51
|
Ma X, Dai Z, Sun K, Zhang Y, Chen J, Yang Y, Tso P, Wu G, Wu Z. Intestinal Epithelial Cell Endoplasmic Reticulum Stress and Inflammatory Bowel Disease Pathogenesis: An Update Review. Front Immunol 2017; 8:1271. [PMID: 29118753 PMCID: PMC5660968 DOI: 10.3389/fimmu.2017.01271] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 09/25/2017] [Indexed: 12/18/2022] Open
Abstract
The intestinal epithelial cells serve essential roles in maintaining intestinal homeostasis, which relies on appropriate endoplasmic reticulum (ER) function for proper protein folding, modification, and secretion. Exogenous or endogenous risk factors with an ability to disturb the ER function can impair the intestinal barrier function and activate inflammatory responses in the host. The last decade has witnessed considerable progress in the understanding of the functional role of ER stress and unfolded protein response (UPR) in the gut homeostasis and its significant contribution to the pathogenesis of inflammatory bowel disease (IBD). Herein, we review recent evidence supporting the viewpoint that deregulation of ER stress and UPR signaling in the intestinal epithelium, including the absorptive cells, Paneth cells, goblet cells, and enteroendocrine cells, mediates the action of genetic or environmental factors driving colitis in experimental animals and IBD patients. In addition, we highlight pharmacologic application of chaperones or small molecules that enhance protein folding and modification capacity or improve the function of the ER. These molecules represent potential therapeutic strategies in the prevention or treatment of IBD through restoring ER homeostasis in intestinal epithelial cells.
Collapse
Affiliation(s)
- Xiaoshi Ma
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing, China
| | - Zhaolai Dai
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing, China
| | - Kaiji Sun
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing, China
| | - Yunchang Zhang
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing, China
| | - Jingqing Chen
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing, China
| | - Ying Yang
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing, China
| | - Patrick Tso
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Institute, University of Cincinnati, Cincinnati, OH, United States
| | - Guoyao Wu
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing, China.,Department of Animal Science, Texas A&M University, College Station, TX, United States
| | - Zhenlong Wu
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing, China.,Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
| |
Collapse
|
52
|
Peck BCE, Shanahan MT, Singh AP, Sethupathy P. Gut Microbial Influences on the Mammalian Intestinal Stem Cell Niche. Stem Cells Int 2017; 2017:5604727. [PMID: 28904533 PMCID: PMC5585682 DOI: 10.1155/2017/5604727] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 08/02/2017] [Indexed: 02/07/2023] Open
Abstract
The mammalian intestinal epithelial stem cell (IESC) niche is comprised of diverse epithelial, immune, and stromal cells, which together respond to environmental changes within the lumen and exert coordinated regulation of IESC behavior. There is growing appreciation for the role of the gut microbiota in modulating intestinal proliferation and differentiation, as well as other aspects of intestinal physiology. In this review, we evaluate the diverse roles of known niche cells in responding to gut microbiota and supporting IESCs. Furthermore, we discuss the potential mechanisms by which microbiota may exert their influence on niche cells and possibly on IESCs directly. Finally, we present an overview of the benefits and limitations of available tools to study niche-microbe interactions and provide our recommendations regarding their use and standardization. The study of host-microbe interactions in the gut is a rapidly growing field, and the IESC niche is at the forefront of host-microbe activity to control nutrient absorption, endocrine signaling, energy homeostasis, immune response, and systemic health.
Collapse
Affiliation(s)
- Bailey C. E. Peck
- Department of Surgery, School of Medicine, University of Michigan, Ann Arbor, MI 48105, USA
| | - Michael T. Shanahan
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Ajeet P. Singh
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Praveen Sethupathy
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
53
|
Hosomi S, Grootjans J, Tschurtschenthaler M, Krupka N, Matute JD, Flak MB, Martinez-Naves E, Gomez Del Moral M, Glickman JN, Ohira M, Lanier LL, Kaser A, Blumberg R. Intestinal epithelial cell endoplasmic reticulum stress promotes MULT1 up-regulation and NKG2D-mediated inflammation. J Exp Med 2017; 214:2985-2997. [PMID: 28747426 PMCID: PMC5626394 DOI: 10.1084/jem.20162041] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Revised: 05/25/2017] [Accepted: 07/10/2017] [Indexed: 12/25/2022] Open
Abstract
Hosomi et al. show that intestinal epithelial cell–specific deletion of X-box–binding protein 1, an unfolded protein response–related transcription factor, results in CHOP-dependent increased expression of specific natural killer group 2 member D (NKG2D) ligands. This activates NKG2D-expressing intraepithelial group 1 ILCs and promotes small intestinal inflammation. Endoplasmic reticulum (ER) stress is commonly observed in intestinal epithelial cells (IECs) and can, if excessive, cause spontaneous intestinal inflammation as shown by mice with IEC-specific deletion of X-box–binding protein 1 (Xbp1), an unfolded protein response–related transcription factor. In this study, Xbp1 deletion in the epithelium (Xbp1ΔIEC) is shown to cause increased expression of natural killer group 2 member D (NKG2D) ligand (NKG2DL) mouse UL16-binding protein (ULBP)–like transcript 1 and its human orthologue cytomegalovirus ULBP via ER stress–related transcription factor C/EBP homology protein. Increased NKG2DL expression on mouse IECs is associated with increased numbers of intraepithelial NKG2D-expressing group 1 innate lymphoid cells (ILCs; NK cells or ILC1). Blockade of NKG2D suppresses cytolysis against ER-stressed epithelial cells in vitro and spontaneous enteritis in vivo. Pharmacological depletion of NK1.1+ cells also significantly improved enteritis, whereas enteritis was not ameliorated in Recombinase activating gene 1−/−;Xbp1ΔIEC mice. These experiments reveal innate immune sensing of ER stress in IECs as an important mechanism of intestinal inflammation.
Collapse
Affiliation(s)
- Shuhei Hosomi
- Department of Medicine, Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.,Department of Gastroenterology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Joep Grootjans
- Department of Medicine, Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.,Department of Gastroenterology and Hepatology, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Markus Tschurtschenthaler
- Department of Medicine, Division of Gastroenterology, University of Cambridge, Cambridge, England, UK
| | - Niklas Krupka
- Department of Medicine, Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Juan D Matute
- Department of Medicine, Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.,Division of Newborn Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Magdalena B Flak
- Department of Medicine, Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Eduardo Martinez-Naves
- Department of Microbiology and Immunology, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| | - Manuel Gomez Del Moral
- Department of Cell Biology, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| | | | - Mizuki Ohira
- Department of Gastroenterology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Lewis L Lanier
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA.,Parker Institute for Cancer Immunotherapy, University of California, San Francisco, San Francisco, CA
| | - Arthur Kaser
- Department of Gastroenterology and Hepatology, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Richard Blumberg
- Department of Medicine, Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
54
|
Torres J, Mehandru S, Colombel JF, Peyrin-Biroulet L. Crohn's disease. Lancet 2017; 389:1741-1755. [PMID: 27914655 DOI: 10.1016/s0140-6736(16)31711-1] [Citation(s) in RCA: 1749] [Impact Index Per Article: 218.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2016] [Revised: 07/09/2016] [Accepted: 07/12/2016] [Indexed: 02/06/2023]
Abstract
Crohn's disease is a chronic inflammatory disease of the gastrointestinal tract, with increasing incidence worldwide. Crohn's disease might result from a complex interplay between genetic susceptibility, environmental factors, and altered gut microbiota, leading to dysregulated innate and adaptive immune responses. The typical clinical scenario is a young patient presenting with abdominal pain, chronic diarrhoea, weight loss, and fatigue. Assessment of disease extent and of prognostic factors for complications is paramount to guide therapeutic decisions. Current strategies aim for deep and long-lasting remission, with the goal of preventing complications, such as surgery, and blocking disease progression. Central to these strategies is the introduction of early immunosuppression or combination therapy with biologicals in high-risk patients, combined with a tight and frequent control of inflammation, and adjustment of therapy on the basis of that assessment (treat to target strategy). The therapeutic armamentarium for Crohn's disease is expanding, and therefore the need to develop biomarkers that can predict response to therapies will become increasingly important for personalised medicine decisions in the near future. In this Seminar, we provide a physician-oriented overview of Crohn's disease in adults, ranging from epidemiology and cause to clinical diagnosis, natural history, patient stratification and clinical management, and ending with an overview of emerging therapies and future directions for research.
Collapse
Affiliation(s)
- Joana Torres
- Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Saurabh Mehandru
- Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Jean-Frédéric Colombel
- Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York City, NY, USA.
| | - Laurent Peyrin-Biroulet
- Department of Gastroenterology, University Hospital of Nancy-Brabois, Vandœuvre-lès-Nancy, France
| |
Collapse
|
55
|
Chairatana P, Nolan EM. Human α-Defensin 6: A Small Peptide That Self-Assembles and Protects the Host by Entangling Microbes. Acc Chem Res 2017; 50:960-967. [PMID: 28296382 DOI: 10.1021/acs.accounts.6b00653] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Human α-defensin 6 (HD6) is a 32-residue cysteine-rich peptide that contributes to innate immunity by protecting the host at mucosal sites. This peptide is produced in small intestinal Paneth cells, stored as an 81-residue precursor peptide named proHD6 in granules, and released into the lumen. One unusual feature of HD6 is that it lacks the broad-spectrum antimicrobial activity observed for other human α-defensins, including the Paneth cell peptide human α-defensin 5 (HD5). HD6 exhibits unprecedented self-assembly properties, which confer an unusual host-defense function. HD6 monomers self-assemble into higher-order oligomers termed "nanonets", which entrap microbes and prevent invasive gastrointestinal pathogens such as Salmonella enterica serovar Typhimurium and Listeria monocytogenes from entering host cells. One possible advantage of this host-defense mechanism is that HD6 helps to keep microbes in the lumen such that they can be excreted or attacked by other components of the immune system, such as recruited neutrophils. In this Account, we report our current understanding of HD6 and focus on work published since 2012 when Bevins and co-workers described the discovery of HD6 nanonets in the literature. First, we present studies that address the biosynthesis, storage, and maturation of HD6, which demonstrate that nature uses a propeptide strategy to spatially and temporally control the formation of HD6 nanonets in the small intestine. The propeptide is stored in Paneth cell granules, and proteolysis occurs during or following release into the lumen, which affords the 32-residue mature peptide that self-assembles. We subsequently highlight structure-function studies that provide a foundation for understanding the molecular basis for why HD6 exhibits unusual self-assembly properties compared with other characterized defensins. The disposition of hydrophobic residues in the HD6 primary structure differs from that of other human α-defensins and is an important structural determinant for oligomerization. Lastly, we consider functional studies that illuminate how HD6 contributes to mucosal immunity. We recently discovered that in addition to blocking bacterial invasion into host epithelial cells by Gram-negative and Gram-positive gastrointestinal pathogens, HD6 suppresses virulence traits displayed by the opportunistic human fungal pathogen Candida albicans. In particular, we found that C. albicans biofilm formation, which causes complications in the treatment of candidiasis, is inhibited by HD6. This observation suggests that HD6 may contribute to intestinal homeostasis by helping to keep C. albicans in its commensal state. We intend for this Account to inspire further biochemical, biophysical, and biological investigations that will advance our understanding of HD6 in mucosal immunity and the host-microbe interaction.
Collapse
Affiliation(s)
- Phoom Chairatana
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Elizabeth M. Nolan
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
56
|
Liu TC, Naito T, Liu Z, VanDussen KL, Haritunians T, Li D, Endo K, Kawai Y, Nagasaki M, Kinouchi Y, McGovern DP, Shimosegawa T, Kakuta Y, Stappenbeck TS. LRRK2 but not ATG16L1 is associated with Paneth cell defect in Japanese Crohn's disease patients. JCI Insight 2017; 2:e91917. [PMID: 28352666 DOI: 10.1172/jci.insight.91917] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND. Morphological patterns of Paneth cells are a prognostic biomarker in Western Crohn's disease (CD) patients, and are associated with autophagy-associated ATG16L1 and NOD2 variants. We hypothesized that genetic determinants of Paneth cell phenotype in other ethnic CD cohorts are distinct but also involved in autophagy. METHODS. We performed a hypothesis-driven analysis of 56 single nucleotide polymorphisms (SNPs) associated with CD susceptibility or known to affect Paneth cell function in 110 Japanese CD patients who underwent ileal resection. We subsequently performed a genome-wide association analysis. Paneth cell phenotype was determined by defensin-5 immunofluorescence. Selected genotype-Paneth cell defect correlations were compared to a Western CD cohort (n = 164). RESULTS. The average percentage of abnormal Paneth cells in Japanese CD was similar to Western CD (P = 0.87), and abnormal Paneth cell phenotype was also associated with early recurrence (P = 0.013). In contrast to Western CD, ATG16L1 T300A was not associated with Paneth cell defect in Japanese CD (P = 0.20). Among the 56 selected SNPs, only LRRK2 M2397T showed significant association with Paneth cell defect (P = 3.62 × 10-4), whereas in the Western CD cohort it was not (P = 0.76). Pathway analysis of LRRK2 and other candidate genes with P less than 5 × 10-4 showed connections with known CD susceptibility genes and links to autophagy and TNF-α networks. CONCLUSIONS. We found dichotomous effects of ATG16L1 and LRRK2 on Paneth cell defect between Japanese and Western CD. Genes affecting Paneth cell phenotype in Japanese CD were also associated with autophagy. Paneth cell phenotype also predicted prognosis in Japanese CD. FUNDING. Helmsley Charitable Trust, Doris Duke Foundation (grant 2014103), Japan Society for the Promotion of Science (KAKENHI grants JP15H04805 and JP15K15284), Crohn's and Colitis Foundation grant 274415, NIH (grants 1R56DK095820, K01DK109081, and UL1 TR000448).
Collapse
Affiliation(s)
- Ta-Chiang Liu
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Takeo Naito
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Zhenqiu Liu
- F. Widjaja Family Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Kelli L VanDussen
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Talin Haritunians
- F. Widjaja Family Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Dalin Li
- F. Widjaja Family Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Katsuya Endo
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yosuke Kawai
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Masao Nagasaki
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Yoshitaka Kinouchi
- Health Administration Center, Center for the Advancement of Higher Education, Tohoku University, Sendai, Japan
| | - Dermot Pb McGovern
- F. Widjaja Family Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Tooru Shimosegawa
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yoichi Kakuta
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Thaddeus S Stappenbeck
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
57
|
Chairatana P, Chiang IL, Nolan EM. Human α-Defensin 6 Self-Assembly Prevents Adhesion and Suppresses Virulence Traits of Candida albicans. Biochemistry 2017; 56:1033-1041. [PMID: 28026958 DOI: 10.1021/acs.biochem.6b01111] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Human α-defensin 6 (HD6) is a host-defense peptide that contributes to intestinal innate immunity and mediates homeostasis at mucosal surfaces by forming noncovalent oligomers that capture bacteria and prevent bacterial invasion of the epithelium. This work illustrates a new role of HD6 in defending the host epithelium against pathogenic microorganisms. We report that HD6 blocks adhesion of Candida albicans to human intestinal epithelial cells and suppresses two C. albicans virulence traits, namely, invasion of human epithelial cells and biofilm formation. Moreover, a comparison of HD6 and a single-point variant F2A that does not form higher-order oligomers demonstrates that the self-assembly properties of HD6 are essential for functional activity against C. albicans. This opportunistic fungal pathogen, which resides in the intestine as a member of the gut microbiota in healthy individuals, can turn virulent and cause a variety of diseases ranging from superficial infections to life-threatening systemic infections. Our results indicate that HD6 may allow C. albicans to persist as a harmless commensal in the gastrointestinal tract. Moreover, HD6 and HD6-inspired molecules may provide a foundation for exploring new antimicrobial strategies that attenuate the virulence traits of C. albicans and other microbial pathogens.
Collapse
Affiliation(s)
- Phoom Chairatana
- Department of Chemistry, Massachusetts Institute of Technology , Cambridge, Massachusetts 02139, United States
| | - I-Ling Chiang
- Department of Chemistry, Massachusetts Institute of Technology , Cambridge, Massachusetts 02139, United States
| | - Elizabeth M Nolan
- Department of Chemistry, Massachusetts Institute of Technology , Cambridge, Massachusetts 02139, United States
| |
Collapse
|
58
|
Ke P, Shao BZ, Xu ZQ, Chen XW, Liu C. Intestinal Autophagy and Its Pharmacological Control in Inflammatory Bowel Disease. Front Immunol 2017; 7:695. [PMID: 28119697 PMCID: PMC5220102 DOI: 10.3389/fimmu.2016.00695] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2016] [Accepted: 12/28/2016] [Indexed: 12/14/2022] Open
Abstract
Intestinal mucosal barrier, mainly composed of the intestinal mucus layer and the epithelium, plays a critical role in nutrient absorption as well as protection from pathogenic microorganisms. It is widely acknowledged that the damage of intestinal mucosal barrier or the disturbance of microorganism balance in the intestinal tract contributes greatly to the pathogenesis and progression of inflammatory bowel disease (IBD), which mainly includes Crohn’s disease and ulcerative colitis. Autophagy is an evolutionarily conserved catabolic process that involves degradation of protein aggregates and damaged organelles for recycling. The roles of autophagy in the pathogenesis and progression of IBD have been increasingly studied. This present review mainly describes the roles of autophagy of Paneth cells, macrophages, and goblet cells in IBD, and finally, several potential therapeutic strategies for IBD taking advantage of autophagy.
Collapse
Affiliation(s)
- Ping Ke
- Department of Pharmacology, Second Military Medical University , Shanghai , China
| | - Bo-Zong Shao
- Department of Pharmacology, Second Military Medical University , Shanghai , China
| | - Zhe-Qi Xu
- Department of Pharmacology, Second Military Medical University , Shanghai , China
| | - Xiong-Wen Chen
- Department of Pharmacology, Second Military Medical University , Shanghai , China
| | - Chong Liu
- Department of Pharmacology, Second Military Medical University , Shanghai , China
| |
Collapse
|
59
|
Abstract
The lining of the gastrointestinal tract needs to be easily accessible to nutrients and, at the same time, defend against pathogens and chemical challenges. This lining is the largest and most vulnerable surface that faces the outside world. To manage the dual problems of effective nutrient conversion and defence, the gut lining has a sophisticated system for detection of individual chemical entities, pathogenic organisms and their products, and physico-chemical properties of its contents. Detection is through specific receptors that signal to the gut endocrine system, the nervous system, the immune system and local tissue defence systems. These effectors, in turn, modify digestive functions and contribute to tissue defence. Receptors for nutrients include taste receptors for sweet, bitter and savoury, free fatty acid receptors, peptide and phytochemical receptors, that are primarily located on enteroendocrine cells. Hormones released by enteroendocrine cells act locally, through the circulation and via the nervous system, to optimise digestion and mucosal health. Pathogen detection is both through antigen presentation to T-cells and through pattern-recognition receptors (PRRs). Activation of PRRs triggers local tissue defence, for example, by causing release of antimicrobials from Paneth cells. Toxic chemicals, including plant toxins, are sensed and then avoided, expelled or metabolised. It continues to be a major challenge to develop a comprehensive understanding of the integrated responses of the gastrointestinal tract to its luminal contents.
Collapse
|
60
|
Collins FL, Schepper JD, Rios-Arce ND, Steury MD, Kang HJ, Mallin H, Schoenherr D, Camfield G, Chishti S, McCabe LR, Parameswaran N. Immunology of Gut-Bone Signaling. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1033:59-94. [PMID: 29101652 PMCID: PMC5749247 DOI: 10.1007/978-3-319-66653-2_5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In recent years a link between the gastrointestinal tract and bone health has started to gain significant attention. Dysbiosis of the intestinal microbiota has been linked to the pathology of a number of diseases which are associated with bone loss. In addition modulation of the intestinal microbiota with probiotic bacteria has revealed to have both beneficial local and systemic effects. In the present chapter, we discuss the intestinal and bone immune systems, explore how intestinal disease affects the immune system, and examine how these pathologic changes could adversely impact bone health.
Collapse
Affiliation(s)
- Fraser L Collins
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | | | - Naiomy Deliz Rios-Arce
- Department of Physiology, Michigan State University, East Lansing, MI, USA
- Comparative Medicine and Integrative Biology Program, East Lansing, MI, USA
| | - Michael D Steury
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Ho Jun Kang
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Heather Mallin
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Daniel Schoenherr
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Glen Camfield
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Saima Chishti
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Laura R McCabe
- Department of Physiology and Department of Radiology, Biomedical Imaging Research Centre, Michigan State University, East Lansing, MI, USA.
| | - Narayanan Parameswaran
- Department of Physiology, Michigan State University, East Lansing, MI, USA.
- Comparative Medicine and Integrative Biology Program, East Lansing, MI, USA.
| |
Collapse
|
61
|
Barron L, Sun RC, Aladegbami B, Erwin CR, Warner BW, Guo J. Intestinal Epithelial-Specific mTORC1 Activation Enhances Intestinal Adaptation After Small Bowel Resection. Cell Mol Gastroenterol Hepatol 2016; 3:231-244. [PMID: 28275690 PMCID: PMC5331783 DOI: 10.1016/j.jcmgh.2016.10.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 10/18/2016] [Indexed: 01/21/2023]
Abstract
BACKGROUND & AIMS Intestinal adaptation is a compensatory response to the massive loss of small intestine after surgical resection. We investigated the role of intestinal epithelial cell-specific mammalian target of rapamycin complex 1 (i-mTORC1) in intestinal adaptation after massive small bowel resection (SBR). METHODS We performed 50% proximal SBR on mice to study adaptation. To manipulate i-mTORC1 activity, Villin-CreER transgenic mice were crossed with tuberous sclerosis complex (TSC)1flox/flox or Raptorflox/flox mice to inducibly activate or inactivate i-mTORC1 activity with tamoxifen. Western blot was used to confirm the activity of mTORC1. Crypt depth and villus height were measured to score adaptation. Immunohistochemistry was used to investigate differentiation and rates of crypt proliferation. RESULTS After SBR, mice treated with systemic rapamycin showed diminished structural adaptation, blunted crypt cell proliferation, and significant body weight loss. Activating i-mTORC1 via TSC1 deletion induced larger hyperproliferative crypts and disorganized Paneth cells without a significant change in villus height. After SBR, ablating TSC1 in intestinal epithelium induced a robust villus growth with much stronger crypt cell proliferation, but similar body weight recovery. Acute inactivation of i-mTORC1 through deletion of Raptor did not change crypt cell proliferation or mucosa structure, but significantly reduced lysozyme/matrix metalloproteinase-7-positive Paneth cell and goblet cell numbers, with increased enteroendocrine cells. Surprisingly, ablation of intestinal epithelial cell-specific Raptor after SBR did not affect adaptation or crypt proliferation, but dramatically reduced body weight recovery after surgery. CONCLUSIONS Systemic, but not intestinal-specific, mTORC1 is important for normal adaptation responses to SBR. Although not required, forced enterocyte mTORC1 signaling after resection causes an enhanced adaptive response.
Collapse
Key Words
- Differentiation
- EGF, epidermal growth factor
- IHC, immunohistochemistry
- MMP, matrix metalloproteinase
- PCR, polymerase chain reaction
- Raptor
- S6K, S6 kinase
- SBR, small bowel resection
- TAM, tamoxifen
- TSC, tuberous sclerosis complex
- TSC1
- WT, wild type
- i-TSC-/-, intestinal epithelial cell–specific tuberous sclerosis complex 1 null mice
- mTOR, mammalian target of rapamycin
- mTORC, mammalian target of rapamycin complex
- p-HH3, phosphorylated histone H3
Collapse
Affiliation(s)
| | | | | | | | | | - Jun Guo
- Correspondence Address correspondence to: Jun Guo, PhD, BJC Institute of Health Room 7118, 425 South Euclid Avenue, St. Louis, Missouri 63110. fax: (314) 747–0610.BJC Institute of Health Room 7118425 South Euclid AvenueSt. LouisMissouri 63110
| |
Collapse
|
62
|
Yu T, Yang HS, Lu XJ, Xia ZS, Ouyang H, Shan TD, Huang CZ, Chen QK. Association of Bactericidal Dysfunction of Paneth Cells in Streptozocin-Induced Diabetic Mice with Insulin Deficiency. Med Sci Monit 2016; 22:3062-72. [PMID: 27572949 PMCID: PMC5013981 DOI: 10.12659/msm.897270] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Type 1 diabetes mellitus (T1DM) is associated with increased risks of enteric infection. Paneth cells constitute the first line of the gut defense. Little is known about the impact of T1DM on the bactericidal function of intestinal Paneth cells. MATERIAL AND METHODS A T1DM mouse model was induced by intraperitoneal injection of streptozocin. The analysis of intestinal microbiota and the mucosal bactericidal assay were conducted to evaluate intestinal innate defense. Numbers of Paneth cells and their expression of related antimicrobial peptides were analyzed. Expression of total insulin receptor (IR) mRNA and relative levels of IR-A/IR-B were analyzed. The primary mouse small intestinal crypt culture was used to analyze the effect of insulin and glucose on the expression of related antimicrobial peptides of Paneth cells. RESULTS In T1DM mice, bacterial loads were increased and there was an alteration in the composition of the intestinal microflora. Exogenous bacteria had better survival in the small bowel of the T1DM mice. The expression of Paneth cell-derived antimicrobial peptides was significantly decreased in the T1DM mice, although the number of Paneth cells was increased. Relative levels of IR-A/IR-B in Paneth cells of diabetic mice were elevated, but the total IR mRNA did not change. Insulin treatment restored the expression of antimicrobial peptides and normalized the microbiota in the gut of T1DM mice. Subsequently, in vitro culture assay demonstrated that insulin rather than glucose was essential for the optimal expression of Paneth cell-derived antimicrobial peptides. CONCLUSIONS The bactericidal function of intestinal Paneth cells was impaired in STZ-induced diabetic mice, resulting in the altered intestinal flora, and insulin was essential for the optimal expression of Paneth cell-derived antimicrobial peptides.
Collapse
Affiliation(s)
- Tao Yu
- Department of Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China (mainland)
| | - Hong-Sheng Yang
- Department of Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China (mainland)
| | - Xi-Ji Lu
- Department of Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China (mainland)
| | - Zhong-Sheng Xia
- Department of Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China (mainland)
| | - Hui Ouyang
- Department of Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China (mainland)
| | - Ti-Dong Shan
- Department of Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China (mainland)
| | - Can-Ze Huang
- Department of Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China (mainland)
| | - Qi-Kui Chen
- Department of Gastroenterology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China (mainland)
| |
Collapse
|
63
|
Elshaer D, Begun J. The role of barrier function, autophagy, and cytokines in maintaining intestinal homeostasis. Semin Cell Dev Biol 2016; 61:51-59. [PMID: 27565684 DOI: 10.1016/j.semcdb.2016.08.018] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 08/10/2016] [Accepted: 08/22/2016] [Indexed: 02/06/2023]
Abstract
Intestinal homeostasis is maintained through the interplay of the intestinal mucosa, local and systemic immune factors, and the microbial content of the gut. The cellular processes of autophagy, endoplasmic reticulum stress, the unfolded protein response and regulation of reactive oxygen species production are required to maintain a balance between pro-inflammatory responses against potential pathogens and a tolerogenic response towards commensal bacteria. Intestinally active cytokines regulate innate immune pathways and cellular pathways within the gut mucosa. Disruption of these processes, or alterations in the cytokine milieu, can result in an improper response to the commensal gut microbial community leading to inappropriate inflammation characteristic of conditions such as inflammatory bowel disease.
Collapse
Affiliation(s)
- Dana Elshaer
- Immunity, Infection and Inflammation Program, Mater Research Institute - University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia.
| | - Jakob Begun
- Immunity, Infection and Inflammation Program, Mater Research Institute - University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia; University of Queensland School of Medicine, Brisbane, Queensland, Australia.
| |
Collapse
|
64
|
Vlantis K, Wullaert A, Polykratis A, Kondylis V, Dannappel M, Schwarzer R, Welz P, Corona T, Walczak H, Weih F, Klein U, Kelliher M, Pasparakis M. NEMO Prevents RIP Kinase 1-Mediated Epithelial Cell Death and Chronic Intestinal Inflammation by NF-κB-Dependent and -Independent Functions. Immunity 2016; 44:553-567. [PMID: 26982364 PMCID: PMC4803910 DOI: 10.1016/j.immuni.2016.02.020] [Citation(s) in RCA: 159] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Revised: 10/25/2015] [Accepted: 12/14/2015] [Indexed: 12/25/2022]
Abstract
Intestinal epithelial cells (IECs) regulate gut immune homeostasis, and impaired epithelial responses are implicated in the pathogenesis of inflammatory bowel diseases (IBD). IEC-specific ablation of nuclear factor κB (NF-κB) essential modulator (NEMO) caused Paneth cell apoptosis and impaired antimicrobial factor expression in the ileum, as well as colonocyte apoptosis and microbiota-driven chronic inflammation in the colon. Combined RelA, c-Rel, and RelB deficiency in IECs caused Paneth cell apoptosis but not colitis, suggesting that NEMO prevents colon inflammation by NF-κB-independent functions. Inhibition of receptor-interacting protein kinase 1 (RIPK1) kinase activity or combined deficiency of Fas-associated via death domain protein (FADD) and RIPK3 prevented epithelial cell death, Paneth cell loss, and colitis development in mice with epithelial NEMO deficiency. Therefore, NEMO prevents intestinal inflammation by inhibiting RIPK1 kinase activity-mediated IEC death, suggesting that RIPK1 inhibitors could be effective in the treatment of colitis in patients with NEMO mutations and possibly in IBD.
Collapse
Affiliation(s)
- Katerina Vlantis
- Institute for Genetics, University of Cologne, 50674 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany; Center for Molecular Medicine (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Andy Wullaert
- Institute for Genetics, University of Cologne, 50674 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany; Center for Molecular Medicine (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Apostolos Polykratis
- Institute for Genetics, University of Cologne, 50674 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany; Center for Molecular Medicine (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Vangelis Kondylis
- Institute for Genetics, University of Cologne, 50674 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany; Center for Molecular Medicine (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Marius Dannappel
- Institute for Genetics, University of Cologne, 50674 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany; Center for Molecular Medicine (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Robin Schwarzer
- Institute for Genetics, University of Cologne, 50674 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany; Center for Molecular Medicine (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Patrick Welz
- Institute for Genetics, University of Cologne, 50674 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany; Center for Molecular Medicine (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Teresa Corona
- Institute for Genetics, University of Cologne, 50674 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany; Center for Molecular Medicine (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Henning Walczak
- Centre for Cell Death, Cancer and Inflammation (CCCI), UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6D, UK
| | - Falk Weih
- Leibniz-Institute for Age Research, Fritz-Lipmann-Institute, 07745 Jena, Germany
| | - Ulf Klein
- Herbert Irving Comprehensive Cancer Center, Departments of Pathology & Cell Biology and Microbiology & Immunology, Columbia University, New York, NY 10032, USA
| | - Michelle Kelliher
- Department of Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Manolis Pasparakis
- Institute for Genetics, University of Cologne, 50674 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany; Center for Molecular Medicine (CMMC), University of Cologne, 50931 Cologne, Germany.
| |
Collapse
|
65
|
Wang L, Li J, Li J, Li R, Lv C, Li S, Mi Y, Zhang C. Identification of the Paneth cells in chicken small intestine. Poult Sci 2016; 95:1631-1635. [DOI: 10.3382/ps/pew079] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 02/01/2016] [Indexed: 12/30/2022] Open
|
66
|
Grootjans J, Lenaerts K, Buurman WA, Dejong CHC, Derikx JPM. Life and death at the mucosal-luminal interface: New perspectives on human intestinal ischemia-reperfusion. World J Gastroenterol 2016; 22:2760-2770. [PMID: 26973414 PMCID: PMC4777998 DOI: 10.3748/wjg.v22.i9.2760] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Revised: 09/24/2015] [Accepted: 12/14/2015] [Indexed: 02/06/2023] Open
Abstract
Intestinal ischemia is a frequently observed phenomenon. Morbidity and mortality rates are extraordinarily high and did not improve over the past decades. This is in part attributable to limited knowledge on the pathophysiology of intestinal ischemia-reperfusion (IR) in man, the paucity in preventive and/or therapeutic options and the lack of early diagnostic markers for intestinal ischemia. To improve our knowledge and solve clinically important questions regarding intestinal IR, we developed a human experimental intestinal IR model. With this model, we were able to gain insight into the mechanisms that allow the human gut to withstand short periods of IR without the development of severe inflammatory responses. The purpose of this review is to overview the most relevant recent advances in our understanding of the pathophysiology of human intestinal IR, as well as the (potential) future clinical implications.
Collapse
|
67
|
Affiliation(s)
- Johan H van Es
- Hubrecht Institute-KNAW & University Medical Center, Utrecht, Uppsalalaan 8, 3584 CT Utrecht, Netherlands
| | - Hans Clevers
- Hubrecht Institute-KNAW & University Medical Center, Utrecht, Uppsalalaan 8, 3584 CT Utrecht, Netherlands.
| |
Collapse
|
68
|
Nolte T, Brander-Weber P, Dangler C, Deschl U, Elwell MR, Greaves P, Hailey R, Leach MW, Pandiri AR, Rogers A, Shackelford CC, Spencer A, Tanaka T, Ward JM. Nonproliferative and Proliferative Lesions of the Gastrointestinal Tract, Pancreas and Salivary Glands of the Rat and Mouse. J Toxicol Pathol 2016; 29:1S-125S. [PMID: 26973378 PMCID: PMC4765498 DOI: 10.1293/tox.29.1s] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The INHAND (International Harmonization of Nomenclature and Diagnostic Criteria for Lesions in Rats and Mice) project is a joint initiative of the Societies of Toxicologic Pathology from Europe (ESTP), Great Britain (BSTP), Japan (JSTP), and North America (STP) to develop an internationally accepted nomenclature and diagnostic criteria for nonproliferative and proliferative lesions in laboratory animals. The purpose of this publication is to provide a standardized nomenclature and diagnostic criteria for classifying lesions in the digestive system including the salivary glands and the exocrine pancreas of laboratory rats and mice. Most lesions are illustrated by color photomicrographs. The standardized nomenclature, the diagnostic criteria, and the photomicrographs are also available electronically on the Internet (http://www.goreni.org/). Sources of material included histopathology databases from government, academia, and industrial laboratories throughout the world. Content includes spontaneous and age related lesions as well as lesions induced by exposure to test items. Relevant infectious and parasitic lesions are included as well. A widely accepted and utilized international harmonization of nomenclature and diagnostic criteria for the digestive system will decrease misunderstandings among regulatory and scientific research organizations in different countries and provide a common language to increase and enrich international exchanges of information among toxicologists and pathologists.
Collapse
Affiliation(s)
- Thomas Nolte
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an
der Riss, 88397, Germany
- Chairman of the Digestive Tract INHAND Committee
| | - Patricia Brander-Weber
- Novartis Institutes for BioMedical Research, Novartis Pharma
AG, CH-4002 Basel, Switzerland
| | - Charles Dangler
- Jackson Laboratory, Bar Harbor, Maine 04609, USA.
Present: Sanofi5 The Mountain Road, Framingham, Massachusetts 01740,
USA
| | - Ulrich Deschl
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an
der Riss, 88397, Germany
| | - Michael R. Elwell
- Covance Laboratories, Inc. 14500 Avion Parkway, Ste 125,
Chantilly, Virginia 20151, USA
| | - Peter Greaves
- University of Leicester, Department of Cancer Studies and
Molecular Medicine, Robert Kilpatrick Clinical Science Building, Leicester Royal
Infirmary, Leicester LE2 7LX, United Kingdom
| | - Richard Hailey
- GlaxoSmithKline PO Box 14164 Durham, North Carolina 27709,
USA
| | | | - Arun R. Pandiri
- Cellular and Molecular Pathology Branch, National Toxicology
Program, National Institute of Environmental Health Sciences, Research Triangle Park,
North Carolina 27709, USA
- Experimental Pathology Laboratories, Inc. PO Box 12766,
Research Triangle Park, North Carolina 27709, USA
| | - Arlin Rogers
- Tufts University, Department of Biomedical Sciences, 274
Tremont Street, Massachusetts 02111, USA
| | - Cynthia C. Shackelford
- Cellular and Molecular Pathology Branch, National Toxicology
Program, National Institute of Environmental Health Sciences, Research Triangle Park,
North Carolina 27709, USA
| | - Andrew Spencer
- Covance Laboratories Ltd, Alnwick Research Centre,
Willowburn Avenue, Alnwick, Northumberland NE66 2JH United Kingdom
| | | | | |
Collapse
|
69
|
Tremblay É, Thibault MP, Ferretti E, Babakissa C, Bertelle V, Bettolli M, Burghardt KM, Colombani JF, Grynspan D, Levy E, Lu P, Mayer S, Ménard D, Mouterde O, Renes IB, Seidman EG, Beaulieu JF. Gene expression profiling in necrotizing enterocolitis reveals pathways common to those reported in Crohn's disease. BMC Med Genomics 2016; 9:6. [PMID: 26801768 PMCID: PMC4722613 DOI: 10.1186/s12920-016-0166-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 01/18/2016] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Necrotizing enterocolitis (NEC) is the most frequent life-threatening gastrointestinal disease experienced by premature infants in neonatal intensive care units. The challenge for neonatologists is to detect early clinical manifestations of NEC. One strategy would be to identify specific markers that could be used as early diagnostic tools to identify preterm infants most at risk of developing NEC or in the event of a diagnostic dilemma of suspected disease. As a first step in this direction, we sought to determine the specific gene expression profile of NEC. METHODS Deep sequencing (RNA-Seq) was used to establish the gene expression profiles in ileal samples obtained from preterm infants diagnosed with NEC and non-NEC conditions. Data were analyzed with Ingenuity Pathway Analysis and ToppCluster softwares. RESULTS Data analysis indicated that the most significant functional pathways over-represented in NEC neonates were associated with immune functions, such as altered T and B cell signaling, B cell development, and the role of pattern recognition receptors for bacteria and viruses. Among the genes that were strongly modulated in neonates with NEC, we observed a significant degree of similarity when compared with those reported in Crohn's disease, a chronic inflammatory bowel disease. CONCLUSIONS Gene expression profile analysis revealed a predominantly altered immune response in the intestine of NEC neonates. Moreover, comparative analysis between NEC and Crohn's disease gene expression repertoires revealed a surprisingly high degree of similarity between these two conditions suggesting a new avenue for identifying NEC biomarkers.
Collapse
Affiliation(s)
- Éric Tremblay
- Department of Anatomy and Cell Biology, Faculté de Médecine et Sciences de la Santé, Université de Sherbrooke, 3001, 12th Avec North, J1H 5N4, Sherbrooke, QC, Canada.
| | - Marie-Pier Thibault
- Department of Anatomy and Cell Biology, Faculté de Médecine et Sciences de la Santé, Université de Sherbrooke, 3001, 12th Avec North, J1H 5N4, Sherbrooke, QC, Canada.
| | - Emanuela Ferretti
- Division of Neonatology, Department of Pediatrics, CHEO, Ottawa, ON, Canada.
| | - Corentin Babakissa
- Department of Pediatrics, Faculté de Médecine et Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada.
| | - Valérie Bertelle
- Division of Neonatology, Department of Pediatrics, Faculté de Médecine et Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada.
| | | | | | | | - David Grynspan
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.
| | - Emile Levy
- Department of Nutrition, Centre de recherche, CHU Sainte-Justine, Université de Montréal, Montréal, QC, Canada.
| | - Peng Lu
- Department of Pediatrics, Erasmus MC-Sophia, Rotterdam, The Netherland.
| | - Sandeep Mayer
- Department of Surgery, Faculté de Médecine et Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada.
| | - Daniel Ménard
- Department of Anatomy and Cell Biology, Faculté de Médecine et Sciences de la Santé, Université de Sherbrooke, 3001, 12th Avec North, J1H 5N4, Sherbrooke, QC, Canada.
| | | | - Ingrid B Renes
- Department of Pediatrics, Erasmus MC-Sophia, Rotterdam, The Netherland.
- Emma Children's Hospital-AMC, Amsterdam, The Netherlands.
| | - Ernest G Seidman
- Division of Gastroenterology, McGill University, Montréal, QC, Canada.
| | - Jean-François Beaulieu
- Department of Anatomy and Cell Biology, Faculté de Médecine et Sciences de la Santé, Université de Sherbrooke, 3001, 12th Avec North, J1H 5N4, Sherbrooke, QC, Canada.
| |
Collapse
|
70
|
Chairatana P, Chu H, Castillo PA, Shen B, Bevins CL, Nolan EM. Proteolysis Triggers Self-Assembly and Unmasks Innate Immune Function of a Human α-Defensin Peptide. Chem Sci 2015; 7:1738-1752. [PMID: 27076903 PMCID: PMC4827351 DOI: 10.1039/c5sc04194e] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Human α-defensin 6 (HD6) is a unique peptide of the defensin family that provides innate immunity in the intestine by self-assembling to form high-order oligomers that entrap bacteria and prevent host cell invasion. Here, we report critical steps in the self-assembly pathway of HD6. We demonstrate that HD6 is localized in secretory granules of small intestinal Paneth cells. HD6 is stored in these granules as an 81-residue propeptide (proHD6), and is recovered from ileal lumen as a 32-residue mature peptide. The propeptide neither forms higher-order oligomers, nor agglutinates bacteria, nor prevents Listeria monocytogenes invasion into epithelial cells. The Paneth cell granules also contain the protease trypsin, and trypsin-catalyzed hydrolysis of proHD6 liberates mature HD6, unmasking its latent activities. This work illustrates a remarkable example of how nature utilizes a propeptide strategy to spatially and temporally control peptide self-assembly, and thereby initiates innate immune function in the human intestine.
Collapse
Affiliation(s)
- Phoom Chairatana
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Hiutung Chu
- Department of Microbiology and Immunology, University of California Davis School of Medicine, Davis, CA 95616, USA
| | - Patricia A Castillo
- Department of Microbiology and Immunology, University of California Davis School of Medicine, Davis, CA 95616, USA
| | - Bo Shen
- Department of General Internal Medicine and Gastroenterology and Hepatology, The Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | - Charles L Bevins
- Department of Microbiology and Immunology, University of California Davis School of Medicine, Davis, CA 95616, USA
| | - Elizabeth M Nolan
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
71
|
Roman M, Madkan VK, Chiu MW. Profile of secukinumab in the treatment of psoriasis: current perspectives. Ther Clin Risk Manag 2015; 11:1767-77. [PMID: 26664127 PMCID: PMC4671779 DOI: 10.2147/tcrm.s79053] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Secukinumab (Cosentyx™) is a human monoclonal IgG1k antibody that has been developed to target and block the actions of IL-17A. It is known that this cytokine is elevated in lesions of psoriasis. Interleukins in the Th17 pathway play a pivotal role in the pathogenesis of psoriasis and have thus become targets for recent biologic drug development. As a monoclonal antibody immune modulator, secukinumab exhibits the expected pharmacokinetic properties of slow subcutaneous absorption, low clearance, and long half-life, although formal studies examining the impact of impaired hepatic or renal function on the overall pharmacokinetic profile have not been conducted. Both Phase II and III clinical trials have demonstrated the effectiveness of secukinumab in the treatment of moderate-to-severe plaque psoriasis, psoriatic arthritis, rheumatoid arthritis, ankylosing spondylitis, and noninfectious uveitis. In June 2015, secukinumab was approved by the US Food and Drug Administration for the treatment of adults with moderate-to-severe plaque psoriasis, with a wealth of clinical trials showcasing its efficacy in improving psoriasis area and severity index scores, and it is superior to other comparable biologics on the market, including the TNF inhibitor etanercept. As such, this review focuses on the marquee clinical trials involving secukinumab treatment of plaque psoriasis, while also exploring this drug's efficacy in treating patients with psoriatic arthritis, a disease that has a well-documented comorbidity in patients diagnosed with moderate-to-severe plaque psoriasis. Finally, the safety and tolerability of this drug in a variety of clinical trials to date have also been reviewed, and will undoubtedly have a large impact on this drug's postmarketing surveillance and future studies regarding its long-term safety.
Collapse
Affiliation(s)
- Michael Roman
- Division of Dermatology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Vandana K Madkan
- Division of Dermatology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Melvin W Chiu
- Division of Dermatology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| |
Collapse
|
72
|
Nakamura K, Sakuragi N, Takakuwa A, Ayabe T. Paneth cell α-defensins and enteric microbiota in health and disease. BIOSCIENCE OF MICROBIOTA FOOD AND HEALTH 2015; 35:57-67. [PMID: 27200259 PMCID: PMC4858879 DOI: 10.12938/bmfh.2015-019] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 10/25/2015] [Indexed: 12/12/2022]
Abstract
Antimicrobial peptides are major effectors of innate immunity of multicellular organisms including humans and play a critical role in host defense, and their importance is widely recognized. The epithelium of the intestine is the largest surface area exposed to the outer environment, including pathogens, toxins and foods. The Paneth cell lineage of intestinal epithelial cells produces and secretes α-defensin antimicrobial peptides and functions in innate enteric immunity by removing pathogens and living symbiotically with commensal microbiota to contribute to intestinal homeostasis. Paneth cells secrete α-defensins, HD5 and HD6 in humans and cryptdins in mice, in response to bacterial, cholinergic and other stimuli. The α-defensins have selective activities against bacteria, eliciting potent microbicidal activities against pathogenic bacteria but minimal or no bactericidal activity against commensal bacteria. Therefore, α-defensins regulate the composition of the intestinal microbiota in vivo and play a role in homeostasis of the entire intestine. Recently, relationships between dysbiosis, or abnormal composition of the intestinal microbiota, and diseases such as inflammatory bowel disease and lifestyle diseases including obesity and atherosclerosis have been reported. Because α-defensins regulate the composition of the intestinal microbiota, Paneth cells and their α-defensins may have a key role as one mechanism linking the microbiota and disease.
Collapse
Affiliation(s)
- Kiminori Nakamura
- Department of Cell Biological Science, Graduate School of Life Science, Faculty of Advanced Life Science, Hokkaido University, Kita-21, Nishi-11, Kita-ku, Sapporo, Hokkaido 001-0021, Japan
| | - Naoya Sakuragi
- Department of Cell Biological Science, Graduate School of Life Science, Faculty of Advanced Life Science, Hokkaido University, Kita-21, Nishi-11, Kita-ku, Sapporo, Hokkaido 001-0021, Japan
| | - Akiko Takakuwa
- Department of Cell Biological Science, Graduate School of Life Science, Faculty of Advanced Life Science, Hokkaido University, Kita-21, Nishi-11, Kita-ku, Sapporo, Hokkaido 001-0021, Japan; Department of Nutrition, Faculty of Nursing and Nutrition, Tenshi College, 3-1-30 Higashi, Kita-13, Higashi-ku, Sapporo, Hokkaido 065-0013, Japan
| | - Tokiyoshi Ayabe
- Department of Cell Biological Science, Graduate School of Life Science, Faculty of Advanced Life Science, Hokkaido University, Kita-21, Nishi-11, Kita-ku, Sapporo, Hokkaido 001-0021, Japan
| |
Collapse
|
73
|
The enteric nervous system neuropeptide, bombesin, reverses innate immune impairments during parenteral nutrition. Ann Surg 2015; 260:432-43; discussion 443-4. [PMID: 25115419 DOI: 10.1097/sla.0000000000000871] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Lack of enteral stimulation during parenteral nutrition (PN) impairs mucosal immunity. Bombesin (BBS), a gastrin-releasing peptide analogue, reverses PN-induced defects in acquired immunity. Paneth cells produce antimicrobial peptides (AMPs) of innate immunity for release after cholinergic stimulation. OBJECTIVE Determine if BBS restores AMPs and bactericidal function during PN. METHODS Intravenously cannulated male ICR mice were randomized to Chow, PN, or PN+BBS (15 μg 3 times daily, n = 7 per group) for 5 days. Ileum was analyzed for AMPs (Protein: sPLA2 by fluorescence, lysozyme and RegIII-γ by western andcryptdin-4 by ELISA; mRNA: all by RT-PCR). Cholinergic stimulated (100 μM bethanechol) ileal specimens assessed Pseudomonas bactericidal activity. Ileum (Chow: n = 7; PN: n = 9; PN+BBS: n = 8) was assessed for Escherichia coli invasion in ex-vivo culture. RESULTS PN significantly decreased most AMPs versus Chow while BBS maintained Chow levels (sPLA2: Chow: 107 + 14*, PN: 44.6 + 7.2, PN+BBS: 78.7 + 13.4* Fl/min/μL/total protein; Lysozyme: Chow: 63.9 + 11.9*, PN: 26.8 + 6.2; PN+BBS: 64.9 + 13.8* lysozyme/total protein; RegIII-γ: Chow: 51.5 + 10.0*, PN: 20.4 + 4.3, PN+BBS: 31.0 + 8.4 RegIII-γ/total protein; Cryptdin-4: Chow: 18.4 + 1.5*, PN: 12.7 + 1.6, PN+BBS: 26.1 + 2.4*† pg/mg [all *P < 0.05 vs PN and †P < 0.05 vs Chow]). Functionally, BBS prevented PN loss of bactericidal activity after cholinergic stimulation (Chow: 25.3 + 3.6*, PN: 13.0 + 3.2; PN+BBS: 27.0 + 4.7* percent bacterial killing, *P < 0.05 vs PN). BBS reduced bacterial invasion in unstimulated tissue barely missing significance (P = 0.06). CONCLUSIONS The enteric nervous system (ENS) controls AMP levels in Paneth cells during PN but mucosal protection by innate immunity requires both ENS and parasympathetic stimulation.
Collapse
|
74
|
Abstract
Recurrence of Crohn's disease (CD) after ileal or colonic resection is common. Many studies have tried to identify predictors of postoperative recurrence (POR) in CD. A wide range of histologic features have been identified, but for most of them, the literature provided conflicting data. In last years, several studies have suggested that histologic findings including inflammatory changes within the enteric nervous system of the resection margin may be associated with CD recurrence. Herein, after briefly summarizing pathophysiology of POR, we review all histological features that have been studied so far: granulomas, histologic appearance at the margin of resection, plexitis, lymphatic vessel density in proximal margin of resection, and morphological analysis of Paneth cells. Granulomas and chronic inflammation at the margin of resection do not seem to predict POR in CD. Active disease at the margin of resection, plexitis, lymphatic vessels density, morphological analysis of Paneth cells may predict POR. Most of these histological features await replication in independent studies. Available evidence indicates that histological findings may be taken into account when developing strategies aimed at preventing postoperative CD recurrence.
Collapse
|
75
|
Pelaseyed T, Bergström JH, Gustafsson JK, Ermund A, Birchenough GMH, Schütte A, van der Post S, Svensson F, Rodríguez-Piñeiro AM, Nyström EEL, Wising C, Johansson MEV, Hansson GC. The mucus and mucins of the goblet cells and enterocytes provide the first defense line of the gastrointestinal tract and interact with the immune system. Immunol Rev 2015; 260:8-20. [PMID: 24942678 DOI: 10.1111/imr.12182] [Citation(s) in RCA: 909] [Impact Index Per Article: 90.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The gastrointestinal tract is covered by mucus that has different properties in the stomach, small intestine, and colon. The large highly glycosylated gel-forming mucins MUC2 and MUC5AC are the major components of the mucus in the intestine and stomach, respectively. In the small intestine, mucus limits the number of bacteria that can reach the epithelium and the Peyer's patches. In the large intestine, the inner mucus layer separates the commensal bacteria from the host epithelium. The outer colonic mucus layer is the natural habitat for the commensal bacteria. The intestinal goblet cells secrete not only the MUC2 mucin but also a number of typical mucus components: CLCA1, FCGBP, AGR2, ZG16, and TFF3. The goblet cells have recently been shown to have a novel gate-keeping role for the presentation of oral antigens to the immune system. Goblet cells deliver small intestinal luminal material to the lamina propria dendritic cells of the tolerogenic CD103(+) type. In addition to the gel-forming mucins, the transmembrane mucins MUC3, MUC12, and MUC17 form the enterocyte glycocalyx that can reach about a micrometer out from the brush border. The MUC17 mucin can shuttle from a surface to an intracellular vesicle localization, suggesting that enterocytes might control and report epithelial microbial challenge. There is communication not only from the epithelial cells to the immune system but also in the opposite direction. One example of this is IL10 that can affect and improve the properties of the inner colonic mucus layer. The mucus and epithelial cells of the gastrointestinal tract are the primary gate keepers and controllers of bacterial interactions with the host immune system, but our understanding of this relationship is still in its infancy.
Collapse
Affiliation(s)
- Thaher Pelaseyed
- Department of Medical Biochemistry, University of Gothenburg, Gothenburg, Sweden
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
76
|
Hosomi S, Kaser A, Blumberg RS. Role of endoplasmic reticulum stress and autophagy as interlinking pathways in the pathogenesis of inflammatory bowel disease. Curr Opin Gastroenterol 2015; 31:81-8. [PMID: 25426970 PMCID: PMC4592163 DOI: 10.1097/mog.0000000000000144] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW The purpose of this study is to provide an overview of the role of endoplasmic reticulum (ER) stress and the unfolded protein response (UPR) in inflammatory bowel disease (IBD). RECENT FINDINGS Human genetic studies have identified several UPR-related genes and autophagy-related genes as IBD risk loci. Impairment of each branch of the UPR causes spontaneous enteritis or creates higher susceptibility for intestinal inflammation in model systems. Deficiency of either UPR or autophagy in small intestinal epithelial cells promotes each other's compensatory engagement, which is especially prominent in Paneth cells such that, in the absence of both, severe spontaneous enteritis emerges. SUMMARY Interactions between the UPR and autophagy exhibit critical synergistic interactions within the intestinal epithelium and especially Paneth cells that are of considerable importance to the maintenance of homeostasis. When dysfunctional in the Paneth cell, spontaneous inflammation can emerge that may extend beyond the epithelium providing direct experimental evidence that subsets of Crohn's disease may emanate from primary Paneth cell disturbances.
Collapse
Affiliation(s)
- Shuhei Hosomi
- Division of Gastroenterology, Hepatology, and Endoscopy, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Arthur Kaser
- Division of Gastroenterology and Hepatology, Department of Medicine, Addenbrooke’s Hospital, University of Cambridge, Cambridge, UK
| | - Richard S. Blumberg
- Division of Gastroenterology, Hepatology, and Endoscopy, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
77
|
|
78
|
Mastroianni JR, Lu W, Selsted ME, Ouellette AJ. Differential Susceptibility of Bacteria to Mouse Paneth Cell α-Defensins under Anaerobic Conditions. Antibiotics (Basel) 2014; 3:493-508. [PMID: 25383215 PMCID: PMC4220453 DOI: 10.3390/antibiotics3040493] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Small intestinal Paneth cells secrete α-defensin peptides, termed cryptdins (Crps) in mice, into the intestinal lumen, where they confer immunity to oral infections and define the composition of the ileal microbiota. In these studies, facultative bacteria maintained under aerobic or anaerobic conditions displayed differential sensitivities to mouse α-defensins under in vitro assay conditions. Regardless of oxygenation, Crps 2 and 3 had robust and similar bactericidal activities against S. typhimurium and S. flexneri, but Crp4 activity against S. flexneri was attenuated in the absence of oxygen. Anaerobic bacteria varied in their susceptibility to Crps 2-4, with Crp4 showing less activity than Crps 2 and 3 against Enterococcus faecalis, and Bacteroides fragilis in anaerobic assays, but Fusobacterium necrophorum was killed only by Crp4 and not by Crps 2 and 3. The influence of anaerobiosis in modulating Crp bactericidal activities in vitro suggests that α-defensin effects on the enteric microbiota may be subject to regulation by local oxygen tension.
Collapse
Affiliation(s)
- Jennifer R. Mastroianni
- Department of Pathology and Laboratory Medicine, Keck School of Medicine of the University of Southern California, USC Norris Cancer Center, Los Angeles, CA 90089-9601, USA; E-Mails: (J.R.M.); (M.E.S.)
| | - Wuyuan Lu
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Institute of Human Virology, Baltimore, MD 21201, USA; E-Mail:
| | - Michael E. Selsted
- Department of Pathology and Laboratory Medicine, Keck School of Medicine of the University of Southern California, USC Norris Cancer Center, Los Angeles, CA 90089-9601, USA; E-Mails: (J.R.M.); (M.E.S.)
| | - André J. Ouellette
- Department of Pathology and Laboratory Medicine, Keck School of Medicine of the University of Southern California, USC Norris Cancer Center, Los Angeles, CA 90089-9601, USA; E-Mails: (J.R.M.); (M.E.S.)
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-323-442-7959
| |
Collapse
|
79
|
Abstract
The intestine represents the largest compartment of the immune system. It is continually exposed to antigens and immunomodulatory agents from the diet and the commensal microbiota, and it is the port of entry for many clinically important pathogens. Intestinal immune processes are also increasingly implicated in controlling disease development elsewhere in the body. In this Review, we detail the anatomical and physiological distinctions that are observed in the small and large intestines, and we suggest how these may account for the diversity in the immune apparatus that is seen throughout the intestine. We describe how the distribution of innate, adaptive and innate-like immune cells varies in different segments of the intestine and discuss the environmental factors that may influence this. Finally, we consider the implications of regional immune specialization for inflammatory disease in the intestine.
Collapse
|
80
|
Loonen LMP, Stolte EH, Jaklofsky MTJ, Meijerink M, Dekker J, van Baarlen P, Wells JM. REG3γ-deficient mice have altered mucus distribution and increased mucosal inflammatory responses to the microbiota and enteric pathogens in the ileum. Mucosal Immunol 2014; 7:939-47. [PMID: 24345802 DOI: 10.1038/mi.2013.109] [Citation(s) in RCA: 139] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Accepted: 11/05/2013] [Indexed: 02/04/2023]
Abstract
REG3γ is considered to have a protective role against infection with Gram-positive bacteria due to its bactericidal activity, but evidence from in vivo studies is lacking. We generated a REG3γ(-/-) mouse, and investigated the effect of lack of REG3γ on intestinal mucus distribution, spatial compartmentalization of bacteria, and expression of innate immunity genes. Infection studies were also performed with Gram-positive and Gram-negative pathogens to investigate the antimicrobial role of REG3γ. REG3γ(-/-) mice display altered mucus distribution, increased bacterial contact with the epithelium, and elevated inflammatory markers in the ileum without histological evidence of pathology. Infection response pathway genes were differentially expressed in both Listeria monocytogenes and Salmonella enteritidis infected REG3γ(-/-) and wild-type (wt) mice. Higher amounts of myeloperoxidase and interleukin-22 transcripts were present in the ileal mucosa of REG3γ(-/-) than wt mice, but translocation to the organs was unaffected. We concluded that REG3γ has a protective role against mucosal infection with pathogenic Listeria and Salmonella in vivo. REG3γ is equally distributed throughout the mucus and its absence results in increased epithelial contact with the microbiota resulting in low-grade inflammation. REG3γ can bind to Gram-negative and Gram-positive bacteria and influence mucus distribution in the ileum, properties which may contribute to mucosal protection.
Collapse
Affiliation(s)
- L M P Loonen
- 1] TI Food and Nutrition, Wageningen, The Netherlands [2] Host-Microbe Interactomics group, Department of Animal Sciences, Wageningen University, Wageningen, The Netherlands
| | - E H Stolte
- Host-Microbe Interactomics group, Department of Animal Sciences, Wageningen University, Wageningen, The Netherlands
| | - M T J Jaklofsky
- Host-Microbe Interactomics group, Department of Animal Sciences, Wageningen University, Wageningen, The Netherlands
| | - M Meijerink
- Host-Microbe Interactomics group, Department of Animal Sciences, Wageningen University, Wageningen, The Netherlands
| | - J Dekker
- TI Food and Nutrition, Wageningen, The Netherlands
| | - P van Baarlen
- Host-Microbe Interactomics group, Department of Animal Sciences, Wageningen University, Wageningen, The Netherlands
| | - J M Wells
- 1] TI Food and Nutrition, Wageningen, The Netherlands [2] Host-Microbe Interactomics group, Department of Animal Sciences, Wageningen University, Wageningen, The Netherlands
| |
Collapse
|
81
|
Erb Downward JR, Falkowski NR, Mason KL, Muraglia R, Huffnagle GB. Modulation of post-antibiotic bacterial community reassembly and host response by Candida albicans. Sci Rep 2014; 3:2191. [PMID: 23846617 PMCID: PMC3709164 DOI: 10.1038/srep02191] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Accepted: 06/14/2013] [Indexed: 12/22/2022] Open
Abstract
The introduction of Candida albicans into cefoperazone-treated mice results in changes in bacterial community reassembly. Our objective was to use high-throughput sequencing to characterize at much greater depth the specific changes in the bacterial microbiome. The colonization of C. albicans significantly altered bacterial community reassembly that was evident at multiple taxonomic levels of resolution. There were marked changes in the levels of Bacteriodetes and Lactobacillaceae. Lachnospiraceae and Ruminococcaceae, the two most abundant bacterial families, did not change in relative proportions after antibiotics, but there were marked genera-level shifts within these two bacterial families. The microbiome shifts occurred in the absence of overt intestinal inflammation. Overall, these experiments demonstrate that the introduction of a single new microbe in numerically inferior numbers into the bacterial microbiome during a broad community disturbance has the potential to significantly alter the subsequent reassembly of the bacterial community as it recovers from that disturbance.
Collapse
Affiliation(s)
- John R Erb Downward
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, USA
| | | | | | | | | |
Collapse
|
82
|
Shuttling of information between the mucosal and luminal environment drives intestinal homeostasis. FEBS Lett 2014; 588:4148-57. [DOI: 10.1016/j.febslet.2014.02.049] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 02/24/2014] [Accepted: 02/25/2014] [Indexed: 12/14/2022]
|
83
|
Shanahan MT, Carroll IM, Gulati AS. Critical design aspects involved in the study of Paneth cells and the intestinal microbiota. Gut Microbes 2014; 5:208-14. [PMID: 24637592 PMCID: PMC4063846 DOI: 10.4161/gmic.27466] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Paneth cells are long-lived secretory cells that reside in the base of the crypts of Lieberkühn of the small intestine. They produce an arsenal of molecules that are involved in numerous biological processes, ranging from the control of gut microbial populations to supporting the intestinal stem cell niche. Because of these important functions, Paneth cell abnormalities are becoming implicated in a variety of disease processes. As such, it is necessary to establish parameters that will allow for the comprehensive study of Paneth cells in health and disease. In this addendum, we highlight critical design aspects involved in the study of Paneth cells and their downstream effects on the intestinal microbiota. The importance of this approach is demonstrated by our recent findings that Nod2 does not regulate mouse Paneth cell antimicrobial function, in contrast to previous reports. This work defines key issues to consider when studying Paneth cells in mouse systems.
Collapse
Affiliation(s)
- Michael T Shanahan
- Department of Medicine; Division of Gastroenterology and Hepatology; University of North Carolina at Chapel Hill; Chapel Hill, NC USA
| | - Ian M Carroll
- Department of Medicine; Division of Gastroenterology and Hepatology; University of North Carolina at Chapel Hill; Chapel Hill, NC USA
| | - Ajay S Gulati
- Department of Pediatrics; Division of Gastroenterology; University of North Carolina at Chapel Hill; Chapel Hill, NC USA,Correspondence to: Ajay S Gulati,
| |
Collapse
|
84
|
Wittkopf N, Neurath MF, Becker C. Immune-epithelial crosstalk at the intestinal surface. J Gastroenterol 2014; 49:375-87. [PMID: 24469679 DOI: 10.1007/s00535-013-0929-4] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 12/18/2013] [Indexed: 02/04/2023]
Abstract
The intestinal tract is one of the most complex organs of the human body. It has to exercise various functions including food and water absorption, as well as barrier and immune regulation. These functions affect not only the gut itself, but influence the overall health of the organism. Diseases involving the gastrointestinal tract such as inflammatory bowel disease and colorectal cancer therefore severely affect the patient's quality of life and can become life-threatening. Intestinal epithelial cells (IECs) play an important role in intestinal inflammation, infection, and cancer development. IECs not only constitute the first barrier in the gut against the lumen, they also constantly signal information about the gut lumen to immune cells, thereby influencing their behaviour. In contrast, by producing various antimicrobial peptides, IECs shape the microbial community within the gut. IECs also respond to cytokines and other mediators of immune cells in the lamina propria. Interactions between epithelial cells and immune cells in the intestine are responsible for gut homeostasis, and modulations of this crosstalk have been reported in studies of gut diseases. This review discusses the wide field of immune-epithelial interactions and shows the importance of immune-epithelial crosstalk in the intestine to gut homeostasis and the overall health status.
Collapse
Affiliation(s)
- Nadine Wittkopf
- Department of Medicine 1, Friedrich-Alexander-University, 91052, Erlangen, Germany,
| | | | | |
Collapse
|
85
|
Kandasamy J, Huda S, Ambalavanan N, Jilling T. Inflammatory signals that regulate intestinal epithelial renewal, differentiation, migration and cell death: Implications for necrotizing enterocolitis. ACTA ACUST UNITED AC 2014; 21:67-80. [PMID: 24533974 DOI: 10.1016/j.pathophys.2014.01.001] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Necrotizing enterocolitis is a disease entity with multiple proposed pathways of pathogenesis. Various combinations of these risk factors, perhaps based on genetic predisposition, possibly lead to the mucosal and epithelial injury that is the hallmark of NEC. Intestinal epithelial integrity is controlled by a tightly regulated balance between proliferation and differentiation of epithelium from intestinal epithelial stem cells and cellular loss by apoptosis. various signaling pathways play a key role in creating and maintaining this balance. The aim of this review article is to outline intestinal epithelial barrier development and structure and the impact of these inflammatory signaling and regulatory pathways as they pertain to the pathogenesis of NEC.
