found to contain the genotoxin island pks associated with development of colorectal cancer. Using intestinal epithelial cells, macrophages, and primary splenocytes, we demonstrate K. pneumoniae 51-5 upregulates expression of proinflammatory genes in vitro. Gnotobiotic experiments in Il10-/- mice demonstrate the neonate isolate induces intestinal inflammation in vivo, with increased expression of proinflammatory genes. Regulation of microbiota assembly revealed K. pneumoniae 51-5 accelerates onset of inflammation in Il10-/- mice, most significantly when microbiota is naturally acquired. Furthermore, K. pneumoniae 51-5 induces DNA damage and cell cycle arrest. Interestingly, K. pneumoniae 51-5 induced tumors in ApcMin/+; Il10-/- mice was not significantly affected by absence of colibactin activating enzyme, ClbP. These findings demonstrate pathogenicity of infant K. pneumoniae isolate is sensitive to microbial colonization status.
Collapse
Affiliation(s)
- Jillian L Pope
- Department of Medicine, University of Florida, Gainesville, Florida, USA
| | - Ye Yang
- Department of Medicine, University of Florida, Gainesville, Florida, USA
| | - Rachel C Newsome
- Department of Medicine, University of Florida, Gainesville, Florida, USA
| | - Wei Sun
- Department of Infectious Diseases and Pathology, College of Veterinary Medicine, University of Florida, Gainesville, Florida, USA
- Department of Immunology & Microbial Disease, Albany Medical College, Albany, New York, USA
| | - Xiaolun Sun
- Department of Medicine, University of Florida, Gainesville, Florida, USA
- Department of Poultry Science, University of Arkanasas, Fayetteville, Arkansas, USA
| | - Maria Ukhanova
- Department of Epidemiology, University of Florida, Gainesville, Florida, USA
| | - Josef Neu
- Department of Medicine, University of Florida, Gainesville, Florida, USA
| | - Jean-Pierre Issa
- Fels Institute for Cancer Research and Molecular Biology, Temple University, Philadelphia, PA, 19140, USA
| | - Volker Mai
- Department of Epidemiology, University of Florida, Gainesville, Florida, USA
| | - Christian Jobin
- Department of Medicine, University of Florida, Gainesville, Florida, USA.
- Department of Anatomy and Cell Biology, University of Florida, Gainesville, Florida, USA.
| |
Collapse
|
575
|
Lu M, Xuan S, Wang Z. Oral microbiota: A new view of body health. FOOD SCIENCE AND HUMAN WELLNESS 2019. [DOI: 10.1016/j.fshw.2018.12.001] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
576
|
Baranzini SE. Insights into microbiome research 5: Mapping is first but function must come next. Mult Scler 2019; 25:193-195. [PMID: 30799701 DOI: 10.1177/1352458518811203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Several efforts identifying differential bacterial prevalence in the gut of multiple sclerosis (MS) patients have been reported and many more are underway. While these are critical first steps in determining the involvement of gut microbes in MS pathogenesis, functional assays (both in vitro and in vivo) are needed to explore the mechanisms underlying the observed changes and ultimately implementing interventional strategies to counter severity, progression or even reduce the chance that it develops in the first place.
Collapse
Affiliation(s)
- Sergio E Baranzini
- Sergio E Baranzini Weill Institute of Neuroscience, Department of Neurology. University of California San Francisco, USA
| |
Collapse
|
577
|
Li W, Xu X, Wen H, Wang Z, Ding C, Liu X, Gao Y, Su H, Zhang J, Han Y, Xia Y, Wang X, Gu H, Yao X. Inverse Association Between the Skin and Oral Microbiota in Atopic Dermatitis. J Invest Dermatol 2019; 139:1779-1787.e12. [PMID: 30802424 DOI: 10.1016/j.jid.2019.02.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 02/01/2019] [Accepted: 02/04/2019] [Indexed: 12/21/2022]
Abstract
Previous studies have shown independently that the skin and gut microbiota are closely associated with atopic dermatitis (AD); however, the microbiota across different habitats of AD patients as an integrated community has not been characterized. In the present study, we comparatively analyzed the structure and function of the microbial communities in the skin, oral cavity, and gut of 172 AD patients and 120 healthy controls through 16S ribosomal RNA gene amplicon sequencing. The skin and oral cavity, but not the gut, of AD patients demonstrated differential reduction in the microbial diversity, and these were distinctly correlated with disease severity. Different degrees of shifts in the community structure were found among different habitats, and the lineage distance between the skin and oral microbiota of AD patients was closer than that observed in the controls. The different habitats of AD patients exhibited site-specific alterations at the genus level, and many oral-specific microbes of AD showed opposing directions of enrichment in the skin and oral cavity. Most interestingly, an inverse association in the functional pathways was found between the skin and oral microbiota of AD patients. Additionally, the alterations of the microbiota in different body sites of AD patients were differentially affected by age.
Collapse
Affiliation(s)
- Wei Li
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, People's Republic of China; Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Xiaoqiang Xu
- Aimigene Institute, Shenzhen, People's Republic of China
| | - He Wen
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, People's Republic of China
| | - Zhifeng Wang
- Aimigene Institute, Shenzhen, People's Republic of China
| | - Chao Ding
- Department of General Surgery, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, People's Republic of China; Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, People's Republic of China
| | - Xiaochun Liu
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, People's Republic of China
| | - Yingxia Gao
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, People's Republic of China
| | - Huichun Su
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, People's Republic of China
| | - Jingxi Zhang
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, People's Republic of China
| | - Yue Han
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, People's Republic of China
| | - Yan Xia
- Aimigene Institute, Shenzhen, People's Republic of China
| | - Xiaokai Wang
- Aimigene Institute, Shenzhen, People's Republic of China
| | - Heng Gu
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, People's Republic of China.
| | - Xu Yao
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, People's Republic of China.
| |
Collapse
|
578
|
Schmidt TSB, Hayward MR, Coelho LP, Li SS, Costea PI, Voigt AY, Wirbel J, Maistrenko OM, Alves RJC, Bergsten E, de Beaufort C, Sobhani I, Heintz-Buschart A, Sunagawa S, Zeller G, Wilmes P, Bork P. Extensive transmission of microbes along the gastrointestinal tract. eLife 2019; 8:e42693. [PMID: 30747106 PMCID: PMC6424576 DOI: 10.7554/elife.42693] [Citation(s) in RCA: 357] [Impact Index Per Article: 59.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 02/03/2019] [Indexed: 12/18/2022] Open
Abstract
The gastrointestinal tract is abundantly colonized by microbes, yet the translocation of oral species to the intestine is considered a rare aberrant event, and a hallmark of disease. By studying salivary and fecal microbial strain populations of 310 species in 470 individuals from five countries, we found that transmission to, and subsequent colonization of, the large intestine by oral microbes is common and extensive among healthy individuals. We found evidence for a vast majority of oral species to be transferable, with increased levels of transmission in colorectal cancer and rheumatoid arthritis patients and, more generally, for species described as opportunistic pathogens. This establishes the oral cavity as an endogenous reservoir for gut microbial strains, and oral-fecal transmission as an important process that shapes the gastrointestinal microbiome in health and disease.
