Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Microbiol ; 3(1): 53-61, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29133884

RESUMO

Oral epithelial cells discriminate between pathogenic and non-pathogenic stimuli, and only induce an inflammatory response when they are exposed to high levels of a potentially harmful microorganism. The pattern recognition receptors (PRRs) in epithelial cells that mediate this differential response are poorly understood. Here, we demonstrate that the ephrin type-A receptor 2 (EphA2) is an oral epithelial cell PRR that binds to exposed ß-glucans on the surface of the fungal pathogen Candida albicans. Binding of C. albicans to EphA2 on oral epithelial cells activates signal transducer and activator of transcription 3 and mitogen-activated protein kinase signalling in an inoculum-dependent manner, and is required for induction of a proinflammatory and antifungal response. EphA2 -/- mice have impaired inflammatory responses and reduced interleukin-17 signalling during oropharyngeal candidiasis, resulting in more severe disease. Our study reveals that EphA2 functions as a PRR for ß-glucans that senses epithelial cell fungal burden and is required for the maximal mucosal inflammatory response to C. albicans.


Assuntos
Candida albicans/metabolismo , Candidíase Bucal/metabolismo , Mucosa Bucal/metabolismo , Receptor EphA2/metabolismo , Receptores de Reconhecimento de Padrão/metabolismo , beta-Glucanas/metabolismo , Animais , Candida albicans/crescimento & desenvolvimento , Candidíase Bucal/patologia , Linhagem Celular , Citocinas/biossíntese , Modelos Animais de Doenças , Endocitose , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/metabolismo , Mediadores da Inflamação/análise , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mucosa Bucal/citologia , Mucosa Bucal/microbiologia , Fosforilação , Receptor EphA2/antagonistas & inibidores , Receptor EphA2/deficiência , Receptores de Reconhecimento de Padrão/antagonistas & inibidores , Receptores de Reconhecimento de Padrão/deficiência , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais
2.
Circulation ; 136(6): 566-582, 2017 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-28487392

RESUMO

BACKGROUND: Atherosclerotic plaque formation results from chronic inflammation and fibroproliferative remodeling in the vascular wall. We previously demonstrated that both human and mouse atherosclerotic plaques show elevated expression of EphA2, a guidance molecule involved in cell-cell interactions and tumorigenesis. METHODS: Here, we assessed the role of EphA2 in atherosclerosis by deleting EphA2 in a mouse model of atherosclerosis (Apoe-/-) and by assessing EphA2 function in multiple vascular cell culture models. After 8 to 16 weeks on a Western diet, male and female mice were assessed for atherosclerotic burden in the large vessels, and plasma lipid levels were analyzed. RESULTS: Despite enhanced weight gain and plasma lipid levels compared with Apoe-/- controls, EphA2-/-Apoe-/- knockout mice show diminished atherosclerotic plaque formation, characterized by reduced proinflammatory gene expression and plaque macrophage content. Although plaque macrophages express EphA2, EphA2 deletion does not affect macrophage phenotype, inflammatory responses, and lipid uptake, and bone marrow chimeras suggest that hematopoietic EphA2 deletion does not affect plaque formation. In contrast, endothelial EphA2 knockdown significantly reduces monocyte firm adhesion under flow. In addition, EphA2-/-Apoe-/- mice show reduced progression to advanced atherosclerotic plaques with diminished smooth muscle and collagen content. Consistent with this phenotype, EphA2 shows enhanced expression after smooth muscle transition to a synthetic phenotype, and EphA2 depletion reduces smooth muscle proliferation, mitogenic signaling, and extracellular matrix deposition both in atherosclerotic plaques and in vascular smooth muscle cells in culture. CONCLUSIONS: Together, these data identify a novel role for EphA2 in atherosclerosis, regulating both plaque inflammation and progression to advanced atherosclerotic lesions. Cell culture studies suggest that endothelial EphA2 contributes to atherosclerotic inflammation by promoting monocyte firm adhesion, whereas smooth muscle EphA2 expression may regulate the progression to advanced atherosclerosis by regulating smooth muscle proliferation and extracellular matrix deposition.


