Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Gut ; 2024 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-38378253

RESUMO

OBJECTIVE: Intestinal fibrosis is considered an inevitable consequence of chronic IBD, leading to stricture formation and need for surgery. During the process of fibrogenesis, extracellular matrix (ECM) components critically regulate the function of mesenchymal cells. We characterised the composition and function of ECM in fibrostenosing Crohn's disease (CD) and control tissues. DESIGN: Decellularised full-thickness intestinal tissue platforms were tested using three different protocols, and ECM composition in different tissue phenotypes was explored by proteomics and validated by quantitative PCR (qPCR) and immunohistochemistry. Primary human intestinal myofibroblasts (HIMFs) treated with milk fat globule-epidermal growth factor 8 (MFGE8) were evaluated regarding the mechanism of their antifibrotic response, and the action of MFGE8 was tested in two experimental intestinal fibrosis models. RESULTS: We established and validated an optimal decellularisation protocol for intestinal IBD tissues. Matrisome analysis revealed elevated MFGE8 expression in CD strictured (CDs) tissue, which was confirmed at the mRNA and protein levels. Treatment with MFGE8 inhibited ECM production in normal control HIMF but not CDs HIMF. Next-generation sequencing uncovered functionally relevant integrin-mediated signalling pathways, and blockade of integrin αvß5 and focal adhesion kinase rendered HIMF non-responsive to MFGE8. MFGE8 prevented and reversed experimental intestinal fibrosis in vitro and in vivo. CONCLUSION: MFGE8 displays antifibrotic effects, and its administration may represent a future approach for prevention of IBD-induced intestinal strictures.

2.
bioRxiv ; 2024 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-38260564

RESUMO

Crohn's disease (CD) has been traditionally viewed as a chronic inflammatory disease that cause gut wall thickening and complications, including fistulas, by mechanisms not understood. By focusing on Parabacteroides distasonis (presumed modern succinate-producing commensal probiotic), recovered from intestinal microfistulous tracts (cavernous fistulous micropathologies CavFT proposed as intermediate between 'mucosal fissures' and 'fistulas') in two patients that required surgery to remove CD-damaged ilea, we demonstrate that such isolates exert pathogenic/pathobiont roles in mouse models of CD. Our isolates are clonally-related; potentially emerging as transmissible in the community and mice; proinflammatory and adapted to the ileum of germ-free mice prone to CD-like ileitis (SAMP1/YitFc) but not healthy mice (C57BL/6J), and cytotoxic/ATP-depleting to HoxB8-immortalized bone marrow derived myeloid cells from SAMP1/YitFc mice when concurrently exposed to succinate and extracts from CavFT-derived E. coli , but not to cells from healthy mice. With unique genomic features supporting recent genetic exchange with Bacteroides fragilis -BGF539, evidence of international presence in primarily human metagenome databases, these CavFT Pdis isolates could represent to a new opportunistic Parabacteroides species, or subspecies (' cavitamuralis' ) adapted to microfistulous niches in CD.

3.
Gastroenterology ; 165(5): 1180-1196, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37507073

RESUMO

BACKGROUND & AIMS: Fibroblasts play a key role in stricture formation in Crohn's disease (CD) but understanding its pathogenesis requires a systems-level investigation to uncover new treatment targets. We studied full-thickness CD tissues to characterize fibroblast heterogeneity and function by generating the first single-cell RNA sequencing (scRNAseq) atlas of strictured bowel and providing proof of principle for therapeutic target validation. METHODS: We performed scRNAseq of 13 fresh full-thickness CD resections containing noninvolved, inflamed nonstrictured, and strictured segments as well as 7 normal non-CD bowel segments. Each segment was separated into mucosa/submucosa or muscularis propria and analyzed separately for a total of 99 tissue samples and 409,001 cells. We validated cadherin-11 (CDH11) as a potential therapeutic target by using whole tissues, isolated intestinal cells, NanoString nCounter, next-generation sequencing, proteomics, and animal models. RESULTS: Our integrated dataset revealed fibroblast heterogeneity in strictured CD with the majority of stricture-selective changes detected in the mucosa/submucosa, but not the muscle layer. Cell-cell interaction modeling revealed CXCL14+ as well as MMP/WNT5A+ fibroblasts displaying a central signaling role in CD strictures. CDH11, a fibroblast cell-cell adhesion molecule, was broadly expressed and up-regulated, and its profibrotic function was validated using NanoString nCounter, RNA sequencing, tissue target expression, in vitro gain- and loss-of-function experiments, proteomics, and knock-out and antibody-mediated CDH11 blockade in experimental colitis. CONCLUSIONS: A full-thickness bowel scRNAseq atlas revealed previously unrecognized fibroblast heterogeneity and interactions in CD strictures and CDH11 was validated as a potential therapeutic target. These results provide a new resource for a better understanding of CD stricture formation and open potential therapeutic developments. This work has been posted as a preprint on Biorxiv under doi: 10.1101/2023.04.03.534781.


Assuntos
Colite , Doença de Crohn , Animais , Doença de Crohn/genética , Doença de Crohn/patologia , Constrição Patológica , Intestinos/patologia , Colite/patologia , Fibroblastos/patologia
4.
Matrix Biol ; 113: 1-21, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36108990

RESUMO

OBJECTIVE: Inflammatory bowel diseases (IBD) cause chronic intestinal damage and extracellular matrix (ECM) remodeling. The ECM may play an active role in inflammation by modulating immune cell functions, including cell adhesion, but this hypothesis has not been tested in IBD. DESIGN: Primary human intestinal myofibroblast (HIMF)-derived ECM from IBD and controls, 3D decellularized colon or ECM molecule-coated scaffolds were tested for their adhesiveness for T cells. Matrisome was analysed via proteomics. Functional integrin blockade was used to investigate the underlying mechanism. Analysis of the pediatric Crohn's disease (CD) RISK inception cohort was used to explore an altered ECM gene expression as a potential predictor for a future complicated disease course. RESULTS: HIMF-derived ECM and 3D decellularized colonic ECM from IBD bound more T cells compared to control. Control HIMFs exposed to the pro-inflammatory cytokines Iinterleukin-1ß (IL-1ß) and tumor necrosis factor (TNF) increased, and to transforming growth factor-ß1 (TGF-ß1) decreased ECM adhesiveness to T cells. Matrisome analysis of the HIMF-derived ECM revealed collagen VI as a major culprit for differences in T cell adhesion. Collagen VI knockdown in HIMF reduced adhesion T cell as did the blockage of integrin αvß1. Elevated gene expression of collagen VI in biopsies of pediatric CD patients was linked to risk for future stricturing disease. CONCLUSION: HIMF-derived ECM in IBD binds a remarkably enhanced number of T cells, which is dependent on Collagen VI and integrin αvß1. Collagen VI expression is a risk factor for a future complicated CD course. Blocking immune cells retention may represent a novel approach to treatment in IBD.


Assuntos
Doenças Inflamatórias Intestinais , Miofibroblastos , Criança , Humanos , Miofibroblastos/metabolismo , Adesividade , Linfócitos T/patologia , Colágeno/metabolismo , Inflamação/metabolismo
5.
Gut ; 71(1): 55-67, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-33468536

RESUMO

OBJECTIVE: Creeping fat, the wrapping of mesenteric fat around the bowel wall, is a typical feature of Crohn's disease, and is associated with stricture formation and bowel obstruction. How creeping fat forms is unknown, and we interrogated potential mechanisms using novel intestinal tissue and cell interaction systems. DESIGN: Tissues from normal, UC, non-strictured and strictured Crohn's disease intestinal specimens were obtained. The muscularis propria matrisome was determined via proteomics. Mesenteric fat explants, primary human preadipocytes and adipocytes were used in multiple ex vivo and in vitro cell migration systems on muscularis propria muscle cell derived or native extracellular matrix. Functional experiments included integrin characterisation via flow cytometry and their inhibition with specific blocking antibodies and chemicals. RESULTS: Crohn's disease muscularis propria cells produced an extracellular matrix scaffold which is in direct spatial and functional contact with the immediately overlaid creeping fat. The scaffold contained multiple proteins, but only fibronectin production was singularly upregulated by transforming growth factor-ß1. The muscle cell-derived matrix triggered migration of preadipocytes out of mesenteric fat, fibronectin being the dominant factor responsible for their migration. Blockade of α5ß1 on the preadipocyte surface inhibited their migration out of mesenteric fat and on 3D decellularised intestinal tissue extracellular matrix. CONCLUSION: Crohn's disease creeping fat appears to result from the migration of preadipocytes out of mesenteric fat and differentiation into adipocytes in response to an increased production of fibronectin by activated muscularis propria cells. These new mechanistic insights may lead to novel approaches for prevention of creeping fat-associated stricture formation.


Assuntos
Adipócitos/patologia , Movimento Celular , Doença de Crohn/patologia , Intestinos/patologia , Músculo Liso/patologia , Adipogenia/fisiologia , Tecido Adiposo/patologia , Diferenciação Celular , Células Cultivadas , Matriz Extracelular/patologia , Fibronectinas/metabolismo , Humanos , Alicerces Teciduais
6.
Inflamm Bowel Dis ; 25(2): 294-305, 2019 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-30295747

RESUMO

Background: Carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) displays multiple activities, among which pathogen binding and angiogenesis are particularly prominent. These same functions are also exerted by Toll- and NOD-like receptors (TLRs and NLRs), which are critical mediators of innate immune responses. We investigated whether a functional inter-relationship exists between CEACAM1 and TLRs and NLRs and its potential impact on induction of intestinal angiogenesis. Methods: This hypothesis was tested using human intestinal microvascular endothelial cells, a unique cell population exposed to microbial products under physiological and pathological conditions. Results: The results show that activation of TLR2/4, TLR4, NOD1, and NOD2 by specific bacterial ligands selectively and differentially upregulates the levels of cellular and soluble CEACAM1 produced by intestinal microvascular endothelial cells. The results also show that CEACAM1 regulates the migration, transmigration, and tube formation of these endothelial cells and mediates vessel sprouting induced by specific TLR and NLR bacterial ligands. Combined, these results demonstrate a close and reciprocal regulatory interaction between CEACAM1 and bacterial products in mediating multiple functions essential to new vessel formation in the gut mucosa. Conclusions: A coordinated and reciprocal interaction of CEACAM1 and microbiota-derived factors is necessary to optimize angiogenesis in the gut mucosa. This suggests that a coordination of endogenous and exogenous innate immune responses is necessary to promote intestinal angiogenesis under physiological and inflammatory conditions such as inflammatory bowel disease.


Assuntos
Antígenos CD/metabolismo , Moléculas de Adesão Celular/metabolismo , Imunidade Inata/imunologia , Mediadores da Inflamação/metabolismo , Doenças Inflamatórias Intestinais/patologia , Mucosa Intestinal/patologia , Microvasos/patologia , Neovascularização Fisiológica , Animais , Antígenos de Bactérias/imunologia , Antígenos CD/genética , Estudos de Casos e Controles , Moléculas de Adesão Celular/antagonistas & inibidores , Moléculas de Adesão Celular/genética , Movimento Celular , Proliferação de Células , Citocinas/metabolismo , Humanos , Doenças Inflamatórias Intestinais/tratamento farmacológico , Doenças Inflamatórias Intestinais/imunologia , Doenças Inflamatórias Intestinais/metabolismo , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Microvasos/imunologia , Microvasos/metabolismo , Proteína Adaptadora de Sinalização NOD1/metabolismo , Proteína Adaptadora de Sinalização NOD2/metabolismo , RNA Interferente Pequeno/genética , Receptor 2 Toll-Like/metabolismo , Receptor 4 Toll-Like/metabolismo
7.
Cell Mol Gastroenterol Hepatol ; 2(3): 358-368.e4, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-27981209

RESUMO

BACKGROUND & AIMS: Crohn's Disease (CD) is a chronic inflammatory disease of the gastrointestinal tract. Fibrosis, a serious complication of CD, occurs when activated intestinal fibroblasts deposit excessive amounts of extracellular matrix (ECM) in affected areas. A major component of the ECM is high-molecular-weight hyaluronan (HA) that, when depolymerized to low-molecular-weight fragments, becomes proinflammatory and profibrotic. Mechanisms for HA degradation are incompletely understood, but the novel protein KIAA1199 recently was discovered to degrade HA. We hypothesized that KIAA1199 protein is increased in CD colon fibroblasts and generates HA fragments that foster inflammation and fibrosis. METHODS: Fibroblasts were isolated from explants of surgically resected colon tissue from CD and non-inflammatory bowel disease control (ND) patients. Protein levels and tissue distribution of KIAA1199 were assessed by immunoblot and immunostaining, and functional HA degradation was measured biochemically. RESULTS: Increased levels of KIAA1199 protein were produced and deposited in the ECM by cultured CD fibroblasts compared with controls. Treatment of fibroblasts with the proinflammatory cytokine interleukin (IL) 6 increased deposition of KIAA1199 in the ECM. CD fibroblasts also produce significantly higher levels of IL6 compared with controls, and antibody blockade of IL6 receptors in CD colon fibroblasts decreased the level of KIAA1199 protein in the ECM. Colon fibroblasts degrade HA, however, small interfering RNA silencing of KIAA1199 abrogated that ability. CONCLUSIONS: CD fibroblasts produce increased levels of KIAA1199 primarily through an IL6-driven autocrine mechanism. This leads to excessive degradation of HA and the generation of proinflammatory HA fragments, which contributes to maintenance of gut inflammation and fibrosis.

8.
Am J Pathol ; 185(9): 2550-62, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26185013

RESUMO

The endothelial-to-mesenchymal transition (EndoMT) is a crucial cellular process during heart development necessary to the formation of cardiac valves. This embryonic process reappears in several pathological situations, such as vascular injury or organ fibrosis of various etiologies, as a mediator of extracellular matrix-producing cells. Because radiation induces both vascular damage and fibrosis, we investigated whether radiation exposure induces EndoMT in primary human intestinal microvascular endothelial cells (HIMECs) and whether EndoMT contributes to radiation-induced rectal damage in humans and in a preclinical model of radiation proctitis in mice. Irradiated HIMECs show phenotypic hallmarks of radiation-induced endothelial cell activation in vitro. Moreover, HIMECs undergo changes in molecular expression pattern compatible with EndoMT, with up-regulation of mesenchymal markers and down-regulation of endothelial markers via transforming growth factor/Smad pathway activation. In vivo, EndoMT readily occurs in the human rectum after radiation therapy for rectal adenocarcinoma. Finally, EndoMT was observed in rectal mucosal and submucosal microvessels in a preclinical model of radiation proctitis in Tie2-green fluorescent protein reporter-expressing mice all along radiation proctitis development, also associated with transforming growth factor/Smad pathway activation. In conclusion, radiation-induced cell activation and tissue inflammation constitute a setting that fosters the phenotypic conversion of endothelial cells into mesenchymal cells. Therefore, EndoMT is identified as a potential participant in radiation-induced gut damage and may represent an interesting therapeutic target in cases of radiation-induced pelvic disease.


Assuntos
Células Endoteliais/metabolismo , Matriz Extracelular/metabolismo , Proctite/metabolismo , Lesões por Radiação/metabolismo , Animais , Biomarcadores/metabolismo , Células Cultivadas , Transição Epitelial-Mesenquimal , Fibrose/metabolismo , Fibrose/patologia , Inflamação/metabolismo , Inflamação/patologia , Camundongos , Proctite/genética , Proctite/patologia , Regulação para Cima/efeitos da radiação
9.
Am J Pathol ; 185(6): 1624-37, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25864926

RESUMO

Intestinal epithelial cell (IEC) death is typical of inflammatory bowel disease (IBD). We investigated: i) whether IEC-released necrotic cell products (proinflammatory mediators) amplify mucosal inflammation, ii) the capacity of necrotic cell lysates from HT29 cells or human IECs to induce human intestinal fibroblasts' (HIF) production of IL-6 and IL-8, and iii) whether IL-1α, released by injured colonocytes, exacerbated experimental IBD. Necrotic cell lysates potently induced HIF IL-6 and IL-8 production independent of Toll-like receptors 2 and 4, receptor for advanced glycation end-products, high-mobility group box 1, uric acid, IL-33, or inflammasome activation. IL-1α was the key IEC-derived necrotic cell product involved in HIF cytokine production. IL-1α-positive cells were identified in the epithelium in human IBD and dextran sulfate sodium (DSS)-induced colitis. IL-1α was detected in the stool of colitic mice before IL-1ß. IL-1α enemas reactivated inflammation after DSS colitis recovery, induced IL-1 receptor expression in subepithelial fibroblasts, and activated de novo inflammation even in mice without overt colitis, after the administration of low-dose DSS. IL-1α amplifies gut inflammation by inducing cytokine production by mesenchymal cells. IL-1α-mediated IEC-fibroblast interaction may be involved in amplifying and perpetuating inflammation, even without obvious intestinal damage. IL-1α may be a target for treating early IBD or preventing the reactivation of IBD.


Assuntos
Colite/metabolismo , Fibroblastos/metabolismo , Inflamação/metabolismo , Interleucina-1alfa/metabolismo , Mucosa Intestinal/metabolismo , Animais , Colite/induzido quimicamente , Colite/patologia , Sulfato de Dextrana , Modelos Animais de Doenças , Fibroblastos/patologia , Células HT29 , Humanos , Inflamação/patologia , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Intestinos/patologia , Camundongos
10.
Gastroenterology ; 148(7): 1405-1416.e3, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25701737

RESUMO

BACKGROUND & AIMS: Defects in colonic epithelial barrier defenses are associated with ulcerative colitis (UC). The proteins that regulate bacterial clearance in the colonic epithelium have not been completely identified. The Drosophila chromosome-associated protein D3 (dCAP-D3) regulates responses to bacterial infection. We examined whether CAP-D3 promotes bacterial clearance in human colonic epithelium. METHODS: Clearance of Salmonella or adherent-invasive Escherichia coli LF82 was assessed by gentamycin protection assays in HT-29 and Caco-2 cells expressing small hairpin RNAs against CAP-D3. We used immunoblot assays to measure levels of CAP-D3 in colonic epithelial cells from patients with UC and healthy individuals (controls). RNA sequencing identified genes activated by CAP-D3. We analyzed the roles of CAP-D3 target genes in bacterial clearance using gentamycin protection and immunofluorescence assays and studies with pharmacologic inhibitors. RESULTS: CAP-D3 expression was reduced in colonic epithelial cells from patients with active UC. Reduced CAP-D3 expression decreased autophagy and impaired intracellular bacterial clearance by HT-29 and Caco-2 colonic epithelial cells. Lower levels of CAP-D3 increased transcription of genes encoding SLC7A5 and SLC3A2, the products of which heterodimerize to form an amino acid transporter in HT-29 cells after bacterial infection; levels of SLC7A5-SLC3A2 were increased in tissues from patients with UC compared with controls. Reduced CAP-D3 in HT-29 cells resulted in earlier recruitment of SLC7A5 to Salmonella-containing vacuoles, increased activity of mTORC1, and increased survival of bacteria. Inhibition of SLC7A5-SLC3A2 or mTORC1 activity rescued the bacterial clearance defects of CAP-D3-deficient cells. CONCLUSIONS: CAP-D3 down-regulates transcription of genes that encode amino acid transporters (SLC7A5 and SLC3A2) to promote bacterial autophagy by colon epithelial cells. Levels of CAP-D3 protein are reduced in patients with active UC; strategies to increase its levels might restore mucosal homeostasis to patients with active UC.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Escherichia coli/fisiologia , Cadeia Pesada da Proteína-1 Reguladora de Fusão/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Transportador 1 de Aminoácidos Neutros Grandes/metabolismo , Salmonella/fisiologia , Adenosina Trifosfatases , Autofagia , Células CACO-2 , Proteínas de Ciclo Celular/genética , Colite Ulcerativa/imunologia , Colite Ulcerativa/metabolismo , Colite Ulcerativa/microbiologia , Doença de Crohn/imunologia , Doença de Crohn/metabolismo , Doença de Crohn/microbiologia , Proteínas de Drosophila , Células Epiteliais/imunologia , Escherichia coli/imunologia , Cadeia Pesada da Proteína-1 Reguladora de Fusão/genética , Regulação da Expressão Gênica , Células HT29 , Humanos , Imunidade Inata , Mucosa Intestinal/imunologia , Transportador 1 de Aminoácidos Neutros Grandes/genética , Alvo Mecanístico do Complexo 1 de Rapamicina , Viabilidade Microbiana , Complexos Multiproteicos/metabolismo , Interferência de RNA , Salmonella/imunologia , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Fatores de Tempo , Transcrição Gênica , Transfecção
11.
Am J Pathol ; 184(12): 3405-14, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25307345

RESUMO

The expression of neurotensin (NT) and its receptor (NTR1) is up-regulated in experimental colitis and inflammatory bowel disease; NT/NTR1 interactions regulate gut inflammation. During active inflammation, metabolic shifts toward hypoxia lead to the activation of hypoxia-inducible factor (HIF)-1, which enhances vascular endothelial growth factor (VEGF) expression, promoting angiogenesis. We hypothesized that NT/NTR1 signaling regulates intestinal manifestations of hypoxia and angiogenesis by promoting HIF-1 transcriptional activity and VEGFα expression in experimental colitis. We studied NTR1 signaling in colitis-associated angiogenesis using 2,4,6-trinitrobenzenesulfonic acid-treated wild-type and NTR1-knockout mice. The effects of NT on HIF-1α and VEGFα were assessed on human colonic epithelial cells overexpressing NTR1 (NCM460-NTR1) and human intestinal microvascular-endothelial cells. NTR1-knockout mice had reduced microvascular density and mucosal integrity score compared with wild-type mice after 2,4,6-trinitrobenzenesulfonic acid treatment. VEGFα mRNA levels were increased in NCM460-NTR1 cells treated with 10(-7) mol/L NT, at 1 and 6 hours post-treatment. NT exposure in NCM460-NTR1 cells caused stabilization, nuclear translocation, and transcriptional activity of HIF-1α in a diacylglycerol kinase-dependent manner. NT did not stimulate tube formation in isolated human intestinal macrovascular endothelial cells but did so in human intestinal macrovascular endothelial cells cocultured with NCM460-NTR1 cells. Our results demonstrate the importance of an NTR1-HIF-1α-VEGFα axis in intestinal angiogenic responses and in the pathophysiology of colitis and inflammatory bowel disease.


Assuntos
Colo/patologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Inflamação/metabolismo , Mucosa Intestinal/metabolismo , Receptores de Neurotensina/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Colite/patologia , Colo/citologia , Modelos Animais de Doenças , Células Endoteliais/citologia , Humanos , Imuno-Histoquímica , Doenças Inflamatórias Intestinais/induzido quimicamente , Doenças Inflamatórias Intestinais/metabolismo , Intestinos/irrigação sanguínea , Masculino , Camundongos , Microcirculação , Neovascularização Patológica , Ácido Trinitrobenzenossulfônico/química , Regulação para Cima
12.
Gastroenterology ; 146(5): 1266-77.e1-9, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24486052

