201
|
Maharshak N, Packey CD, Ellermann M, Manick S, Siddle JP, Huh EY, Plevy S, Sartor RB, Carroll IM. Altered enteric microbiota ecology in interleukin 10-deficient mice during development and progression of intestinal inflammation. Gut Microbes 2013; 4:316-24. [PMID: 23822920 PMCID: PMC3744516 DOI: 10.4161/gmic.25486] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Inflammatory bowel diseases (IBD) result from dysregulated immune responses toward microbial and perhaps other luminal antigens in a genetically susceptible host, and are associated with altered composition and diversity of the intestinal microbiota. The interleukin 10-deficient (IL-10 (-/-) ) mouse has been widely used to model human IBD; however the specific alterations that occur in the intestinal microbiota of this mouse model during the onset of colonic inflammation have not yet been defined. The aim of our study was to define the changes in diversity and composition that occur in the intestinal microbiota of IL-10 (-/-) mice during the onset and progression of colonic inflammation. We used high throughput sequencing of the 16S rRNA gene to characterize the diversity and composition of formerly germ-free, wild-type and IL-10 (-/-) mice associated with the same intestinal microbiota over time. Following two weeks of colonization with a specific pathogen-free (SPF) microbiota we observed a significant increase in the diversity and richness of the intestinal microbiota of wild-type mice. In contrast, a progressive decrease in diversity and richness was observed at three and four weeks in IL-10 (-/-) mice. This decrease in diversity and richness was mirrored by an increase in Proteobacteria and Escherichia coli in IL-10 (-/-) mice. An increase in E. coli was also observed in conventionally raised IL-10 (-/-) mice at the point of colonic inflammation. Our data reports the sequential changes in diversity and composition of the intestinal microbiota in an immune-mediated mouse model that may help provide insights into the primary vs. secondary role of dysbiosis in human IBD patients.
Collapse
Affiliation(s)
- Nitsan Maharshak
- Division of Gastroenterology and Hepatology; Center for Gastrointestinal Biology and Disease; University of North Carolina at Chapel Hill; Chapel Hill, NC USA,Department of Gastroenterology and Liver diseases; Tel Aviv Sourasky Medical Center; Sackler School of Medicine; Tel Aviv University; Tel Aviv, Israel
| | - Christopher D. Packey
- Division of Gastroenterology and Hepatology; Center for Gastrointestinal Biology and Disease; University of North Carolina at Chapel Hill; Chapel Hill, NC USA,Department of Microbiology and Immunology; University of North Carolina at Chapel Hill; Chapel Hill, NC USA
| | - Melissa Ellermann
- Division of Gastroenterology and Hepatology; Center for Gastrointestinal Biology and Disease; University of North Carolina at Chapel Hill; Chapel Hill, NC USA,Department of Microbiology and Immunology; University of North Carolina at Chapel Hill; Chapel Hill, NC USA
| | - Sayeed Manick
- Division of Gastroenterology and Hepatology; Center for Gastrointestinal Biology and Disease; University of North Carolina at Chapel Hill; Chapel Hill, NC USA
| | - Jennica P. Siddle
- Division of Gastroenterology and Hepatology; Center for Gastrointestinal Biology and Disease; University of North Carolina at Chapel Hill; Chapel Hill, NC USA
| | - Eun Young Huh
- Division of Gastroenterology and Hepatology; Center for Gastrointestinal Biology and Disease; University of North Carolina at Chapel Hill; Chapel Hill, NC USA
| | - Scott Plevy
- Division of Gastroenterology and Hepatology; Center for Gastrointestinal Biology and Disease; University of North Carolina at Chapel Hill; Chapel Hill, NC USA,Department of Microbiology and Immunology; University of North Carolina at Chapel Hill; Chapel Hill, NC USA
| | - R. Balfour Sartor
- Division of Gastroenterology and Hepatology; Center for Gastrointestinal Biology and Disease; University of North Carolina at Chapel Hill; Chapel Hill, NC USA,Department of Microbiology and Immunology; University of North Carolina at Chapel Hill; Chapel Hill, NC USA
| | - Ian M. Carroll
- Division of Gastroenterology and Hepatology; Center for Gastrointestinal Biology and Disease; University of North Carolina at Chapel Hill; Chapel Hill, NC USA,Correspondence to: Ian M. Carroll,
| |
Collapse
|
202
|
Abstract
The microbiome has captured the attention of scientists from multiple research fields including ecology, immunology, microbiology and cancer biology. The microbial community living in the gastrointestinal tract is the most abundant and diverse niche of the human body and it is not surprising that microbiome research has predominantly focused upon this organ system. In this addendum, we summarize the latest developments in microbiome research on inflammatory bowel diseases and colorectal cancer. In addition, we highlight our recent findings that chronic intestinal inflammation modulates microbial community composition and the development of colorectal cancer. Our findings redefine the paradigm of inflammation-associated cancer by illuminating the key role of bacteria in development of colorectal cancer.
Collapse
|
203
|
|
204
|
Abstract
Although abundant indirect evidence suggests that environmental factors contribute to the pathogenesis of inflammatory bowel disease (IBD), the search for specific etiological agents has been largely disappointing. Early life factors have been hypothesized to influence the risk of IBD; however, new data from the Nurses Health Study showed no associations between breastfeeding, gestational age, and birthweight and adult-onset IBD.
Collapse
|
205
|
Diet, microbes, and host genetics: the perfect storm in inflammatory bowel diseases. J Gastroenterol 2013; 48:315-21. [PMID: 23475322 PMCID: PMC3698420 DOI: 10.1007/s00535-013-0777-2] [Citation(s) in RCA: 112] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Accepted: 01/28/2013] [Indexed: 02/07/2023]
Abstract
The incidence of inflammatory bowel diseases (IBD), as well as other inflammatory conditions, has dramatically increased over the past half century. While many studies have shown that IBD exhibits a genetic component via genome-wide association studies, genetic drift alone cannot account for this increase, and other factors, such as those found in the environment must play a role, suggesting a "multiple hit" phenomenon that precipitates disease. One major environmental factor, dietary intake, has shifted to a high fat, high carbohydrate Western-type diet in developing nations, nearly in direct correlation with the increasing incidence of IBD. Recent evidence suggests that specific changes in dietary intake have led to a shift in the composite human gut microbiota, resulting in the emergence of pathobionts that can thrive under specific conditions. In the genetically susceptible host, the emerging pathobionts can lead to increasing incidence and severity of IBD and other inflammatory disorders. Since the gut microbiota is plastic and responds to dietary modulations, the use of probiotics, prebiotics, and/or dietary alterations are all intriguing complementary therapeutic approaches to alleviate IBD symptoms. However, the interactions are complex and it is unlikely that a one-size-fits all approach can be utilized across all populations affected by IBD. Exploration into and thoroughly understanding the interactions between host and microbes, primarily in the genetically susceptible host, will help define strategies that can be tailored to an individual as we move towards an era of personalized medicine to treat IBD.
Collapse
|
206
|
Schirbel A, Kessler S, Rieder F, West G, Rebert N, Asosingh K, McDonald C, Fiocchi C. Pro-angiogenic activity of TLRs and NLRs: a novel link between gut microbiota and intestinal angiogenesis. Gastroenterology 2013; 144:613-623.e9. [PMID: 23149220 PMCID: PMC3578104 DOI: 10.1053/j.gastro.2012.11.005] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2012] [Revised: 10/12/2012] [Accepted: 11/07/2012] [Indexed: 12/23/2022]
Abstract
BACKGROUND & AIMS In intestinal inflammation the gut microbiota induces an innate immune response by activating epithelial and immune cells that initiate or maintain inflammation. We investigated whether the microbiota also can activate local microvascular cells and induce angiogenesis. METHODS Human intestinal microvascular endothelial cells (HIMEC) and human intestinal fibroblasts (HIF) were exposed to bacterial ligands specific for Toll-like receptor (TLR)2/6 and 4, and NOD1 and NOD2, and cell proliferation, migration, transmigration, tube formation, and production of pro-angiogenic factors were measured. The ability of the ligands to induce ex vivo vessel sprouting in an aortic ring assay and in vivo angiogenesis using a collagen gel assay also were assessed. RESULTS Bacterial ligands induced proliferation, migration, transmigration, tube formation of HIMEC, vessel sprouting, and in vivo angiogenesis; they also stimulated production of angiogenic factors from HIMEC and HIF, and HIF-derived angiogenic factors promoted HIMEC proliferation. To various degrees, all ligands induced angiogenic responses, but these were ligand- and cell type-dependent. Responses were mediated through receptor interacting protein-2 (RIP2)- and tumor necrosis factor receptor-associated factor 6 (TRAF6)-dependent signaling, involved the mitogen-activated protein kinase (MAPK) and nuclear factor-κB (NF-κB) pathways and the up-regulation of vascular endothelial growth factor receptor 2 (VEGF-R2) and focal adhesion kinase (FAK). Knockdown of RIP2 and TRAF6 by RNA interference and neutralization of interleukin-8, basic fibroblast growth factor, and vascular endothelial growth factor inhibited TLR-/NOD-like receptor-induced HIMEC angiogenesis. CONCLUSIONS The gut microbiota can selectively activate mucosal endothelial and mesenchymal cells to promote specific angiogenic responses in a TLR- and NOD-like receptor-dependent fashion. This innate immunity-mediated response may expand the mucosal microvascular network, foster immune cell recruitment, and contribute to chronic intestinal inflammation.
Collapse
Affiliation(s)
- Anja Schirbel
- Department of Pathobiology, Lerner Research Institute Cleveland Clinic Foundation, Cleveland, USA,Department of Hepatology and Gastroenterology, Campus Virchow-Klinikum, Charité -Universitätsmedizin, Berlin, Germany
| | - Sean Kessler
- Department of Pathobiology, Lerner Research Institute Cleveland Clinic Foundation, Cleveland, USA
| | - Florian Rieder
- Department of Pathobiology, Lerner Research Institute Cleveland Clinic Foundation, Cleveland, USA,Department of Gastroenterology, Digestive Disease Institute, Cleveland Clinic Foundation, Cleveland, USA
| | - Gail West
- Department of Pathobiology, Lerner Research Institute Cleveland Clinic Foundation, Cleveland, USA
| | - Nancy Rebert
- Department of Pathobiology, Lerner Research Institute Cleveland Clinic Foundation, Cleveland, USA
| | - Kewal Asosingh
- Department of Pathobiology, Lerner Research Institute Cleveland Clinic Foundation, Cleveland, USA
| | - Christine McDonald
- Department of Pathobiology, Lerner Research Institute Cleveland Clinic Foundation, Cleveland, USA
| | - Claudio Fiocchi
- Department of Pathobiology, Lerner Research Institute Cleveland Clinic Foundation, Cleveland, USA,Department of Gastroenterology, Digestive Disease Institute, Cleveland Clinic Foundation, Cleveland, USA
| |
Collapse
|
207
|
Draft Genome Sequence of Enterococcus faecalis PC1.1, a Candidate Probiotic Strain Isolated from Human Feces. GENOME ANNOUNCEMENTS 2013; 1:genomeA00160-12. [PMID: 23469340 PMCID: PMC3587934 DOI: 10.1128/genomea.00160-12] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Accepted: 12/21/2012] [Indexed: 12/24/2022]
Abstract
Enterococcus faecalis is commonly isolated from the gastrointestinal tract of healthy infants and adults, where it contributes to host health and well-being. We describe here the draft genome sequence of E. faecalis PC1.1, a candidate probiotic strain isolated from human feces.