Assuntos
Aterosclerose/patologia , Receptor EphA2/genética , Animais , Aorta/metabolismo , Aorta/patologia , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Aterosclerose/metabolismo , Linhagem da Célula , Proliferação de Células , Células Cultivadas , Modelos Animais de Doenças , Feminino , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Inflamação , Macrófagos/citologia , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Knockout , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/metabolismo , Fenótipo , Placa Aterosclerótica/patologia , Receptor EphA2/deficiência , Receptor EphA2/metabolismo , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo
3.
PLoS One ; 10(7): e0130975, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26177500

RESUMO

OBJECTIVES: To investigate the role of EphA2 in malignant cellular behavior in renal cell carcinoma (RCC) cells and whether FAK/RhoA signaling can act as downstream effectors of EphA2 on RCC cells. METHODS: Expression of EphA2 protein in non-metastatic RCC (Caki-2 and A498), metastatic RCC cells (Caki-1 and ACHN), HEK-293 cells and prostate cancer cells (PC-3 and DU-145; positive controls of EphA2 expression) was evaluated by Western blot. Changes in mRNA or protein expression of EphA2, FAK or membrane-bound RhoA following EphA2, FAK or RhoA small interfering RNA (siRNA) transfection were determined by reverse transcription polymerase chain reaction or Western blot. The effect of siRNA treatment on cellular viability, apoptosis and invasion was analyzed by cell counting kit-8, Annexin-V and modified Matrigel-Boyden assays, respectively. RESULTS: In all RCC cell lines, the expression of EphA2 protein was detectable at variable levels; however, in HEK-293 cells, EphA2 expression was very low. Treatment with EphA2 siRNA significantly reduced the expression of EphA2 mRNA and protein in all RCC cell lines. For non-metastatic RCC cells (Caki-2 and A498) but not metastatic RCC cells (Caki-1 and ACHN), cellular viability, invasiveness, resistance to apoptosis, expression of membrane-bound RhoA protein and FAK phosphorylation were significantly decreased in EphA2 siRNA-treated cells compared to the control. In non-metastatic RCC cells, FAK siRNA significantly attenuated the invasiveness, resistance to apoptosis, as well as expression of membrane-bound RhoA protein without changing protein expression of EphA2. RhoA siRNA significantly decreased the malignant cellular behavior and expression of membrane-bound RhoA protein without changing EphA2 protein expression or FAK phosphorylation. CONCLUSIONS: Our data provide the first functional evidence that the EphA2/FAK/RhoA signaling pathway plays a critical role in the malignant cellular behavior of RCC and appears to be functional particularly in the early stage of malignant progression of non-metastatic RCC.


Assuntos
Carcinoma de Células Renais/patologia , Neoplasias Renais/patologia , Receptor EphA2/metabolismo , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Sobrevivência Celular , Quinase 1 de Adesão Focal/metabolismo , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Metástase Neoplásica , Fosforilação , RNA Interferente Pequeno/genética , Receptor EphA2/deficiência , Receptor EphA2/genética , Transdução de Sinais , Proteína rhoA de Ligação ao GTP/metabolismo
4.
Mol Cancer Res ; 13(3): 524-37, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25504371

RESUMO

UNLABELLED: Angiogenic remodeling during embryonic development and in adult tissue homeostasis is orchestrated by cooperative signaling between several distinct molecular pathways, which are often exploited by tumors. Indeed, tumors upregulate proangiogenic molecules while simultaneously suppressing angiostatic pathways to recruit blood vessels for growth, survival, and metastatic spread. Understanding how cancers exploit proangiogenic and antiangiogenic signals is a key step in developing new, molecularly targeted antiangiogenic therapies. While EphA2, a receptor tyrosine kinase (RTK), is required for VEGF-induced angiogenesis, the mechanism through which these pathways intersect remains unclear. Slit2 expression is elevated in EphA2-deficient endothelium, and here it is reported that inhibiting Slit activity rescues VEGF-induced angiogenesis in cell culture and in vivo, as well as VEGF-dependent tumor angiogenesis, in EphA2-deficient endothelial cells and animals. Moreover, blocking Slit activity or Slit2 expression in EphA2-deficient endothelial cells restores VEGF-induced activation of Src and Rac, both of which are required for VEGF-mediated angiogenesis. These data suggest that EphA2 suppression of Slit2 expression and Slit angiostatic activity enables VEGF-induced angiogenesis in vitro and in vivo, providing a plausible mechanism for impaired endothelial responses to VEGF in the absence of EphA2 function. IMPLICATIONS: Modulation of angiostatic factor Slit2 by EphA2 receptor regulates endothelial responses to VEGF-mediated angiogenesis and tumor neovascularization.


Assuntos
Endotélio/irrigação sanguínea , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Neoplasias Mamárias Experimentais/irrigação sanguínea , Neovascularização Fisiológica , Proteínas do Tecido Nervoso/metabolismo , Receptor EphA2/deficiência , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Células Cultivadas , Técnicas de Cocultura , Meios de Cultivo Condicionados , Endotélio/metabolismo , Feminino , Pulmão/irrigação sanguínea , Pulmão/citologia , Neoplasias Mamárias Experimentais/metabolismo , Camundongos , Transdução de Sinais
5.
Cardiovasc Diabetol ; 13: 114, 2014 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-25166508

RESUMO

BACKGROUND: We have previously shown that EphrinA1/EphA expression profile changes in response to myocardial infarction (MI), exogenous EphrinA1-Fc administration following MI positively influences wound healing, and that deletion of the EphA2 Receptor (EphA2-R) exacerbates injury and remodeling. To determine whether or not ephrinA1-Fc would be of therapeutic value in the hyperglycemic infarcted heart, it is critical to evaluate how ephrinA1/EphA signaling changes in the hyperglycemic myocardium in response to MI. METHODS: Streptozotocin (STZ)-induced hyperglycemia in wild type (WT) and EphA2-receptor mutant (EphA2-R-M) mice was initiated by an intraperitoneal injection of STZ (150 mg/kg) 10 days before surgery. MI was induced by permanent ligation of the left anterior descending coronary artery and analyses were performed at 4 days post-MI. ANOVAs with Student-Newman Keuls multiple comparison post-hoc analysis illustrated which groups were significantly different, with significance of at least p < 0.05. RESULTS: Both WT and EphA2-R-M mice responded adversely to STZ, but only hyperglycemic EphA2-R-M mice had lower ejection fraction (EF) and fractional shortening (FS). At 4 days post-MI, we observed greater post-MI mortality in EphA2-R-M mice compared with WT and this was greater still in the EphA2-R-M hyperglycemic mice. Although infarct size was greater in hyperglycemic WT mice vs normoglycemic mice, there was no difference between hyperglycemic EphA2-R-M mice and normoglycemic EphA2-R-M mice. The hypertrophic response that normally occurs in viable myocardium remote to the infarct was noticeably absent in epicardial cardiomyocytes and cardiac dysfunction worsened in hyperglycemic EphA2-R-M hearts post-MI. The characteristic interstitial fibrotic response in the compensating myocardium remote to the infarct also did not occur in hyperglycemic EphA2-R-M mouse hearts to the same extent as that observed in the hyperglycemic WT mouse hearts. Differences in neutrophil and pan-leukocyte infiltration and serum cytokines implicate EphA2-R in modulation of injury and the differences in ephrinA1 and EphA6-R expression in governing this are discussed. CONCLUSIONS: We conclude that EphA2-mutant mice are more prone to hyperglycemia-induced increased injury, decreased survival, and worsened LV remodeling due to impaired wound healing.


Assuntos
Modelos Animais de Doenças , Hiperglicemia/metabolismo , Hiperglicemia/mortalidade , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/mortalidade , Receptor EphA2/deficiência , Animais , Masculino , Camundongos , Camundongos Knockout , Taxa de Sobrevida/tendências
6.
Cardiovasc Pathol ; 23(3): 169-74, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24561077

RESUMO

BACKGROUND: The inflammatory response of vascular endothelial cells plays important roles in the initiation and progression of atherosclerotic lesions. EphA2 receptor activation promotes the endothelial cell inflammatory response, and its expression is increased in the endothelial cell layer of atherosclerotic plaques. However, the association between EphA2 and atherosclerosis has not been determined. METHODS: Eight-week-old male ApoE(-/-) mice were systemically infected with adenoassociated virus serotype 9 carrying a small hairpin RNA specifically targeting the EphA2 gene to knock down EphA2 expression in aortic endothelial cells. These mice were then fed a high-cholesterol diet for 12 weeks. Blood was collected for the measurement of plasma lipids. The aortas were harvested to evaluate the atherosclerotic lesion size, macrophage components, and expression of proinflammatory genes using Oil Red O staining, immunofluorescence staining, and molecular biology analysis. RESULTS: The lesions formed in the entire aorta and aortic sinus of the ApoE(-/-) mice with EphA2 knockdown were significantly smaller than those in the control mice (10.7%±3.1% versus 25.1%±4.2%; 0.51±0.02mm(2) versus 0.85±0.03mm(2); n=10; P<.05). Furthermore, the lesions in the ApoE(-/-) mice with EphA2 knockdown displayed reduced inflammation compared with the control mice, as reflected by the decreased macrophage infiltration (8.2%±2.9% versus 22.7%±4%; n=10; P<.05); decreased nuclear factor-κß activation; and diminished expression of vascular cell adhesion molecule-1, E-selectin, and monocyte chemotactic protein-1 (all P<.05). CONCLUSIONS: Our data demonstrate that the EphA2 receptor silencing attenuates the extent and inflammation of atherosclerotic lesions in ApoE(-/-) mice. Thus, EphA2 knockdown in endothelial cells represents a novel therapeutic strategy for patients with atherosclerosis.


Assuntos
Aorta/metabolismo , Doenças da Aorta/prevenção & controle , Apolipoproteínas E/deficiência , Aterosclerose/prevenção & controle , Placa Aterosclerótica , Receptor EphA2/deficiência , Animais , Aorta/patologia , Doenças da Aorta/genética , Doenças da Aorta/metabolismo , Doenças da Aorta/patologia , Apolipoproteínas E/genética , Aterosclerose/genética , Aterosclerose/metabolismo , Aterosclerose/patologia , Colesterol na Dieta , Modelos Animais de Doenças , Mediadores da Inflamação/metabolismo , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Camundongos Knockout , Interferência de RNA , Receptor EphA2/genética
7.
Mol Vis ; 19: 254-66, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23401654

RESUMO

PURPOSE: The cells of the mammalian lens must be carefully organized and regulated to maintain clarity. Recent studies have identified the Eph receptor ligand ephrin-A5 as a major contributor to lens development, as mice lacking ephrin-A5 develop abnormal lenses, resulting in cataracts. As a follow-up to our previous study on the cataracts observed in ephrin-A5(-/-) animals, we have further examined the morphological and molecular changes in the ephrin-A5(-/-) lens. METHODS: Wild-type and ephrin-A5(-/-) eyes at various ages were fixed, sectioned, and examined using histological techniques. Protein expression and localization were determined using immunohistochemistry and western blot analysis. RESULTS: Lens abnormalities in the ephrin-A5(-/-) animals are observed at postnatal stages, with lens opacity occurring by postnatal day 21. Structural defects in the lens are first observed in the outer lens fiber cell region where cells in the ephrin-A5(-/-) lens are severely disorganized. Ephrin-A5 and the Eph receptor EphA2 are expressed during early ocular development and continue to be expressed into postnatal stages. The cataracts in the ephrin-A5(-/-) mutants occur regardless of the presence of the CP49 mutation. CONCLUSIONS: In this follow-up study, we have uncovered additional details explicating the mechanisms underlying ephrin-A5 function in the lens. Furthermore, elucidation of the expression of ephrin-A5 and the Eph receptor EphA2 in the lens supports a fundamental role for this receptor-ligand complex in lens development. These observations, in concert with our previous study, strongly suggest that ephrin-A5 has a critical role in postnatal lens fiber organization to maintain lens transparency.


Assuntos
Efrina-A5/deficiência , Efrina-A5/genética , Regulação da Expressão Gênica no Desenvolvimento , Cristalino/metabolismo , Cristalino/patologia , Animais , Catarata/etiologia , Catarata/metabolismo , Catarata/patologia , Efrina-A5/metabolismo , Proteínas do Olho/genética , Proteínas do Olho/metabolismo , Proteínas de Filamentos Intermediários/genética , Proteínas de Filamentos Intermediários/metabolismo , Cristalino/crescimento & desenvolvimento , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Mutação , Receptor EphA2/deficiência , Receptor EphA2/genética , Receptor EphA2/metabolismo , Índice de Gravidade de Doença
8.
PLoS One ; 8(1): e53528, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23308246

RESUMO

Ephrin (Eph) signaling within the central nervous system is known to modulate axon guidance, synaptic plasticity, and to promote long-term potentiation. We investigated the potential involvement of EphA2 receptors in ischemic stroke-induced brain inflammation in a mouse model of focal stroke. Cerebral ischemia was induced in male C57Bl6/J wild-type (WT) and EphA2-deficient (EphA2(-/-)) mice by middle cerebral artery occlusion (MCAO; 60 min), followed by reperfusion (24 or 72 h). Brain infarction was measured using triphenyltetrazolium chloride staining. Neurological deficit scores and brain infarct volumes were significantly less in EphA2(-/-) mice compared with WT controls. This protection by EphA2 deletion was associated with a comparative decrease in brain edema, blood-brain barrier damage, MMP-9 expression and leukocyte infiltration, and higher expression levels of the tight junction protein, zona occludens-1. Moreover, EphA2(-/-) brains had significantly lower levels of the pro-apoptotic proteins, cleaved caspase-3 and BAX, and higher levels of the anti-apoptotic protein, Bcl-2 as compared to WT group. We confirmed that isolated WT cortical neurons express the EphA2 receptor and its ligands (ephrin-A1-A3). Furthermore, expression of all four proteins was increased in WT primary cortical neurons following 24 h of glucose deprivation, and in the brains of WT mice following stroke. Glucose deprivation induced less cell death in primary neurons from EphA2(-/-) compared with WT mice. In conclusion, our data provide the first evidence that the EphA2 receptor directly contributes to blood-brain barrier damage and neuronal death following ischemic stroke.


Assuntos
Infarto Encefálico/genética , Isquemia Encefálica/genética , Córtex Cerebral/metabolismo , Neurônios/metabolismo , Receptor EphA2/genética , Animais , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/patologia , Infarto Encefálico/metabolismo , Infarto Encefálico/patologia , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Caspase 3/genética , Caspase 3/metabolismo , Córtex Cerebral/patologia , Efrinas/genética , Efrinas/metabolismo , Regulação da Expressão Gênica , Glucose/deficiência , Infarto da Artéria Cerebral Média/patologia , Masculino , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Neurônios/patologia , Receptor EphA2/deficiência , Traumatismo por Reperfusão/patologia , Transdução de Sinais , Sais de Tetrazólio , Proteína da Zônula de Oclusão-1/genética , Proteína da Zônula de Oclusão-1/metabolismo , Proteína X Associada a bcl-2/genética , Proteína X Associada a bcl-2/metabolismo
9.
Am J Respir Cell Mol Biol ; 46(1): 40-7, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21799118

RESUMO

Stimulation by the ephrin-A1 ligand of the EphA2 receptor increases endothelial permeability. Lung injury increases the expression of EphA2, but the role of EphA2 in such injury is not well understood. To determine whether EphA2 contributes to changes in permeability and inflammation in the injured lung, we studied wild-type (WT) and EphA2 knockout (KO) mice, using isolated, perfused lung (IPL) preparations and a model of bleomycin-induced lung injury. We also studied the response of endothelial cells to ephrin-A1. In the IPL preparations, ephrin-A1 increased the filtration coefficient in WT mice, but not in EphA2 KO mice, demonstrating that EphA2 regulates vascular permeability. In early bleomycin injury in WT mice, the expression of both EphA2 and ephrin-A1 increased. EphA2 KO animals were protected from lung injury, showing less water and alveolar protein in the lungs than WT mice, consistent with reduced permeability. Bleomycin caused less accumulation of lung leukocytes in EphA2 KO animals than in WT animals, suggesting that EphA2 regulates inflammation. To determine whether EphA2 deficiency alters the production of chemokines, CXCL1 and CCL2 in the lungs were measured. After bleomycin injury, EphA2 KO animals produced less CXCL1 and CCL2 than WT animals. Because NF-κß mediates the production of chemokines, the effect of the ephrin-A1 ligand on the activation of NF-κß and the expression of chemokines was measured in endothelial cells. Ephrin-a1 significantly increased NF-κß nuclear translocation and the expression of chemokine mRNA. This study demonstrates that the expression of EphA2 increases in the injured lung, and not only contributes to changes in permeability, but also plays a previously unrecognized role in promoting inflammatory responses.


Assuntos
Bleomicina/toxicidade , Permeabilidade Capilar/fisiologia , Efrina-A1/metabolismo , Efrina-A1/farmacologia , Lesão Pulmonar/metabolismo , Receptor EphA2/deficiência , Receptor EphA2/metabolismo , Animais , Permeabilidade Capilar/genética , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Quimiocina CXCL1/genética , Quimiocina CXCL1/metabolismo , Células Endoteliais/metabolismo , Efrina-A1/biossíntese , Efrina-A1/genética , Técnicas de Inativação de Genes , Humanos , Inflamação/genética , Inflamação/metabolismo , Leucócitos/metabolismo , Lesão Pulmonar/induzido quimicamente , Lesão Pulmonar/genética , Camundongos , Camundongos Knockout , NF-kappa B/metabolismo , RNA Mensageiro/genética , Receptor EphA2/genética , Água/metabolismo
10.
PLoS One ; 6(11): e28147, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22140528

RESUMO

Recent genetic studies show that the Eph/ephrin bidirectional signaling pathway is associated with both congenital and age-related cataracts in mice and humans. We have investigated the molecular mechanisms of cataractogenesis and the roles of ephrin-A5 and EphA2 in the lens. Ephrin-A5 knockout ⁻/⁻ mice often display anterior polar cataracts while EphA2⁻/⁻ lenses show very mild cortical or nuclear cataracts at weaning age. The anterior polar cataract of ephrin-A5⁻/⁻ lenses is correlated with multilayers of aberrant cells that express alpha smooth muscle actin, a marker for mesenchymal cells. Only select fiber cells are altered in ephrin-A5⁻/⁻ lenses. Moreover, the disruption of membrane-associated ß-catenin and E-cadherin junctions is observed in ephrin-A5⁻/⁻ lens central epithelial cells. In contrast, EphA2⁻/⁻ lenses display normal monolayer epithelium while disorganization is apparent in all lens fiber cells. Immunostaining of ephrin-A5 proteins, highly expressed in lens epithelial cells, were not colocalized with EphA2 proteins, mainly expressed in lens fiber cells. Besides the previously reported function of ephrin-A5 in lens fiber cells, this work suggests that ephrin-A5 regulates ß-catenin signaling and E-cadherin to prevent lens anterior epithelial cells from undergoing the epithelial-to-mesenchymal transition while EphA2 is essential for controlling the organization of lens fiber cells through an unknown mechanism. Ephrin-A5 and EphA2 likely interacting with other members of Eph/ephrin family to play diverse functions in lens epithelial cells and/or fiber cells.


Assuntos
Efrina-A5/metabolismo , Cristalino/metabolismo , Receptor EphA2/metabolismo , Receptores da Família Eph/metabolismo , Transdução de Sinais , Animais , Cápsula Anterior do Cristalino/metabolismo , Cápsula Anterior do Cristalino/patologia , Caderinas/metabolismo , Catarata/metabolismo , Catarata/patologia , Efrina-A5/deficiência , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Proteínas de Fluorescência Verde/metabolismo , Cristalino/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Confocal , Fenótipo , Transporte Proteico , Receptor EphA2/deficiência , beta Catenina/metabolismo
11.
Am J Pathol ; 174(6): 2388-99, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19443703

RESUMO

Both Eph receptors and ephrin ligands have been implicated in blood vessel and neuronal development. Recent studies suggested that EphA2 inhibition reduces tumor angiogenesis, but its role in blood vessel development and inflammation is unclear. We examined these issues using either airways of pathogen-free, EphA2-deficient mice at various ages or EphA2-deficient mice whose airways were inflamed by either Mycoplasma pulmonis infection or ovalbumin sensitization and challenge. EphA2-deficient mice had fewer capillaries, a greater number of endothelial sprouts, and greater capillary diameters than age-matched, wild-type control mice. Moreover, capillaries in EphA2-deficient mice had significantly less pericyte coverage, suggesting abnormal interactions between endothelial cells and pericytes. These differences were apparent in early postnatal life but decreased during progression into adulthood. In inflamed airways, significantly more angiogenesis and lymphangiogenesis, a greater number of infiltrating leukocytes, and higher expression levels of inflammatory cytokine mRNA were present in EphA2-deficient mice after M. pulmonis infection. Additionally, in allergic airway inflammation with ovalbumin sensitization and challenge, a greater number of lymphatic sprouts and infiltrating leukocytes, higher mRNA expression levels of TH2 cytokines and chemokines related to allergic airway inflammation, and enhanced airway hyper-responsiveness were present in EphA2-deficient mice. We conclude that defective pericyte coverage causes capillary defects, abundant endothelial sprouts, and thick capillary diameters in EphA2-deficient mice, indicating that these animals have exaggerated responses to airway inflammation.


Assuntos
Capilares/patologia , Linfangiogênese , Neovascularização Fisiológica , Pneumonia/patologia , Receptor EphA2/deficiência , Alérgenos/imunologia , Animais , Capilares/crescimento & desenvolvimento , Citocinas/biossíntese , Células Endoteliais/patologia , Imuno-Histoquímica , Camundongos , Camundongos Mutantes , Infecções por Mycoplasma/imunologia , Infecções por Mycoplasma/patologia , Mycoplasma pulmonis , Ovalbumina/imunologia , Pericitos/patologia , Pneumonia/genética , Pneumonia/metabolismo , Receptor EphA2/genética , Hipersensibilidade Respiratória/induzido quimicamente , Hipersensibilidade Respiratória/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Th2/imunologia
12.
Cancer Biol Ther ; 8(11): 1027-34, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19395869

RESUMO

EphA2 gene silencing has been shown to result in antitumor efficacy. Here we considered whether silencing additional targets downstream of EphA2 would further enhance the therapeutic effect. EphA2 targeted siRNA was tested in combination with either FAK or Src targeted siRNA using DOPC nanoliposomes in orthotopic models of ovarian carcinoma. The effects of therapy were determined by changes in tumor weight, proliferation (Ki-67), and microvessel density (CD31). In our initial in vivo study, EphA2 plus FAK silencing resulted in the greatest reduction in tumor growth (by 73%, p < 0.005) as compared to control siRNA alone. In the SKOV3ip1 and HeyA8 ovarian cancer models, EphA2 siRNA-DOPC treatment resulted in a 50-67% decrease in tumor growth (p < 0.02, for both), and FAK siRNA-DOPC resulted in a 61-62% decrease in tumor growth (p < 0.009, p < 0.05, respectively). EphA2 plus FAK siRNA-DOPC treatment resulted in a significant reduction (SKOV3ip1: 76%, p < 0.007, HeyA8: 90%, p < 0.003) in tumor growth compared to control siRNA-DOPC. Combination treatment with EphA2 + FAK siRNA-DOPC resulted in significant decreases in tumor cell proliferation (p < 0.001) and microvessel density compared to control siRNA-DOPC (80%; p < 0.001), or the monotherapy groups (p values <0.001). These data suggest that the antitumor efficacy of in vivo EphA2 targeting is enhanced in combination with FAK silencing. Dual targeting of EphA2 and FAK may have therapeutic implications for ovarian cancer management.


Assuntos
Proteína-Tirosina Quinases de Adesão Focal/genética , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/terapia , Receptor EphA2/genética , Inibidores da Angiogênese/genética , Animais , Apoptose/genética , Processos de Crescimento Celular/genética , Modelos Animais de Doenças , Feminino , Proteína-Tirosina Quinases de Adesão Focal/deficiência , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Humanos , Imuno-Histoquímica , Antígeno Ki-67/metabolismo , Camundongos , Camundongos Nus , Neoplasias Ovarianas/enzimologia , Neoplasias Ovarianas/patologia , RNA Interferente Pequeno/genética , Receptor EphA2/deficiência
13.
Mol Biol Cell ; 20(10): 2572-81, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19321667

RESUMO

Eph receptor tyrosine kinases, including EphA2, are expressed in the mammary gland. However, their role in mammary gland development remains poorly understood. Using EphA2-deficient animals, we demonstrate for the first time that EphA2 receptor function is required for mammary epithelial growth and branching morphogenesis. Loss of EphA2 decreased penetration of mammary epithelium into fat pad, reduced epithelial proliferation, and inhibited epithelial branching. These defects appear to be intrinsic to loss of EphA2 in epithelium, as transplantation of EphA2-deficient mammary tissue into wild-type recipient stroma recapitulated these defects. In addition, HGF-induced mammary epithelial branching morphogenesis was significantly reduced in EphA2-deficient cells relative to wild-type cells, which correlated with elevated basal RhoA activity. Moreover, inhibition of ROCK kinase activity in EphA2-deficient mammary epithelium rescued branching defects in primary three-dimensional cultures. These results suggest that EphA2 receptor acts as a positive regulator in mammary gland development, functioning downstream of HGF to regulate branching through inhibition of RhoA. Together, these data demonstrate a positive role for EphA2 during normal mammary epithelial proliferation and branching morphogenesis.


Assuntos
Glândulas Mamárias Animais/enzimologia , Glândulas Mamárias Animais/crescimento & desenvolvimento , Morfogênese , Receptor EphA2/metabolismo , Animais , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Efrina-A1/metabolismo , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/enzimologia , Epitélio/efeitos dos fármacos , Epitélio/enzimologia , Fator de Crescimento de Hepatócito/farmacologia , Ligantes , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/efeitos dos fármacos , Camundongos , Modelos Biológicos , Morfogênese/efeitos dos fármacos , Receptor EphA2/deficiência , Proteína rhoA de Ligação ao GTP/metabolismo
14.
Int J Cancer ; 124(6): 1366-71, 2009 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-19089910

RESUMO

The Eph receptor A2 (EphA2) is overexpressed in a range of human epithelial cancers, a phenotype that is associated with cancer cell proliferation, progression and angiogenesis. Mouse models of mammary neoplasia have confirmed the role of EphA2 as mice carrying a knockout allele of EphA2 were resistant to breast cancer, a phenotype that was associated with interactions between EphA2 and ErbB2. We investigated in vivo the role of EphA2 in GI cancer. To determine whether EphA2 influences intestinal tumorigenesis, we used qRT-PCR to examine the mRNA expression levels of EphA2 in tumors from the small intestine and colon of Apc(Min/+) mice. We found that EphA2 was significantly up-regulated in tumors from both regions when compared with normal control tissues. We then evaluated the spatial expression patterns of EphA2 protein using immunohistochemistry in both the small intestine and colon and found that in normal tissues EphA2 was robustly expressed in highly differentiated cells, such as cells of the villi, but that EphA2 expression was largely absent from the stem cell niche and proliferative zones of intestinal crypts. In contrast, in tumors EphA2 was broadly expressed. Finally, we created a strain of Apc(Min/+) mice carrying a genetic knockout of the EphA2 gene. These mice developed significantly fewer and smaller tumors in both the small and large intestine. Overall, our results indicate that EphA2 plays an oncogenic role in the mammalian intestine suggesting that strategies to target EphA2 activity may offer new therapeutic modalities for colorectal cancer.


Assuntos
Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Neoplasias/genética , Neoplasias/patologia , Receptor EphA2/deficiência , Animais , Mapeamento Cromossômico , Cromossomos Humanos Par 1 , Primers do DNA , Progressão da Doença , Feminino , Genótipo , Humanos , Intestinos/citologia , Intestinos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Metástase Neoplásica/genética , Reação em Cadeia da Polimerase , Receptor EphA2/genética
15.
Cancer Res ; 66(14): 7050-8, 2006 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-16849550

RESUMO

EphA2 receptor tyrosine kinase is frequently overexpressed in different human cancers, suggesting that it may promote tumor development and progression. However, evidence also exists that EphA2 may possess antitumorigenic properties, raising a critical question on the role of EphA2 kinase in tumorigenesis in vivo. We report here that deletion of EphA2 in mouse led to markedly enhanced susceptibility to 7,12-dimethylbenz(a)anthracene/12-O-tetradecanoylphorbol-13-acetate (DMBA/TPA) two-stage skin carcinogenesis. EphA2-null mice developed skin tumors with an increased frequency and shortened latency. Moreover, tumors in homozygous knockout mice grew faster and were twice as likely to show invasive malignant progression. Haploinsufficiency of EphA2 caused an intermediate phenotype in tumor development but had little effects on invasive progression. EphA2 and ephrin-A1 exhibited compartmentalized expression pattern in mouse skin that localized EphA2/ephrin-A1 interactions to the basal layer of epidermis, which was disrupted in tumors. Loss of EphA2 increased tumor cell proliferation, whereas apoptosis was not affected. In vitro, treatment of primary keratinocytes from wild-type mice with ephrin-A1 suppressed cell proliferation and inhibited extracellular signal-regulated kinase 1/2 (ERK1/2) activities. Both effects were abolished in EphA2-null keratinocytes, suggesting that loss of ERK inhibition by EphA2 may be one of the contributing mechanisms for increased tumor susceptibility. Interestingly, despite its tumor suppressive function, EphA2 was overexpressed in skin tumors compared with surrounding normal skin in wild-type mice, similar to the observations in human cancers. EphA2 overexpression may represent a compensatory feedback mechanism during tumorigenesis. Together, these results show that EphA2 is a novel tumor suppressor gene in mammalian skin.


Assuntos
Transformação Celular Neoplásica/metabolismo , Receptor EphA2/deficiência , Neoplasias Cutâneas/enzimologia , Pele/enzimologia , 9,10-Dimetil-1,2-benzantraceno , Animais , Carcinógenos , Processos de Crescimento Celular/fisiologia , Ativação Enzimática , Efrina-A1/biossíntese , Efrina-A1/genética , Efrina-A1/farmacologia , Feminino , Predisposição Genética para Doença , Queratinócitos/citologia , Queratinócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Receptor EphA2/biossíntese , Receptor EphA2/genética , Pele/efeitos dos fármacos , Neoplasias Cutâneas/induzido quimicamente , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia , Acetato de Tetradecanoilforbol
16.
Nat Neurosci ; 8(8): 1022-7, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16025107

RESUMO

In mammals, retinal ganglion cell (RGC) projections initially intermingle and then segregate into a stereotyped pattern of eye-specific layers in the dorsal lateral geniculate nucleus (dLGN). Here we found that in mice deficient for ephrin-A2, ephrin-A3 and ephrin-A5, eye-specific inputs segregated but the shape and location of eye-specific layers were profoundly disrupted. In contrast, mice that lacked correlated retinal activity did not segregate eye-specific inputs. Inhibition of correlated neural activity in ephrin mutants led to overlapping retinal projections that were located in inappropriate regions of the dLGN. Thus, ephrin-As and neural activity act together to control patterning of eye-specific retinogeniculate layers.


Assuntos
Padronização Corporal/fisiologia , Efrina-A2/fisiologia , Efrina-A3/fisiologia , Efrina-A5/fisiologia , Corpos Geniculados/fisiologia , Células Ganglionares da Retina/fisiologia , Transmissão Sináptica/fisiologia , Animais , Mapeamento Encefálico , Efrina-A2/deficiência , Efrina-A3/deficiência , Efrina-A5/deficiência , Camundongos , Camundongos Knockout , Receptor EphA2/deficiência , Receptor EphA3/deficiência , Receptor EphA5/deficiência , Vias Visuais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...