RESUMO

BACKGROUND & AIMS: Patients with eosinophilic esophagitis (EoE) often become dysphagic from the combination of organ fibrosis and motor abnormalities. We investigated mechanisms of dysphagia, assessing the response of human esophageal fibroblasts (HEFs), human esophageal muscle cells (HEMCs), and esophageal muscle strips to eosinophil-derived products. METHODS: Biopsy specimens were collected via endoscopy from the upper, middle, and lower thirds of the esophagus of 18 patients with EoE and 21 individuals undergoing endoscopy for other reasons (controls). Primary cultures of esophageal fibroblasts and muscle cells were derived from 12 freshly resected human esophagectomy specimens. Eosinophil distribution was investigated by histologic analyses of full-thickness esophageal tissue. Active secretion of EoE-related mediators was assessed from medium underlying mucosal biopsy cultures. We quantified production of fibronectin and collagen I by HEF and HEMC in response to eosinophil products. We also measured the expression of intercellular adhesion molecule-1 and vascular cell adhesion molecule-1 by, and adhesion of human eosinophils to, HEFs and HEMCs. Eosinophil products were tested in an esophageal muscle contraction assay. RESULTS: Activated eosinophils were present in all esophageal layers. Significantly higher concentrations of eosinophil-related mediators were secreted spontaneously in mucosal biopsy specimens from patients with EoE than controls. Exposure of HEFs and HEMCs to increasing concentrations of eosinophil products or co-culture with eosinophils caused HEFs and HEMCs to increase secretion of fibronectin and collagen I; this was inhibited by blocking transforming growth factor ß1 and p38 mitogen-activated protein kinase signaling. Eosinophil binding to HEFs and HEMCs increased after incubation of mesenchymal cells with eosinophil-derived products, and decreased after blockade of transforming growth factor ß1 and p38 mitogen-activated protein kinase blockade. Eosinophil products reduced electrical field-induced contraction of esophageal muscle strips, but not acetylcholine-induced contraction. CONCLUSIONS: In an analysis of tissues samples from patients with EoE, we linked the presence and activation state of eosinophils in EoE with altered fibrogenesis and motility of esophageal fibroblasts and muscle cells. This process might contribute to the development of dysphagia.


Assuntos
Citocinas/metabolismo , Transtornos de Deglutição/etiologia , Deglutição , Esofagite Eosinofílica/complicações , Eosinófilos/imunologia , Contração Muscular , Células Th2/imunologia , Fator de Crescimento Transformador beta1/metabolismo , Idoso , Biópsia , Estudos de Casos e Controles , Adesão Celular , Comunicação Celular , Células Cultivadas , Técnicas de Cocultura , Colágeno Tipo I/metabolismo , Transtornos de Deglutição/imunologia , Transtornos de Deglutição/metabolismo , Transtornos de Deglutição/patologia , Transtornos de Deglutição/fisiopatologia , Esofagite Eosinofílica/imunologia , Esofagite Eosinofílica/metabolismo , Esofagite Eosinofílica/patologia , Esofagite Eosinofílica/fisiopatologia , Eosinófilos/metabolismo , Esofagoscopia , Feminino , Fibroblastos/imunologia , Fibroblastos/metabolismo , Fibroblastos/patologia , Fibronectinas/metabolismo , Fibrose , Humanos , Molécula 1 de Adesão Intercelular/metabolismo , Masculino , Pessoa de Meia-Idade , Mucosa/imunologia , Mucosa/metabolismo , Mucosa/patologia , Miócitos de Músculo Liso/imunologia , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Células Th2/metabolismo , Molécula 1 de Adesão de Célula Vascular/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
13.
Inflamm Bowel Dis ; 19(7): 1354-64, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23635716

RESUMO

BACKGROUND: Fibrosis of the intestine is currently an irreversible complication of inflammatory bowel disease; yet, little is understood of the underlying pathogenesis and antifibrotic strategies remain elusive. To develop effective therapies, knowledge of the mechanism of transcription and excessive deposition of type I collagen, a hallmark of fibrosis, is needed. We have shown previously that endothelial-to-mesenchymal transition (EndoMT) contributes to the pool of intestinal fibrotic cells and that a cytokine cocktail (interleukin 1-ß, tumor necrosis factor α, and transforming growth factor ß) induces collagen I alpha 2 (COL1A2) mRNA and protein. METHODS: Chromatin immunoprecipitation assays on pure cultures of human intestinal mucosal endothelial cells undergoing EndoMT were performed with antibodies to specific histone modifications and RNA polymerase II. Reverse transcriptase-PCR was used to quantify the levels of Col1A2 and endothelial-specific von Willebrand factor (vWF) mRNA. RESULTS: We showed that cytokines induce selective chromatin modifications (histone 4 hyperacetylation, and hypermethylation of histone 3) and phosphorylated RNA polymerase II at the COL1A2 promoter. Hypoacetylated and hypomethylated histone 3 was detected on the repressed vWF gene. Prolonged exposure to cytokines (16 days) retained hyperacetylation of select lysines in H4 on the COL1A2 promoter. Removal of cytokines after 16 days and continued culture for 10 days showed persistent hyperacetylation at lysine 16 in histone H4. CONCLUSIONS: This is the first study to show that COL1A2 gene expression is associated with cytokine-induced, temporally ordered, and persistent chromatin modifications and suggests that these are important determinants of gene expression in EndoMT and intestinal fibrosis.


Assuntos
Cromatina/genética , Colágeno Tipo I/genética , Endotélio Vascular/efeitos dos fármacos , Fibrose/tratamento farmacológico , Interleucina-1beta/farmacologia , Mucosa Intestinal/patologia , Mesoderma/efeitos dos fármacos , Fator de Crescimento Transformador beta1/farmacologia , Fator de Necrose Tumoral alfa/farmacologia , Western Blotting , Células Cultivadas , Imunoprecipitação da Cromatina , Colágeno Tipo I/metabolismo , Metilação de DNA , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Fibrose/metabolismo , Fibrose/patologia , Histonas/genética , Humanos , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Lisina/genética , Mesoderma/metabolismo , Mesoderma/patologia , Regiões Promotoras Genéticas , RNA Polimerase II/metabolismo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transcrição Gênica
14.
Gastroenterology ; 144(3): 613-623.e9, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23149220

RESUMO

BACKGROUND & AIMS: In intestinal inflammation the gut microbiota induces an innate immune response by activating epithelial and immune cells that initiate or maintain inflammation. We investigated whether the microbiota also can activate local microvascular cells and induce angiogenesis. METHODS: Human intestinal microvascular endothelial cells (HIMEC) and human intestinal fibroblasts (HIF) were exposed to bacterial ligands specific for Toll-like receptor (TLR)2/6 and 4, and NOD1 and NOD2, and cell proliferation, migration, transmigration, tube formation, and production of pro-angiogenic factors were measured. The ability of the ligands to induce ex vivo vessel sprouting in an aortic ring assay and in vivo angiogenesis using a collagen gel assay also were assessed. RESULTS: Bacterial ligands induced proliferation, migration, transmigration, tube formation of HIMEC, vessel sprouting, and in vivo angiogenesis; they also stimulated production of angiogenic factors from HIMEC and HIF, and HIF-derived angiogenic factors promoted HIMEC proliferation. To various degrees, all ligands induced angiogenic responses, but these were ligand- and cell type-dependent. Responses were mediated through receptor interacting protein-2 (RIP2)- and tumor necrosis factor receptor-associated factor 6 (TRAF6)-dependent signaling, involved the mitogen-activated protein kinase (MAPK) and nuclear factor-κB (NF-κB) pathways and the up-regulation of vascular endothelial growth factor receptor 2 (VEGF-R2) and focal adhesion kinase (FAK). Knockdown of RIP2 and TRAF6 by RNA interference and neutralization of interleukin-8, basic fibroblast growth factor, and vascular endothelial growth factor inhibited TLR-/NOD-like receptor-induced HIMEC angiogenesis. CONCLUSIONS: The gut microbiota can selectively activate mucosal endothelial and mesenchymal cells to promote specific angiogenic responses in a TLR- and NOD-like receptor-dependent fashion. This innate immunity-mediated response may expand the mucosal microvascular network, foster immune cell recruitment, and contribute to chronic intestinal inflammation.


Assuntos
Intestinos/irrigação sanguínea , Intestinos/microbiologia , Metagenoma/fisiologia , Neovascularização Fisiológica , Proteína Adaptadora de Sinalização NOD1/metabolismo , Proteína Adaptadora de Sinalização NOD2/metabolismo , Receptores Toll-Like/metabolismo , Proliferação de Células/efeitos dos fármacos , Meios de Cultivo Condicionados/farmacologia , Fibroblastos/fisiologia , Humanos , NF-kappa B/metabolismo , Interferência de RNA , Proteína Serina-Treonina Quinase 2 de Interação com Receptor/genética , Proteína Serina-Treonina Quinase 2 de Interação com Receptor/metabolismo , Fator 6 Associado a Receptor de TNF/metabolismo , Migração Transendotelial e Transepitelial/efeitos dos fármacos , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
15.
Gastroenterology ; 143(4): 1017-26.e9, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22749932

RESUMO

BACKGROUND & AIMS: Defective apoptosis of lamina propria T cells (LPTs) is involved in the pathogenesis of Crohn's disease. Survivin, a member of the inhibitors of apoptosis family, prevents cell death and regulates cell division. Survivin has been studied extensively in cancer, but little is known about its role in Crohn's disease. METHODS: LPTs were isolated from mucosal samples of patients with Crohn's disease or ulcerative colitis and healthy individuals (controls). LPTs were activated with interleukin-2 or via CD3, CD2, and CD28 signaling, and cultured at 42°C to induce heat shock. Survivin expression was assessed by immunohistochemistry, confocal microscopy, and immunoblotting; survivin levels were reduced by RNA interference. Cell viability, apoptosis, and proliferation were measured by trypan blue exclusion, annexin-V/7-Aminoactinomycin D staining, and uptake of [3]thymidine, respectively. RESULTS: LPTs from patients with Crohn's disease had higher levels of survivin than LPTs from patients with ulcerative colitis or controls. RNA knockdown of survivin in LPTs inhibited their proliferation and promoted apoptosis. Levels of survivin were low in LPTs from patients with ulcerative colitis and controls as a result of ubiquitin-mediated proteasome degradation. In LPTs from patients with Crohn's disease, survivin bound to the heat shock protein (HSP)90, and therefore was resistant to proteasome degradation. Incubating LPTs with 17-N-allylamino-17-demethoxygeldanamycin, an inhibitor of HSP90, reduced levels of survivin and induced apoptosis. CONCLUSIONS: Levels of survivin are increased in LPTs from patients with Crohn's disease (compared with ulcerative colitis and controls) because survivin interacts with HSP90 and prevents proteasome degradation. This allows LPTs to avoid apoptosis. Strategies to restore apoptosis to these cells might be developed to treat patients with Crohn's disease.


Assuntos
Colite Ulcerativa/metabolismo , Doença de Crohn/metabolismo , Proteínas de Choque Térmico HSP90/metabolismo , Proteínas Inibidoras de Apoptose/metabolismo , Linfócitos T/metabolismo , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Proteínas Reguladoras de Apoptose , Antígenos CD2/metabolismo , Antígenos CD28/metabolismo , Complexo CD3/metabolismo , Complexo CD3/farmacologia , Núcleo Celular/metabolismo , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Colite Ulcerativa/imunologia , Doença de Crohn/imunologia , Citoplasma/metabolismo , Endopeptidase K/farmacologia , Feminino , Humanos , Proteínas Inibidoras de Apoptose/genética , Interleucina-2/farmacologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Masculino , Pessoa de Meia-Idade , Proteínas Mitocondriais , Fosforilação/efeitos dos fármacos , Proteólise , Interferência de RNA , RNA Mensageiro/metabolismo , Transdução de Sinais , Survivina , Linfócitos T/efeitos dos fármacos , Linfócitos T/enzimologia , Ubiquitinação , Adulto Jovem
16.
PLoS One ; 6(5): e20454, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21637825

