1
|
Fajardo C RJ, Clavijo C, Díaz GJ, Cadavid LF. Tissue distribution and expression dynamics of trefoil factor genes in the hydroid Hydractinia symbiolongicarpus. Gene 2024; 929:148824. [PMID: 39103057 DOI: 10.1016/j.gene.2024.148824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 07/26/2024] [Accepted: 08/02/2024] [Indexed: 08/07/2024]
Abstract
Proteins of the trefoil factor family (TFF) participate in mucosal repair and are formed by single or tandemly repeated trefoil domains. TFFs have been extensively studied in mammals and amphibians, but they have not been functionally characterized in other animals. Here we report the identification of two genes expressed in the hydroid Hydractinia symbiolongicarpus, predicted to encode trefoil domain-containing peptides, one with four trefoil domains in tandem and the other one with a trefoil domain flanked by two ShKT domains. Differential expression analyses by qPCR after an immune challenge and an induced mechanical damage, reveal that the former gene (hysyTFF) had no significant changes in expression after the inductions. However, the latter (hysyTFF-like) was overexpressed after three hours post immune challenge and was downregulated after the first hour post epithelial damage. Immunoblot analyses using specific IgY antibodies revealed that hysyTFF is secreted as a high molecular weight complex. Finally, whole mount immunofluorescence assays showed that hysyTFF was predominantly expressed in the endoderm of stolons and polyps, and sparsely in the ectoderm of both polyps and larvae. Thus, the tissue distribution and expression dynamics of trefoil factor genes in H. symbiolongicarpus suggest that hysyTFF is part of an ancient mechanism of epithelial restitution, and the newly reported hysyTFF-like might act as an immune effector gene, perhaps encoding an antibacterial peptide.
Collapse
Affiliation(s)
- R Johana Fajardo C
- Departamento de Biología, Facultad de Ciencias, Universidad Nacional de Colombia - Sede Bogotá, 111321 Bogotá, Colombia
| | - Carlos Clavijo
- Departamento de Biología, Facultad de Ciencias, Universidad Nacional de Colombia - Sede Bogotá, 111321 Bogotá, Colombia
| | - Gonzalo J Díaz
- Departamento de Salud Animal, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional de Colombia - Sede Bogotá, 111321 Bogotá, Colombia
| | - Luis F Cadavid
- Instituto de Genética, Universidad Nacional de Colombia - Sede Bogotá, 111321 Bogotá, Colombia.
| |
Collapse
|
2
|
Liebing E, Krug SM, Neurath MF, Siegmund B, Becker C. Wall of Resilience: How the Intestinal Epithelium Prevents Inflammatory Onslaught in the Gut. Cell Mol Gastroenterol Hepatol 2024; 19:101423. [PMID: 39461590 PMCID: PMC11720114 DOI: 10.1016/j.jcmgh.2024.101423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 10/17/2024] [Accepted: 10/18/2024] [Indexed: 10/29/2024]
Abstract
The intestinal epithelium forms the boundary between the intestinal immune system in the lamina propria and the outside world, the intestinal lumen, which contains a diverse array of microbial and environmental antigens. Composed of specialized cells, this epithelial monolayer has an exceptional turnover rate. Differentiated epithelial cells are released into the intestinal lumen within a few days, at the villus tip, a process that requires strict regulation. Dysfunction of the epithelial barrier increases the intestinal permeability and paves the way for luminal antigens to pass into the intestinal serosa. Stem cells at the bottom of Lieberkühn crypts provide a constant supply of mature epithelial cells. Differentiated intestinal epithelial cells exhibit a diverse array of mechanisms that enable communication with surrounding cells, fortification against microorganisms, and orchestration of nutrient absorption and hormonal balance. Furthermore, tight junctions regulate paracellular permeability properties, and their disruption can lead to an impairment of the intestinal barrier, allowing inflammation to develop or further progress. Intestinal epithelial cells provide a communication platform through which they maintain homeostasis with a spectrum of entities including immune cells, neuronal cells, and connective tissue cells. This homeostasis can be disrupted in disease, such as inflammatory bowel disease. Patients suffering from inflammatory bowel disease show an impaired gut barrier, dysregulated cellular communication, and aberrant proliferation and demise of cells. This review summarizes the individual cellular and molecular mechanisms pivotal for upholding the integrity of the intestinal epithelial barrier and shows how these can be disrupted in diseases, such as inflammatory bowel disease.
Collapse
Affiliation(s)
- Eva Liebing
- Department of Medicine 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; Deutsches Zentrum Immuntherapie, Erlangen, Germany
| | - Susanne M Krug
- Clinical Physiology/Nutritional Medicine, Charité-Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Markus F Neurath
- Department of Medicine 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; Deutsches Zentrum Immuntherapie, Erlangen, Germany
| | - Britta Siegmund
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Christoph Becker
- Department of Medicine 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; Deutsches Zentrum Immuntherapie, Erlangen, Germany.
| |
Collapse
|
3
|
Lin Z, Wan X, Zhang T, Huo H, Zhang X, Li K, Bei W, Guo J, Yang Y. Trefoil factor 3: New highlights in chronic kidney disease research. Cell Signal 2022; 100:110470. [PMID: 36122885 DOI: 10.1016/j.cellsig.2022.110470] [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: 07/22/2022] [Revised: 09/10/2022] [Accepted: 09/13/2022] [Indexed: 11/28/2022]
Abstract
Trefoil factor 3 (TFF3, also known as intestinal trefoil factor) is a small-molecule peptide containing a typical trefoil structure. TFF3 has several biological effects, such as wound healing, immune regulation, neuroprotection, and cell migration and proliferation promotion. Although TFF3 binding sites were identified in rat kidneys more than a decade ago, the specific effects of this small-molecule peptide on kidneys remain unclear. Until recently, much of the research on TFF3 in the kidney field has focused exclusively on its role as a biomarker. Notably, a large prospective randomized study of patients with 29 common clinical diseases revealed that chronic kidney disease (CKD) was associated with the highest serum TFF3 levels, which were 3-fold higher than in acute gastroenteritis, which had the second-highest levels. Examination of each stage of CKD revealed that urine and serum TFF3 levels significantly increased with the progression of CKD. These results suggest that the role of TFF3 in CKD needs further research. The present review summarizes the renal physiological expression, biological functions, and downstream signaling of TFF3, as well as the upstream events that lead to high expression of TFF3 in CKD.
Collapse
Affiliation(s)
- Ziyang Lin
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Science and Technology Building, 280 Waihuan East Road, Guangzhou Higher Education Mega, Guangzhou, China
| | - Xiaofen Wan
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Science and Technology Building, 280 Waihuan East Road, Guangzhou Higher Education Mega, Guangzhou, China
| | - Tao Zhang
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Science and Technology Building, 280 Waihuan East Road, Guangzhou Higher Education Mega, Guangzhou, China
| | - Hongyan Huo
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Science and Technology Building, 280 Waihuan East Road, Guangzhou Higher Education Mega, Guangzhou, China
| | - Xiaoyu Zhang
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Science and Technology Building, 280 Waihuan East Road, Guangzhou Higher Education Mega, Guangzhou, China
| | - Kunping Li
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Science and Technology Building, 280 Waihuan East Road, Guangzhou Higher Education Mega, Guangzhou, China
| | - Weijian Bei
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Science and Technology Building, 280 Waihuan East Road, Guangzhou Higher Education Mega, Guangzhou, China
| | - Jiao Guo
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Science and Technology Building, 280 Waihuan East Road, Guangzhou Higher Education Mega, Guangzhou, China
| | - Yiqi Yang
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Science and Technology Building, 280 Waihuan East Road, Guangzhou Higher Education Mega, Guangzhou, China.
| |
Collapse
|
4
|
Yang Y, Lin Z, Lin Q, Bei W, Guo J. Pathological and therapeutic roles of bioactive peptide trefoil factor 3 in diverse diseases: recent progress and perspective. Cell Death Dis 2022; 13:62. [PMID: 35039476 PMCID: PMC8763889 DOI: 10.1038/s41419-022-04504-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 12/23/2021] [Accepted: 12/30/2021] [Indexed: 12/16/2022]
Abstract
Trefoil factor 3 (TFF3) is the last small-molecule peptide found in the trefoil factor family, which is mainly secreted by intestinal goblet cells and exerts mucosal repair effect in the gastrointestinal tract. Emerging evidence indicated that the TFF3 expression profile and biological effects changed significantly in pathological states such as cancer, colitis, gastric ulcer, diabetes mellitus, non-alcoholic fatty liver disease, and nervous system disease. More importantly, mucosal protection would no longer be the only effect of TFF3, it gradually exhibits carcinogenic activity and potential regulatory effect of nervous and endocrine systems, but the inner mechanisms remain unclear. Understanding the molecular function of TFF3 in specific diseases might provide a new insight for the clinical development of novel therapeutic strategies. This review provides an up-to-date overview of the pathological effects of TFF3 in different disease and discusses the binding proteins, signaling pathways, and clinical application.
Collapse
Affiliation(s)
- Yiqi Yang
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Science and Technology Building, 280 Waihuan East Road, Guangzhou Higher Education Mega, Guangzhou, China
| | - Ziyang Lin
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Science and Technology Building, 280 Waihuan East Road, Guangzhou Higher Education Mega, Guangzhou, China
| | - Quanyou Lin
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Science and Technology Building, 280 Waihuan East Road, Guangzhou Higher Education Mega, Guangzhou, China
| | - Weijian Bei
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Science and Technology Building, 280 Waihuan East Road, Guangzhou Higher Education Mega, Guangzhou, China
| | - Jiao Guo
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Science and Technology Building, 280 Waihuan East Road, Guangzhou Higher Education Mega, Guangzhou, China.
| |
Collapse
|
5
|
Adewale B, Heintz JR, Pastore CF, Rossi HL, Hung LY, Rahman N, Bethony J, Diemert D, Babatunde JA, Herbert DR. Parasitic helminth infections in humans modulate Trefoil Factor levels in a manner dependent on the species of parasite and age of the host. PLoS Negl Trop Dis 2021; 15:e0009550. [PMID: 34662329 PMCID: PMC8553090 DOI: 10.1371/journal.pntd.0009550] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 10/28/2021] [Accepted: 10/07/2021] [Indexed: 11/19/2022] Open
Abstract
Helminth infections, including hookworms and Schistosomes, can cause severe disability and death. Infection management and control would benefit from identification of biomarkers for early detection and prognosis. While animal models suggest that Trefoil Factor Family proteins (TFF2 and TFF3) and interleukin-33 (IL-33) -driven type 2 immune responses are critical mediators of tissue repair and worm clearance in the context of hookworm infection, very little is known about how they are modulated in the context of human helminth infection. We measured TFF2, TFF3, and IL-33 levels in serum from patients in Brazil infected with Hookworm and/or Schistosomes, and compared them to endemic and non-endemic controls. TFF2 was specifically elevated by Hookworm infection in females, not Schistosoma or co-infection. This elevation was correlated with age, but not worm burden. TFF3 was elevated by Schistosoma infection and found to be generally higher in females. IL-33 was not significantly altered by infection. To determine if this might apply more broadly to other species or regions, we measured TFFs and cytokine levels (IFNγ, TNFα, IL-33, IL-13, IL-1β, IL-17A, IL-22, and IL-10) in both the serum and urine of Nigerian school children infected with S. haematobium. We found that serum levels of TFF2 and 3 were reduced by infection, likely in an age dependent manner. In the serum, only IL-10 and IL-13 were significantly increased, while in urine IFN-γ, TNF-α, IL-13, IL-1β, IL-22, and IL-10 were significantly increased in by infection. Taken together, these data support a role for TFF proteins in human helminth infection. Billions of people are infected with parasitic helminths across the globe, especially in resource poor regions. These infections can result in severe developmental delay, disability, and death. Adequate management of helminth infection would benefit from the identification of host biomarkers in easily obtained samples (e.g. serum or urine) that correlate to infection state. Our goal was to determine if specific proteins involved in tissue repair and immune modulation are altered by infection with specific helminth species in Brazil (hookworm and S. mansoni species of blood fluke) or Nigeria (S. haematobium species of blood fluke). One of these proteins, Trefoil Factor 2 (TFF2), was elevated in the serum of hookworm infected women from Brazil, while another, TFF3 is higher in women than men, but also increased by S. mansoni blood fluke infection. In contrast, both TFFs were decreased in the serum of Nigerian children infected by S. haematobium, while many pro-inflammatory cytokines were increased in the urine, where the eggs emerge from host tissue.
Collapse
Affiliation(s)
- Babatunde Adewale
- Public Health Department, Nigerian Institute of Medical Research, Yaba, Lagos, Nigeria
| | - Jonathan R. Heintz
- University of Pennsylvania, Perlman School of Medicine Biostatistics Analysis Center, Philadelphia, Pennsylvania, United States of America
| | - Christopher F. Pastore
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania, United States of Amerca
| | - Heather L. Rossi
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania, United States of Amerca
| | - Li-Yin Hung
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania, United States of Amerca
- Department of Medicine, Division of Experimental Medicine, University of California, San Francisco, San Francisco, California, United States of Amerca
| | - Nurudeen Rahman
- Public Health Department, Nigerian Institute of Medical Research, Yaba, Lagos, Nigeria
| | - Jeff Bethony
- Department of Microbiology, Immunology & Tropical Medicine, George Washington University Medical Center, Washington, District of Columbia, United States of Amerca
| | - David Diemert
- Department of Microbiology, Immunology & Tropical Medicine, George Washington University Medical Center, Washington, District of Columbia, United States of Amerca
| | - James Ayorinde Babatunde
- Department of Biochemistry & Nutrition, Nigerian Institute of Medical Research, Yaba, Lagos, Nigeria
| | - De’Broski R. Herbert
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania, United States of Amerca
- Department of Medicine, Division of Experimental Medicine, University of California, San Francisco, San Francisco, California, United States of Amerca
- * E-mail:
| |
Collapse
|
6
|
Yin B, Tian-Chu H, Ling-Fen X. Protection by microRNA-7a-5p Antagomir Against Intestinal Mucosal Injury Related to the JNK Pathway in TNBS-Induced Experimental Colitis. THE TURKISH JOURNAL OF GASTROENTEROLOGY : THE OFFICIAL JOURNAL OF TURKISH SOCIETY OF GASTROENTEROLOGY 2021; 32:431-436. [PMID: 34231472 PMCID: PMC8975295 DOI: 10.5152/tjg.2021.20746] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 11/22/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Increasing evidence shows that microRNA-7a-5p (miR-7a-5p) plays an important role in regulating the inflammatory process in inflammatory bowel disease (IBD). How miR-7a-5p contributes to this process is poorly defined. The purpose of this study was to examine whether miR-7a-5p regulates 2,4,6-trinitrobenzene sulfonic acid (TNBS)-induced inflammatory responses via the JNK pathway. METHODS Colitis was induced in male mice by intracolonic administration of TNBS; mice were divided into 3 groups: normal control (NC), TNBS, and miR-7a-5p antagomir-treated group. Inflammatory responses were estimated by disease activity index (DAI) and histological scores. The relative expressions of miR-7a-5p and tight junction protein, ZO-1, were detected by RT-qPCR. Western blot assays were used to estimate the level of JNK pathway proteins and ZO-1. After miRNA-antagomir injection, the extent of colonic tissue injury and expression levels of ZO-1 and JNK in intestinal tissue were compared. RESULTS miR-7a-5p and p-JNK expression were higher in the intestinal tissue of the TNBS group as compared to NC. Inhibition of the expression of miR-7a-5p resulted in significantly decreased expression of p-JNK but increased expression of ZO-1 and promoted the recovery of intestinal mucosa. CONCLUSION This work demonstrates a correlation between the JNK pathway and miR-7a-5p in TNBS-induced experimental colitis in mice, which may provide a new research direction for the treatment of IBD.
Collapse
Affiliation(s)
- Bao Yin
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China; Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Huang Tian-Chu
- Key Laboratory of Congenital Malformation, Ministry of Health, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Xu Ling-Fen
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| |
Collapse
|
7
|
Chen X, Liu L, Chen W, Qin F, Zhou F, Yang H. Ziyuglycoside II Inhibits Rotavirus Induced Diarrhea Possibly via TLR4/NF-κB Pathways. Biol Pharm Bull 2021; 43:932-937. [PMID: 32475915 DOI: 10.1248/bpb.b19-00771] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Rotavirus (RV) induced diarrhea has been a major reason affecting children healthy under 5 years old especially in developing countries. Although specific vaccines have preventive effects, antiviral therapy is essential for the diarrhea patients. Ziyuglycoside II is a traditional Chinese herb which has been proven to possess anti-virus effects. This study aimed to investigate the roles of Ziyuglycoside II in rotavirus-induced diarrhea and the underlying molecular mechanism. We found that normal MA104 cells treated with RV became swollen and gather together. However, Ziyuglycoside II treatment inhibited cell growth in a dose- and time dependent manner and suppressed RV replication. Moreover, Ziyuglycoside II reversed RV-induced downregulation of anti-inflammatory cytokine interleukin (IL)-10 and upregulation of pro-inflammatory factors, such as interferon-γ (IFN-γ), IL-1β, IL-6, and tumor necrosis factor (TNF-α). Moreover, Ziyuglycoside II administration and ribavirin blocked toll-like receptor 4 (TLR4)/nuclear factor kappa-B (NF-κB) signaling pathway both in mRNA and protein level, which was paralleled with immunohistochemical assay. Additionally, Ziyuglycoside II administration improved diarrhea symptoms and decreased diarrhea scores. Ziyuglycoside II and ribavirin inhibited the apoptosis of small intestine epithelial cells induced by RV. Taken together, RV treatment induced diarrhea. Ziyuglycoside II administration inhibited TLR4/NF-κB pathway and inflammatory response and improved RV-induced diarrhea. The inhibitory effects of Ziyuglycoside II on RV-induced diarrhea predicted Ziyuglycoside II may be a promising drug for diarrhea.
Collapse
Affiliation(s)
- Xiaolan Chen
- College of Veterinary Pharmaceutical, Jiangsu Agri-animal Husbandry Vocational College
| | - Li Liu
- College of Veterinary Pharmaceutical, Jiangsu Agri-animal Husbandry Vocational College
| | - Wei Chen
- College of Veterinary Medicine, Jiangsu Agri-animal Husbandry Vocational College
| | - Feng Qin
- College of Veterinary Pharmaceutical, Jiangsu Agri-animal Husbandry Vocational College
| | - Fang Zhou
- College of Veterinary Pharmaceutical, Jiangsu Agri-animal Husbandry Vocational College
| | - Haifeng Yang
- College of Veterinary Pharmaceutical, Jiangsu Agri-animal Husbandry Vocational College
| |
Collapse
|
8
|
Teng X, Yang Y, Liu L, Yang L, Wu J, Sun M, Xu L. Evaluation of inflammatory bowel disease activity in children using serum trefoil factor peptide. Pediatr Res 2020; 88:792-795. [PMID: 32120375 DOI: 10.1038/s41390-020-0812-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 02/04/2020] [Accepted: 02/08/2020] [Indexed: 11/09/2022]
Abstract
BACKGROUND The diagnosis of IBD and evaluation of treatment require endoscopy, which is difficult in children. This study evaluated the use of TFF3 as a biomarker. METHODS Permeability of the intestinal mucosa and serum TFF3 were assayed and colon tissue was harvested 7 days after inducing IBD in mice with TNBSA. TFF3 was monitored in 51 pediatric IBD patients stratified by active disease or remission and in 20 healthy children. Mucosal healing was assessed by the Simple Endoscopic Score for Crohn Disease and Baron scores in CD and UC patients. RESULTS Histological evaluation revealed transmural inflammation of the colon in IBD model mice. Permeability of the intestinal mucosa and serum TFF3 were both higher in TNBSA-treated than in control mice (P < 0.05). TFF3 was higher in children with active IBD than in those in remission and in healthy children (P < 0.05). TFF3 was positively correlated with the SES-CD score (P < 0.05) but not with either the pediatric CDAI score or the serum CRP. The sensitivity of serum TFF3 for monitoring CD activity was 100% and the specificity was 76.2%. CONCLUSIONS TFF3 level increased with CD activity, which is of significance for diagnosis and for evaluation of mucosal healing. TFF3 could also be a marker in pediatric UC, as TFF3 positively correlated with UCAI. IMPACT The diagnosis and evaluation of IBD is difficult; endoscopy provides objective assessment; TFF3 can be a useful marker instead of endoscopy. TFF3 was increased in active CD of children; TFF3 can be used as a clinical marker of pediatric CD; TFF3 can diagnose and evaluate mucosal healing of CD. Pediatrician should pay attention to clinical marker; TFF3 level may be a key evaluation of mucosal healing of CD; the value of diagnosis of TFF3 in CD is important.
Collapse
Affiliation(s)
- Xu Teng
- Department of Pediatric Gastroenterology, Shengjing Hospital of China Medical University, 110004, Shenyang, Liaoning, China
| | - Yuming Yang
- Department of Pediatric Gastroenterology, Shengjing Hospital of China Medical University, 110004, Shenyang, Liaoning, China
- Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Lu Liu
- Department of Pediatric Gastroenterology, Shengjing Hospital of China Medical University, 110004, Shenyang, Liaoning, China
| | - Lijuan Yang
- Department of Pediatric Gastroenterology, Shengjing Hospital of China Medical University, 110004, Shenyang, Liaoning, China
- Dalian Children's Hospital, Dalian, China
| | - Jie Wu
- Department of Pediatric Gastroenterology, Shengjing Hospital of China Medical University, 110004, Shenyang, Liaoning, China
| | - Mei Sun
- Department of Pediatric Gastroenterology, Shengjing Hospital of China Medical University, 110004, Shenyang, Liaoning, China
| | - Lingfen Xu
- Department of Pediatric Gastroenterology, Shengjing Hospital of China Medical University, 110004, Shenyang, Liaoning, China.
| |
Collapse
|
9
|
Wang J, Zhu G, Sun C, Xiong K, Yao T, Su Y, Fang H. TAK-242 ameliorates DSS-induced colitis by regulating the gut microbiota and the JAK2/STAT3 signaling pathway. Microb Cell Fact 2020; 19:158. [PMID: 32762699 PMCID: PMC7412642 DOI: 10.1186/s12934-020-01417-x] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Accepted: 07/30/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The goal of the present study was to investigate the effects of TAK-242 on the gut microbiota and the TLR4/JAK2/STAT3 signaling pathway in mice with dextran sulfate sodium (DSS)-induced colitis. RESULTS At the phylum level, Bacteroidetes, Firmicutes, Actinobacteria, Cyanobacteria, Epsilonbacteraeota and Proteobacteria were the primary microbiota in the five groups. TAK-242 treatment significantly enhanced Verrucomicrobia and Actinobacteria; significantly decreased Cyanobacteria, Epsilonbacteraeota and Proteobacteria; and particularly promoted the growth of Akkermansia. TAK-242 markedly alleviated DSS-induced colitis symptoms and colonic lesions by promoting IL-10 release, inhibiting IL-17 release, downregulating TLR4 and JAK2/STAT3 mRNA and protein expression and increasing JAK2/STAT3 phosphorylation. CONCLUSION TAK-242 modulates the structure of the gut microbiota in colitis and may be a novel therapeutic candidate for ulcerative colitis.
Collapse
Affiliation(s)
- Jiajia Wang
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China.
| | - Guannan Zhu
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Cheng Sun
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Kangwei Xiong
- Department of Gastroenterology and Hepatology, The Second Hospital of Anhui Medical University, Hefei, Anhui, China
- Center for Gut Microbiota Research, The Second Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Tingting Yao
- Department of Gastroenterology and Hepatology, The Second Hospital of Anhui Medical University, Hefei, Anhui, China
- Center for Gut Microbiota Research, The Second Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yuan Su
- Department of Gastroenterology and Hepatology, The Second Hospital of Anhui Medical University, Hefei, Anhui, China
- Center for Gut Microbiota Research, The Second Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Haiming Fang
- Department of Gastroenterology and Hepatology, The Second Hospital of Anhui Medical University, Hefei, Anhui, China.
- Center for Gut Microbiota Research, The Second Hospital of Anhui Medical University, Hefei, Anhui, China.
| |
Collapse
|
10
|
Guo J, Yang LJ, Sun M, Xu LF. Inhibiting microRNA-7 Expression Exhibited a Protective Effect on Intestinal Mucosal Injury in TNBS-Induced Inflammatory Bowel Disease Animal Model. Inflammation 2019; 42:2267-2277. [PMID: 31494796 DOI: 10.1007/s10753-019-01091-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
This study aimed to explore the expression and correlation of microRNA-7 (miR-7) and trefoil factor 3 (TFF3) genes and proteins in inflammatory bowel disease (IBD) mouse models and to elucidate the effect of miR-7 inhibition in the intestinal mucosa in IBD models. A TNBS-induced IBD mouse model was established. Changes in intestinal inflammation were observed by HE staining, and the expression levels of miR-7 and TFF3 were detected by RT-PCR. After miRNA-antagomir injection, the degree of colonic tissue damage and the expression levels of miR-7 and TFF3 in intestinal tissues were compared. TNBS-induced IBD mice showed significant weight loss, significantly decreased disease activity index (DAI), and a significantly increased pathological damage score. miR-7 was highly expressed in the colon tissue of IBD mice, and TFF3 was downregulated. Inhibition of the expression of miR-7 improved the stool characteristics and fecal occult blood (OB) of IBD mice, significantly increased the expression of TFF3 protein, and decreased the pathological damage scores. In the IBD mouse model, miR-7 posttranscriptionally regulates TFF3. The inhibition of miR-7 expression improves some clinical manifestations of IBD mice, reduces the pathological damage of the intestinal mucosa, and shows a protective effect in IBD.
Collapse
Affiliation(s)
- Jing Guo
- Department of Pediatrics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, 110004, Liaoning, People's Republic of China
| | - Li-Juan Yang
- Dalian Children's Hospital, Dalian, 116000, Liaoning, People's Republic of China
| | - Mei Sun
- Department of Pediatrics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, 110004, Liaoning, People's Republic of China
| | - Ling-Fen Xu
- Department of Pediatrics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, 110004, Liaoning, People's Republic of China.
| |
Collapse
|
11
|
Rashidian A, Rashki A, Abdollahi A, Haddadi NS, Chamanara M, Mumtaz F, Dehpour AR. Dapsone reduced acetic acid-induced inflammatory response in rat colon tissue through inhibition of NF-kB signaling pathway. Immunopharmacol Immunotoxicol 2019; 41:607-613. [DOI: 10.1080/08923973.2019.1678635] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Amir Rashidian
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Asma Rashki
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Abdollahi
- Department of Pathology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nazgol-Sadat Haddadi
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohsen Chamanara
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology, School of Medicine, Aja University of Medical Sciences, Tehran, Iran
| | - Faiza Mumtaz
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad Reza Dehpour
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
12
|
Belle NM, Ji Y, Herbine K, Wei Y, Park J, Zullo K, Hung LY, Srivatsa S, Young T, Oniskey T, Pastore C, Nieves W, Somsouk M, Herbert DR. TFF3 interacts with LINGO2 to regulate EGFR activation for protection against colitis and gastrointestinal helminths. Nat Commun 2019; 10:4408. [PMID: 31562318 PMCID: PMC6764942 DOI: 10.1038/s41467-019-12315-1] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 08/26/2019] [Indexed: 12/14/2022] Open
Abstract
Intestinal epithelial cells (IEC) have important functions in nutrient absorption, barrier integrity, regeneration, pathogen-sensing, and mucus secretion. Goblet cells are a specialized cell type of IEC that secrete Trefoil factor 3 (TFF3) to regulate mucus viscosity and wound healing, but whether TFF3-responsiveness requires a receptor is unclear. Here, we show that leucine rich repeat receptor and nogo-interacting protein 2 (LINGO2) is essential for TFF3-mediated functions. LINGO2 immunoprecipitates with TFF3, co-localizes with TFF3 on the cell membrane of IEC, and allows TFF3 to block apoptosis. We further show that TFF3-LINGO2 interactions disrupt EGFR-LINGO2 complexes resulting in enhanced EGFR signaling. Excessive basal EGFR activation in Lingo2 deficient mice increases disease severity during colitis and augments immunity against helminth infection. Conversely, TFF3 deficiency reduces helminth immunity. Thus, TFF3-LINGO2 interactions de-repress inhibitory LINGO2-EGFR complexes, allowing TFF3 to drive wound healing and immunity.
Collapse
Affiliation(s)
- Nicole Maloney Belle
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, 19140, USA
| | - Yingbiao Ji
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, 19140, USA
| | - Karl Herbine
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, 19140, USA
| | - Yun Wei
- Division of Experimental Medicine, University of California, San Francisco, San Francisco, CA, 94110, USA.,Department of Inflammation and Oncology, Amgen Inc., 1120 Veterans Boulevard, South San Francisco, CA, 94080, USA
| | - JoonHyung Park
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, 19140, USA
| | - Kelly Zullo
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, 19140, USA
| | - Li-Yin Hung
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, 19140, USA.,Division of Experimental Medicine, University of California, San Francisco, San Francisco, CA, 94110, USA
| | - Sriram Srivatsa
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, 19140, USA
| | - Tanner Young
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, 19140, USA
| | - Taylor Oniskey
- Division of Experimental Medicine, University of California, San Francisco, San Francisco, CA, 94110, USA
| | - Christopher Pastore
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, 19140, USA
| | - Wildaliz Nieves
- Division of Gastroenterology at ZSFG, University of California, San Francisco, San Francisco, CA, 94110, USA
| | - Ma Somsouk
- Division of Gastroenterology at ZSFG, University of California, San Francisco, San Francisco, CA, 94110, USA
| | - De'Broski R Herbert
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, 19140, USA. .,Division of Experimental Medicine, University of California, San Francisco, San Francisco, CA, 94110, USA.
| |
Collapse
|
13
|
Hormdee D, Prajaneh S, Kampichai A, Tak R, Chaiyarit P. Prolonged Suppressive Effects of Periodontitis on Salivary TFF3 Production. Eur J Dent 2019; 13:193-198. [PMID: 31466117 PMCID: PMC6777149 DOI: 10.1055/s-0039-1693949] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Objective
As a follow-up to our previous study that demonstrated decreased salivary trefoil factor family 3 (TFF3) peptide levels in chronic periodontitis patients, this current study aimed to observe the effects of nonsurgical periodontal treatment on salivary TFF3 peptides in patients with periodontal diseases.
Materials and Methods
Eighty-seven volunteers that comprised of 30 individuals with healthy periodontium, 31 with gingivitis, and 26 with chronic periodontitis were considered for the study. Prior to periodontal treatment, a general periodontal examination was performed along with collection of saliva samples from each volunteer. Nonsurgical periodontal treatments were provided to patients with gingivitis and periodontitis. Two weeks post-treatment, saliva samples were recollected, and the periodontal status was re-evaluated. Salivary TFF3 concentrations were measured by enzyme-linked immunosorbent assay.
Statistical Analysis
Mann–Whitney U test was used when the investigated data were not normally distributed. Chi-squared test was used when dealing with categorical data. Kruskal–Wallis test with post-hoc corrections was used to compare data among the three investigated groups. Two-tailed
p
< 0.05 was considered as statistically significant.
Results
Prior to the periodontal treatment, salivary TFF3 concentrations in patients with gingivitis and periodontitis were significantly lower than those with healthy periodontium. Two weeks post-treatment, increased levels of salivary TFF3 were observed in patients with gingivitis, whereas the concentrations decreased in patients with chronic periodontitis.
Conclusion
This study demonstrated the effects of periodontal disease on the production of salivary TFF3 peptides. Interestingly, nonsurgical periodontal treatment also affected the recovery of salivary TFF3 peptides but varied in their outcomes between gingivitis and periodontitis patients.
Collapse
Affiliation(s)
- Doosadee Hormdee
- Division of Periodontology, Department of Oral Biomedical Sciences, Faculty of Dentistry, Khon Kaen University, Khon Kaen, Thailand.,Research Group of Chronic Inflammatory Oral Diseases and Systemic Diseases Associated with Oral Health, Khon Kaen University, Thailand
| | - Saengsome Prajaneh
- Division of Periodontology, Department of Oral Biomedical Sciences, Faculty of Dentistry, Khon Kaen University, Khon Kaen, Thailand
| | | | - Ranuch Tak
- Faculty of Dentistry, International University, Phnom Penh, Cambodia
| | - Ponlatham Chaiyarit
- Research Group of Chronic Inflammatory Oral Diseases and Systemic Diseases Associated with Oral Health, Khon Kaen University, Thailand.,Division of Oral Diagnosis, Department of Oral Biomedical Sciences, Faculty of Dentistry, Khon Kaen University, Khon Kaen, Thailand
| |
Collapse
|
14
|
Bian X, Liu X, Liu J, Zhao Y, Li H, Zhang L, Li P, Gao Y. Hepatoprotective effect of chiisanoside from Acanthopanax sessiliflorus against LPS/D-GalN-induced acute liver injury by inhibiting NF-κB and activating Nrf2/HO-1 signaling pathways. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2019; 99:3283-3290. [PMID: 30552777 DOI: 10.1002/jsfa.9541] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 11/12/2018] [Accepted: 12/11/2018] [Indexed: 06/09/2023]
Abstract
BACKGROUND In China, Acanthopanax sessiliflorus is a delicious wild vegetable. It is also used to treat inflammation and pain. Chiisanoside (CSS) is the main constituent of the leaf of A. sessiliflorus. Combined use of lipopolysaccharide and d-galactosamine (LPS/D-GalN) can induce acute liver failure in human beings, and there are no reports on the protective effect of CSS against LPS/D-GalN-induced acute liver injury in mice. RESULTS Chiisanoside pretreatment evidently reduced the activities of alanine transaminase (ALT) and aspartate transaminase (AST) in the changes induced by LPS/D-GalN, and these histopathological changes induced by LPS/GalN were significantly weakened. Catalase (CAT), glutathione (GSH), and superoxide dismutase (SOD) activities increased, and malondialdehyde (MDA) activity decreased after CSS treatment compared with LPS/D-GalN treatment. Pretreatment with CSS also inhibited the expression levels of inflammatory factors. The administration of CSS prevented the phosphorylated expression of inhibitor kappa B (IκB) kinase, and led to a significant increase in heme oxygenase-1 (HO-1) expression and nuclear factor erythroid 2-related factor2 (Nrf2) nuclear translocation. CONCLUSION The protective effects of CSS are attributed to its antioxidative effect and inflammatory suppression in Nuclear factor kappa beta (NF-κB) and Nrf2/HO-1 signaling pathways. Chiisanoside might therefore be a potential ingredient for drug and food development against acute liver injury in the future. © 2018 Society of Chemical Industry.
Collapse
Affiliation(s)
- Xingbo Bian
- College of Chinese Medicinal Materials, Jilin Agriculture University, Changchun, China
| | | | | | - Yan Zhao
- College of Chinese Medicinal Materials, Jilin Agriculture University, Changchun, China
| | | | - Lianxue Zhang
- College of Chinese Medicinal Materials, Jilin Agriculture University, Changchun, China
| | | | - Yugang Gao
- College of Chinese Medicinal Materials, Jilin Agriculture University, Changchun, China
| |
Collapse
|
15
|
Protective Effects of Different Kinds of Filtered Water on Hypertensive Mouse by Suppressing Oxidative Stress and Inflammation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:2917387. [PMID: 30622665 PMCID: PMC6304849 DOI: 10.1155/2018/2917387] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 09/29/2018] [Accepted: 10/21/2018] [Indexed: 11/23/2022]
Abstract
Oxidative stress and inflammation play an important role in hypertensive animals and patients. Hydrogen plays a role of antioxidation and anti-inflammation. Calcium and magnesium play an important role in reducing hypertension and antioxidant. Filtered water contains abundant hydrogen and a large number of other essential elements of the human body. We investigated the protective effects of filtered water on hypertensive mice. To establish hypertension model, ICR mice were administered with N′-nitro-L-arginine methyl ester (L-NAME) hydrochloride 64 mg/kg per day for 1 month. The hypertensive mice were, respectively, administered with pure water, tap water, and filtered water for 2 months. Lipid peroxidation, antioxidant enzymatic activity, endothelin-1 (ET-1), angiotensin II (Ang II), and proinflammatory cytokines (TNF-α, IL-6, and IL-1β) were assessed. Expressions of phosphorylated NF-κB P65 in the kidney were analyzed by western blot. qRT-PCR analysis was adopted to determine the expression levels of the proinflammatory cytokines and NF-κB P65. The results demonstrated that filtered water can reduce the blood pressure. Filtered water treatment restored the activity of antioxidant enzymes, downregulated ET-1, and Ang II in the serum of mice. Filtered water treatment suppressed proinflammatory cytokines and decreased the mRNA expression of TNF-α, IL-6, IL-1β, and NF-κB P65. Consumption of filtered water inhibited the expression of NF-κB P65. This suggests that filtered water can reduce the blood pressure. The protection mechanisms include downregulating oxidative stress and inhibiting inflammation, which is partly due to the inhibition of the NF-κB signaling pathway.
Collapse
|
16
|
Tanaka K, Sugiyama H, Yamanari T, Mise K, Morinaga H, Kitagawa M, Onishi A, Ogawa-Akiyama A, Tanabe K, Eguchi J, Ohmoto Y, Shikata K, Wada J. Renal expression of trefoil factor 3 mRNA in association with tubulointerstitial fibrosis in IgA nephropathy. Nephrology (Carlton) 2018; 23:855-862. [PMID: 29987860 PMCID: PMC6174951 DOI: 10.1111/nep.13444] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/04/2018] [Indexed: 12/13/2022]
Abstract
Aim Trefoil factor 3 (TFF3) is a small peptide that is involved in mucosal protection. TFF3 is widely expressed in multiple tissues including kidney tissue. Previous studies have reported that the levels of urinary TFF3 are significantly increased in patients with chronic kidney disease. The aim of this study is to detect the TFF3 mRNA in kidney and elucidate the relationship between renal TFF3 mRNA and tubulointerstitial fibrosis in IgA nephropathy (IgAN). Methods We investigated the renal mRNA expression of TFF3 by real‐time PCR analysis in biopsy specimens from patients with IgAN, other glomerulonephritis (OGN) and minor glomerular abnormalities (MGA). We also determined the renal localization of TFF3 and the levels of urinary TFF3 by immunostaining and ELISA, respectively. Results The renal TFF3 mRNA expression was significantly associated with the urinary TFF3 secretion and the tubulointerstitial fibrosis score in the IgAN group alone. Immunostaining of the renal specimen of IgAN patients revealed that TFF3 is located in the renal tubular epithelial cells. The locations were almost the same as those that showed uromodulin positivity; specifically, the thick ascending limb (TAL) of the loop of Henle and the early portion of the distal tubule. The urinary TFF3 levels were positively correlated with the levels of urinary biomarkers of tubulointerstitial injury in such patients. Conclusion Renal TFF3 mRNA is associated with renal tubulointerstitial fibrosis in IgAN patients. The TFF3 located in the renal tubular epithelial cells may play a role in the progression of tubulointerstitial fibrosis in IgAN patients. Trefoil factor 3 is a peptide that, generally speaking, has protective functions in epithelial biology. This study reports that TFF3 is increased in IgA nephropathy and correlates with injury. Whether TFF3 is functionally a counter‐regulatory, protective factor or whether its overexpression denotes a pathogenic role remains an outstanding question.
Collapse
Affiliation(s)
- Keiko Tanaka
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Hitoshi Sugiyama
- Department of Human Resource Development of Dialysis Therapy for Kidney Disease, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Toshio Yamanari
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Koki Mise
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Hiroshi Morinaga
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Masashi Kitagawa
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Akifumi Onishi
- Department of Human Resource Development of Dialysis Therapy for Kidney Disease, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Ayu Ogawa-Akiyama
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Katsuyuki Tanabe
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Jun Eguchi
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | | | - Kenichi Shikata
- Center for Innovative Clinical Medicine, Okayama University Hospital, Okayama, Japan
| | - Jun Wada
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
17
|
Kordjazy N, Haj-Mirzaian A, Haj-Mirzaian A, Rohani MM, Gelfand EW, Rezaei N, Abdolghaffari AH. Role of toll-like receptors in inflammatory bowel disease. Pharmacol Res 2018; 129:204-215. [PMID: 29155256 DOI: 10.1016/j.phrs.2017.11.017] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 10/05/2017] [Accepted: 11/14/2017] [Indexed: 12/15/2022]
Abstract
Inflammatory bowel disease (IBD) is the chronic inflammation of the gastrointestinal tract. Recently, studies of the interplay between the adaptive and innate immune responses have provided a better understanding of the immunopathogenesis of inflammatory disorders such as IBD, as well as identification of novel targets for more potent interventions. Toll-like receptors (TLRs) are a class of proteins that play a significant role in the innate immune system and are involved in inflammatory processes. Activation of TLR signal transduction pathways lead to the induction of numerous genes that function in host defense, including those for inflammatory cytokines, chemokines, and antigen presenting molecules. It was proposed that TLR mutations and dysregulation are major contributing factors to the predisposition and susceptibility to IBD. Thus, modulating TLRs represent an innovative immunotherapeutic approach in IBD therapy. This article outlines the role of TLRs in IBD, focusing on both animal and human studies; the role of TLR-targeted agonists or antagonists as potential therapeutic agents in the different stages of the disease is discussed.
Collapse
Affiliation(s)
- Nastaran Kordjazy
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Arvin Haj-Mirzaian
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Shahid Beheshti Universtity of Medical Sciences, Tehran, Iran
| | - Arya Haj-Mirzaian
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Mojtaba Rohani
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Erwin W Gelfand
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, CO, USA
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Boston, MA, USA.
| | - Amir Hossein Abdolghaffari
- Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Tehran, Iran; Department of Pharmacology, Pharmaceutical Sciences Branch, Islamic Azad University, Tehran, Iran; Department of Toxicology and Pharmacology, Faculty of Pharmacy and Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences, Tehran, Iran; Gastrointestinal Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| |
Collapse
|
18
|
Allaire JM, Morampudi V, Crowley SM, Stahl M, Yu H, Bhullar K, Knodler LA, Bressler B, Jacobson K, Vallance BA. Frontline defenders: goblet cell mediators dictate host-microbe interactions in the intestinal tract during health and disease. Am J Physiol Gastrointest Liver Physiol 2018; 314:G360-G377. [PMID: 29122749 PMCID: PMC5899238 DOI: 10.1152/ajpgi.00181.2017] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Goblet cells (GCs) are the predominant secretory epithelial cells lining the luminal surface of the mammalian gastrointestinal (GI) tract. Best known for their apical release of mucin 2 (Muc2), which is critical for the formation of the intestinal mucus barrier, GCs have often been overlooked for their active contributions to intestinal protection and host defense. In part, this oversight reflects the limited tools available to study their function but also because GCs have long been viewed as relatively passive players in promoting intestinal homeostasis and host defense. In light of recent studies, this perspective has shifted, as current evidence suggests that Muc2 as well as other GC mediators are actively released into the lumen to defend the host when the GI tract is challenged by noxious stimuli. The ability of GCs to sense and respond to danger signals, such as bacterial pathogens, has recently been linked to inflammasome signaling, potentially intrinsic to the GCs themselves. Moreover, further work suggests that GCs release Muc2, as well as other mediators, to modulate the composition of the gut microbiome, leading to both the expansion as well as the depletion of specific gut microbes. This review will focus on the mechanisms by which GCs actively defend the host from noxious stimuli, as well as describe advanced technologies and new approaches by which their responses can be addressed. Taken together, we will highlight current insights into this understudied, yet critical, aspect of intestinal mucosal protection and its role in promoting gut defense and homeostasis.