Collapse
|
208
|
Wang X, Allen TD, Yang Y, Moore DR, Huycke MM. Cyclooxygenase-2 generates the endogenous mutagen trans-4-hydroxy-2-nonenal in Enterococcus faecalis-infected macrophages. Cancer Prev Res (Phila) 2013; 6:206-16. [PMID: 23321929 DOI: 10.1158/1940-6207.capr-12-0350] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Infection of macrophages by the human intestinal commensal Enterococcus faecalis generates DNA damage and chromosomal instability in mammalian cells through bystander effects. These effects are characterized by clastogenesis and damage to mitotic spindles in target cells and are mediated, in part, by trans-4-hydroxy-2-nonenal (4-HNE). In this study, we investigated the role of COX and lipoxygenase (LOX) in producing this reactive aldehyde using E. faecalis-infected macrophages and interleukin (IL)-10-knockout mice colonized with this commensal. 4-HNE production by E. faecalis-infected macrophages was significantly reduced by COX and LOX inhibitors. The infection of macrophages led to decreased Cox1 and Alox5 expression whereas COX-2 and 4-HNE increased. Silencing Alox5 and Cox1 with gene-specific siRNAs had no effect on 4-HNE production. In contrast, silencing Cox2 significantly decreased 4-HNE production by E. faecalis-infected macrophages. Depleting intracellular glutathione increased 4-HNE production by these cells. Next, to confirm COX-2 as a source for 4-HNE, we assayed the products generated by recombinant human COX-2 and found 4-HNE in a concentration-dependent manner using arachidonic acid as a substrate. Finally, tissue macrophages in colon biopsies from IL-10-knockout mice colonized with E. faecalis were positive for COX-2 by immunohistochemical staining. This was associated with increased staining for 4-HNE protein adducts in surrounding stroma. These data show that E. faecalis, a human intestinal commensal, can trigger macrophages to produce 4-HNE through COX-2. Importantly, it reinforces the concept of COX-2 as a procarcinogenic enzyme capable of damaging DNA in target cells through bystander effects that contribute to colorectal carcinogenesis.
Collapse
Affiliation(s)
- Xingmin Wang
- Veterans Affairs Medical Center, 921 N.E. 13 Street, Oklahoma City, OK 73104, USA.
| | | | | | | | | |
Collapse
|
209
|
Abstract
The human body is populated by an extremely diverse group of microbes that live in a symbiotic relationship with their host. Among these, intestinal commensals are the most abundant, induce homeostatic mucosal immune responses, and fulfill physiologic functions that benefit the host. In some cases, gut symbionts, including Escherichia coli, may contribute to the pathogenesis of chronic intestinal inflammation by causing dysregulated immune activation in genetically susceptible hosts. Although immune responses to bacterial products are well-characterized, the impact of intestinal inflammation on the function of commensal luminal microbes is only beginning to be elucidated. We recently reported that chronic intestinal inflammation induces commensal E. coli to upregulate stress response genes that paradoxically limit their growth in vivo. Herein, we discuss our findings in the context of host-microbial interactions in health and disease and a developing paradigm that may distinguish pathogens from commensals.
Collapse
Affiliation(s)
- Sandrine Tchaptchet
- Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, USA
| | | |
Collapse
|
210
|
Abstract
Recognition of microorganisms by pattern-recognition receptors (PRRs) is the primary component of innate immunity that is responsible for the maintenance of host-microbial interactions in intestinal mucosa. Dysregulation in host-commensal interactions has been implicated as the central pathogenesis of inflammatory bowel disease (IBD), which predisposes to developing colorectal cancer. Recent animal studies have begun to outline some unique physiology and pathology involving each PRR signaling in the intestine. The major roles played by PRRs in the gut appear to be the regulation of the number and the composition of commensal bacteria, epithelial proliferation, and mucosal permeability in response to epithelial injury. In addition, PRR signaling in lamina propria immune cells may be involved in induction of inflammation in response to invasion of pathogens. Because some PRR-deficient mice have shown variable susceptibility to colitis, the outcome of intestinal inflammation may be modified depending on PRR signaling in epithelial cells, immune cells, and the composition of commensal flora. Through recent findings in animal models of IBD, this review will discuss how abnormal PRR signaling may contribute to the pathogenesis of inflammation and inflammation-associated tumorigenesis in the intestine.
Collapse
|
211
|
Nemoto H, Kataoka K, Ishikawa H, Ikata K, Arimochi H, Iwasaki T, Ohnishi Y, Kuwahara T, Yasutomo K. Reduced diversity and imbalance of fecal microbiota in patients with ulcerative colitis. Dig Dis Sci 2012; 57:2955-64. [PMID: 22623042 DOI: 10.1007/s10620-012-2236-y] [Citation(s) in RCA: 114] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2011] [Accepted: 05/02/2012] [Indexed: 02/08/2023]
Abstract
BACKGROUND Clinical observations and experimental colitis models have indicated the importance of intestinal bacteria in the etiology of ulcerative colitis (UC), but a causative bacterial agent has not been identified. AIM To determine how intestinal bacteria are associated with UC, fecal microbiota and other components were compared for UC patients and healthy adults. METHODS Fresh feces were collected from 48 UC patients. Fecal microbiota were analyzed by use of terminal-restriction fragment length polymorphism (T-RFLP), real-time PCR, and culture. The concentrations of organic acids, indole, and ammonia, and pH and moisture, which are indicators of the intestinal environment, were measured and compared with healthy control data. RESULTS T-RFLP data divided the UC patients into four clusters; one cluster was obtained for healthy subjects. The diversity of fecal microbiota was significantly lower in UC patients. There were significantly fewer Bacteroides and Clostridium subcluster XIVab, and the amount of Enterococcus was higher in UC patients than in healthy subjects. The fecal concentration of organic acids was significantly lower in UC patients who were in remission. CONCLUSION UC patients have imbalances in the intestinal environment-less diversity of fecal microbiota, lower levels of major anaerobic bacteria (Bacteroides and Clostridium subcluster XIVab), and a lower concentration of organic acids.
Collapse
Affiliation(s)
- Hideyuki Nemoto
- Department of Immunology and Parasitology, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima 770-8503, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
212
|
Blumberg R, Powrie F. Microbiota, disease, and back to health: a metastable journey. Sci Transl Med 2012; 4:137rv7. [PMID: 22674557 PMCID: PMC5020897 DOI: 10.1126/scitranslmed.3004184] [Citation(s) in RCA: 220] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Alterations in the composition of the commensal microbiota have been observed in many complex diseases. Understanding the basis for these changes, how they relate to disease risk or activity, and the mechanisms by which the symbiotic state of colonization resistance and host homeostasis is restored is critical for future therapies aimed at manipulating the microbiota.
Collapse
Affiliation(s)
- Richard Blumberg
- Gastroenterology Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA.
| | | |
Collapse
|
213
|
Yang Y, Wang X, Moore DR, Lightfoot SA, Huycke MM. TNF-α mediates macrophage-induced bystander effects through Netrin-1. Cancer Res 2012; 72:5219-29. [PMID: 22915753 DOI: 10.1158/0008-5472.can-12-1463] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Macrophage-induced bystander effects have been implicated as an important mediator of chromosomal instability and colon cancer triggered by Enterococcus faecalis, a human intestinal commensal bacteria. There is little understanding about how inflammatory cytokines mediate bystander effects, but questions in this area are important because of the pivotal contributions made by inflammatory processes to cancer initiation and progression. Here, we report that the central proinflammatory cytokine TNF-α acts as a diffusible mediator of the bystander effects induced by macrophages, an effect caused by a proliferation of macrophages that trigger epithelial cell production of Netrin-1, a neuronal guidance molecule. TNF-α-mediated bystander assays used a murine coculture system of primary colonic epithelial cells and E. faecalis-infected macrophages (in vitro), with an interleukin 10 (IL-10)-deficient mouse model of colon cancer that involves long-term colonization with E. faecalis (in vivo). In cell cocultures, we observed increased expression of the TNF-α receptor Tnfrsf1b and Netrin-1. These effects were blocked by anti-TNF-α antibody or by pretreatment with an inhibitor of NF-κB signaling. RNAi-mediated attenuation of Tnfrsf1b decreased TNF-α-induced netrin-1 production and augmented epithelial cell apoptosis in culture. Extending these observations, colon biopsies from E. faecalis-colonized IL-10(-/-) mice exhibited crypt hyperplasia and increased staining for macrophages, TNF-α, netrin-1, NF-κB, Tnfrsf1b, and the proliferation marker proliferating cell nuclear antigen while also displaying a reduction in epithelial cell apoptosis. Together, our results define a pathway for macrophage-induced bystander effects in which TNF-α triggers TNFRSF1b receptor signaling leading to increased production of Netrin-1, crypt hyperplasia, and decreased epithelial cell apoptosis. In elucidating an important commensal-associated proinflammatory mechanism in the intestinal microenvironment, our work highlights the role of Netrin-1 and a specific TNF-α receptor as candidate targets to prevent or treat colorectal cancer.
Collapse
Affiliation(s)
- Yonghong Yang
- The Muchmore Laboratories for Infectious Diseases Research, Research Service, Department of Veterans Affairs Medical Center, Oklahoma City, OK 73104, USA
| | | | | | | | | |
Collapse
|
214
|
Arthur JC, Perez-Chanona E, Mühlbauer M, Tomkovich S, Uronis JM, Fan TJ, Campbell BJ, Abujamel T, Dogan B, Rogers AB, Rhodes JM, Stintzi A, Simpson KW, Hansen JJ, Keku TO, Fodor AA, Jobin C. Intestinal inflammation targets cancer-inducing activity of the microbiota. Science 2012; 338:120-3. [PMID: 22903521 DOI: 10.1126/science.1224820] [Citation(s) in RCA: 1640] [Impact Index Per Article: 126.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Inflammation alters host physiology to promote cancer, as seen in colitis-associated colorectal cancer (CRC). Here, we identify the intestinal microbiota as a target of inflammation that affects the progression of CRC. High-throughput sequencing revealed that inflammation modifies gut microbial composition in colitis-susceptible interleukin-10-deficient (Il10(-/-)) mice. Monocolonization with the commensal Escherichia coli NC101 promoted invasive carcinoma in azoxymethane (AOM)-treated Il10(-/-) mice. Deletion of the polyketide synthase (pks) genotoxic island from E. coli NC101 decreased tumor multiplicity and invasion in AOM/Il10(-/-) mice, without altering intestinal inflammation. Mucosa-associated pks(+) E. coli were found in a significantly high percentage of inflammatory bowel disease and CRC patients. This suggests that in mice, colitis can promote tumorigenesis by altering microbial composition and inducing the expansion of microorganisms with genotoxic capabilities.
Collapse
Affiliation(s)
- Janelle C Arthur
- Department of Medicine, Pharmacology and Immunology-Microbiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
215
|
The colitis-associated transcriptional profile of commensal Bacteroides thetaiotaomicron enhances adaptive immune responses to a bacterial antigen. PLoS One 2012; 7:e42645. [PMID: 22880065 PMCID: PMC3411805 DOI: 10.1371/journal.pone.0042645] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Accepted: 07/11/2012] [Indexed: 01/01/2023] Open
Abstract
Background Inflammatory bowel diseases (IBD) may be caused in part by aberrant immune responses to commensal intestinal microbes including the well-characterized anaerobic gut commensal Bacteroides thetaiotaomicron (B. theta). Healthy, germ-free HLA-B27 transgenic (Tg) rats develop chronic colitis when colonized with complex gut commensal bacteria whereas non-transgenic (nTg) rats remain disease-free. However, the role of B. theta in causing disease in Tg rats is unknown nor is much known about how gut microbes respond to host inflammation. Methods Tg and nTg rats were monoassociated with a human isolate of B. theta. Colonic inflammation was assessed by histologic scoring and tissue pro-inflammatory cytokine measurement. Whole genome transcriptional profiling of B. theta recovered from ceca was performed using custom GeneChips and data analyzed using dChip, Significance Analysis of Microarrays, and Gene Set Enrichment Analysis (GSEA) software. Western Blots were used to determine adaptive immune responses to a differentially expressed B. theta gene. Results B. theta monoassociated Tg rats, but not nTg or germ-free controls, developed chronic colitis. Transcriptional profiles of cecal B. theta were significantly different in Tg vs. nTg rats. GSEA revealed that genes in KEGG canonical pathways involved in bacterial growth and metabolism were downregulated in B. theta from Tg rats with colitis though luminal bacterial concentrations were unaffected. Bacterial genes in the Gene Ontology molecular function “receptor activity”, most of which encode nutrient binding proteins, were significantly upregulated in B. theta from Tg rats and include a SusC homolog that induces adaptive immune responses in Tg rats. Conclusions B. theta induces colitis in HLA-B27 Tg rats, which is associated with regulation of bacterial genes in metabolic and nutrient binding pathways that may affect host immune responses. These studies of the host-microbial dialogue may lead to the identification of novel microbial targets for IBD therapies.
Collapse
|
216
|
|
217
|
Abstract
PURPOSE OF REVIEW Human colitis-associated cancers (CAC) represent a heterogeneous group of conditions in which multiple oncogenic pathways are involved. In this article, we review the latest studies using genetic, chemical, bacterial and innate immune-mediated experimental models of CAC. RECENT FINDINGS Using the azoxymethane-dextran sodium sulfate model, wound healing pathways seem to be required in the development of CAC. There is also an emerging understanding that commensal and/or pathogenic bacteria can promote tumorigenesis, through T cell and TLR-mediated inflammation. Using specific transgenic mice (villin-CD98, T cell SMAD7, villin-TLR4) or specific knockout mice, investigators have determined that derangements in epithelial or innate and adaptive immune pathways can result in CAC. Subtle perturbations in epithelial repair - both too little or too exuberant - can render mice susceptible to tumorigenesis. SUMMARY With the aid of animal models, we have witnessed a rapid expansion of our knowledge of the molecular and immunologic mechanisms underlying inflammatory cancers. Though animal models have contributed a discrete amount of information to our understanding of tumorigenesis in the setting of intestinal inflammation, it is clear that no single animal model will be able to adequately recapitulate the pathogenesis of complex colorectal cancers, but each model gets us one step closer to comprehending the nature of CAC.
Collapse
|
218
|
Lee SE, Li X, Kim JCK, Lee J, González-Navajas JM, Hong SH, Park IK, Rhee JH, Raz E. Type I interferons maintain Foxp3 expression and T-regulatory cell functions under inflammatory conditions in mice. Gastroenterology 2012; 143:145-54. [PMID: 22475534 PMCID: PMC3729390 DOI: 10.1053/j.gastro.2012.03.042] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2011] [Revised: 03/12/2012] [Accepted: 03/15/2012] [Indexed: 12/22/2022]
Abstract
BACKGROUND & AIMS Foxp3(+) T-regulatory cells (Tregs) maintain intestinal homeostasis under conditions of continuous challenge with inflammatory microbes. However, plasticity of the Treg population under certain conditions has been reported; Foxp3(+) Tregs can be converted to Foxp3(-) CD4(+) T cells. METHODS We used mice with a T cell-induced colitis model to study the regulatory role of type I interferons (IFNs) in adaptive immunity. We transferred CD4(+)CD45RB(hi) (RB(hi)) T cells, with or without CD4(+)CD45RB(lo) CD25(+) T cells, from wild-type or IFN-αβR(-/-) mice into Rag1(-/-) recipients. We analyzed induction of colitis by flow cytometry, confocal microscopy, and enzyme-linked immunosorbent assay and reverse-transcription polymerase chain reaction analyses. IFN-αβR(-/-)Rag(-/-) mice were given injections of recombinant IFN-α following transfer of IFN-αβR(-/-) RB(hi) T cells and CD4(+)Foxp3(+) cells from Foxp3-eGFP mice. RESULTS Signaling by type I IFNs was required for maintenance of Foxp3 expression and the suppressive activity of Tregs in mice. Transfer of CD4(+)CD45RB(lo)CD25(+) Tregs from IFN-αβR(-/-) mice did not prevent T-cell induction of colitis in mice. Foxp3 expression by Tregs transferred from IFN-αβR(-/-) mice was significantly lower than that of Tregs from wild-type mice. Administration of recombinant IFN-α reduced T cell-mediated colitis by increasing the number of Foxp3(+) Tregs and their suppressive functions. CONCLUSIONS Type I IFNs regulate intestinal homeostasis by maintaining Foxp3 expression on Tregs in colons of mice under inflammatory conditions.
Collapse
Affiliation(s)
- Shee Eun Lee
- Department of Medicine, University of California San Diego, La Jolla, California 92093, USA.
| | - Xiangli Li
- Department of Medicine, University of California San Diego, La Jolla, California
| | - Joanna C. K. Kim
- Department of Medicine, University of California San Diego, La Jolla, California
| | - Jongdae Lee
- Department of Medicine, University of California San Diego, La Jolla, California
| | - Jose M. González-Navajas
- Department of Medicine, University of California San Diego, La Jolla, California,Biomedical Research Network Center (CIBERehd), National Institute of Health Carlos III, Madrid, Spain
| | - Seol Hee Hong
- Clinical Vaccine R&D Center, Chonnam National University, Gwangju, Korea,Dental Science Research Institute, Chonnam National University, Gwangju, Korea
| | - In-Kyu Park
- Clinical Vaccine R&D Center, Chonnam National University, Gwangju, Korea
| | - Joon Haeng Rhee
- Clinical Vaccine R&D Center, Chonnam National University, Gwangju, Korea
| | - Eyal Raz
- Department of Medicine, University of California San Diego, La Jolla, California
| |
Collapse
|
219
|
Abstract
The interaction of the host with its abundant intestinal microbiota is complex and engages most of the cells in the intestinal mucosa. The inflammatory bowel diseases appear to be disorders of the host immune response to the microbiota. This is supported by data from induced gene mutations in mice and more recently by the identification of gene variants in humans that result in IBD or IBD susceptibility. These genetic studies have provided insights into the cells and molecular pathways involved in the host-microbiota dialog. This review discusses the innate, adaptive, and regulatory immune response to the microbiota in the context of the mouse and human genes that are involved in maintaining intestinal homeostasis and preventing inflammation. These data continue to support the hypothesis that inflammatory bowel disease results from a dysregulated adaptive immune response, particularly a CD4 T-cell response, to the microbiota. The microbiota itself is an active participant in these homeostatic processes. The microbiota composition is perturbed during inflammation, resulting in a dysbiosis that may induce or perpetuate inflammation. However, host genotype and the environment have a major impact on the shape of such dysbiosis, as well as upon which members of the microbiota stimulate pathogenic immune responses.
Collapse
Affiliation(s)
- Charles O. Elson
- Departments of Medicine and Microbiology; University of Alabama at Birmingham; Birmingham, AL USA,Correspondence to: Charles O. Elson,
| | - Yingzi Cong
- Departments of Microbiology/Immunology and Pathology; University of Texas Medical Branch; Galveston, TX USA
| |
Collapse
|
220
|
Papa E, Docktor M, Smillie C, Weber S, Preheim SP, Gevers D, Giannoukos G, Ciulla D, Tabbaa D, Ingram J, Schauer DB, Ward DV, Korzenik JR, Xavier RJ, Bousvaros A, Alm EJ. Non-invasive mapping of the gastrointestinal microbiota identifies children with inflammatory bowel disease. PLoS One 2012; 7:e39242. [PMID: 22768065 PMCID: PMC3387146 DOI: 10.1371/journal.pone.0039242] [Citation(s) in RCA: 219] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2012] [Accepted: 05/17/2012] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Pediatric inflammatory bowel disease (IBD) is challenging to diagnose because of the non-specificity of symptoms; an unequivocal diagnosis can only be made using colonoscopy, which clinicians are reluctant to recommend for children. Diagnosis of pediatric IBD is therefore frequently delayed, leading to inappropriate treatment plans and poor outcomes. We investigated the use of 16S rRNA sequencing of fecal samples and new analytical methods to assess differences in the microbiota of children with IBD and other gastrointestinal disorders. METHODOLOGY/PRINCIPAL FINDINGS We applied synthetic learning in microbial ecology (SLiME) analysis to 16S sequencing data obtained from i) published surveys of microbiota diversity in IBD and ii) fecal samples from 91 children and young adults who were treated in the gastroenterology program of Children's Hospital (Boston, USA). The developed method accurately distinguished control samples from those of patients with IBD; the area under the receiver-operating-characteristic curve (AUC) value was 0.83 (corresponding to 80.3% sensitivity and 69.7% specificity at a set threshold). The accuracy was maintained among data sets collected by different sampling and sequencing methods. The method identified taxa associated with disease states and distinguished patients with Crohn's disease from those with ulcerative colitis with reasonable accuracy. The findings were validated using samples from an additional group of 68 patients; the validation test identified patients with IBD with an AUC value of 0.84 (e.g. 92% sensitivity, 58.5% specificity). CONCLUSIONS/SIGNIFICANCE Microbiome-based diagnostics can distinguish pediatric patients with IBD from patients with similar symptoms. Although this test can not replace endoscopy and histological examination as diagnostic tools, classification based on microbial diversity is an effective complementary technique for IBD detection in pediatric patients.
