1
|
Hurtado-Lorenzo A, Swantek JL. The landscape of new therapeutic opportunities for IBD. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2024; 101:1-83. [PMID: 39521596 DOI: 10.1016/bs.apha.2024.10.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
This chapter presents an overview of the emerging strategies to address the unmet needs in the management of inflammatory bowel diseases (IBD). IBD poses significant challenges, as over half of patients experience disease progression despite interventions, leading to irreversible complications, and a substantial proportion do not respond to existing therapies, such as biologics. To overcome these limitations, we describe a diverse array of novel therapeutic approaches. In the area of immune homeostasis restoration, the focus is on targeting cytokine networks, leukocyte trafficking, novel immune pathways, and cell therapies involving regulatory T cells and mesenchymal stem cells (MSC). Recognizing the critical role of impaired intestinal barrier integrity in IBD, we highlight therapies aimed at restoring barrier function and promoting mucosal healing, such as those targeting cell proliferation, tight junctions, and lipid mediators. Addressing the challenges posed by fibrosis and fistulas, we describe emerging targets for reversing fibrosis like kinase and cytokine inhibitors and nuclear receptor agonists, as well as the potential of MSC for fistulas. The restoration of a healthy gut microbiome, through strategies like fecal microbiota transplantation, rationally defined bacterial consortia, and targeted antimicrobials, is also highlighted. We also describe innovative approaches to gut-targeted drug delivery to enhance efficacy and minimize side effects. Reinforcing these advancements is the critical role of precision medicine, which emphasizes the use of multiomics analysis for the discovery of biomarkers to enable personalized IBD care. Overall, the emerging landscape of therapeutic opportunities for IBD holds great potential to surpass the therapeutic ceiling of current treatments.
Collapse
Affiliation(s)
- Andrés Hurtado-Lorenzo
- Translational Research & IBD Ventures, Research Department, Crohn's & Colitis Foundation, New York, NY, United States.
| | - Jennifer L Swantek
- Translational Research & IBD Ventures, Research Department, Crohn's & Colitis Foundation, New York, NY, United States
| |
Collapse
|
2
|
Toto F, Marangelo C, Scanu M, De Angelis P, Isoldi S, Abreu MT, Cucchiara S, Stronati L, Del Chierico F, Putignani L. A Novel Microbial Dysbiosis Index and Intestinal Microbiota-Associated Markers as Tools of Precision Medicine in Inflammatory Bowel Disease Paediatric Patients. Int J Mol Sci 2024; 25:9618. [PMID: 39273567 PMCID: PMC11395508 DOI: 10.3390/ijms25179618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 08/30/2024] [Accepted: 09/03/2024] [Indexed: 09/15/2024] Open
Abstract
Recent evidence indicates that the gut microbiota (GM) has a significant impact on the inflammatory bowel disease (IBD) progression. Our aim was to investigate the GM profiles, the Microbial Dysbiosis Index (MDI) and the intestinal microbiota-associated markers in relation to IBD clinical characteristics and disease state. We performed 16S rRNA metataxonomy on both stools and ileal biopsies, metabolic dysbiosis tests on urine and intestinal permeability and mucosal immunity activation tests on the stools of 35 IBD paediatric patients. On the GM profile, we assigned the MDI to each patient. In the statistical analyses, the MDI was correlated with clinical parameters and intestinal microbial-associated markers. In IBD patients with high MDI, Gemellaceae and Enterobacteriaceae were increased in stools, and Fusobacterium, Haemophilus and Veillonella were increased in ileal biopsies. Ruminococcaceae and WAL_1855D were enriched in active disease condition; the last one was also positively correlated to MDI. Furthermore, the MDI results correlated with PUCAI and Matts scores in ulcerative colitis patients (UC). Finally, in our patients, we detected metabolic dysbiosis, intestinal permeability and mucosal immunity activation. In conclusion, the MDI showed a strong association with both severity and activity of IBD and a positive correlation with clinical scores, especially in UC. Thus, this evidence could be a useful tool for the diagnosis and prognosis of IBD.
Collapse
Affiliation(s)
- Francesca Toto
- Immunology, Rheumatology and Infectious Diseases Research Area, Unit of Microbiome, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
| | - Chiara Marangelo
- Immunology, Rheumatology and Infectious Diseases Research Area, Unit of Microbiome, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
| | - Matteo Scanu
- Immunology, Rheumatology and Infectious Diseases Research Area, Unit of Microbiome, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
| | - Paola De Angelis
- Digestive Endoscopy and Surgery Unit, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
| | - Sara Isoldi
- Pediatric Gastroenterology and Hepatology Unit, Santobono-Pausilipon Children's Hospital, 80122 Naples, Italy
| | - Maria Teresa Abreu
- Crohn's and Colitis Center, Division of Digestive Health and Liver Diseases, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Salvatore Cucchiara
- Maternal Child Health Department, Pediatric Gastroenterology and Liver Unit, Sapienza University of Rome, 00185 Rome, Italy
| | - Laura Stronati
- Department of Molecular Medicine, Sapienza University of Rome, 00185 Rome, Italy
| | - Federica Del Chierico
- Immunology, Rheumatology and Infectious Diseases Research Area, Unit of Microbiome, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
| | - Lorenza Putignani
- Unit of Microbiology and Diagnostic Immunology, Unit of Microbiomics and Immunology, Rheumatology and Infectious Diseases Research Area, Unit of Microbiome, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
| |
Collapse
|
3
|
Berard M, Chassain K, Méry C, Gillaizeau F, Carton T, Humeau H, Navasiolava N, Rocour S, Schurgers L, Kempf M, Martin L. Changes in the gut microbiota of pseudoxanthoma elasticum patients. Ann Dermatol Venereol 2024; 151:103290. [PMID: 39003978 DOI: 10.1016/j.annder.2024.103290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 03/11/2024] [Accepted: 05/13/2024] [Indexed: 07/16/2024]
Abstract
OBJECTIVE Pseudoxanthoma elasticum (PXE) is a rare autosomal disorder with a variable phenotype that may be modulated by environmental factors. Plasma vitamin K (VK) levels may be involved in the ectopic calcification process observed in PXE. Since VK2 is predominantly produced by the gut microbiota, we hypothesized that changes in the gut microbiota of PXE patients might exacerbate the calcification process and disease symptoms. METHODS Twenty PXE patients were included in the study and 60 gut microbiota profiles from the Biofortis laboratory database were used as controls. RESULTS The Rhodospirillaceae family was more abundant in the PXE group while the Sphingomonadaceae family was more abundant in the control group. In a PXE severity subgroup analysis, microbiota dispersion was lower in "severe" than in "non-severe" patients, which was confirmed by permutation multivariate analysis of variance at the phylum, family and genus ranks. However, no significant association was found in a model incorporating relative abundance of bacterial families, severity score, and different blood and fecal VK species. CONCLUSION These results suggest slight compositional changes in the gut microbiota of PXE patients. Further studies are needed to substantiate their impact on VK metabolism and the calcification process.
Collapse
Affiliation(s)
- M Berard
- National Reference Center for PXE (MAGEC Nord), Dept. of Dermatology, Angers University Hospital, F-49000 Angers, France
| | - K Chassain
- National Reference Center for PXE (MAGEC Nord), Dept. of Dermatology, Angers University Hospital, F-49000 Angers, France
| | - C Méry
- Biofortis SAS, 44800 Saint Herblain, France
| | | | - T Carton
- Biofortis SAS, 44800 Saint Herblain, France
| | - H Humeau
- National Reference Center for PXE (MAGEC Nord), Dept. of Dermatology, Angers University Hospital, F-49000 Angers, France
| | - N Navasiolava
- National Reference Center for PXE (MAGEC Nord), Dept. of Dermatology, Angers University Hospital, F-49000 Angers, France
| | - S Rocour
- National Reference Center for PXE (MAGEC Nord), Dept. of Dermatology, Angers University Hospital, F-49000 Angers, France
| | - L Schurgers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, University of Maastricht, Netherlands
| | - M Kempf
- Laboratory of Bacteriology, Dept. of Infectious Agents, Angers University Hospital, F-49000 Angers, France; Nantes University, Angers University, INSERM, CNRS, Immunology and New Concepts in ImmunoTherapy, INCIT, UMR 1302/EMR6001, F-44000 Nantes, France
| | - L Martin
- National Reference Center for PXE (MAGEC Nord), Dept. of Dermatology, Angers University Hospital, F-49000 Angers, France; Angers University, MitoVasc (INSERM U1083, CNRS 6015), SFR ICAT, F-49000 Angers, France.
| |
Collapse
|
4
|
Cortes GM, Marcialis MA, Bardanzellu F, Corrias A, Fanos V, Mussap M. Inflammatory Bowel Disease and COVID-19: How Microbiomics and Metabolomics Depict Two Sides of the Same Coin. Front Microbiol 2022; 13:856165. [PMID: 35391730 PMCID: PMC8981987 DOI: 10.3389/fmicb.2022.856165] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 02/21/2022] [Indexed: 12/11/2022] Open
Abstract
The integrity of the gastrointestinal tract structure and function is seriously compromised by two pathological conditions sharing, at least in part, several pathogenetic mechanisms: inflammatory bowel diseases (IBD) and coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. IBD and COVID-19 are marked by gut inflammation, intestinal barrier breakdown, resulting in mucosal hyperpermeability, gut bacterial overgrowth, and dysbiosis together with perturbations in microbial and human metabolic pathways originating changes in the blood and fecal metabolome. This review compared the most relevant metabolic and microbial alterations reported from the literature in patients with IBD with those in patients with COVID-19. In both diseases, gut dysbiosis is marked by the prevalence of pro-inflammatory bacterial species and the shortfall of anti-inflammatory species; most studies reported the decrease in Firmicutes, with a specific decrease in obligately anaerobic producers short-chain fatty acids (SCFAs), such as Faecalibacterium prausnitzii. In addition, Escherichia coli overgrowth has been observed in IBD and COVID-19, while Akkermansia muciniphila is depleted in IBD and overexpressed in COVID-19. In patients with COVID-19, gut dysbiosis continues after the clearance of the viral RNA from the upper respiratory tract and the resolution of clinical symptoms. Finally, we presented and discussed the impact of gut dysbiosis, inflammation, oxidative stress, and increased energy demand on metabolic pathways involving key metabolites, such as tryptophan, phenylalanine, histidine, glutamine, succinate, citrate, and lipids.
Collapse
Affiliation(s)
- Gian Mario Cortes
- Neonatal Intensive Care Unit, Department of Surgical Sciences, University of Cagliari, Monserrato, Italy
| | - Maria Antonietta Marcialis
- Neonatal Intensive Care Unit, Department of Surgical Sciences, University of Cagliari, Monserrato, Italy
| | - Flaminia Bardanzellu
- Neonatal Intensive Care Unit, Department of Surgical Sciences, University of Cagliari, Monserrato, Italy
| | - Angelica Corrias
- Neonatal Intensive Care Unit, Department of Surgical Sciences, University of Cagliari, Monserrato, Italy
| | - Vassilios Fanos
- Neonatal Intensive Care Unit, Department of Surgical Sciences, University of Cagliari, Monserrato, Italy
| | - Michele Mussap
- Laboratory Medicine, Department of Surgical Sciences, School of Medicine, University of Cagliari, Monserrato, Italy
| |
Collapse
|
5
|
Honig G, Larkin PB, Heller C, Hurtado-Lorenzo A. Research-Based Product Innovation to Address Critical Unmet Needs of Patients with Inflammatory Bowel Diseases. Inflamm Bowel Dis 2021; 27:S1-S16. [PMID: 34791292 PMCID: PMC8922161 DOI: 10.1093/ibd/izab230] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Indexed: 12/09/2022]
Abstract
Despite progress in recent decades, patients with inflammatory bowel diseases face many critical unmet needs, demonstrating the limitations of available treatment options. Addressing these unmet needs will require interventions targeting multiple aspects of inflammatory bowel disease pathology, including disease drivers that are not targeted by available therapies. The vast majority of late-stage investigational therapies also focus primarily on a narrow range of fundamental mechanisms. Thus, there is a pressing need to advance to clinical stage differentiated investigational therapies directly targeting a broader range of key mechanistic drivers of inflammatory bowel diseases. In addition, innovations are critically needed to enable treatments to be tailored to the specific underlying abnormal biological pathways of patients; interventions with improved safety profiles; biomarkers to develop prognostic, predictive, and monitoring tests; novel devices for nonpharmacological approaches such as minimally invasive monitoring; and digital health technologies. To address these needs, the Crohn's & Colitis Foundation launched IBD Ventures, a venture philanthropy-funding mechanism, and IBD Innovate®, an innovative, product-focused scientific conference. This special IBD Innovate® supplement is a collection of articles reflecting the diverse and exciting research and development that is currently ongoing in the inflammatory bowel disease field to deliver innovative and differentiated products addressing critical unmet needs of patients. Here, we highlight the pipeline of new product opportunities currently advancing at the preclinical and early clinical development stages. We categorize and describe novel and differentiated potential product opportunities based on their potential to address the following critical unmet patient needs: (1) biomarkers for prognosis of disease course and prediction/monitoring of treatment response; (2) restoration of eubiosis; (3) restoration of barrier function and mucosal healing; (4) more effective and safer anti-inflammatories; (5) neuromodulatory and behavioral therapies; (6) management of disease complications; and (7) targeted drug delivery.
Collapse
|
6
|
Dong JY, Xia KJ, Liang W, Liu LL, Yang F, Fang XS, Xiong YJ, Wang L, Zhou ZJ, Li CY, Zhang WD, Wang JY, Chen DP. Ginsenoside Rb1 alleviates colitis in mice via activation of endoplasmic reticulum-resident E3 ubiquitin ligase Hrd1 signaling pathway. Acta Pharmacol Sin 2021; 42:1461-1471. [PMID: 33268823 PMCID: PMC8379258 DOI: 10.1038/s41401-020-00561-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 10/19/2020] [Indexed: 12/11/2022]
Abstract
Endoplasmic reticulum (ER) homeostasis is regulated by ER-resident E3 ubiquitin ligase Hrd1, which has been implicated in inflammatory bowel disease (IBD). Ginsenoside Rb1 (GRb1) is the major ginsenoside in ginseng with multiple pharmacological activities. In this study we investigated the role of Hrd1 in IBD and its regulation by GRb1. Two mouse colitis models were established to mimic human IBD: drinking water containing dextran sodium sulfate (DSS) as well as intra-colonic infusion of 2, 4, 6-trinitrobenzene sulfonic acid (TNBS). Colitis mice were treated with GRb1 (20, 40 mg·kg-1·d-1, ig) or a positive control drug sulfasalazine (500 mg·kg-1·d-1, ig) for 7 days. The model mice showed typical colitis symptoms and pathological changes in colon tissue. In addition to significant inflammatory responses and cell apoptosis in colon tissue, colon epithelial expression of Hrd1 was significantly decreased, the expression of ER stress markers GRP78, PERK, CHOP, and caspase 12 was increased, and the expression of Fas was increased (Fas was removed by Hrd1-induced ubiquitination). These changes were partially, or completely, reversed by GRb1 administration, whereas injection of Hrd1 inhibitor LS102 (50 mg·kg-1· d-1, ip, for 6 days) exacerbated colitis symptoms in colitis mice. GRb1 administration not only normalized Hrd1 expression at both the mRNA and protein levels, but also alleviated the ER stress response, Fas-related apoptosis, and other colitis symptoms. In intestinal cell line IEC-6, the expression of Hrd1 was significantly decreased by LPS treatment, but was normalized by GRb1 (200 μM). GRb1 alleviated LPS-induced ER stress and cell apoptosis in IEC-6 cells, and GRb1 action was inhibited by knockdown of Hrd1 using small interfering RNA. In summary, these results reveal a pathological role of Hrd1 in colitis, and provide a novel insight into alternative treatment of colitis using GRb1 activating Hrd1 signaling pathway.
Collapse
|
7
|
Yang Y, Sheng Y, Wang J, Zhou X, Guan Q, Shen H, Li W, Ruan S. Aureusidin derivative CNQX inhibits chronic colitis inflammation and mucosal barrier damage by targeting myeloid differentiation 2 protein. J Cell Mol Med 2021; 25:7257-7269. [PMID: 34184406 PMCID: PMC8335670 DOI: 10.1111/jcmm.16755] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 05/17/2021] [Accepted: 06/09/2021] [Indexed: 12/14/2022] Open
Abstract
Our previous study has found that aureusidin can inhibit inflammation by targeting myeloid differentiation 2 (MD2) protein. Structural optimization of aureusidin gave rise to a derivative named CNQX. LPS was used to induce inflammation in intestinal macrophages; flow cytometry, PI staining and Hoechst 33342 staining were used to detect the apoptotic level of macrophages; enzyme-linked immunosorbent assay (ELISA) was utilized to detect the expression level of inflammatory factors (including IL-1β, IL-18 and TNF-α); immunofluorescence staining was used to investigate the expression of MD2; Western blot was employed to measure the protein level of TLR4, MD2, MyD88 and p-P65. As a result, CNQX with IC50 of 2.5 μM can significantly inhibit the inflammatory damage of macrophages, decrease apoptotic level, reduce the expression level of inflammatory factors and simultaneously decrease the expression level of TLR4, MD2, MyD88 as well as p-P65. Caco-2 cell line was used to simulate the intestinal mucosal barrier in vitro, LPS was employed to induce cell injury in Caco-2 (to up-regulate barrier permeability), and CNQX with IC50 of 2.5 μl was used for intervention. Flow cytometry was used to detect the apoptotic level of Caco-2 cells, trans-epithelial electric resistance (TEER) was measured, FITC-D was used to detect the permeability of the intestinal mucosa, and Western blot was used to detect the expression levels of tight junction proteins (including occludin, claudin-1, MyD88, TLR4 and MD2). As a result, CNQX decreased the apoptotic level of Caco-2 cells, increased TEER value, decreased the expression levels of MyD88, TLR4 and MD2, and increased the protein levels of tight junction proteins (including occludin and claudin-1). C57BL/6 wild-type mice were treated with drinking water containing Dextran sulphate sodium (DSS) to establish murine chronic colitis model. After CQNX intervention, we detected the bodyweight, DAI score and H&E tissue staining to evaluate the life status and pathological changes. Immunohistochemistry (IHC) staining was used to detect the expression of MD2 protein, tight junction protein (including occludin and claudin-1). Transmission electron microscopy and FITC-D were used to detect intestinal mucosal permeability. Western blot was used to detect the expression levels of tight junction proteins (including occludin, claudin-1, MyD88, TLR4 and MD2) in the intestinal mucosa tissue. Consequently, CNQX can inhibit the intestinal inflammatory response in mice with colitis, inhibit the mucosal barrier injury, increase the expression of tight junction proteins (including occludin and claudin-1) and decrease the expression levels of MyD88, TLR4 and MD2. Mechanistically, pull-down and immunoprecipitation assays showed that CNQX can inhibit the activation of TLR4/MD2-NF-κB by binding to MD2 protein. Collectively, in this study, we found that CNQX can suppress the activation of TLR4 signals by targeting MD2 protein, thereby inhibiting inflammation and mucosal barrier damage of chronic colitis.
Collapse
Affiliation(s)
- Yi Yang
- Department of PharmacyThe Second Affiliated Hospital of Jiaxing UniversityJiaxingChina
| | - Yongjia Sheng
- Department of PharmacyThe Second Affiliated Hospital of Jiaxing UniversityJiaxingChina
| | - Jin Wang
- Department of PharmacyThe Second Affiliated Hospital of Jiaxing UniversityJiaxingChina
| | - Xiaohong Zhou
- Department of PharmacyThe Second Affiliated Hospital of Jiaxing UniversityJiaxingChina
| | - Qiaobing Guan
- Department of PharmacyThe Second Affiliated Hospital of Jiaxing UniversityJiaxingChina
| | - Heping Shen
- Department of PharmacyThe Second Affiliated Hospital of Jiaxing UniversityJiaxingChina
| | - Wenyan Li
- Department of PharmacyThe Second Affiliated Hospital of Jiaxing UniversityJiaxingChina
| | - Shuiliang Ruan
- Department of Center LaboratoryThe Second Affiliated Hospital of Jiaxing UniversityJiaxingChina
| |
Collapse
|
8
|
Du G, Huang H, Zhu Q, Ying L. Effects of cat ownership on the gut microbiota of owners. PLoS One 2021; 16:e0253133. [PMID: 34133453 PMCID: PMC8208556 DOI: 10.1371/journal.pone.0253133] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 05/28/2021] [Indexed: 12/16/2022] Open
Abstract
Pet ownership is an essential environmental exposure that might influence the health of the owner. This study’s primary objectives were to explore the effects of cat ownership on the gut microbial diversity and composition of owners. Raw data from the American Gut Project were obtained from the SRA database. A total of 214 Caucasian individuals (111 female) with cats and 214 individuals (111 female) without cats were used in the following analysis. OTU number showed significant alteration in the Cat group and Female_cat group, compared with that of the no cat (NC) group and Female_ NC group, respectively. Compared with the NC group, the microbial phylum Proteobacteria was significantly decreased in the Cat group. The microbial families Alcaligenaceae and Pasteurellaceae were significantly reduced, while Enterobacteriaceae and Pseudomonadaceae were significantly increased in the Cat group. Fifty metabolic pathways were predicted to be significantly changed in the Cat group. Twenty-one and 13 metabolic pathways were predicted to be significantly changed in the female_cat and male_cat groups, respectively. Moreover, the microbial phylum Cyanobacteria was significantly decreased, while the families Alcaligenaceae, Pseudomonadaceae and Enterobacteriaceae were significantly changed in the normal weight cat group. In addition, 41 and 7 metabolic pathways were predicted to be significantly changed in the normal-weight cat and overweight cat groups, respectively. Therefore, this study demonstrated that cat ownership could influence owners’ gut microbiota composition and function, especially in the female group and normal-weight group.
Collapse
Affiliation(s)
- Guankui Du
- Department of Biochemistry and Molecular Biology, Hainan Medical University, Haikou, China
- The Key Laboratory of Molecular Biology, Hainan Medical University, Haikou, China
- * E-mail: (GKD); (YL)
| | - Hairong Huang
- School of Public Health, Hainan Medical University, Haikou, China
| | - Qiwei Zhu
- The Key Laboratory of Molecular Biology, Hainan Medical University, Haikou, China
| | - Li Ying
- Haikou Customs, Haikou, China
- * E-mail: (GKD); (YL)
| |
Collapse
|
9
|
Aldars-García L, Chaparro M, Gisbert JP. Systematic Review: The Gut Microbiome and Its Potential Clinical Application in Inflammatory Bowel Disease. Microorganisms 2021; 9:microorganisms9050977. [PMID: 33946482 PMCID: PMC8147118 DOI: 10.3390/microorganisms9050977] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 04/22/2021] [Accepted: 04/29/2021] [Indexed: 02/07/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic relapsing-remitting systemic disease of the gastrointestinal tract. It is well established that the gut microbiome has a profound impact on IBD pathogenesis. Our aim was to systematically review the literature on the IBD gut microbiome and its usefulness to provide microbiome-based biomarkers. A systematic search of the online bibliographic database PubMed from inception to August 2020 with screening in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines was conducted. One-hundred and forty-four papers were eligible for inclusion. There was a wide heterogeneity in microbiome analysis methods or experimental design. The IBD intestinal microbiome was generally characterized by reduced species richness and diversity, and lower temporal stability, while changes in the gut microbiome seemed to play a pivotal role in determining the onset of IBD. Multiple studies have identified certain microbial taxa that are enriched or depleted in IBD, including bacteria, fungi, viruses, and archaea. The two main features in this sense are the decrease in beneficial bacteria and the increase in pathogenic bacteria. Significant differences were also present between remission and relapse IBD status. Shifts in gut microbial community composition and abundance have proven to be valuable as diagnostic biomarkers. The gut microbiome plays a major role in IBD, yet studies need to go from casualty to causality. Longitudinal designs including newly diagnosed treatment-naïve patients are needed to provide insights into the role of microbes in the onset of intestinal inflammation. A better understanding of the human gut microbiome could provide innovative targets for diagnosis, prognosis, treatment and even cure of this relevant disease.
Collapse
Affiliation(s)
- Laila Aldars-García
- Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP), Universidad Autónoma de Madrid, 28006 Madrid, Spain; (L.A.-G.); (M.C.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), 28006 Madrid, Spain
| | - María Chaparro
- Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP), Universidad Autónoma de Madrid, 28006 Madrid, Spain; (L.A.-G.); (M.C.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), 28006 Madrid, Spain
| | - Javier P. Gisbert
- Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP), Universidad Autónoma de Madrid, 28006 Madrid, Spain; (L.A.-G.); (M.C.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), 28006 Madrid, Spain
- Correspondence: ; Tel.: +34-913-093-911; Fax: +34-915-204-013
| |
Collapse
|
10
|
Zhu YT, Yuan YZ, Feng QP, Hu MY, Li WJ, Wu X, Xiang SY, Yu SQ. Food emulsifier polysorbate 80 promotes the intestinal absorption of mono-2-ethylhexyl phthalate by disturbing intestinal barrier. Toxicol Appl Pharmacol 2021; 414:115411. [PMID: 33476678 DOI: 10.1016/j.taap.2021.115411] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 01/12/2021] [Accepted: 01/15/2021] [Indexed: 12/16/2022]
Abstract
Di-2-ethylhexyl phosphate (DEHP) and its main toxic metabolite mono-2-ethylhexyl phthalate (MEHP) are the typical endocrine disrupting chemicals (EDCs) and widely affect human health. Our previous research reported that synthetic nonionic dietary emulsifier polysorbate 80 (P80, E433) had the promotional effect on the oral absorption of DEHP in rats. The aim of this study was to explore its mechanism of promoting oral absorption, focusing on the mucus barrier and mucosal barrier of the small intestine. A small molecule fluorescent probe 5-aminofluorescein-MEHP (MEHP-AF) was used as a tracker of MEHP in vivo and in vitro. First of all, we verified that P80 promoted the bioavailability of MEHP-AF in the long-term and low-dose exposure of MEHP-AF with P80 as a result of increasing the intestinal absorption of MEHP-AF. Afterwards, experimental results from Western blot, qPCR, immunohistochemistry, and immunofluorescence showed that P80 decreased the expression of proteins (mucus protein mucin-2, tight junction proteins claudin-1 and occludin) related to mucus barrier and mucosal barrier in the intestine, changed the integrity of intestinal epithelial cell, and increased the permeability of intestinal epithelial mucosa. These results indicated that P80 promoted the oral absorption of MEHP-AF by altering the intestinal mucus barrier and mucosal barrier. These findings are of great importance for assessing the safety risks of some food emulsifiers and clarifying the absorption mechanism of chemical pollutants in food, especially for EDCs.
Collapse
Affiliation(s)
- Yu-Ting Zhu
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210046, People's Republic of China
| | - Yi-Zhen Yuan
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210046, People's Republic of China
| | - Qiu-Ping Feng
- College of Food Sciences and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210046, People's Republic of China
| | - Meng-Yuan Hu
- College of Food Sciences and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210046, People's Republic of China
| | - Wen-Jie Li
- College of Food Sciences and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210046, People's Republic of China
| | - Xiu Wu
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210046, People's Republic of China
| | - Su-Yun Xiang
- College of Food Sciences and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210046, People's Republic of China.
| | - Shu-Qin Yu
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210046, People's Republic of China.
| |
Collapse
|
11
|
Gokulan K, Kumar A, Lahiani MH, Sutherland VL, Cerniglia CE, Khare S. Differential Toxicological Outcome of Corn Oil Exposure in Rats and Mice as Assessed by Microbial Composition, Epithelial Permeability, and Ileal Mucosa-Associated Immune Status. Toxicol Sci 2021; 180:89-102. [PMID: 33263755 DOI: 10.1093/toxsci/kfaa177] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Studies to evaluate the toxicity of xenobiotics on the human gut microbiome and related health effects require a diligent selection of (1) an appropriate animal model to facilitate toxicity assessment in predicting human exposure, and (2) an appropriate non-interfering vehicle for the administration of water insoluble compounds. In biomedical studies with water insoluble xenobiotics, corn oil is one of the most commonly used nonaqueous vehicles. This study evaluated the suitability of corn oil as a vehicle in adult female Sprague Dawley rats and adult CD-1 mice; the rodent models that are often utilized in toxicological studies. We studied the host response in terms of change in the intestinal microbiome and mRNA expression of intestinal permeability and immune response-related genes when water (control) and corn oil (2 ml/kg) were administered as a vehicle through oral gavage. The results showed that the use of corn oil as a vehicle has no adverse impact in rats for either the immune response or the intestinal microbial population. On the other hand, mice treated with corn oil showed changes in bacterial community adhered to the ileum, as well as changes in the mRNA expression of intestinal permeability-related and ileal mucosa-associated immune response genes. Overall, results of this study suggest that the type of rodent species and vehicle used in toxicological risk assessments of xenobiotics studies should be taken into consideration in the experimental setup and study design.
Collapse
Affiliation(s)
- Kuppan Gokulan
- Division of Microbiology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, Arkansas 72079, USA
| | - Amit Kumar
- Division of Microbiology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, Arkansas 72079, USA
| | - Mohamed H Lahiani
- Division of Microbiology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, Arkansas 72079, USA
| | - Vicki L Sutherland
- National Institute of Environmental Health Sciences, National Institute of Health, Research Triangle Park, North Carolina 27709, USA
| | - Carl E Cerniglia
- Division of Microbiology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, Arkansas 72079, USA
| | - Sangeeta Khare
- Division of Microbiology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, Arkansas 72079, USA
| |
Collapse
|
12
|
Vaga S, Lee S, Ji B, Andreasson A, Talley NJ, Agréus L, Bidkhori G, Kovatcheva-Datchary P, Park J, Lee D, Proctor G, Ehrlich SD, Nielsen J, Engstrand L, Shoaie S. Compositional and functional differences of the mucosal microbiota along the intestine of healthy individuals. Sci Rep 2020; 10:14977. [PMID: 32917913 PMCID: PMC7486370 DOI: 10.1038/s41598-020-71939-2] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 08/17/2020] [Indexed: 12/14/2022] Open
Abstract
Gut mucosal microbes evolved closest to the host, developing specialized local communities. There is, however, insufficient knowledge of these communities as most studies have employed sequencing technologies to investigate faecal microbiota only. This work used shotgun metagenomics of mucosal biopsies to explore the microbial communities' compositions of terminal ileum and large intestine in 5 healthy individuals. Functional annotations and genome-scale metabolic modelling of selected species were then employed to identify local functional enrichments. While faecal metagenomics provided a good approximation of the average gut mucosal microbiome composition, mucosal biopsies allowed detecting the subtle variations of local microbial communities. Given their significant enrichment in the mucosal microbiota, we highlight the roles of Bacteroides species and describe the antimicrobial resistance biogeography along the intestine. We also detail which species, at which locations, are involved with the tryptophan/indole pathway, whose malfunctioning has been linked to pathologies including inflammatory bowel disease. Our study thus provides invaluable resources for investigating mechanisms connecting gut microbiota and host pathophysiology.
Collapse
Affiliation(s)
- Stefania Vaga
- Centre for Host-Microbiome Interactions, Dental Institute, King's College London, London, UK
| | - Sunjae Lee
- Centre for Host-Microbiome Interactions, Dental Institute, King's College London, London, UK
| | - Boyang Ji
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Anna Andreasson
- Stress Research Institute, Stockholm University, Stockholm, Sweden
- Department of Psychology, Macquarie University, Macquarie Park, NSW, Australia
- Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | | | - Lars Agréus
- Division of Family Medicine and Primary Care, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Gholamreza Bidkhori
- Centre for Host-Microbiome Interactions, Dental Institute, King's College London, London, UK
| | - Petia Kovatcheva-Datchary
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, 41345, Gothenburg, Sweden
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Scientific Research Center for Translational Medicine, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Junseok Park
- Department of Bio and Brain Engineering, KAIST, 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Doheon Lee
- Department of Bio and Brain Engineering, KAIST, 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Gordon Proctor
- Centre for Host-Microbiome Interactions, Dental Institute, King's College London, London, UK
| | | | - Jens Nielsen
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden.
- BioInnovation Institute, Ole Maaløes Vej 3, 2200, Copenhagen N, Denmark.
| | - Lars Engstrand
- Centre for Translational Microbiome Research (CTMR), Department of Microbiology, Tumor and Cell Biology, & Science for Life Laboratory, Karolinska Institute, Stockholm, Sweden.
| | - Saeed Shoaie
- Centre for Host-Microbiome Interactions, Dental Institute, King's College London, London, UK.
- Science for Life Laboratory, KTH-Royal Institute of Technology, Tomtebodavägen 23A, 17165, Solna, Sweden.
| |
Collapse
|
13
|
Zhao C, Wang D, Wu M, Luo Y, Yang M, Guo J, Zhang H, Zhang X. Tumor necrosis factor ligand-related molecule 1A affects the intestinal mucosal barrier function by promoting Th9/interleukin-9 expression. J Int Med Res 2020; 48:300060520926011. [PMID: 32567429 PMCID: PMC7309405 DOI: 10.1177/0300060520926011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 04/20/2020] [Indexed: 01/15/2023] Open
Abstract
OBJECTIVES To investigate the effect of tumor necrosis factor ligand-related molecule 1A (TL1A) on the intestinal mucosal barrier in mice with chronic colitis. METHODS Male TL1A-overexpressing transgenic mice and male C57BL/6 wild-type mice were used to establish a dextran sodium sulfate (DSS)-induced colitis model. The expression of occludin and claudin-1 was observed. Bacterial distribution in the intestinal mucosa and Th9/interleukin (IL)-9 expression were detected. In vitro co-culture systems of naive CD4+ T cells and Caco-2 cells were established and TL1A was added. Changes in transepithelial electrical resistance and IL-9 expression were measured. CD4+IL-9 cells were detected by flow cytometry. RESULTS DSS mice showed a significant down-regulation of occludin and claudin-1 compared with controls. Expression levels of occludin, zonulin-1, and claudin-1 in the Caco-2+TGF-β+IL-4+TL1A group were significantly lower than in the Caco-2+TGF-β+IL-4 group. Bacterial distribution was clearly disordered in the DSS group. Transmembrane resistance of the Caco-2+TGF-β+IL-4+TL1A group was significantly lower and IL-9 expression significantly higher than in the Caco-2+TGF-β+IL-4 group. CONCLUSIONS TL1A overexpression promotes destruction of the intestinal mucosal barrier in mice with chronic colitis. The underlying mechanism may be associated with the promoting role of TL1A in Th9/IL-9 expression, which further destroys the mucosal barrier.
Collapse
Affiliation(s)
- Caihong Zhao
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei, China
- Department of Gastroenterology, Harrison International Peace Hospital, Hengshui, Hebei, China
| | - Dong Wang
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei, China
| | - Mengyao Wu
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei, China
| | - Yuxin Luo
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei, China
| | - Mingyue Yang
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei, China
| | - Jinbo Guo
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei, China
| | - Hong Zhang
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei, China
| | - Xiaolan Zhang
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei, China
| |
Collapse
|
14
|
Bluemel S, Wang L, Kuelbs C, Moncera K, Torralba M, Singh H, Fouts DE, Schnabl B. Intestinal and hepatic microbiota changes associated with chronic ethanol administration in mice. Gut Microbes 2020; 11:265-275. [PMID: 30982395 PMCID: PMC7524386 DOI: 10.1080/19490976.2019.1595300] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 01/31/2019] [Accepted: 03/07/2019] [Indexed: 02/07/2023] Open
Abstract
Alcohol-induced liver disease is closely related to translocation of bacterial products and bacteria from the intestine to the liver. However, it is not known whether bacterial translocation to the liver depends on certain intestinal microbiota changes that would predispose bacteria to translocate to the liver. In this study, we investigated the microbiota in the jejunum, ileum, cecum, feces and liver of mice subjected to chronic ethanol feeding using a Lieber DeCarli diet model of chronic ethanol feeding for 8 weeks. We demonstrate that chronic ethanol administration changes alpha diversity in the ileum and the liver and leads to compositional changes especially in the ileum. This is largely driven by an increase in gram-negative phyla - the source of endotoxins. Moreover, gram-negative Prevotella not only increased in the mucus layer of the ileum but also in liver samples. These results suggest that bacterial translocation to the liver might be associated with microbiota changes in the distal gastrointestinal tract.
Collapse
Affiliation(s)
- Sena Bluemel
- Department of Medicine, University of California San Diego, La Jolla, USA
- Division of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Lirui Wang
- Department of Medicine, University of California San Diego, La Jolla, USA
- Department of Medicine, VA San Diego Healthcare System, San Diego, USA
| | - Claire Kuelbs
- Rockville Campus, J. Craig Venter Institute, Rockville, USA
| | - Kelvin Moncera
- Rockville Campus, J. Craig Venter Institute, Rockville, USA
| | | | - Harinder Singh
- Rockville Campus, J. Craig Venter Institute, Rockville, USA
| | | | - Bernd Schnabl
- Department of Medicine, University of California San Diego, La Jolla, USA
- Department of Medicine, VA San Diego Healthcare System, San Diego, USA
| |
Collapse
|
15
|
Zheng Y, Hu G, Wu W, Qiu L, Bing X, Chen J. Time-dependent gut microbiota analysis of juvenile Oreochromis niloticus by dietary supplementation of resveratrol. Arch Microbiol 2019; 202:43-53. [PMID: 31463601 DOI: 10.1007/s00203-019-01712-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 06/24/2019] [Accepted: 07/31/2019] [Indexed: 02/07/2023]
Abstract
To evaluate the changes in bacterial diversity at various time points under resveratrol supplementation, we aimed to investigate the diversification of gut microbiota and the changes in total genetic diversity. We performed 16S rDNA gene sequencing at different time points (15, 30, and 45 days) to analyze the gut microbiota of tilapia. Fusobacteria, Proteobacteria, and Bacteroidetes (15 days) or Cyanobacteria (30 and 45 days) were found to be the three most abundant phyla. Cyanobacteria (15 and 30 days), Proteobacteria (15 days), Firmicutes and Chlamydiae (30 and 45 days), Planctomycetes (30 days), Bacteroidetes, Actinobacteria, and Fusobacteria (45 days) in the 0.05 g/kg RES group increased as compared to that in the controls. Proteobacteria and Cyanobacteria significantly decreased and increased at 30 and 45 days, respectively, while the reverse pattern was observed at 15 days. The Bacteroidetes:Firmicutes and Proteobacteria:Cyanobacteria ratios were significantly increased (15 and 45 days, P < 0.05) and decreased (30 days, P < 0.05). RES supplementation did not affect the richness and diversity of the gut microbiota in tilapia. Our findings may contribute to the development of strategies for the management of diseases.
Collapse
Affiliation(s)
- Yao Zheng
- Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences/Fishery Eco-Environment Monitoring Center of Lower Reaches of Yangtze River, Ministry of Agriculture/Laboratory of Quality and Safety Risk Assessment for Aquatic Products on Environmental Factors (Wuxi), Ministry of Agriculture, No. 9 Shanshui East Rd., Wuxi, Jiangsu, 214081, People's Republic of China
| | - Gengdong Hu
- Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences/Fishery Eco-Environment Monitoring Center of Lower Reaches of Yangtze River, Ministry of Agriculture/Laboratory of Quality and Safety Risk Assessment for Aquatic Products on Environmental Factors (Wuxi), Ministry of Agriculture, No. 9 Shanshui East Rd., Wuxi, Jiangsu, 214081, People's Republic of China
| | - Wei Wu
- Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences/Fishery Eco-Environment Monitoring Center of Lower Reaches of Yangtze River, Ministry of Agriculture/Laboratory of Quality and Safety Risk Assessment for Aquatic Products on Environmental Factors (Wuxi), Ministry of Agriculture, No. 9 Shanshui East Rd., Wuxi, Jiangsu, 214081, People's Republic of China
| | - Liping Qiu
- Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences/Fishery Eco-Environment Monitoring Center of Lower Reaches of Yangtze River, Ministry of Agriculture/Laboratory of Quality and Safety Risk Assessment for Aquatic Products on Environmental Factors (Wuxi), Ministry of Agriculture, No. 9 Shanshui East Rd., Wuxi, Jiangsu, 214081, People's Republic of China
| | - Xuwen Bing
- Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences/Fishery Eco-Environment Monitoring Center of Lower Reaches of Yangtze River, Ministry of Agriculture/Laboratory of Quality and Safety Risk Assessment for Aquatic Products on Environmental Factors (Wuxi), Ministry of Agriculture, No. 9 Shanshui East Rd., Wuxi, Jiangsu, 214081, People's Republic of China.
| | - Jiazhang Chen
- Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences/Fishery Eco-Environment Monitoring Center of Lower Reaches of Yangtze River, Ministry of Agriculture/Laboratory of Quality and Safety Risk Assessment for Aquatic Products on Environmental Factors (Wuxi), Ministry of Agriculture, No. 9 Shanshui East Rd., Wuxi, Jiangsu, 214081, People's Republic of China. .,Key Laboratory of Control of Quality and Safety for Aquatic Products, Ministry of Agriculture, Beijing, 100141, People's Republic of China.
| |
Collapse
|
16
|
Harris KG, Chang EB. The intestinal microbiota in the pathogenesis of inflammatory bowel diseases: new insights into complex disease. Clin Sci (Lond) 2018; 132:2013-2028. [PMID: 30232239 PMCID: PMC6907688 DOI: 10.1042/cs20171110] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 08/30/2018] [Accepted: 09/04/2018] [Indexed: 12/16/2022]
Abstract
Inflammatory bowel diseases (IBD) are a group of chronic diseases of increasing worldwide prevalence characterized by gastrointestinal (GI) inflammation leading to debilitating symptoms and complications. The contribution of the intestinal microbiota to the pathogenesis and etiology of these diseases is an area of active research interest. Here, we discuss key mechanisms underlying the chronic inflammation seen in IBD as well as evidence implicating the intestinal microbiota in the development and potentiation of that inflammation. We also discuss recently published work in areas of interest within the field of microbial involvement in IBD pathogenesis - the importance of proper microecology within the GI tract, the evidence that the intestinal microbiota transduces environmental and genetic risk factors for IBD, and the mechanisms by which microbial products contribute to communication between microbe and host. There is an extensive body of published research on the evidence for microbial involvement in IBD; the goal of this review is to highlight the growing edges of the field where exciting and innovative research is pushing the boundaries of the conceptual framework of the role of the intestinal microbiota in IBD pathogenesis.
Collapse
Affiliation(s)
| | - Eugene B Chang
- Department of Medicine, University of Chicago, Chicago, IL 60637, U.S.A.
| |
Collapse
|
17
|
Kroschwald S, Chiu CY, Heydeck D, Rohwer N, Gehring T, Seifert U, Lux A, Rothe M, Weylandt KH, Kuhn H. Female mice carrying a defective Alox15 gene are protected from experimental colitis via sustained maintenance of the intestinal epithelial barrier function. Biochim Biophys Acta Mol Cell Biol Lipids 2018; 1863:866-880. [PMID: 29702245 DOI: 10.1016/j.bbalip.2018.04.019] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 04/17/2018] [Accepted: 04/21/2018] [Indexed: 12/19/2022]
Abstract
Lipoxygenases (ALOXs) are involved in the regulation of cellular redox homeostasis. They also have been implicated in the biosynthesis of pro- and anti-inflammatory lipid mediators and play a role in the pathogenesis of inflammatory diseases, which constitute a major health challenge owing to increasing incidence and prevalence in all industrialized countries around the world. To explore the pathophysiological role of Alox15 (leukocyte-type 12-LOX) in mouse experimental colitis we tested the impact of systemic inactivation of the Alox15 gene on the extent of dextrane sulfate sodium (DSS) colitis. We found that in wildtype mice expression of the Alox15 gene was augmented during DSS-colitis while expression of other Alox genes (Alox5, Alox15b) was hardly altered. Systemic Alox15 (leukocyte-type 12-LOX) deficiency induced less severe colitis symptoms and suppressed in vivo formation of 12-hydroxyeicosatetraenoic acid (12-HETE), the major Alox15 (leukocyte-type 12-LOX) product in mice. These alterations were paralleled by reduced expression of pro-inflammatory gene products, by sustained expression of the zonula occludens protein 1 (ZO-1) and by a less impaired intestinal epithelial barrier function. These results are consistent with in vitro incubations of colon epithelial cells, in which addition of 12S-HETE compromised enantioselectively transepithelial electric resistance. Consistent with these data transgenic overexpression of human ALOX15 intensified the inflammatory symptoms. In summary, our results indicate that systemic Alox15 (leukocyte-type 12-LOX) deficiency protects mice from DSS-colitis. Since exogenous 12-HETE compromises the expression of the tight junction protein ZO-1 the protective effect has been related to a less pronounced impairment of the intestinal epithelial barrier function.
Collapse
Affiliation(s)
- Saskia Kroschwald
- Institute of Biochemistry, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Chariteplatz 1, D-10117 Berlin, Germany; Institute for Molecular and Clinical Immunology, Medical Faculty of the Otto-von-Guericke-University, Leipziger Str. 44, 39120 Magdeburg, Germany
| | - Cheng-Ying Chiu
- Division of Medicine, Department of Hepatology, Gastroenterology and Metabolism, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Dagmar Heydeck
- Institute of Biochemistry, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Chariteplatz 1, D-10117 Berlin, Germany
| | - Nadine Rohwer
- Division of Medicine, Department of Hepatology, Gastroenterology and Metabolism, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Tatjana Gehring
- Institute of Biochemistry, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Chariteplatz 1, D-10117 Berlin, Germany
| | - Ulrike Seifert
- Institute for Molecular and Clinical Immunology, Medical Faculty of the Otto-von-Guericke-University, Leipziger Str. 44, 39120 Magdeburg, Germany
| | - Anke Lux
- Institute for Molecular and Clinical Immunology, Medical Faculty of the Otto-von-Guericke-University, Leipziger Str. 44, 39120 Magdeburg, Germany
| | - Michael Rothe
- Lipidomix GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany
| | - Karsten-Henrich Weylandt
- Division of Medicine, Department of Hepatology, Gastroenterology and Metabolism, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Augustenburger Platz 1, 13353 Berlin, Germany; Division of Medicine, Department of Gastroenterology and Oncology, Ruppiner Kliniken, Brandenburg Medical School, 16816 Neuruppin, Germany.