Collapse
Affiliation(s)
- Joannie M. Allaire
- 1Division of Gastroenterology, Department of Pediatrics, BC Children’s Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Vijay Morampudi
- 1Division of Gastroenterology, Department of Pediatrics, BC Children’s Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Shauna M. Crowley
- 1Division of Gastroenterology, Department of Pediatrics, BC Children’s Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Martin Stahl
- 1Division of Gastroenterology, Department of Pediatrics, BC Children’s Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Hongbing Yu
- 1Division of Gastroenterology, Department of Pediatrics, BC Children’s Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kirandeep Bhullar
- 1Division of Gastroenterology, Department of Pediatrics, BC Children’s Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Leigh A. Knodler
- 2Paul G. Allen School for Global Animal Health, College of Veterinary Medicine, Washington State University, Pullman, Washington
| | - Brian Bressler
- 3Division of Gastroenterology, Department of Medicine, St. Paul’s Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kevan Jacobson
- 1Division of Gastroenterology, Department of Pediatrics, BC Children’s Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Bruce A. Vallance
- 1Division of Gastroenterology, Department of Pediatrics, BC Children’s Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
19
|
Xie H, Guo JH, An WM, Tian ST, Yu HP, Yang XL, Wang HM, Guo Z. Diagnostic value evaluation of trefoil factors family 3 for the early detection of colorectal cancer. World J Gastroenterol 2017; 23:2159-2167. [PMID: 28405143 PMCID: PMC5374127 DOI: 10.3748/wjg.v23.i12.2159] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 03/10/2017] [Accepted: 03/17/2017] [Indexed: 02/06/2023] Open
Abstract
AIM The purpose of this study was to evaluate the diagnostic value of trefoil factor family 3 (TFF3) for the early detection of colorectal cancer (CC). METHODS Serum TFF3 and carcino-embryonic antigen (CEA) were detected in 527 individuals, including 115 healthy control (HC), 198 colorectal adenoma (CA), and 214 CC individuals in the training group. RESULTS Serum TFF3 showed no significant correlation with age, gender, or tumor location but showed significant correlation with the tumor stage. Serum TFF3 in the CC group was significantly higher than in the HC or CA group. The AUC values of TFF3 for discriminating between HC and CC and between CA and CC were 0.930 (0.903, 0.958) and 0.834 (0.796, 0.873). A multivariate model combining TFF3 and CEA was built. Compared to TFF3 or CEA alone, the multivariate model showed significant improvement (P < 0.001). For discriminating between HC and CC, HC and early stage CC, HC and advanced stage CC, CA and CC, CA and early stage CC, and CA and advanced stage CC in the training group, the sensitivities were 92.99%, 91.46%, 93.18%, 73.83%, 76.83%, and 81.82%, and the specificities were 91.30%, 91.30%, 93.91%, 88.38%, 77.27%, and 88.38%, respectively. After validation, the sensitivities were 89.39%, 85.71%, 90.79%, 72.73%, 71.43%, and 78.95%, and the specificities were 87.85%, 87.85%, 2.52%, 87.85%, 80.77%, and 87.50%, respectively. CONCLUSION The multivariate diagnostic model that included TFF3 and CEA showed significant improvement over the conventional biomarker CEA and might provide a potential method for the early detection of CC.
Collapse
|
20
|
Hanisch C, Sharbati J, Kutz-Lohroff B, Huber O, Einspanier R, Sharbati S. TFF3-dependent resistance of human colorectal adenocarcinoma cells HT-29/B6 to apoptosis is mediated by miR-491-5p regulation of lncRNA PRINS. Cell Death Discov 2017; 3:16106. [PMID: 28149533 PMCID: PMC5279457 DOI: 10.1038/cddiscovery.2016.106] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 12/17/2016] [Indexed: 12/21/2022] Open
Abstract
Tumour necrosis factor-α (TNF-α) is a double-edged cytokine associated with pathogenesis of inflammatory-related cancers being also able to induce cancer cell death. In the process of tumour development or metastasis, cancer cells can become resistant to TNF-α. In trefoil factor 3 (TFF3) overexpressing colorectal adenocarcinoma cells (HT-29/B6), we observed enhanced resistance against TNF-α/interferon gamma-induced apoptosis. TFF3 is a secreted small peptide that supports intestinal tissue repair but is also involved in intestinal tumour progression and scattering. We hypothesised that TFF3 rescues intestinal epithelial cancer cells from TNF-α-induced apoptosis by involving regulatory RNA networks. In silico-based expression analysis revealed TFF3-mediated regulation of selected microRNAs as well as long non-coding RNAs (lncRNAs), whereas miR-491-5p was identified to target the lncRNA ‘psoriasis susceptibility-related RNA gene induced by stress’ (PRINS). RNA interference-based gain- and loss-of-function experiments examined miR-491-PRINS axis to exert the TFF3-mediated phenotype. Chemical inhibition of selected pathways showed that phosphatidylinositol 3-kinase/AKT accounts for TFF3-mediated downregulation of miR-491-5p and accumulation of PRINS. Moreover, we showed that PRINS colocalises with PMAIP1 (NOXA) in nuclei of HT-29/B6 possessing inhibitory effects. Immunoprecipitation experiments proved molecular interaction of PMAIP1 with PRINS. Our study provides an insight into RNA regulatory networks that determine resistance of colorectal cancer cells to apoptosis.
Collapse
Affiliation(s)
- Carlos Hanisch
- Department of Veterinary Medicine, Institute of Veterinary Biochemistry, Freie Universität Berlin , Berlin, Germany
| | - Jutta Sharbati
- Department of Veterinary Medicine, Institute of Veterinary Biochemistry, Freie Universität Berlin, Berlin, Germany; Lise Meitner School of Science, Berlin, Germany
| | - Barbara Kutz-Lohroff
- Department of Veterinary Medicine, Institute of Veterinary Biochemistry, Freie Universität Berlin , Berlin, Germany
| | - Otmar Huber
- Institute of Biochemistry II, Jena University Hospital , Jena, Germany
| | - Ralf Einspanier
- Department of Veterinary Medicine, Institute of Veterinary Biochemistry, Freie Universität Berlin , Berlin, Germany
| | - Soroush Sharbati
- Department of Veterinary Medicine, Institute of Veterinary Biochemistry, Freie Universität Berlin , Berlin, Germany
| |
Collapse
|
21
|
Abstract
Trefoil factor (TFF) peptides, with a 40-amino acid motif and including six conserved cysteine residues that form intramolecular disulfide bonds, are a family of mucin-associated secretory molecules mediating many physiological roles that maintain and restore gastrointestinal (GI) mucosal homeostasis. TFF peptides play important roles in response to GI mucosal injury and inflammation. In response to acute GI mucosal injury, TFF peptides accelerate cell migration to seal the damaged area from luminal contents, whereas chronic inflammation leads to increased TFF expression to prevent further progression of disease. Although much evidence supports the physiological significance of TFF peptides in mucosal defenses, the molecular and cellular mechanisms of TFF peptides in the GI epithelium remain largely unknown. In this review, we summarize the functional roles of TFF1, 2, and 3 and illustrate their action mechanisms, focusing on defense mechanisms in the GI tract.
Collapse
Affiliation(s)
- Eitaro Aihara
- Department of Molecular and Cellular Physiology, University of Cincinnati, Cincinnati, Ohio 45267;
| | - Kristen A Engevik
- Department of Molecular and Cellular Physiology, University of Cincinnati, Cincinnati, Ohio 45267;
| | - Marshall H Montrose
- Department of Molecular and Cellular Physiology, University of Cincinnati, Cincinnati, Ohio 45267;
| |
Collapse
|
22
|
Zhang Z, Liu J, Shen P, Cao Y, Lu X, Gao X, Fu Y, Liu B, Zhang N. Zanthoxylum bungeanum pericarp extract prevents dextran sulfate sodium-induced experimental colitis in mice via the regulation of TLR4 and TLR4-related signaling pathways. Int Immunopharmacol 2016; 41:127-135. [DOI: 10.1016/j.intimp.2016.10.021] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 10/27/2016] [Accepted: 10/27/2016] [Indexed: 02/07/2023]
|
23
|
Inaba Y, Ueno N, Numata M, Zhu X, Messer JS, Boone DL, Fujiya M, Kohgo Y, Musch MW, Chang EB. Soluble bioactive microbial mediators regulate proteasomal degradation and autophagy to protect against inflammation-induced stress. Am J Physiol Gastrointest Liver Physiol 2016; 311:G634-G647. [PMID: 27514476 PMCID: PMC5142193 DOI: 10.1152/ajpgi.00092.2016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 08/07/2016] [Indexed: 01/31/2023]
Abstract
Bifidobacterium breve and other Gram-positive gut commensal microbes protect the gastrointestinal epithelium against inflammation-induced stress. However, the mechanisms whereby these bacteria accomplish this protection are poorly understood. In this study, we examined soluble factors derived from Bifidobacterium breve and their impact on the two major protein degradation systems within intestinal epithelial cells, proteasomes and autophagy. Conditioned media from gastrointestinal Gram-positive, but not Gram-negative, bacteria activated autophagy and increased expression of the autophagy proteins Atg5 and Atg7 along with the stress response protein heat shock protein 27. Specific examination of media conditioned by the Gram-positive bacterium Bifidobacterium breve (Bb-CM) showed that this microbe produces small molecules (<3 kDa) that increase expression of the autophagy proteins Atg5 and Atg7, activate autophagy, and inhibit proteasomal enzyme activity. Upregulation of autophagy by Bb-CM was mediated through MAP kinase signaling. In vitro studies using C2BBe1 cells silenced for Atg7 and in vivo studies using mice conditionally deficient in intestinal epithelial cell Atg7 showed that Bb-CM-induced cytoprotection is dependent on autophagy. Therefore, this work demonstrates that Gram-positive bacteria modify protein degradation programs within intestinal epithelial cells to promote their survival during stress. It also reveals the therapeutic potential of soluble molecules produced by these microbes for prevention and treatment of gastrointestinal disease.
Collapse
Affiliation(s)
- Yuhei Inaba
- Department of Medicine, Inflammatory Bowel Disease Research Center, The University of Chicago, Chicago, Illinois; Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Asahikawa, Japan
| | - Nobuhiro Ueno
- Department of Medicine, Inflammatory Bowel Disease Research Center, The University of Chicago, Chicago, Illinois; Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Asahikawa, Japan
| | - Masatsugu Numata
- Department of Medicine, Inflammatory Bowel Disease Research Center, The University of Chicago, Chicago, Illinois; Division of Life Style and Digestive Diseases, Kagoshima Medical University, Kagoshima, Japan
| | - Xiaorong Zhu
- Department of Medicine, Inflammatory Bowel Disease Research Center, The University of Chicago, Chicago, Illinois
| | - Jeannette S Messer
- Department of Medicine, Inflammatory Bowel Disease Research Center, The University of Chicago, Chicago, Illinois
| | - David L Boone
- Department of Microbiology and Immunology, Indiana University School of Medicine-South Bend, South Bend, Indiana
| | - Mikihiro Fujiya
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Asahikawa, Japan
| | - Yutaka Kohgo
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Asahikawa, Japan
| | - Mark W Musch
- Department of Medicine, Inflammatory Bowel Disease Research Center, The University of Chicago, Chicago, Illinois
| | - Eugene B Chang
- Department of Medicine, Inflammatory Bowel Disease Research Center, The University of Chicago, Chicago, Illinois;
| |
Collapse
|
24
|
Alpinetin attenuates inflammatory responses by suppressing TLR4 and NLRP3 signaling pathways in DSS-induced acute colitis. Sci Rep 2016; 6:28370. [PMID: 27321991 PMCID: PMC4913257 DOI: 10.1038/srep28370] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Accepted: 06/03/2016] [Indexed: 12/15/2022] Open
Abstract
Alpinetin, a composition of Alpinia katsumadai Hayata, has been reported to have a number of biological properties, such as antibacterial, antitumor and other important therapeutic activities. However, the effect of alpinetin on inflammatory bowel disease (IBD) has not yet been reported. The purpose of this study was to investigate the anti-inflammatory effect and mechanism of alpinetin on dextran sulfate sodium (DSS)-induced colitis in mice. In vivo, DSS-induced mice colitis model was established by giving mice drinking water containing 5% (w/v) DSS for 7 days. Alpinetin (25, 50 and 100 mg/kg) were administered once a day by intraperitoneal injection 3 days before DSS treatment. In vitro, phorbol myristate acetate (PMA)-differentiated monocytic THP-1 macrophages were treated with alpinetin and stimulated by lipopolysaccharide (LPS). The results showed that alpinetin significantly attenuated diarrhea, colonic shortening, histological injury, myeloperoxidase (MPO) activity and the expressions of tumor necrosis factor (TNF-α) and interleukin (IL-1β) production in mice. In vitro, alpinetin markedly inhibited LPS-induced TNF-α and IL-1β production, as well as Toll-like receptor 4 (TLR4) mediated nuclear transcription factor-kappaB (NF-κB) and NOD-like receptor protein 3 (NLRP3) inflammasome activation. In conclusion, this study demonstrated that alpinetin had protective effects on DSS-induced colitis and may be a promising therapeutic reagent for colitis treatment.
Collapse
|
25
|
Atorvastatin attenuates TNBS-induced rat colitis: the involvement of the TLR4/NF-kB signaling pathway. Inflammopharmacology 2016; 24:109-18. [PMID: 27038922 DOI: 10.1007/s10787-016-0263-6] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 03/18/2016] [Indexed: 12/13/2022]
Abstract
AIM The aim of the present study is to explore whether atorvastatin improves intestinal inflammation through the inhibition of the TLR4/NFkB signaling pathway in TNBS-induced rat colitis. METHODS Acute colitis was induced by intra-rectal administration of 100 mg/kg TNBS dissolved in 0.25 ml of 50 % ethanol. Twenty four hours after colitis induction, saline, atorvastatin (20 and 40 mg/kg) and sulfasalazine (100 mg/kg) were given to the animals by oral route. This was repeated daily for 1 week. Body weight changes, macroscopic and microscopic lesions were assessed. MPO and TNF-α activities were detected by immunohistochemistry (IHC) and the expression level of TLR4, MyD88 and NF-κB p65 proteins were measured by western blotting analysis. RESULTS Atorvastatin and sulfasalazine reduced the body weight loss, macroscopic and microscopic lesions. Additionally, both drugs decreased the expression of MPO and TNF-α positive cells in the colon tissue. Furthermore, they inhibited the TNBS-induced expression of TLR4, MyD88 and NF-κB p65 proteins. CONCLUSIONS It is suggested that the anti-inflammatory effect of atorvastatin on TNBS-induced rat colitis may involve the inhibition of the TLR4/NFkB signaling pathway.
Collapse
|
26
|
Busch M, Dünker N. Trefoil factor family peptides – friends or foes? Biomol Concepts 2015; 6:343-59. [DOI: 10.1515/bmc-2015-0020] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 08/20/2015] [Indexed: 12/13/2022] Open
Abstract
AbstractTrefoil factor family (TFF) peptides are a group of molecules bearing a characteristic three-loop trefoil domain. They are mainly secreted in mucous epithelia together with mucins but are also synthesized in the nervous system. For many years, TFF peptides were only known for their wound healing and protective function, e.g. in epithelial protection and restitution. However, experimental evidence has emerged supporting a pivotal role of TFF peptides in oncogenic transformation, tumorigenesis and metastasis. Deregulated expression of TFF peptides at the gene and protein level is obviously implicated in numerous cancers, and opposing functions as oncogenes and tumor suppressors have been described. With regard to the regulation of TFF expression, epigenetic mechanisms as well as the involvement of various miRNAs are new, promising aspects in the field of cancer research. This review will summarize current knowledge about the expression and regulation of TFF peptides and the involvement of TFF peptides in tumor biology and cancerogenesis.
Collapse
Affiliation(s)
- Maike Busch
- 1Medical Faculty, Institute for Anatomy II, Department of Neuroanatomy, University of Duisburg-Essen, Hufelandstr. 55, D-45122 Essen, Germany
| | - Nicole Dünker
- 1Medical Faculty, Institute for Anatomy II, Department of Neuroanatomy, University of Duisburg-Essen, Hufelandstr. 55, D-45122 Essen, Germany
| |
Collapse
|
27
|
Fu T, Znalesniak EB, Kalinski T, Möhle L, Biswas A, Salm F, Dunay IR, Hoffmann W. TFF Peptides Play a Role in the Immune Response Following Oral Infection of Mice with Toxoplasma Gondii. Eur J Microbiol Immunol (Bp) 2015; 5:221-31. [PMID: 26495133 PMCID: PMC4598890 DOI: 10.1556/1886.2015.00028] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 08/27/2015] [Indexed: 01/22/2023] Open
Abstract
The peptide trefoil factor family 3 (TFF3) is a major constituent of the intestinal mucus, playing an important role in the repair of epithelial surfaces. To further understand the role of TFF3 in the protection of intestinal epithelium, we tested the influence of TFF3 in a murine Toxoplasma gondii-induced ileitis model. Surprisingly, TFF3KO mice showed a reduced immune response in the ileum when compared to wild-type animals. Interleukin-12 and interferon-γ expression levels as well as the number of CD4+ lymphocytes were reduced in the infected TFF3KO mice. These effects were in line with the trend of elevated parasite levels in the ileum. Moreover, TFF1 expression was upregulated in the spleen of infected mice. These initial results indicate that TFF3 is involved in the immune pathology of T. gondii infection-induced intestinal inflammation. Thus far, the mechanisms of how TFF3 influences the immune response are not fully understood. Further studies should identify if TFF3 affects mucus sensing of dendritic cells and how TFF3 is involved in regulating the immune response as an intrinsic secretory peptide of immune cells.
Collapse
Affiliation(s)
- Ting Fu
- Institute of Molecular Biology and Medicinal Chemistry, Otto-von-Guericke University Magdeburg , Germany
| | - Eva B Znalesniak
- Institute of Molecular Biology and Medicinal Chemistry, Otto-von-Guericke University Magdeburg , Germany
| | - Thomas Kalinski
- Institute of Pathology, Otto-von-Guericke University Magdeburg , Germany
| | - Luisa Möhle
- Institute of Medical Microbiology and Hygiene, Otto-von-Guericke University Magdeburg , Germany
| | - Aindrila Biswas
- Institute of Medical Microbiology and Hygiene, Otto-von-Guericke University Magdeburg , Germany
| | - Franz Salm
- Institute of Molecular Biology and Medicinal Chemistry, Otto-von-Guericke University Magdeburg , Germany
| | - Ildiko Rita Dunay
- Institute of Medical Microbiology and Hygiene, Otto-von-Guericke University Magdeburg , Germany
| | - Werner Hoffmann
- Institute of Molecular Biology and Medicinal Chemistry, Otto-von-Guericke University Magdeburg , Germany
| |
Collapse
|
28
|
Liu SQ, Ma XL, Qin G, Liu Q, Li YC, Wu YH. Trans-system mechanisms against ischemic myocardial injury. Compr Physiol 2015; 5:167-92. [PMID: 25589268 DOI: 10.1002/cphy.c140026] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
A mammalian organism possesses a hierarchy of naturally evolved protective mechanisms against ischemic myocardial injury at the molecular, cellular, and organ levels. These mechanisms comprise regional protective processes, including upregulation and secretion of paracrine cell-survival factors, inflammation, angiogenesis, fibrosis, and resident stem cell-based cardiomyocyte regeneration. There are also interactive protective processes between the injured heart, circulation, and selected remote organs, defined as trans-system protective mechanisms, including upregulation and secretion of endocrine cell-survival factors from the liver and adipose tissue as well as mobilization of bone marrow, splenic, and hepatic cells to the injury site to mediate myocardial protection and repair. The injured heart and activated remote organs exploit molecular and cellular processes, including signal transduction, gene expression, cell proliferation, differentiation, migration, mobilization, and/or extracellular matrix production, to establish protective mechanisms. Both regional and trans-system cardioprotective mechanisms are mediated by paracrine and endocrine messengers and act in coordination and synergy to maximize the protective effect, minimize myocardial infarction, and improve myocardial function, ensuring the survival and timely repair of the injured heart. The concept of the trans-system protective mechanisms may be generalized to other organ systems-injury in one organ may initiate regional as well as trans-system protective responses, thereby minimizing injury and ensuring the survival of the entire organism. Selected trans-system processes may serve as core protective mechanisms that can be exploited by selected organs in injury. These naturally evolved protective mechanisms are the foundation for developing protective strategies for myocardial infarction and injury-induced disorders in other organ systems.
Collapse
Affiliation(s)
- Shu Q Liu
- Biomedical Engineering Department, Northwestern University, Evanston, Illinois Department of Emergency Medicine, Thomas Jefferson University Hospitals, Philadelphia, Pennsylvania Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois Carbohydrate and Lipid Metabolism Research Laboratory, College of Life Science and Technology, Dalian University, Dalian, China Department of Medicine, Division of Biological Sciences, The University of Chicago, Chicago, Illinois
| | | | | | | | | | | |
Collapse
|
29
|
Protective effect of oroxylin A against lipopolysaccharide and/or D-galactosamine-induced acute liver injury in mice. J Surg Res 2015; 195:522-8. [PMID: 25818981 DOI: 10.1016/j.jss.2015.01.047] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2014] [Revised: 01/13/2015] [Accepted: 01/26/2015] [Indexed: 11/21/2022]
Abstract
BACKGROUND Oroxylin A, a natural flavonoid isolated from Scutellariae baicalensis, has been reported to possess a wide spectrum of pharmacologic activities. However, the effects of oroxylin A on liver injury are poor understood. The purpose of this study was to investigate the effects of oroxylin A on acute liver injury in mice induced by lipopolysaccharide and/or D-galactosamine (LPS and/or D-GalN). METHODS Mice acute liver injury model was induced by LPS (50 μg/kg) and/or GalN (800 mg/kg). Serum alanine aminotransferase, aspartate aminotransferase, and tumor necrosis factor-α levels, hepatic tissue histology, malondialdehyde content, and myeloperoxidase activity were analyzed. Meanwhile, nuclear factor kappa B (NF-κB), heme oxygenase-1 (HO-1), and nuclear factor erythroid2-related factor 2 (Nrf2) expression were detected by Western blotting. RESULTS The results showed that oroxylin A dose-dependently inhibited LPS and/or GalN-induced serum alanine aminotransferase, aspartate aminotransferase, and tumor necrosis factor-α levels. Hepatic malondialdehyde content and myeloperoxidase activity were also suppressed by oroxylin A. We also found that oroxylin A inhibited LPS and/or GalN-induced toll like receptor 4 (TLR4) expression and NF-κB activation. In addition, oroxylin A upregulated the expression of Nrf2 and HO-1 in a dose-dependent manner. CONCLUSIONS In conclusion, oroxylin A protected against LPS and/or GalN-induced liver injury through activating Nrf2 and inhibiting TLR4 signaling pathway.
Collapse
|
30
|
Lin X, Liu ZF, Ding YH, Wang F, Pan HQ, Li J. Effect of IL-22 on expression of intestinal trefoil factor in intestinal epithelial cells. Shijie Huaren Xiaohua Zazhi 2015; 23:1290. [DOI: 10.11569/wcjd.v23.i8.1290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
31
|
The anti-inflammation effect of baicalin on experimental colitis through inhibiting TLR4/NF-κB pathway activation. Int Immunopharmacol 2014; 23:294-303. [PMID: 25239813 DOI: 10.1016/j.intimp.2014.09.005] [Citation(s) in RCA: 163] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 08/19/2014] [Accepted: 09/05/2014] [Indexed: 12/25/2022]
Abstract
BACKGROUND Baicalin holds a protective effect on inflammatory responses in several diseases. However, its molecular mechanism of anti-inflammatory activity on ulcerative colitis (UC) remains unknown. The present study was conducted to verify whether the anti-inflammation effect of baicalin on experimental colitis is via inhibiting TLR4/NF-κB pathway activation. METHODS The inflammatory response in RAW264.7 cells was induced by LPS and in rats by intrarectal administration of TNBS. Western blot analysis was carried out to examine toll-like receptor 4 (TLR4), NF-κB, p-NF-κB p65, IκB and p-IκB protein expressions in cells. Furthermore, intercellular adhesion molecule-1 (ICAM-1), monocyte chemoattractant protein-1 (MCP-1), cyclo-oxygenase-2 (Cox-2), tumor necrosis factor-α (TNF-α), interleukin (IL)-1β and IL-6 levels in cell supernatant and rat serum were detected by appropriate kits. An immunohistochemical assay was applied to examine TNF-α and IL-1β protein expression in colon tissues and TLR4 and p-NF-κB p65 protein expressions in RAW264.7 cells. RESULTS Baicalin ameliorates the considered inflammatory symptoms of induced colitis. It could also down-regulate pro-inflammatory mediators in the colon mucosa. The decline in the production of pro-inflammatory cytokines was correlated with the decrease in mucosal TLR4 protein expression. The expression of p-NF-κB p65 protein was significantly decreased, which correlated with a similar decrease in p-IκB protein. Consistent with the in vivo results, baicalin blocked LPS-stimulated nuclear translocation of p-NF-κB p65 in mouse macrophage RAW264.7 cells. CONCLUSIONS The present study indicates for the first time that the mechanism for baicalin on abrogating experimental colitis was targeted inhibition of the TLR4/NF-κB pathway activation.
Collapse
|
32
|
Li P, Turner JH. Chronic rhinosinusitis without nasal polyps is associated with increased expression of trefoil factor family peptides. Int Forum Allergy Rhinol 2014; 4:571-6. [DOI: 10.1002/alr.21334] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 02/08/2014] [Accepted: 03/15/2014] [Indexed: 01/10/2023]
Affiliation(s)
- Ping Li
- Department of Otolaryngology-Head and Neck Surgery; Vanderbilt University School of Medicine; Nashville TN
| | - Justin H. Turner
- Department of Otolaryngology-Head and Neck Surgery; Vanderbilt University School of Medicine; Nashville TN
| |
Collapse
|
33
|
Aamann L, Vestergaard EM, Grønbæk H. Trefoil factors in inflammatory bowel disease. World J Gastroenterol 2014; 20:3223-3230. [PMID: 24696606 PMCID: PMC3964394 DOI: 10.3748/wjg.v20.i12.3223] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Revised: 01/24/2014] [Accepted: 02/20/2014] [Indexed: 02/06/2023] Open
Abstract
Inflammatory bowel disease (IBD), which comprises ulcerative colitis and Crohn’s disease, is characterized by inflammation of the gastrointestinal tract. The trefoil factors 1, 2, and 3 (TFF1-3) are a family of peptides that play important roles in the protection and repair of epithelial surfaces, including the gastrointestinal tract. TFFs may be involved in IBD pathogenesis and are a potential treatment option. In the present review, we describe the TFF family and their potential role in IBD by summarizing the current knowledge of their expression, possible function and pharmacological role in IBD.
Collapse
|
34
|
Zhang J, Dou W, Zhang E, Sun A, Ding L, Wei X, Chou G, Mani S, Wang Z. Paeoniflorin abrogates DSS-induced colitis via a TLR4-dependent pathway. Am J Physiol Gastrointest Liver Physiol 2014; 306:G27-36. [PMID: 24232001 PMCID: PMC3920084 DOI: 10.1152/ajpgi.00465.2012] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Paeonia lactiflora Pall is one of the most well-known herbs in China, Korea, and Japan for more than 1,200 years. Paeoniflorin, the major bioactive component of peony root, has recently been reported to have anticolitic activity. However, the underlying molecular mechanism is unclear. The present study was to explore the possible mechanism of paeoniflorin in attenuating dextran sulfate sodium (DSS)-induced colitis. Pre- and coadministration of paeoniflorin significantly reduced the severity of colitis and resulted in downregulation of several inflammatory parameters in the colon, including the activity of myeloperoxidase (MPO), the levels of TNF-α and IL-6, and the mRNA expression of proinflammatory mediators (MCP-1, Cox2, IFN-γ, TNF-α, IL-6, and IL-17). The decline in the activation of NF-κB p65, ERK, JNK, and p38 MAPK correlated with a decrease in mucosal Toll-like receptor 4 (TLR4) but not TLR2 or TLR5 expression. In accordance with the in vivo results, paeoniflorin downregulated TLR4 expression, blocked nuclear translocation of NF-κB p65, and reduced the production of IL-6 in LPS-stimulated mouse macrophage RAW264.7 cells. Transient transfection assay performed in LPS-stimulated human colon cancer HT-29 cells indicated that paeoniflorin inhibits NF-κB transcriptional activity in a dose-dependent manner. TLR4 knockdown and overexpression experiments demonstrated a requirement for TLR4 in paeoniflorin-mediated downregulation of inflammatory cytokines. Thus, for the first time, the present study indicates that paeoniflorin abrogates DSS-induced colitis via decreasing the expression of TLR4 and suppressing the activation of NF-κB and MAPK pathways.
Collapse
Affiliation(s)
- Jingjing Zhang
- 1200 Cailun Rd., Rm. 5301, Shanghai Univ. of TCM, Shanghai 201203, China.
| | - Wei Dou
- 1Shanghai Key Laboratory of Complex Prescription and MOE Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China; ,3Departments of Medicine and Genetics, Albert Einstein College of Medicine, New York, New York; and
| | - Eryun Zhang
- 1Shanghai Key Laboratory of Complex Prescription and MOE Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China; ,2Department of Pharmacognosy, China Pharmaceutical University, Nanjing, China;
| | - Aning Sun
- 1Shanghai Key Laboratory of Complex Prescription and MOE Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China; ,2Department of Pharmacognosy, China Pharmaceutical University, Nanjing, China;
| | - Lili Ding
- 1Shanghai Key Laboratory of Complex Prescription and MOE Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China;
| | - Xiaohui Wei
- 1Shanghai Key Laboratory of Complex Prescription and MOE Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China;
| | - Guixin Chou
- 4Shanghai R&D Center for Standardization of Traditional Chinese Medicine, Shanghai, China
| | - Sridhar Mani
- 3Departments of Medicine and Genetics, Albert Einstein College of Medicine, New York, New York; and
| | - Zhengtao Wang
- 1Shanghai Key Laboratory of Complex Prescription and MOE Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China; ,2Department of Pharmacognosy, China Pharmaceutical University, Nanjing, China; ,4Shanghai R&D Center for Standardization of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
35
|
Biasi F, Leonarduzzi G, Oteiza PI, Poli G. Inflammatory bowel disease: mechanisms, redox considerations, and therapeutic targets. Antioxid Redox Signal 2013; 19:1711-47. [PMID: 23305298 PMCID: PMC3809610 DOI: 10.1089/ars.2012.4530] [Citation(s) in RCA: 215] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Oxidative stress is thought to play a key role in the development of intestinal damage in inflammatory bowel disease (IBD), because of its primary involvement in intestinal cells' aberrant immune and inflammatory responses to dietary antigens and to the commensal bacteria. During the active disease phase, activated leukocytes generate not only a wide spectrum of pro-inflammatory cytokines, but also excess oxidative reactions, which markedly alter the redox equilibrium within the gut mucosa, and maintain inflammation by inducing redox-sensitive signaling pathways and transcription factors. Moreover, several inflammatory molecules generate further oxidation products, leading to a self-sustaining and auto-amplifying vicious circle, which eventually impairs the gut barrier. The current treatment of IBD consists of long-term conventional anti-inflammatory therapy and often leads to drug refractoriness or intolerance, limiting patients' quality of life. Immune modulators or anti-tumor necrosis factor α antibodies have recently been used, but all carry the risk of significant side effects and a poor treatment response. Recent developments in molecular medicine point to the possibility of treating the oxidative stress associated with IBD, by designing a proper supplementation of specific lipids to induce local production of anti-inflammatory derivatives, as well as by developing biological therapies that target selective molecules (i.e., nuclear factor-κB, NADPH oxidase, prohibitins, or inflammasomes) involved in redox signaling. The clinical significance of oxidative stress in IBD is now becoming clear, and may soon lead to important new therapeutic options to lessen intestinal damage in this disease.
Collapse
Affiliation(s)
- Fiorella Biasi
- 1 Department of Clinical and Biological Sciences, University of Turin , San Luigi Gonzaga Hospital, Orbassano, Italy
| | | | | | | |
Collapse
|
36
|
Trefoil factor 3 as an endocrine neuroprotective factor from the liver in experimental cerebral ischemia/reperfusion injury. PLoS One 2013; 8:e77732. [PMID: 24204940 PMCID: PMC3799633 DOI: 10.1371/journal.pone.0077732] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Accepted: 09/08/2013] [Indexed: 01/30/2023] Open
Abstract
Cerebral ischemia, while causing neuronal injury, can activate innate neuroprotective mechanisms, minimizing neuronal death. In this report, we demonstrate that experimental cerebral ischemia/reperfusion injury in the mouse causes upregulation of the secretory protein trefoil factor 3 (TFF3) in the hepatocyte in association with an increase in serum TFF3. Partial hepatectomy (~60% liver resection) immediately following cerebral injury significantly lowered the serum level of TFF3, suggesting a contribution of the liver to the elevation of serum TFF3. Compared to wild-type mice, TFF3-/- mice exhibited a significantly higher activity of caspase 3 and level of cell death in the ischemic cerebral lesion, a larger fraction of cerebral infarcts, and a smaller fraction of the injured cerebral hemisphere, accompanied by severer forelimb motor deficits. Intravenous administration of recombinant TFF3 reversed changes in cerebral injury and forelimb motor function due to TFF3 deficiency. These observations suggest an endocrine neuroprotective mechanism involving TFF3 from the liver in experimental cerebral ischemia/reperfusion injury.
Collapse
|
37
|
Barrera GJ, Sanchez G, Gonzalez JE. Trefoil factor 3 isolated from human breast milk downregulates cytokines (IL8 and IL6) and promotes human beta defensin (hBD2 and hBD4) expression in intestinal epithelial cells HT-29. Bosn J Basic Med Sci 2013. [PMID: 23198942 DOI: 10.17305/bjbms.2012.2448] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Trefoil factors (TFF) are secretory products of mucin producing cells. They play a key role in the maintenance of the surface integrity of oral mucosa and enhance healing of the gastrointestinal mucosa by a process called restitution. TFF comprises the gastric peptides (TFF1), spasmolytic peptide (TFF2), and the intestinal trefoil factor (TFF3). They have an important and necessary role in epithelial restitution within the gastrointestinal tract. Significant amounts of TFF are present in human milk. This study aimed to determine a possible correlation between TFF3 isolated from human breast milk and levels of cytokines (IL8 and IL6) and defensins (hBD2 and hBD4) in intestinal epithelial cells HT-29 treated with trefoil. Samples of human milk were collected within 2-4 weeks postpartum from healthy human mothers (18-30-years-old) by manual breast massage, and TFF3 was purified by ammonium sulfate precipitation, isoelectric precipitation, DEAE-chromatography, and gel filtration. In this work we measured the concentrations and mRNA levels of cytokines and defensins by immunoassay (ELISA) and semiquantitative RT-PCR technique, respectively. Also we measured the peroxidase activity. We present the first evidence of human milk TFF3 purification. Here we show that the presence of TFF3 isolated from milk strongly correlates with downregulation of IL8 and IL6 in human intestinal epithelial cells. On the other hand, TFF3 activated the epithelial cells in culture to produce beta defensins 2 (hBD2) and beta defensins 4 (hBD4). These findings suggest that TFF can activate intestinal epithelial cells and could actively participate in the immune system of breastfed babies by inducing the production of peptides related to innate defence, such as defensins.
Collapse
Affiliation(s)
- Girolamo Jose Barrera
- Laboratorio de Biotecnologia Aplicada L.B.A., Av. Don Julio Centeno, CC metro Plaza, M17-M18, San Diego, Edo. Carabobo, Venezuela., Postal Number 2001.
| | | | | |
Collapse
|
38
|
Protective effect of naringenin against experimental colitis via suppression of Toll-like receptor 4/NF-κB signalling. Br J Nutr 2013; 110:599-608. [PMID: 23506745 DOI: 10.1017/s0007114512005594] [Citation(s) in RCA: 171] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Naringenin, one of the most abundant flavonoids in citrus, grapefruits and tomatoes, has been used as a traditional anti-inflammatory agent for centuries. However, the molecular mechanism of naringenin in intestinal inflammation remains unknown so far. The present study investigated a molecular basis for the protective effect of naringenin in dextran sulphate sodium-induced murine colitis. Pre-administration of naringenin significantly reduced the severity of colitis and resulted in down-regulation of pro-inflammatory mediators (inducible NO synthase (iNOS), intercellular adhesion molecule-1 (ICAM-1), monocyte chemoattractant protein-1 (MCP-1), cyclo-oxygenase-2 (Cox2), TNF-α and IL-6 mRNA) in the colon mucosa. The decline in the production of pro-inflammatory cytokines, specifically TNF-α and IL-6, correlated with a decrease in mucosal Toll-like receptor 4 (TLR4) mRNA and protein. Phospho-NF-κB p65 protein was significantly decreased, which correlated with a similar decrease in phospho-IκBα protein. Consistent with the in vivo results, naringenin exposure blocked lipopolysaccharide-stimulated nuclear translocation of NF-κB p65 in mouse macrophage RAW264.7 cells. In addition, in vitro NF-κB reporter assays performed on human colonic HT-29 cells exposed to naringenin demonstrated a significant inhibition of TNF-α-induced NF-κB luciferase expression. Thus, for the first time, the present study indicates that targeted inhibition of the TLR4/NF-κB signalling pathway might be an important mechanism for naringenin in abrogating experimental colitis.
Collapse
|
39
|
Dai SX, Zou Y, Feng YL, Liu HB, Zheng XB. Baicalin down-regulates the expression of macrophage migration inhibitory factor (MIF) effectively for rats with ulcerative colitis. Phytother Res 2011; 26:498-504. [PMID: 21887805 DOI: 10.1002/ptr.3581] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2011] [Revised: 05/08/2011] [Accepted: 05/11/2011] [Indexed: 12/17/2022]
Abstract
The purpose of this study was to investigate whether baicalin, a Chinese herbal extract, down-regulates the expression of macrophage migration inhibitory factor (MIF), an inflammatory factor that regulates the function of macrophages (MΦ), in rats with trinitrobenzene sulphonic acid (TNBS)-induced ulcerative colitis (UC). The results showed that baicalin simultaneously down-regulated the expression of MIF, the quantity of MΦs and the amount of MΦ-related cytokines, including macrophage chemotactic factor-1 (MCP-1, CCL2) and macrophage inflammatory protein-3α (MIP-3α, CCL20), in rats with UC. There was no statistical difference between baicailin and mesalazine in down-regulating the expression of MIF. Our study demonstrated that baicalin, an inexpensive but effective monomer, could be a new and alternative pharmaceutical for UC.
Collapse
Affiliation(s)
- Shi-Xue Dai
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | | | | | | | | |
Collapse
|
40
|
Shi H, Ren J, Xu H, Pan J, Hao X, Barlow LL, Dong W. Upregulated expression of hITF in Crohn's disease and screening of hITF Interactant by a yeast two-hybrid system. Dig Dis Sci 2010; 55:2929-2939. [PMID: 20499178 DOI: 10.1007/s10620-010-1227-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2009] [Accepted: 03/25/2010] [Indexed: 12/16/2022]
Abstract
AIMS To study the expression of human intestinal trefoil factor (hITF) mRNA in Crohn's disease and to screen the cellular proteins that can interact with the hITF protein by a yeast two-hybrid system in order to explore the mechanism of hITF in protecting intestinal mucosa from injury. METHODS Seventy-eight patients underwent double-balloon enteroscopy (DBE). Expression of hITF mRNA was detected by quantitative real-time polymerase chain reaction analysis (qRT-PCR). The hITF gene was amplified by PCR and cloned into vector pDEST32. The yeast cells cotransformed with pDEST32-hITF and the human jejunal cDNA library were plated in a selective SC-Leu-Trp-His-Ura medium. The subsequent screen was performed with χ-gal detection, and true-positive clones were sequenced and analyzed with bioinformatics. Co-immunoprecipitation (Co-IP) was performed to confirm the binding of putative proteins to the hITF protein. RESULTS Thirty-nine patients were diagnosed with Crohn's disease. We found that the expression of hITF mRNA is significantly increased in Crohn's disease compared to normal controls. A total of ten colonies were selected and sequenced. Among these, six colonies were Homo sapiens zinc finger protein 193 (ZNF193), three colonies were Homo sapiens Aldo-keto reductase family 1C 1 (AKR1C1), and one colony was of an unknown gene. A reverse two-hybrid experiment and Co-IP indicated that ZNF193 and AKR1C1 might interact with hITF. CONCLUSIONS The expression of hITF mRNA is increased in Crohn's disease. ZNF193 and AKR1C1 are proteins that can interact with the hITF protein by a yeast two-hybrid system and Co-IP, hITF may contribute to the mucosal repair through this interaction.
Collapse
Affiliation(s)
- Huaxiu Shi
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | | | | | | | | | | | | |
Collapse
|
41
|
Pimentel-Nunes P, Soares JB, Roncon-Albuquerque R, Dinis-Ribeiro M, Leite-Moreira AF. Toll-like receptors as therapeutic targets in gastrointestinal diseases. Expert Opin Ther Targets 2010; 14:347-68. [PMID: 20146632 DOI: 10.1517/14728221003642027] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
IMPORTANCE OF THE FIELD Toll-like receptors (TLRs) are innate immunity receptors that recognize several different antigens, initiating immunological/inflammatory responses. Recent evidence associates numerous pathophysiological processes and diseases with dysregulated activation of these receptors, conferring a potential therapeutic value to their modulation. AREAS COVERED IN THIS REVIEW The aim of this systematic review that covers literature from the past 10 years is to address the role of TLRs in the pathophysiology of gastrointestinal (GI) diseases as well as the therapeutic potential of modulating TLRs' signaling pathways in GI pathology. WHAT THE READER WILL GAIN This review shows that TLRs play an important role in the pathophysiology of several GI diseases and that modulating TLRs signaling pathways may have an enormous therapeutic potential. Different methods for modulation of TLRs' activity in GI tract, with direct agonists/antagonists but also with non-specific substances, like antibiotics or probiotics, are presented. TAKE HOME MESSAGE Even though TLRs modulators have been used for therapy in some GI diseases, further research, particularly in humans, is needed in order to establish the precise role of the different TLRs in the diverse GI diseases and to motivate clinical trials that consider TLRs as therapeutic targets in GI pathology.
Collapse
Affiliation(s)
- Pedro Pimentel-Nunes
- Department of Physiology, Cardiovascular Research & Development Unit, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319, Portugal.
| | | | | | | | | |
Collapse
|