Collapse
Affiliation(s)
- Eliseo Papa
- Harvard/MIT Health Science and Technology Institute, Cambridge, Massachusetts, United States of America
| | - Michael Docktor
- Inflammatory Bowel Disease Center, Children’s Hospital Boston, Boston, Massachusetts, United States of America
| | - Christopher Smillie
- Computational and Systems Biology Initiative, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Sarah Weber
- Inflammatory Bowel Disease Center, Children’s Hospital Boston, Boston, Massachusetts, United States of America
| | - Sarah P. Preheim
- Department of Civil and Environmental Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Dirk Gevers
- The Broad Institute, 7 Cambridge Center, Cambridge, Massachusetts, United States of America
| | - Georgia Giannoukos
- The Broad Institute, 7 Cambridge Center, Cambridge, Massachusetts, United States of America
| | - Dawn Ciulla
- The Broad Institute, 7 Cambridge Center, Cambridge, Massachusetts, United States of America
| | - Diana Tabbaa
- The Broad Institute, 7 Cambridge Center, Cambridge, Massachusetts, United States of America
| | - Jay Ingram
- Inflammatory Bowel Disease Center, Children’s Hospital Boston, Boston, Massachusetts, United States of America
| | - David B. Schauer
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Doyle V. Ward
- The Broad Institute, 7 Cambridge Center, Cambridge, Massachusetts, United States of America
| | - Joshua R. Korzenik
- Gastrointestinal Unit, Center for Inflammatory Bowel Disease, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Ramnik J. Xavier
- The Broad Institute, 7 Cambridge Center, Cambridge, Massachusetts, United States of America
- Gastrointestinal Unit, Center for Inflammatory Bowel Disease, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Center for Computational and Integrative Biology, Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Athos Bousvaros
- Inflammatory Bowel Disease Center, Children’s Hospital Boston, Boston, Massachusetts, United States of America
| | - Eric J. Alm
- Department of Civil and Environmental Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- The Broad Institute, 7 Cambridge Center, Cambridge, Massachusetts, United States of America
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| |
Collapse
|
221
|
Benjamin JL, Hedin CRH, Koutsoumpas A, Ng SC, McCarthy NE, Prescott NJ, Pessoa-Lopes P, Mathew CG, Sanderson J, Hart AL, Kamm MA, Knight SC, Forbes A, Stagg AJ, Lindsay JO, Whelan K. Smokers with active Crohn's disease have a clinically relevant dysbiosis of the gastrointestinal microbiota. Inflamm Bowel Dis 2012; 18:1092-100. [PMID: 22102318 DOI: 10.1002/ibd.21864] [Citation(s) in RCA: 164] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Accepted: 07/24/2011] [Indexed: 12/11/2022]
Abstract
BACKGROUND Patients with Crohn's disease (CD) have an intestinal dysbiosis with components of the microbiota exerting differential immune effects. Smoking is associated with an increased incidence of CD, more frequent relapse, and greater burden of surgery. This study aimed to investigate the association between smoking and the intestinal microbiota in patients with active CD. METHODS Patients with active CD (n = 103) and healthy controls (n = 66) were recruited and demographic and clinical data recorded including current smoking behavior. Fecal samples were collected and analyzed by fluorescent in situ hybridization using probes targeting 16S rRNA of bacteria previously shown to be altered in active CD (bifidobacteria, bacteroides, Clostridium coccoides-Eubacterium rectale, Escherichia coli, and Faecalibacterium prausnitzii). RESULTS In total, 29/101 (29%) patients with CD and 8/58 (14%) controls were current smokers (P = 0.032). Following multivariate analysis, smoking was found to have a significant and independent effect on the microbiota of patients with CD, with higher Bacteroides-Prevotella in smokers (38.4%) compared with nonsmokers (28.1%) (F((1,93)) = 12.6, P = 0.001). Healthy controls who smoked also had higher Bacteroides-Prevotella (34.8%) than nonsmokers (24.1%) (F((1,55)) = 4.5, P = 0.038). In the pooled multivariate analysis, patients with CD had higher bifidobacteria (F((1,156)) = 30.5, P < 0.001), higher Bacteroides-Prevotella (F((1,156)) = 6.5, P = 0.012), and lower F. prausnitzii (F((1,156)) = 3.8, P = 0.052) compared with healthy controls. CONCLUSIONS Smokers have luminal microbiota that consist of significantly higher bacteroides. Investigation of whether this is one mechanism through which the negative effects of smoking on CD are mediated is warranted.
Collapse
Affiliation(s)
- Jane L Benjamin
- King's College London, School of Medicine, Diabetes and Nutritional Sciences Division, London, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
222
|
Chinen T, Rudensky AY. The effects of commensal microbiota on immune cell subsets and inflammatory responses. Immunol Rev 2012; 245:45-55. [PMID: 22168413 DOI: 10.1111/j.1600-065x.2011.01083.x] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Billions of years of coevolution shaped the mutually beneficial relationships between metazoans and symbiotic commensal microorganisms. Commensal microorganisms profoundly affect the physiology of the host and provide the host with survival advantages in several ways, while they could also trigger pathogenic immune responses and threaten the well-being of the host. Recent advances in DNA sequencing technology enabled the analysis of commensal microbiota, and improvements in the techniques of culturing gut-resident microorganisms and of rearing gnotobiotic rodents have made it possible to assess the effect of individual component of microbial communities on host physiology. In this review, we discuss the current understanding of the interactions of commensal microbiota with the host immune system.
Collapse
Affiliation(s)
- Takatoshi Chinen
- Howard Hughes Medical Institute and Immunology Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA
| | | |
Collapse
|
223
|
Wang X, Yang Y, Moore DR, Nimmo SL, Lightfoot SA, Huycke MM. 4-hydroxy-2-nonenal mediates genotoxicity and bystander effects caused by Enterococcus faecalis-infected macrophages. Gastroenterology 2012; 142:543-551.e7. [PMID: 22108198 PMCID: PMC3371374 DOI: 10.1053/j.gastro.2011.11.020] [Citation(s) in RCA: 292] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2011] [Revised: 10/31/2011] [Accepted: 11/07/2011] [Indexed: 12/02/2022]
Abstract
BACKGROUND & AIMS Enterococcus faecalis is a human intestinal commensal that produces extracellular superoxide and promotes chromosome instability via macrophage-induced bystander effects. We investigated the ability of 4-hydroxy-2-nonenal (4-HNE), a diffusible breakdown product of ω-6 polyunsaturated fatty acids, to mediate these effects. METHODS 4-HNE was purified from E faecalis-infected macrophages; its genotoxicity was assessed in human colon cancer (HCT116) and primary murine colon epithelial (YAMC) cell lines. RESULTS 4-HNE induced G(2)-M cell cycle arrest, led to formation γH2AX foci, and disrupted the mitotic spindle in both cell lines. Binucleate tetraploid cells that formed after incubation with 4-HNE were associated with the activation of stathmin and microtubule catastrophe. Silencing glutathione S-transferase α4, a scavenger of 4-HNE, increased the susceptibility of epithelial cells to 4-HNE-induced genotoxicity. Interleukin-10 knockout mice colonized with superoxide-producing E faecalis developed inflammation and colorectal cancer, whereas colonization with a superoxide-deficient strain resulted in inflammation but not cancer. 4-HNE-protein adducts were found in the lamina propria and macrophages in areas of colorectal inflammation. CONCLUSIONS 4-HNE can act as an autochthonous mitotic spindle poison in normal colonic epithelial and colon cancer cells. This finding links the macrophage-induced bystander effects to colorectal carcinogenesis.
Collapse
Affiliation(s)
- Xingmin Wang
- The Muchmore Laboratories for Infectious Diseases Research, Research Service, Oklahoma City, OK 73104,Department of Radiation Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104
| | - Yonghong Yang
- The Muchmore Laboratories for Infectious Diseases Research, Research Service, Oklahoma City, OK 73104,Department of Radiation Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104
| | - Danny R. Moore
- The Muchmore Laboratories for Infectious Diseases Research, Research Service, Oklahoma City, OK 73104,Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104
| | - Susan L. Nimmo
- Department of Chemistry & Biochemistry, University of Oklahoma, Norman, OK 73019, USA
| | - Stanley A. Lightfoot
- Pathology and Laboratory Service, Department of Veterans Affairs Medical Center, Oklahoma City, OK 73104,Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104
| | - Mark M. Huycke
- The Muchmore Laboratories for Infectious Diseases Research, Research Service, Oklahoma City, OK 73104,Department of Radiation Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104,Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104,Corresponding author: Mark M. Huycke, M.D., Veterans Affairs Medical Center, 921 N.E. 13th Street, Oklahoma City, OK 73104,
| |
Collapse
|
224
|
Presley LL, Ye J, Li X, LeBlanc J, Zhang Z, Ruegger PM, Allard J, McGovern D, Ippoliti A, Roth B, Cui X, Jeske DR, Elashoff D, Goodglick L, Braun J, Borneman J. Host-microbe relationships in inflammatory bowel disease detected by bacterial and metaproteomic analysis of the mucosal-luminal interface. Inflamm Bowel Dis 2012; 18:409-17. [PMID: 21698720 PMCID: PMC3179764 DOI: 10.1002/ibd.21793] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Accepted: 05/16/2011] [Indexed: 12/16/2022]
Abstract
BACKGROUND Host-microbe interactions at the intestinal mucosal-luminal interface (MLI) are critical factors in the biology of inflammatory bowel disease (IBD). METHODS To address this issue, we performed a series of investigations integrating analysis of the bacteria and metaproteome at the MLI of Crohn's disease, ulcerative colitis, and healthy human subjects. After quantifying these variables in mucosal specimens from a first sample set, we searched for bacteria exhibiting strong correlations with host proteins. This assessment identified a small subset of bacterial phylotypes possessing this host interaction property. Using a second and independent sample set, we tested the association of disease state with levels of these 14 "host interaction" bacterial phylotypes. RESULTS A high frequency of these bacteria (35%) significantly differentiated human subjects by disease type. Analysis of the MLI metaproteomes also yielded disease classification with exceptional confidence levels. Examination of the relationships between the bacteria and proteins, using regularized canonical correlation analysis (RCCA), sorted most subjects by disease type, supporting the concept that host-microbe interactions are involved in the biology underlying IBD. Moreover, this correlation analysis identified bacteria and proteins that were undetected by standard means-based methods such as analysis of variance, and identified associations of specific bacterial phylotypes with particular protein features of the innate immune response, some of which have been documented in model systems. CONCLUSIONS These findings suggest that computational mining of mucosa-associated bacteria for host interaction provides an unsupervised strategy to uncover networks of bacterial taxa and host processes relevant to normal and disease states. (Inflamm Bowel Dis 2012;).
Collapse
Affiliation(s)
- Laura L. Presley
- Department of Plant Pathology and Microbiology, University of California, Riverside, CA, 92521, USA
| | - Jingxiao Ye
- Department of Plant Pathology and Microbiology, University of California, Riverside, CA, 92521, USA
| | - Xiaoxiao Li
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - James LeBlanc
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Zhanpan Zhang
- Department of Statistics, University of California, Riverside, CA, 92521, USA
| | - Paul M. Ruegger
- Department of Plant Pathology and Microbiology, University of California, Riverside, CA, 92521, USA
| | - Jeff Allard
- Department of Medicine, Division of Digestive Disease, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Dermot McGovern
- Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Andrew Ippoliti
- Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Bennett Roth
- Department of Medicine, Division of Digestive Disease, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Xinping Cui
- Department of Statistics, University of California, Riverside, CA, 92521, USA
| | - Daniel R. Jeske
- Department of Statistics, University of California, Riverside, CA, 92521, USA
| | - David Elashoff
- Department of Human Genetics and Department of Biostatistics, Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Lee Goodglick
- Department of Pathology and Laboratory Medicine, Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Jonathan Braun
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
- Department of Pathology and Laboratory Medicine, Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - James Borneman
- Department of Plant Pathology and Microbiology, University of California, Riverside, CA, 92521, USA
| |
Collapse
|
225
|
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
|
226
|
Smith P, Siddharth J, Pearson R, Holway N, Shaxted M, Butler M, Clark N, Jamontt J, Watson RP, Sanmugalingam D, Parkinson SJ. Host genetics and environmental factors regulate ecological succession of the mouse colon tissue-associated microbiota. PLoS One 2012; 7:e30273. [PMID: 22272321 PMCID: PMC3260280 DOI: 10.1371/journal.pone.0030273] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2011] [Accepted: 12/12/2011] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND The integration of host genetics, environmental triggers and the microbiota is a recognised factor in the pathogenesis of barrier function diseases such as IBD. In order to determine how these factors interact to regulate the host immune response and ecological succession of the colon tissue-associated microbiota, we investigated the temporal interaction between the microbiota and the host following disruption of the colonic epithelial barrier. METHODOLOGY/PRINCIPAL FINDINGS Oral administration of DSS was applied as a mechanistic model of environmental damage of the colon and the resulting inflammation characterized for various parameters over time in WT and Nod2 KO mice. RESULTS In WT mice, DSS damage exposed the host to the commensal flora and led to a migration of the tissue-associated bacteria from the epithelium to mucosal and submucosal layers correlating with changes in proinflammatory cytokine profiles and a progressive transition from acute to chronic inflammation of the colon. Tissue-associated bacteria levels peaked at day 21 post-DSS and declined thereafter, correlating with recruitment of innate immune cells and development of the adaptive immune response. Histological parameters, immune cell infiltration and cytokine biomarkers of inflammation were indistinguishable between Nod2 and WT littermates following DSS, however, Nod2 KO mice demonstrated significantly higher tissue-associated bacterial levels in the colon. DSS damage and Nod2 genotype independently regulated the community structure of the colon microbiota. CONCLUSIONS/SIGNIFICANCE The results of these experiments demonstrate the integration of environmental and genetic factors in the ecological succession of the commensal flora in mammalian tissue. The association of Nod2 genotype (and other host polymorphisms) and environmental factors likely combine to influence the ecological succession of the tissue-associated microflora accounting in part for their association with the pathogenesis of inflammatory bowel diseases.
Collapse
Affiliation(s)
- Philip Smith
- Novartis Institutes for Biomedical Research, Gastrointestinal Disease Area, Horsham, West Sussex, United Kingdom
| | - Jay Siddharth
- Novartis Institutes for Biomedical Research, Gastrointestinal Disease Area, Horsham, West Sussex, United Kingdom
| | - Ruth Pearson
- Novartis Institutes for Biomedical Research, Gastrointestinal Disease Area, Horsham, West Sussex, United Kingdom
| | - Nicholas Holway
- Novartis Institutes for Biomedical Research Information Technology, Horsham, West Sussex, United Kingdom
| | - Mark Shaxted
- Novartis Institutes for Biomedical Research, Gastrointestinal Disease Area, Horsham, West Sussex, United Kingdom
| | - Matt Butler
- Novartis Institutes for Biomedical Research, Gastrointestinal Disease Area, Horsham, West Sussex, United Kingdom
| | - Natalie Clark
- Novartis Institutes for Biomedical Research, Gastrointestinal Disease Area, Horsham, West Sussex, United Kingdom
| | - Joanna Jamontt
- Novartis Institutes for Biomedical Research, Gastrointestinal Disease Area, Horsham, West Sussex, United Kingdom
| | - Robert P. Watson
- Novartis Institutes for Biomedical Research, Gastrointestinal Disease Area, Horsham, West Sussex, United Kingdom
| | - Devika Sanmugalingam
- Novartis Institutes for Biomedical Research, Gastrointestinal Disease Area, Horsham, West Sussex, United Kingdom
| | - Scott J. Parkinson
- Novartis Institutes for Biomedical Research, Gastrointestinal Disease Area, Horsham, West Sussex, United Kingdom
- Novartis Institutes for Biomedical Research, Developmental and Molecular Pathways, Basel, Switzerland
- * E-mail:
| |
Collapse
|
227
|
Bleich A, Hansen AK. Time to include the gut microbiota in the hygienic standardisation of laboratory rodents. Comp Immunol Microbiol Infect Dis 2012; 35:81-92. [PMID: 22257867 DOI: 10.1016/j.cimid.2011.12.006] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2011] [Revised: 11/17/2011] [Accepted: 12/19/2011] [Indexed: 02/06/2023]
Abstract
The gut microbiota (GM) composition and its impact on animal experiments has become currently dramatically relevant in our days: (1) recent progress in metagenomic technologies, (2) the availability of large scale quantitative analyses to characterize even subtle phenotypes, (3) the limited diversity of laboratory rodent GM due to strict barriers at laboratory animal vendors, and (4) the availability of up to 300.000 different transgenic mouse strains from different sources displaying a huge variety in their GM composition. In this review the GM is described as a variable in animal experiments which need to be reduced for scientific as well as ethical reasons, and strategies how to implement this in routine diagnostic procedures are proposed. We conclude that we have both enough information available to state that the GM has an essential impact on animal models, as well as the methods available to start dealing with these impacts.
Collapse
Affiliation(s)
- André Bleich
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany, Hannover, Germany.
| | | |
Collapse
|
228
|
Sun X, Threadgill D, Jobin C. Campylobacter jejuni induces colitis through activation of mammalian target of rapamycin signaling. Gastroenterology 2012; 142:86-95.e5. [PMID: 21963787 PMCID: PMC3253301 DOI: 10.1053/j.gastro.2011.09.042] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2011] [Revised: 08/30/2011] [Accepted: 09/20/2011] [Indexed: 12/02/2022]
Abstract
BACKGROUND & AIMS Campylobacter jejuni is the worldwide leading cause of bacterial-induced enteritis. The molecular and cellular events that lead to campylobacteriosis are poorly understood. We identify mammalian target of rapamycin (mTOR) as a signaling pathway that leads to C jejuni-induced intestinal inflammation. METHODS Germ-free (control) or conventionally derived Il10(-/-) mice that express enhanced green fluorescent protein (EGFP) under the control of nuclear factor κB (Il10(-/-); NF-κB(EGFP) mice) were infected with C jejuni (10(9) colony-forming units/mouse) for 12 days; their responses were determined using histologic, semiquantitative reverse-transcription polymerase chain reaction, fluorescence in situ hybridization, transmission electron microscopy, and tissue culture analyses. mTOR signaling was blocked by daily intraperitoneal injections of the pharmacologic inhibitor rapamycin (1.5 mg/kg). CD4(+) T cells were depleted by intraperitoneal injections of antibodies against CD4 (0.5 mg/mouse every 3 days). Bacterial survival in splenocytes was measured using a gentamycin killing assay. RESULTS C jejuni induced intestinal inflammation, which correlated with activation of mTOR signaling and neutrophil infiltration. The inflamed intestines of these mice had increased levels of interleukin-1β, Cxcl2, interleukin-17a, and EGFP; C jejuni localized to colons and extraintestinal tissues of infected Il10(-/-); NF-κB(EGFP) mice compared with controls. Rapamycin, administered before or after introduction of C jejuni, blocked C jejuni-induced intestinal inflammation and bacterial accumulation. LC3II processing and killing of C jejuni were increased in splenocytes incubated with rapamycin compared with controls. CONCLUSIONS mTOR signaling mediates C jejuni-induced colitis in Il10(-/-) mice, independently of T-cell activation. Factors involved in mTOR signaling might be therapeutic targets for campylobacteriosis.
Collapse
Affiliation(s)
- Xiaolun Sun
- Department of Medicine, University of North Carolina, Chapel Hill, NC
| | - Deborah Threadgill
- Department of Microbiology, North Carolina State University, Raleigh, NC
| | - Christian Jobin
- Department of Medicine, University of North Carolina, Chapel Hill, NC,Department of Pharmacology, University of North Carolina, Chapel Hill, NC,Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC
| |
Collapse
|
229
|
Dextran sodium sulfate-induced inflammation alters the expression of proteins by intestinal Escherichia coli strains in a gnotobiotic mouse model. Appl Environ Microbiol 2011; 78:1513-22. [PMID: 22210207 DOI: 10.1128/aem.07340-11] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
To identify Escherichia coli proteins involved in adaptation to intestinal inflammation, mice were monoassociated with the colitogenic E. coli strain UNC or with the probiotic E. coli strain Nissle. Intestinal inflammation was induced by treating the mice with 3.5% dextran sodium sulfate (DSS). Differentially expressed proteins in E. coli strains collected from cecal contents were identified by 2-dimensional difference gel electrophoresis. In both strains, acute inflammation led to the downregulation of pathways involved in carbohydrate breakdown and energy generation. Accordingly, DSS-treated mice had lower concentrations of bacterial fermentation products in their cecal contents than control mice. Differentially expressed proteins also included the Fe-S cluster repair protein NfuA, the tryptophanase TnaA, and the uncharacterized protein YggE. NfuA expression was 3-fold higher in E. coli strains from DSS-treated than from control mice. Reporter experiments confirmed the induction of nfuA in response to iron deprivation, mimicking Fe-S cluster destruction by inflammation. YggE expression, which has been reported to reduce the intracellular level of reactive oxygen species, was 4- to 8-fold higher in E. coli Nissle than in E. coli UNC. This was confirmed by in vitro reporter gene assays indicating that Nissle is better equipped to cope with oxidative stress than UNC. Nissle isolated from DSS-treated and control mice had TnaA levels 4- to 7-fold-higher than those of UNC. Levels of indole resulting from the TnaA reaction were higher in control animals associated with E. coli Nissle. Because of its anti-inflammatory effect, indole is hypothesized to be involved in the extension of the remission phase in ulcerative colitis described for E. coli Nissle.
Collapse
|
230
|
Patwa LG, Fan TJ, Tchaptchet S, Liu Y, Lussier YA, Sartor RB, Hansen JJ. Chronic intestinal inflammation induces stress-response genes in commensal Escherichia coli. Gastroenterology 2011; 141:1842-51.e1-10. [PMID: 21726510 PMCID: PMC3624969 DOI: 10.1053/j.gastro.2011.06.064] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2010] [Revised: 06/13/2011] [Accepted: 06/21/2011] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Intestinal microbes induce homeostatic mucosal immune responses, but can also cause inappropriate immune activation in genetically susceptible hosts. Although immune responses to bacterial products have been studied extensively, little is known about how intestinal inflammation affects functions of commensal luminal microbes. METHODS Microarrays and real-time polymerase chain reaction were used to profile transcriptional changes in luminal bacteria from wild-type and IL-10(-/-) mice monoassociated with a nonpathogenic, murine isolate of Escherichia coli (NC101, which causes colitis in gnotobiotic IL-10(-/-) mice). Colonic inflammation and innate and adaptive immune responses were measured in wild-type and IL-10(-/-) mice monoassociated with mutant NC101 that lack selected, up-regulated genes, and in IL-10(-/-) mice that were colonized with a combination of mutant and parental NC101. We measured intracellular survival of bacteria within primary macrophages from mice and resulting production of tumor necrosis factor. RESULTS Bacteria from IL-10(-/-) mice with colitis had significant up-regulation of the stress-response regulon, including the small heat shock proteins IbpA and IbpB that protect E coli from oxidative stress, compared to healthy, wild-type controls. In IL-10(-/-) mice, expression of ibpAB reduced histologic signs of colon inflammation, secretion of interleukin-12/23p40 in colonic explant cultures, serologic reactivity to NC101 antigens, and secretion of interferon-gamma by stimulated mesenteric lymph node cells. Infection of primary macrophages by bacteria that express ibpAB was associated with decreased intracellular survival and reduced secretion of tumor necrosis factor. CONCLUSIONS Chronic intestinal inflammation causes functional alterations in gene expression in commensal gut bacterium (E coli NC101). Further studies of these expression patterns might identify therapeutic targets for patients with inflammatory bowel diseases.
Collapse
Affiliation(s)
- Laura G. Patwa
- Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA,Department of Medicine, Division of Gastroenterology and Hepatology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Ting-Jia Fan
- Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA,Department of Medicine, Division of Gastroenterology and Hepatology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Sandrine Tchaptchet
- Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA,Department of Medicine, Division of Gastroenterology and Hepatology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yang Liu
- Department of Medicine, Center for Biomedical Informatics and Section of Genetic Medicine, University of Chicago, Chicago, IL 60637, USA,Institute for Genomics and Systems Biology and Institute for Translational Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Yves A. Lussier
- Department of Medicine, Center for Biomedical Informatics and Section of Genetic Medicine, University of Chicago, Chicago, IL 60637, USA,Institute for Genomics and Systems Biology and Institute for Translational Medicine, University of Chicago, Chicago, IL 60637, USA
| | - R. Balfour Sartor
- Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA,Department of Medicine, Division of Gastroenterology and Hepatology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA,Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jonathan J. Hansen
- Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA,Department of Medicine, Division of Gastroenterology and Hepatology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
231
|
Abstract
Inflammatory bowel disease is a chronic inflammatory disease of the gut which manifests as ulcerative colitis or Crohn's disease. One of the most studied animal models of spontaneous Crohn's disease is the senescence-accelerated mouse (SAMP1/Yit strain) model. In SAMP1/Yit mice, although many immunological responses are perturbed, some evidence suggests that the primary defect lies in the epithelial cell barrier. In the process of studying epithelial permeability, we observed that the stomach in SAMP1/Yit mice also had increased permeability. Upon further examination, these mice were shown to have marked, chronic gastritis with focal to diffuse aggregates of mononuclear cells of mixed lineages. These aggregates were located predominantly in the oxyntic mucosa, with occasional lesions in the forestomach but with relatively fewer cellular infiltrates in the antral mucosa. Real-time RT PCR showed an increase in several helper T cell (Th cell)-derived pro-inflammatory cytokines in the gastric mucosa of SAMP1/Yit mice. However, many of the cells in the aggregates of SAMP1/Yit mice were B cells. SAMP1/Yit B cells exacerbate ileitis when co-transferred into immunodeficient recipients. The gastritis also reflects a contribution by B cells. As SAMP1/Yit mice were derived from AKR mice, we examined AKR mice and determined that they too have an increased occurrence of gastritis, although they do not develop ileitis. B cells contributed to the gastric inflammation in these mice also. Thus, SAMP1/Yit mice display gastritis as well as ileitis, and B cells appear to play a role in the pathogenesis of inflammation at both sites. This review will discuss some of the mechanisms that may account for these different manifestations of gastrointestinal disease.
Collapse
|
232
|
Mahendran V, Riordan SM, Grimm MC, Tran TAT, Major J, Kaakoush NO, Mitchell H, Zhang L. Prevalence of Campylobacter species in adult Crohn's disease and the preferential colonization sites of Campylobacter species in the human intestine. PLoS One 2011; 6:e25417. [PMID: 21966525 PMCID: PMC3179513 DOI: 10.1371/journal.pone.0025417] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2011] [Accepted: 09/02/2011] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION Crohn's disease (CD) and ulcerative colitis (UC) are the two major forms of inflammatory bowel disease (IBD). A high prevalence of Campylobacter concisus was previously detected in paediatric CD and adult UC. Currently, the prevalence of C. concisus in adult CD and the preferential colonization sites of Campylobacter species in the human intestine are unknown. In this study, we examined the prevalence of Campylobacter species in biopsies collected from multiple anatomic sites of adult patients with IBD and controls. METHODS Three hundred and one biopsies collected from ileum, caecum, descending colon and rectum of 28 patients IBD (15 CD and 13 UC) and 33 controls were studied. Biopsies were used for DNA extraction and detection of Campylobacter species by PCR-sequencing and Campylobacter cultivation. RESULTS A significantly higher prevalence of C. concisus in colonic biopsies of patients with CD (53%) was detected as compared with the controls (18%). Campylobacter genus-PCR positivity and C. concisus positivity in patients with UC were 85% and 77% respectively, being significantly higher than that in the controls (48% and 36%). C. concisus was more often detected in descending colonic and rectal biopsies from patients with IBD in comparison to the controls. C. concisus was isolated from patients with IBD. CONCLUSION The high intestinal prevalence of C. concisus in patients with IBD, particularly in the proximal large intestine, suggests that future studies are needed to investigate the possible involvement of C. concisus in a subgroup of human IBD. To our knowledge, this is the first report of the association between adult CD and C. concisus as well as the first study of the preferential colonization sites of C. concisus in the human intestine.
Collapse
Affiliation(s)
- Vikneswari Mahendran
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| | - Stephen M. Riordan
- Gastrointestinal and Liver Unit, The Prince of Wales Hospital, Sydney, Australia
- Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Michael C. Grimm
- St George Clinical School, University of New South Wales, Sydney, Australia
| | - Thi Anh Tuyet Tran
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| | - Joelene Major
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| | - Nadeem O. Kaakoush
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| | - Hazel Mitchell
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| | - Li Zhang
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| |
Collapse
|
233
|
Miyoshi J, Yajima T, Okamoto S, Matsuoka K, Inoue N, Hisamatsu T, Shimamura K, Nakazawa A, Kanai T, Ogata H, Iwao Y, Mukai M, Hibi T. Ectopic expression of blood type antigens in inflamed mucosa with higher incidence of FUT2 secretor status in colonic Crohn's disease. J Gastroenterol 2011; 46:1056-63. [PMID: 21725903 DOI: 10.1007/s00535-011-0425-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2011] [Accepted: 04/12/2011] [Indexed: 02/04/2023]
Abstract
BACKGROUND Host-intestinal microbial interaction plays an important role in the pathogenesis of inflammatory bowel diseases (IBDs). The surface molecules of the intestinal epithelium act as receptors for bacterial adhesion and regulate the intestinal bacteria. Some known receptors are the mucosal blood type antigens, which are regulated by the fucosyltransferase2 (FUT2) gene, and individuals who express these antigens in the gastrointestinal tract are called secretors. Recent research has revealed that the FUT2 gene is associated with Crohn's disease (CD) in western populations. METHODS To clarify the contribution of mucosal blood type antigens in IBD, we determined the incidence of five previously reported single-nucleotide polymorphisms of the FUT2 gene in Japanese patients. We also used immunohistochemistry to investigate the antigen expression in mucosal specimens from IBD patients and animal models. RESULTS Genetic analysis revealed that all of the patients with colonic CD were secretors, whereas the incidence of secretors was 80, 80, 67, and 80%, respectively, for the control, ileocolonic CD, ileal CD, and ulcerative colitis groups (P = 0.036). Abnormal expression of blood type antigens was observed only in colonic CD. Interleukin-10⁻/⁻ mice, but not dextran sulfate sodium colitis mice, had enhanced colonic expression of blood type antigens, and the expression of these antigens preceded the development of colitis in the interleukin-10⁻/⁻ mice. CONCLUSIONS FUT2 secretor status was associated with colonic-type CD. This finding, taken together with the immunohistochemistry data, suggests that the abnormal expression of blood type antigens in the colon may be a unique and essential factor for colonic CD.
Collapse
Affiliation(s)
- Jun Miyoshi
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
234
|
Steck N, Hoffmann M, Sava IG, Kim SC, Hahne H, Tonkonogy SL, Mair K, Krueger D, Pruteanu M, Shanahan F, Vogelmann R, Schemann M, Kuster B, Sartor RB, Haller D. Enterococcus faecalis metalloprotease compromises epithelial barrier and contributes to intestinal inflammation. Gastroenterology 2011; 141:959-71. [PMID: 21699778 DOI: 10.1053/j.gastro.2011.05.035] [Citation(s) in RCA: 214] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2010] [Revised: 05/01/2011] [Accepted: 05/12/2011] [Indexed: 12/21/2022]
Abstract
BACKGROUND & AIMS Matrix metalloproteases (MMPs) mediate pathogenesis of chronic intestinal inflammation. We characterized the role of the gelatinase (GelE), a metalloprotease from Enterococcus faecalis, in the development of colitis in mice. METHODS Germ-free, interleukin-10-deficient (IL-10(-/-)) mice were monoassociated with the colitogenic E faecalis strain OG1RF and isogenic, GelE-mutant strains. Barrier function was determined by measuring E-cadherin expression, transepithelial electrical resistance (TER), and translocation of permeability markers in colonic epithelial cells and colon segments from IL-10(-/-) and TNF(ΔARE/Wt) mice. GelE specificity was shown with the MMP inhibitor marimastat. RESULTS Histologic analysis (score 0-4) of E faecalis monoassociated IL-10(-/-) mice revealed a significant reduction in colonic tissue inflammation in the absence of bacteria-derived GelE. We identified cleavage sites for GelE in the sequence of recombinant mouse E-cadherin, indicating that it might be degraded by GelE. Experiments with Ussing chambers and purified GelE revealed the loss of barrier function and extracellular E-cadherin in mice susceptible to intestinal inflammation (IL-10(-/-) and TNF(ΔARE/Wt) mice) before inflammation developed. Colonic epithelial cells had reduced TER and increased translocation of permeability markers after stimulation with GelE from OG1RF or strains of E faecalis isolated from patients with Crohn's disease and ulcerative colitis. CONCLUSIONS The metalloprotease GelE, produced by commensal strains of E faecalis, contributes to development of chronic intestinal inflammation in mice that are susceptible to intestinal inflammation (IL-10(-/-) and TNF(ΔARE/Wt) mice) by impairing epithelial barrier integrity.
Collapse
Affiliation(s)
- Natalie Steck
- Chair for Biofunctionality, ZIEL-Research Center for Nutrition and Food Science, CDD Center for Diet and Disease, Technische Universität München, Freising-Weihenstephan, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
235
|
Frank DN, Zhu W, Sartor RB, Li E. Investigating the biological and clinical significance of human dysbioses. Trends Microbiol 2011; 19:427-34. [PMID: 21775143 DOI: 10.1016/j.tim.2011.06.005] [Citation(s) in RCA: 135] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2011] [Revised: 06/08/2011] [Accepted: 06/20/2011] [Indexed: 12/13/2022]
Abstract
Culture-independent microbiological technologies that interrogate complex microbial populations without prior axenic culture, coupled with high-throughput DNA sequencing, have revolutionized the scale, speed and economics of microbial ecological studies. Their application to the medical realm has led to a highly productive merger of clinical, experimental and environmental microbiology. The functional roles played by members of the human microbiota are being actively explored through experimental manipulation of animal model systems and studies of human populations. In concert, these studies have appreciably expanded our understanding of the composition and dynamics of human-associated microbial communities (microbiota). Of note, several human diseases have been linked to alterations in the composition of resident microbial communities, so-called dysbiosis. However, how changes in microbial communities contribute to disease etiology remains poorly defined. Correlation of microbial composition represents integration of only two datasets (phenotype and microbial composition). This article explores strategies for merging the human microbiome data with multiple additional datasets (e.g. host single nucleotide polymorphisms and host gene expression) and for integrating patient-based data with results from experimental animal models to gain deeper understanding of how host-microbe interactions impact disease.
Collapse
Affiliation(s)
- Daniel N Frank
- Division of Infectious Diseases, School of Medicine, University of Colorado, School of Medicine, Aurora, CO 80045, USA.
| | | | | | | |
Collapse
|
236
|
Sydora BC, McFarlane SM, Doyle JSG, Fedorak RN. Neonatal exposure to fecal antigens reduces intestinal inflammation. Inflamm Bowel Dis 2011; 17:899-906. [PMID: 20824814 DOI: 10.1002/ibd.21453] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2010] [Accepted: 06/09/2010] [Indexed: 12/17/2022]
Abstract
BACKGROUND A role for bacterial antigens in the pathogenesis of inflammatory bowel disease (IBD) has been established in enhanced humoral and cellular immune response to ubiquitous antigens of the enteric flora. However, we have recently shown that bacterial antigens in the absence of live bacteria cannot initiate an intestinal inflammation in IBD-prone interleukin (IL)-10 gene-deficient mice. The objective was to investigate whether neonatal exposure to antigens of their own endogenous flora can tolerize mice to bacterial antigens. METHODS IL-10 gene-deficient neonates were injected intraperitoneally within 72 hours of birth with a sterile solution of bacterial lysates prepared from fecal material of either conventionally raised mice (contains bacterial antigens) or axenic mice (lacks bacterial antigens). The onset of intestinal inflammation was monitored as the appearance of occult blood in the stool in weekly hemoccult analysis. Mice were sacrificed between age 15 and 19 weeks and tested for histopathologic injury, intestinal inflammation, and systemic response to bacterial antigens. RESULTS In mice neonatally exposed to bacterial antigens the onset of intestinal inflammation was delayed and the incidence of histopathologic injury at age 18 weeks was reduced. In addition, mice injected with lysates from conventionally raised mice exhibited decreased release of proinflammatory cytokines (interferon gamma [IFN-γ] and IL-17) in intestinal tissue and demonstrated reduced bacteria-stimulated systemic responses when compared to mice injected with lysates derived from bacteria-free, axenic mice. CONCLUSIONS Neonatal intraperitoneal injection of antigens from the commensal flora causes long-lasting changes in systemic and mucosal immune responses resulting in delayed onset of intestinal inflammation and injury in IBD-prone IL-10 gene-deficient mice.
Collapse
Affiliation(s)
- Beate C Sydora
- Division of Gastroenterology, Department of Medicine, University of Alberta, Center of Excellence for Gastrointestinal Inflammation and Immunity Research, Edmonton, Alberta, Canada.
| | | | | | | |
Collapse
|
237
|
Sartor RB. Key questions to guide a better understanding of host-commensal microbiota interactions in intestinal inflammation. Mucosal Immunol 2011; 4:127-32. [PMID: 21248723 DOI: 10.1038/mi.2010.87] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Co-evolution with an extremely complex commensal enteric microbiota has helped shape mammalian mucosal immune responses. A yet incompletely defined subset of intestinal bacteria is required to stimulate chronic, immune-mediated intestinal inflammation, including human Crohn's disease, and intestinal microbiota composition is altered in a characteristic manner by the inflammatory response to create a dysbiotic relationship of protective vs. aggressive bacteria. We pose a number of questions regarding host interactions with the enteric microbiota, including influences of inflammation, host genetics, early environmental exposure, and diet on microbial composition and function, and conversely, the effect of bacterial metabolism, enteric fungi and viruses, and endogenous protective bacterial species on host immune and inflammatory responses. These questions are designed to stimulate research that will promote a better understanding of host-microbial interactions in the intestine and promote targeted novel therapeutic interventions.
Collapse
Affiliation(s)
- R B Sartor
- Department of Medicine/Division of Gastroenterology and Hepatology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.
| |
Collapse
|
238
|
Abstract
Gut microbiota contains about 10(14) bacterial cells classified within 4 bacterial phyla, namely Firmicutes, Bacteroidetes, Actinobacteria, and Proteobacteria. Much of the information has been generated through the application of nucleic acid-based methodologies (16S rRNA) which provide a cornerstone of microbial taxonomy. Inflammatory bowel disease (IBD) involves a dysregulated immune response to the gut microbiota in genetically predisposed hosts. Experimental animal models of colitis provide the best evidence that bacteria present in the bowel of the animals have an essential role in the pathogenesis of colitis since in most models, germ-free animals do not develop disease. Moreover, in the immunodeficient mouse model of colitis called TRUC (T-bet-/- x RAG2-/-), a colitogenic gut microbiota is selected and can be transmitted to mice with intact immunity and induce colitis. Current interest therefore focuses on the bacterial community as the source of antigens that fuel the chronic inflammation seen in IBD. Dysbiosis, an imbalance between harmful and protective bacteria, has been evoked and investigated in IBD. Thus, besides the classical pathogens, gut microbiota can drive pathogenicity via two mechanisms: an expansion of 'pro-inflammatory' species or a restriction in the protective compounds of the microbiota. Complexity of the microbiota suggests that both mechanisms may contribute to chronic gut inflammation in IBD.
Collapse
Affiliation(s)
- P Seksik
- Gastroenterology and Nutrition Department, Hôpital Saint Antoine, AP-HP, Paris, France.
| |
Collapse
|
239
|
Xia Y, Chen HQ, Zhang M, Jiang YQ, Hang XM, Qin HL. Effect of Lactobacillus plantarum LP-Onlly on gut flora and colitis in interleukin-10 knockout mice. J Gastroenterol Hepatol 2011; 26:405-11. [PMID: 21261733 DOI: 10.1111/j.1440-1746.2010.06498.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND AIM Probiotics are used in the therapy of inflammatory bowel disease. This study aimed to determine the effects of probiotic Lactobacillus plantarum LP-Onlly (LP) on gut flora and colitis in interleukin-10 knockout (IL-10(-/-) ) mice, a model of spontaneous colitis. METHODS IL-10(-/-) and wild-type mice were used at 8 weeks of age and LP by gavage was administered at a dose of 10(9) cells/day per mice for 4 weeks. Mice were maintained for another one week without LP treatment. The colonic tissues were collected for histological and ultrastructural analysis at death after 4 weeks treatment of LP, and the feces were collected at 1-week intervals throughout the experiment for the analysis of gut flora and LP using selective culture-based techniques. RESULTS Compared with control mice, IL-10(-/-) mice developed a severe intestinal inflammation and tissue damage, and had an abnormal composition of gut microflora. LP administration attenuated colitis with the decreased inflammatory scoring and histological injury in the colon of IL-10(-/-) mice. In addition, LP administration increased the numbers of beneficial total bifidobacteria and lactobacilli, and decreased the numbers of potential pathogenic enterococci and Clostridium perfringens, although the decrease of coliforms was not significant after LP treatment in IL-10(-/-) mice. CONCLUSIONS Oral administration of LP was effective in the treatment of colitis, with the direct modification of gut microflora in IL-10(-/-) mice. This probiotic strain could be used as a potential adjuvant in the therapy of inflammatory bowel disease, although further studies are required in human.
Collapse
Affiliation(s)
- Yang Xia
- Department of Surgery, Affiliated Sixth People's Hospital, Shanghai Jiaotong University, Shanghai, China
| | | | | | | | | | | |
Collapse
|
240
|
Hoffmann M, Messlik A, Kim SC, Sartor RB, Haller D. Impact of a probiotic Enterococcus faecalis
in a gnotobiotic mouse model of experimental colitis. Mol Nutr Food Res 2011; 55:703-13. [DOI: 10.1002/mnfr.201000361] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2010] [Revised: 11/12/2010] [Accepted: 11/15/2010] [Indexed: 12/12/2022]
|
241
|
Abstract
Gut microbes play a major role in carcinogenesis of the gastrointestinal tract. We and others have shown in mouse models that colonic bacteria also influence the development of extraintestinal cancers including hepatocellular and mammary carcinomas. Microbes such as Helicobacter hepaticus invoke a proinflammatory microenvironment in the lower bowel that may extend to distant organs, often in the absence of histologically evident inflammation. Innate immunity plays a crucial role in the promotion of liver cancer and other systemic diseases by gut microbes. Additional mechanisms include type 1 adaptive immunity, altered metabolism, and oxidative stress. Emerging links between host genetics, gut microbes, inflammatory bowel disease and colorectal cancer also may prove useful for the correlation of specific bacterial populations with extraintestinal neoplasms. Interruption of deleterious host-microbe networks through judicious use of antibiotics and targeted molecular therapies may help reduce the incidence of liver, breast, and other human cancers.
Collapse
|
242
|
Abstract
Recently, an unprecedented effort has been directed at understanding the interplay between chronic inflammation and development of cancer, with the case of inflammatory bowel disease (IBD)-associated colorectal cancer at the forefront of this research endeavor. The last decade has been particularly fertile, with the discovery of numerous innovative paradigms linking various inflammatory, proliferative, and innate and adaptive immune signaling pathways to the development of colorectal cancer. Because of the preponderant role of the intestinal microbiota in the initiation and progression of IBD, recent efforts have been directed at understanding the relationship between bacteria and colorectal cancer. The microbiota and its collective genome, the microbiome, form a diverse and complex ecological community that profoundly impacts intestinal homeostasis and disease states. This review will discuss the differential influence of the microbiota on the development of IBD-associated colorectal cancer and highlight the role of innate immune sensor-dependent as well as -independent mechanisms in this pathology.
Collapse
Affiliation(s)
- Janelle C Arthur
- Department of Medicine and the Center for Gastrointestinal Biology and Disease, University of North Carolina, Chapel Hill, North Carolina 27599-7080, USA
| | | |
Collapse
|
243
|
Abstract
Colorectal cancer is a major cause of cancer-related morbidity and mortality in the United States and many other regions of the world. Our understanding of the pathogenesis of colorectal cancer, from the precursor adenomatous polyp to adenocarcinoma, has evolved rapidly. Colorectal carcinogenesis is a sequential process characterized by the accumulation of multiple genetic and molecular alterations in colonic epithelial cells. However, the development of colorectal cancer involves more then just a genetic predisposition. External or environmental factors presumably play a significant role, and inflammatory bowel diseases, obesity, alcohol consumption, and a diet high in fat and low in fiber have all been implicated as risk factors for the development of either colonic adenomas or carcinomas. We are becoming increasingly aware of microbes as causes of malignancies. This article reviews the various microbes that have been associated with the development of colorectal carcinomas.
Collapse
Affiliation(s)
- Nazia Hasan
- Department of Medicine, NYU School of Medicine, New York, NY 10016, USA
| | | | | |
Collapse
|
244
|
Pawlowski SW, Calabrese G, Kolling GL, Platts-Mills J, Freire R, AlcantaraWarren C, Liu B, Sartor RB, Guerrant RL. Murine model of Clostridium difficile infection with aged gnotobiotic C57BL/6 mice and a BI/NAP1 strain. J Infect Dis 2010; 202:1708-12. [PMID: 20977342 PMCID: PMC3057484 DOI: 10.1086/657086] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2010] [Accepted: 05/04/2010] [Indexed: 11/03/2022] Open
Abstract
The increased incidence and severity of Clostridium difficile infection (CDI) in older adults (age, ≥65 years) corresponds with the emergence of the BI/NAP1 strain, making elucidation of the host immune response extremely important. We therefore infected germ-free C57BL/6 mice aged 7-14 months with a BI/NAP1 strain and monitored the mice for response. Infected mice were moribund 48-72 h after infection and developed gross and histological cecitis and colitis and elevated concentrations of keratinocyte chemoattractant, interleukin 1β, monocyte chemotactic protein 1, and granulocyte colony-stimulating factor and decreased levels of interferon γ, interleukin 12 p40, interleukin 12 p70, and interleukin 10 compared with controls. We conclude that aged, germ-free C57BL/6 mice are susceptible to fulminant CDI from a BI/NAP1 strain and represent a novel model to further elucidate the host immune response to acute CDI.
Collapse
Affiliation(s)
- S W Pawlowski
- Center For Global Health, Division of Infectious Diseases and International Health, Department of Internal Medicine, University of Virginia, Charlottesville, VA 22908, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
245
|
Sartor RB. Genetics and environmental interactions shape the intestinal microbiome to promote inflammatory bowel disease versus mucosal homeostasis. Gastroenterology 2010; 139:1816-9. [PMID: 21029802 DOI: 10.1053/j.gastro.2010.10.036] [Citation(s) in RCA: 138] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
246
|
Uno JK, Rao KN, Matsuoka K, Sheikh SZ, Kobayashi T, Li F, Steinbach EC, Sepulveda AR, Vanhaesebroeck B, Balfour Sartor R, Plevy SE. Altered macrophage function contributes to colitis in mice defective in the phosphoinositide-3 kinase subunit p110δ. Gastroenterology 2010; 139:1642-53, 1653.e1-6. [PMID: 20637203 PMCID: PMC2967619 DOI: 10.1053/j.gastro.2010.07.008] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2010] [Revised: 06/07/2010] [Accepted: 07/08/2010] [Indexed: 02/01/2023]
Abstract
BACKGROUND & AIMS Innate immune responses are crucial for host defense against pathogens but need to be tightly regulated to prevent chronic inflammation. Initial characterization of mice with a targeted inactivating mutation in the p110δ subunit of phosphoinositide 3-kinase (PI3K p110δ(D910A/D910A)) revealed defects in B- and T-cell signaling and chronic colitis. Here, we further characterize features of inflammatory bowel diseases in these mice and investigate underlying innate immune defects. METHODS Colons and macrophages from PI3K p110δ(D910A/D910A) mice were evaluated for colonic inflammation and innate immune dysfunction. Colonic p110δ messenger RNA expression was examined in interleukin (IL)-10(-/-) and wild-type germ-free mice during transition to a conventional microbiota. To assess polygenic impact on development of colitis, p110δ(D910A/D910A) mice were backcrossed to IL-10(-/-) mice. RESULTS A mild spontaneous colitis was shown in PI3K p110δ(D910A/D910A) mice at 8 weeks, with inflammation increasing with age. An inflammatory mucosal and systemic cytokine profile was characterized by expression of IL-12/23. In PI3K p110δ(D910A/D910A) macrophages, augmented toll-like receptor signaling and defective bactericidal activity were observed. Consistent with an important homeostatic role for PI3K p110δ, wild-type mice raised in a germ-free environment markedly up-regulated colonic PI3K p110δ expression with the introduction of the enteric microbiota; however, colitis-prone IL-10(-/-) mice did not. Moreover, PI3K p110δ(D910A/D910A) mice crossed to IL-10(-/-) mice developed severe colitis at an early age. CONCLUSIONS This study describes a novel model of experimental colitis that highlights the importance of PI3K p110δ in maintaining mucosal homeostasis and could provide insight into the pathogenesis of human inflammatory bowel disease.
Collapse
Affiliation(s)
- Jennifer K. Uno
- Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA.,Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Kavitha N. Rao
- Department of Immunology, University of Pittsburgh School of Medicine. Pittsburgh, PA 15213, USA
| | - Katsuyoshi Matsuoka
- Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Shehzad Z. Sheikh
- Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA.,Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Taku Kobayashi
- Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Fengling Li
- Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA.,Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Erin C. Steinbach
- Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA.,Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Antonia R. Sepulveda
- Department of Pathology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Bart Vanhaesebroeck
- Centre for Cell Signalling, Institute of Cancer, Queen Mary University of London, Charterhouse Square, London, UK
| | - R Balfour Sartor
- Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA.,Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Scott E. Plevy
- Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA.,Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| |
Collapse
|
247
|
Chen LF, Miao YL, Du Y, Li HN, Xiao YL. Significance of CEACAM6, Syndecan-1, PDGFA and HLA-DRB5 expression in patients with ulcerative colitis. Shijie Huaren Xiaohua Zazhi 2010; 18:2971-2975. [DOI: 10.11569/wcjd.v18.i28.2971] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To analyze the significance of the expression of carcinoembryonic antigen-related cell adhesion molecule 6 (CEACAM6), Syndecan-1, platelet-derived growth factor alpha (PDGFA) and HLA-DRB5 genes in the development and progression of ulcerative colitis.
METHODS: The mRNA expression of CEACAM6, Syndecan-1, PDGFA and HLA-DRB5 was detected by semiquantitative RT-PCR in peripheral blood mononuclear cells (PBMC) and colon specimens from 21 patients with ulcerative colitis.
RESULTS: The mRNA expression levels of CEACAM6, Syndecan-1 and PDGFA in patients with ulcerative colitis were significantly higher than those in normal controls (0.77 ± 0.23 vs 0.58 ± 0.14, 1.16 ± 0.39 vs 0.85 ± 0.16, 0.90 ± 0.18 vs 0.78 ± 0.13, all P < 0.01). The expression of HLA-DRB5 mRNA was also up-regulated in patients with ulcerative colitis compared with normal controls (0.58 ± 0.19 vs 0.42 ± 0.19, P < 0.01).
CONCLUSION: CEACAM6, Syndecan-1, PDGFA and HLA-DRB5 are highly expressed in ulcerative colitis and may therefore play an important role in the pathogenesis of ulcerative colitis.
Collapse
|
248
|
Knoch B, Barnett MPG, Cooney J, McNabb WC, Barraclough D, Laing W, Roy NC. Dietary oleic acid as a control fatty acid for polyunsaturated fatty acid intervention studies: a transcriptomics and proteomics investigation using interleukin-10 gene-deficient mice. Biotechnol J 2010; 5:1226-40. [PMID: 20872728 DOI: 10.1002/biot.201000066] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2010] [Revised: 09/06/2010] [Accepted: 09/08/2010] [Indexed: 01/22/2023]
Abstract
Oleic acid (OA) has been used as a control fatty acid in dietary polyunsaturated fatty acid (PUFA) intervention studies due to its lack of effect on eiconasoid biosynthesis. Since the effect of OA as a control fatty acid has not yet been investigated for transcriptomics and proteomics studies, this study aimed to test whether colonic transcriptome and proteome profiles associated with colitis development in mice fed a linoleic acid-rich corn oil-AIN-76A diet (Il10(-/-) compared to C57 mice) where similar to those of OA-fed Il10(-/-) compared to C57 mice (genotype comparison). A close clustering of colonic gene and protein expression profiles between the mice fed the AIN-76A or OA diet was observed. Inflammation-induced regulatory processes associated with cellular and humoral immune responses, cellular stress response and metabolic processes related to energy utilization were identified in Il10(-/-) compared to C57 mice fed either diet. Thus OA was considered as a suitable control unsaturated fatty acid for use in multi-omics PUFA studies. The second aim of this study was to test the effect of an OA-enriched AIN-76A diet compared to a linoleic acid-rich corn oil-AIN-76A diet on colonic transcriptome and proteome changes within Il10(-/-) or C57 mice (diet comparison). Overall, there was a limited concordance observed between measureable transcriptomics and proteomics profiles for genotype and diet comparisons. This underlines the importance and validity of a systems biology approach to understand the effects of diet on gene expression as a function of the genotype.
Collapse
Affiliation(s)
- Bianca Knoch
- Agri-Foods & Health, Food & Textiles Group, AgResearch, Palmerston North, New Zealand
| | | | | | | | | | | | | |
Collapse
|
249
|
McGovern DPB, Jones MR, Taylor KD, Marciante K, Yan X, Dubinsky M, Ippoliti A, Vasiliauskas E, Berel D, Derkowski C, Dutridge D, Fleshner P, Shih DQ, Melmed G, Mengesha E, King L, Pressman S, Haritunians T, Guo X, Targan SR, Rotter JI. Fucosyltransferase 2 (FUT2) non-secretor status is associated with Crohn's disease. Hum Mol Genet 2010; 19:3468-76. [PMID: 20570966 PMCID: PMC2916706 DOI: 10.1093/hmg/ddq248] [Citation(s) in RCA: 296] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2010] [Revised: 06/03/2010] [Accepted: 06/10/2010] [Indexed: 12/20/2022] Open
Abstract
Genetic variation in both innate and adaptive immune systems is associated with Crohn's disease (CD) susceptibility, but much of the heritability to CD remains unknown. We performed a genome-wide association study (GWAS) in 896 CD cases and 3204 healthy controls all of Caucasian origin as defined by multidimensional scaling. We found supportive evidence for 21 out of 40 CD loci identified in a recent CD GWAS meta-analysis, including two loci which had only nominally achieved replication (rs4807569, 19p13; rs991804, CCL2/CCL7). In addition, we identified associations with genes involved in tight junctions/epithelial integrity (ASHL, ARPC1A), innate immunity (EXOC2), dendritic cell biology [CADM1 (IGSF4)], macrophage development (MMD2), TGF-beta signaling (MAP3K7IP1) and FUT2 (a physiological trait that regulates gastrointestinal mucosal expression of blood group A and B antigens) (rs602662, P=3.4x10(-5)). Twenty percent of Caucasians are 'non-secretors' who do not express ABO antigens in saliva as a result of the FUT2 W134X allele. We demonstrated replication in an independent cohort of 1174 CD cases and 357 controls between the four primary FUT2 single nucleotide polymorphisms (SNPs) and CD (rs602662, combined P-value 4.90x10(-8)) and also association with FUT2 W143X (P=2.6x10(-5)). Further evidence of the relevance of this locus to CD pathogenesis was demonstrated by the association of the original four SNPs and CD in the recently published CD GWAS meta-analysis (rs602662, P=0.001). These findings strongly implicate this locus in CD susceptibility and highlight the role of the mucus layer in the development of CD.
Collapse
Affiliation(s)
- Dermot P B McGovern
- Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
250
|
Nutrigenomics and IBD: the intestinal microbiota at the cross-road between inflammation and metabolism. J Clin Gastroenterol 2010; 44 Suppl 1:S6-9. [PMID: 20535026 DOI: 10.1097/mcg.0b013e3181dd8b76] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Nutrition-related factors together with components of the gut-associated microbial ecosystem (gut microbiota) emerge as prime environmental triggers for the development and modification of lifestyle-related chronic diseases including chronic inflammatory disorders of the gastro-intestinal tract such as Crohn's disease and ulcerative colitis. Although a variety of susceptibility genes were identified in genome-wide association studies, the impact of environmental factors in initiating or promoting the development of these complex diseases are unknown. Nutrigenomics is a transdisciplinary approach to understand the subtle but contentious impact of nutrition and/or microbes as prime environmental triggers in shaping the dynamic range between health and diseases. Profiling technologies such as transcriptomics, proteomics, and metabonomics at the interface of the host's genetic make-up and its metabolic phenotype are implemented to identify cellular and molecular targets to develop novel hypothesise with respect to the functional role of diet and gut bacteria in modulating chronic degenerative diseases including inflammatory bowel disease.
Collapse
|