| | - Hartmut Kuhn
- Institute of Biochemistry, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Chariteplatz 1, D-10117 Berlin, Germany.
| |
Collapse
|
18
|
The Potential Effect of Chinese Herbal Formula Hongqijiangzhi Fang in Improving NAFLD: Focusing on NLRP3 Inflammasome and Gut Microbiota. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:5378961. [PMID: 29675053 PMCID: PMC5841032 DOI: 10.1155/2018/5378961] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 01/23/2018] [Indexed: 12/12/2022]
Abstract
The present study investigates the potential therapeutic mechanism underlying the effects of the Chinese herbal formula Hongqijiangzhi Fang (HJF) on nonalcoholic fatty liver disease (NAFLD) in rats. Male Sprague Dawley (SD) rats were randomly divided into 4 groups (n = 8): control group was fed a normal diet, three other groups were fed high-fat diets (HFD), and the two treatment groups were intragastrically given a compound probiotic or HJF during the molding time. After 16 w, related indices were detected. The results showed that HJF significantly reduced abdominal aorta serum cholesterol (TC), triglyceride (TG), low-density lipoprotein (LDL), IL-1β, and IL-18, portal venous serum lipopolysaccharide (LPS), and liver TC and TG levels in HFD-fed rats. HJF ameliorated hepatic steatosis in the liver and improved the intestinal barrier in HFD-fed rats. Activation of the NLRP3 inflammasome was reduced by HJF in HFD-fed rats. Additionally, the abundances of A. muciniphila (Verrucomicrobiaceae), F. rappini (Helicobacteraceae), and Enterobacteriaceae bacteria significantly decreased in HJF-treated HFD-fed rats. In conclusion, these result suggested that the Chinese herbal formula HJF reduced hepatic steatosis maybe through decreasing certain gut bacteria (such as Enterobacteriaceae bacteria and F. rappini), alleviating intestinal endotoxemia and reducing NLRP3 inflammasome activation.
Collapse
|
19
|
Chiodini RJ, Dowd SE, Barron JN, Galandiuk S, Davis B, Glassing A. Transitional and temporal changes in the mucosal and submucosal intestinal microbiota in advanced Crohn's disease of the terminal ileum. J Med Microbiol 2018; 67:549-559. [PMID: 29458679 DOI: 10.1099/jmm.0.000690] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
PURPOSE Crohn's disease is a chronic debilitating intestinal syndrome of unknown aetiology that is thought to result in part from an imbalance (dysbiosis) of the intestinal microbial populations, known as the microbiota. In this study we sought to compare the microbiota at the mucosal and submucosal levels at the resection margin in Crohn's disease to those in other intestinal dysbiotic disease controls to determine the level of bacterial translocation. METHODOLOGY 16S microbiota sequencing was performed on DNA extracted from mucosal and submucosal samples from resected intestinal tissues from Crohn's disease and controls. RESULTS Grossly normal appearing tissue at the resection margin showed early signs, suggesting bacterial translocation, with two bacterial families having penetrated the mucosal surfaces. In contrast, 4 and 13 bacterial families were present within submucosal tissues at the disease centre and disease margin, respectively. Although there was no significant difference in biodiversity, there was increased bacterial richness in the Crohn's disease group as compared to non-IBD controls. CONCLUSION The presence and/or absence of certain bacteria suggested disease-specific ecological or micro-environmental pressures driving or excluding certain organisms in Crohn's disease. The data suggest that several of the dysbiotic conditions previously reported for Crohn's disease are not unique but common to general dysbiosis. The examination of multiple intestinal sites in advanced disease may provide a spectrum of disease from early onset at the resection margin to active disease at the disease margin and late-stage fibrostenotic disease at the centre of the lesion, and a unique etiopathogenic view of Crohn's disease.
Collapse
Affiliation(s)
- Rodrick J Chiodini
- Department of Biological and Physical Sciences, Montana State University-Billings, Billings, MT, USA.,St Vincent Healthcare, Sisters of Charity of Leavenworth Health System, Billings, MT, USA
| | - Scot E Dowd
- Molecular Research (Mr DNA), Shallowater, TX, USA
| | - James N Barron
- Department of Biological and Physical Sciences, Montana State University-Billings, Billings, MT, USA
| | - Susan Galandiuk
- Department of Surgery, University of Louisville, Louisville, KY, USA
| | - Brian Davis
- Department of Surgery, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, USA
| | - Angela Glassing
- Department of Biological and Physical Sciences, Montana State University-Billings, Billings, MT, USA
| |
Collapse
|
20
|
Souza AL, Fiorini Aguiar SL, Gonçalves Miranda MC, Lemos L, Freitas Guimaraes MA, Reis DS, Vieira Barros PA, Veloso ES, Carvalho TG, Ribeiro FM, Ferreira E, Cara DC, Gomes-Santos AC, Faria AMC. Consumption of Diet Containing Free Amino Acids Exacerbates Colitis in Mice. Front Immunol 2017; 8:1587. [PMID: 29209321 PMCID: PMC5701921 DOI: 10.3389/fimmu.2017.01587] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 11/03/2017] [Indexed: 12/25/2022] Open
Abstract
Dietary proteins can influence the maturation of the immune system, particularly the gut-associated lymphoid tissue, when consumed from weaning to adulthood. Moreover, replacement of dietary proteins by amino acids at weaning has been shown to impair the generation of regulatory T cells in the gut as well as immune activities such as protective response to infection, induction of oral and nasal tolerance as well as allergic responses. Polymeric and elemental diets are used in the clinical practice, but the specific role of intact proteins and free amino acids during the intestinal inflammation are not known. It is plausible that these two dietary nitrogen sources would yield distinct immunological outcomes since proteins are recognized by the immune system as antigens and amino acids do not bind to antigen-recognition receptors but instead to intracellular receptors such as mammalian target of rapamycin (mTOR). In this study, our aim was to evaluate the effects of consumption of an amino acid-containing diet (AA diet) versus a control protein-containing diet in adult mice at steady state and during colitis development. We showed that consumption of a AA diet by adult mature mice lead to various immunological changes including decrease in the production of serum IgG as well as increase in the levels of IL-6, IL-17A, TGF-β, and IL-10 in the small and large intestines. It also led to changes in the intestinal morphology, to increase in intestinal permeability, in the number of total and activated CD4+ T cells in the small intestine as well as in the frequency of proliferating cells in the colon. Moreover, consumption of AA diet during and prior to development of dextran sodium sulfate-induced colitis exacerbated gut inflammation. Administration of rapamycin during AA diet consumption prevented colitis exacerbation suggesting that mTOR activation was involved in the effects triggered by the AA diet. Therefore, our study suggests that different outcomes can result from the use of diets containing either intact proteins or free amino acids such as elemental, semielemental, and polymeric diets during intestinal inflammation. These results may contribute to the design of nutritional therapeutic intervention for inflammatory bowel diseases.
Collapse
Affiliation(s)
- Adna Luciana Souza
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Centro das Ciências Biológicas e da Saúde, Universidade Federal do Oeste da Bahia, Barreiras, Brazil
| | - Sarah Leão Fiorini Aguiar
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Luisa Lemos
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Daniela Silva Reis
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Emerson Soares Veloso
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Fabiola Mara Ribeiro
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Enio Ferreira
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Denise Carmona Cara
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ana Cristina Gomes-Santos
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Centro Universitário UNA, Belo Horizonte, Brazil
| | - Ana Maria Caetano Faria
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
21
|
Spadoni I, Fornasa G, Rescigno M. Organ-specific protection mediated by cooperation between vascular and epithelial barriers. Nat Rev Immunol 2017; 17:761-773. [PMID: 28869253 DOI: 10.1038/nri.2017.100] [Citation(s) in RCA: 130] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Immune privilege is a complex process that protects organs from immune-mediated attack and damage. It is accomplished by a series of cellular barriers that both control immune cell entry and promote the development of tolerogenic immune cells. In this Review, we describe the vascular endothelial and epithelial barriers in organs that are commonly considered to be immune privileged, such as the brain and the eye. We compare these classical barriers with barriers in the intestine, which share features with barriers of immune-privileged organs, such as the capacity to induce tolerance and to protect from external insults. We suggest that when intestinal barriers break down, disruption of other barriers at distant sites can ensue, and this may underlie the development of various neurological, metabolic and intestinal disorders.
Collapse
Affiliation(s)
- Ilaria Spadoni
- Department of Experimental Oncology, European Institute of Oncology, 20139 Milan, Italy
| | - Giulia Fornasa
- Department of Experimental Oncology, European Institute of Oncology, 20139 Milan, Italy
| | - Maria Rescigno
- Department of Experimental Oncology, European Institute of Oncology, 20139 Milan, Italy.,Dipartimento di Scienze della Salute, Università degli Studi di Milano, 20122 Milan, Italy
| |
Collapse
|
22
|
The Interplay between Defensins and Microbiota in Crohn's Disease. Mediators Inflamm 2017; 2017:8392523. [PMID: 28246439 PMCID: PMC5299173 DOI: 10.1155/2017/8392523] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 12/16/2016] [Accepted: 01/04/2017] [Indexed: 02/08/2023] Open
Abstract
Crohn's disease (CD) is a chronic inflammation of the intestinal mucosa, characterized by periods of acute recurrence and remission. Depending on the specific region affected, CD is classified as ileal CD or colonic CD. It is largely accepted that the intestinal microbiota is involved in the onset of the pathology. Indeed, a reduced immune tolerance to components of the intestinal commensal microbiota and inflammation of the intestinal barrier typifies patients with CD. Several studies have shown defective expression of intestinal antimicrobial peptides (AMPs) in patients with CD compared to controls, particularly defensins. A reduction in α-defensins is observed in ileal CD, while β-defensins are increased in colonic CD. In addition to an immunological basis, the disease is frequently associated with genetic alterations including mutations of NOD2 gene. Several therapeutic strategies to circumvent the dysfunction observed in CD are currently under investigation. These include the use of delivery systems to administer endogenous AMPs and the engineering of peptidomimetics that could ameliorate the severity of CD. In this review, the role defensins play in CD and the strategies aimed at overcoming bacterial resistance will be discussed.
Collapse
|
23
|
Joly Condette C, Elion Dzon B, Hamdad F, Biendo M, Bach V, Khorsi-Cauet H. Use of molecular typing to investigate bacterial translocation from the intestinal tract of chlorpyrifos-exposed rats. Gut Pathog 2016; 8:50. [PMID: 27826358 PMCID: PMC5097847 DOI: 10.1186/s13099-016-0129-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 10/12/2016] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Human are confronted on a daily basis with contaminant pesticide residues in food, water and other components of the environment. Although the digestive system is the first organ to come into contact with food contaminants, very few data are available on the impact of low-dose pesticide exposure during the in utero and postnatal periods on intestinal bacterial translocation (BT). Previous studies have revealed that chlorpyrifos (CPF) exposure is associated with intestinal dysbiosis and the contamination of sterile organs. Here, molecular typing was used to investigate intestinal bacterial translocation in rats exposed to chlorpyrifos in utero and during lactation. The translocated bacteria were profiled, and CPF tolerance and antibiotic resistance traits were determined. METHODS A total of 72 intestinal segments and extra-intestinal organs were obtained from 14 CPF-exposed rats. The samples were cultured to isolate bacterial strains that had tolerated treatment with 1 or 5 mg CPF/kg bodyweight/day in vivo. Strains were identified using matrix-assisted laser desorption/ionization (MALDI) Biotyper. The disk diffusion method was used to determine the antibiotic susceptibility. The isolates were genotyped with PCR assays for the enterobacterial repetitive intergenic consensus sequence and random amplification polymorphic DNA. RESULTS Bacterial translocation was confirmed for 7 of the 31 strains (22.6 %) isolated from extra-intestinal sites. Overall, the most prevalent bacteria were Staphylococcus aureus (55.5 % of the 72 intestinal and extra-intestinal isolates), Enterococcus faecalis (27.7 %) and Bacillus cereus (9.8 %). 5 % of the S. aureus isolates displayed methicillin resistance. Seventy two strains were identified phenotypically, and seven translocated strains (mainly S. aureus) were identified by genotyping. Genotypically confirmed translocation was mainly observed found in pesticide-exposed groups (6 out of 7). CONCLUSION BT from the intestinal tract colonized normally sterile extra-intestinal organs in CPF-exposed rats. Our findings validate the use of molecular typing for the assessment of intestinal BT in CPF-exposed rats during critical periods of development.
Collapse
Affiliation(s)
- Claire Joly Condette
- Laboratoire PeriTox UMR I 01, Faculty of Medicine, Centre Universitaire de Recherche Scientifique, Université de Picardie Jules Verne, Avenue René Laёnnec, 80054 Amiens cedex 1, France
- Laboratoire LNPC EA4666, Faculty of Medicine, Centre Universitaire de Recherche Scientifique, Université de Picardie Jules Verne, Avenue René Laёnnec, 80054 Amiens cedex 1, France
| | - Bertin Elion Dzon
- Laboratoire PeriTox UMR I 01, Faculty of Medicine, Centre Universitaire de Recherche Scientifique, Université de Picardie Jules Verne, Avenue René Laёnnec, 80054 Amiens cedex 1, France
| | - Farida Hamdad
- Laboratoire de Bactériologie, Centre Hospitalier Universitaire Amiens Picardie, Avenue René Laёnnec, Salouёl, 80054 Amiens cedex 1, France
| | - Maurice Biendo
- Laboratoire PeriTox UMR I 01, Faculty of Medicine, Centre Universitaire de Recherche Scientifique, Université de Picardie Jules Verne, Avenue René Laёnnec, 80054 Amiens cedex 1, France
- Laboratoire de Bactériologie, Centre Hospitalier Universitaire Amiens Picardie, Avenue René Laёnnec, Salouёl, 80054 Amiens cedex 1, France
| | - Véronique Bach
- Laboratoire PeriTox UMR I 01, Faculty of Medicine, Centre Universitaire de Recherche Scientifique, Université de Picardie Jules Verne, Avenue René Laёnnec, 80054 Amiens cedex 1, France
| | - Hafida Khorsi-Cauet
- Laboratoire PeriTox UMR I 01, Faculty of Medicine, Centre Universitaire de Recherche Scientifique, Université de Picardie Jules Verne, Avenue René Laёnnec, 80054 Amiens cedex 1, France
| |
Collapse
|
24
|
Research Advance in Intestinal Mucosal Barrier and Pathogenesis of Crohn's Disease. Gastroenterol Res Pract 2016; 2016:9686238. [PMID: 27651792 PMCID: PMC5019909 DOI: 10.1155/2016/9686238] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 07/07/2016] [Accepted: 08/15/2016] [Indexed: 01/30/2023] Open
Abstract
To date, the etiology and pathogenesis of Crohn's disease (CD) have not been fully elucidated. It is widely accepted that genetic, immune, and environment factors are closely related to the development of CD. As an important defensive line for human body against the environment, intestinal mucosa is able to protect the homeostasis of gut bacteria and alleviate the intestinal inflammatory and immune response. It is evident that the dysfunction of intestinal mucosa barriers plays a crucial role in CD initiation and development. Yet researches are insufficient on intestinal mucosal barrier's action in the prevention of CD onset. This article summarizes the research advances about the correlations between the disorders of intestinal mucosal barriers and CD.
Collapse
|