1
|
Touny AA, Venkataraman B, Ojha S, Pessia M, Subramanian VS, Hariharagowdru SN, Subramanya SB. Phytochemical Compounds as Promising Therapeutics for Intestinal Fibrosis in Inflammatory Bowel Disease: A Critical Review. Nutrients 2024; 16:3633. [PMID: 39519465 PMCID: PMC11547603 DOI: 10.3390/nu16213633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 10/20/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND/OBJECTIVE Intestinal fibrosis, a prominent consequence of inflammatory bowel disease (IBD), presents considerable difficulty owing to the absence of licensed antifibrotic therapies. This review assesses the therapeutic potential of phytochemicals as alternate methods for controlling intestinal fibrosis. Phytochemicals, bioactive molecules originating from plants, exhibit potential antifibrotic, anti-inflammatory, and antioxidant activities, targeting pathways associated with inflammation and fibrosis. Compounds such as Asperuloside, Berberine, and olive phenols have demonstrated potential in preclinical models by regulating critical signaling pathways, including TGF-β/Smad and NFκB, which are integral to advancing fibrosis. RESULTS The main findings suggest that these phytochemicals significantly reduce fibrotic markers, collagen deposition, and inflammation in various experimental models of IBD. These phytochemicals may function as supplementary medicines to standard treatments, perhaps enhancing patient outcomes while mitigating the adverse effects of prolonged immunosuppressive usage. Nonetheless, additional clinical trials are necessary to validate their safety, effectiveness, and bioavailability in human subjects. CONCLUSIONS Therefore, investigating phytochemicals may lead to crucial advances in the formulation of innovative treatment approaches for fibrosis associated with IBD, offering a promising avenue for future therapeutic development.
Collapse
Affiliation(s)
- Aya A. Touny
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (A.A.T.); (B.V.); (M.P.); (S.N.H.)
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, Ahram Canadian University, Giza 12581, Egypt
| | - Balaji Venkataraman
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (A.A.T.); (B.V.); (M.P.); (S.N.H.)
| | - Shreesh Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates;
| | - Mauro Pessia
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (A.A.T.); (B.V.); (M.P.); (S.N.H.)
| | | | - Shamanth Neralagundi Hariharagowdru
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (A.A.T.); (B.V.); (M.P.); (S.N.H.)
- Zayed Bin Sultan Center for Health Sciences, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Sandeep B. Subramanya
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (A.A.T.); (B.V.); (M.P.); (S.N.H.)
- Zayed Bin Sultan Center for Health Sciences, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| |
Collapse
|
2
|
Pallares-Mendez R, Brassetti A, Bove AM, Simone G. Insights into the Interplay between the Urinary Microbiome and Bladder Cancer: A Comprehensive Review. J Clin Med 2024; 13:4927. [PMID: 39201069 PMCID: PMC11355659 DOI: 10.3390/jcm13164927] [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: 07/17/2024] [Revised: 08/11/2024] [Accepted: 08/18/2024] [Indexed: 09/02/2024] Open
Abstract
New insights in the urinary microbiome have led to a better understanding being built of the shifts in bacterial representations from health to disease; these hold promise as markers for diagnosis and therapeutic responses. Although several efforts have been made to identify a "core urinary microbiome", different fingerprints have been identified in men and women that shift with age. The main bacterial groups overall include Firmicutes, Actinobacteria, Fusobacteria, and Bacteroidetes. Although patients with bladder cancer have a microbiome that is similar to that of healthy individuals, differences have been observed at the species level with Fusobacterium nucleatum and Ralstonia, and at the genus level with Cutibacterium. Different bacterial representations may influence extracellular matrix composition, affecting tumor metastatic spreading and tumorigenic metalloproteinase expression. Furthermore, gene expression affecting targets of immune therapy, such as PD-L1, has been associated with changes in bacterial representations and therapeutic response to BCG. This comprehensive review aims to examine the influence of the urinary microbiome in bladder cancer.
Collapse
Affiliation(s)
| | - Aldo Brassetti
- Department of Urology, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi, 53, 00128 Rome, Italy; (R.P.-M.)
| | | | | |
Collapse
|
3
|
Watanabe D, Kamada N. Contribution of the Gut Microbiota to Intestinal Fibrosis in Crohn's Disease. Front Med (Lausanne) 2022; 9:826240. [PMID: 35198577 PMCID: PMC8859331 DOI: 10.3389/fmed.2022.826240] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/13/2022] [Indexed: 12/16/2022] Open
Abstract
In Crohn's disease (CD), intestinal fibrosis is a critical determinant of a patient's prognosis. Although inflammation may be a prerequisite for the initiation of intestinal fibrosis, research shows that the progression or continuation of intestinal fibrosis can occur independently of inflammation. Thus, once initiated, intestinal fibrosis may persist even if medical treatment controls inflammation. Clearly, an understanding of the pathophysiological mechanisms of intestinal fibrosis is required to diminish its occurrence. Accumulating evidence suggests that the gut microbiota contributes to the pathogenesis of intestinal fibrosis. For example, the presence of antibodies against gut microbes can predict which CD patients will have intestinal complications. In addition, microbial ligands can activate intestinal fibroblasts, thereby inducing the production of extracellular matrix. Moreover, in various animal models, bacterial infection can lead to the development of intestinal fibrosis. In this review, we summarize the current knowledge of the link between intestinal fibrosis in CD and the gut microbiota. We highlight basic science and clinical evidence that the gut microbiota can be causative for intestinal fibrosis in CD and provide valuable information about the animal models used to investigate intestinal fibrosis.
Collapse
Affiliation(s)
- Daisuke Watanabe
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Nobuhiko Kamada
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
- WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
| |
Collapse
|
4
|
Zhan S, Li N, Liu C, Mao R, Wu D, Li T, Chen M, Zhuang X, Zeng Z. Intestinal Fibrosis and Gut Microbiota: Clues From Other Organs. Front Microbiol 2021; 12:694967. [PMID: 34335525 PMCID: PMC8322786 DOI: 10.3389/fmicb.2021.694967] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 06/24/2021] [Indexed: 12/11/2022] Open
Abstract
Fibrosis is a complex and difficult to elucidate pathological process with no available therapies. Growing evidence implicates intestinal microbiota in the occurrence and development of fibrosis, and the potential mechanisms involved in different organs have been explored in several studies. In this review, we summarize the causative and preventive effects of gut microbiota on intestinal fibrosis, as well as the relationships between gut microbiota and fibrosis in other organs. Interestingly, several colonized microbes are associated with fibrosis via their structural components and metabolic products. They may also play essential roles in regulating inflammation and fibroblast activation or differentiation, which modulates extracellular matrix formation. While the relationships between intestinal fibrosis and gut microbiota remain unclear, lessons can be drawn from the effects of gut microbiota on hepatic, cardiac, nephritic, and pulmonary fibrosis. Various intestinal microbes alterations have been detected in different fibrotic organs; however, the results were heterogeneous. Mechanisms by which the intestinal microbiota regulate fibrotic processes in other organs, such as novel metabolic products or specific microbes, are also discussed. The specific microbiota associated with fibrosis in other organs could instruct future studies aiming to discover prospective mechanisms regulating intestinal fibrosis.
Collapse
Affiliation(s)
- Shukai Zhan
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Na Li
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Caiguang Liu
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ren Mao
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Dongxuan Wu
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Tong Li
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Minhu Chen
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaojun Zhuang
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhirong Zeng
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
5
|
Jenik K, Alkie TN, Moore E, Dejong JD, Lee LEJ, DeWitte-Orr SJ. Characterization of a bovine intestinal myofibroblast cell line and stimulation using phytoglycogen-based nanoparticles bound to inosine monophosphate. In Vitro Cell Dev Biol Anim 2021; 57:86-94. [PMID: 33474688 DOI: 10.1007/s11626-020-00536-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 11/24/2020] [Indexed: 12/19/2022]
Abstract
The goal of the present study was to characterize a novel bovine intestinal myofibroblast (BT-IMF) cell line isolated from a fetal bovine intestine. This cell type is of importance as intestinal myofibroblasts play a key role in controlling intestinal epithelial cell proliferation, intestinal regulation, wound healing, epithelial cell turnover, and structural support. The present work demonstrates that BT-IMF cells could be successfully cryopreserved and thawed and cultured past 25 passages. Immunocytochemical staining of the BT-IMF cell line was positive for vimentin and smooth muscle actin (α-SMA) and negative for pancytokeratin, suggesting that the cells are myofibroblastic in type. Growth kinetic experiments demonstrate that hydrocortisone negatively impacts BT-IMF growth and non-essential amino acids enhance its proliferation. Inosine monophosphate (IMP) is a dietary nucleotide and is essential for supporting animal health. Stimulation with IMP bound to a novel phytoglycogen-based nanocarrier (IMP-NP) showed enhanced cell proliferation. BT-IMF provides a new tool for studying bovine cells in vitro and may be of particular interest for cultured meat manufacturing in the future.
Collapse
Affiliation(s)
- K Jenik
- Department of Biology, Wilfrid Laurier University, Waterloo, ON, Canada
| | - T N Alkie
- Department of Biology, Wilfrid Laurier University, Waterloo, ON, Canada.,Department of Health Sciences, Wilfrid Laurier University, Waterloo, ON, Canada
| | - E Moore
- Glysantis Inc, Guelph, ON, Canada
| | - J D Dejong
- Department of Health Sciences, Wilfrid Laurier University, Waterloo, ON, Canada.,Glysantis Inc, Guelph, ON, Canada
| | - L E J Lee
- Faculty of Science, University of the Fraser Valley, Abbotsford, BC, Canada
| | - S J DeWitte-Orr
- Department of Biology, Wilfrid Laurier University, Waterloo, ON, Canada. .,Department of Health Sciences, Wilfrid Laurier University, Waterloo, ON, Canada.
| |
Collapse
|
6
|
Yersiniabactin-Producing Adherent/Invasive Escherichia coli Promotes Inflammation-Associated Fibrosis in Gnotobiotic Il10-/- Mice. Infect Immun 2019; 87:IAI.00587-19. [PMID: 31481410 PMCID: PMC6803345 DOI: 10.1128/iai.00587-19] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 08/22/2019] [Indexed: 12/16/2022] Open
Abstract
Fibrosis is a significant complication of intestinal disorders associated with microbial dysbiosis and pathobiont expansion, notably Crohn’s disease (CD). Mechanisms that favor fibrosis are not well understood, and therapeutic strategies are limited. Here we demonstrate that colitis-susceptible Il10-deficient mice develop inflammation-associated fibrosis when monoassociated with adherent/invasive Escherichia coli (AIEC) that harbors the yersiniabactin (Ybt) pathogenicity island. Fibrosis is a significant complication of intestinal disorders associated with microbial dysbiosis and pathobiont expansion, notably Crohn’s disease (CD). Mechanisms that favor fibrosis are not well understood, and therapeutic strategies are limited. Here we demonstrate that colitis-susceptible Il10-deficient mice develop inflammation-associated fibrosis when monoassociated with adherent/invasive Escherichia coli (AIEC) that harbors the yersiniabactin (Ybt) pathogenicity island. Inactivation of Ybt siderophore production in AIEC nearly abrogated fibrosis development in inflamed mice. In contrast, inactivation of Ybt import through its cognate receptor FyuA enhanced fibrosis severity. This corresponded with increased colonic expression of profibrogenic genes prior to the development of histological disease, therefore suggesting causality. fyuA-deficient AIEC also exhibited greater localization within subepithelial tissues and fibrotic lesions that was dependent on Ybt biosynthesis and corresponded with increased fibroblast activation in vitro. Together, these findings suggest that Ybt establishes a profibrotic environment in the host in the absence of binding to its cognate receptor and indicate a direct link between intestinal AIEC and the induction of inflammation-associated fibrosis.
Collapse
|
7
|
Lutz C, Weder B, Hünerwadel A, Fagagnini S, Lang B, Beerenwinkel N, Rossel JB, Rogler G, Misselwitz B, Hausmann M. Myeloid differentiation primary response gene (MyD) 88 signalling is not essential for intestinal fibrosis development. Sci Rep 2017; 7:17678. [PMID: 29247242 PMCID: PMC5732165 DOI: 10.1038/s41598-017-17755-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 11/29/2017] [Indexed: 01/15/2023] Open
Abstract
Dysregulation of the immune response to microbiota is associated with inflammatory bowel disease (IBD), which can trigger intestinal fibrosis. MyD88 is a key component of microbiota signalling but its influence on intestinal fibrosis has not been clarified. Small bowel resections from donor-mice were transplanted subcutaneously into the neck of recipients C57BL/6 B6-MyD88tm1 Aki (MyD88-/-) and C57BL/6-Tg(UBC-green fluorescence protein (GFP))30Scha/J (GFP-Tg). Grafts were explanted up to 21 days after transplantation. Collagen layer thickness was determined using Sirius Red stained slides. In the mouse model of fibrosis collagen deposition and transforming growth factor-beta 1 (TGF-β1) expression was equal in MyD88+/+ and MyD88-/-, indicating that MyD88 was not essential for fibrogenesis. Matrix metalloproteinase (Mmp)9 expression was significantly decreased in grafts transplanted into MyD88-/- recipients compared to MyD88+/+ recipients (0.2 ± 0.1 vs. 153.0 ± 23.1, respectively, p < 0.05), similarly recruitment of neutrophils was significantly reduced (16.3 ± 4.5 vs. 25.4 ± 3.1, respectively, p < 0.05). Development of intestinal fibrosis appears to be independent of MyD88 signalling indicating a minor role of bacterial wall compounds in the process which is in contrast to published concepts and theories. Development of fibrosis appears to be uncoupled from acute inflammation.
Collapse
Affiliation(s)
- C Lutz
- Department of Gastroenterology and Hepatology, University Hospital, Zurich, Switzerland
| | - B Weder
- Department of Gastroenterology and Hepatology, University Hospital, Zurich, Switzerland
| | - A Hünerwadel
- Department of Gastroenterology and Hepatology, University Hospital, Zurich, Switzerland
| | - S Fagagnini
- Department of Gastroenterology and Hepatology, University Hospital, Zurich, Switzerland
| | - B Lang
- Department of Biosystems Sciences and Engineering, ETH Zurich, Basel, Switzerland.,SIB Swiss Institute of Bioinformatics, Basel, Switzerland
| | - N Beerenwinkel
- Department of Biosystems Sciences and Engineering, ETH Zurich, Basel, Switzerland.,SIB Swiss Institute of Bioinformatics, Basel, Switzerland
| | - J B Rossel
- Institute of Social and Preventive Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - G Rogler
- Department of Gastroenterology and Hepatology, University Hospital, Zurich, Switzerland
| | - B Misselwitz
- Department of Gastroenterology and Hepatology, University Hospital, Zurich, Switzerland
| | - M Hausmann
- Department of Gastroenterology and Hepatology, University Hospital, Zurich, Switzerland.
| |
Collapse
|
8
|
Schmiedlin-Ren P, Reingold LJ, Broxson CS, Rittershaus AC, Brudi JS, Adler J, Owens SR, Zimmermann EM. Anti-TNFα alters the natural history of experimental Crohn's disease in rats when begun early, but not late, in disease. Am J Physiol Gastrointest Liver Physiol 2016; 311:G688-G698. [PMID: 27562059 PMCID: PMC5142192 DOI: 10.1152/ajpgi.00216.2015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 08/16/2016] [Indexed: 02/06/2023]
Abstract
Anti-TNFα therapy decreases inflammation in Crohn's disease (CD). However, its ability to decrease fibrosis and alter the natural history of CD is not established. Anti-TNF-α prevents inflammation and fibrosis in the peptidoglycan-polysaccharide (PG-PS) model of CD. Here we studied anti-TNF-α in a treatment paradigm. PG-PS or human serum albumin (HSA; control) was injected into bowel wall of anesthetized Lewis rats at laparotomy. Mouse anti-mouse TNF-α or vehicle treatment was begun day (d)1, d7, or d14 postlaparotomy. Rats were euthanized d21-23. Gross abdominal and histologic findings were scored. Cecal levels of relevant mRNAs were measured by quantitative real-time PCR. There was a stepwise loss of responsiveness when anti-TNFα was begun on d7 and d14 compared with d1 that was seen in the percent decrease in the median gross abdominal score and histologic inflammation score in PG-PS-injected rats [as %decrease; gross abdominal score: d1 = 75% (P = 0.003), d7 = 57% (P = 0.18), d14 = no change (P = 0.99); histologic inflammation: d1 = 57% (P = 0.006), d7 = 50% (P = 0.019), d14 = no change (P = 0.99)]. This was also reflected in changes in IL-1β, IL-6, TNF-α, IGF-I, TGF-β1, procollagen I, and procollagen III mRNAs that were decreased or trended downward in PG-PS-injected animals given anti-TNF-α beginning d1 or d7 compared with vehicle-treated rats; there was no effect if anti-TNF-α was begun d14. This change in responsiveness to anti-TNFα therapy was coincident with a major shift in the cytokine milieu observed on d14 in the PG-PS injected rats (vehicle treated). Our data are consistent with the clinical observation that improved outcomes occur when anti-TNF-α therapy is initiated early in the course of CD.
Collapse
Affiliation(s)
- Phyllissa Schmiedlin-Ren
- 1Division of Gastroenterology, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, Michigan;
| | - Laura J. Reingold
- 1Division of Gastroenterology, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, Michigan;
| | - Christopher S. Broxson
- 4Division of Gastroenterology, Hepatology and Nutrition, Department of Internal Medicine, University of Florida Health System, Gainesville, Florida
| | - Ahren C. Rittershaus
- 2Department of Pathology, University of Michigan Health System, Ann Arbor, Michigan;
| | - Josh S. Brudi
- 1Division of Gastroenterology, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, Michigan;
| | - Jeremy Adler
- 3Division of Gastroenterology, Department of Pediatrics and Communicable Diseases, University of Michigan Health System, Ann Arbor, Michigan; and
| | - Scott R. Owens
- 2Department of Pathology, University of Michigan Health System, Ann Arbor, Michigan;
| | - Ellen M. Zimmermann
- 1Division of Gastroenterology, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, Michigan; ,4Division of Gastroenterology, Hepatology and Nutrition, Department of Internal Medicine, University of Florida Health System, Gainesville, Florida
| |
Collapse
|
9
|
Alfano M, Canducci F, Nebuloni M, Clementi M, Montorsi F, Salonia A. The interplay of extracellular matrix and microbiome in urothelial bladder cancer. Nat Rev Urol 2016; 13:77-90. [PMID: 26666363 PMCID: PMC7097604 DOI: 10.1038/nrurol.2015.292] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Many pathological changes in solid tumours are caused by the accumulation of genetic mutations and epigenetic molecular alterations. In addition, tumour progression is profoundly influenced by the environment surrounding the transformed cells. The interplay between tumour cells and their microenvironment has been recognized as one of the key determinants of cancer development and is being extensively investigated. Data suggest that both the extracellular matrix and the microbiota represent microenvironments that contribute to the onset and progression of tumours. Through the introduction of omics technologies and pyrosequencing analyses, a detailed investigation of these two microenvironments is now possible. In urological research, assessment of their dysregulation has become increasingly important to provide diagnostic, prognostic and predictive biomarkers for urothelial bladder cancer. Understanding the roles of the extracellular matrix and microbiota, two key components of the urothelial mucosa, in the sequelae of pathogenic events that occur in the development and progression of urothelial carcinomas will be important to overcome the shortcomings in current bladder cancer treatment strategies.
Collapse
Affiliation(s)
- Massimo Alfano
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS Ospedale San Raffaele, Via Olgettina 60, 20132 Milan Italy
| | - Filippo Canducci
- Department of Biotechnology and Life Sciences, Università degli Studi dell'Insubria, Via Dunant 3, 21100 Varese Italy
| | - Manuela Nebuloni
- Department of Clinical Sciences, Pathology Unit, L. Sacco Hospital, Università degli Studi di Milano, Via Giovanni Battista Grassi 74, 20157 Milan Italy
| | - Massimo Clementi
- Università Vita-Salute San Raffaele, Via Olgettina 60, 20132 Milan Italy
| | - Francesco Montorsi
- Università Vita-Salute San Raffaele, Via Olgettina 60, 20132 Milan Italy
| | - Andrea Salonia
- Università Vita-Salute San Raffaele, Via Olgettina 60, 20132 Milan Italy
| |
Collapse
|
10
|
Grillo AR, Scarpa M, D'Incà R, Brun P, Scarpa M, Porzionato A, De Caro R, Martines D, Buda A, Angriman I, Palù G, Sturniolo GC, Castagliuolo I. TAK1 is a key modulator of the profibrogenic phenotype of human ileal myofibroblasts in Crohn's disease. Am J Physiol Gastrointest Liver Physiol 2015; 309:G443-54. [PMID: 26185333 DOI: 10.1152/ajpgi.00400.2014] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 07/06/2015] [Indexed: 01/31/2023]
Abstract
Transforming growth factor (TGF)-β-activated kinase 1 (TAK1) signaling can mediate inflammatory responses as well as tissue remodeling. Intestinal mucosal myofibroblast (IMF) activation drives gut fibrosis in Crohn's disease (CD); however, the molecular pathways involved are largely unknown. Thus we investigated the yet-unknown expression and function of TAK1 in human CD-associated fibrosis. Ileal surgical specimens, ileal biopsies, and IMF isolated from controls and CD patients were analyzed for TAK1 and its active phosphorylated form (pTAK1) by Western blotting, immunohistochemistry, and real-time quantitative PCR. TAK1 pharmacological inhibition and silencing were used to assess its role in collagen and inflammatory cytokine synthesis in IMF. TAK1 and pTAK1 levels increased in ileum specimens from CD patients compared with controls and correlated to tissue fibrosis. Similarly, TAK1 mRNA in ileal biopsies of CD patients correlated with fibrogenic marker expression but not inflammatory cytokines. CD-derived IMF showed higher TAK1 and pTAK1 expression associated with increased collagen1(α)1 mRNA levels compared with control IMF. TGF-β1 promoted pTAK1 nuclear translocation and collagen synthesis. TAK1 inhibition or silencing significantly reduced TGF-β1-stimulated collagen production and normalized the profibrogenic phenotype of CD-derived IMF. Taken together, these data suggest that TAK1 activation and nuclear translocation induce and maintain a fibrogenic phenotype in the IMF. Thus the TAK1 signaling pathway may represent a suitable target to design new, antifibrotic therapies.
Collapse
Affiliation(s)
- Alessia Rosaria Grillo
- Department of Molecular Medicine, University of Padova, Padova, Italy; Department of Surgery Oncology and Gastroenterology, University of Padova, Padova, Italy; and
| | - Melania Scarpa
- Oncological Surgery Unit, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
| | - Renata D'Incà
- Department of Surgery Oncology and Gastroenterology, University of Padova, Padova, Italy; and
| | - Paola Brun
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Marco Scarpa
- Oncological Surgery Unit, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
| | - Andrea Porzionato
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Raffaele De Caro
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Diego Martines
- Department of Surgery Oncology and Gastroenterology, University of Padova, Padova, Italy; and
| | - Andrea Buda
- Department of Surgery Oncology and Gastroenterology, University of Padova, Padova, Italy; and
| | - Imerio Angriman
- Department of Surgery Oncology and Gastroenterology, University of Padova, Padova, Italy; and
| | - Giorgio Palù
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Giacomo Carlo Sturniolo
- Department of Surgery Oncology and Gastroenterology, University of Padova, Padova, Italy; and
| | | |
Collapse
|
11
|
Role of intestinal myofibroblasts in HIV-associated intestinal collagen deposition and immune reconstitution following combination antiretroviral therapy. AIDS 2015; 29:877-88. [PMID: 25784439 DOI: 10.1097/qad.0000000000000636] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVE To investigate the potential role of mucosal intestinal myofibroblasts (IMFs) in HIV and associated fibrosis in gut-associated lymphoid tissue. DESIGN Profibrotic changes within the secondary lymphoid organs and mucosa have been implicated in failed immune reconstitution following effective combination antiretroviral therapy (cART). Microbial translocation is believed to be sustaining these systemic inflammatory pathways. IMFs are nonprofessional antigen-presenting cells with both immunoregulatory and mesenchymal functions that are ideally positioned to respond to translocating microbial antigen. METHODS Duodenal biopsies, obtained from patients naive to cART, underwent trichrome staining and were examined for tissue growth factor-beta (TGF-β) expression. Combined immunostaining and second harmonic generation analysis were used to determine IMF activation and collagen deposition. Confocal microscopy was performed to examine IMF activation and Toll-like receptor (TLR)4 expression. Finally, primary IMF cultures were stimulated with lipopolysaccharide to demonstrate the expression of the inflammatory biomarkers. RESULTS The expression of the fibrosis-promoting molecule, TGF-β1, is significantly increased in duodenal biopsies from HIV patients naïve to cART, and negatively correlated with subsequent peripheral CD4(+) recovery. The increase in TGF-β1 coincided with an increase in collagen deposition in the duodenal mucosa in the tissue area adjacent to the IMFs. We also observed that IMFs expressed TLR4 and had an activated phenotype since they were positive for fibroblast activation protein. Finally, stimulation of IMFs from HIV patients with TLR4 resulted in significantly increased expression of profibrotic molecules, TGF-β1, and interleukin-6. CONCLUSION Our data support the hypothesis that activated IMFs may be among the major cells contributing to the profibrotic changes, and thus, the establishment and maintenance of systemic inflammation interfering with immune reconstitution in HIV patients.
Collapse
|
12
|
Esser-von Bieren J, Volpe B, Sutherland DB, Bürgi J, Verbeek JS, Marsland BJ, Urban JF, Harris NL. Immune antibodies and helminth products drive CXCR2-dependent macrophage-myofibroblast crosstalk to promote intestinal repair. PLoS Pathog 2015; 11:e1004778. [PMID: 25806513 PMCID: PMC4373753 DOI: 10.1371/journal.ppat.1004778] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 03/03/2015] [Indexed: 12/12/2022] Open
Abstract
Helminth parasites can cause considerable damage when migrating through host tissues, thus making rapid tissue repair imperative to prevent bleeding and bacterial dissemination particularly during enteric infection. However, how protective type 2 responses targeted against these tissue-disruptive multicellular parasites might contribute to homeostatic wound healing in the intestine has remained unclear. Here, we observed that mice lacking antibodies (Aid-/-) or activating Fc receptors (Fcrg-/-) displayed impaired intestinal repair following infection with the murine helminth Heligmosomoides polygyrus bakeri (Hpb), whilst transfer of immune serum could partially restore chemokine production and rescue wound healing in Aid-/- mice. Impaired healing was associated with a reduced expression of CXCR2 ligands (CXCL2/3) by macrophages (MΦ) and myofibroblasts (MF) within intestinal lesions. Whilst antibodies and helminths together triggered CXCL2 production by MΦ in vitro via surface FcR engagement, chemokine secretion by intestinal MF was elicited by helminths directly via Fcrg-chain/dectin2 signaling. Blockade of CXCR2 during Hpb challenge infection reproduced the delayed wound repair observed in helminth infected Aid-/- and Fcrg-/- mice. Finally, conditioned media from human MΦ stimulated with infective larvae of the helminth Ascaris suum together with immune serum, promoted CXCR2-dependent scratch wound closure by human MF in vitro. Collectively our findings suggest that helminths and antibodies instruct a chemokine driven MΦ-MF crosstalk to promote intestinal repair, a capacity that may be harnessed in clinical settings of impaired wound healing. To complete their lifecycles, helminth parasites have to migrate through tissues such as the skin, lung, liver and intestine. This migration causes severe tissue damage, resulting in the need for rapid repair to restore the integrity and function of damaged tissues. Protective type 2 immune responses against helminths can repair acute lung damage, but they can also promote liver fibrosis. However, how protective immune mechanisms might contribute to wound healing during enteric nematode infection has remained unclear. Here we show that during a protective antibody response, where helminth larvae are trapped in the intestinal mucosa, macrophages and myofibroblasts secrete chemokines, which promote the repair of helminth-caused lesions. Chemokine secretion by macrophages was triggered by antibodies and helminth products, whilst myofibroblasts produced chemokines directly in response to innate recognition of helminth products. The same chemokines that instructed intestinal repair in mice were also secreted by human macrophages, when co-cultured with immune serum and helminths. Finally, human myofibroblasts closed in vitro scratch wounds more rapidly, when stimulated with the chemokine secretions of helminth-antibody activated human macrophages. Thus, our findings reveal a novel mechanism, by which a protective antibody response can promote the repair of intestinal injury during helminth infection.
Collapse
Affiliation(s)
- Julia Esser-von Bieren
- Laboratory of Intestinal Immunology, Global Health Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Center of Allergy and Environment (ZAUM), member of the German Center for Lung Research (DZL), Technische Universität and Helmholtz Center Munich, Munich, Germany
| | - Beatrice Volpe
- Laboratory of Intestinal Immunology, Global Health Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- * E-mail:
| | - Duncan B. Sutherland
- Laboratory of Intestinal Immunology, Global Health Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- * E-mail:
| | - Jérôme Bürgi
- Laboratory of Cell and Membrane Biology, Global Health Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - J. Sjef Verbeek
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Benjamin J. Marsland
- Faculty of Biology and Medicine, University of Lausanne, Service de Pneumologie, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Joseph F. Urban
- Diet, Genomics, & Immunology Laboratory, Beltsville Human Nutrition Research Center, Agricultural Research Service, United States Department of Agriculture, Beltsville, Maryland, United States of America
| | - Nicola L. Harris
- Laboratory of Intestinal Immunology, Global Health Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
13
|
Rieder F, Kessler S, Sans M, Fiocchi C. Animal models of intestinal fibrosis: new tools for the understanding of pathogenesis and therapy of human disease. Am J Physiol Gastrointest Liver Physiol 2012; 303:G786-801. [PMID: 22878121 PMCID: PMC4073977 DOI: 10.1152/ajpgi.00059.2012] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Fibrosis is a serious condition complicating chronic inflammatory processes affecting the intestinal tract. Advances in this field that rely on human studies have been slow and seriously restricted by practical and logistic reasons. As a consequence, well-characterized animal models of intestinal fibrosis have emerged as logical and essential systems to better define and understand the pathophysiology of fibrosis. In point of fact, animal models allow the execution of mechanistic studies as well as the implementation of clinical trials with novel, pathophysiology-based therapeutic approaches. This review provides an overview of the currently available animal models of intestinal fibrosis, taking into consideration the methods of induction, key characteristics of each model, and underlying mechanisms. Currently available models will be classified into seven categories: spontaneous, gene-targeted, chemical-, immune-, bacteria-, and radiation-induced as well as postoperative fibrosis. Each model will be discussed in regard to its potential to create research opportunities to gain insights into the mechanisms of intestinal fibrosis and stricture formation and assist in the development of effective and specific antifibrotic therapies.
Collapse
Affiliation(s)
- Florian Rieder
- 1Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio; ,2Department of Gastroenterology and Hepatology, Digestive Disease Institute, Cleveland Clinic, Cleveland, Ohio; and
| | - Sean Kessler
- 1Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio;
| | - Miquel Sans
- 3Service of Gastroenterology, Centro Medico Teknon, Barcelona, Spain
| | - Claudio Fiocchi
- 1Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio; ,2Department of Gastroenterology and Hepatology, Digestive Disease Institute, Cleveland Clinic, Cleveland, Ohio; and
| |
Collapse
|
14
|
Inoue K, Naito Y, Takagi T, Hayashi N, Hirai Y, Mizushima K, Horie R, Fukumoto K, Yamada S, Harusato A, Hirata I, Omatsu T, Yoshida N, Uchiyama K, Ishikawa T, Handa O, Konishi H, Wakabayashi N, Yagi N, Ichikawa H, Kokura S, Yoshikawa T. Daikenchuto, a Kampo medicine, regulates intestinal fibrosis associated with decreasing expression of heat shock protein 47 and collagen content in a rat colitis model. Biol Pharm Bull 2012; 34:1659-65. [PMID: 22040876 DOI: 10.1248/bpb.34.1659] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Heat shock protein (HSP) 47 may play an important role in the pathogenesis of intestinal fibrosis. Daikenchuto (DKT), a traditional Japanese herbal (Kampo) medicine, has been reported to ameliorate intestinal inflammation. The aims of this study were to determine time-course profiles of several parameters of fibrosis in a rat model, to confirm the HSP47-expressing cells in the colon, and finally to evaluate DKT's effects on intestinal fibrosis. Colitis was induced in male Wistar rats weighing 200 g using an enema of trinitrobenzene sulfonic acid (TNBS). HSP47 localization was determined by immunohistochemistry. Colonic inflammation and fibrosis were assessed by macroscopic, histological, morphometric, and immunohistochemical analyses. Colonic mRNA expression of transforming growth factor β1 (TGF-β1), HSP47, and collagen type I were assessed by real time-polymerase chain reaction (PCR). DKT was administered orally once a day from 8 to 14 d after TNBS administration. The colon was removed on the 15th day. HSP47 immunoreactivity was coexpressed with α-smooth muscle actin-positive cells located in the subepithelial space. Intracolonic administration of TNBS resulted in grossly visible ulcers. Colonic inflammation persisted for 6 weeks, and fibrosis persisted for 4 weeks after cessation of TNBS treatment. The expression levels of mRNA and proteins for TGF-β1, HSP47, and collagen I were elevated in colonic mucosa treated with TNBS. These fibrosis markers indicated that DKT treatment significantly inhibited TNBS-induced fibrosis. These findings suggest that DKT reduces intestinal fibrosis associated with decreasing expression of HSP47 and collagen content in the intestine.
Collapse
Affiliation(s)
- Ken Inoue
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Abstract
Wound healing is an appropriate response to inflammation and tissue injury in the gastrointestinal tract. If wound healing responses are excessive, perpetuated, or prolonged, they lead to fibrosis, distortion of tissue architecture, and loss of function. This introductory editorial and the minireviews or reviews in this themes series highlight the diversity in severity and location of fibrosis in response to gastrointestinal inflammation. The multiplicity of cellular and molecular mediators and new players, including stem cells or extracellular matrix-producing cells derived from nonmesenchymal cell types, is reviewed. Comparisons of inflammation-induced fibrosis across organ systems and the need for integrated and systems-based molecular approaches, new imaging modalities, well-characterized animal models, cell culture models, and improved diagnostic or predictive markers are reviewed. To date, intestinal fibrosis has received much less attention than inflammation in terms of defining mechanisms and underlying causes. This themes series aims to illustrate the importance of research in this area in gastrointestinal health and disease.
Collapse
Affiliation(s)
- Claudio Fiocchi
- 1Department of Gastroenterology and Hepatology, Digestive Disease Institute, Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio; and
| | - P. Kay Lund
- 2Department of Cell and Molecular Physiology, Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
16
|
Medina C, Santos-Martinez MJ, Santana A, Paz-Cabrera MC, Johnston MJ, Mourelle M, Salas A, Guarner F. Transforming growth factor-beta type 1 receptor (ALK5) and Smad proteins mediate TIMP-1 and collagen synthesis in experimental intestinal fibrosis. J Pathol 2011; 224:461-72. [PMID: 21465486 DOI: 10.1002/path.2870] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2010] [Revised: 12/15/2010] [Accepted: 01/31/2011] [Indexed: 12/16/2022]
Abstract
Transforming growth factor β (TGF-β) is known to play a key role in intestinal fibrosis; however, the underlying mechanisms are not well understood. TGF-β signal transduction is through TGF-β receptors, including the TGF-β type 1 receptor. Most cell types contain a TGF-β type 1 receptor form known as activin receptor-like kinase 5 (ALK5), which propagates the signal to the nucleus through the phosphorylation of Smad2 and Smad3 proteins. Therefore, we assessed the effect of the disruption of TGF-β/ALK5/Smad signalling by an ALK5 inhibitor (SD-208) in two experimental animal models of intestinal fibrosis: anaerobic bacteria- and trinitrobenzensulphonic acid-induced colitis. In addition, isolated myofibroblasts were pretreated with SD-208 and exposed to recombinant TGF-β1. Finally, myofibroblasts were transfected with ALK5, Smad2, and Smad3-specific siRNA. Up-regulation of ALK5 and TIMP-1, phosphorylation of Smad2 and Smad3 proteins, and increased intestinal wall collagen deposition were found in both experimental animal models. These effects were decreased by SD-208. TGF-β1 treatment also induced phosphorylation of Smad2 and Smad3 and up-regulation of ALK5 protein, TIMP-1, and α2 type 1 collagen gene expression in isolated myofibroblasts. Again these effects were inhibited by SD-208. Also, ALK5, Smad2, and Smad3 siRNA abolished the induction of TIMP-1 and α2 type 1 collagen. Our findings provide evidence that the TGF-β/ALK5/Smad pathway participates in the pathogenesis of experimental intestinal fibrosis. These data show promise for the development of an effective therapeutic intervention in this condition.
Collapse
Affiliation(s)
- Carlos Medina
- Department of Pharmacology, Trinity College Dublin, Dublin 2, Ireland.
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Chatterjee M, Saluja R, Tewari S, Barthwal MK, Goel SK, Dikshit M. Augmented nitric oxide generation in neutrophils: Oxidative and pro-inflammatory implications in hypertension. Free Radic Res 2009; 43:1195-204. [DOI: 10.3109/10715760903247256] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
18
|
The toll-like receptor signaling molecule Myd88 contributes to pancreatic beta-cell homeostasis in response to injury. PLoS One 2009; 4:e5063. [PMID: 19357791 PMCID: PMC2666970 DOI: 10.1371/journal.pone.0005063] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2008] [Accepted: 02/19/2009] [Indexed: 11/19/2022] Open
Abstract
Commensal flora and pathogenic microbes influence the incidence of diabetes in animal models yet little is known about the mechanistic basis of these interactions. We hypothesized that Myd88, an adaptor molecule in the Toll-like-receptor (TLR) pathway, regulates pancreatic beta-cell function and homeostasis. We first examined beta-cells histologically and found that Myd88-/- mice have smaller islets in comparison to C57Bl/6 controls. Myd88-/- mice were nonetheless normoglycemic both at rest and after an intra-peritoneal glucose tolerance test (IPGTT). In contrast, after low-dose streptozotocin (STZ) challenge, Myd88-/-mice had an abnormal IPGTT relative to WT controls. Furthermore, Myd88-/- mice suffer enhanced beta-cell apoptosis and have enhanced hepatic damage with delayed recovery upon low-dose STZ treatment. Finally, we treated WT mice with broad-spectrum oral antibiotics to deplete their commensal flora. In WT mice, low dose oral lipopolysaccharide, but not lipotichoic acid or antibiotics alone, strongly promoted enhanced glycemic control. These data suggest that Myd88 signaling and certain TLR ligands mediate a homeostatic effect on beta-cells primarily in the setting of injury.
Collapse
|
19
|
Walton KLW, Holt L, Sartor RB. Lipopolysaccharide activates innate immune responses in murine intestinal myofibroblasts through multiple signaling pathways. Am J Physiol Gastrointest Liver Physiol 2009; 296:G601-11. [PMID: 19136385 PMCID: PMC2660170 DOI: 10.1152/ajpgi.00022.2008] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Myofibroblasts (MF) play an important role in intestinal wound healing. A compromised epithelial barrier exposes intestinal subepithelial MF to luminal bacterial products. However, responses of murine intestinal MF to bacterial adjuvants and potential roles of intestinal MF in innate immune responses are not well defined. Our aims in this study were to determine innate immune responses and intracellular signaling pathways of intestinal MF exposed to LPS, a prototypic Toll-like receptor (TLR) ligand. Expression of TLR4 in primary murine intestinal MF cultures was confirmed by RT-PCR and Western blotting. LPS-induced secretion of prostaglandin E2 (PGE2), interleukin (IL)-6, and keratinocyte-derived chemokines (KC) was measured by ELISA. Intracellular responses to LPS were assessed by Western blotting for NF-kappaB p65, Ikappa-Balpha, Akt, p38 MAP kinase, and cyclooxygenase-2 (COX-2). LPS induced rapid phosphorylation of NF-kappaB p65, Akt, and p38 MAPK and degradation of Ikappa-Balpha. LPS induced expression of COX-2 and secretion of PGE2 (2.0+/-0.8-fold induction vs. unstimulated cells), IL-6 (6.6+/-0.4-fold induction), and KC (12.5+/-0.4-fold induction). Inhibition of phosphoinositide-3 (PI3)-kinase, p38 MAPK, or NF-kappaB pathways reduced LPS-induced PGE2, IL-6, and KC secretion. These studies show that primary murine intestinal MF respond to LPS, evidenced by activation of NF-kappaB, PI3-kinase, and MAPK signaling pathways and secretion of proinflammatory molecules. Inhibition of these pathways attenuated LPS-dependent PGE2, IL-6, and KC production, indicating that LPS activates MF by multiple signaling pathways. These data support the hypothesis that MF are a component of the innate immune system and may exert paracrine effects on adjacent epithelial and immune cells by responding to luminal bacterial adjuvants.
Collapse
Affiliation(s)
- Kristen L. W. Walton
- Department of Medicine and Center for Gastrointestinal Biology, University of North Carolina, Chapel Hill, North Carolina
| | - Lisa Holt
- Department of Medicine and Center for Gastrointestinal Biology, University of North Carolina, Chapel Hill, North Carolina
| | - R. Balfour Sartor
- Department of Medicine and Center for Gastrointestinal Biology, University of North Carolina, Chapel Hill, North Carolina
| |
Collapse
|
20
|
|
21
|
Simmons JG, Ling Y, Wilkins H, Fuller CR, D’Ercole AJ, Fagin J, Lund PK. Cell-specific effects of insulin receptor substrate-1 deficiency on normal and IGF-I-mediated colon growth. Am J Physiol Gastrointest Liver Physiol 2007; 293:G995-1003. [PMID: 17823215 PMCID: PMC2267759 DOI: 10.1152/ajpgi.00537.2006] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Insulin-like growth factor I (IGF-I) potently stimulates intestinal growth. Insulin receptor substrate-1 (IRS-1) mediates proliferative and antiapoptotic actions of IGF-I in cell lines, but its in vivo relevance in intestine is not defined. This study tested the hypothesis that there is cell type-specific dependence on IRS-1 as a mediator of IGF-I action. Length, mass, crypt cell proliferation, and apoptosis were measured in small intestine and colon of IRS-1-null mice and wild-type (WT) littermates and in colon of IRS-1-null or WT mice expressing IGF-I transgenes. Expression of IGF-I receptor and signaling intermediates was examined in intestine of WT and IRS-1-null mice, cultured intestinal epithelial cells, and myofibroblasts. Absolute IRS-1 deficiency reduced mucosal mass in jejunum and colon, but effects were more pronounced in colon. Muscularis mass was decreased in both segments. In IGF-I transgenics, IRS-1 deficiency significantly attenuated IGF-I-stimulated growth of colonic mucosa and abolished antiapoptotic but not mitogenic effects of IGF-I transgene on crypt cells. IGF-I-induced muscularis growth was unaffected by IRS-1 deficiency. In intestinal epithelial cells, IRS-1 was expressed at higher levels than IRS-2 and was preferentially activated by IGF-I. In contrast, IGF-I activated both IRS-1 and IRS-2 in intestinal myofibroblasts and IRS-2 activation was upregulated in IRS-1-null myofibroblasts. We conclude that the intestinal epithelium but not the muscularis requires IRS-1 for normal trophic actions of IGF-I and that IRS-1 is required for antiapoptotic but not mitogenic effects of IGF-I in the intestinal crypts in vivo.
Collapse
Affiliation(s)
- J. G. Simmons
- Department of Cell and Molecular Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Y. Ling
- Department of Cell and Molecular Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - H. Wilkins
- Department of Cell and Molecular Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - C. R. Fuller
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - A. J. D’Ercole
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - James Fagin
- Department of Medicine, University of Cincinnati, Cincinnati, Ohio
| | - P. K. Lund
- Department of Cell and Molecular Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
22
|
Wang Y, Wang L, Iordanov H, Swietlicki EA, Zheng Q, Jiang S, Tang Y, Levin MS, Rubin DC. Epimorphin(-/-) mice have increased intestinal growth, decreased susceptibility to dextran sodium sulfate colitis, and impaired spermatogenesis. J Clin Invest 2006; 116:1535-46. [PMID: 16710473 PMCID: PMC1462938 DOI: 10.1172/jci25442] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2005] [Accepted: 03/28/2006] [Indexed: 01/02/2023] Open
Abstract
Dynamic and reciprocal epithelial-mesenchymal interactions are critical for the normal morphogenesis and maintenance of epithelia. Epimorphin has been identified as a unique molecule expressed by mesenchymal cells and myofibroblasts and has putative morphogenetic effects in multiple epithelial tissues, including intestine, skin, mammary gland, lung, gallbladder, and liver. To define the in vivo role of epimorphin, we created epimorphin-null mice by targeted inactivation of the epimorphin gene. Male epimorphin-/- mice are sterile due to abnormal testicular development and impaired spermatogenesis. Intestinal growth is increased in epimorphin-/- mice due to augmented crypt cell proliferation and crypt fission during the neonatal (suckling) period, mediated at least in part by changes in bone morphogenetic protein (Bmp) and Wnt/beta-catenin signaling pathways. Colonic mucosal injury and colitis induced by dextran sodium sulfate (DSS) are ameliorated in epimorphin-/- mice, probably due to the increased proliferative capacity of the epimorphin-/- colon. These in vivo findings support the notion that epimorphin is a key stromal regulator of epithelial cell proliferation and growth in the intestine. In addition, our studies demonstrate a novel and critical role for epimorphin in regulating testicular development and growth as well as spermatogenesis.
Collapse
Affiliation(s)
- Yuan Wang
- Department of Medicine and
Speciality Care Service Line, St. Louis VA Medical Center, St. Louis, Missouri, USA.
Department of Molecular Biology and Pharmacology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Lihua Wang
- Department of Medicine and
Speciality Care Service Line, St. Louis VA Medical Center, St. Louis, Missouri, USA.
Department of Molecular Biology and Pharmacology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Hristo Iordanov
- Department of Medicine and
Speciality Care Service Line, St. Louis VA Medical Center, St. Louis, Missouri, USA.
Department of Molecular Biology and Pharmacology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Elzbieta A. Swietlicki
- Department of Medicine and
Speciality Care Service Line, St. Louis VA Medical Center, St. Louis, Missouri, USA.
Department of Molecular Biology and Pharmacology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Qun Zheng
- Department of Medicine and
Speciality Care Service Line, St. Louis VA Medical Center, St. Louis, Missouri, USA.
Department of Molecular Biology and Pharmacology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Shujun Jiang
- Department of Medicine and
Speciality Care Service Line, St. Louis VA Medical Center, St. Louis, Missouri, USA.
Department of Molecular Biology and Pharmacology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Yuzhu Tang
- Department of Medicine and
Speciality Care Service Line, St. Louis VA Medical Center, St. Louis, Missouri, USA.
Department of Molecular Biology and Pharmacology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Marc S. Levin
- Department of Medicine and
Speciality Care Service Line, St. Louis VA Medical Center, St. Louis, Missouri, USA.
Department of Molecular Biology and Pharmacology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Deborah C. Rubin
- Department of Medicine and
Speciality Care Service Line, St. Louis VA Medical Center, St. Louis, Missouri, USA.
Department of Molecular Biology and Pharmacology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
23
|
Adegboyega PA, Ololade O, Saada J, Mifflin R, Di Mari JF, Powell DW. Subepithelial myofibroblasts express cyclooxygenase-2 in colorectal tubular adenomas. Clin Cancer Res 2005; 10:5870-9. [PMID: 15355919 DOI: 10.1158/1078-0432.ccr-0431-03] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Recent data support the hypothesis that the inducible isoform of cyclooxygenase (COX-2) plays a role in the early stages of colonic carcinogenesis and that nonsteroidal anti-inflammatory drugs (NSAIDs) retard the development of colon cancer by modulating COX-2. However, the cell types responsible for producing COX-2 in colorectal adenomas remain a subject of controversy. EXPERIMENTAL DESIGN COX-2 expression in normal colonic mucosa (n = 50), hyperplastic polyps (n = 43), sporadic adenomas (n = 67), and invasive colonic adenocarcinoma (n = 39) was studied in formalin-fixed and paraffin-embedded tissue sections from endoscopy biopsy and colonic resection specimens. Immunohistochemistry (avidin-biotin complex technique with double immunolabeling) was used to identify the phenotypes of COX-2-producing cells. RESULTS In colorectal adenomas, increased expression of COX-2 was detected and localized to alpha smooth muscle actin ( proportional, variant SMA)-positive subepithelial stromal cells (myofibroblasts) in the periluminal region of the lamina propria in 63 (94%) of 67 cases. In contrast, in normal colonic mucosa and in hyperplastic polyps with intact epithelium, COX-2 expression was found only in macrophages and endothelial cells. In areas in which the surface epithelium was ulcerated in normal mucosa as well as hyperplastic or neoplastic polyps, COX-2 expression was increased in granulation tissue (and present in macrophages, endothelium, and myofibroblasts). In invasive carcinoma, COX-2 expression in myofibroblasts was limited to the adenomatous portion of the tumor and was detected in 62% of cases (n = 39). In addition, focal expression of COX-2 by malignant epithelial cells was observed in 23% of invasive adenocarcinoma. CONCLUSIONS These results show that increased COX-2 expression in sporadic adenoma of the colon is common and is localized specifically to subepithelial intestinal myofibroblasts. These findings further support the hypothesis that myofibroblasts are important target cells for NSAID-mediated chemoprevention of colorectal cancer.
Collapse
Affiliation(s)
- Patrick A Adegboyega
- Department of Pathology, 2190 John Sealy Annex, Mail Route 0588, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-0588, USA.
| | | | | | | | | | | |
Collapse
|
24
|
Dieleman LA, Hoentjen F, Qian BF, Sprengers D, Tjwa E, Torres MF, Torrice CD, Sartor RB, Tonkonogy SL. Reduced ratio of protective versus proinflammatory cytokine responses to commensal bacteria in HLA-B27 transgenic rats. Clin Exp Immunol 2004; 136:30-9. [PMID: 15030511 PMCID: PMC1808999 DOI: 10.1111/j.1365-2249.2004.02410.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Germ-free HLA-B27 transgenic (TG) rats do not develop colitis, but colonization with specific pathogen-free (SPF) bacteria induces colitis accompanied by immune activation. To study host-dependent immune responses to commensal caecal bacteria we investigated cytokine profiles in mesenteric lymph node (MLN) cells from HLA-B27 TG versus nontransgenic (non-TG) littermates after in vitro stimulation with caecal bacterial lysates (CBL). Supernatants from CBL-stimulated unseparated T- or B- cell-depleted MLN cells from HLA-B27 TG and non-TG littermates were analysed for IFN-gamma, IL-12, TNF, IL-10 and TGF-beta production. Our results show that unfractionated TG MLN cells stimulated with CBL produced more IFN-gamma, IL-12 and TNF than did non-TG MLN cells. In contrast, CBL-stimulated non-TG MLN cells produced more IL-10 and TGF-beta. T cell depletion abolished IFN-gamma and decreased IL-12 production, but did not affect IL-10 and TGF-beta production. Conversely, neither IL-10 nor TGF-beta was produced in cultures of B cell-depleted MLN. In addition, CD4(+) T cells enriched from MLN of HLA-B27 TG but not from non-TG rats produced IFN-gamma when cocultured with CBL-pulsed antigen presenting cells from non-TG rats. Interestingly, IL-10 and TGF-beta, but not IFN-gamma, IL-12 and TNF were produced by MLN cells from germ-free TG rats. These results indicate that the colitis that develops in SPF HLA-B27 TG rats is accompanied by activation of IFN-gamma-producing CD4(+) T cells that respond to commensal bacteria. However, B cell cytokine production in response to components of commensal intestinal microorganisms occurs in the absence of intestinal inflammation.
Collapse
Affiliation(s)
- L A Dieleman
- Center for Gastrointestinal Biology and Disease, University of North Carolina, Chapel Hill, NC, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Chitapanarux T, Chen SL, Lee H, Melton AC, Yee HF. C-type natriuretic peptide induces human colonic myofibroblast relaxation. Am J Physiol Gastrointest Liver Physiol 2004; 286:G31-6. [PMID: 12958024 DOI: 10.1152/ajpgi.00325.2003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Intestinal response to injury requires coordinated regulation of the tension exerted by subepithelial myofibroblasts (SEM). However, the signals governing relaxation of intestinal SEM have not been investigated. Our aim was to test the hypothesis that signal transduction pathways initiated by C-type natriuretic peptide (CNP) induce intestinal SEM relaxation. We directly quantified the effects of CNP on isometric tension exerted by cultured human colonic SEM. We also measured the effects of CNP on cGMP content, myosin regulatory light chain (MLC) phosphorylation, and cytosolic Ca2+ concentration. CNP induced relaxation of SEM within 10 s. By 10 min, relaxation reached a plateau that was sustained for 2 h. CNP-induced relaxation was saturable, with a maximal decrease in tension (51.7 +/- 3.8 dyn) observed at 250 nM. SEM relaxation in response to CNP constituted approximately 23% of total basal tension. CNP increased intracellular cGMP content and reduced MLC phosphorylation. Effects of CNP on cGMP and MLC exhibited the same dose dependence as CNP-induced relaxation. MLC phosphorylation decreased within 2 min of CNP exposure and was sustained for at least 45 min. CNP also stimulated a large transient increase in cytosolic Ca2+ concentration that occurred within 30 s and was nearly complete by 1 min. We also observed that calyculin-A, a potent inhibitor of MLC phosphatase, completely abolished the reduction in MLC phosphorylation induced by CNP. These results suggest that CNP induces intestinal SEM relaxation through cGMP-associated reductions in MLC phosphorylation. Moreover, these findings raise the possibility that CNP plays a role in intestinal wound healing.
Collapse
Affiliation(s)
- Taned Chitapanarux
- Department of Medicine, CURE Digestive Diseases Research, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA 90095, USA
| | | | | | | | | |
Collapse
|
26
|
Kitani A, Fuss I, Nakamura K, Kumaki F, Usui T, Strober W. Transforming growth factor (TGF)-beta1-producing regulatory T cells induce Smad-mediated interleukin 10 secretion that facilitates coordinated immunoregulatory activity and amelioration of TGF-beta1-mediated fibrosis. ACTA ACUST UNITED AC 2003; 198:1179-88. [PMID: 14557415 PMCID: PMC2194234 DOI: 10.1084/jem.20030917] [Citation(s) in RCA: 200] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Interleukin (IL)-10 and transforming growth factor (TGF)-β1 are suppressor cytokines that frequently occur together during a regulatory T cell response. Here we used a one gene doxycycline (Dox)-inducible plasmid encoding TGF-β1 to analyze this association and test its utility. In initial studies, we showed that intranasal administration of this plasmid (along with Dox) led to the appearance of TGF-β1–producing cells (in spleen and lamina propria) and the almost concomitant appearance of IL-10–producing cells. Moreover, we showed that these cells exert Dox-regulated suppression of the T helper cell (Th)1-mediated inflammation in trinitrobenzene sulfonic acid colitis. In subsequent in vitro studies using retroviral TGF-β1 expression, we established that IL-10 production by Th1 cells occurs after exposure to TGF-β1 from either an endogenous or exogenous source. In addition, using a self-inactivating retrovirus luciferase reporter construct we showed that TGF-β1 induces Smad4, which then binds to and activates the IL-10 promoter. Furthermore, intranasal TGF-β1 plasmid administration ameliorates bleomycin-induced fibrosis in wild-type but not IL-10–deficient mice, strongly suggesting that the amelioration is IL-10 dependent and that IL-10 protects mice from TGF-β1–mediated fibrosis. Taken together, these findings suggest that the induction of IL-10 by TGF-β1 is not fortuitous, but instead fulfills important requirements of TGF-β1 function after its secretion by regulatory T cells.
Collapse
Affiliation(s)
- Atsushi Kitani
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Building 10, Room 11N238, 10 Center Drive, Bethesda, MD 20892, USA
| | | | | | | | | | | |
Collapse
|
27
|
McKaig BC, McWilliams D, Watson SA, Mahida YR. Expression and regulation of tissue inhibitor of metalloproteinase-1 and matrix metalloproteinases by intestinal myofibroblasts in inflammatory bowel disease. THE AMERICAN JOURNAL OF PATHOLOGY 2003; 162:1355-60. [PMID: 12651627 PMCID: PMC1851243 DOI: 10.1016/s0002-9440(10)63931-4] [Citation(s) in RCA: 151] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Intestinal fibrosis and strictures frequently occur in Crohn's disease but not ulcerative colitis. We have recently shown that, compared to myofibroblasts obtained from normal and ulcerative colitis tissue, myofibroblasts isolated from fibrotic Crohn's disease mucosal samples express significantly lower amounts of transforming growth factor (TGF)-beta 3, but the expression of TGF-beta 2 was significantly greater. We now report that in myofibroblast cultures established from fibrotic Crohn's disease mucosal samples there is significantly higher constitutive expression of tissue inhibitor of metalloproteinase (TIMP)-1 compared to similar cells isolated from normal or ulcerative colitis tissue. Myofibroblasts derived from normal mucosa and from mucosa affected by ulcerative colitis or Crohn's disease also expressed matrix metalloproteinase (MMP)-1, MMP-2, and MMP-3 but did not express MMP-9. Recombinant (r) TGF-beta 1 and rTGF-beta 2, but not rTGF-beta 3, induced expression of TIMP-1 in normal intestinal myofibroblasts. These studies illustrate a potential mechanism by which differential expression of isoforms of TGF-beta may lead to excessive deposition of extracellular matrix and stricture formation via TIMP-1-mediated inhibition of MMP activity.
Collapse
Affiliation(s)
- Brian C McKaig
- Division of Gastroenterology, University Hospital, Queen's Medical Centre, Nottingham, United Kingdom.
| | | | | | | |
Collapse
|
28
|
Chamaillard M, Philpott D, Girardin SE, Zouali H, Lesage S, Chareyre F, Bui TH, Giovannini M, Zaehringer U, Penard-Lacronique V, Sansonetti PJ, Hugot JP, Thomas G. Gene-environment interaction modulated by allelic heterogeneity in inflammatory diseases. Proc Natl Acad Sci U S A 2003; 100:3455-60. [PMID: 12626759 PMCID: PMC152314 DOI: 10.1073/pnas.0530276100] [Citation(s) in RCA: 240] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
CARD15 is a major susceptibility gene for a frequent multifactorial chronic inflammatory bowel disorder, Crohn disease (CD). By using NF-kappaB activation assays, the cytosolic CARD15 was shown to efficiently detect bacterial peptidoglycan (PGN), reminiscent of the PGN recognition protein surveillance mechanism in Drosophila. The 3 CD-associated variants and 13 additional variants carried by CD patients demonstrated impaired PGN-dependent response revealing null, hypomorphic, or dominant-negative properties. Quantitative parametrization of this response, computed from the patients' CARD15 genotypes, was predictive of several variable CD manifestations. In contrast, CARD15 alleles associated with Blau's syndrome promoted PGN-independent NF-kappaB activation, an observation that accounts for the minimal microbial input in the etiology of this dominant, monogenic inflammatory disorder affecting solely aseptic sites.
Collapse
Affiliation(s)
- Mathias Chamaillard
- Fondation Jean Dausset, Centre d'Etude du Polymorphisme Humain, 27 Rue Juliette Dodu, 75010 Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Ng EK, Panesar N, Longo WE, Shapiro MJ, Kaminski DL, Tolman KC, Mazuski JE. Human intestinal epithelial and smooth muscle cells are potent producers of IL-6. Mediators Inflamm 2003; 12:3-8. [PMID: 12745542 PMCID: PMC1781593 DOI: 10.1080/0962935031000096917] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Interleukin-6 (IL-6), a pluripotent cytokine, has traditionally been considered the product of proinflammatory cells. However, many other cell types have been shown to produce IL-6. Since intestinal inflammation is commonly associated with a vigorous systemic inflammatory response, we hypothesized that intestinal epithelial and smooth muscle cells might contribute to that response by producing IL-6. We therefore studied the capacity of differentiated human intestinal epithelial and smooth muscle cell lines to produce IL-6 in response to various proinflammatory stimuli. MATERIALS AND METHODS CCL-241, a human intestinal epithelial cell line, and HISM, a human intestinal muscle cell line, were grown to confluency and then treated for 24 h with various concentrations of lipopolysaccharide, Clostridium difficile culture extract containing both toxin A and toxin B, recombinant human tumor necrosis factor-alpha (TNF-alpha), or recombinant human interleukin-1 beta (IL-1beta). Supernatants were then collected for IL-6 determination using an enzyme-linked immunosorbent assay. Cell numbers were determined using a Coulter counter. For comparison, parallel studies were performed using phorbol ester-primed U-937 and THP-1 human macrophage cell lines. RESULTS Both human intestinal epithelial and smooth muscle cells produced IL-6 under basal conditions. In HISM cells, but not in CCL-241 cells, IL-6 release was increased slightly by treatment with C. difficile culture extract containing both toxin A and toxin B and with lipopolysaccharide. In both cell lines, IL-6 production was profoundly stimulated by treatment with IL-1beta and less so with TNF-alpha. Combinations of high-dose TNF-alpha and IL-1beta may have a slightly additive, but not synergistic, effect on IL-6 release. The amount of IL-6 produced by IL-1-stimulated intestinal cell lines was 70-fold higher than that produced by stimulated macrophage cell lines. CONCLUSIONS; Both intestinal epithelial and smooth muscle cells demonstrate the ability to release significant amounts of IL-6. The profound response to IL-1beta and TNF-alpha stimulation by both cell lines suggests that human intestinal parenchymal cells, influenced by paracrine mediators liberated from proinflammatory cells, might significantly contribute to the overall systemic inflammatory response by producing IL-6.
Collapse
Affiliation(s)
- Edmond K Ng
- Department of Surgery, Saint Louis University School of Medicine, St Louis, MO, USA
| | | | | | | | | | | | | |
Collapse
|
30
|
Simmons JG, Pucilowska JB, Keku TO, Lund PK. IGF-I and TGF-beta1 have distinct effects on phenotype and proliferation of intestinal fibroblasts. Am J Physiol Gastrointest Liver Physiol 2002; 283:G809-18. [PMID: 12181198 DOI: 10.1152/ajpgi.00057.2002] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Insulin-like growth factor I (IGF-I) and transforming growth factor-beta1 (TGF-beta1) are upregulated in myofibroblasts at sites of fibrosis in experimental enterocolitis and in Crohn's disease (CD). We compared the sites of expression of IGF-I and TGF-beta1 in a rat peptidoglycan-polysaccharide (PG-PS) model of chronic granulomatous enterocolitis and fibrosis. We used the human colonic CCD-18Co fibroblast/myofibroblast cell line to test the hypothesis that TGF-beta1 and IGF-I interact to regulate proliferation, collagen synthesis, and activated phenotype typified by expression of alpha-smooth muscle actin and organization into stress fibers. IGF-I potently stimulated while TGF-beta1 inhibited basal DNA synthesis. TGF-beta1 and IGF-I each had similar but not additive effects to induce type I collagen. TGF-beta1 but not IGF-I potently stimulated expression of alpha-smooth muscle actin and stress fiber formation. IGF-I in combination with TGF-beta1 attenuated stress fiber formation without reducing alpha-smooth muscle actin expression. Stress fibers were not a prerequisite for increased collagen synthesis. TGF-beta1 upregulated IGF-I mRNA, which led us to examine the effects of IGF-I in cells previously activated by TGF-beta1 pretreatment. IGF-I potently stimulated proliferation of TGF-beta1-activated myofibroblasts without reversing activated fibrogenic phenotype. We conclude that TGF-beta1 and IGF-I both stimulate type I collagen synthesis but have differential effects on activated phenotype and proliferation. We propose that during intestinal inflammation, regulation of activated phenotype and proliferation may require sequential actions of TGF-beta1 and IGF-I, but they may act in concert to increase collagen deposition.
Collapse
Affiliation(s)
- James G Simmons
- Department of Cell and Molecular Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7545, USA.
| | | | | | | |
Collapse
|
31
|
Thiagarajah JR, Griffiths NM, Pedley KC, Naftalin RJ. Evidence for modulation of pericryptal sheath myofibroblasts in rat descending colon by transforming growth factor beta and angiotensin II. BMC Gastroenterol 2002; 2:4. [PMID: 11872151 PMCID: PMC65696 DOI: 10.1186/1471-230x-2-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2001] [Accepted: 02/12/2002] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Absorption of water and Na+ in descending colonic crypts is dependent on the barrier function of the surrounding myofibroblastic pericryptal sheath. Here the effects of high and low Na+ diets and exposure to whole body ionising radiation on the growth and activation of the descending colonic pericryptal myofibroblasts are evaluated. In addition the effect of a post-irradiation treatment with the angiotensin converting enzyme inhibitor Captopril was investigated. METHODS The levels of Angiotensin II type 1 receptor (AT1), ACE, collagen type IV, transforming growth factor-beta type 1 receptor (TGF-betaR1), OB cadherin and alpha-smooth muscle actin in both descending colon and caecum were evaluated, using immunocytochemistry and confocal microscopy, in rats fed on high and low Na+ diets (LS). These parameters were also determined during 3 months post-irradiation with 8Gy from a 60Co source in the presence and absence of the angiotensin converting enzyme inhibitor, Captopril. RESULTS Increases in AT1 receptor (135.6% +/- 18.3, P < 0.001); ACE (70.1% +/- 13.1, P < 0.001); collagen type IV (49.6% +/- 15.3, P < 0.001); TGF-+/-beta1 receptors (291.0% +/- 26.5, P < 0.001); OB-cadherin (26.3% +/- 13.8, P < 0.05) and alpha-smooth muscle actin (82.5% +/- 12.4, P < 0.001) were observed in the pericryptal myofibroblasts of the descending colon after LS diet. There are also increases in AT1 receptor and TGF-beta1 receptor, smooth muscle actin and collagen type IV after irradiation. Captopril reduced all these effects of irradiation on the pericryptal sheath and also decreased the amount of collagen and smooth muscle actin in control rats (P < 0.001). CONCLUSIONS These results demonstrate an activation of descending colonic myofibroblasts to trophic stimuli, or irradiation, which can be attenuated by Captopril, indicative of local trophic control by angiotensin II and TGF-beta release.
Collapse
Affiliation(s)
- Jay R Thiagarajah
- Division of Physiology, School of Biomedical Sciences, King's College London, Guys Campus, London, UK
| | - Nina M Griffiths
- Institut de Protection et de Sûreté Nucléaire, Fontenay aux Roses, France
| | - Kevin C Pedley
- Division of Physiology, School of Biomedical Sciences, King's College London, Guys Campus, London, UK
| | - Richard J Naftalin
- Division of Physiology, School of Biomedical Sciences, King's College London, Guys Campus, London, UK
| |
Collapse
|
32
|
Kougias P, Wei D, Rice PJ, Ensley HE, Kalbfleisch J, Williams DL, Browder IW. Normal human fibroblasts express pattern recognition receptors for fungal (1-->3)-beta-D-glucans. Infect Immun 2001; 69:3933-8. [PMID: 11349061 PMCID: PMC98428 DOI: 10.1128/iai.69.6.3933-3938.2001] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Fungal cell wall glucans nonspecifically stimulate various aspects of innate immunity. Glucans are thought to mediate their effects via interaction with membrane receptors on macrophages, neutrophils, and NK cells. There have been no reports of glucan receptors on nonimmune cells. We investigated the binding of a water-soluble glucan in primary cultures of normal human dermal fibroblasts (NHDF). Membranes from NHDF exhibited saturable binding with an apparent dissociation constant (K(D)) of 8.9 +/- 1.9 microg of protein per ml and a maximum binding of 100 +/- 8 resonance units. Competition studies demonstrated the presence of at least two glucan binding sites on NHDF. Glucan phosphate competed for all binding sites, with a K(D) of 5.6 microM (95% confidence interval [CI], 3.0 to 11 microM), while laminarin competed for 69% +/- 6% of binding sites, with a K(D) of 3.7 microM (95% CI, 1.9 to 7.3 microM). Glucan (1 microg/ml) stimulated fibroblast NF-kappaB nuclear binding activity and interleukin 6 (IL-6) gene expression in a time-dependent manner. NF-kappaB was activated at 4, 8, and 12 h, while IL-6 mRNA levels were increased by 48% at 8 h. This is the first report of pattern recognition receptors for glucan on human fibroblasts and the first demonstration of glucan binding sites on cells other than leukocytes. It also provides the first evidence that glucans can directly modulate the functional activity of NHDF. These results provide new insights into the mechanisms by which the host recognizes and responds to fungal (1-->3)-beta-D-glucans and suggests that the response to glucans may not be confined to cells of the immune system.
Collapse
Affiliation(s)
- P Kougias
- Departments of Surgery, James H. Quillen College of Medicine, Johnson City, Tennessee 37614, USA
| | | | | | | | | | | | | |
Collapse
|
33
|
Pucilowska JB, McNaughton KK, Mohapatra NK, Hoyt EC, Zimmermann EM, Sartor RB, Lund PK. IGF-I and procollagen alpha1(I) are coexpressed in a subset of mesenchymal cells in active Crohn's disease. Am J Physiol Gastrointest Liver Physiol 2000; 279:G1307-22. [PMID: 11093955 DOI: 10.1152/ajpgi.2000.279.6.g1307] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
This study tested the hypothesis that insulin-like growth factor I (IGF-I) expression is increased at sites of fibrosis in diseased intestine of patients with Crohn's disease (CD). IGF-I mRNA was quantified by RNase protection assay in uninvolved and involved intestine of 13 CD patients (10 ileum, 3 colon) and 7 ulcerative colitis (UC) patients (colon). In situ hybridization histochemistry compared the localization of IGF-I and procollagen alpha1(I) mRNAs. Masson's trichrome staining and immunohistochemistry for IGF-I precursor, alpha-smooth muscle actin (A), vimentin (V), desmin (D), and c-kit were used to examine the mesenchymal cell subtypes that express IGF-I and collagen in uninvolved and involved ileum and colon of CD patients and "normal" ileum and colon from noninflammatory controls. IGF-I mRNA was elevated in involved ileum and colon of patients with CD but not in involved colon of patients with UC. IGF-I and procollagen alpha1(I) mRNA showed overlapping distribution within fibrotic submucosa and muscularis propria of involved CD ileum and colon. In involved CD intestine, increased IGF-I precursor expression localized to mesenchymal cells in regions of tissue disorganization and fibrosis in muscularis mucosa, submucosa, and muscularis propria. In these regions, there were increased numbers of V(+) cells relative to normal or uninvolved intestine. Increased IGF-I expression was localized to cells with a phenotype typical of fibroblasts (V(+)/A(-)/D(-)), myofibroblasts (V(+)/A(+)/D(+)), and, to a lesser extent, cells with normal enteric smooth muscle phenotype (V(-)/A(+)/D(+)). We conclude that increased IGF-I expression in multiple mesenchymal cell subtypes and increased numbers of cells with fibroblast/myofibroblast phenotype are involved in fibrosis associated with CD.
Collapse
Affiliation(s)
- J B Pucilowska
- Department of Cell and Molecular Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7545, USA.
| | | | | | | | | | | | | |
Collapse
|
34
|
Chen K, Wei Y, Sharp GC, Braley‐Mullen H. Characterization of thyroid fibrosis in a murine model of granulomatous experimental autoimmune thyroiditis. J Leukoc Biol 2000. [DOI: 10.1189/jlb.68.6.828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Kemin Chen
- Department of Internal Medicine, University of Missouri, School of Medicine, Columbia, Missouri
| | - Yongzhong Wei
- Department of Internal Medicine, University of Missouri, School of Medicine, Columbia, Missouri
| | - Gordon C. Sharp
- Department of Internal Medicine, University of Missouri, School of Medicine, Columbia, Missouri
- Department of Pathology, University of Missouri, School of Medicine, Columbia, Missouri
| | - Helen Braley‐Mullen
- Department of Internal Medicine, University of Missouri, School of Medicine, Columbia, Missouri
- VA Research Service, University of Missouri, School of Medicine, Columbia, Missouri
- Molecular Microbiology & Immunology, University of Missouri, School of Medicine, Columbia, Missouri
| |
Collapse
|
35
|
Han DS, Li F, Holt L, Connolly K, Hubert M, Miceli R, Okoye Z, Santiago G, Windle K, Wong E, Sartor RB. Keratinocyte growth factor-2 (FGF-10) promotes healing of experimental small intestinal ulceration in rats. Am J Physiol Gastrointest Liver Physiol 2000; 279:G1011-22. [PMID: 11052999 DOI: 10.1152/ajpgi.2000.279.5.g1011] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Keratinocyte growth factor-2 (KGF-2, repifermin) is a homolog of KGF-1 with epithelial mitogenic activities. We investigated the therapeutic role of KGF-2 in intestinal ulceration and its mechanisms of protection. KGF-2 (0.3-5 mg/kg) was administered before or after induction of small intestinal ulceration by indomethacin (Indo) in prevention and treatment protocols. In acute studies, KGF-2 was injected for up to 7 days before or daily for 5 days after Indo. In a 15-day chronic study, KGF-2 was injected intravenously daily beginning before or 7 days after Indo. Injury was evaluated by blinded macroscopic and microscopic inflammatory scores, epithelial BrdU staining, tissue IL-1beta, PGE(2), and hydroxyproline concentrations, and collagen type I RNA expression. In vitro effects of KGF-2 were evaluated by epithelial cellular proliferation, restitution of wounded monolayers, PGE(2) secretion, and expression of COX-2 and collagen mRNA. Intravenous KGF-2 significantly decreased acute intestinal injury by all parameters and significantly decreased chronic ulceration. Pretreatment, daily infusion, and delayed treatment were effective. KGF-2 promoted in vitro epithelial restitution with only modest effects on epithelial cell proliferation, stimulated COX-2 expression in cultured epithelial cells, and upregulated in vitro and in vivo PGE(2) production. KGF-2 did not affect in vivo fibrosis, although it induced collagen expression in cultured intestinal myofibroblasts. These results suggest that KGF-2 inhibits intestinal inflammation by stimulating epithelial restitution and protective PGs.
Collapse
Affiliation(s)
- D S Han
- Center for Gastrointestinal Biology and Disease, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Pucilowska JB, Williams KL, Lund PK. Fibrogenesis. IV. Fibrosis and inflammatory bowel disease: cellular mediators and animal models. Am J Physiol Gastrointest Liver Physiol 2000; 279:G653-9. [PMID: 11005750 DOI: 10.1152/ajpgi.2000.279.4.g653] [Citation(s) in RCA: 118] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The cellular mediators of intestinal fibrosis and the relationship between fibrosis and normal repair are not understood. Identification of the types of intestinal mesenchymal cells that produce collagen during normal healing and fibrosis is vital for elucidating the answers to these questions. Acute injury may cause normal mesenchymal cells to convert to a fibrogenic phenotype that is not maintained during normal healing but may lead to fibrosis when inappropriately sustained. Proliferation of normal or fibrogenic mesenchymal cells may lead to muscularis overgrowth associated with fibrosis. The presence of increased numbers of vimentin-positive cells within fibrotic, hypertrophied muscularis in Crohn's disease suggests that changes in mesenchymal cell phenotype and number may indeed be associated with fibrosis. Fibrosis is induced in rats by peptidoglycan polysaccharides or trinitrobenzene sulfonic acid-ethanol administration, but inducing fibrosis in mice has been technically challenging. The development of current mouse models of colitis, such as dextran sodium sulfate or trinitrobenzene sulfonic acid-ethanol administration, into models of fibrosis will allow us to use genetic manipulation to study molecular mediators of fibrosis.
Collapse
Affiliation(s)
- J B Pucilowska
- Department of Cell and Molecular Physiology and Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7545, USA
| | | | | |
Collapse
|