Collapse
Affiliation(s)
- Jegen Kandasamy
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Alabama at Birmingham, USA
| | - Shehzad Huda
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Alabama at Birmingham, USA
| | - Namasivayam Ambalavanan
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Alabama at Birmingham, USA
| | - Tamas Jilling
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Alabama at Birmingham, USA.
| |
Collapse
|
86
|
VanDussen KL, Liu TC, Li D, Towfic F, Modiano N, Winter R, Haritunians T, Taylor KD, Dhall D, Targan SR, Xavier RJ, McGovern DPB, Stappenbeck TS. Genetic variants synthesize to produce paneth cell phenotypes that define subtypes of Crohn's disease. Gastroenterology 2014; 146:200-9. [PMID: 24076061 PMCID: PMC3899786 DOI: 10.1053/j.gastro.2013.09.048] [Citation(s) in RCA: 144] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Revised: 09/11/2013] [Accepted: 09/16/2013] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Genetic susceptibility loci for Crohn's disease (CD) are numerous, complex, and likely interact with undefined components of the environment. It has been a challenge to link the effects of particular loci to phenotypes of cells associated with pathogenesis of CD, such as Paneth cells. We investigated whether specific phenotypes of Paneth cells associated with particular genetic susceptibility loci can be used to define specific subtypes of CD. METHODS We performed a retrospective analysis of 119 resection specimens collected from patients with CD at 2 separate medical centers. Paneth cell phenotypes were classified as normal or abnormal (with disordered, diminished, diffuse, or excluded granule phenotypes) based on lysozyme-positive secretory granule morphology. To uncover the molecular basis of the Paneth cell phenotypes, we developed methods to determine transcriptional profiles from whole-thickness and laser-capture microdissected, formalin-fixed, paraffin-embedded tissue sections. RESULTS The proportion of abnormal Paneth cells was associated with the number of CD-associated NOD2 risk alleles. The cumulative number of NOD2 and ATG16L1 risk alleles had an additive effect on the proportion of abnormal Paneth cells. Unsupervised clustering analysis of demographic and Paneth cell data divided patients into 2 principal subgroups, defined by high and low proportions of abnormal Paneth cells. The disordered and diffuse abnormal Paneth cell phenotypes were associated with an altered transcriptional signature of immune system activation. We observed an inverse correlation between abnormal Paneth cells and presence of granuloma. In addition, high proportions of abnormal Paneth cells were associated with shorter time to disease recurrence after surgery. CONCLUSIONS Histologic analysis of Paneth cell phenotypes can be used to divide patients with CD into subgroups with distinct pathognomonic and clinical features.
Collapse
Affiliation(s)
- Kelli L. VanDussen
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ta-Chiang Liu
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Dalin Li
- The F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA,Medical Genetics Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Fadi Towfic
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Harvard Medical School and Broad Institute, Boston, MA 02115, USA
| | - Nir Modiano
- The F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Rachel Winter
- The F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Talin Haritunians
- The F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA,Medical Genetics Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Kent D. Taylor
- Medical Genetics Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Deepti Dhall
- Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Stephan R. Targan
- The F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Ramnik J. Xavier
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Harvard Medical School and Broad Institute, Boston, MA 02115, USA
| | - Dermot P. B. McGovern
- The F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA,Medical Genetics Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA,Corresponding Authors: Thaddeus S. Stappenbeck, 660 S. Euclid Ave, Box 8118, St. Louis, MO 63110, Phone: 314-362-4214, . Dermot P. B. McGovern, 8797 Beverly Blvd, Suite 300, Los Angeles, CA, 90048,
| | - Thaddeus S. Stappenbeck
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA,Corresponding Authors: Thaddeus S. Stappenbeck, 660 S. Euclid Ave, Box 8118, St. Louis, MO 63110, Phone: 314-362-4214, . Dermot P. B. McGovern, 8797 Beverly Blvd, Suite 300, Los Angeles, CA, 90048,
| |
Collapse
|
87
|
Abstract
The gastrointestinal tract presents the largest and most vulnerable surface to the outside world. Simultaneously, it must be accessible and permeable to nutrients and must defend against pathogens and potentially injurious chemicals. Integrated responses to these challenges require the gut to sense its environment, which it does through a range of detection systems for specific chemical entities, pathogenic organisms and their products (including toxins), as well as physicochemical properties of its contents. Sensory information is then communicated to four major effector systems: the enteroendocrine hormonal signalling system; the innervation of the gut, both intrinsic and extrinsic; the gut immune system; and the local tissue defence system. Extensive endocrine-neuro-immune-organ-defence interactions are demonstrable, but under-investigated. A major challenge is to develop a comprehensive understanding of the integrated responses of the gut to the sensory information it receives. A major therapeutic opportunity exists to develop agents that target the receptors facing the gut lumen.
Collapse
Affiliation(s)
- John B Furness
- Department of Anatomy & Neuroscience, University of Melbourne, Grattan Street, Parkville, Vic 3010, Australia
| | | | | | | | | |
Collapse
|
88
|
Abstract
The complex community of colonizing microbes inhabiting the mucosal surfaces of mammals is vital to homeostasis and normal physiology in the host. When the composition of this microbiota is unfavorably altered, termed dysbiosis, the host is rendered more susceptible to a variety of chronic diseases. In the mammalian small intestine, specialized secretory epithelial cells, named Paneth cells, produce a variety of secreted antimicrobial peptides that fundamentally influence the composition of the microbiota. Recent investigations have identified numerous genetic and environmental factors that can disrupt normal Paneth cell function, resulting in compromised antimicrobial peptide secretion and consequent dysbiosis. These findings suggest that Paneth cell dysfunction should be considered a common cause of dysbiosis.
Collapse
Affiliation(s)
- Nita H Salzman
- Division of Gastroenterology, Department of Pediatrics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, United States.
| | | |
Collapse
|
89
|
Abstract
The intestinal mucosa interfaces with a complex, dense community of microorganisms, including hundreds of species of resident microbiota and many transient microbes entering from food- and water-borne sources. In the small intestine, Paneth cells (specialized secretory epithelial cells) produce abundant quantities of α-defensins and several other antibiotic peptides. Human Paneth cells make two α-defensins: HD5 and HD6. Data from in vivo models indicate that Paneth cell α-defensins play a pivotal role in defense from food- and water-borne pathogens in the intestine. The mechanism by which these two α-defensins protect from enteric pathogens is quite distinct. HD5 is a potent antimicrobial that kills target microbes by membrane disruption, whereas HD6 is newly discovered to self-assemble to form fibrils and nanonets that surround and entangle bacteria. Recent data suggest that HD5 also serves to help shape the composition of the colonizing microbiota. Studies in humans suggest that reduced expression of HD5 and HD6 is a fundamental feature of ileal Crohn's disease. Mechanistically, the link between reduced Paneth cell α-defensin expression and ileal Crohn's disease pathogenesis may be a result of the weakened mucosal antimicrobial defense and/or alterations in the composition of commensal microbiota.
Collapse
Affiliation(s)
- Charles L Bevins
- Department of Microbiology and Immunology, School of Medicine, University of California Davis, Davis, Calif., USA
| |
Collapse
|
90
|
Abstract
Paneth cells are highly specialized epithelial cells of the small intestine, where they coordinate many physiological functions. First identified more than a century ago on the basis of their readily discernible secretory granules by routine histology, these cells are located at the base of the crypts of Lieberkühn, tiny invaginations that line the mucosal surface all along the small intestine. Investigations over the past several decades determined that these cells synthesize and secrete substantial quantities of antimicrobial peptides and proteins. More recent studies have determined that these antimicrobial molecules are key mediators of host-microbe interactions, including homeostatic balance with colonizing microbiota and innate immune protection from enteric pathogens. Perhaps more intriguing, Paneth cells secrete factors that help sustain and modulate the epithelial stem and progenitor cells that cohabitate in the crypts and rejuvenate the small intestinal epithelium. Dysfunction of Paneth cell biology contributes to the pathogenesis of chronic inflammatory bowel disease.
Collapse
Affiliation(s)
- Hans C Clevers
- Hubrecht Institute-KNAW, University Medical Center Utrecht, Uppsalalaan, Utrecht 3584CT, The Netherlands.
| | | |
Collapse
|
91
|
Lucero CM, Fallert Junecko B, Klamar CR, Sciullo LA, Berendam SJ, Cillo AR, Qin S, Sui Y, Sanghavi S, Murphey-Corb MA, Reinhart TA. Macaque paneth cells express lymphoid chemokine CXCL13 and other antimicrobial peptides not previously described as expressed in intestinal crypts. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2013; 20:1320-8. [PMID: 23803902 PMCID: PMC3754526 DOI: 10.1128/cvi.00651-12] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Accepted: 06/12/2013] [Indexed: 12/30/2022]
Abstract
CXCL13 is a constitutively expressed chemokine that controls migration of immune cells to lymphoid follicles. Previously, we found CXCL13 mRNA levels increased in rhesus macaque spleen tissues during AIDS. This led us to examine the levels and locations of CXCL13 by detailed in situ methods in cynomolgus macaque lymphoid and intestinal tissues. Our results revealed that there were distinct localization patterns of CXCL13 mRNA compared to protein in germinal centers. These patterns shifted during the course of simian immunodeficiency virus (SIV) infection, with increased mRNA expression within and around follicles during AIDS compared to uninfected or acutely infected animals. Unexpectedly, CXCL13 expression was also found in abundance in Paneth cells in crypts throughout the small intestine. Therefore, we expanded our analyses to include chemokines and antimicrobial peptides (AMPs) not previously demonstrated to be expressed by Paneth cells in intestinal tissues. We examined the expression patterns of multiple chemokines, including CCL25, as well as α-defensin 6 (DEFA6), β-defensin 2 (BDEF2), rhesus θ-defensin 1 (RTD-1), and Reg3γ in situ in intestinal tissues. Of the 10 chemokines examined, CXCL13 was unique in its expression by Paneth cells. BDEF2, RTD-1, and Reg3γ were also expressed by Paneth cells. BDEF2 and RTD-1 previously have not been shown to be expressed by Paneth cells. These findings expand our understanding of mucosal immunology, innate antimicrobial defenses, homeostatic chemokine function, and host protective mechanisms against microbial translocation.
Collapse
Affiliation(s)
- Carissa M. Lucero
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Beth Fallert Junecko
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Cynthia R. Klamar
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Lauren A. Sciullo
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Stella J. Berendam
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Anthony R. Cillo
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Shulin Qin
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Yongjun Sui
- National Cancer Institute, Bethesda, Maryland, USA
| | - Sonali Sanghavi
- King Edward Memorial Hospital and Research Center, Rasta Peth, Pune, India
| | - Michael A. Murphey-Corb
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Todd A. Reinhart
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
92
|
Abstract
Mucin-type O-glycans are the primary constituents of mucins that are expressed on various mucosal sites of the body, especially the bacteria-laden intestinal tract. Mucins are the main components of mucus, which is secreted by goblet cells and forms a protective homeostatic barrier between the resident microbiota and the underlying immune cells in the colon. However, the specific role of mucin-type O-glycans in mucus barrier function has been uncertain. Recent studies utilizing mice deficient in key glycosyltransferases involved in O-glycan biosynthesis on intestinal mucins have underscored the importance of mucin-type O-glycosylation in mucus barrier function. This review will highlight recent advances in our understanding of mucin-type O-glycan function in the mucus barrier and how they promote mutualism with our resident microbiota.
Collapse
Affiliation(s)
- Kirk S B Bergstrom
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | | |
Collapse
|
93
|
Abstract
Mucins--large, highly glycosylated proteins--are important for the luminal protection of the gastrointestinal tract. Enterocytes have their apical surface covered by transmembrane mucins and goblet cells produce the secreted gel-forming mucins that form mucus. The small intestine has a single unattached mucus layer, which in cystic fibrosis becomes attached, accounting for the intestinal manifestations of this disease. The stomach and colon have two layers of mucus; the inner layer is attached and the outer layer is less dense and unattached. In the colon, the outer mucus layer is the habitat for commensal bacteria. The inner mucus layer is impervious to bacteria and is renewed every hour by surface goblet cells. The crypt goblet cells have the ability to restitute the mucus layer by secretion, for example after an ischaemic challenge. Proteases of certain parasites and some bacteria can cleave mucins and dissolve the mucus as part of their pathogenicity. The inner mucus layer can, however, also become penetrable to bacteria by several other mechanisms, including aberrations in the immune system. When bacteria reach the epithelial surface, the immune system is activated and inflammation is triggered. This mechanism might occur in some types of ulcerative colitis.
Collapse
|
94
|
Cabrera S, Fernández AF, Mariño G, Aguirre A, Suárez MF, Español Y, Vega JA, Laurà R, Fueyo A, Fernández-García MS, Freije JMP, Kroemer G, López-Otín C. ATG4B/autophagin-1 regulates intestinal homeostasis and protects mice from experimental colitis. Autophagy 2013; 9:1188-200. [PMID: 23782979 DOI: 10.4161/auto.24797] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The identification of inflammatory bowel disease (IBD) susceptibility genes by genome-wide association has linked this pathology to autophagy, a lysosomal degradation pathway that is crucial for cell and tissue homeostasis. Here, we describe autophagy-related 4B, cysteine peptidase/autophagin-1 (ATG4B) as an essential protein in the control of inflammatory response during experimental colitis. In this pathological condition, ATG4B protein levels increase in parallel with the induction of autophagy. Moreover, ATG4B expression is significantly reduced in affected areas of the colon from IBD patients. Consistently, atg4b (-/-) mice present Paneth cell abnormalities, as well as an increased susceptibility to DSS-induced colitis. atg4b-deficient mice exhibit significant alterations in proinflammatory cytokines and mediators of the immune response to bacterial infections, which are reminiscent of those found in patients with Crohn disease or ulcerative colitis. Additionally, antibiotic treatments and bone marrow transplantation from wild-type mice reduced colitis in atg4b (-/-) mice. Taken together, these results provided additional evidence for the importance of autophagy in intestinal pathologies and describe ATG4B as a novel protective protein in inflammatory colitis. Finally, we propose that atg4b-null mice are a suitable model for in vivo studies aimed at testing new therapeutic strategies for intestinal diseases associated with autophagy deficiency.
Collapse
Affiliation(s)
- Sandra Cabrera
- Departamento de Bioquímica y Biología Molecular; Facultad de Medicina; Instituto Universitario de Oncología (IUOPA); Universidad de Oviedo; Oviedo, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
95
|
Stow JL, Murray RZ. Intracellular trafficking and secretion of inflammatory cytokines. Cytokine Growth Factor Rev 2013; 24:227-39. [PMID: 23647915 DOI: 10.1016/j.cytogfr.2013.04.001] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The secretion of cytokines by immune cells plays a significant role in determining the course of an inflammatory response. The levels and timing of each cytokine released are critical for mounting an effective but confined response, whereas excessive or dysregulated inflammation contributes to many diseases. Cytokines are both culprits and targets for effective treatments in some diseases. The multiple points and mechanisms that have evolved for cellular control of cytokine secretion highlight the potency of these mediators and the fine tuning required to manage inflammation. Cytokine production in cells is regulated by cell signaling, and at mRNA and protein synthesis levels. Thereafter, the intracellular transport pathways and molecular trafficking machinery have intricate and essential roles in dictating the release and activity of cytokines. The trafficking machinery and secretory (exocytic) pathways are complex and highly regulated in many cells, involving specialized membranes, molecules and organelles that enable these cells to deliver cytokines to often-distinct areas of the cell surface, in a timely manner. This review provides an overview of secretory pathways - both conventional and unconventional - and key families of trafficking machinery. The prevailing knowledge about the trafficking and secretion of a number of individual cytokines is also summarized. In conclusion, we present emerging concepts about the functional plasticity of secretory pathways and their modulation for controlling cytokines and inflammation.
Collapse
Affiliation(s)
- Jennifer L Stow
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia.
| | | |
Collapse
|
96
|
Glycobiome: bacteria and mucus at the epithelial interface. Best Pract Res Clin Gastroenterol 2013; 27:25-38. [PMID: 23768550 DOI: 10.1016/j.bpg.2013.03.001] [Citation(s) in RCA: 161] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Accepted: 03/08/2013] [Indexed: 02/06/2023]
Abstract
The human gastrointestinal tract is colonised with a dense and diverse microbial community, that is an important player in human health and physiology. Close to the epithelial cells the mucosal microbiota is separated from the host with a thin lining of host derived glycans, including the cell surface glycocalyx and the extracellular secreted mucus. The mucosa-associated microbial composition differs from the luminal content and could be particularly important for nutrient exchange, communication with the host, development of the immune system, and resistance against invading pathogens. The mucosa-associated microbiota has adapted to the glycan rich environment by the production of mucus-degrading enzymes and mucus-binding extracellular proteins, and include mucus-degrading specialists such as Akkermansia muciniphila and Bacteroides thetaiotaomicron. This review is focussed on the host-microbe interactions within the glycan landscape at the epithelial interface and considers the spatial organisation and composition of the mucosa-associated microbiota in health and disease.
Collapse
|
97
|
Affiliation(s)
- Hans Clevers
- Hubrecht Institute and University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
98
|
Pinheiro da Silva F, Machado MCC. Antimicrobial peptides: clinical relevance and therapeutic implications. Peptides 2012; 36:308-14. [PMID: 22659161 DOI: 10.1016/j.peptides.2012.05.014] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2012] [Revised: 05/23/2012] [Accepted: 05/23/2012] [Indexed: 12/14/2022]
Abstract
Antimicrobial peptides (AMPs) are molecules that provide protection against environmental pathogens, acting against a large number of microorganisms, including bacteria, fungi, yeast, virus and others. Two major groups of antimicrobial peptides are found in humans: cathelicidins and defensins. Recently, several studies have furnished information that besides their role in infection diseases, antimicrobial peptides play a role in diseases as diverse as inflammatory disorders, autoimmunity and cancer. Here, we discuss the role of antimicrobial peptides and vitamin D have in such complex diseases and propose their use should be more explored in the diagnosis and treatment of such conditions.
Collapse
|
99
|
Chu H, Pazgier M, Jung G, Nuccio SP, Castillo PA, de Jong MF, Winter MG, Winter SE, Wehkamp J, Shen B, Salzman NH, Underwood MA, Tsolis RM, Young GM, Lu W, Lehrer RI, Bäumler AJ, Bevins CL. Human α-defensin 6 promotes mucosal innate immunity through self-assembled peptide nanonets. Science 2012; 337:477-81. [PMID: 22722251 DOI: 10.1126/science.1218831] [Citation(s) in RCA: 311] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Defensins are antimicrobial peptides that contribute broadly to innate immunity, including protection of mucosal tissues. Human α-defensin (HD) 6 is highly expressed by secretory Paneth cells of the small intestine. However, in contrast to the other defensins, it lacks appreciable bactericidal activity. Nevertheless, we report here that HD6 affords protection against invasion by enteric bacterial pathogens in vitro and in vivo. After stochastic binding to bacterial surface proteins, HD6 undergoes ordered self-assembly to form fibrils and nanonets that surround and entangle bacteria. This self-assembly mechanism occurs in vivo, requires histidine-27, and is consistent with x-ray crystallography data. These findings support a key role for HD6 in protecting the small intestine against invasion by diverse enteric pathogens and may explain the conservation of HD6 throughout Hominidae evolution.
Collapse
Affiliation(s)
- Hiutung Chu
- Department of Microbiology and Immunology, School of Medicine, University of California, Davis, CA 95616, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
100
|
Gulati AS, Shanahan MT, Arthur JC, Grossniklaus E, von Furstenberg RJ, Kreuk L, Henning SJ, Jobin C, Sartor RB. Mouse background strain profoundly influences Paneth cell function and intestinal microbial composition. PLoS One 2012; 7:e32403. [PMID: 22384242 PMCID: PMC3288091 DOI: 10.1371/journal.pone.0032403] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2011] [Accepted: 01/30/2012] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Increasing evidence supports the central role of Paneth cells in maintaining intestinal host-microbial homeostasis. However, the direct impact of host genotype on Paneth cell function remains unclear. Here, we characterize key differences in Paneth cell function and intestinal microbial composition in two widely utilized, genetically distinct mouse strains (C57BL/6 and 129/SvEv). In doing so, we demonstrate critical influences of host genotype on Paneth cell activity and the enteric microbiota. METHODOLOGY AND PRINCIPAL FINDINGS Paneth cell numbers were determined by flow cytometry. Antimicrobial peptide (AMP) expression was evaluated using quantitative reverse-transcriptase polymerase chain reaction (qRT-PCR), acid urea-polyacrylamide gel electrophoresis, and mass spectrometry. Effects of mouse background on microbial composition were assessed by reciprocal colonization of germ-free mice from both background strains, followed by compositional analysis of resultant gut bacterial communities using terminal restriction fragment length polymorphism analysis and 16 S qPCR. Our results revealed that 129/SvEv mice possessed fewer Paneth cells and a divergent AMP profile relative to C57BL/6 counterparts. Novel 129/SvEv á-defensin peptides were identified, including Defa2/18v, Defa11, Defa16, and Defa18. Host genotype profoundly affected the global profile of the intestinal microbiota, while both source and host factors were found to influence specific bacterial groups. Interestingly, ileal α-defensins from 129/SvEv mice displayed attenuated antimicrobial activity against pro-inflammatory E. coli strains, a bacterial species found to be expanded in these animals. CONCLUSIONS AND SIGNIFICANCE This work establishes the important impact of host genotype on Paneth cell function and the composition of the intestinal microbiota. It further identifies specific AMP and microbial alterations in two commonly used inbred mouse strains that have varying susceptibilities to a variety of disorders, ranging from obesity to intestinal inflammation. This will be critical for future studies utilizing these murine backgrounds to study the effects of Paneth cells and the intestinal microbiota on host health and disease.
Collapse
Affiliation(s)
- Ajay S Gulati
- Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, North Carolina, United States of America.
| | | | | | | | | | | | | | | | | |
Collapse
|