Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 170
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 120(4): e2218162120, 2023 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-36669099

RESUMO

Resolution of inflammation and mucosal wound healing are crucial processes required to re-establish homeostasis following injury of mucosal tissues. Maresin-2 (MaR2), a lipid specialized pro-resolving mediator derived from omega-3 polyunsaturated fatty acid, has been reported to promote resolution of inflammation. However, a potential role for MaR2 in regulating mucosal repair remains undefined. Using lipidomic analyses, we demonstrate biosynthesis of MaR2 in healing intestinal mucosal wounds in vivo. Importantly, administration of exogenous MaR2 promoted mucosal repair following dextran sulfate sodium-induced colitis or biopsy-induced colonic mucosal injury. Functional analyses revealed that MaR2 promotes mucosal wound repair by driving intestinal epithelial migration through activation of focal cell-matrix adhesion signaling in primary human intestinal epithelial cells. Because of its labile nature, MaR2 is easily degradable and requires ultracold storage to maintain functionality. Thus, we created thermostable polylactic acid MaR2 nanoparticles that retain biological activity following extended storage at 4 °C or above. Taken together, these results establish MaR2 as a potent pro-repair lipid mediator with broad therapeutic potential for use in promoting mucosal repair in inflammatory diseases.


Assuntos
Colite , Nanopartículas , Humanos , Colite/induzido quimicamente , Colite/tratamento farmacológico , Intestinos , Mucosa Intestinal/fisiologia , Inflamação , Sulfato de Dextrana/efeitos adversos
2.
Gastroenterology ; 166(1): 103-116.e9, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37716376

RESUMO

BACKGROUND & AIMS: CXADR-like membrane protein (CLMP) is structurally related to coxsackie and adenovirus receptor. Pathogenic variants in CLMP gene have been associated with congenital short bowel syndrome, implying a role for CLMP in intestinal development. However, the contribution of CLMP to regulating gut development and homeostasis is unknown. METHODS: In this study, we investigated CLMP function in the colonic epithelium using complementary in vivo and in vitro approaches, including mice with inducible intestinal epithelial cell (IEC)-specific deletion of CLMP (ClmpΔIEC), intestinal organoids, IECs with overexpression, or loss of CLMP and RNA sequencing data from individuals with colorectal cancer. RESULTS: Loss of CLMP enhanced IEC proliferation and, conversely, CLMP overexpression reduced proliferation. Xenograft experiments revealed increased tumor growth in mice implanted with CLMP-deficient colonic tumor cells, and poor engraftment was observed with CLMP-overexpressing cells. ClmpΔIEC mice showed exacerbated tumor burden in an azoxymethane and dextran sulfate sodium-induced colonic tumorigenesis model, and CLMP expression was reduced in human colorectal cancer samples. Mechanistic studies revealed that CLMP-dependent regulation of IEC proliferation is linked to signaling through mTOR-Akt-ß-catenin pathways. CONCLUSIONS: These results reveal novel insights into CLMP function in the colonic epithelium, highlighting an important role in regulating IEC proliferation, suggesting tumor suppressive function in colon cancer.


Assuntos
Colite , Neoplasias do Colo , Animais , Humanos , Camundongos , Proliferação de Células , Colite/induzido quimicamente , Colite/metabolismo , Neoplasias do Colo/patologia , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus , Células Epiteliais/patologia , Mucosa Intestinal/patologia , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo
3.
Proc Natl Acad Sci U S A ; 117(17): 9477-9482, 2020 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-32300016

RESUMO

Resolution of intestinal inflammation and wound repair are active processes that mediate epithelial healing at mucosal surfaces. Lipid molecules referred to as specialized proresolving mediators (SPMs) play an important role in the restorative response. Resolvin E1 (RvE1), a SPM derived from omega-3 fatty acids, has been reported to dampen intestinal inflammation by promoting anti-inflammatory responses including increased neutrophil spherocytosis and macrophage production of IL-10. Despite these observations, a role for RvE1 in regulating intestinal epithelial cell migration and proliferation during mucosal wound repair has not been explored. Using an endoscopic biopsy-based wound healing model, we report that RvE1 is locally produced in response to intestinal mucosal injury. Exposure of intestinal epithelial cells to RvE1 promoted wound repair by increasing cellular proliferation and migration through activation of signaling pathways including CREB, mTOR, and Src-FAK. Additionally, RvE1-triggered activation of the small GTPase Rac1 led to increased intracellular reactive oxygen species (ROS) production, cell-matrix adhesion, and cellular protrusions at the leading edge of migrating cells. Furthermore, in situ administration of RvE1-encapsulated synthetic targeted polymeric nanoparticles into intestinal wounds promoted mucosal repair. Together, these findings demonstrate that RvE1 functions as a prorepair lipid mediator by increasing intestinal epithelial cell migration and proliferation, and highlight potential therapeutic applications for this SPM to promote mucosal healing in the intestine.


Assuntos
Ácido Eicosapentaenoico/análogos & derivados , Mucosa Intestinal/metabolismo , Cicatrização/fisiologia , Animais , Adesão Celular , Linhagem Celular , Colo , Ácido Eicosapentaenoico/metabolismo , Ácido Eicosapentaenoico/farmacologia , Humanos , Mucosa Intestinal/patologia , Camundongos , Camundongos Endogâmicos C57BL , Nanopartículas , Neuropeptídeos , Organoides , Espécies Reativas de Oxigênio , Proteínas rac1 de Ligação ao GTP
4.
Trans Am Clin Climatol Assoc ; 132: 22-33, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36196189

RESUMO

Neutrophils (PMNs) play a critical role in innate immunity, yet many pathologic conditions are associated with dysregulated infiltration of PMNs into tissues. In the gut, robust PMN accumulation and migration across the intestinal epithelium closely correlates with clinical symptoms in conditions such as ulcerative colitis. While much is known about how PMNs migrate out of blood vessels, far less is understood about how PMNs traverse epithelial barriers. Until fairly recently, in vitro models of PMN transepithelial migration (TEpM) across cultured intestinal epithelial cell lines provided many of the insights into the molecular basis of TEpM. However, innovative animal models have provided new avenues for investigating in vivo mechanisms regulating PMN TEpM. This report will highlight molecular insights gained from studies on PMN TEpM and provide a rationale for developing tissue targeted strategies directed at reducing pathologic consequences of dysregulated PMN trafficking in the gut.


Assuntos
Neutrófilos , Migração Transendotelial e Transepitelial , Animais , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia
5.
Am J Pathol ; 190(10): 2029-2038, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32958140

RESUMO

N-formyl peptide receptors (FPRs) serve as phagocyte pattern-recognition receptors that play a crucial role in the regulation of host defense against infection. Epithelial cells also express FPRs, and their activation during inflammation or injury results in enhanced epithelial migration and proliferation and improved mucosal wound repair. However, signaling mechanisms that govern epithelial FPR1 activity are not well understood. This study identified a novel FPR1-interacting protein, WD40 repeat protein (WDR)-26, which negatively regulates FPR1-mediated wound healing in intestinal epithelial cells. We show that WDR26-mediated inhibition of wound repair is mediated through the inhibition of Rac family small GTPase 1 and cell division cycle 42 activation, as well as downstream intracellular reactive oxygen species production. Furthermore, on FPR1 activation with N-formyl-methionyl-leucyl phenylalanine, WDR26 dissociates from FPR1, resulting in the activation of downstream cell division cycle 42/Rac family small GTPase 1 signaling, increased epithelial cell migration, and mucosal wound repair. These findings elucidate a novel regulatory function of WDR26 in FPR1-mediated wound healing in intestinal epithelial cells.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Células Epiteliais/metabolismo , Intestinos/patologia , Cicatrização/fisiologia , Movimento Celular/fisiologia , Humanos , Mucosa Intestinal/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Receptores de Formil Peptídeo/metabolismo
6.
FASEB J ; 34(2): 2326-2343, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31907993

RESUMO

Polymorphonuclear neutrophils (PMNs) play a critical role in the innate immune response to invading pathogens. However, dysregulated mucosal trafficking of PMNs and associated epithelial tissue damage is a pathological hallmark of numerous inflammatory conditions including inflammatory bowel disease. The glycoprotein CD11b/CD18 plays a well-described role in regulating PMN transepithelial migration and PMN inflammatory functions. Previous studies have demonstrated that targeting of the N-linked glycan Lewis X on CD11b blocks PMN transepithelial migration (TEpM). Given evidence of glycosylation-dependent regulation of CD11b/CD18 function, we performed MALDI TOF Mass Spectrometry (MS) analyses on CD11b/CD18 purified from human PMNs. Unusual glycan epitopes identified on CD11b/CD18 included high Mannose oligosaccharides recognized by the Galanthus Nivalis lectin and biantennary galactosylated N-glycans recognized by the Phaseolus Vulgaris erythroagglutinin lectin. Importantly, we show that selective targeting of glycans on CD11b with such lectins results in altered intracellular signaling events that inhibit TEpM and differentially affect key PMN inflammatory functions including phagocytosis, superoxide release and apoptosis. Taken together, these data demonstrate that discrete glycan motifs expressed on CD11b/CD18 such as biantennary galactose could represent novel targets for selective manipulation of CD11b function and reduction of PMN-associated tissue damage in chronic inflammatory diseases.


Assuntos
Antígeno CD11b/imunologia , Antígenos CD18/imunologia , Epitopos/imunologia , Neutrófilos/imunologia , Antígeno CD11b/química , Antígenos CD18/química , Epitopos/química , Humanos , Lectinas de Ligação a Manose/química , Lectinas de Ligação a Manose/farmacologia , Neutrófilos/química , Fagocitose , Fito-Hemaglutininas/química , Fito-Hemaglutininas/farmacologia , Lectinas de Plantas/química , Lectinas de Plantas/farmacologia , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Superóxidos/química , Superóxidos/imunologia , Migração Transendotelial e Transepitelial
7.
FASEB J ; 34(5): 7089-7102, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32275114

RESUMO

There is compelling evidence implicating intestinal permeability in the pathogenesis of nonalcoholic steatohepatitis (NASH), but the underlying mechanisms remain poorly understood. Here we examined the role of bile acids (BA) in western diet (WD)-induced loss of colonic epithelial barrier (CEB) function in mice with a genetic impairment in intestinal epithelial barrier function, junctional adhesion molecule A knockout mice, F11r-/- . WD-fed knockout mice developed severe NASH, which was associated with increased BA concentration in the cecum and loss of CEB function. Analysis of cecal BA composition revealed selective increases in primary unconjugated BAs in the WD-fed mice, which correlated with increased abundance of microbial taxa linked to BA metabolism. In vitro permeability assays revealed that chenodeoxycholic acid (CDCA), which was elevated in the cecum of WD-fed mice, increased paracellular permeability, while the BA-binding resin sevelamer hydrochloride protected against CDCA-induced loss of barrier function. Sequestration of intestinal BAs by in vivo delivery of sevelamer to WD-fed knockout mice attenuated colonic mucosal inflammation and improved CEB. Sevelamer also reduced hepatic inflammation and fibrosis, and improved metabolic derangements associated with NASH. Collectively, these findings highlight a hitherto unappreciated role for BAs in WD-induced impairment of the intestinal epithelial barrier in NASH.


Assuntos
Ácidos e Sais Biliares/metabolismo , Colo/metabolismo , Dieta Ocidental/efeitos adversos , Hepatopatia Gordurosa não Alcoólica/metabolismo , Animais , Células CACO-2 , Moléculas de Adesão Celular/deficiência , Moléculas de Adesão Celular/genética , Colo/patologia , Modelos Animais de Doenças , Humanos , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Hepatopatia Gordurosa não Alcoólica/etiologia , Hepatopatia Gordurosa não Alcoólica/patologia , Permeabilidade , Receptores de Superfície Celular/deficiência , Receptores de Superfície Celular/genética , Sevelamer/administração & dosagem
8.
Immunity ; 37(3): 563-73, 2012 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-22981539

RESUMO

Mice lacking junctional adhesion molecule A (JAM-A, encoded by F11r) exhibit enhanced intestinal epithelial permeability, bacterial translocation, and elevated colonic lymphocyte numbers, yet do not develop colitis. To investigate the contribution of adaptive immune compensation in response to increased intestinal epithelial permeability, we examined the susceptibility of F11r(-/-)Rag1(-/-) mice to acute colitis. Although negligible contributions of adaptive immunity in F11r(+/+)Rag1(-/-) mice were observed, F11r(-/-)Rag1(-/-) mice exhibited increased microflora-dependent colitis. Elimination of T cell subsets and cytokine analyses revealed a protective role for TGF-ß-producing CD4(+) T cells in F11r(-/-) mice. Additionally, loss of JAM-A resulted in elevated mucosal and serum IgA that was dependent upon CD4(+) T cells and TGF-ß. Absence of IgA in F11r(+/+)Igha(-/-) mice did not affect disease, whereas F11r(-/-)Igha(-/-) mice displayed markedly increased susceptibility to acute injury-induced colitis. These data establish a role for adaptive immune-mediated protection from acute colitis under conditions of intestinal epithelial barrier compromise.


Assuntos
Imunidade Adaptativa/imunologia , Colite/imunologia , Mucosa Intestinal/imunologia , Intestinos/imunologia , Imunidade Adaptativa/genética , Animais , Translocação Bacteriana/genética , Translocação Bacteriana/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/imunologia , Colite/induzido quimicamente , Colite/genética , Sulfato de Dextrana , Epitélio/imunologia , Epitélio/metabolismo , Feminino , Citometria de Fluxo , Expressão Gênica , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/imunologia , Imunoglobulina A/genética , Imunoglobulina A/imunologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Intestinos/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Permeabilidade , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/imunologia , Fator de Crescimento Transformador beta/metabolismo
9.
J Hepatol ; 73(5): 1013-1022, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32540177

RESUMO

BACKGROUND & AIMS: The heterodimeric integrin receptor α4ß7 regulates CD4 T cell recruitment to inflamed tissues, but its role in the pathogenesis of non-alcoholic steatohepatitis (NASH) is unknown. Herein, we examined the role of α4ß7-mediated recruitment of CD4 T cells to the intestine and liver in NASH. METHODS: Male littermate F11r+/+ (control) and junctional adhesion molecule A knockout F11r-/- mice were fed a normal diet or a western diet (WD) for 8 weeks. Liver and intestinal tissues were analyzed by histology, quantitative reverse transcription PCR (qRT-PCR), 16s rRNA sequencing and flow cytometry. Colonic mucosa-associated microbiota were analyzed using 16s rRNA sequencing. Liver biopsies from patients with NASH were analyzed by confocal imaging and qRT-PCR. RESULTS: WD-fed knockout mice developed NASH and had increased hepatic and intestinal α4ß7+ CD4 T cells relative to control mice who developed mild hepatic steatosis. The increase in α4ß7+ CD4 T cells was associated with markedly higher expression of the α4ß7 ligand mucosal addressin cell adhesion molecule 1 (MAdCAM-1) in the colonic mucosa and livers of WD-fed knockout mice. Elevated MAdCAM-1 expression correlated with increased mucosa-associated Proteobacteria in the WD-fed knockout mice. Antibiotics reduced MAdCAM-1 expression indicating that the diet-altered microbiota promoted colonic and hepatic MAdCAM-1 expression. α4ß7 blockade in WD-fed knockout mice significantly decreased α4ß7+ CD4 T cell recruitment to the intestine and liver, attenuated hepatic inflammation and fibrosis, and improved metabolic indices. MAdCAM-1 blockade also reduced hepatic inflammation and fibrosis in WD-fed knockout mice. Hepatic MAdCAM-1 expression was elevated in patients with NASH and correlated with higher expression of α4 and ß7 integrins. CONCLUSIONS: These findings establish α4ß7/MAdCAM-1 as a critical axis regulating NASH development through colonic and hepatic CD4 T cell recruitment. LAY SUMMARY: Non-alcoholic steatohepatitis (NASH) is an advanced and progressive form of non-alcoholic fatty liver disease (NAFLD), and despite its growing incidence no therapies currently exist to halt NAFLD progression. Herein, we show that blocking integrin receptor α4ß7-mediated recruitment of CD4 T cells to the intestine and liver not only attenuates hepatic inflammation and fibrosis, but also improves metabolic derangements associated with NASH. These findings provide evidence for the potential therapeutic application of α4ß7 antibody in the treatment of human NASH.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Dieta Ocidental/efeitos adversos , Integrinas/metabolismo , Mucosa Intestinal/imunologia , Fígado/imunologia , Hepatopatia Gordurosa não Alcoólica/etiologia , Hepatopatia Gordurosa não Alcoólica/imunologia , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Linfócitos T CD4-Positivos/metabolismo , Moléculas de Adesão Celular/antagonistas & inibidores , Moléculas de Adesão Celular/deficiência , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Modelos Animais de Doenças , Microbioma Gastrointestinal/genética , Humanos , Integrinas/antagonistas & inibidores , Integrinas/imunologia , Fígado/patologia , Masculino , Camundongos , Camundongos Knockout , Mucoproteínas/antagonistas & inibidores , Mucoproteínas/metabolismo , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , Hepatopatia Gordurosa não Alcoólica/patologia , RNA Ribossômico 16S/genética , Receptores de Superfície Celular/deficiência , Receptores de Superfície Celular/genética
10.
FASEB J ; 33(12): 13632-13643, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31585047

RESUMO

Mucosal wound repair is coordinated by dynamic crosstalk between endogenous and exogenous mediators and specific receptors on epithelial cells and infiltrating immune cells. One class of such receptor-ligand pairs involves formyl peptide receptors (FPRs) that have been shown to influence inflammatory response and repair. Here we explored the role of murine Fpr2/3, an ortholog of human FPR2/receptor for lipoxin A4 (ALX), in orchestrating intestinal mucosal repair. Compared with wild-type (WT) mice, Fpr2/3-/- mice exhibited delayed recovery from acute experimental colitis and perturbed repair after biopsy-induced colonic mucosal injury. Decreased numbers of infiltrating monocytes were observed in healing wounds from Fpr2/3-/- mice compared with WT animals. Bone marrow transplant experiments revealed that Fpr2/3-/- monocytes showed a competitive disadvantage when infiltrating colonic wounds. Moreover, Fpr2/3-/- monocytes were defective in chemotactic responses to the chemokine CC chemokine ligand (CCL)20, which is up-regulated during early phases of inflammation. Analysis of Fpr2/3-/- monocytes revealed altered expression of the CCL20 receptor CC chemokine receptor (CCR)6, suggesting that Fpr2/3 regulates CCL20-CCR6-mediated monocyte chemotaxis to sites of mucosal injury in the gut. These findings demonstrate an important contribution of Fpr2/3 in facilitating monocyte recruitment to sites of mucosal injury to influence wound repair.-Birkl, D., O'Leary, M. N., Quiros, M., Azcutia, V., Schaller, M., Reed, M., Nishio, H., Keeney, J., Neish, A. S., Lukacs, N. W., Parkos, C. A., Nusrat, A. Formyl peptide receptor 2 regulates monocyte recruitment to promote intestinal mucosal wound repair.


Assuntos
Movimento Celular , Inflamação/terapia , Mucosa Intestinal/fisiologia , Monócitos/metabolismo , Receptores de Formil Peptídeo/fisiologia , Cicatrização , Animais , Transplante de Medula Óssea , Quimiocina CCL20/genética , Quimiocina CCL20/metabolismo , Colite/induzido quimicamente , Colite/metabolismo , Colite/patologia , Sulfato de Dextrana/toxicidade , Inflamação/etiologia , Inflamação/patologia , Mucosa Intestinal/lesões , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/citologia , Receptores CCR6/genética , Receptores CCR6/metabolismo
11.
Immunol Rev ; 273(1): 94-111, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27558330

RESUMO

Polymorphonuclear neutrophils (PMNs) are innate immune system cells that play an essential role in eradicating invading pathogens. PMN migration to sites of infection/inflammation requires exiting the microcirculation and subsequent crossing of epithelial barriers in mucosa-lined organs such as the lungs and intestines. Although these processes usually occur without significant damage to surrounding host tissues, dysregulated/excessive PMN transmigration and resultant bystander-tissue damage are characteristic of numerous mucosal inflammatory disorders. Mechanisms controlling PMN extravasation have been well characterized, but the molecular details regarding regulation of PMN migration across mucosal epithelia are poorly understood. Given that PMN migration across mucosal epithelia is strongly correlated with disease symptoms in many inflammatory mucosal disorders, enhanced understanding of the mechanisms regulating PMN transepithelial migration should provide insights into clinically relevant tissue-targeted therapies aimed at ameliorating PMN-mediated bystander-tissue damage. This review will highlight current understanding of the molecular interactions between PMNs and mucosal epithelia and the associated functional consequences.


Assuntos
Movimento Celular , Células Epiteliais/fisiologia , Imunidade nas Mucosas , Inflamação/imunologia , Neutrófilos/imunologia , Animais , Terapia Biológica , Comunicação Celular , Humanos , Infiltração de Neutrófilos
12.
Immunity ; 32(3): 392-402, 2010 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-20303298

RESUMO

Inflammatory cytokines have been proposed to regulate epithelial homeostasis during intestinal inflammation. We report here that interferon-gamma (IFN-gamma) regulates the crucial homeostatic functions of cell proliferation and apoptosis through serine-threonine protein kinase AKT-beta-catenin and Wingless-Int (Wnt)-beta-catenin signaling pathways. Short-term exposure of intestinal epithelial cells to IFN-gamma resulted in activation of beta-catenin through AKT, followed by induction of the secreted Wnt inhibitor Dkk1. Consequently, we observed an increase in Dkk1-mediated apoptosis upon extended IFN-gamma treatment and reduced proliferation through depletion of the Wnt coreceptor LRP6. These effects were enhanced by tumor necrosis factor-alpha (TNF-alpha), suggesting synergism between the two cytokines. Consistent with these results, colitis in vivo was associated with decreased beta-catenin-T cell factor (TCF) signaling, loss of plasma membrane-associated LRP6, and reduced epithelial cell proliferation. Proliferation was partially restored in IFN-gamma-deficient mice. Thus, we propose that IFN-gamma regulates intestinal epithelial homeostasis by sequential regulation of converging beta-catenin signaling pathways.


Assuntos
Células Epiteliais/imunologia , Homeostase , Interferon gama/imunologia , Intestinos/imunologia , Transdução de Sinais , beta Catenina/metabolismo , Animais , Apoptose , Linhagem Celular , Proliferação de Células , Colite/genética , Colite/imunologia , Colite/metabolismo , Colite/patologia , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Interferon gama/deficiência , Interferon gama/metabolismo , Mucosa Intestinal/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Wnt/metabolismo
13.
J Immunol ; 196(1): 34-8, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26590314

RESUMO

IL-1 family members are central mediators of host defense. In this article, we show that the novel IL-1 family member IL-36γ was expressed during experimental colitis and human inflammatory bowel disease. Germ-free mice failed to induce IL-36γ in response to dextran sodium sulfate (DSS)-induced damage, suggesting that gut microbiota are involved in its induction. Surprisingly, IL-36R-deficient (Il1rl2(-/-)) mice exhibited defective recovery following DSS-induced damage and impaired closure of colonic mucosal biopsy wounds, which coincided with impaired neutrophil accumulation in the wound bed. Failure of Il1rl2(-/-) mice to recover from DSS-induced damage was associated with a profound reduction in IL-22 expression, particularly by colonic neutrophils. Defective recovery of Il1rl2(-/-) mice could be rescued by an aryl hydrocarbon receptor agonist, which was sufficient to restore IL-22 expression and promote full recovery from DSS-induced damage. These findings implicate the IL-36/IL-36R axis in the resolution of intestinal mucosal wounds.


Assuntos
Colite/imunologia , Interleucina-1/biossíntese , Interleucinas/biossíntese , Receptores de Interleucina/imunologia , Cicatrização/imunologia , Animais , Colite/induzido quimicamente , Colite/microbiologia , Colo/imunologia , Colo/lesões , Sulfato de Dextrana , Helicobacter hepaticus/patogenicidade , Humanos , Inflamação/imunologia , Inflamação/patologia , Doenças Inflamatórias Intestinais/imunologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infiltração de Neutrófilos/imunologia , Neutrófilos/imunologia , Neutrófilos/metabolismo , Receptores de Hidrocarboneto Arílico/agonistas , Receptores de Interleucina/genética , Cicatrização/genética , Interleucina 22
14.
Gastroenterology ; 151(4): 616-32, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27436072

RESUMO

The intestinal tract is lined by a single layer of columnar epithelial cells that forms a dynamic, permeable barrier allowing for selective absorption of nutrients, while restricting access to pathogens and food-borne antigens. Precise regulation of epithelial barrier function is therefore required for maintaining mucosal homeostasis and depends, in part, on barrier-forming elements within the epithelium and a balance between pro- and anti-inflammatory factors in the mucosa. Pathologic states, such as inflammatory bowel disease, are associated with a leaky epithelial barrier, resulting in excessive exposure to microbial antigens, recruitment of leukocytes, release of soluble mediators, and ultimately mucosal damage. An inflammatory microenvironment affects epithelial barrier properties and mucosal homeostasis by altering the structure and function of epithelial intercellular junctions through direct and indirect mechanisms. We review our current understanding of complex interactions between the intestinal epithelium and immune cells, with a focus on pathologic mucosal inflammation and mechanisms of epithelial repair. We discuss leukocyte-epithelial interactions, as well as inflammatory mediators that affect the epithelial barrier and mucosal repair. Increased knowledge of communication networks between the epithelium and immune system will lead to tissue-specific strategies for treating pathologic intestinal inflammation.


Assuntos
Comunicação Celular , Células Epiteliais/fisiologia , Junções Intercelulares/fisiologia , Mucosa Intestinal/fisiopatologia , Leucócitos/fisiologia , Homeostase , Humanos , Doenças Inflamatórias Intestinais/patologia , Doenças Inflamatórias Intestinais/fisiopatologia , Mucosa Intestinal/patologia
15.
Gastroenterology ; 151(4): 733-746.e12, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27342212

RESUMO

BACKGROUND & AIMS: There is evidence from clinical studies that compromised intestinal epithelial permeability contributes to the development of nonalcoholic steatohepatitis (NASH), but the exact mechanisms are not clear. Mice with disruption of the gene (F11r) encoding junctional adhesion molecule A (JAM-A) have defects in intestinal epithelial permeability. We used these mice to study how disruption of the intestinal epithelial barrier contributes to NASH. METHODS: Male C57BL/6 (control) or F11r(-/-) mice were fed a normal diet or a diet high in saturated fat, fructose, and cholesterol (HFCD) for 8 weeks. Liver and intestinal tissues were collected and analyzed by histology, quantitative reverse-transcription polymerase chain reaction, and flow cytometry. Intestinal epithelial permeability was assessed in mice by measuring permeability to fluorescently labeled dextran. The intestinal microbiota were analyzed using 16S ribosomal RNA sequencing. We also analyzed biopsy specimens from proximal colons of 30 patients with nonalcoholic fatty liver disease (NAFLD) and 19 subjects without NAFLD (controls) undergoing surveillance colonoscopy. RESULTS: F11r(-/-) mice fed a HFCD, but not a normal diet, developed histologic and pathologic features of severe NASH including steatosis, lobular inflammation, hepatocellular ballooning, and fibrosis, whereas control mice fed a HFCD developed only modest steatosis. Interestingly, there were no differences in body weight, ratio of liver weight:body weight, or glucose homeostasis between control and F11r(-/-) mice fed a HFCD. In these mice, liver injury was associated with significant increases in mucosal inflammation, tight junction disruption, and intestinal epithelial permeability to bacterial endotoxins, compared with control mice or F11r(-/-) mice fed a normal diet. The HFCD led to a significant increase in inflammatory microbial taxa in F11r(-/-) mice, compared with control mice. Administration of oral antibiotics or sequestration of bacterial endotoxins with sevelamer hydrochloride reduced mucosal inflammation and restored normal liver histology in F11r(-/-) mice fed a HFCD. Protein and transcript levels of JAM-A were significantly lower in the intestinal mucosa of patients with NAFLD than without NAFLD; decreased expression of JAM-A correlated with increased mucosal inflammation. CONCLUSIONS: Mice with defects in intestinal epithelial permeability develop more severe steatohepatitis after a HFCD than control mice, and colon tissues from patients with NAFLD have lower levels of JAM-A and higher levels of inflammation than subjects without NAFLD. These findings indicate that intestinal epithelial barrier function and microbial dysbiosis contribute to the development of NASH. Restoration of intestinal barrier integrity and manipulation of gut microbiota might be developed as therapeutic strategies for patients with NASH.


Assuntos
Moléculas de Adesão Celular/deficiência , Dieta Hiperlipídica/efeitos adversos , Hepatopatia Gordurosa não Alcoólica/genética , Receptores de Superfície Celular/deficiência , Animais , Colesterol , Dieta Hiperlipídica/métodos , Carboidratos da Dieta , Modelos Animais de Doenças , Disbiose/complicações , Disbiose/genética , Frutose , Microbioma Gastrointestinal/genética , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Fígado/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Hepatopatia Gordurosa não Alcoólica/microbiologia , Hepatopatia Gordurosa não Alcoólica/patologia , Permeabilidade , Reação em Cadeia da Polimerase Via Transcriptase Reversa
16.
Am J Pathol ; 186(6): 1404-16, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27083514

RESUMO

In recent years, it has become clear that innate immune cells termed neutrophils act as double-edged swords by playing essential roles in clearing infection but also causing tissue damage, yet being critical for wound healing. Neutrophil recruitment to sites of injured tissue or infection has been well studied, and many of the molecular events that regulate passage of leukocytes out of the microcirculation are now understood. However, after exiting the circulation, the molecular details that regulate neutrophil passage to end targets, such mucosal surfaces, are just beginning to be appreciated. Given that migration of neutrophils across mucosal epithelia is associated with disease symptoms and disruption of critical barrier function in disorders such as inflammatory bowel disease, there has been long-standing interest in understanding the molecular basis and functional consequences of neutrophil-epithelial interactions. It is a great honor that my work was recognized by the Rous-Whipple Award this past year, giving me the opportunity to summarize what we have learned during the past few decades about leukocyte interactions with epithelial cells.


Assuntos
Células Epiteliais/fisiologia , Neutrófilos/fisiologia , Animais , Movimento Celular/fisiologia , Células Epiteliais/citologia , Humanos , Neutrófilos/citologia , Cicatrização/fisiologia
17.
Am J Pathol ; 186(2): 297-311, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26687991

RESUMO

Polymorphonuclear leukocytes (PMNs) are innate immune cells whose principal function is to migrate from the blood to sites of inflammation, where they exert crucial anti-infectious and immunomodulatory effects. However, dysregulated migration of PMNs into mucosal epithelial tissues is characteristic of chronic inflammatory disorders, including inflammatory bowel disease. Carbohydrate-mediated binding interactions between PMN Lewis glycans and endothelial glycan-binding proteins are critical for initial migration of PMN out of the vasculature. However, the role of Lewis glycans during transepithelial migration (TEM) has not been well characterized. Herein, we show that antibody blockade of Lewis X (Le(x)) displayed as terminal glycan residues on the PMN surface blocks chemotaxis and TEM while enhancing PMN-adhesive interactions with intestinal epithelia. Unexpectedly, targeting of subterminal Le(x) residues within glycan chains had no effect on PMN migration or adhesive interactions. There was increased surface expression of Le(x) on PMN after TEM, and blockade of terminal Le(x) regulated post-migratory PMN functions, increasing PMN phagocytosis and the surface mobilization of azurophilic (CD63, myeloperoxidase, and neutrophil elastase) and specific (CD66b and lactoferrin) granule markers. These findings suggest that terminal Le(x) represents a potential target for regulating PMN trafficking and function in inflamed mucosa. Furthermore, given its abundant expression on migrating PMN, Le(x) may be a rational target for modulating inflammation in diseases where dysregulated PMN influx is associated with host tissue damage.


Assuntos
Mucosa Intestinal/metabolismo , Antígenos CD15/imunologia , Neutrófilos/metabolismo , Fagocitose/imunologia , Migração Transendotelial e Transepitelial/imunologia , Adesão Celular/imunologia , Células Cultivadas , Quimiotaxia/imunologia , Epitélio/metabolismo , Humanos , Doenças Inflamatórias Intestinais/metabolismo , Mucosa Intestinal/patologia
18.
FASEB J ; 30(12): 4007-4020, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27553226

RESUMO

Neutrophil [polymorphonuclear leukocyte (PMN)] transepithelial migration (TEM) is a hallmark of inflammatory mucosal disorders. PMN TEM is associated with epithelial injury; however, mechanisms involved in this process are not well defined. The current work describes a new mechanism whereby deposition of PMN membrane-derived microparticles (PMN-MPs) onto intestinal epithelial cells (IECs) during TEM leads to loss of epithelial cadherins, thus promoting epithelial injury and increased PMN recruitment. PMN-MPs secreted by activated PMNs during TEM displayed a high level of enzymatically active matrix metalloproteinase 9 (MMP-9), and were capable of mediating potent effects on IEC integrity. Isolated PMN-MPs efficiently bound to IEC monolayers and induced cleavage of desmoglein-2 (DSG-2) but not E-cadherin, leading to disruption of IEC intercellular adhesions. Furthermore, PMN-MP binding to intestinal epithelium in vitro in transwell assays and in vivo in ligated intestinal loop preparations facilitated increases in PMN TEM. These effects were MMP-9 dependent and were reversed in the presence of specific pharmacological inhibitors. Finally, we demonstrated that IEC Dsg-2 serves as a barrier for migrating PMNs, and its removal by PMN-MP-associated MMP-9 facilitates PMN trafficking across epithelial layers. Our findings thus implicate PMN-MPs in PMN-mediated inflammation and epithelial damage, as observed in inflammatory disorders of mucosal surfaces.-Butin-Israeli, V., Houser, M. C., Feng, M., Thorp, E. B., Nusrat, A., Parkos, C. A, Sumagin, R. Deposition of microparticles by neutrophils onto inflamed epithelium: a new mechanism to disrupt epithelial intercellular adhesions and promote transepithelial migration.


Assuntos
Células Epiteliais/citologia , Epitélio/metabolismo , Neutrófilos/metabolismo , Migração Transendotelial e Transepitelial/fisiologia , Animais , Caderinas/metabolismo , Adesão Celular , Micropartículas Derivadas de Células/metabolismo , Células Cultivadas , Humanos , Camundongos Endogâmicos C57BL
19.
Am J Physiol Gastrointest Liver Physiol ; 311(3): G458-65, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27418680

RESUMO

Intestinal wounds often occur during inflammatory and ischemic disorders of the gut. To repair damage, intestinal epithelial cells must rapidly spread and migrate to cover exposed lamina propria, events that involve redox signaling. Wounds are subject to extensive redox alterations, particularly resulting from H2O2 produced in the adjacent tissue by both the epithelium and emigrating leukocytes. The mechanisms governing these processes are not fully understood, particularly at the level of protein signaling. Crk-associated substrate, or Cas, is an important signaling protein known to modulate focal adhesion and actin cytoskeletal dynamics, whose association with Crk is regulated by Abl kinase, a ubiquitously expressed tyrosine kinase. We sought to evaluate the role of Abl regulation of Cas at the level of cell spreading and migration during wound closure. As a model, we used intestinal epithelial cells exposed to H2O2 or scratch wounded to assess the Abl-Cas signaling pathway. We characterized the localization of phosphorylated Cas in mouse colonic epithelium under baseline conditions and after biopsy wounding the mucosa. Analysis of actin and focal adhesion dynamics by microscopy or biochemical analysis after manipulating Abl kinase revealed that Abl controls redox-dependent Cas phosphorylation and localization to influence cell spreading and migration. Collectively, our data shed new light on redox-sensitive protein signaling modules controlling intestinal wound healing.


Assuntos
Movimento Celular/fisiologia , Colo/patologia , Proteína Substrato Associada a Crk/metabolismo , Células Epiteliais/fisiologia , Mucosa Intestinal/citologia , Animais , Biópsia , Células CACO-2 , Proteína Substrato Associada a Crk/genética , Regulação da Expressão Gênica , Humanos , Mucosa Intestinal/patologia , Camundongos , Oxirredução , Fosforilação , Ferimentos e Lesões
20.
Am J Pathol ; 185(8): 2206-18, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26216285

RESUMO

The intestinal epithelium is a dynamic barrier that maintains the distinct environments of intestinal tissue and lumen. Epithelial barrier function is defined principally by tight junctions, which, in turn, depend on the regulated expression of claudin family proteins. Claudins are expressed differentially during intestinal epithelial cell (IEC) differentiation. However, regulatory mechanisms governing claudin expression during epithelial differentiation are incompletely understood. We investigated the molecular mechanisms regulating claudin-7 during IEC differentiation. Claudin-7 expression is increased as epithelial cells differentiate along the intestinal crypt-luminal axis. By using model IECs we observed increased claudin-7 mRNA and nascent heteronuclear RNA levels during differentiation. A screen for potential regulators of the CLDN7 gene during IEC differentiation was performed using a transcription factor/DNA binding array, CLDN7 luciferase reporters, and in silico promoter analysis. We identified hepatocyte nuclear factor 4α as a regulatory factor that bound endogenous CLDN7 promoter in differentiating IECs and stimulated CLDN7 promoter activity. These findings support a role of hepatocyte nuclear factor 4α in controlling claudin-7 expression during IEC differentiation.


Assuntos
Diferenciação Celular/genética , Claudinas/metabolismo , Células Epiteliais/metabolismo , Fator 4 Nuclear de Hepatócito/metabolismo , Mucosa Intestinal/metabolismo , Células CACO-2 , Claudinas/genética , Células Epiteliais/citologia , Regulação da Expressão Gênica , Células HT29 , Fator 4 Nuclear de Hepatócito/genética , Humanos , Mucosa Intestinal/citologia , Regiões Promotoras Genéticas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA