Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell ; 186(18): 3793-3809.e26, 2023 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-37562401

RESUMO

Hepatocytes, the major metabolic hub of the body, execute functions that are human-specific, altered in human disease, and currently thought to be regulated through endocrine and cell-autonomous mechanisms. Here, we show that key metabolic functions of human hepatocytes are controlled by non-parenchymal cells (NPCs) in their microenvironment. We developed mice bearing human hepatic tissue composed of human hepatocytes and NPCs, including human immune, endothelial, and stellate cells. Humanized livers reproduce human liver architecture, perform vital human-specific metabolic/homeostatic processes, and model human pathologies, including fibrosis and non-alcoholic fatty liver disease (NAFLD). Leveraging species mismatch and lipidomics, we demonstrate that human NPCs control metabolic functions of human hepatocytes in a paracrine manner. Mechanistically, we uncover a species-specific interaction whereby WNT2 secreted by sinusoidal endothelial cells controls cholesterol uptake and bile acid conjugation in hepatocytes through receptor FZD5. These results reveal the essential microenvironmental regulation of hepatic metabolism and its human-specific aspects.


Assuntos
Células Endoteliais , Fígado , Animais , Humanos , Camundongos , Células Endoteliais/metabolismo , Hepatócitos/metabolismo , Células de Kupffer/metabolismo , Fígado/citologia , Fígado/metabolismo , Hepatopatia Gordurosa não Alcoólica/metabolismo , Fibrose/metabolismo
2.
Proc Natl Acad Sci U S A ; 119(33): e2203318119, 2022 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-35939687

RESUMO

γδ T cells are an abundant T cell population at the mucosa and are important in providing immune surveillance as well as maintaining tissue homeostasis. However, despite γδ T cells' origin in the thymus, detailed mechanisms regulating γδ T cell development remain poorly understood. N6-methyladenosine (m6A) represents one of the most common posttranscriptional modifications of messenger RNA (mRNA) in mammalian cells, but whether it plays a role in γδ T cell biology is still unclear. Here, we show that depletion of the m6A demethylase ALKBH5 in lymphocytes specifically induces an expansion of γδ T cells, which confers enhanced protection against gastrointestinal Salmonella typhimurium infection. Mechanistically, loss of ALKBH5 favors the development of γδ T cell precursors by increasing the abundance of m6A RNA modification in thymocytes, which further reduces the expression of several target genes including Notch signaling components Jagged1 and Notch2. As a result, impairment of Jagged1/Notch2 signaling contributes to enhanced proliferation and differentiation of γδ T cell precursors, leading to an expanded mature γδ T cell repertoire. Taken together, our results indicate a checkpoint role of ALKBH5 and m6A modification in the regulation of γδ T cell early development.


Assuntos
Homólogo AlkB 5 da RNA Desmetilase , Linfócitos Intraepiteliais , RNA Mensageiro , Homólogo AlkB 5 da RNA Desmetilase/genética , Homólogo AlkB 5 da RNA Desmetilase/metabolismo , Animais , Linfócitos Intraepiteliais/enzimologia , Linfócitos Intraepiteliais/imunologia , Proteína Jagged-1/metabolismo , Camundongos , Camundongos Knockout , RNA Mensageiro/metabolismo , Receptor Notch2/metabolismo , Transdução de Sinais/genética
3.
Sci Adv ; 8(12): eabl5723, 2022 03 25.
Artigo em Inglês | MEDLINE | ID: mdl-35333576

RESUMO

Colonic mucosal barrier dysfunction is one of the major causes of inflammatory bowel disease (IBD). However, the mechanisms underlying mucosal barrier dysfunction are poorly understood. N6-methyladenosine (m6A) mRNA modification is an important modulator of epitranscriptional regulation of gene expression, participating in multiple physiological and pathological processes. However, the function of m6A modification in colonic epithelial cells and stem cells is unknown. Here, we show that m6A modification is essential for maintaining the homeostatic self-renewal in colonic stem cells. Specific deletion of the methyltransferase 14 (Mettl14) gene in mouse colon resulted in colonic stem cell apoptosis, causing mucosal barrier dysfunction and severe colitis. Mechanistically, we revealed that Mettl14 restricted colonic epithelial cell death by regulating the stability of Nfkbia mRNA and modulating the NF-κB pathway. Our results identified a previously unidentified role for m6A modification in colonic epithelial cells and stem cells, suggesting that m6A modification may be a potential therapeutic target for IBD.


Assuntos
Colo , NF-kappa B , Animais , Apoptose/genética , Colo/metabolismo , Colo/patologia , Células Epiteliais/metabolismo , Homeostase , Camundongos , NF-kappa B/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
4.
Nat Commun ; 13(1): 192, 2022 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-35017482

RESUMO

A key challenge in analyzing single cell RNA-sequencing data is the large number of false zeros, where genes actually expressed in a given cell are incorrectly measured as unexpressed. We present a method based on low-rank matrix approximation which imputes these values while preserving biologically non-expressed genes (true biological zeros) at zero expression levels. We provide theoretical justification for this denoising approach and demonstrate its advantages relative to other methods on simulated and biological datasets.


Assuntos
Algoritmos , RNA/genética , Análise de Sequência de RNA/estatística & dados numéricos , Animais , Linfócitos B/citologia , Linfócitos B/metabolismo , Brônquios/citologia , Brônquios/metabolismo , Conjuntos de Dados como Assunto , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Humanos , Células Matadoras Naturais/citologia , Células Matadoras Naturais/metabolismo , Camundongos , Monócitos/citologia , Monócitos/metabolismo , Cultura Primária de Células , RNA/metabolismo , RNA-Seq , Análise de Célula Única , Linfócitos T/citologia , Linfócitos T/metabolismo
5.
Nature ; 592(7855): 606-610, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33658717

RESUMO

Intestinal stromal cells are known to modulate the propagation and differentiation of intestinal stem cells1,2. However, the precise cellular and molecular mechanisms by which this diverse stromal cell population maintains tissue homeostasis and repair are poorly understood. Here we describe a subset of intestinal stromal cells, named MAP3K2-regulated intestinal stromal cells (MRISCs), and show that they are the primary cellular source of the WNT agonist R-spondin 1 following intestinal injury in mice. MRISCs, which are epigenetically and transcriptomically distinct from subsets of intestinal stromal cells that have previously been reported3-6, are strategically localized at the bases of colon crypts, and function to maintain LGR5+ intestinal stem cells and protect against acute intestinal damage through enhanced R-spondin 1 production. Mechanistically, this MAP3K2 specific function is mediated by a previously unknown reactive oxygen species (ROS)-MAP3K2-ERK5-KLF2 axis to enhance production of R-spondin 1. Our results identify MRISCs as a key component of an intestinal stem cell niche that specifically depends on MAP3K2 to augment WNT signalling for the regeneration of damaged intestine.


Assuntos
Mucosa Intestinal/citologia , MAP Quinase Quinase Quinase 2/metabolismo , Nicho de Células-Tronco , Células Estromais/citologia , Animais , Antígenos CD34 , Colite/patologia , Colite/prevenção & controle , Epigênese Genética , Feminino , Mucosa Intestinal/patologia , Fatores de Transcrição Kruppel-Like/metabolismo , Masculino , Camundongos , Espécies Reativas de Oxigênio/metabolismo , Tetraspanina 28 , Trombospondinas/biossíntese , Trombospondinas/metabolismo , Antígenos Thy-1
6.
Nature ; 580(7804): 524-529, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32322056

RESUMO

The initiation of an intestinal tumour is a probabilistic process that depends on the competition between mutant and normal epithelial stem cells in crypts1. Intestinal stem cells are closely associated with a diverse but poorly characterized network of mesenchymal cell types2,3. However, whether the physiological mesenchymal microenvironment of mutant stem cells affects tumour initiation remains unknown. Here we provide in vivo evidence that the mesenchymal niche controls tumour initiation in trans. By characterizing the heterogeneity of the intestinal mesenchyme using single-cell RNA-sequencing analysis, we identified a population of rare pericryptal Ptgs2-expressing fibroblasts that constitutively process arachidonic acid into highly labile prostaglandin E2 (PGE2). Specific ablation of Ptgs2 in fibroblasts was sufficient to prevent tumour initiation in two different models of sporadic, autochthonous tumorigenesis. Mechanistically, single-cell RNA-sequencing analyses of a mesenchymal niche model showed that fibroblast-derived PGE2 drives the expansion οf a population of Sca-1+ reserve-like stem cells. These express a strong regenerative/tumorigenic program, driven by the Hippo pathway effector Yap. In vivo, Yap is indispensable for Sca-1+ cell expansion and early tumour initiation and displays a nuclear localization in both mouse and human adenomas. Using organoid experiments, we identified a molecular mechanism whereby PGE2 promotes Yap dephosphorylation, nuclear translocation and transcriptional activity by signalling through the receptor Ptger4. Epithelial-specific ablation of Ptger4 misdirected the regenerative reprogramming of stem cells and prevented Sca-1+ cell expansion and sporadic tumour initiation in mutant mice, thereby demonstrating the robust paracrine control of tumour-initiating stem cells by PGE2-Ptger4. Analyses of patient-derived organoids established that PGE2-PTGER4 also regulates stem-cell function in humans. Our study demonstrates that initiation of colorectal cancer is orchestrated by the mesenchymal niche and reveals a mechanism by which rare pericryptal Ptgs2-expressing fibroblasts exert paracrine control over tumour-initiating stem cells via the druggable PGE2-Ptger4-Yap signalling axis.


Assuntos
Carcinogênese , Neoplasias Colorretais/patologia , Intestinos/patologia , Mesoderma/patologia , Células-Tronco Neoplásicas/patologia , Comunicação Parácrina , Nicho de Células-Tronco , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Antígenos Ly/metabolismo , Ácido Araquidônico/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proliferação de Células , Neoplasias Colorretais/metabolismo , Ciclo-Oxigenase 2/metabolismo , Dinoprostona/metabolismo , Feminino , Fibroblastos/metabolismo , Fibroblastos/patologia , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Masculino , Proteínas de Membrana/metabolismo , Mesoderma/metabolismo , Camundongos , Células-Tronco Neoplásicas/metabolismo , Organoides/metabolismo , Organoides/patologia , Receptores de Prostaglandina E Subtipo EP4/metabolismo , Análise de Célula Única , Proteínas de Sinalização YAP
8.
Cell ; 180(1): 50-63.e12, 2020 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-31923399

RESUMO

Mucosal barrier immunity is essential for the maintenance of the commensal microflora and combating invasive bacterial infection. Although immune and epithelial cells are thought to be the canonical orchestrators of this complex equilibrium, here, we show that the enteric nervous system (ENS) plays an essential and non-redundant role in governing the antimicrobial protein (AMP) response. Using confocal microscopy and single-molecule fluorescence in situ mRNA hybridization (smFISH) studies, we observed that intestinal neurons produce the pleiotropic cytokine IL-18. Strikingly, deletion of IL-18 from the enteric neurons alone, but not immune or epithelial cells, rendered mice susceptible to invasive Salmonella typhimurium (S.t.) infection. Mechanistically, unbiased RNA sequencing and single-cell sequencing revealed that enteric neuronal IL-18 is specifically required for homeostatic goblet cell AMP production. Together, we show that neuron-derived IL-18 signaling controls tissue-wide intestinal immunity and has profound consequences on the mucosal barrier and invasive bacterial killing.


Assuntos
Imunidade nas Mucosas/imunologia , Interleucina-18/imunologia , Mucosa Intestinal/imunologia , Animais , Citocinas/imunologia , Sistema Nervoso Entérico/imunologia , Sistema Nervoso Entérico/metabolismo , Células Epiteliais/imunologia , Feminino , Células Caliciformes/imunologia , Interleucina-18/biossíntese , Mucosa Intestinal/metabolismo , Intestino Delgado/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/imunologia , Ratos , Ratos Sprague-Dawley , Infecções por Salmonella/imunologia , Salmonella typhimurium/imunologia , Transdução de Sinais/imunologia
9.
Sci Rep ; 7(1): 2397, 2017 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-28546545

RESUMO

The human growth hormone (hGH) minigene used for transgene stabilization in mice has been recently identified to be locally expressed in the tissues where transgenes are active and associated with phenotypic alterations. Here we extend these findings by analyzing the effect of the hGH minigene in TgC6hp55 transgenic mice which express the human TNFR1 under the control of the mesenchymal cell-specific CollagenVI promoter. These mice displayed a fully penetrant phenotype characterized by growth enhancement accompanied by perturbations in metabolic, skeletal, histological and other physiological parameters. Notably, this phenotype was independent of TNF-TNFR1 signaling since the genetic ablation of either Tnf or Tradd did not rescue the phenotype. Further analyses showed that the hGH minigene was expressed in several tissues, also leading to increased hGH protein levels in the serum. Pharmacological blockade of GH signaling prevented the development of the phenotype. Our results indicate that the unplanned expression of the hGH minigene in CollagenVI expressing mesenchymal cells can lead through local and/or systemic mechanisms to enhanced somatic growth followed by a plethora of primary and/or secondary effects such as hyperphagia, hypermetabolism, disturbed glucose homeostasis, altered hematological parameters, increased bone formation and lipid accumulation in metabolically critical tissues.


Assuntos
Expressão Gênica , Hormônio do Crescimento Humano/genética , Fenótipo , Transgenes , Animais , Colágeno Tipo VI/genética , Feminino , Regulação da Expressão Gênica , Glucose/metabolismo , Hormônio do Crescimento Humano/metabolismo , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Regiões Promotoras Genéticas , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo
10.
Differentiation ; 92(3): 116-131, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27165847

RESUMO

In this Review we summarize our current understanding of the biology of mesenchymal cells of the intestinal lamina propria focusing mainly on fibroblasts and myofibroblasts. The topics covered include 1) the embryonic origin of mesenchymal cells of the intestinal lamina propria and their heterogeneity in adults, 2) the role of the mesenchyme in intestinal development, 3) the physiological function of fibroblasts and myofibroblasts in adults as part of the intestinal stem cell niche and the mucosal immune system and 4) the involvement of fibroblasts and myofibroblasts in epithelial homeostasis upon injury and in the pathogenesis of diseases such as Inflammatory Bowel Diseases, fibrosis and cancer. We emphasize studies addressing the function of intestinal mesenchymal cells in vivo, and also discuss major open questions and current challenges in this field.


Assuntos
Fibroblastos/citologia , Mucosa Intestinal/patologia , Intestinos/patologia , Miofibroblastos/citologia , Células Estromais/citologia , Animais , Humanos , Mucosa Intestinal/fisiopatologia , Intestinos/fisiopatologia , Mesoderma/metabolismo
11.
Proc Natl Acad Sci U S A ; 111(43): E4658-67, 2014 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-25316791

RESUMO

Tumor progression locus-2 (Tpl2) kinase is a major inflammatory mediator in immune cell types recently found to be genetically associated with inflammatory bowel diseases (IBDs). Here we show that Tpl2 may exert a dominant homeostatic rather than inflammatory function in the intestine mediated specifically by subepithelial intestinal myofibroblasts (IMFs). Mice with complete or IMF-specific Tpl2 ablation are highly susceptible to epithelial injury-induced colitis showing impaired compensatory proliferation in crypts and extensive ulcerations without significant changes in inflammatory responses. Following epithelial injury, IMFs sense innate or inflammatory signals and activate, via Tpl2, the cyclooxygenase-2 (Cox-2)-prostaglandin E2 (PGE2) pathway, which we show here to be essential for the epithelial homeostatic response. Exogenous PGE2 administration rescues mice with complete or IMF-specific Tpl2 ablation from defects in crypt function and susceptibility to colitis. We also show that Tpl2 expression is decreased in IMFs isolated from the inflamed ileum of IBD patients indicating that Tpl2 function in IMFs may be highly relevant to human disease. The IMF-mediated mechanism we propose also involves the IBD-associated genes IL1R1, MAPK1, and the PGE2 receptor-encoding PTGER4. Our results establish a previously unidentified myofibroblast-specific innate pathway that regulates intestinal homeostasis and may underlie IBD susceptibility in humans.


Assuntos
Ciclo-Oxigenase 2/metabolismo , Dinoprostona/metabolismo , Epitélio/metabolismo , Homeostase , Intestinos/patologia , MAP Quinase Quinase Quinases/metabolismo , Miofibroblastos/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Animais , Linhagem da Célula , Proliferação de Células/efeitos dos fármacos , Colite/enzimologia , Colite/imunologia , Colite/patologia , Sulfato de Dextrana , Dinoprostona/administração & dosagem , Dinoprostona/farmacologia , Suscetibilidade a Doenças , Ativação Enzimática/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Epitélio/patologia , Homeostase/efeitos dos fármacos , Humanos , Imunidade Inata/efeitos dos fármacos , Inflamação/patologia , Doenças Inflamatórias Intestinais/enzimologia , Doenças Inflamatórias Intestinais/patologia , MAP Quinase Quinase Quinases/deficiência , Camundongos Endogâmicos C57BL , Modelos Biológicos , Miofibroblastos/efeitos dos fármacos , Miofibroblastos/enzimologia , Miofibroblastos/patologia , Fenótipo , Proteínas Proto-Oncogênicas/deficiência , Transdução de Sinais/efeitos dos fármacos
12.
Int Immunol ; 26(9): 509-15, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24821262

RESUMO

An important function of intestinal epithelial cells (IECs) is to maintain the integrity of the mucosal barrier. Inflammation challenges the integrity of the mucosal barrier and the intestinal epithelium needs to adapt to a multitude of signals in order to perform the complex process of maintenance and restitution of its barrier function. Dysfunctions in epithelial barrier integrity and restoration contribute to the pathogenesis of inflammatory bowel diseases (IBDs) such as Crohn's disease and ulcerative colitis. Mucosal healing has developed to a significant treatment goal in IBD. In this review, we would like to highlight physiologic and pathologic adaptations of the intestinal epithelium to inflammation, exemplified by its responses to TNF-α. A large body of literature exists that highlights the diverse effects of this cytokine on IECs. TNF-α modulates intestinal mucus secretion and constitution. TNF-α stimulation modulates paracellular flow via tight junctional control. TNF-α induces intracellular signaling cascades that determine significant cell fate decisions such as survival, cell death or proliferation. TNF-α impacts epithelial wound healing in ErbB- and Wnt-dependent pathways while also importantly guiding immune cell attraction and function. We selected important studies from recent years with a focus on functional in vivo data providing crucial insights into the complex process of intestinal homeostasis.


Assuntos
Imunidade nas Mucosas , Doenças Inflamatórias Intestinais/imunologia , Mucosa Intestinal/imunologia , Fator de Necrose Tumoral alfa/imunologia , Via de Sinalização Wnt/imunologia , Animais , Receptores ErbB/imunologia , Humanos , Inflamação/imunologia , Inflamação/patologia , Doenças Inflamatórias Intestinais/patologia , Mucosa Intestinal/patologia , Proteínas Wnt/imunologia
13.
J Clin Invest ; 122(11): 4231-42, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23064365

RESUMO

The tumor microenvironment plays a significant role in colitis-associated cancer (CAC). Intestinal myofibroblasts (IMFs) are cells in the intestinal lamina propria secreting factors that are known to modulate carcinogenesis; however, the physiological role of IMFs and signaling pathways influencing CAC have remained unknown. Tumor progression locus 2 (Tpl2) is a MAPK that regulates inflammatory and oncogenic pathways. In this study we addressed the role of Tpl2 in CAC using complete and tissue-specific ablation of Tpl2 in mutant mice. Tpl2-deficient mice did not exhibit significant differences in inflammatory burdens following azoxymethane (AOM)/dextran sodium sulfate (DSS) administration compared with wild-type mice; however, the mutant mice developed significantly increased numbers and sizes of tumors, associated with enhanced epithelial proliferation and decreased apoptosis. Cell-specific ablation of Tpl2 in IMFs, but not in intestinal epithelial or myeloid cells, conferred a similar susceptibility to adenocarcinoma formation. Tpl2-deficient IMFs upregulated HGF production and became less sensitive to the negative regulation of HGF by TGF-ß3. In vivo inhibition of HGF-mediated c-Met activation blocked early, enhanced colon dysplasia in Tpl2-deficient mice, indicating that Tpl2 normally suppresses the HGF/c-Met pathway. These findings establish a mesenchyme-specific role for Tpl2 in the regulation of HGF production and suppression of epithelial tumorigenesis.


Assuntos
Adenocarcinoma/metabolismo , Transformação Celular Neoplásica/metabolismo , Colite/metabolismo , Fator de Crescimento de Hepatócito/metabolismo , Mucosa Intestinal/metabolismo , Neoplasias Intestinais/metabolismo , MAP Quinase Quinase Quinases/metabolismo , Miofibroblastos/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Transdução de Sinais , Adenocarcinoma/etiologia , Adenocarcinoma/genética , Adenocarcinoma/patologia , Animais , Azoximetano/efeitos adversos , Azoximetano/farmacologia , Carcinógenos/farmacologia , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Colite/induzido quimicamente , Colite/complicações , Colite/genética , Colite/patologia , Sulfato de Dextrana/efeitos adversos , Sulfato de Dextrana/farmacologia , Fator de Crescimento de Hepatócito/genética , Neoplasias Intestinais/etiologia , Neoplasias Intestinais/genética , Neoplasias Intestinais/patologia , Intestinos/patologia , MAP Quinase Quinase Quinases/genética , Camundongos , Camundongos Knockout , Mucosa/metabolismo , Mucosa/patologia , Miofibroblastos/patologia , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-met/genética , Proteínas Proto-Oncogênicas c-met/metabolismo
14.
Proc Natl Acad Sci U S A ; 108(13): 5396-401, 2011 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-21402942

RESUMO

TNF plays a crucial role in the pathogenesis of Crohn disease. Dysregulated TNF production in mice that bear the genetic deletion of the TNF AU-rich regulatory elements (ARE) (Tnf(ΔARE/+) mice) results in TNF receptor I (TNFRI)-dependent spontaneous Crohn-like pathology. Current concepts consider intestinal epithelial cell (IEC) responses to TNF to be critical for intestinal pathology, but the potential contribution of IEC-derived TNF in disease pathogenesis has not been addressed. In this study we examined whether IEC are sufficient as cellular targets or sources of TNF in the development of intestinal pathology. Using IEC-specific reactivation of a hypomorphic Tnf(ΔAREneo) allele in mice, we show that selective chronic overproduction of TNF by IEC suffices to cause full development of Crohn-like pathology. Epithelial TNF overexpression leads to early activation of the underlying intestinal myofibroblast, a cell type previously identified as a sufficient target of TNF for disease development in the Tnf(ΔARE) model. By contrast, restricted TNFRI expression on IEC although sufficient to confer IEC apoptosis after acute exogenous TNF administration, fails to induce pathology following chronic specific targeting of IEC by endogenous TNF in Tnf(ΔARE/+) mice. Our results argue against IEC being early and sufficient responders to chronic TNF-mediated pathogenic signals and suggest that proinflammatory aberrations leading to chronic TNF production by IEC may initiate pathology in Crohn disease.


Assuntos
Doença de Crohn/patologia , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Intestinos/citologia , Intestinos/patologia , Fator de Necrose Tumoral alfa/metabolismo , Animais , Doença de Crohn/metabolismo , Células Epiteliais/citologia , Humanos , Inflamação/metabolismo , Inflamação/patologia , Mucosa Intestinal/metabolismo , Mesoderma/citologia , Mesoderma/metabolismo , Camundongos , Camundongos Transgênicos , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Fator de Necrose Tumoral alfa/genética
15.
Gastroenterology ; 134(7): 2025-35, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18439426

RESUMO

BACKGROUND & AIMS: In the present work, we address the requirement for intestinal-specific homing molecules, the chemokine/chemokine receptor pair CCL25/CCR9 and beta7 integrin, in the pathogenesis of the CD8(+) T cell-dependent Tnf(DeltaARE) mouse model of Crohn's-like inflammatory bowel disease. METHODS: We investigated by flow cytometry lymphocyte recruitment in the intestinal epithelium and lamina propria (LP); cytokine production by intraepithelial and LP lymphocytes; and peripheral expression of CCR9, alpha4beta7, and alphaEbeta7 integrin. The functional significance of CCL25/CCR9 and beta7 integrin in inflammatory lymphocyte recruitment and intestinal disease development was assessed in Tnf(DeltaARE) mice genetically lacking these molecules. RESULTS: Intestinal inflammation in the Tnf(DeltaARE) mice is associated with early reduction of CD8alphaalpha-expressing intraepithelial lymphocytes, decreased T helper cell 1 and increased T helper cell 17 responses by LP CD4(+) lymphocytes, increased alphaEbeta7 integrin expression in peripheral activated/memory intestinal-homing CD8alphabeta lymphocytes, and predominance of tumor necrosis factor/interferon-gamma-producing CD8alphabeta lymphocytes in the epithelium. Although CCL25/CCR9 have been strongly implicated in T-lymphocyte recruitment to the small intestine, inflammatory pathology develops unperturbed in the genetic absence of CCL25/CCR9. Furthermore, CD8alphabeta lymphocyte recruitment in the intestinal epithelium and inflammatory infiltration in the LP are not impaired in CCR9- or CCL25-deficient Tnf(DeltaARE) mice. In contrast, genetic ablation of beta7 integrin results in complete amelioration of intestinal pathology. CONCLUSIONS: Our findings demonstrate that development of intestinal inflammation in the Tnf(DeltaARE) mice is critically dependent on beta7 integrin-mediated T-lymphocyte recruitment, whereas the function of the CCL25/CCR9 axis appears dispensable in this model.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Quimiocinas CC/metabolismo , Quimiotaxia de Leucócito , Colo/imunologia , Doença de Crohn/imunologia , Cadeias beta de Integrinas/metabolismo , Receptores CCR/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Antígenos CD8/metabolismo , Quimiocinas CC/genética , Colo/patologia , Doença de Crohn/genética , Doença de Crohn/patologia , Modelos Animais de Doenças , Citometria de Fluxo , Cadeias beta de Integrinas/genética , Integrinas/metabolismo , Interferon gama/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Receptores CCR/genética , Linfócitos T Auxiliares-Indutores/imunologia , Fatores de Tempo , Fator de Necrose Tumoral alfa/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...