RESUMO

BACKGROUND: NKX2-3 is associated with inflammatory bowel disease (IBD). NKX2-3 is expressed in microvascular endothelial cells and the muscularis mucosa of the gastrointestinal tract. Human intestinal microvascular endothelial cells (HIMECs) are actively involved in the pathogenesis of IBD and IBD-associated microvascular dysfunction. To understand the cellular function of NKX2-3 and its potential role underlying IBD pathogenesis, we investigated the genes regulated by NKX2-3 in HIMEC using cDNA microarray. METHODOLOGY/PRINCIPAL FINDINGS: NKX2-3 expression was suppressed by shRNA in two HIMEC lines and gene expression was profiled by cDNA microarray. Pathway Analysis was used to identify gene networks according to biological functions and associated pathways. Validation of microarray and genes expression in intestinal tissues was assessed by RT-PCR. NKX2-3 regulated genes are involved in immune and inflammatory response, cell proliferation and growth, metabolic process, and angiogenesis. Several inflammation and angiogenesis related signaling pathways that play important roles in IBD were regulated by NKX2-3, including endothelin-1 and VEGF-PI3K/AKT-eNOS. Expression levels of NKX2-3, VEGFA, PI3K, AKT, and eNOS are increased in intestinal tissues from IBD patients and expression levels of EDN1 are decreased in intestinal tissues from IBD patients. These results demonstrated the important roles of NKX2-3, VEGF, PI3K, AKT, eNOS, and EDN1 in IBD pathogenesis. Correlation analysis showed a positive correlation between mRNA expression of NKX2-3 and VEGFA and a negative correlation between mRNA expression of NKX2-3 and EDN1 in intestinal tissues from IBD patients. CONCLUSION/RELEVANCE: NKX2-3 may play an important role in IBD pathogenesis by regulating endothelin-1 and VEGF signaling in HIMECs.


Assuntos
Células Endoteliais/metabolismo , Endotelina-1/genética , Regulação da Expressão Gênica , Proteínas de Homeodomínio/metabolismo , Microvasos/patologia , Fatores de Transcrição/metabolismo , Transcrição Gênica , Fator A de Crescimento do Endotélio Vascular/genética , Adulto , Linhagem Celular , Demografia , Células Endoteliais/enzimologia , Endotelina-1/metabolismo , Feminino , Técnicas de Silenciamento de Genes , Redes Reguladoras de Genes/genética , Proteínas de Homeodomínio/genética , Humanos , Doenças Inflamatórias Intestinais/genética , Doenças Inflamatórias Intestinais/patologia , Intestinos/patologia , Masculino , Pessoa de Meia-Idade , Óxido Nítrico Sintase Tipo III/genética , Óxido Nítrico Sintase Tipo III/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Reprodutibilidade dos Testes , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/genética , Fatores de Transcrição/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
17.
Clin Transl Sci ; 1(1): 57-61, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-20443819

RESUMO

Increased hyaluronan (HA) deposition is a common feature of inflamed tissues, including inflammatory bowel disease (IBD)-involved intestines. However, whether HA accumulation promotes or is the result of intestinal inflammation is unknown. Using the mouse dextran sulfate sodium (DSS)-induced experimental model of colitis, we investigated changes in HA deposition in the colon over time in conjunction with evolving pathological changes of tissue architecture. Profound changes in colon HA deposition occurred within 3-7 days of oral DSS administration and, more important, they preceded the inflammatory infiltrate. Interestingly, HA deposition within blood vessels of the colon is observed as early as 3 days during the course of colitis induction, well before any significant inflammatory infiltrate. HA deposition is also observed in blood vessels of inflamed human colon of IBD patients. We determined that human intestinal endothelial cells generate HA in response to proinflammatory stimuli by demonstrating a TNF-alpha-induced increase in hyaluronan synthase-3 mRNA expression and the accumulation of HA cable-like structures that are adhesive for leukocytes. Additionally, IBD mucosal endothelial cells produce higher levels of cell surface HA in response to TNF-alpha than non-IBD control cells. Therefore, HA deposition is an early event in inflamed gut tissue, preceding and likely promoting leukocyte infiltration.


Assuntos
Colite/metabolismo , Ácido Hialurônico/metabolismo , Inflamação , Doenças Inflamatórias Intestinais/metabolismo , Mucosa Intestinal/metabolismo , Leucócitos/citologia , Animais , Sulfato de Dextrana/química , Células Endoteliais/citologia , Endotélio/patologia , Endotélio Vascular/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fator de Necrose Tumoral alfa/metabolismo
18.
Gastroenterology ; 132(1): 139-53, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17241867

RESUMO

BACKGROUND & AIMS: Fractalkine (FKN/CX3CL1) is a unique chemokine combining adhesive and chemotactic properties. We investigated FKN production by the mucosal microvasculature in inflammatory bowel disease (IBD), its capacity for leukocyte recruitment into the gut, and the number of CX3CR1+ cells in the circulation and mucosa of IBD patients. METHODS: The expression of FKN by human intestinal microvascular endothelial cells (HIMECs) and CX3CR1 by circulating cells was evaluated by flow cytometry, and mucosal CX3CR1+ cells were enumerated by immunohistochemistry. The capacity of FKN to mediate leukocyte binding to HIMECs was assessed by immunoblockade, and to induce HIMEC transmigration by a Transwell system. RESULTS: The spontaneously low HIMEC FKN expression was enhanced markedly by tumor necrosis factor-alpha plus interferon-gamma stimulation, or direct leukocyte contact. This effect was significantly stronger in IBD than control HIMECs. Up-regulation of HIMEC FKN expression was dependent on p38 and extracellular signal-regulated kinase phosphorylation, as was abrogated by selective mitogen-activated protein kinase inhibitors. Circulating T cells contained significantly higher numbers of CX3CR1+ cells in active IBD than inactive IBD or healthy subjects, and IBD mucosa contained significantly more CX3CR1+ cells than control mucosa. Antibody-blocking experiments showed that FKN was a major contributor to T- and monocytic-cell adhesion to HIMECs. Finally, FKN enhanced the expression of active beta1 integrin on leukocytes and mediated leukocyte HIMEC transmigration. CONCLUSIONS: In view of the capacity of FKN to mediate leukocyte adhesion, chemoattraction, and transmigration, its increased production by mucosal microvascular cells and increased numbers of circulating and mucosal CX3CR1+ cells in IBD point to a significant role of FKN in disease pathogenesis.


Assuntos
Movimento Celular/imunologia , Quimiocinas CX3C/metabolismo , Células Endoteliais/citologia , Doenças Inflamatórias Intestinais/imunologia , Proteínas de Membrana/metabolismo , Receptores de Quimiocinas/metabolismo , Linfócitos T/citologia , Receptor 1 de Quimiocina CX3C , Adesão Celular/imunologia , Células Cultivadas , Quimiocina CX3CL1 , Quimiocinas CX3C/imunologia , Citocinas/farmacologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/imunologia , Citometria de Fluxo , Humanos , Doenças Inflamatórias Intestinais/patologia , Integrina beta1/metabolismo , Mucosa Intestinal/irrigação sanguínea , Mucosa Intestinal/imunologia , Células Jurkat , Sistema de Sinalização das MAP Quinases/imunologia , Proteínas de Membrana/imunologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Receptores de Superfície Celular/metabolismo , Receptores de Quimiocinas/imunologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Regulação para Cima/imunologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
19.
Gastroenterology ; 132(1): 154-65, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17241868

RESUMO

BACKGROUND & AIMS: Gastroesophageal reflux disease is a condition frequently associated with esophagitis and motor abnormalities. Recent evidence suggests that proinflammatory cytokines, such as interleukin (IL)-1beta and IL-6, may be implicated because they reduce esophageal muscle contractility, but these results derive from in vitro or animal models of esophagitis. This study used human esophageal cells and tissues to identify the cellular source of cytokines in human esophagitis investigate whether cytokines can be induced by gastric refluxate, and examine whether esophageal tissue- or cell-derived mediators affect muscle contractility. METHODS: Endoscopic mucosal biopsy specimens were obtained from patients with and without esophagitis, organ-cultured, and undernatants were assessed for cytokine content. The cytokine profile of esophageal epithelial, fibroblast, and muscle cells was analyzed, and esophageal mucosa and cell products were tested in an esophageal circular muscle contraction assay. RESULTS: The mucosa of esophagitis patients produced significantly greater amounts of IL-1beta and IL-6 compared with those of control patients. Cultured esophageal epithelial cells produced IL-6, as did fibroblasts and muscle cells. Epithelial cells exposed to buffered, but not denatured, gastric juice produced IL-6. Undernatants of mucosal biopsy cultures from esophagitis patients reduced esophageal muscle contraction, as did supernatants from esophageal epithelial cell cultures. CONCLUSIONS: The human esophagus produces cytokines capable of reducing contractility of esophageal muscle cells. Exposure to gastric juice is sufficient to stimulate esophageal epithelial cells to produce IL-6, a cytokine able to alter esophageal contractility. These results indicate that classic cytokines are important mediators of the motor disturbances associated with human esophageal inflammation.


Assuntos
Transtornos da Motilidade Esofágica/imunologia , Transtornos da Motilidade Esofágica/patologia , Refluxo Gastroesofágico/imunologia , Refluxo Gastroesofágico/patologia , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Animais , Biópsia , Gatos , Linhagem Celular , Esofagite/imunologia , Esofagite/patologia , Esôfago/citologia , Suco Gástrico , Humanos , Interleucina-1beta/genética , Interleucina-6/genética , Queratinócitos/citologia , Queratinócitos/metabolismo , Mucosa/imunologia , Mucosa/patologia , Contração Muscular , Músculo Liso/fisiologia , Técnicas de Cultura de Órgãos , Reação em Cadeia da Polimerase , RNA Mensageiro/metabolismo
20.
Gastroenterology ; 130(7): 2060-73, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16762629

RESUMO

BACKGROUND & AIMS: Angiogenesis is a critical component of neoplastic and chronic inflammatory disorders, but whether angiogenesis also occurs in inflammatory bowel disease (IBD) has yet to be established. We assessed mucosal vascularization, expression of endothelial alphaVbeta3 integrin, angiogenic factors, and their bioactivity in Crohn's disease (CD) and ulcerative colitis (UC) mucosa. METHODS: Mucosal endothelium was immunostained for CD31 and factor VIII and quantified by digital morphometry. alphaVbeta3 expression was studied in vivo by confocal microscopy and in vitro by flow cytometric analysis of human intestinal microvascular endothelial cells (HIMECs). Vascular endothelial growth factor (VEGF), interleukin (IL)-8, and bFGF levels were measured in mucosal extracts and cells and angiogenic bioactivity shown by induction of HIMEC migration and the corneal and chorioallantoic membrane angiogenesis assays. RESULTS: Microvessel density was increased in IBD mucosa. Endothelial alphaVbeta3 was strongly expressed in IBD but only sporadically in normal mucosa and was up-regulated in HIMECs by VEGF, tumor necrosis factor alpha, and bFGF. IBD mucosal extracts induced a significantly higher degree of HIMEC migration than control mucosa, and this response was mostly dependent on IL-8 and less on basic fibroblast growth factor or vascular endothelial growth factor. Compared with normal mucosa, IBD mucosal extracts induced a potent angiogenic response in both the corneal and chorioallantoic membrane assays. CONCLUSIONS: These results provide morphological, phenotypic and functional evidence of potent angiogenic activity in both CD and UC mucosa, indicating that the local microvasculature undergoes an intense process of inflammation-dependent angiogenesis. Thus, angiogenesis appears to be an integral component of IBD pathogenesis, providing the practical and conceptual framework for anti-angiogenic therapies in IBD.


Assuntos
Mediadores da Inflamação/análise , Doenças Inflamatórias Intestinais/patologia , Mucosa Intestinal/citologia , Neovascularização Patológica/patologia , Biópsia por Agulha , Estudos de Casos e Controles , Células Cultivadas , Quimiotaxia , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Feminino , Fator 2 de Crescimento de Fibroblastos/análise , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Doenças Inflamatórias Intestinais/etiologia , Integrinas/análise , Interleucina-8/análise , Mucosa Intestinal/irrigação sanguínea , Mucosa Intestinal/patologia , Masculino , Microcirculação/fisiologia , Estudos de Amostragem , Sensibilidade e Especificidade , Fator A de Crescimento do Endotélio Vascular/análise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA