1
|
Dang K, Zhang N, Gao H, Wang G, Liang H, Xue M. Influence of intestinal microecology in the development of gout or hyperuricemia and the potential therapeutic targets. Int J Rheum Dis 2023; 26:1911-1922. [PMID: 37606177 DOI: 10.1111/1756-185x.14888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 08/08/2023] [Accepted: 08/09/2023] [Indexed: 08/23/2023]
Abstract
Gout and hyperuricemia are common metabolic diseases. Patients with purine metabolism disorder and/or decreased uric acid excretion showed increased uric acid levels in the blood. The increase of uric acid in the blood leads to the deposition of urate crystals in tissues, joints, and kidneys, and causes gout. Recent studies have revealed that imbalance of the intestinal microecology is closely related to the occurrence and development of hyperuricemia and gout. Disorder of the intestinal flora often occurs in patients with gout, and high purine and high fructose may induce the disorder of intestinal flora. Short-chain fatty acids and endotoxins produced by intestinal bacteria are closely related to the inflammatory response of gout. This article summarizes the characteristics of intestinal microecology in patients or animal models with hyperuricemia or gout, and explores the relationship between intestinal microecology and gout or hyperuricemia from the aspect of the intestinal barrier, intestinal microorganisms, intestinal metabolites, and intestinal immune system. We also review the current status of hyperuricemia treatment by targeting intestinal microecology.
Collapse
Affiliation(s)
- Kai Dang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Nan Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Haiqi Gao
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Guifa Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Hui Liang
- Department of Human Nutrition, College of Public Health, Qingdao University, Qingdao, China
| | - Meilan Xue
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| |
Collapse
|
2
|
Shemtov SJ, Emani R, Bielska O, Covarrubias AJ, Verdin E, Andersen JK, Winer DA. The intestinal immune system and gut barrier function in obesity and ageing. FEBS J 2023; 290:4163-4186. [PMID: 35727858 PMCID: PMC9768107 DOI: 10.1111/febs.16558] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 04/29/2022] [Accepted: 06/20/2022] [Indexed: 08/13/2023]
Abstract
Obesity and ageing predispose to numerous, yet overlapping chronic diseases. For example, metabolic abnormalities, including insulin resistance (IR) and type 2 diabetes (T2D) are important causes of morbidity and mortality. Low-grade chronic inflammation of tissues, such as the liver, visceral adipose tissue and neurological tissues, is considered a significant contributor to these chronic diseases. Thus, it is becoming increasingly important to understand what drives this inflammation in affected tissues. Recent evidence, especially in the context of obesity, suggests that the intestine plays an important role as the gatekeeper of inflammatory stimuli that ultimately fuels low-grade chronic tissue inflammation. In addition to metabolic diseases, abnormalities in the intestinal mucosal barrier have been linked to a range of other chronic inflammatory conditions, such as neurodegeneration and ageing. The flow of inflammatory stimuli from the gut is in part controlled by local immunological inputs impacting the intestinal barrier. Here, we will review the impact of obesity and ageing on the intestinal immune system and its downstream consequences on gut barrier function, which is strongly implicated in the pathogenesis of obesity and age-related diseases. In particular, we will discuss the effects of age-related intestinal dysfunction on neurodegenerative diseases.
Collapse
Affiliation(s)
- Sarah J. Shemtov
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California, USA
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945, USA
| | - Rohini Emani
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945, USA
| | - Olga Bielska
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945, USA
| | - Anthony J. Covarrubias
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095 USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, 90095 USA
| | - Eric Verdin
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945, USA
| | - Julie K. Andersen
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945, USA
| | - Daniel A. Winer
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California, USA
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945, USA
- Division of Cellular & Molecular Biology, Diabetes Research Group, Toronto General Research Institute (TGRI), University Health Network, 101 College Street, Toronto, ON, M5G 1L7, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, 1 King’s College Circle, Toronto, ON, M5S 1A8, Canada
- Department of Immunology, University of Toronto, 1 King’s College Circle, Toronto, ON, M5S 1A8, Canada
| |
Collapse
|
3
|
Shukla PK, Rao RG, Meena AS, Giorgianni F, Lee SC, Raju P, Shashikanth N, Shekhar C, Beranova S, Balazs L, Tigyi G, Gosain A, Rao R. Paneth cell dysfunction in radiation injury and radio-mitigation by human α-defensin 5. Front Immunol 2023; 14:1174140. [PMID: 37638013 PMCID: PMC10448521 DOI: 10.3389/fimmu.2023.1174140] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 07/14/2023] [Indexed: 08/29/2023] Open
Abstract
Introduction The mechanism underlying radiation-induced gut microbiota dysbiosis is undefined. This study examined the effect of radiation on the intestinal Paneth cell α-defensin expression and its impact on microbiota composition and mucosal tissue injury and evaluated the radio-mitigative effect of human α-defensin 5 (HD5). Methods Adult mice were subjected to total body irradiation, and Paneth cell α-defensin expression was evaluated by measuring α-defensin mRNA by RT-PCR and α-defensin peptide levels by mass spectrometry. Vascular-to-luminal flux of FITC-inulin was measured to evaluate intestinal mucosal permeability and endotoxemia by measuring plasma lipopolysaccharide. HD5 was administered in a liquid diet 24 hours before or after irradiation. Gut microbiota was analyzed by 16S rRNA sequencing. Intestinal epithelial junctions were analyzed by immunofluorescence confocal microscopy and mucosal inflammatory response by cytokine expression. Systemic inflammation was evaluated by measuring plasma cytokine levels. Results Ionizing radiation reduced the Paneth cell α-defensin expression and depleted α-defensin peptides in the intestinal lumen. α-Defensin down-regulation was associated with the time-dependent alteration of gut microbiota composition, increased gut permeability, and endotoxemia. Administration of human α-defensin 5 (HD5) in the diet 24 hours before irradiation (prophylactic) significantly blocked radiation-induced gut microbiota dysbiosis, disruption of intestinal epithelial tight junction and adherens junction, mucosal barrier dysfunction, and mucosal inflammatory response. HD5, administered 24 hours after irradiation (treatment), reversed radiation-induced microbiota dysbiosis, tight junction and adherens junction disruption, and barrier dysfunction. Furthermore, HD5 treatment also prevents and reverses radiation-induced endotoxemia and systemic inflammation. Conclusion These data demonstrate that radiation induces Paneth cell dysfunction in the intestine, and HD5 feeding prevents and mitigates radiation-induced intestinal mucosal injury, endotoxemia, and systemic inflammation.