Collapse
Affiliation(s)
- Thomas SB Schmidt
- Structural and Computational Biology UnitEuropean Molecular Biology LaboratoryHeidelbergGermany
| | - Matthew R Hayward
- Structural and Computational Biology UnitEuropean Molecular Biology LaboratoryHeidelbergGermany
| | - Luis P Coelho
- Structural and Computational Biology UnitEuropean Molecular Biology LaboratoryHeidelbergGermany
| | - Simone S Li
- Structural and Computational Biology UnitEuropean Molecular Biology LaboratoryHeidelbergGermany
| | - Paul I Costea
- Structural and Computational Biology UnitEuropean Molecular Biology LaboratoryHeidelbergGermany
| | - Anita Y Voigt
- Structural and Computational Biology UnitEuropean Molecular Biology LaboratoryHeidelbergGermany
| | - Jakob Wirbel
- Structural and Computational Biology UnitEuropean Molecular Biology LaboratoryHeidelbergGermany
| | - Oleksandr M Maistrenko
- Structural and Computational Biology UnitEuropean Molecular Biology LaboratoryHeidelbergGermany
| | - Renato JC Alves
- Structural and Computational Biology UnitEuropean Molecular Biology LaboratoryHeidelbergGermany
- Joint PhD programmeEuropean Molecular Biology Laboratory and Faculty of Biosciences, Heidelberg UniversityHeidelbergGermany
| | - Emma Bergsten
- Department of Gastroenterology and EA7375 -EC2M3APHP and UPEC Université Paris-Est CréteilCréteilFrance
| | - Carine de Beaufort
- Luxembourg Centre for Systems BiomedicineLuxembourgLuxembourg
- Clinique PédiatriqueCentre Hospitalier de LuxembourgLuxembourgLuxembourg
| | - Iradj Sobhani
- Department of Gastroenterology and EA7375 -EC2M3APHP and UPEC Université Paris-Est CréteilCréteilFrance
| | | | - Shinichi Sunagawa
- Structural and Computational Biology UnitEuropean Molecular Biology LaboratoryHeidelbergGermany
| | - Georg Zeller
- Structural and Computational Biology UnitEuropean Molecular Biology LaboratoryHeidelbergGermany
| | - Paul Wilmes
- Luxembourg Centre for Systems BiomedicineLuxembourgLuxembourg
| | - Peer Bork
- Structural and Computational Biology UnitEuropean Molecular Biology LaboratoryHeidelbergGermany
- Max Delbrück Centre for Molecular MedicineBerlinGermany
- Molecular Medicine Partnership Unit (MMPU)European Molecular Biology Laboratory and University Hospital HeidelbergHeidelbergGermany
- Department of Bioinformatics, BiocenterUniversity of WürzburgWürzburgGermany
| |
Collapse
|
579
|
van der Meulen TA, Harmsen HJ, Vila AV, Kurilshikov A, Liefers SC, Zhernakova A, Fu J, Wijmenga C, Weersma RK, de Leeuw K, Bootsma H, Spijkervet FK, Vissink A, Kroese FG. Shared gut, but distinct oral microbiota composition in primary Sjögren's syndrome and systemic lupus erythematosus. J Autoimmun 2019; 97:77-87. [DOI: 10.1016/j.jaut.2018.10.009] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 10/12/2018] [Accepted: 10/16/2018] [Indexed: 12/11/2022]
|
580
|
The microbiome and immunodeficiencies: Lessons from rare diseases. J Autoimmun 2019; 98:132-148. [PMID: 30704941 DOI: 10.1016/j.jaut.2019.01.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Revised: 01/21/2019] [Accepted: 01/23/2019] [Indexed: 12/20/2022]
Abstract
Primary immunodeficiencies (PIDs) are inherited disorders of the immune system, associated with a considerable increase in susceptibility to infections. PIDs can also predispose to malignancy, inflammation and autoimmunity. There is increasing awareness that some aspects of the immune dysregulation in PIDs may be linked to intestinal microbiota. Indeed, the gut microbiota and its metabolites have been shown to influence immune functions and immune homeostasis both locally and systemically. Recent studies have indicated that genetic defects causing PIDs lead to perturbations in the conventional mechanisms underlying homeostasis in the gut, resulting in poor immune surveillance at the intestinal barrier, which associates with altered intestinal permeability and bacterial translocation. Consistently, a substantial proportion of PID patients presents with clinically challenging IBD-like pathology. Here, we describe the current body of literature reporting on dysbiosis of the gut microbiota in different PIDs and how this can be either the result or cause of immune dysregulation. Further, we report how infections in PIDs enhance pathobionts colonization and speculate how, in turn, pathobionts may be responsible for increased disease susceptibility and secondary infections in these patients. The potential relationship between the microbial composition in the intestine and other sites, such as the oral cavity and skin, is also highlighted. Finally, we provide evidence, in preclinical models of PIDs, for the efficacy of microbiota manipulation to ameliorate disease complications, and suggest that the potential use of dietary intervention to correct dysbiotic flora in PID patients may hold promise.
Collapse
|
581
|
Khatun A, Sakurai M, Sakai Y, Tachibana M, Ohara N, Morimoto M. Mycobacterial infection induces eosinophilia and production of α-defensin by eosinophils in mice. J Vet Med Sci 2019; 81:138-142. [PMID: 30473572 PMCID: PMC6361637 DOI: 10.1292/jvms.18-0619] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
It has been well known in humans that eosinophil infiltration into the site of inflammation and eosinophilia occur in mycobacterial infections. However, the role of eosinophils against the
mycobacterium is unclear. We showed in previous study that in situ mouse eosinophils infiltrated into tissues produce α-defensin, an anti-bacterial peptide. We investigated
in this study whether eosinophils reacting to mycobacteria produce α-defensin in mice and whether it can be used as a model. We showed that mycobacterial infection induced blood eosinophilia
and infiltration of α-defensin producing eosinophils that to surround mycobacteria at the site of infection. These findings were usually seen during human mycobacterial infection. We
established a good model to study host defense mechanism against mycobacteria through α-defensin via eosinophils.
Collapse
Affiliation(s)
- Afia Khatun
- Laboratory of Veterinary Pathology, Joint Faculty of Veterinary Medicine, Yamaguchi University, 1677-1, Yoshida, Yamaguchi 753-8515, Japan
| | - Masashi Sakurai
- Laboratory of Veterinary Pathology, Joint Faculty of Veterinary Medicine, Yamaguchi University, 1677-1, Yoshida, Yamaguchi 753-8515, Japan
| | - Yusuke Sakai
- Laboratory of Veterinary Pathology, Joint Faculty of Veterinary Medicine, Yamaguchi University, 1677-1, Yoshida, Yamaguchi 753-8515, Japan
| | - Masato Tachibana
- Department of Oral Microbiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata, Kita-ku, Okayama 700-8558, Japan
| | - Naoya Ohara
- Department of Oral Microbiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata, Kita-ku, Okayama 700-8558, Japan
| | - Masahiro Morimoto
- Laboratory of Veterinary Pathology, Joint Faculty of Veterinary Medicine, Yamaguchi University, 1677-1, Yoshida, Yamaguchi 753-8515, Japan
| |
Collapse
|
582
|
|
583
|
A defined commensal consortium elicits CD8 T cells and anti-cancer immunity. Nature 2019; 565:600-605. [PMID: 30675064 DOI: 10.1038/s41586-019-0878-z] [Citation(s) in RCA: 779] [Impact Index Per Article: 129.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 12/05/2018] [Indexed: 02/07/2023]
Abstract
There is a growing appreciation for the importance of the gut microbiota as a therapeutic target in various diseases. However, there are only a handful of known commensal strains that can potentially be used to manipulate host physiological functions. Here we isolate a consortium of 11 bacterial strains from healthy human donor faeces that is capable of robustly inducing interferon-γ-producing CD8 T cells in the intestine. These 11 strains act together to mediate the induction without causing inflammation in a manner that is dependent on CD103+ dendritic cells and major histocompatibility (MHC) class Ia molecules. Colonization of mice with the 11-strain mixture enhances both host resistance against Listeria monocytogenes infection and the therapeutic efficacy of immune checkpoint inhibitors in syngeneic tumour models. The 11 strains primarily represent rare, low-abundance components of the human microbiome, and thus have great potential as broadly effective biotherapeutics.
Collapse
|
584
|
Britton GJ, Contijoch EJ, Mogno I, Vennaro OH, Llewellyn SR, Ng R, Li Z, Mortha A, Merad M, Das A, Gevers D, McGovern DPB, Singh N, Braun J, Jacobs JP, Clemente JC, Grinspan A, Sands BE, Colombel JF, Dubinsky MC, Faith JJ. Microbiotas from Humans with Inflammatory Bowel Disease Alter the Balance of Gut Th17 and RORγt + Regulatory T Cells and Exacerbate Colitis in Mice. Immunity 2019; 50:212-224.e4. [PMID: 30650377 PMCID: PMC6512335 DOI: 10.1016/j.immuni.2018.12.015] [Citation(s) in RCA: 368] [Impact Index Per Article: 61.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 10/03/2018] [Accepted: 12/13/2018] [Indexed: 12/13/2022]
Abstract
Microbiota are thought to influence the development and progression of inflammatory bowel disease (IBD), but determining generalizable effects of microbiota on IBD etiology requires larger-scale functional analyses. We colonized germ-free mice with intestinal microbiotas from 30 healthy and IBD donors and determined the homeostatic intestinal T cell response to each microbiota. Compared to microbiotas from healthy donors, transfer of IBD microbiotas into germ-free mice increased numbers of intestinal Th17 cells and Th2 cells and decreased numbers of RORγt+ Treg cells. Colonization with IBD microbiotas exacerbated disease in a model where colitis is induced upon transfer of naive T cells into Rag1-/- mice. The proportions of Th17 and RORγt+ Treg cells induced by each microbiota were predictive of human disease status and accounted for disease severity in the Rag1-/- colitis model. Thus, an impact on intestinal Th17 and RORγt+ Treg cell compartments emerges as a unifying feature of IBD microbiotas, suggesting a general mechanism for microbial contribution to IBD pathogenesis.
Collapse
Affiliation(s)
- Graham J Britton
- Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Eduardo J Contijoch
- Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ilaria Mogno
- Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Olivia H Vennaro
- Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sean R Llewellyn
- Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ruby Ng
- Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Zhihua Li
- Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Arthur Mortha
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Miriam Merad
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anuk Das
- Janssen Human Microbiome Institute, Janssen Research and Development, LLC, Spring House, PA, USA
| | - Dirk Gevers
- Janssen Human Microbiome Institute, Janssen Research and Development, LLC, Spring House, PA, USA
| | - Dermot P B McGovern
- Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Namita Singh
- Pediatric Gastroenterology and Inflammatory Bowel Disease, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jonathan Braun
- UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - Jonathan P Jacobs
- Division of Digestive Diseases, Department of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Jose C Clemente
- Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ari Grinspan
- Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bruce E Sands
- Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jean-Frederic Colombel
- Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Marla C Dubinsky
- Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Pediatric Gastroenterology and Hepatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jeremiah J Faith
- Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
585
|
Nakamoto N, Sasaki N, Aoki R, Miyamoto K, Suda W, Teratani T, Suzuki T, Koda Y, Chu PS, Taniki N, Yamaguchi A, Kanamori M, Kamada N, Hattori M, Ashida H, Sakamoto M, Atarashi K, Narushima S, Yoshimura A, Honda K, Sato T, Kanai T. Gut pathobionts underlie intestinal barrier dysfunction and liver T helper 17 cell immune response in primary sclerosing cholangitis. Nat Microbiol 2019; 4:492-503. [PMID: 30643240 DOI: 10.1038/s41564-018-0333-1] [Citation(s) in RCA: 301] [Impact Index Per Article: 50.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 11/29/2018] [Indexed: 02/07/2023]
Abstract
Primary sclerosing cholangitis (PSC) is a chronic inflammatory liver disease and its frequent complication with ulcerative colitis highlights the pathogenic role of epithelial barrier dysfunction. Intestinal barrier dysfunction has been implicated in the pathogenesis of PSC, yet its underlying mechanism remains unknown. Here, we identify Klebsiella pneumonia in the microbiota of patients with PSC and demonstrate that K. pneumoniae disrupts the epithelial barrier to initiate bacterial translocation and liver inflammatory responses. Gnotobiotic mice inoculated with PSC-derived microbiota exhibited T helper 17 (TH17) cell responses in the liver and increased susceptibility to hepatobiliary injuries. Bacterial culture of mesenteric lymph nodes in these mice isolated K. pneumoniae, Proteus mirabilis and Enterococcus gallinarum, which were prevalently detected in patients with PSC. A bacterial-organoid co-culture system visualized the epithelial-damaging effect of PSC-derived K. pneumoniae that was associated with bacterial translocation and susceptibility to TH17-mediated hepatobiliary injuries. We also show that antibiotic treatment ameliorated the TH17 immune response induced by PSC-derived microbiota. These results highlight the role of pathobionts in intestinal barrier dysfunction and liver inflammation, providing insights into therapeutic strategies for PSC.
Collapse
Affiliation(s)
- Nobuhiro Nakamoto
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Shinanomachi, Tokyo, Japan
| | - Nobuo Sasaki
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Shinanomachi, Tokyo, Japan.,AMED-CREST, Japan Agency for Medical Research and Development, Tokyo, Japan
| | - Ryo Aoki
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Shinanomachi, Tokyo, Japan.,Institute of Health Science, Ezaki Glico Co., Ltd, Osaka, Japan
| | - Kentaro Miyamoto
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Shinanomachi, Tokyo, Japan.,Miyarisan Pharmaceutical Co., Ltd, Tokyo, Japan
| | - Wataru Suda
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinanomachi, Tokyo, Japan.,Laboratory of Metagenomics, Department of Computational Biology and Medical Sciences, The University of Tokyo, Chiba, Japan
| | - Toshiaki Teratani
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Shinanomachi, Tokyo, Japan
| | - Takahiro Suzuki
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Shinanomachi, Tokyo, Japan.,Miyarisan Pharmaceutical Co., Ltd, Tokyo, Japan
| | - Yuzo Koda
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Shinanomachi, Tokyo, Japan.,Research Unit/Immunology & Inflammation, Sohyaku Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, Kanagawa, Japan
| | - Po-Sung Chu
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Shinanomachi, Tokyo, Japan
| | - Nobuhito Taniki
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Shinanomachi, Tokyo, Japan
| | - Akihiro Yamaguchi
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Shinanomachi, Tokyo, Japan
| | - Mitsuhiro Kanamori
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinanomachi, Tokyo, Japan
| | - Nobuhiko Kamada
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Masahira Hattori
- Laboratory of Metagenomics, Department of Computational Biology and Medical Sciences, The University of Tokyo, Chiba, Japan.,Cooperative Major in Advanced Health Science, Graduate School of Advanced Science and Engineering, Faculty of Science and Engineering, Waseda University, Tokyo, Japan
| | - Hiroshi Ashida
- Department of Bacterial Infection and Host Response, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Michiie Sakamoto
- Department of Pathology, Keio University School of Medicine, Shinanomachi, Tokyo, Japan
| | - Koji Atarashi
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinanomachi, Tokyo, Japan.,RIKEN Center for Integrative Medical Sciences, Laboratory for Gut Homeostasis, Kanagawa, Japan
| | - Seiko Narushima
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinanomachi, Tokyo, Japan.,RIKEN Center for Integrative Medical Sciences, Laboratory for Gut Homeostasis, Kanagawa, Japan
| | - Akihiko Yoshimura
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinanomachi, Tokyo, Japan
| | - Kenya Honda
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinanomachi, Tokyo, Japan.,RIKEN Center for Integrative Medical Sciences, Laboratory for Gut Homeostasis, Kanagawa, Japan
| | - Toshiro Sato
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Shinanomachi, Tokyo, Japan.
| | - Takanori Kanai
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Shinanomachi, Tokyo, Japan. .,AMED-CREST, Japan Agency for Medical Research and Development, Tokyo, Japan.
| |
Collapse
|
586
|
Dinakaran V, Mandape SN, Shuba K, Pratap S, Sakhare SS, Tabatabai MA, Smoot DT, Farmer-Dixon CM, Kesavalu LN, Adunyah SE, Southerland JH, Gangula PR. Identification of Specific Oral and Gut Pathogens in Full Thickness Colon of Colitis Patients: Implications for Colon Motility. Front Microbiol 2019; 9:3220. [PMID: 30666239 PMCID: PMC6330997 DOI: 10.3389/fmicb.2018.03220] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 12/11/2018] [Indexed: 12/31/2022] Open
Abstract
Impaired colon motility is one of the leading problems associated with inflammatory bowel disease (IBD). An expanding body of evidence supports the role of microbiome in normal gut function and in progression of IBD. The objective of this work is to determine whether diseased full thickness colon specimens, including the neuromuscular region (critical for colon motility function), contain specific oral and gut pathogens. In addition, we compared the differences in colon microbiome between Caucasians (CA) and African Americans (AA). Thirty-nine human full thickness colon (diseased colon and adjacent healthy colon) specimens were collected from Crohn's Colitis (CC) or Ulcerative Colitis (UC) patients while they underwent elective colon surgeries. We isolated and analyzed bacterial ribosomal RNA (rRNA) from colon specimens by amplicon sequencing of the 16S rRNA gene region. The microbiome proportions were quantified into Operational Taxonomic Units (OTUs) by analysis with Quantitative Insights Into Microbial ecology (QIIME) platform. Two hundred twenty-eight different bacterial species were identified by QIIME analysis. However, we could only decipher the species name of fifty-three bacteria. Our results show that proportion of non-detrimental bacteria in CC or UC colon samples were altered compared to adjacent healthy colon specimens. We further show, for the first time in full thickness colon specimens, that microbiome of CC and UC diseased specimens is dominated by putative oral pathogens belonging to the Phyla Firmicutes (Streptococcus, Staphylococcus, Peptostreptococcus), and Fusobacteria (Fusobacterium). In addition, we have identified patterns of differences in microbiome levels between CA and AA specimens with potential implications for health disparities research. Overall, our results suggest a significant association between oral and gut microbes in the modulation of colon motility in colitis patients.
Collapse
Affiliation(s)
- Vasudevan Dinakaran
- Department of ODS & Research, School of Dentistry, Meharry Medical College, Nashville, TN, United States
| | - Sammed N Mandape
- Bioinformatics Core, School of Graduate Studies/Research & School of Medicine, Meharry Medical College, Nashville, TN, United States
| | - Kristina Shuba
- Department of ODS & Research, School of Dentistry, Meharry Medical College, Nashville, TN, United States
| | - Siddharth Pratap
- Bioinformatics Core, School of Graduate Studies/Research & School of Medicine, Meharry Medical College, Nashville, TN, United States
| | - Shruti S Sakhare
- Bioinformatics Core, School of Graduate Studies/Research & School of Medicine, Meharry Medical College, Nashville, TN, United States
| | - Mohammad Ali Tabatabai
- Department of Public Health, School of Graduate Studies & Research, Meharry Medical College, Nashville, TN, United States
| | - Duane T Smoot
- Department of Internal Medicine, Division of Gastroenterology & Hepatology, Meharry Medical College, Nashville, TN, United States
| | - Cherae M Farmer-Dixon
- Department of ODS & Research, School of Dentistry, Meharry Medical College, Nashville, TN, United States
| | - Lakshmyya N Kesavalu
- Department of Periodontology, College of Dentistry, University of Florida, Gainesville, FL, United States
| | - Samuel Evans Adunyah
- Department of Biochemistry, Cancer Biology, Neuroscience & Pharmacology, Meharry Medical College, Nashville, TN, United States
| | - Janet Hayes Southerland
- Department of Nutrition Metabolism & Oral Surgery, University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Pandu R Gangula
- Department of ODS & Research, School of Dentistry, Meharry Medical College, Nashville, TN, United States
| |
Collapse
|
587
|
CHEN NAN, JIN TONGTONG, LIU WENNING, ZHU DONGQING, CHEN YINGYING, SHEN YUELIANG, LING ZONGXIN, WANG HONGJIE, ZHANG LIPING. Gastric Microbiota Alteration in Klebsiella pneumoniae-Caused Liver Abscesses Mice. Pol J Microbiol 2019; 68:247-254. [PMID: 31250595 PMCID: PMC7256812 DOI: 10.33073/pjm-2019-026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 03/14/2019] [Accepted: 03/24/2019] [Indexed: 12/30/2022] Open
Abstract
Gastric microbiota provides a biological barrier against the invasion of foreign pathogens from the oral cavity, playing a vital role in maintaining gastrointestinal health. Klebsiella spp. of oral origin causes various infections not only in gastrointestinal tract but also in other organs, with Klebsiella pneumoniae serotype K1 resulting in a liver abscess (KLA) through oral inoculation in mice. However, the relationship between gastric microbiota and the extra-gastrointestinal KLA infection is not clear. In our study, a 454 pyrosequencing analysis of the bacterial 16S rRNA gene shows that the composition of gastric mucosal microbiota in mice with or without KLA infection varies greatly after oral inoculation with K. pneumoniae serotype K1 isolate. Interestingly, only several bacteria taxa show a significant change in gastric mucosal microbiota of KLA mice, including the decreased abundance of Bacteroides, Alisptipes and increased abundance of Streptococcus. It is worth noting that the abundance of Klebsiella exhibits an obvious increase in KLA mice, which might be closely related to KLA infection. At the same time, the endogenous antibiotics, defensins, involved in the regulation of the bacterial microbiota also show an increase in stomach and intestine. All these findings indicate that liver abscess caused by K. pneumoniae oral inoculation has a close relationship with gastric microbiota, which might provide important information for future clinical treatment. Gastric microbiota provides a biological barrier against the invasion of foreign pathogens from the oral cavity, playing a vital role in maintaining gastrointestinal health. Klebsiella spp. of oral origin causes various infections not only in gastrointestinal tract but also in other organs, with Klebsiella pneumoniae serotype K1 resulting in a liver abscess (KLA) through oral inoculation in mice. However, the relationship between gastric microbiota and the extra-gastrointestinal KLA infection is not clear. In our study, a 454 pyrosequencing analysis of the bacterial 16S rRNA gene shows that the composition of gastric mucosal microbiota in mice with or without KLA infection varies greatly after oral inoculation with K. pneumoniae serotype K1 isolate. Interestingly, only several bacteria taxa show a significant change in gastric mucosal microbiota of KLA mice, including the decreased abundance of Bacteroides, Alisptipes and increased abundance of Streptococcus. It is worth noting that the abundance of Klebsiella exhibits an obvious increase in KLA mice, which might be closely related to KLA infection. At the same time, the endogenous antibiotics, defensins, involved in the regulation of the bacterial microbiota also show an increase in stomach and intestine. All these findings indicate that liver abscess caused by K. pneumoniae oral inoculation has a close relationship with gastric microbiota, which might provide important information for future clinical treatment.
Collapse
Affiliation(s)
- NAN CHEN
- Central Laboratory, Affiliated Hospital of Hebei University and Baoding, China
- Department of Medical Microbiology, Medicine College, Hebei University, Baoding, China
| | - TONG-TONG JIN
- Central Laboratory, Affiliated Hospital of Hebei University and Baoding, China
- Department of Medical Microbiology, Medicine College, Hebei University, Baoding, China
| | - WEN-NING LIU
- Central Laboratory, Affiliated Hospital of Hebei University and Baoding, China
- Department of Medical Microbiology, Medicine College, Hebei University, Baoding, China
| | - DONG-QING ZHU
- Central Laboratory, Affiliated Hospital of Hebei University and Baoding, China
- Department of Medical Microbiology, Medicine College, Hebei University, Baoding, China
| | - YING-YING CHEN
- Department of Pharmacology, Zhejiang Medical College, Zhejiang University, Hangzhou, China
| | - YUE-LIANG SHEN
- Department of Pharmacology, Zhejiang Medical College, Zhejiang University, Hangzhou, China
| | - ZONG-XIN LING
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - HONG-JIE WANG
- Central Laboratory, Affiliated Hospital of Hebei University and Baoding, China
- Department of Medical Microbiology, Medicine College, Hebei University, Baoding, China
| | - LI-PING ZHANG
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Hebei University, Baoding, China
- College of Life Sciences, Hebei University, Baoding, China
| |
Collapse
|
588
|
Horisberger A, La Rosa S, Zurcher JP, Zimmermann S, Spertini F, Coukos G, Obeid M. A severe case of refractory esophageal stenosis induced by nivolumab and responding to tocilizumab therapy. J Immunother Cancer 2018; 6:156. [PMID: 30587227 PMCID: PMC6307169 DOI: 10.1186/s40425-018-0481-0] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 12/13/2018] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND The prevalence of esophageal stenosis caused by immune checkpoint inhibitors in the context of induced immune mucositis and esophagitis is extremely rare. CASE PRESENTATION We report the case of a patient with stage IV pulmonary adenocarcinoma treated for 6 months with nivolumab who developed bilateral sterile conjunctivitis followed by oropharyngeal mucositis and esophagitis complicated by a severe esophageal stenosis. The laryngeal margin and hypopharyngeal mucosa appeared highly inflammatory with fibrinous deposits. Esophagogastroduodenoscopy revealed mucositis with a scar-like structure immediately below the upper esophageal sphincter with nonulcerative mucosa and an inflammatory aspect of the entire esophagus. No involvement of the stomach was observed. Oropharynx biopsies displayed marked lymphocytic T cell-infiltration with several foci of monocellular necrosis in the squamous epithelium. No morphologic evidence of adenocarcinoma and no signs of mycotic, bacterial or viral infection were noted. A blood sample revealed a discrete increase in the erythrocyte sedimentation rate (ESR) with no eosinophilia or leukocytosis. Liver and kidney function panel tests were normal. A thoracoabdominal CT scan reported no evidence of disease recurrence. Despite multiple boluses of methylprednisolone and high doses of prednisone continued for several months, the patient experienced very rapid symptomatological reappearance during three steroid tapering attempts and aggravation of his esophageal stenosis to an aphagic stage, requiring a nasogastric tube. This long course of high-dose corticosteroid treatment was complicated with osteoporosis-induced fractures with several spontaneous compressions of thoracolumbar vertebrae requiring an enlarged T10 to L5 cementoplasty. Anti-IL-6 blockade therapy with tocilizumab resulted in excellent clinical response, allowing the total resolution of the immune-related adverse events (irAEs) and leading to successful steroid tapering. CONCLUSIONS Herein, we describe the first case of a patient who developed autoimmune mucositis and esophagitis complicated by a severe refractory esophageal stenosis induced during treatment by nivolumab, which completely resolved after personalized treatment with tocilizumab, suggesting a role of IL-6 blockade in the management of severe steroid refractory esophageal stenosis and more broadly in refractory immune-related adverse events.
Collapse
Affiliation(s)
- Alice Horisberger
- Department of Medicine, Division of Immunology and Allergy, Lausanne University Hospital CHUV, rue du Bugnon 46, CH-1011, Lausanne, Switzerland
| | - Stefano La Rosa
- Service of Clinical Pathology, Institute of Pathology, Lausanne University Hospital CHUV, rue du Bugnon 25, CH-1011, Lausanne, Switzerland
| | - Jean-Philippe Zurcher
- Department of Oncology, Lausanne University Hospital CHUV, rue du Bugnon 46, CH-1011, Lausanne, Switzerland
| | - Stefan Zimmermann
- Department of Oncology, Lausanne University Hospital CHUV, rue du Bugnon 46, CH-1011, Lausanne, Switzerland
| | - Francois Spertini
- Department of Medicine, Division of Immunology and Allergy, Lausanne University Hospital CHUV, rue du Bugnon 46, CH-1011, Lausanne, Switzerland
| | - George Coukos
- Department of Oncology, Lausanne University Hospital CHUV, rue du Bugnon 46, CH-1011, Lausanne, Switzerland
- Ludwig Institute for Cancer Research, University of Lausanne, chemin des Boveresses 155, CH-1066, Epalinges, Switzerland
| | - Michel Obeid
- Department of Medicine, Division of Immunology and Allergy, Lausanne University Hospital CHUV, rue du Bugnon 46, CH-1011, Lausanne, Switzerland.
- Vaccination and Immunotherapy Center, Lausanne University Hospital CHUV, rue du Bugnon 17, CH-1011, Lausanne, Switzerland.
- Medical school Pitié-Salpêtrière, Sorbonne University, 91 Boulevard de l'Hôpital, F-75013, Paris, France.
| |
Collapse
|
589
|
Deshpande NP, Riordan SM, Castaño-Rodríguez N, Wilkins MR, Kaakoush NO. Signatures within the esophageal microbiome are associated with host genetics, age, and disease. MICROBIOME 2018; 6:227. [PMID: 30558669 PMCID: PMC6297961 DOI: 10.1186/s40168-018-0611-4] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 11/29/2018] [Indexed: 05/11/2023]
Abstract
BACKGROUND The esophageal microbiome has been proposed to be involved in a range of diseases including the esophageal adenocarcinoma cascade; however, little is currently known about its function and relationship to the host. Here, the esophageal microbiomes of 106 prospectively recruited patients were assessed using 16S rRNA and 18S rRNA amplicon sequencing as well as shotgun sequencing, and associations with age, gender, proton pump inhibitor use, host genetics, and disease were tested. RESULTS The esophageal microbiome was found to cluster into functionally distinct community types (esotypes) defined by the relative abundances of Streptococcus and Prevotella. While age was found to be a significant factor driving microbiome composition, bacterial signatures and functions such as enrichment with Gram-negative oral-associated bacteria and microbial lactic acid production were associated with the early stages of the esophageal adenocarcinoma cascade. Non-bacterial microbes such as archaea, Candida spp., and bacteriophages were also identified in low abundance in the esophageal microbiome. Specific host SNPs in NOTCH2, STEAP2-AS1, and NREP were associated with the composition of the esophageal microbiome in our cohort. CONCLUSIONS This study provides the most comprehensive assessment of the esophageal microbiome to date and identifies novel signatures and host markers that can be investigated further in the context of esophageal adenocarcinoma development.
Collapse
Affiliation(s)
- Nandan P Deshpande
- Systems Biology Initiative, School of Biotechnology and Biomolecular Sciences, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Stephen M Riordan
- Gastrointestinal and Liver Unit, The Prince of Wales Hospital, Randwick, NSW, 2031, Australia
| | | | - Marc R Wilkins
- Systems Biology Initiative, School of Biotechnology and Biomolecular Sciences, UNSW Sydney, Sydney, NSW, 2052, Australia
- School of Biotechnology and Biomolecular Sciences, UNSW Sydney, Sydney, NSW, 2052, Australia
- Ramaciotti Centre for Gene Function Analysis, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Nadeem O Kaakoush
- School of Medical Sciences, UNSW Sydney, Kensington, Sydney, NSW, 2052, Australia.
| |
Collapse
|
590
|
Abstract
Acute anterior uveitis (AAU) and the spondyloarthritis (SpA) subtypes ankylosing spondylitis, reactive arthritis and psoriatic arthritis are among the inflammatory diseases affected by the biology of the intestinal microbiome. In this Review, the relationship between AAU, SpA and the microbiome is discussed, with a focus on the major SpA risk gene HLA-B*27 and how it is associated with both intestinal tolerance and the loss of ocular immune privilege that can accompany AAU. We provide four potential mechanisms to account for how dysbiosis, barrier function and immune response contribute to the development of ocular inflammation and the pathogenesis of AAU. Finally, potential therapeutic avenues to target the microbiota for the clinical management of AAU and SpA are outlined.
Collapse
Affiliation(s)
- James T Rosenbaum
- Departments of Ophthalmology, Medicine and Cell Biology, Oregon Health and Science University, Portland, OR, USA
- Legacy Devers Eye Institute, Portland, OR, USA
| | - Mark Asquith
- Department of Medicine, Oregon Health and Science University, Portland, OR, USA.
| |
Collapse
|
591
|
Qualitative and Quantitative DNA- and RNA-Based Analysis of the Bacterial Stomach Microbiota in Humans, Mice, and Gerbils. mSystems 2018; 3:mSystems00262-18. [PMID: 30505943 PMCID: PMC6247015 DOI: 10.1128/msystems.00262-18] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 10/30/2018] [Indexed: 02/06/2023] Open
Abstract
Clinical stomach interventions, such as acid inhibition or bypass surgery, have been linked to fecal microbiota alterations. We demonstrate that the stomach microbiota largely overlaps those of adjacent gastrointestinal locations and identify gradual decreases and increases in the relative abundances of specific bacteria within the stomach, suggesting selective enrichment and depletion. Moreover, similarities between stomach and esophagus samples are proportional to the concentrations of Streptococcus (Firmicutes) in the stomach. The relative abundance of Firmicutes in the stomach, compared to that of Bacteroidetes, is increased in RNA relative to DNA, indicating higher transcriptional activity. Moreover, increased absolute bacterial loads are associated with decreased relative abundance of Firmicutes and higher relative abundance of Bacteroidetes. Our findings characterize the stomach microbiota as influenced by Bacteroidetes influx against a background of transcriptionally more active Firmicutes. Human, mouse, and gerbil stomach microbiotas differ at lower taxonomic levels, which might affect the utility of these model organisms. Clinical interventions in the stomach have been linked to fecal microbiota alterations, suggesting a function of the stomach in gastrointestinal (GI) homeostasis. We sought to determine the taxonomic bacterial biogeography of the upper GI tract, including different sites within the human stomach (cardia, corpus, and antrum), adjacent upstream (esophagus) and downstream (duodenum) locations, and luminal contents (aspirate), as well as whole-stomach samples from mice and gerbils. Qualitative and quantitative DNA- and RNA-based taxonomic microbiota analyses were combined to study the relationship of relative and absolute bacterial abundances and transcriptionally active bacterial microbiota components in the stomach of humans and mice. Stomach microbiota compositions resembled those of esophagus and duodenum. However, along the descending GI tract, the relative abundances of specific oropharyngeal commensals decreased (Streptococcus) or increased (Rothia mucilaginosa, Porphyromonas, and Lachnospiraceae). Furthermore, the compositional similarity (weighted UniFrac) between stomach aspirates and esophageal biopsy samples increased with gastric Streptococcus relative abundance. In both human aspirate and mouse stomach samples, Firmicutes were more abundant among transcriptionally active bacteria than Bacteroidetes. The relative abundance of Firmicutes in the stomach was negatively correlated and that of Bacteroidetes was positively correlated with absolute bacterial abundance, suggesting a disproportionate increase of Bacteroidetes over Firmicutes at higher bacterial densities. Human, mouse, and gerbil stomach samples showed similarities at higher taxonomic levels but differences at lower taxonomic levels. Our findings suggest selective enrichment and depletion of specific bacterial taxa in the stomach and Firmicutes being transcriptionally more active than Bacteroidetes that increase in relative abundance with total bacterial load. IMPORTANCE Clinical stomach interventions, such as acid inhibition or bypass surgery, have been linked to fecal microbiota alterations. We demonstrate that the stomach microbiota largely overlaps those of adjacent gastrointestinal locations and identify gradual decreases and increases in the relative abundances of specific bacteria within the stomach, suggesting selective enrichment and depletion. Moreover, similarities between stomach and esophagus samples are proportional to the concentrations of Streptococcus (Firmicutes) in the stomach. The relative abundance of Firmicutes in the stomach, compared to that of Bacteroidetes, is increased in RNA relative to DNA, indicating higher transcriptional activity. Moreover, increased absolute bacterial loads are associated with decreased relative abundance of Firmicutes and higher relative abundance of Bacteroidetes. Our findings characterize the stomach microbiota as influenced by Bacteroidetes influx against a background of transcriptionally more active Firmicutes. Human, mouse, and gerbil stomach microbiotas differ at lower taxonomic levels, which might affect the utility of these model organisms.
Collapse
|
592
|
Sorini C, Cardoso RF, Gagliani N, Villablanca EJ. Commensal Bacteria-Specific CD4 + T Cell Responses in Health and Disease. Front Immunol 2018; 9:2667. [PMID: 30524431 PMCID: PMC6256970 DOI: 10.3389/fimmu.2018.02667] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 10/29/2018] [Indexed: 12/14/2022] Open
Abstract
Over the course of evolution, mammalian body surfaces have adapted their complex immune system to allow a harmless coexistence with the commensal microbiota. The adaptive immune response, in particular CD4+ T cell-mediated, is crucial to maintain intestinal immune homeostasis by discriminating between harmless (e.g., dietary compounds and intestinal microbes) and harmful stimuli (e.g., pathogens). To tolerate food molecules and microbial components, CD4+ T cells establish a finely tuned crosstalk with the environment whereas breakdown of these mechanisms might lead to chronic disease associated with mucosal barriers and beyond. How commensal-specific immune responses are regulated and how these molecular and cellular mechanisms can be manipulated to treat chronic disorders is yet poorly understood. In this review, we discuss current knowledge of the regulation of commensal bacteria-specific CD4+ T cells. We place particular focus on the key role of commensal-specific CD4+ T cells in maintaining tolerance while efficiently eradicating local and systemic infections, with a focus on factors that trigger their aberrant activation.
Collapse
Affiliation(s)
- Chiara Sorini
- Immunology and Allergy Unit, Department of Medicine Solna, Karolinska Institute, Stockholm, Sweden
| | - Rebeca F. Cardoso
- Immunology and Allergy Unit, Department of Medicine Solna, Karolinska Institute, Stockholm, Sweden
| | - Nicola Gagliani
- Immunology and Allergy Unit, Department of Medicine Solna, Karolinska Institute, Stockholm, Sweden
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Eduardo J. Villablanca
- Immunology and Allergy Unit, Department of Medicine Solna, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
593
|
Sonoda A, Kamiyama N, Ozaka S, Gendo Y, Ozaki T, Hirose H, Noguchi K, Saechue B, Sachi N, Sakai K, Mizukami K, Hidano S, Murakami K, Kobayashi T. Oral administration of antibiotics results in fecal occult bleeding due to metabolic disorders and defective proliferation of the gut epithelial cell in mice. Genes Cells 2018; 23:1043-1055. [PMID: 30353943 DOI: 10.1111/gtc.12649] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 09/21/2018] [Accepted: 10/09/2018] [Indexed: 12/29/2022]
Abstract
Antibiotics sometimes exert adverse effects on the pathogenesis of colitis due to the dysbiosis resulting from the disruption of gut homeostasis. However, the precise mechanisms underlying colitogenic effects of antibiotic-induced colitis are largely unknown. Here, we show a novel murine fecal occult bleeding model induced by the combinatorial treatment of ampicillin and vancomycin, which is accompanied by an enlarged cecum, upregulation of pro-inflammatory cytokines IL-6 and IL-12, a reduction in Ki-67-positive epithelial cell number and an increase in the apoptotic cell number in the colon. Moreover, gas chromatography-tandem mass analysis showed that various kinds of metabolites, including glutamic acid and butyric acid, were significantly decreased in the cecal contents. In addition, abundance of butyric acid producer Clostridiales was dramatically reduced in the enlarged cecum. Interestingly, supplementation of monosodium glutamate or its precursor glutamine suppressed colonic IL-6 and IL-12, protected from cell apoptosis and prevented fecal occult blood indicating that the reduced level of glutamic acid is a possible mechanism of antibiotic-induced fecal occult bleeding. Our data showed a novel mechanism of antibiotic-induced fecal occult bleeding providing a new insight into the clinical application of glutamic acid for the treatment of antibiotic-induced colitis.
Collapse
Affiliation(s)
- Akira Sonoda
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Yufu, Japan.,Department of Gastroenterology, Faculty of Medicine, Oita University, Yufu, Japan
| | - Naganori Kamiyama
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Yufu, Japan
| | - Sotaro Ozaka
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Yufu, Japan.,Department of Gastroenterology, Faculty of Medicine, Oita University, Yufu, Japan
| | - Yoshiko Gendo
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Yufu, Japan
| | - Takashi Ozaki
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Yufu, Japan
| | - Haruna Hirose
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Yufu, Japan
| | - Kaori Noguchi
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Yufu, Japan
| | - Benjawan Saechue
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Yufu, Japan
| | - Nozomi Sachi
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Yufu, Japan
| | - Kumiko Sakai
- Department of Division of Life Science Research, Faculty of Medicine, Oita University, Yufu, Japan
| | - Kazuhiro Mizukami
- Department of Gastroenterology, Faculty of Medicine, Oita University, Yufu, Japan.,Clinical Training Institute for Interns, Faculty of Medicine, Oita University, Yufu, Japan
| | - Shinya Hidano
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Yufu, Japan
| | - Kazunari Murakami
- Department of Gastroenterology, Faculty of Medicine, Oita University, Yufu, Japan
| | - Takashi Kobayashi
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Yufu, Japan
| |
Collapse
|
594
|
Adolph TE, Effenberger M, Tilg H. How our understanding of the microbiome has changed the way we look at liver diseases. Biomark Med 2018; 12:1203-1206. [PMID: 30444134 DOI: 10.2217/bmm-2018-0266] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Timon E Adolph
- Department of Internal Medicine I, Gastroenterology, Hepatology, Metabolism & Endocrinology, Medical University Innsbruck, Innsbruck, Austria
| | - Maria Effenberger
- Department of Internal Medicine I, Gastroenterology, Hepatology, Metabolism & Endocrinology, Medical University Innsbruck, Innsbruck, Austria
| | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Metabolism & Endocrinology, Medical University Innsbruck, Innsbruck, Austria
| |
Collapse
|
595
|
Khatun A, Sakurai M, Okada K, Sakai Y, Morimoto M. Detection of α-defensin in eosinophils in helminth-infected mouse model. J Vet Med Sci 2018; 80:1887-1894. [PMID: 30393268 PMCID: PMC6305521 DOI: 10.1292/jvms.18-0601] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
α-defensin is a potent antimicrobial peptide secreted from intestinal mucosal epithelial
cells, such as Paneth cells, and affects not only bacteria but also parasites and fungi.
Recently, human eosinophils have also been shown to produce α-defensin, but no studies
have been done on other animals. In this study, we attempted to detect α-defensin protein
in mouse eosinophils infiltrating the intestinal mucosa during a helminth infection using
Zamboni fixation and immunohistochemistry. Most of the eosinophils infiltrating the
intestinal mucosa during helminth infection were positive for α-defensin. The expression
level of α-defensin mRNA was 50 fold that in the control. Meanwhile, the number of Paneth
cells was doubled, and their α-defensin fluorescence intensity was increased. These
results suggested that eosinophils are also important producers of α-defensin, such as
Paneth cells in mice, and that α-defensin produced from eosinophils might be involved in
defensive mechanisms against helminths. Moreover, the experimental system used in this
study is a good model to study the generation of α-defensin by eosinophils.
Collapse
Affiliation(s)
- Afia Khatun
- Laboratory of Veterinary Pathology, Joint Faculty of Veterinary Medicine, Yamaguchi University, 1677-1 Yoshida, Yamaguchi 753-8515, Japan
| | - Masashi Sakurai
- Laboratory of Veterinary Pathology, Joint Faculty of Veterinary Medicine, Yamaguchi University, 1677-1 Yoshida, Yamaguchi 753-8515, Japan
| | - Kazuki Okada
- North Lab, 8-35 Hondori, 2-chome kita, Shiroishi-ku, Sapporo, Hokkaido 003-0027, Japan
| | - Yusuke Sakai
- Laboratory of Veterinary Pathology, Joint Faculty of Veterinary Medicine, Yamaguchi University, 1677-1 Yoshida, Yamaguchi 753-8515, Japan
| | - Masahiro Morimoto
- Laboratory of Veterinary Pathology, Joint Faculty of Veterinary Medicine, Yamaguchi University, 1677-1 Yoshida, Yamaguchi 753-8515, Japan
| |
Collapse
|
596
|
Abstract
PURPOSE OF REVIEW In this review, we discuss recent advances into delineating the dual role of intestinal phagocytes in health and during intestinal disease. We further discuss the key role of gut-resident macrophages in recognition of bacterial and fungal microbiota in the gut. RECENT FINDINGS Inflammatory bowel disease (IBD) commonly manifests with pathologic changes in the composition of gut bacterial and fungal microbiota. Intestinal macrophages are key regulators of the balance between tolerogenic immunity and inflammation. Recent studies have highlighted the role of resident intestinal macrophages in the control of commensal fungi and bacteria in the steady state and during dysbiosis. The dual role of these cells in maintaining intestinal homeostasis and responding to microbiota dysbiosis during inflammation is being increasingly studied. SUMMARY It is becoming increasingly clear that an aberrant proinflammatory response to microbiota by infiltrating monocytes plays a role in the development of intestinal inflammation. Intestinal mononuclear phagocytes with characteristics of macrophages play an important role in limiting fungal and bacterial overgrowth under these conditions, but can be influenced by the inflammatory environment to further propel inflammation. Better understanding of the interaction of intestinal macrophages with host microbiota including commensal fungi and bacteria, provides an opportunity for the development of more targeted therapies for IBD.
Collapse
|
597
|
Schirmer M, Denson L, Vlamakis H, Franzosa EA, Thomas S, Gotman NM, Rufo P, Baker SS, Sauer C, Markowitz J, Pfefferkorn M, Oliva-Hemker M, Rosh J, Otley A, Boyle B, Mack D, Baldassano R, Keljo D, LeLeiko N, Heyman M, Griffiths A, Patel AS, Noe J, Kugathasan S, Walters T, Huttenhower C, Hyams J, Xavier RJ. Compositional and Temporal Changes in the Gut Microbiome of Pediatric Ulcerative Colitis Patients Are Linked to Disease Course. Cell Host Microbe 2018; 24:600-610.e4. [PMID: 30308161 PMCID: PMC6277984 DOI: 10.1016/j.chom.2018.09.009] [Citation(s) in RCA: 185] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 06/28/2018] [Accepted: 09/03/2018] [Indexed: 12/11/2022]
Abstract
Evaluating progression risk and determining optimal therapy for ulcerative colitis (UC) is challenging as many patients exhibit incomplete responses to treatment. As part of the PROTECT (Predicting Response to Standardized Colitis Therapy) Study, we evaluated the role of the gut microbiome in disease course for 405 pediatric, new-onset, treatment-naive UC patients. Patients were monitored for 1 year upon treatment initiation, and microbial taxonomic composition was analyzed from fecal samples and rectal biopsies. Depletion of core gut microbes and expansion of bacteria typical of the oral cavity were associated with baseline disease severity. Remission and refractory disease were linked to species-specific temporal changes that may be implicative of therapy efficacy, and a pronounced increase in microbiome variability was observed prior to colectomy. Finally, microbial associations with disease-associated serological markers suggest host-microbial interactions in UC. These insights will help improve existing treatments and develop therapeutic approaches guiding optimal medical care.
Collapse
Affiliation(s)
- Melanie Schirmer
- The Broad Institute of MIT and Harvard, Infectious Disease and Microbiome, Cambridge, MA 02142, USA; Harvard T.H. Chan School of Public Health, Biostatistics Department, Boston, MA 02115, USA
| | - Lee Denson
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Hera Vlamakis
- The Broad Institute of MIT and Harvard, Infectious Disease and Microbiome, Cambridge, MA 02142, USA; Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Eric A Franzosa
- The Broad Institute of MIT and Harvard, Infectious Disease and Microbiome, Cambridge, MA 02142, USA; Harvard T.H. Chan School of Public Health, Biostatistics Department, Boston, MA 02115, USA
| | - Sonia Thomas
- Collaborative Studies Coordinating Center, Department of Biostatistics, University of North Carolina, Chapel Hill, NC 27516, USA; RTI International, Biostatistics and Epidemiology Division, Research Triangle Park, NC 27709, USA
| | - Nathan M Gotman
- Collaborative Studies Coordinating Center, Department of Biostatistics, University of North Carolina, Chapel Hill, NC 27516, USA
| | - Paul Rufo
- Children's Hospital Boston, Boston, MA 02115, USA
| | - Susan S Baker
- Women and Children's Hospital of Buffalo WCHOB, Buffalo, NY 14222, USA
| | - Cary Sauer
- Emory Children's Center, Atlanta, GA 30322, USA
| | - James Markowitz
- Cohen Children's Medical Center, Pediatric Gastroenterology, New York, NY 11040, USA
| | - Marian Pfefferkorn
- Riley Children's Hospital Indiana University, School of Medicine, Section of Gastroenterology/Hepatology/Nutrition, Indianapolis, IN 46202, USA
| | - Maria Oliva-Hemker
- Johns Hopkins Children's Center, Department of Pediatrics, Baltimore, MD 21287, USA
| | - Joel Rosh
- Goryeb Children's Hospital/Atlantic Health, Pediatric Gastroenterology, Morristown, NJ 07960, USA
| | - Anthony Otley
- IWK Health Centre, Division of Gastroenterology and Nutrition, Halifax, NS B3K 6R8, Canada
| | - Brendan Boyle
- Nationwide Children's Hospital, Pediatrics, Columbus, OH 43205, USA
| | - David Mack
- Children's Hospital of Eastern Ontario and University of Ottawa, Department of Pediatrics, Ottawa, ON K1H 8L1, Canada
| | - Robert Baldassano
- Children's Hospital of Philadelphia CHOP, Pediatric Gastroenterologist, Philadelphia, PA 19104, USA
| | - David Keljo
- UPMC Children's Hospital of Pittsburgh, Department of Pediatrics, Pittsburgh, PA 15224, USA
| | - Neal LeLeiko
- Hasbro Children's Hospital, Pediatric Gastroenterology, Providence, RI 02903, USA
| | - Melvin Heyman
- University of California at San Francisco, Pediatric Gastroenterology, San Francisco, CA 94158, USA
| | - Anne Griffiths
- Sickkids Hospital, University of Toronto, Gastroenterology, Hepatology and Nutrition, Toronto, ON M5G 1X8, Canada
| | - Ashish S Patel
- UT Southwestern, Department of Pediatrics, Dallas, TX 75390, USA
| | - Joshua Noe
- Medical College of Wisconsin, Gastroenterology, Milwaukee, WI 53226, USA
| | | | - Thomas Walters
- Sickkids Hospital, University of Toronto, Gastroenterology, Hepatology and Nutrition, Toronto, ON M5G 1X8, Canada
| | - Curtis Huttenhower
- The Broad Institute of MIT and Harvard, Infectious Disease and Microbiome, Cambridge, MA 02142, USA; Harvard T.H. Chan School of Public Health, Biostatistics Department, Boston, MA 02115, USA
| | - Jeffrey Hyams
- Connecticut Children's Medical Center, Division of Digestive Diseases, Hartford, CT 06106, USA
| | - Ramnik J Xavier
- The Broad Institute of MIT and Harvard, Infectious Disease and Microbiome, Cambridge, MA 02142, USA; Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Boston, MA 02114, USA; Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA.
| |
Collapse
|
598
|
Sokol H, Mahlaoui N, Aguilar C, Bach P, Join-Lambert O, Garraffo A, Seksik P, Danion F, Jegou S, Straube M, Lenoir C, Neven B, Moshous D, Blanche S, Pigneur B, Goulet O, Ruemmele F, Suarez F, Beaugerie L, Pannier S, Mazingue F, Lortholary O, Galicier L, Picard C, de Saint Basile G, Latour S, Fischer A. Intestinal dysbiosis in inflammatory bowel disease associated with primary immunodeficiency. J Allergy Clin Immunol 2018; 143:775-778.e6. [PMID: 30312711 DOI: 10.1016/j.jaci.2018.09.021] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Revised: 09/20/2018] [Accepted: 09/28/2018] [Indexed: 10/28/2022]
Affiliation(s)
- Harry Sokol
- Sorbonne Universités, UPMC Univ. Paris 06, École normale supérieure, PSL Research University, CNRS, INSERM, Assistance Publique Hôpitaux de Paris (APHP), Laboratoire des Biomolécules (LBM), rue de Chaligny, Paris, France; INRA, UMR1319 Micalis & AgroParisTech, Jouy en Josas, France; Department of Gastroenterology, Saint Antoine Hospital, AP-HP, UPMC Univ. Paris 06, Paris, France; French National Reference Center for Primary Immune Deficiency (CEREDIH), Necker-Enfants Malades University Hospital, APHP, Paris, France
| | - Nizar Mahlaoui
- French National Reference Center for Primary Immune Deficiency (CEREDIH), Necker-Enfants Malades University Hospital, APHP, Paris, France; Paediatric Haematology-Immunology and Rheumatology Department, Hôpital Necker-Enfants Malades, APHP, Paris, France
| | - Claire Aguilar
- Infectious Diseases and Tropical Medicine Department Paris Descartes University, Necker-Pasteur Infectious Diseases Center, Necker-Enfants Malades Hospital APHP, Paris, France; INSERM UMR 1163, Imagine Institute, Paris, France
| | - Perrine Bach
- French National Reference Center for Primary Immune Deficiency (CEREDIH), Necker-Enfants Malades University Hospital, APHP, Paris, France
| | - Olivier Join-Lambert
- Microbiology Department, Paris Descartes University, Necker-Enfants Malades Hospital, APHP, Paris, France
| | - Aurélie Garraffo
- Infectious Diseases and Tropical Medicine Department Paris Descartes University, Necker-Pasteur Infectious Diseases Center, Necker-Enfants Malades Hospital APHP, Paris, France
| | - Philippe Seksik
- Sorbonne Universités, UPMC Univ. Paris 06, École normale supérieure, PSL Research University, CNRS, INSERM, Assistance Publique Hôpitaux de Paris (APHP), Laboratoire des Biomolécules (LBM), rue de Chaligny, Paris, France; Department of Gastroenterology, Saint Antoine Hospital, AP-HP, UPMC Univ. Paris 06, Paris, France
| | - François Danion
- Infectious Diseases and Tropical Medicine Department Paris Descartes University, Necker-Pasteur Infectious Diseases Center, Necker-Enfants Malades Hospital APHP, Paris, France
| | - Sarah Jegou
- Sorbonne Universités, UPMC Univ. Paris 06, École normale supérieure, PSL Research University, CNRS, INSERM, Assistance Publique Hôpitaux de Paris (APHP), Laboratoire des Biomolécules (LBM), rue de Chaligny, Paris, France
| | - Marjolene Straube
- Sorbonne Universités, UPMC Univ. Paris 06, École normale supérieure, PSL Research University, CNRS, INSERM, Assistance Publique Hôpitaux de Paris (APHP), Laboratoire des Biomolécules (LBM), rue de Chaligny, Paris, France
| | | | - Bénédicte Neven
- Paediatric Haematology-Immunology and Rheumatology Department, Hôpital Necker-Enfants Malades, APHP, Paris, France; INSERM UMR 1163, Imagine Institute, Paris, France; Paris Descartes University, Sorbonne Paris Cité, Imagine Institute, Paris, France
| | - Despina Moshous
- Paediatric Haematology-Immunology and Rheumatology Department, Hôpital Necker-Enfants Malades, APHP, Paris, France; INSERM UMR 1163, Imagine Institute, Paris, France; Paris Descartes University, Sorbonne Paris Cité, Imagine Institute, Paris, France
| | - Stéphane Blanche
- Paediatric Haematology-Immunology and Rheumatology Department, Hôpital Necker-Enfants Malades, APHP, Paris, France; Paris Descartes University, Sorbonne Paris Cité, Imagine Institute, Paris, France
| | - Bénédicte Pigneur
- INSERM UMR 1163, Imagine Institute, Paris, France; Paediatric Gastroenterology Hepatology and Nutrition Department, Paris Descartes University, Hôpital Necker-Enfants Malades, APHP, Paris, France
| | - Olivier Goulet
- INSERM UMR 1163, Imagine Institute, Paris, France; Paediatric Gastroenterology Hepatology and Nutrition Department, Paris Descartes University, Hôpital Necker-Enfants Malades, APHP, Paris, France
| | - Frank Ruemmele
- INSERM UMR 1163, Imagine Institute, Paris, France; Paediatric Gastroenterology Hepatology and Nutrition Department, Paris Descartes University, Hôpital Necker-Enfants Malades, APHP, Paris, France
| | - Felipe Suarez
- French National Reference Center for Primary Immune Deficiency (CEREDIH), Necker-Enfants Malades University Hospital, APHP, Paris, France; INSERM UMR 1163, Imagine Institute, Paris, France; Hematology Department, Paris Descartes University, Necker-Enfants Malades University Hospital, APHP, Paris, France
| | - Laurent Beaugerie
- Department of Gastroenterology, Saint Antoine Hospital, AP-HP, UPMC Univ. Paris 06, Paris, France
| | - Stéphanie Pannier
- Study Center for Primary Immunodeficiencies, Necker-Enfants Malades University Hospital, APHP, Paris, France; Department of Orthopedic Pediatrics, Necker-Enfants Malades University Hospital, APHP, Paris, France
| | - Françoise Mazingue
- Department of Pediatrics, Hôpital Jeanne de Flandre, University Hospital of Lille, Lille, France
| | - Olivier Lortholary
- French National Reference Center for Primary Immune Deficiency (CEREDIH), Necker-Enfants Malades University Hospital, APHP, Paris, France; Infectious Diseases and Tropical Medicine Department Paris Descartes University, Necker-Pasteur Infectious Diseases Center, Necker-Enfants Malades Hospital APHP, Paris, France; Paris Descartes University, Sorbonne Paris Cité, Imagine Institute, Paris, France
| | - Lionel Galicier
- Department of Clinical Immunology, Saint-Louis Hospital, APHP, Paris, France
| | - Capucine Picard
- French National Reference Center for Primary Immune Deficiency (CEREDIH), Necker-Enfants Malades University Hospital, APHP, Paris, France; INSERM UMR 1163, Imagine Institute, Paris, France; Paris Descartes University, Sorbonne Paris Cité, Imagine Institute, Paris, France; Study Center for Primary Immunodeficiencies, Necker-Enfants Malades University Hospital, APHP, Paris, France
| | - Geneviève de Saint Basile
- INSERM UMR 1163, Imagine Institute, Paris, France; Paris Descartes University, Sorbonne Paris Cité, Imagine Institute, Paris, France; Study Center for Primary Immunodeficiencies, Necker-Enfants Malades University Hospital, APHP, Paris, France
| | - Sylvain Latour
- INSERM UMR 1163, Imagine Institute, Paris, France; Paris Descartes University, Sorbonne Paris Cité, Imagine Institute, Paris, France
| | - Alain Fischer
- French National Reference Center for Primary Immune Deficiency (CEREDIH), Necker-Enfants Malades University Hospital, APHP, Paris, France; Paediatric Haematology-Immunology and Rheumatology Department, Hôpital Necker-Enfants Malades, APHP, Paris, France; INSERM UMR 1163, Imagine Institute, Paris, France; Paris Descartes University, Sorbonne Paris Cité, Imagine Institute, Paris, France; Collège de France, Paris, France.
| |
Collapse
|
599
|
Amphiregulin-producing γδ T cells are vital for safeguarding oral barrier immune homeostasis. Proc Natl Acad Sci U S A 2018; 115:10738-10743. [PMID: 30279177 PMCID: PMC6196490 DOI: 10.1073/pnas.1802320115] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Loss of oral barrier homeostasis leads to the development of periodontitis, the most common chronic inflammatory condition of mankind. Therefore, it is important to better understand the immune mediators acting at this unique barrier to safeguard tissue integrity. Here we identify a vital role for γδ T cells in constraining pathological inflammation at the oral barrier, as the absence of γδ T cells resulted in enhanced pathology during periodontitis. We show that oral barrier γδ T cells produce the reparative cytokine Amphiregulin, administration of which rescued the elevated oral pathology of tcrδ−/− mice. Collectively, we identify a pathway controlling oral immunity mediated by barrier-resident γδ T cells, highlighting that these cells are crucial guards of oral barrier immune homeostasis. γδ T cells are enriched at barrier sites such as the gut, skin, and lung, where their roles in maintaining barrier integrity are well established. However, how these cells contribute to homeostasis at the gingiva, a key oral barrier and site of the common chronic inflammatory disease periodontitis, has not been explored. Here we demonstrate that the gingiva is policed by γδ T cells with a T cell receptor (TCR) repertoire that diversifies during development. Gingival γδ T cells accumulated rapidly after birth in response to barrier damage, and strikingly, their absence resulted in enhanced pathology in murine models of the oral inflammatory disease periodontitis. Alterations in bacterial communities could not account for the increased disease severity seen in γδ T cell-deficient mice. Instead, gingival γδ T cells produced the wound healing associated cytokine amphiregulin, administration of which rescued the elevated oral pathology of tcrδ−/− mice. Collectively, our results identify γδ T cells as critical constituents of the immuno-surveillance network that safeguard gingival tissue homeostasis.
Collapse
|
600
|
Harris KG, Chang EB. The intestinal microbiota in the pathogenesis of inflammatory bowel diseases: new insights into complex disease. Clin Sci (Lond) 2018; 132:2013-2028. [PMID: 30232239 PMCID: PMC6907688 DOI: 10.1042/cs20171110] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 08/30/2018] [Accepted: 09/04/2018] [Indexed: 12/16/2022]
Abstract
Inflammatory bowel diseases (IBD) are a group of chronic diseases of increasing worldwide prevalence characterized by gastrointestinal (GI) inflammation leading to debilitating symptoms and complications. The contribution of the intestinal microbiota to the pathogenesis and etiology of these diseases is an area of active research interest. Here, we discuss key mechanisms underlying the chronic inflammation seen in IBD as well as evidence implicating the intestinal microbiota in the development and potentiation of that inflammation. We also discuss recently published work in areas of interest within the field of microbial involvement in IBD pathogenesis - the importance of proper microecology within the GI tract, the evidence that the intestinal microbiota transduces environmental and genetic risk factors for IBD, and the mechanisms by which microbial products contribute to communication between microbe and host. There is an extensive body of published research on the evidence for microbial involvement in IBD; the goal of this review is to highlight the growing edges of the field where exciting and innovative research is pushing the boundaries of the conceptual framework of the role of the intestinal microbiota in IBD pathogenesis.
Collapse
Affiliation(s)
| | - Eugene B Chang
- Department of Medicine, University of Chicago, Chicago, IL 60637, U.S.A.
| |
Collapse
|