Collapse
Affiliation(s)
- Pradeep K. Shukla
- College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Roshan G. Rao
- College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Avtar S. Meena
- College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Francesco Giorgianni
- College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Sue Chin Lee
- College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Preeti Raju
- College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Nitesh Shashikanth
- College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Chandra Shekhar
- College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Sarka Beranova
- College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Louisa Balazs
- College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Gabor Tigyi
- College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Ankush Gosain
- College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - RadhaKrishna Rao
- College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|
4
|
Barreto E Barreto L, Rattes IC, da Costa AV, Gama P. Paneth cells and their multiple functions. Cell Biol Int 2022; 46:701-710. [PMID: 35032139 DOI: 10.1002/cbin.11764] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 12/30/2021] [Accepted: 01/08/2022] [Indexed: 01/08/2023]
Abstract
The small intestine mucosa is lined by specialized cells that form the crypt-villus axis, which expands its surface. Among the six intestinal epithelial cell types, the Paneth cell is located at the base of the crypt, and it contains numerous granules in its cytoplasm, composed of antimicrobial peptides, such as defensins and lysozyme, and growth factors, such as EGF, TGF-alpha, and Wnt ligands. Together, these elements act in the defense against microorganisms, regulation of intestinal microbiota, maintenance, and regulation of stem cell identity. Pathologies that target Paneth cells can disturb such defense activity, but they also affect the maintenance of stem cell niche. In that way, Crohn's disease, necrotizing enterocolitis, and graft-versus-host disease promote a reduction of Paneth cell population, and consequently of secretion of their products into the lumen of the crypts, making the affected organism predisposed to infections and dysbiosis. Additionally, the emergence of new intestinal cells is also decreased. This review aims to address the main characteristics of Paneth cells, highlighting their multiple functions and the importance of their preservation to ensure bowel homeostasis. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Laylla Barreto E Barreto
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Isadora Campos Rattes
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Aline Vasques da Costa
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Patrícia Gama
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| |
Collapse
|
5
|
Wada Y, Kodama M, Mizukami K, Okimoto T, Fuchino T, Tsutsumi K, Fukuda M, Hirashita Y, Fukuda K, Okamoto K, Ogawa R, Kushima R, Murakami K. Differences in Regression Patterns of Complete and Incomplete Intestinal Metaplasia at Ten Years after Helicobacter pylori Eradication. Acta Histochem Cytochem 2021; 54:185-194. [PMID: 35023881 PMCID: PMC8727845 DOI: 10.1267/ahc.21-00069] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 10/01/2021] [Indexed: 11/22/2022] Open
Abstract
This study was conducted to reveal the reversibility of subtype of intestinal metaplasia (IM) and Paneth cells after H. pylori eradication (HPE). Among 75 patients, we retrospectively examined the proportions of patients with complete type of IM (CIM), incomplete type of IM (IIM) and Paneth cells in their biopsy specimens obtained from the greater curvature of the antrum (A2) and the greater curvature of the middle corpus (B2) before and during a follow-up period of 10 years after HPE. Immunohistochemistry was used to determine IM type. Compared to before HPE, the proportion of patients with CIM did not decrease significantly during the 10-year follow-up after HPE both in A2 (32% vs. 21.3%, P = 0.13) and in B2 (6.7% vs. 2.7%, P = 0.60). IIM rates in A2 was significantly lower during this time (26.7% vs. 10.7%, P = 0.04), whereas no patients showed IIM in B2 before HPE. The proportion of patients with Paneth cells decreased significantly in A2 after 3, 8, and 9 years of HPE and in B2 after 4, 6 and 9 years of HPE (P < 0.05 for all). Thus, IIM and Paneth cells regressed during a period of 10 years after HPE.
Collapse
Affiliation(s)
- Yasuhiro Wada
- Department of Gastroenterology, Faculty of Medicine, Oita University, Yufu, Oita, Japan
| | - Masaaki Kodama
- Department of Gastroenterology, Faculty of Medicine, Oita University, Yufu, Oita, Japan
- Faculty of Welfare and Health Science, Oita University, Oita, Japan
| | - Kazuhiro Mizukami
- Department of Gastroenterology, Faculty of Medicine, Oita University, Yufu, Oita, Japan
| | - Tadayoshi Okimoto
- Department of Gastroenterology, Faculty of Medicine, Oita University, Yufu, Oita, Japan
| | - Takafumi Fuchino
- Department of Gastroenterology, Faculty of Medicine, Oita University, Yufu, Oita, Japan
| | - Koshiro Tsutsumi
- Department of Gastroenterology, Faculty of Medicine, Oita University, Yufu, Oita, Japan
| | - Masahide Fukuda
- Department of Gastroenterology, Faculty of Medicine, Oita University, Yufu, Oita, Japan
| | - Yuka Hirashita
- Department of Gastroenterology, Faculty of Medicine, Oita University, Yufu, Oita, Japan
| | - Kensuke Fukuda
- Department of Gastroenterology, Faculty of Medicine, Oita University, Yufu, Oita, Japan
| | - Kazuhisa Okamoto
- Department of Gastroenterology, Faculty of Medicine, Oita University, Yufu, Oita, Japan
| | - Ryo Ogawa
- Department of Gastroenterology, Faculty of Medicine, Oita University, Yufu, Oita, Japan
| | - Ryoji Kushima
- Department of Pathology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Kazunari Murakami
- Department of Gastroenterology, Faculty of Medicine, Oita University, Yufu, Oita, Japan
| |
Collapse
|
6
|
Sun T, Xue M, Yang J, Pei Z, Zhang N, Qin K, Liang H. Metabolic regulation mechanism of fucoidan via intestinal microecology in diseases. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2021; 101:4456-4463. [PMID: 33682122 DOI: 10.1002/jsfa.11202] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 02/23/2021] [Accepted: 03/07/2021] [Indexed: 06/12/2023]
Abstract
The intestinal microecology is an extremely complex ecosystem consisting of gut microbiota, intestinal mucosa and the intestinal immune system. The intestinal microecology performs several important functions and is considered to be an essential 'organ' because it plays an important role in regulating human metabolism. Fucoidan contains a large amount of fucose and galactose residues, as well as various other neutral and acidic monosaccharides. Fucoidan particularly effects tumors, inflammatory bowel disease, diabetes and obesity by repairing intestinal mucosal damage and improving the intestinal microecological environment. It has been proposed that fucoidan could be used as a prebiotic agent for pharmaceutical and functional foods. In this review, we elucidate the potential mechanisms of the metabolic regulation of fucoidan with respect to the intestinal microecology of diseases. © 2021 Society of Chemical Industry.
Collapse
Affiliation(s)
- Ting Sun
- Basic Medical College, Qingdao University of Medicine, Qingdao, China
| | - Meilan Xue
- Basic Medical College, Qingdao University of Medicine, Qingdao, China
| | - Jia Yang
- Basic Medical College, Qingdao University of Medicine, Qingdao, China
| | - Zhongqian Pei
- Basic Medical College, Qingdao University of Medicine, Qingdao, China
| | - Nan Zhang
- Basic Medical College, Qingdao University of Medicine, Qingdao, China
| | - Kunpeng Qin
- Basic Medical College, Qingdao University of Medicine, Qingdao, China
| | - Hui Liang
- Department of Human Nutrition, College of Public Health, Qingdao University of Medicine, Qingdao, China
| |
Collapse
|
7
|
Liu TC, Kern JT, Jain U, Sonnek NM, Xiong S, Simpson KF, VanDussen KL, Winkler ES, Haritunians T, Malique A, Lu Q, Sasaki Y, Storer C, Diamond MS, Head RD, McGovern DPB, Stappenbeck TS. Western diet induces Paneth cell defects through microbiome alterations and farnesoid X receptor and type I interferon activation. Cell Host Microbe 2021; 29:988-1001.e6. [PMID: 34010595 DOI: 10.1016/j.chom.2021.04.004] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 12/22/2020] [Accepted: 04/09/2021] [Indexed: 02/07/2023]
Abstract
Intestinal Paneth cells modulate innate immunity and infection. In Crohn's disease, genetic mutations together with environmental triggers can disable Paneth cell function. Here, we find that a western diet (WD) similarly leads to Paneth cell dysfunction through mechanisms dependent on the microbiome and farnesoid X receptor (FXR) and type I interferon (IFN) signaling. Analysis of multiple human cohorts suggests that obesity is associated with Paneth cell dysfunction. In mouse models, consumption of a WD for as little as 4 weeks led to Paneth cell dysfunction. WD consumption in conjunction with Clostridium spp. increased the secondary bile acid deoxycholic acid levels in the ileum, which in turn inhibited Paneth cell function. The process required excess signaling of both FXR and IFN within intestinal epithelial cells. Our findings provide a mechanistic link between poor diet and inhibition of gut innate immunity and uncover an effect of FXR activation in gut inflammation.
Collapse
Affiliation(s)
- Ta-Chiang Liu
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA.
| | - Justin T Kern
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Umang Jain
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Naomi M Sonnek
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Shanshan Xiong
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Katherine F Simpson
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Kelli L VanDussen
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Emma S Winkler
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Talin Haritunians
- The F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles 90048, USA
| | - Atika Malique
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Qiuhe Lu
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Yo Sasaki
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Chad Storer
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Michael S Diamond
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA; Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Richard D Head
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Dermot P B McGovern
- The F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles 90048, USA
| | - Thaddeus S Stappenbeck
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA.
| |
Collapse
|
8
|
Bilsborough J, Targan SR, Snapper SB. Therapeutic Targets in Inflammatory Bowel Disease: Current and Future. ACTA ACUST UNITED AC 2016. [DOI: 10.1038/ajgsup.2016.18] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
9
|
Abstract
Macrophages are found in all tissues and regulate tissue morphogenesis during development through trophic and scavenger functions. The colony stimulating factor-1 (CSF-1) receptor (CSF-1R) is the major regulator of tissue macrophage development and maintenance. In combination with receptor activator of nuclear factor κB (RANK), the CSF-1R also regulates the differentiation of the bone-resorbing osteoclast and controls bone remodeling during embryonic and early postnatal development. CSF-1R-regulated macrophages play trophic and remodeling roles in development. Outside the mononuclear phagocytic system, the CSF-1R directly regulates neuronal survival and differentiation, the development of intestinal Paneth cells and of preimplantation embryos, as well as trophoblast innate immune function. Consistent with the pleiotropic roles of the receptor during development, CSF-1R deficiency in most mouse strains causes embryonic or perinatal death and the surviving mice exhibit multiple developmental and functional deficits. The CSF-1R is activated by two dimeric glycoprotein ligands, CSF-1, and interleukin-34 (IL-34). Homozygous Csf1-null mutations phenocopy most of the deficits of Csf1r-null mice. In contrast, Il34-null mice have no gross phenotype, except for decreased numbers of Langerhans cells and microglia, indicating that CSF-1 plays the major developmental role. Homozygous inactivating mutations of the Csf1r or its ligands have not been reported in man. However, heterozygous inactivating mutations in the Csf1r lead to a dominantly inherited adult-onset progressive dementia, highlighting the importance of CSF-1R signaling in the brain.
Collapse
Affiliation(s)
- Violeta Chitu
- Albert Einstein College of Medicine, Bronx, NY, United States
| | | |
Collapse
|
10
|
Shenoy S. Genetic risks and familial associations of small bowel carcinoma. World J Gastrointest Oncol 2016; 8:509-519. [PMID: 27326320 PMCID: PMC4909452 DOI: 10.4251/wjgo.v8.i6.509] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 02/02/2016] [Accepted: 03/14/2016] [Indexed: 02/05/2023] Open
Abstract
Adenocarcinoma of small intestines (SBA) is a relatively rare malignancy with poor outcomes due to delayed diagnosis. Fifty percent of patients have metastases on presentation and therefore early detection and treatment offers the best long term outcomes. Certain genetic polyposis syndromes and familial diseases are associated with increased risks for SBA. These include familial adenomatous polyposis (FAP), Lynch syndromes (LS), Juvenile polyposis syndrome, Peutz-Jeghers syndrome, Crohn's disease (CD) and celiac disease. Mutations in APC gene, Mismatch repair genes, STK11 gene, and SMAD4 gene have been implicated for the genetic diseases respectively. While there are no specific inherited genetic mutations for CD, genome-wide association studies have established over 140 loci associated with CD. CpG island mutations with defects in mismatch repair genes have been identified in celiac disease. Significant diagnostic advances have occurred in the past decade and intuitively, it would seem beneficial to use these advanced modalities for surveillance of these patients. At present it is debatable and no clear data exists to support this approach except for established guidelines to diagnose duodenal polyps in FAP, and LS. Here we discuss the genetic alterations, cancer risks, signaling mechanisms and briefly touch the surveillance modalities available for these genetic and clinical syndromes. English language articles from PubMed/Medline and Embase was searched were collected using the phrases "small-bowel adenocarcinoma, genetics, surveillance, familial adenomatous polyposis, lynch syndromes, Peutz-Jeghers syndrome, juvenile polyposis syndrome, CD and celiac disease". Figures, tables and schematic diagram to illustrate pathways are included in the review.
Collapse
|
11
|
Chen W, Frankel WL, Cronley KM, Yu L, Zhou X, Yearsley MM. Significance of paneth cell metaplasia in Barrett esophagus: a morphologic and clinicopathologic study. Am J Clin Pathol 2015; 143:665-71. [PMID: 25873500 DOI: 10.1309/ajcpvujmcvbc9pkm] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
OBJECTIVES The metaplastic intestinal epithelium in Barrett esophagus (BE) occasionally contains Paneth cells; however, little is known regarding the prevalence and significance of Paneth cell metaplasia (PCM) in BE. METHODS We evaluated 757 esophageal biopsy specimens with intestinal metaplasia (IM) for PCM. Outcome analysis was performed in 299 cases with complete clinical data using multinomial logistic regression. RESULTS Thirty-one percent (234/757) of the IM cases showed PCM. Paneth cells are decreased when BE epithelium becomes increasingly dysplastic. Long-segment BE shows significantly more PCM than short-segment BE. On follow-up biopsies, patients without PCM (NPCM) are three times more likely to regress than patients with PCM, regardless of dysplasia, BE segment length, age, or sex. However, there is no significant difference in terms of progression to dysplasia/adenocarcinoma between the PCM and NPCM groups. CONCLUSIONS The presence of PCM is associated with less disease regression and is not associated with more disease progression.
Collapse
Affiliation(s)
- Wei Chen
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus
| | - Wendy L. Frankel
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus
| | - Kevin M. Cronley
- Department of Gastroenterology, Hepatology and Nutrition, The Ohio State University Wexner Medical Center, Columbus
| | - Lianbo Yu
- Center for Biostatistics, The Ohio State University Wexner Medical Center, Columbus
| | - Xiaoping Zhou
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus
| | - Martha M. Yearsley
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus
| |
Collapse
|
12
|
Zimmer J, Hobkirk J, Mohamed F, Browning MJ, Stover CM. On the Functional Overlap between Complement and Anti-Microbial Peptides. Front Immunol 2015; 5:689. [PMID: 25646095 PMCID: PMC4298222 DOI: 10.3389/fimmu.2014.00689] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 12/22/2014] [Indexed: 12/19/2022] Open
Abstract
Intriguingly, activated complement and anti-microbial peptides share certain functionalities; lytic, phagocytic, and chemo-attractant activities and each may, in addition, exert cell instructive roles. Each has been shown to have distinct LPS detoxifying activity and may play a role in the development of endotoxin tolerance. In search of the origin of complement, a functional homolog of complement C3 involved in opsonization has been identified in horseshoe crabs. Horseshoe crabs possess anti-microbial peptides able to bind to acyl chains or phosphate groups/saccharides of endotoxin, LPS. Complement activity as a whole is detectable in marine invertebrates. These are also a source of anti-microbial peptides with potential pharmaceutical applicability. Investigating the locality for the production of complement pathway proteins and their role in modulating cellular immune responses are emerging fields. The significance of local synthesis of complement components is becoming clearer from in vivo studies of parenchymatous disease involving specifically generated, complement-deficient mouse lines. Complement C3 is a central component of complement activation. Its provision by cells of the myeloid lineage varies. Their effector functions in turn are increased in the presence of anti-microbial peptides. This may point to a potentiating range of activities, which should serve the maintenance of health but may also cause disease. Because of the therapeutic implications, this review will consider closely studies dealing with complement activation and anti-microbial peptide activity in acute inflammation (e.g., dialysis-related peritonitis, appendicitis, and ischemia).
Collapse
Affiliation(s)
- Jana Zimmer
- Department of Infectious Diseases - Medical Microbiology and Hygiene, Ruprecht-Karls-University of Heidelberg , Heidelberg , Germany
| | - James Hobkirk
- Department of Academic Endocrinology, Diabetes and Metabolism, Hull York Medical School, University of Hull , Hull , UK
| | - Fatima Mohamed
- Department of Infection, Immunity and Inflammation, University of Leicester , Leicester , UK
| | - Michael J Browning
- Department of Infection, Immunity and Inflammation, University of Leicester , Leicester , UK ; Department of Immunology, Leicester Royal Infirmary , Leicester , UK
| | - Cordula M Stover
- Department of Infection, Immunity and Inflammation, University of Leicester , Leicester , UK
| |
Collapse
|
13
|
Kremer B, Mariman R, van Erk M, Lagerweij T, Nagelkerken L. Temporal colonic gene expression profiling in the recurrent colitis model identifies early and chronic inflammatory processes. PLoS One 2012; 7:e50388. [PMID: 23226271 PMCID: PMC3511545 DOI: 10.1371/journal.pone.0050388] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2012] [Accepted: 10/18/2012] [Indexed: 12/30/2022] Open
Abstract
The recurrent TNBS-colitis model in BALB/c mice has been proposed as a model of Inflammatory Bowel Disease with a shifting pattern of local cytokines with the expression of Th1 cytokines during the early phase, Th17 cytokines during the intermediate phase and Th2 cytokines during late fibrotic stages. In this study, we evaluated the development of pathology in time–in conjunction with genome-wide gene expression in the colons–in response to three weekly intrarectal instillations of TNBS. During this time-frame mice develop colitis with extensive cellular infiltration of (sub)mucosa and mildly to moderately affected crypt architecture. These pathological processes were sensitive to local treatment with budesonide. Gene expression profiling confirmed an acute phase response after each intrarectal TNBS-challenge. In addition, a chronic inflammatory process developed over time particularly evident from a gradual increase in expression of mast cell related genes. The changes in pathological hallmarks were consistent with a temporal expression of mRNA encoding a selection of chemokines. In conclusion, the early stages of the recurrent TNBS-colitis model reflect several aspects of inflammatory bowel disease which are sensitive to immunomodulation.
Collapse
Affiliation(s)
- Bas Kremer
- Department of Metabolic Health Research, TNO, Leiden, The Netherlands.
| | | | | | | | | |
Collapse
|
14
|
The goblet cell is the cellular source of the anti-microbial angiogenin 4 in the large intestine post Trichuris muris infection. PLoS One 2012; 7:e42248. [PMID: 22970115 PMCID: PMC3435386 DOI: 10.1371/journal.pone.0042248] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2012] [Accepted: 07/04/2012] [Indexed: 01/10/2023] Open
Abstract
Background Mouse angiogenin 4 (Ang4) has previously been described as a Paneth cell–derived antimicrobial peptide important in epithelial host defence in the small intestine. However, a source for Ang4 in the large intestine, which is devoid of Paneth cells, has not been defined. Methodology/Principal Findings Analysis was performed on Ang4 expression in colonic tissue by qPCR and immunohistochemistry following infection with the large intestine dwelling helminth parasite Trichuris muris. This demonstrated an increase in expression of the peptide following infection of resistant BALB/c mice. Further, histological analysis of colonic tissue revealed the cellular source of this Ang4 to be goblet cells. To elucidate the mechanism of Ang4 expression immunohistochemistry and qPCR for Ang4 was performed on colonic tissue from T. muris infected mouse mutants. Experiments comparing C3H/HeN and C3H/HeJ mice, which have a natural inactivating mutation of TLR4, revealed that Ang4 expression is TLR4 independent. Subsequent experiments with IL-13 and IL-4 receptor alpha deficient mice demonstrated that goblet cell expression of Ang4 is controlled either directly or indirectly by IL-13. Conclusions The cellular source of mouse Ang4 in the colon following T. muris infection is the goblet cell and expression is under the control of IL-13.
Collapse
|
15
|
Rumio C, Sommariva M, Sfondrini L, Palazzo M, Morelli D, Viganò L, De Cecco L, Tagliabue E, Balsari A. Induction of Paneth cell degranulation by orally administered Toll-like receptor ligands. J Cell Physiol 2012; 227:1107-13. [PMID: 21567398 DOI: 10.1002/jcp.22830] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The secretory activity of Paneth cells is related to the bacterial milieu in the small intestine; however, the molecules involved in inducing Paneth cell secretion of enzymes and antimicrobial peptides are not well-defined. Mice treated orally with CpG-oligodeoxynucleotide (ODN), an agonist of Toll-like receptor (TLR) 9, showed rapid and massive Paneth cell degranulation. CpG-ODN-induced degranulation was not observed in TLR9(-/-) mice or in chimeric TLR9(-/-) mice reconstituted with wild-type (WT) bone marrow, but was observed in WT mice reconstituted with TLR9(-/-) bone marrow, indicating a role for TLR9-expressing gastrointestinal cells in CpG recognition. The TLR3 agonist polyinosinic-polycytidylic acid also induced rapid degranulation, whereas the TLR4 and TLR5 agonists LPS and flagellin, respectively, induced late degranulation mediated by TNF-α. Our evidence that TLR9 and TLR3 agonists induce Paneth cell degranulation points to the need for further studies of the mechanisms underlying Paneth cell function as an avenue toward preventing infection and treating inflammatory bowel diseases.
Collapse
Affiliation(s)
- Cristiano Rumio
- Department of Human Morphology and Biomedical Sciences Città Studi University of Milan, Milan, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Pahl R, Brunke G, Steubesand N, Schubert S, Böttner M, Wedel T, Jürgensen C, Hampe J, Schäfer H, Zeissig S, Schreiber S, Rosenstiel P, Reiss K, Arlt A. IL-1β and ADAM17 are central regulators of β-defensin expression in Candida esophagitis. Am J Physiol Gastrointest Liver Physiol 2011; 300:G547-53. [PMID: 21233274 DOI: 10.1152/ajpgi.00251.2010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Candida albicans resides on epithelial surfaces as part of the physiological microflora. However, under certain conditions, it may cause life-threatening infections, including Candida sepsis. We have recently shown that human β-defensins (hBDs) hBD-2 and hBD-3 are upregulated in Candida esophagitis and that this antifungal host response is distinctly regulated by NF-κB and MAPK/activator protein-1 (AP-1) pathways. Here, we show that C. albicans induces hBD-2 through an autocrine IL-1β loop and that activation of the epidermal growth factor receptor (EGFR) by endogenous transforming growth factor-α (TGF-α) is a crucial event in the induction of hBD-3. To further dissect upstream signaling events, we investigated expression of the central sheddases for EGFR ligands ADAM10 and ADAM17 in the healthy and infected esophagus. Next, we used pharmaceutical inhibitors and small-interfering RNA-mediated knock down of ADAM10 and ADAM17 to reveal that ADAM17-induced shedding of TGF-α is a crucial step in the induction of hBD-3 expression in response to Candida infection. In conclusion, we describe for the first time an autocrine IL-1β loop responsible for the induction of hBD-2 expression and an ADAM17-TGF-α-EGFR-MAPK/AP-1 pathway leading to hBD-3 upregulation in the course of a Candida infection of the esophagus.
Collapse
Affiliation(s)
- Rene Pahl
- Department of Internal Medicine I, University of Kiel, Germany University Hospital Schleswig-Holstein, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Kim HJ, Jung JR, Kim HJ, Lee SY, Chang IH, Lee TJ, Kim W, Myung SC. Expression of human β-defensin-2 in the prostate. BJU Int 2010; 107:144-9. [DOI: 10.1111/j.1464-410x.2010.09469.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
18
|
Jäger S, Stange EF, Wehkamp J. Antimicrobial peptides in gastrointestinal inflammation. Int J Inflam 2010; 2010:910283. [PMID: 21151692 PMCID: PMC2992817 DOI: 10.4061/2010/910283] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2010] [Accepted: 08/18/2010] [Indexed: 12/19/2022] Open
Abstract
Acute and chronic inflammations of mucosal surfaces are complex events in which the effector mechanisms of innate and adaptive immune systems interact with pathogenic and commensal bacteria. The role of constitutive and inducible antimicrobial peptides in intestinal inflammation has been investigated thoroughly over the recent years, and their involvement in various disease states is expanded ever more. Especially in the intestines, a critical balance between luminal bacteria and the antimicrobial peptides is essential, and a breakdown in barrier function by impaired production of defensins is already implicated in Crohn's disease. In this paper, we focus on the role of antimicrobial peptides in inflammatory processes along the gastrointestinal tract, while considering the resident and pathogenic flora encountered at the specific sites. The role of antimicrobial peptides in the primary events of inflammatory bowel diseases receives special attention.
Collapse
Affiliation(s)
- Simon Jäger
- Department of Internal Medicine I, Robert Bosch Hospital, Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Auerbachstr. 112, 70376 Stuttgart, Germany
| | | | | |
Collapse
|
19
|
Defensin-mRNA expression in the upper gastrointestinal tract is modulated in children with celiac disease and Helicobacter pylori-positive gastritis. J Pediatr Gastroenterol Nutr 2010; 50:596-600. [PMID: 20400909 DOI: 10.1097/mpg.0b013e3181cd26cd] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVES Defensins are expressed in epithelial cells as cationic peptides with antimicrobial properties. Because of their role as immunologically important effector molecules, their contribution in maintaining a stable microenvironment in the gastrointestinal tract has recently received much attention. The present study was designed to further characterize expression patterns of defensins in diseases of the upper gastrointestinal tract in children, particularly in Helicobacter pylori (Hp)-associated gastritis or celiac disease (CD). PATIENTS AND METHODS Semiquantitative real-time reverse transcriptase-polymerase chain reaction (PCR) was carried out with gene-specific primers for human beta-defensin 1 to 6 (hBD1 to 6) and human alpha-defensin 5 and 6 (hD5 and 6) in mucosal biopsies of children diagnosed as having CD (n = 11; 4.2-16.2 years) or Hp gastritis (n = 18; 3.2-16.7 years). Levels of expression were compared with those of healthy individuals (n = 21; 2.8-14.6 years). Expression levels in Hp-infected specimens were furthermore compared with those with histologic inflammation not associated with Hp infection (n = 30; 3.6-15.7 years). RESULTS Expression of hBD2 was upregulated in the antrum and corpus of patients with Hp gastritis, whereas inflammation without detection of Hp was not associated with any change in defensin gene expression. In patients with CD, expression of hBD2 was upregulated in the antrum, whereas hBD1 and 4 were downregulated in duodenal biopsies. CONCLUSIONS Different pathological conditions of the upper gastrointestinal tract lead to specific modulations of defensin gene expression in children. Especially the pathophysiological role of hBD2 in Hp infection and hBD1 and 4 in CD warrant further attention.
Collapse
|
20
|
Abstract
IMPORTANCE OF THE FIELD Inflammatory bowel diseases are chronic inflammatory diseases that comprise of two forms - Crohn's disease (CD) and ulcerative colitis (UC) - characterized by aberrant responses to luminal bacteria in genetically susceptible individuals. Whereas inflammation is limited to the large intestine in patients with UC, CD can affect all parts of the gastrointestinal tract. During disease exacerbations, pharmacological or surgical intervention is usually needed to re-establish remission; however, current therapeutic interventions cannot cure CD. As a subgroup of patients with CD will not be able to remain in remission with available drugs or suffer from side effects, new therapeutic strategies are needed. AREAS COVERED IN THIS REVIEW This review focuses on emerging drugs in the treatment of CD and reviews data on their efficacy and safety. An extensive review of the available literature was undertaken using MEDLINE to identify relevant studies. WHAT THE READER WILL GAIN The reader will learn about current therapeutic strategies in patients with CD and gain insights into emerging new drugs. TAKE HOME MESSAGE As modification of the clinical course of CD becomes the therapeutic paradigm, potential future treatments have to induce mucosal healing in order to prevent long-term complications. New biologics show promising results.
Collapse
Affiliation(s)
- Ulrike Strauch
- University Hospital of Regensburg, Department of Internal Medicine I, Franz-Josef-Strauss Allee 11, Regensburg 93042, Germany
| | | |
Collapse
|
21
|
Untersmayr E, Bises G, Starkl P, Bevins CL, Scheiner O, Boltz-Nitulescu G, Wrba F, Jensen-Jarolim E. The high affinity IgE receptor Fc epsilonRI is expressed by human intestinal epithelial cells. PLoS One 2010; 5:e9023. [PMID: 20126404 PMCID: PMC2814858 DOI: 10.1371/journal.pone.0009023] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2009] [Accepted: 01/14/2010] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND IgE antibodies play a paramount role in the pathogenesis of various intestinal disorders. To gain insights in IgE-mediated pathophysiology of the gut, we investigated the expression of the high affinity IgE receptor Fc epsilonRI in human intestinal epithelium. METHODOLOGY/PRINCIPAL FINDINGS Fc epsilonRI alpha-chain, as detected by immunohistochemistry, was positive in epithelial cells for eight of eleven (8/11) specimens from colon cancer patients and 5/11 patients with inflammation of the enteric mucosa. The Fc epsilonRIalpha positive epithelial cells co-expressed Fc epsilonRIgamma, whereas with one exception, none of the samples was positive for the beta-chain in the epithelial layer. The functionality of Fc epsilonRI was confirmed in situ by human IgE binding. In experiments with human intestinal tumor cell lines, subconfluent Caco-2/TC7 and HCT-8 cells were found to express the alpha- and gamma-chains of Fc epsilonRI and to bind IgE, whereas confluent cells were negative for gamma-chains. CONCLUSIONS/SIGNIFICANCE Our data provide the first evidence that the components of a functional Fc epsilonRI are in vitro expressed by the human intestinal epithelial cells depending on differentiation and, more importantly, in situ in epithelia of patients with colon cancer or gastrointestinal inflammations. Thus, a contribution of Fc epsilonRI either to immunosurveillance or pathophysiology of the intestinal epithelium is suggested.
Collapse
Affiliation(s)
- Eva Untersmayr
- Department of Pathophysiology, Center of Pathophysiology, Infectiology and Immunology, Medical University Vienna, Vienna, Austria.
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Wolfs TGAM, Derikx JPM, Hodin CMIM, Vanderlocht J, Driessen A, de Bruïne AP, Bevins CL, Lasitschka F, Gassler N, van Gemert WG, Buurman WA. Localization of the lipopolysaccharide recognition complex in the human healthy and inflamed premature and adult gut. Inflamm Bowel Dis 2010; 16:68-75. [PMID: 20014022 DOI: 10.1002/ibd.20995] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Microbiota in the intestinal lumen provide an abundant source of potentially detrimental antigens, including lipopolysaccharide (LPS), a potent immunostimulatory product of Gram-negative bacteria recognized by the host via TLR-4 and MD-2. An aberrant immune response to LPS or other bacterial antigens has been linked to inflammatory bowel disease (IBD) and necrotizing enterocolitis (NEC). METHODS We investigated which cells express MD-2 in the normal and inflamed ileum from neonates and adults by immunohistochemistry. Moreover, MD-2 and TLR4 mRNA expression in normal adult ileum was studied by reverse-transcription polymerase chain reaction (RT-PCR) on cells isolated by laser capture microdissection. RESULTS Premature infants did not show MD-2 expression either in epithelial cells or in the lamina propria. Similarly, MD-2 was absent in epithelial cells and lamina propria inflammatory cells in preterm infants with NEC. MD-2 protein in the healthy term neonatal and adult ileum was predominantly expressed by Paneth cells and some resident inflammatory cells in the lamina propria. MD-2 and TLR-4 mRNA expression was restricted to crypt cells. Also in IBD, Paneth cells were still the sole MD-2-expressing epithelial cells, whereas inflammatory cells (mainly plasma cells) were responsible for the vast majority of the MD-2 expression. CONCLUSIONS The absence of MD-2 in the immature neonatal gut suggests impaired LPS sensing, which could predispose neonates to NEC upon microbial colonization of the immature intestine. The apparent expression of MD-2 by Paneth cells supports the critical concept that these cells respond to luminal bacterial products in order to maintain homeostasis with the intestinal microbiota in vivo.
Collapse
Affiliation(s)
- Tim G A M Wolfs
- Department of Surgery, Nutrition and Toxicology Research Institute Maastricht (NUTRIM), Maastricht University Medical Centre, Maastricht, the Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Lapis K. Role of antimicrobial peptides (AMP) and pattern recognition receptors (PRR) in the intestinal mucosa homeostasis. Orv Hetil 2009; 150:2146-9. [DOI: 10.1556/oh.2009.28737] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Homeostasis and integrity of bowel mucosa is assured by well controlled mechanical, biochemical and immunological mechanisms. First line of defense is presented by the antimicrobial peptides (AMP), which form a continuous layer on the bowel surface, produced by intestinal specific (Paneth) and non-specific epithelial cells. AMPs have a significant antimicrobial, antifungal and antiviral, as well as immunomodulatory effects. Next line of defense is the pattern recognition receptors (PRR), which allows identifying conservative molecular patterns of different pathogens, and starts antimicrobial and inflammatory mechanisms through gene-expression induction. We review the most recent knowledge and studies concerning these mechanisms.
Collapse
Affiliation(s)
- Károly Lapis
- Semmelweis Egyetem, Általános Orvostudományi Kar I. Patológiai és Kísérleti Rákkutató Intézet Budapest Üllői út 26. 1085
| |
Collapse
|
24
|
Huynh D, Dai XM, Nandi S, Lightowler S, Trivett M, Chan CK, Bertoncello I, Ramsay RG, Stanley ER. Colony stimulating factor-1 dependence of paneth cell development in the mouse small intestine. Gastroenterology 2009; 137:136-144.e1443. [PMID: 19303020 PMCID: PMC2706482 DOI: 10.1053/j.gastro.2009.03.004] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2008] [Revised: 03/03/2009] [Accepted: 03/10/2009] [Indexed: 12/25/2022]
Abstract
BACKGROUND & AIMS Paneth cells (PCs) secrete defensins and antimicrobial enzymes that contribute to innate immunity against pathogen infections within the mucosa of the small intestine. We examined the role of colony stimulating factor-1 (CSF-1) in PC development. METHODS CSF-1-deficient and CSF-1 receptor (CSF-1R)-deficient mice and administration of neutralizing anti-CSF-1R antibody were used to study the requirement of CSF-1 for the development of epithelial cells of the small intestine. CSF-1 transgenic reporter mice and mice that express only the membrane-spanning, cell-surface CSF-1 isoform were used to investigate regulation by systemic versus local CSF-1. RESULTS Mice deficient in CSF-1 or CSF-1R had greatly reduced numbers of mature PCs. PCs express the CSF-1R, and administration of anti-CSF-1R antibody to neonatal mice significantly reduced the number of PCs. Analysis of transgenic CSF-1 reporter mice showed that CSF-1-expressing cells are in close proximity to PCs. CSF-1/CSF-1R-deficient mice also had reduced numbers of the proliferating epithelial cell progenitors and lamina propria macrophages. Expression of the membrane-spanning, cell-surface CSF-1 isoform in CSF-1-deficient mice completely rescued the deficiencies of PCs, proliferating progenitors, and lamina propria macrophages. CONCLUSIONS These results indicate local regulation by CSF-1 of PC development, either directly, in a juxtacrine/paracrine manner, or indirectly, by lamina propria macrophages. Therefore, CSF-1R hyperstimulation could be involved in hyperproliferative disorders of the small intestine, such as Crohn's disease and ulcerative colitis.
Collapse
Affiliation(s)
- Duy Huynh
- Peter MacCallum Cancer Centre, East Melbourne, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Lipopolysaccharide-binding protein: localization in secretory granules of Paneth cells in the mouse small intestine. Histochem Cell Biol 2009; 131:727-32. [DOI: 10.1007/s00418-009-0572-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/03/2009] [Indexed: 12/18/2022]
|
26
|
Andreu P, Peignon G, Slomianny C, Taketo MM, Colnot S, Robine S, Lamarque D, Laurent-Puig P, Perret C, Romagnolo B. A genetic study of the role of the Wnt/β-catenin signalling in Paneth cell differentiation. Dev Biol 2008; 324:288-96. [DOI: 10.1016/j.ydbio.2008.09.027] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2008] [Revised: 08/21/2008] [Accepted: 09/17/2008] [Indexed: 12/21/2022]
|
27
|
Schwab M, Reynders V, Loitsch S, Steinhilber D, Schröder O, Stein J. The dietary histone deacetylase inhibitor sulforaphane induces human beta-defensin-2 in intestinal epithelial cells. Immunology 2008; 125:241-251. [PMID: 18373608 PMCID: PMC2561129 DOI: 10.1111/j.1365-2567.2008.02834.x] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2007] [Revised: 02/13/2007] [Accepted: 02/15/2008] [Indexed: 12/17/2022] Open
Abstract
Antimicrobial peptides like human beta-defensin-2 (HBD-2) play an important role in the innate immune system protecting the intestinal mucosa against bacterial invasion. The dietary histone deacetylase (HDAC) inhibitors sulforaphane (SFN) and butyrate have received a great deal of attention because of their ability to simultaneously modulate multiple cellular targets involved in cellular protection. In this study the influence of SFN and butyrate on HBD-2 expression as well as the molecular pathways involved in SFN-mediated induction of HBD-2 were scrutinized. Treatment of Caco-2, HT-29 and SW480 cells with SFN led to a time- and dose-dependent upregulation of HBD-2 mRNA expression as determined by semi-quantitative reverse transcription-polymerase chain reaction. Moreover, HBD-2 protein production increased in response to SFN, measured by enzyme-linked immunosorbent assay. Induction of HBD-2 was also observed in response to butyrate. Immunofluorescence analysis revealed that the protein was localized in the cytosol. Coincubation of SFN with a vitamin D receptor (VDR), or an extracellular-regulated kinase 1/2 or a nuclear factor-kappaB inhibitor all reduced HBD-2 mRNA upregulation. In contrast, transfection of cells with a dominant-negative peroxisome proliferator-activated receptor gamma (PPARgamma) mutant vector to inhibit PPARgamma wild-type action and inhibition of p38 mitogen-activated protein kinase (MAPK) signalling did not affect SFN-mediated upregulation of HBD-2 mRNA. Moreover, SFN induced the expression of VDR, PPARgamma and phosphorylated ERK1/2 but did not affect p38 MAPK activation. The data clearly demonstrate for the first time that the dietary HDAC inhibitor SFN is able to induce antimicrobial peptides in colonocytes. In this process HBD-2 expression is regulated via VDR, mitogen-activated protein kinase kinase/extracellular-regulated kinase and nuclear factor-kappaB signalling.
Collapse
Affiliation(s)
- Markus Schwab
- First Department of Medicine-ZAFES, Johann Wolfgang Goethe-University FrankfurtFrankfurt am Main, Germany
| | - Veerle Reynders
- First Department of Medicine-ZAFES, Johann Wolfgang Goethe-University FrankfurtFrankfurt am Main, Germany
| | - Stefan Loitsch
- First Department of Medicine-ZAFES, Johann Wolfgang Goethe-University FrankfurtFrankfurt am Main, Germany
| | - Dieter Steinhilber
- Institute of Pharmaceutical Chemistry-ZAFES, Johann Wolfgang Goethe-University FrankfurtFrankfurt am Main, Germany
| | - Oliver Schröder
- First Department of Medicine-ZAFES, Johann Wolfgang Goethe-University FrankfurtFrankfurt am Main, Germany
| | - Jürgen Stein
- First Department of Medicine-ZAFES, Johann Wolfgang Goethe-University FrankfurtFrankfurt am Main, Germany
| |
Collapse
|
28
|
George MD, Wehkamp J, Kays RJ, Leutenegger CM, Sabir S, Grishina I, Dandekar S, Bevins CL. In vivo gene expression profiling of human intestinal epithelial cells: analysis by laser microdissection of formalin fixed tissues. BMC Genomics 2008; 9:209. [PMID: 18457593 PMCID: PMC2394537 DOI: 10.1186/1471-2164-9-209] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2007] [Accepted: 05/05/2008] [Indexed: 01/20/2023] Open
Abstract
Background The small intestinal epithelium mediates vital functions of nutrient absorption and host defense. The spatial organization of the epithelial cells along the crypt-villus axis segregates them into regions of specialized function. However, the differences in transcriptional programming and the molecular machinery that governs the migration, adhesion, and differentiation of intestinal epithelial cell lineages in humans remain under-explored. To increase our understanding of these mechanisms, we have evaluated gene expression patterns of ileal epithelial cells isolated by laser capture microdissection from either the villus epithelial or crypt cell regions of healthy human small intestinal mucosa. Expression profiles in villus and crypt epithelium were determined by DNA microarray, quantitative real-time PCR, and immunohistochemistry based methods. The expression levels of selected epithelial biomarkers were also compared between gastrointestinal tissues. Results Previously established biomarkers as well as a novel and distinct set of genes believed to be linked to epithelial cell motility, adhesion, and differentiation were found to be enriched in each of the two corresponding cell populations (GEO accession: GSE10629). Additionally, high baseline expression levels of innate antimicrobials, alpha defensin 5 (HD5) and regenerating islet-derived 3 alpha (Reg3A), were detected exclusively within the small bowel crypt, most notably in the ileum in comparison to other sites along the gastrointestinal tract. Conclusion The elucidation of differential gene expression patterns between crypt and villus epithelial cell lineages in human ileal tissue provides novel insights into the molecular machinery that mediates their functions and spatial organization. Moreover, our findings establish an important framework of knowledge for future investigations of human gastrointestinal diseases.
Collapse
Affiliation(s)
- Michael D George
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis CA 95616, USA.
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Abstract
PURPOSE OF REVIEW According to the current paradigm, ulcerative colitis and Crohn's disease occur in genetically predisposed individuals because of dysregulated immune responses against intraluminal bacterial antigens. Data have recently accumulated supporting alternative hypotheses for the pathogenesis of inflammatory bowel disease. Here, we present novel immunogenetic pathways and discuss their impact on traditional understanding of inflammatory bowel disease. RECENT FINDINGS In the gastrointestinal tract the innate immune system contains intraluminal bacteria locally, avoiding invasion of the deeper layers and preventing induction of long-standing proinflammatory responses. Failure of this protective function of the innate immune system appears to be the primary defect in inflammatory bowel disease, as a result of impairment of NOD2 signaling or other unidentified deficiencies. The adaptive immune response that ensues was thought to be strictly differentiated between T-helper-1 mediated in Crohn's disease and T-helper-2 mediated in ulcerative colitis. This concept is rapidly changing, however, in light of recent evidence suggesting that tissue injury in inflammatory bowel disease is mediated by novel effector pathways, the most prominent of which is the interleukin-23/Th17 axis. SUMMARY Elucidation of the pathways that underlie chronic intestinal inflammation will facilitate the development of new treatments with increased specificity and probably with decreased toxicity.
Collapse
Affiliation(s)
- Giorgos Bamias
- First Department of Propaedeutic Medicine, Laiko General Hospital, Athens, Greece, and Digestive Health Center of Excellence, University of Virginia Health System, Charlottesville, Virginia 22908, USA
| | | |
Collapse
|