Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 118(29)2021 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-34266948

RESUMO

Hypoxia is an important phenomenon in solid tumors that contributes to metastasis, tumor microenvironment (TME) deregulation, and resistance to therapies. The receptor tyrosine kinase AXL is an HIF target, but its roles during hypoxic stress leading to the TME deregulation are not well defined. We report here that the mammary gland-specific deletion of Axl in a HER2+ mouse model of breast cancer leads to a normalization of the blood vessels, a proinflammatory TME, and a reduction of lung metastases by dampening the hypoxic response in tumor cells. During hypoxia, interfering with AXL reduces HIF-1α levels altering the hypoxic response leading to a reduction of hypoxia-induced epithelial-to-mesenchymal transition (EMT), invasion, and production of key cytokines for macrophages behaviors. These observations suggest that inhibition of Axl generates a suitable setting to increase immunotherapy. Accordingly, combining pharmacological inhibition of Axl with anti-PD-1 in a preclinical model of HER2+ breast cancer reduces the primary tumor and metastatic burdens, suggesting a potential therapeutic approach to manage HER2+ patients whose tumors present high hypoxic features.


Assuntos
Neoplasias da Mama/genética , Neoplasias da Mama/imunologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/imunologia , Imunoterapia , Proteínas Proto-Oncogênicas/genética , Receptores Proteína Tirosina Quinases/genética , Animais , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/fisiopatologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Feminino , Deleção de Genes , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Marcação de Genes , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Inibidores de Checkpoint Imunológico/administração & dosagem , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Camundongos , Metástase Neoplásica/tratamento farmacológico , Metástase Neoplásica/genética , Metástase Neoplásica/imunologia , Receptor de Morte Celular Programada 1/genética , Receptor de Morte Celular Programada 1/imunologia , Proteínas Proto-Oncogênicas/imunologia , Receptores Proteína Tirosina Quinases/imunologia , Microambiente Tumoral/efeitos dos fármacos , Receptor Tirosina Quinase Axl
2.
Cell Mol Life Sci ; 79(1): 37, 2021 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-34971428

RESUMO

The roles of nitric oxide (NO) and endothelial NO synthase (eNOS) in the regulation of angiogenesis are well documented. However, the involvement of eNOS in the sprouting of endothelial tip-cells at the vascular front during sprouting angiogenesis remains poorly defined. In this study, we show that downregulation of eNOS markedly inhibits VEGF-stimulated migration of endothelial cells but increases their polarization, as evidenced by the reorientation of the Golgi in migrating monolayers and by the fewer filopodia on tip cells at ends of sprouts in endothelial cell spheroids. The effect of eNOS inhibition on EC polarization was prevented in Par3-depleted cells. Importantly, downregulation of eNOS increased the expression of polarity genes, such as PARD3B, PARD6A, PARD6B, PKCΖ, TJP3, and CRB1 in endothelial cells. In retinas of eNOS knockout mice, vascular development is retarded with decreased vessel density and vascular branching. Furthermore, tip cells at the extremities of the vascular front have a marked reduction in the number of filopodia per cell and are more oriented. In a model of oxygen-induced retinopathy (OIR), eNOS deficient mice are protected during the initial vaso-obliterative phase, have reduced pathological neovascularization, and retinal endothelial tip cells have fewer filopodia. Single-cell RNA sequencing of endothelial cells from OIR retinas revealed enrichment of genes related to cell polarity in the endothelial tip-cell subtype of eNOS deficient mice. These results indicate that inhibition of eNOS alters the polarity program of endothelial cells, which increases cell polarization, regulates sprouting angiogenesis and normalizes pathological neovascularization during retinopathy.


Assuntos
Neovascularização Patológica , Neovascularização Fisiológica , Óxido Nítrico Sintase Tipo III/fisiologia , Retina/metabolismo , Neovascularização Retiniana , Vasos Retinianos , Animais , Bovinos , Linhagem Celular , Movimento Celular , Polaridade Celular , Células Endoteliais , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Retina/citologia , Retina/patologia , Neovascularização Retiniana/metabolismo , Neovascularização Retiniana/patologia , Vasos Retinianos/citologia , Vasos Retinianos/patologia
3.
Proc Natl Acad Sci U S A ; 116(10): 4538-4547, 2019 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-30787185

RESUMO

Diabetic macular edema is a major complication of diabetes resulting in loss of central vision. Although heightened vessel leakiness has been linked to glial and neuronal-derived factors, relatively little is known on the mechanisms by which mature endothelial cells exit from a quiescent state and compromise barrier function. Here we report that endothelial NOTCH1 signaling in mature diabetic retinas contributes to increased vascular permeability. By providing both human and mouse data, we show that NOTCH1 ligands JAGGED1 and DELTA LIKE-4 are up-regulated secondary to hyperglycemia and activate both canonical and rapid noncanonical NOTCH1 pathways that ultimately disrupt endothelial adherens junctions in diabetic retinas by causing dissociation of vascular endothelial-cadherin from ß-catenin. We further demonstrate that neutralization of NOTCH1 ligands prevents diabetes-induced retinal edema. Collectively, these results identify a fundamental process in diabetes-mediated vascular permeability and provide translational rational for targeting the NOTCH pathway (primarily JAGGED1) in conditions characterized by compromised vascular barrier function.


Assuntos
Permeabilidade Capilar , Retinopatia Diabética/patologia , Receptor Notch1/metabolismo , Transdução de Sinais , Proteínas Adaptadoras de Transdução de Sinal/biossíntese , Animais , Antígenos CD/metabolismo , Caderinas/metabolismo , Proteínas de Ligação ao Cálcio/biossíntese , Ativação Enzimática , Hiperglicemia/metabolismo , Proteína Jagged-1/biossíntese , Camundongos , Óxido Nítrico/biossíntese , Vasos Retinianos/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Quinases da Família src/metabolismo
4.
J Biol Chem ; 292(29): 12178-12191, 2017 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-28600358

RESUMO

Developmental angiogenesis and the maintenance of the blood-brain barrier involve endothelial cell adhesion, which is linked to cytoskeletal dynamics. GPR124 (also known as TEM5/ADGRA2) is an adhesion G protein-coupled receptor family member that plays a pivotal role in brain angiogenesis and in ensuring a tight blood-brain barrier. However, the signaling properties of GPR124 remain poorly defined. Here, we show that ectopic expression of GPR124 promotes cell adhesion, additive to extracellular matrix-dependent effect, coupled with filopodia and lamellipodia formation and an enrichment of a pool of the G protein-coupled receptor at actin-rich cellular protrusions containing VASP, a filopodial marker. Accordingly, GPR124-expressing cells also displayed increased activation of both Rac and Cdc42 GTPases. Mechanistically, we uncover novel direct interactions between endogenous GPR124 and the Rho guanine nucleotide exchange factors Elmo/Dock and intersectin (ITSN). Small fragments of either Elmo or ITSN1 that bind GPR124 blocked GPR124-induced cell adhesion. In addition, Gßγ interacts with the C-terminal tail of GPR124 and promotes the formation of a GPR124-Elmo complex. Furthermore, GPR124 also promotes the activation of the Elmo-Dock complex, as measured by Elmo phosphorylation on a conserved C-terminal tyrosine residue. Interestingly, Elmo and ITSN1 also interact with each other independently of their GPR124-recognition regions. Moreover, endogenous phospho-Elmo and ITSN1 co-localize with GPR124 at lamellipodia of adhering endothelial cells, where GPR124 expression contributes to polarity acquisition during wound healing. Collectively, our results indicate that GPR124 promotes cell adhesion via Elmo-Dock and ITSN. This constitutes a previously unrecognized complex formed of atypical and conventional Rho guanine nucleotide exchange factors for Rac and Cdc42 that is putatively involved in GPR124-dependent angiogenic responses.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Endotélio Vascular/metabolismo , Processamento de Proteína Pós-Traducional , Receptores Acoplados a Proteínas G/metabolismo , Proteínas rac de Ligação ao GTP/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas Adaptadoras de Transporte Vesicular/química , Animais , Células COS , Adesão Celular , Células Cultivadas , Chlorocebus aethiops , Endotélio Vascular/citologia , Células HEK293 , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Fosforilação , Domínios e Motivos de Interação entre Proteínas , Multimerização Proteica , Transporte Proteico , Pseudópodes/metabolismo , Interferência de RNA , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/genética , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Proteínas rac de Ligação ao GTP/química
5.
Mol Cell ; 39(3): 468-76, 2010 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-20705246

RESUMO

Disruption of adherens junctions between endothelial cells results in compromised endothelial barrier function and in altered angiogenesis. Nitric oxide (NO) produced by endothelial NO synthase (eNOS) is essential for increased vascular permeability induced by vascular endothelial growth factor (VEGF). However, the molecular mechanisms by which NO modulates endothelial permeability remain elusive. Here, we show that, within adherens junctions, beta-catenin is a substrate for S-nitrosylation by NO. Stimulation of endothelial cells with VEGF induces S-nitrosylation of beta-catenin, which is dependent on expression and activity of eNOS. Furthermore, VEGF-induced S-nitrosylation of beta-catenin is inhibited in eNOS(-/-) mice. We identify Cys619, located within the VE-cadherin interaction site, as the major S-nitrosylation locus in response to VEGF. Inhibition of S-nitrosylation at Cys619 prevents NO-dependent dissociation of beta-catenin from VE-cadherin and disassembly of adherens junction complexes and inhibits VEGF-stimulated endothelial permeability. Thus, we identify S-nitrosylation of beta-catenin as a modulator of intercellular contacts between endothelial cells.


Assuntos
Permeabilidade Capilar/fisiologia , Células Endoteliais/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Óxido Nítrico/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , beta Catenina/metabolismo , Junções Aderentes/genética , Junções Aderentes/metabolismo , Animais , Antígenos CD/genética , Antígenos CD/metabolismo , Caderinas/genética , Caderinas/metabolismo , Permeabilidade Capilar/efeitos dos fármacos , Células Cultivadas , Cisteína/genética , Cisteína/metabolismo , Camundongos , Camundongos Knockout , Óxido Nítrico/genética , Óxido Nítrico Sintase Tipo III/genética , Fator A de Crescimento do Endotélio Vascular/farmacologia , beta Catenina/genética
6.
Mol Cell Proteomics ; 15(5): 1511-25, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26846344

RESUMO

VEGF and angiopoietin-1 (Ang-1) are essential factors to promote angiogenesis through regulation of a plethora of signaling events in endothelial cells (ECs). Although pathways activated by VEGF and Ang-1 are being established, the unique signaling nodes conferring specific responses to each factor remain poorly defined. Thus, we conducted a large-scale comparative phosphoproteomic analysis of signaling pathways activated by VEGF and Ang-1 in ECs using mass spectrometry. Analysis of VEGF and Ang-1 networks of regulated phosphoproteins revealed that the junctional proteins ZO-1, ZO-2, JUP and p120-catenin are part of a cluster of proteins phosphorylated following VEGF stimulation that are linked to MAPK1 activation. Down-regulation of these junctional proteins led to MAPK1 activation and accordingly, increased proliferation of ECs stimulated specifically by VEGF, but not by Ang-1. We identified ZO-1 as the central regulator of this effect and showed that modulation of cellular ZO-1 levels is necessary for EC proliferation during vascular development of the mouse postnatal retina. In conclusion, we uncovered ZO-1 as part of a signaling node activated by VEGF, but not Ang-1, that specifically modulates EC proliferation during angiogenesis.


Assuntos
Angiopoietina-1/metabolismo , Células Endoteliais/citologia , Proteômica/métodos , Retina/crescimento & desenvolvimento , Fator A de Crescimento do Endotélio Vascular/metabolismo , Proteína da Zônula de Oclusão-1/metabolismo , Animais , Bovinos , Linhagem Celular , Proliferação de Células , Células Endoteliais/metabolismo , Regulação da Expressão Gênica , Humanos , Espectrometria de Massas/métodos , Camundongos , Neovascularização Fisiológica , Fosfoproteínas/metabolismo , Retina/metabolismo , Transdução de Sinais
7.
Blood ; 120(16): 3371-81, 2012 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-22936663

RESUMO

Angiogenic sprouting requires that cell-cell contacts be maintained during migration of endothelial cells. Angiopoietin-1 (Ang-1) and vascular endothelial growth factor act oppositely on endothelial cell junctions. We found that Ang-1 promotes collective and directional migration and, in contrast to VEGF, induces the formation of a complex formed of atypical protein kinase C (PKC)-ζ and ß-catenin at cell-cell junctions and at the leading edge of migrating endothelial cells. This complex brings Par3, Par6, and adherens junction proteins at the front of migrating cells to locally activate Rac1 in response to Ang-1. The colocalization of PKCζ and ß-catenin at leading edge along with PKCζ-dependent stabilization of cell-cell contacts promotes directed and collective endothelial cell migration. Consistent with these results, down-regulation of PKCζ in endothelial cells alters Ang-1-induced sprouting in vitro and knockdown in developing zebrafish results in intersegmental vessel defects caused by a perturbed directionality of tip cells and by loss of cell contacts between tip and stalk cells. These results reveal that PKCζ and ß-catenin function in a complex at adherens junctions and at the leading edge of migrating endothelial cells to modulate collective and directional migration during angiogenesis.


Assuntos
Angiopoietina-1/farmacologia , Movimento Celular/fisiologia , Endotélio Vascular/metabolismo , Neovascularização Fisiológica/fisiologia , Proteína Quinase C/metabolismo , beta Catenina/metabolismo , Junções Aderentes/metabolismo , Animais , Animais Geneticamente Modificados , Aorta/citologia , Aorta/metabolismo , Células COS , Bovinos , Movimento Celular/efeitos dos fármacos , Polaridade Celular , Células Cultivadas , Chlorocebus aethiops , Embrião não Mamífero/citologia , Embrião não Mamífero/metabolismo , Endotélio Vascular/citologia , Imunofluorescência , Junções Intercelulares/metabolismo , Microinjeções , Cicatrização , Peixe-Zebra/genética , Peixe-Zebra/crescimento & desenvolvimento , Peixe-Zebra/metabolismo
8.
Nat Commun ; 15(1): 4405, 2024 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-38782923

RESUMO

Zonula occludens-1 (ZO-1) is involved in the regulation of cell-cell junctions between endothelial cells (ECs). Here we identify the ZO-1 protein interactome and uncover ZO-1 interactions with RNA-binding proteins that are part of stress granules (SGs). Downregulation of ZO-1 increased SG formation in response to stress and protected ECs from cellular insults. The ZO-1 interactome uncovered an association between ZO-1 and Y-box binding protein 1 (YB-1), a constituent of SGs. Arsenite treatment of ECs decreased the interaction between ZO-1 and YB-1, and drove SG assembly. YB-1 expression is essential for SG formation and for the cytoprotective effects induced by ZO-1 downregulation. In the developing retinal vascular plexus of newborn mice, ECs at the front of growing vessels express less ZO-1 but display more YB-1-positive granules than ECs located in the vascular plexus. Endothelial-specific deletion of ZO-1 in mice at post-natal day 7 markedly increased the presence of YB-1-positive granules in ECs of retinal blood vessels, altered tip EC morphology and vascular patterning, resulting in aberrant endothelial proliferation, and arrest in the expansion of the retinal vasculature. Our findings suggest that, through its interaction with YB-1, ZO-1 controls SG formation and the response of ECs to stress during angiogenesis.


Assuntos
Células Endoteliais , Proteína 1 de Ligação a Y-Box , Proteína da Zônula de Oclusão-1 , Animais , Proteína 1 de Ligação a Y-Box/metabolismo , Proteína 1 de Ligação a Y-Box/genética , Proteína da Zônula de Oclusão-1/metabolismo , Proteína da Zônula de Oclusão-1/genética , Camundongos , Humanos , Células Endoteliais/metabolismo , Grânulos de Estresse/metabolismo , Neovascularização Fisiológica , Vasos Retinianos/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Angiogênese , Fatores de Transcrição
9.
J Biol Chem ; 287(51): 43180-90, 2012 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-23105101

RESUMO

Protein-tyrosine phosphatase (PTP)-PEST (PTPN12) is ubiquitously expressed. It is essential for normal embryonic development and embryonic viability in mice. Herein we addressed the involvement of PTP-PEST in endothelial cell functions using a combination of genetic and biochemical approaches. By generating primary endothelial cells from an inducible PTP-PEST-deficient mouse, we found that PTP-PEST is not needed for endothelial cell differentiation and proliferation or for the control of endothelial cell permeability. Nevertheless, it is required for integrin-mediated adhesion and migration of endothelial cells. PTP-PEST-deficient endothelial cells displayed increased tyrosine phosphorylation of Cas, paxillin, and Pyk2, which were previously also implicated in integrin functions. By eliminating PTP-PEST in endothelial cells in vivo, we obtained evidence that expression of PTP-PEST in endothelial cells is required for normal vascular development and embryonic viability. Therefore, PTP-PEST is a key regulator of integrin-mediated functions in endothelial cells seemingly through its capacity to control Cas, paxillin, and Pyk2. This function explains at least in part the essential role of PTP-PEST in embryonic development and viability.


Assuntos
Vasos Sanguíneos/embriologia , Permeabilidade da Membrana Celular , Movimento Celular , Embrião de Mamíferos/enzimologia , Células Endoteliais/citologia , Células Endoteliais/enzimologia , Proteína Tirosina Fosfatase não Receptora Tipo 12/metabolismo , Animais , Adesão Celular , Diferenciação Celular , Proliferação de Células , Forma Celular , Embrião de Mamíferos/citologia , Feminino , Quinase 2 de Adesão Focal/metabolismo , Genótipo , Integrinas/metabolismo , Junções Intercelulares/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Paxilina/metabolismo , Fosforilação , Proteína Tirosina Fosfatase não Receptora Tipo 12/deficiência
10.
Arterioscler Thromb Vasc Biol ; 32(10): 2484-92, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22859491

RESUMO

OBJECTIVE: Vascular endothelial growth factor (VEGF) signaling to endothelial NO synthase (eNOS) plays a central role in angiogenesis. In endothelial cells (ECs), heat-shock protein 90 (Hsp90) is also a regulator of eNOS activity. Our study is designed to determine whether modulation of the activator of Hsp90 ATPase 1 (AHA1) regulates the function of Hsp90 in ECs. METHODS AND RESULTS: We show that eNOS phosphorylation on Ser-1179 after VEGF stimulation is significantly reduced in ECs transfected with a small interfering RNA against AHA1. Accordingly, VEGF-stimulated NO production, endothelial permeability, cell migration, and EC invasion in Matrigel implants in mice are reduced in small interfering RNA against AHA1-treated conditions. Furthermore, the induction of eNOS association with Hsp90 after VEGF stimulation is decreased in AHA1-downregulated cells. We also demonstrate that modulation of Hsp90 activity by AHA1 regulates phosphorylation of Hsp90 on Tyr-300. Interestingly, the association of AHA1 with Hsp90 is increased after c-Src-mediated phosphorylation of Hsp90 on Tyr-300. Finally, we show that overexpression of AHA1 in ECs promotes association of eNOS and Hsp90, phosphorylation of Ser-1179 of eNOS, increases NO production, and cell migration. CONCLUSIONS: These results reveal that modulation of Hsp90 activity by AHA1 regulates VEGF signaling to eNOS and angiogenesis.


Assuntos
Endotélio Vascular/metabolismo , Proteínas de Choque Térmico HSP90/metabolismo , Chaperonas Moleculares/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Células COS , Bovinos , Movimento Celular/fisiologia , Células Cultivadas , Chlorocebus aethiops , Regulação para Baixo/fisiologia , Endotélio Vascular/citologia , Humanos , Técnicas In Vitro , Modelos Animais , Neovascularização Fisiológica/fisiologia , Óxido Nítrico/metabolismo , Fosforilação
11.
J Cell Sci ; 123(Pt 24): 4221-30, 2010 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-21081647

RESUMO

Carcinoembryonic antigen cell adhesion molecule-1 (CEACAM1) is an immunoglobulin-like cell surface co-receptor expressed on epithelial, hematopoietic and endothelial cells. CEACAM1 functions as an adhesion molecule, mainly binding to itself or other members of the CEA family. We and others have previously shown that CEACAM1 is crucial for in vivo vascular integrity during ischemic neo-vascularization. Here, we have deciphered the roles of CEACAM1 in normal and pathological vascularization. We have found that Ceacam1-/- mice exhibit a significant increase in basal vascular permeability related to increased basal Akt and endothelial nitric oxide synthase (eNOS) activation in primary murine lung endothelial cells (MLECs). Moreover, CEACAM1 deletion in MLECs inhibits VEGF-mediated nitric oxide (NO) production, consistent with defective VEGF-dependent in vivo permeability in Ceacam1-/- mice. In addition, Ceacam1-null mice exhibit increased permeability of tumor vasculature. Finally, we demonstrate that CEACAM1 is tyrosine-phosphorylated upon VEGF treatment in a SHP-1- and Src-dependent manner, and that the key residues of the long cytoplasmic domain of CEACAM1 are crucial for CEACAM1 phosphorylation and NO production. This data represents the first report, to our knowledge, of a functional link between CEACAM1 and the VEGFR2/Akt/eNOS-mediated vascular permeability pathway.


Assuntos
Permeabilidade Capilar , Antígeno Carcinoembrionário/metabolismo , Motivos de Aminoácidos , Animais , Aorta/efeitos dos fármacos , Aorta/patologia , Aorta/ultraestrutura , Permeabilidade Capilar/efeitos dos fármacos , Antígeno Carcinoembrionário/química , Bovinos , Células Cultivadas , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/enzimologia , Ativação Enzimática/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Pulmão/citologia , Neoplasias Pulmonares/irrigação sanguínea , Neoplasias Pulmonares/patologia , Camundongos , Óxido Nítrico/biossíntese , Óxido Nítrico Sintase Tipo III/metabolismo , Fosforilação/efeitos dos fármacos , Fosfotirosina/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 6/metabolismo , Relação Estrutura-Atividade , Fator A de Crescimento do Endotélio Vascular/farmacologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Quinases da Família src/metabolismo
12.
Cancer Cell ; 4(1): 31-9, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12892711

RESUMO

Tumor vasculature is hyperpermeable to macromolecules compared to normal vasculature; however, the relationship between tumor hyperpermeability and tumor progression is poorly understood. Here we show that a cell-permeable peptide derived from caveolin-1, termed cavtratin, reduces microvascular hyperpermeability and delays tumor progression in mice. These antipermeability and antitumor actions of cavtratin occur in the absence of direct cytostatic or antiangiogenic effects. Cavtratin blocks microvascular permeability by inhibiting endothelial nitric oxide synthase (eNOS), as the antipermeability and antitumor actions of cavtratin are markedly diminished in eNOS knockout mice. Our results support the concepts that hyperpermeability of tumor blood vessels contributes to tumor progression and that blockade of eNOS may be exploited as a novel target for antitumor therapy.


Assuntos
Permeabilidade Capilar , Carcinoma Hepatocelular/prevenção & controle , Carcinoma Pulmonar de Lewis/prevenção & controle , Caveolinas/uso terapêutico , Neovascularização Fisiológica/fisiologia , Fragmentos de Peptídeos/uso terapêutico , Animais , Carcinoma Hepatocelular/irrigação sanguínea , Carcinoma Hepatocelular/patologia , Carcinoma Pulmonar de Lewis/irrigação sanguínea , Carcinoma Pulmonar de Lewis/patologia , Caveolina 1 , Progressão da Doença , Endotélio Vascular/citologia , Inibidores Enzimáticos/farmacologia , Neoplasias Hepáticas Experimentais/irrigação sanguínea , Neoplasias Hepáticas Experimentais/patologia , Neoplasias Hepáticas Experimentais/prevenção & controle , Neoplasias Pulmonares/irrigação sanguínea , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/prevenção & controle , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Nus , Óxido Nítrico Sintase/antagonistas & inibidores , Óxido Nítrico Sintase Tipo II , Óxido Nítrico Sintase Tipo III , Fator A de Crescimento do Endotélio Vascular/fisiologia
13.
Mol Pharmacol ; 80(3): 407-15, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21642394

RESUMO

Excessive synthesis of reactive oxygen species contributes to the pathology of many human diseases and originates from changes in the expression and posttranslational regulation of the transmembrane NADPH oxidases (Noxes). Nox5 is a novel Nox isoform whose activity is regulated by intracellular calcium levels. We have reported that the activity and calcium-sensitivity of Nox5 can also be modulated by direct phosphorylation. However, the kinases that phosphorylate Nox5 have not been identified, and thus, the goal of this study was to determine whether calcium-activated kinases such as calcium/calmodulin-dependent kinase II (CAMKII) are involved. We found that Nox5 activity in bovine aortic endothelial cells was suppressed by two doses of the CAMKII inhibitor 2-(N-[2-hydroxyethyl])-N-(4-methoxybenzenesulfonyl)amino-N-(4-chlorocinnamyl)-N-methylamine (KN-93). In cotransfected COS-7 cells, wild-type and constitutively active CAMKII, but not a dominant-negative, robustly increased basal Nox5 activity. The ability of CAMKII to increase Nox5 activity was also observed with fixed calcium concentrations in an isolated enzyme activity assay. CAMKII did not elevate intracellular calcium or activate other Nox enzymes. In vitro phosphorylation assays revealed that CAMKII can directly phosphorylate Nox5 on Thr494 and Ser498 as detected by phosphorylation state-specific antibodies. Mass spectrometry (MS) analysis revealed the phosphorylation of additional, novel sites at Ser475, Ser502, and Ser675. Of these phosphorylation sites, mutation of only Ser475 to alanine prevented CAMKII-induced increases in Nox5 activity. The ability of CAMKIIα to phosphorylate Ser475 in intact cells was supported by the binding of Nox5 to phosphoprotein-affinity columns and via MS/MS analysis. Together, these results suggest that CAMKII can positively regulate Nox5 activity via the phosphorylation of Ser475.


Assuntos
Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , NADPH Oxidases/metabolismo , Animais , Western Blotting , Células COS , Cálcio/metabolismo , Bovinos , Células Cultivadas , Chlorocebus aethiops , Eletroforese em Gel de Poliacrilamida , Ativação Enzimática , Fosforilação , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
14.
J Biol Chem ; 285(32): 24591-9, 2010 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-20529868

RESUMO

Vascular endothelial growth factor (VEGF) induces angiogenesis and regulates endothelial function via production and release of nitric oxide (NO), an important signaling molecule. The molecular basis leading to NO production involves phosphatidylinositiol-3 kinase (PI3K), Akt, and endothelial nitric-oxide synthase (eNOS) activation. In this study, we have examined whether small GTP-binding proteins of the ADP-ribosylation factor (ARF) family act as molecular switches to regulate signaling cascades activated by VEGF in endothelial cells. Our results show that this growth factor can promote the rapid and transient activation of ARF1. In endothelial cells, this GTPase is present on dynamic plasma membrane ruffles. Inhibition of ARF1 expression, using RNA interference, markedly impaired VEGF-dependent eNOS phosphorylation and NO production by preventing the activation of the PI3K/Akt signaling axis. Furthermore, our data indicate that phosphorylation of Tyr(801), on VEGF receptor 2, is essential for activating Src- and ARF1-dependent signaling events leading to NO release from endothelial cells. Lastly, this mediator is known to regulate a broad variety of endothelial cell functions. Depletion of ARF1 markedly inhibits VEGF-dependent increase of vascular permeability as well as capillary tubule formation, a process important for angiogenesis. Taken together, our data indicate that ARF1 is a novel modulator of VEGF-stimulated NO release and signaling in endothelial cells.


Assuntos
Fator 1 de Ribosilação do ADP/metabolismo , Células Endoteliais/metabolismo , Regulação Enzimológica da Expressão Gênica , Óxido Nítrico Sintase Tipo III/metabolismo , Óxido Nítrico/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Aorta/citologia , Bovinos , Ativação Enzimática , Humanos , Sistema de Sinalização das MAP Quinases , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/metabolismo
15.
Blood ; 114(15): 3343-51, 2009 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-19564638

RESUMO

Vascular endothelial growth factor (VEGF) is a potent angiogenic cytokine that also increases vascular permeability. Nitric oxide (NO) released from endothelial cells, after activation of endothelial NO synthase (eNOS), contributes to proangiogenic and permeability effects of VEGF. Angiopoietin-1 (Ang-1), via Tie2 receptors, shares many of the proangiogenic properties of VEGF on endothelial cells. However, in contrast to VEGF, Ang-1 protects blood vessels from increased plasma leakage, which contributes to their stabilization. Because eNOS-derived NO is central to increased permeability in response to VEGF, we investigated whether Ang-1 interferes with VEGF signaling to eNOS. We demonstrate that Ang-1 stimulation of endothelial cells inhibits VEGF-induced NO release and transendothelial permeability. In contrast to VEGF stimulation, Ang-1 causes a marked protein kinase C (PKC)-dependent increase in phosphorylation of eNOS on the inhibitory Thr(497). Furthermore, using pharmacologic inhibitors, overexpression studies, and small interfering RNA-mediated gene silencing, we demonstrate that atypical PKC zeta is responsible for phosphorylation of eNOS on Thr(497) in response to Ang-1. In addition, PKC zeta knockdown abrogates the capacity of Ang-1 to inhibit VEGF-induced NO release and endothelial permeability. Thus, inhibition of NO production by Ang-1, via phosphorylation of eNOS on Thr(497) by PKC zeta, is responsible, at least in part, for inhibition of VEGF-stimulated endothelial permeability by Ang-1.


Assuntos
Angiopoietina-1/farmacologia , Permeabilidade Capilar/efeitos dos fármacos , Endotélio Vascular/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Proteína Quinase C/metabolismo , Fator A de Crescimento do Endotélio Vascular/farmacologia , Animais , Permeabilidade Capilar/genética , Bovinos , Células Endoteliais , Técnicas de Silenciamento de Genes , Inativação Gênica , Humanos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo III/genética , Fosforilação/efeitos dos fármacos , Proteína Quinase C/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
16.
Nat Med ; 9(3): 357-62, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12598894

RESUMO

Small polybasic peptides derived from the transduction domains of certain proteins, such as the third alpha-helix of the Antennapedia (Antp) homeodomain, can cross the cell membrane through a receptor-independent mechanism. These cell-permeable molecules have been used as 'Trojan horses' to introduce biologically active cargo molecules such as DNA, peptides or proteins into cells. Using these cell-permeable peptides, we have developed an efficient and simple method to increase virally mediated gene delivery and protein expression in vitro and in vivo. Here, we show that cell-permeable peptides increase viral cell entry, improve gene expression at reduced titers of virus and improve efficacy of therapeutically relevant genes in vivo.


Assuntos
Técnicas de Transferência de Genes , Proteínas Nucleares , Peptídeos/metabolismo , Fatores de Transcrição , Replicação Viral/fisiologia , Adenoviridae/genética , Adenoviridae/metabolismo , Sequência de Aminoácidos , Animais , Proteína do Homeodomínio de Antennapedia , Artérias/citologia , Artérias/metabolismo , Células COS , Corantes Fluorescentes/metabolismo , Regulação da Expressão Gênica , Terapia Genética , Membro Posterior/irrigação sanguínea , Proteínas de Homeodomínio/química , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Isquemia , Dados de Sequência Molecular , Músculo Esquelético/metabolismo , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo III , Peptídeos/química , Peptídeos/genética , Proteínas Recombinantes de Fusão/metabolismo , Alinhamento de Sequência
17.
Arterioscler Thromb Vasc Biol ; 29(6): 902-8, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19423865

RESUMO

OBJECTIVE: Sphingosine-1-phosphate (S1P) is a potent bioactive phospholipid responsible for a variety of vascular cell responses. Hypoxia-inducible factor-1 (HIF-1) is a transcriptional activator of genes essential for adaptation to low oxygen. S1P and HIF-1 are both important mediators of vascular cell responses such as migation, proliferation, and survival. Studies have shown that nonhypoxic stimuli can activate HIF-1 in oxygenated conditions. Here, we attempt to determine whether S1P can modulate the vascular activation of HIF-1. METHODS AND RESULTS: We show that in vascular endothelial and smooth muscle cells, activation of the S1P type-2 receptor by S1P strongly increases HIF-1 alpha protein levels, the active subunit of HIF-1. This is achieved through pVHL-independent stabilization of HIF-1 alpha. We demonstrate that the HIF-1 nuclear complex, formed on S1P stimulation, is transcriptionally active and specifically binds to a hypoxia-responsive elements. Moreover, S1P activates the expression of genes known to be closely regulated by HIF-1. CONCLUSIONS: Our results identify S1P as a novel and potent nonhypoxic activator of HIF-1. We believe that understanding the role played by HIF-1 in S1P gene regulation will have a strong impact on different aspects of vascular biology.


Assuntos
Células Endoteliais/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Lisofosfolipídeos/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Esfingosina/análogos & derivados , Animais , Bovinos , Hipóxia Celular , Núcleo Celular/metabolismo , Células Cultivadas , Regulação da Expressão Gênica , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Camundongos , Estabilidade Proteica , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Wistar , Receptores de Lisoesfingolipídeo/metabolismo , Esfingosina/metabolismo , Fatores de Tempo , Transcrição Gênica , Transfecção
18.
Can J Physiol Pharmacol ; 88(10): 968-76, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20962896

RESUMO

Vascular endothelial growth factor (VEGF)-mediated inflammation requires the synthesis of acute platelet-activating factor (PAF) by endothelial cells (ECs). We previously reported that VEGF-mediated PAF synthesis involves the activation of the homodimeric tyrosine kinase receptor VEGFR-2/R-2, leading to the recruitment of p38 and p42/p44 mitogen-activated protein kinases (MAPKs) and activation of secreted group V phospholipase A2 (sPLA2-V). We have also reported that VEGF-A165-mediated prostacyclin (PGI2) synthesis requires VEGFR-1/R-2 heterodimeric receptor activation. Selective activation of VEGF receptors can coordinate the synthesis of pro-PAF and anti-PGI2 inflammatory factors. It is unknown which VEGFR-2 tyrosine phosphorylation site(s) contribute(s) to PAF synthesis. Bovine aortic endothelial cells (BAECs) were transfected with pcDNA vectors encoding for native VEGF receptor-2 (VEGFR-2) cDNA or VEGFR-2 cDNA containing tyrosine phosphorylation sites mutated into phenylalanine residues (Y801F, Y1059F, Y1175F, Y1214F); an empty pcDNA vector was used as a negative control. Treatment of pcDNA-transfected BAECs with VEGF (10⁻9 mol/L) for 15 min increased PAF synthesis by 180%. In BAECs transfected with pcDNA vectors encoding mutated Y801F, Y1059F, Y1175F, or Y1214F VEGFR-2 cDNA, we observed a marked reduction of VEGF-mediated PAF synthesis by 38%, 46%, 69%, and 31%, respectively, compared with BAECs transfected with pcDNA vector encoding VEGFR-2 cDNA. Our data provide a novel insight as to the mechanisms by which VEGF promotes PAF synthesis.


Assuntos
Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Fator de Ativação de Plaquetas/biossíntese , Tirosina/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Sítios de Ligação , Western Blotting , Bovinos , Células Cultivadas , Endotélio Vascular/citologia , Mutagênese Sítio-Dirigida , Fosforilação , Transfecção , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética
19.
Mol Biol Cell ; 18(11): 4659-68, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17855507

RESUMO

Nitric oxide (NO) release from endothelial cells, via endothelial NO synthase (eNOS) activation, is central to the proangiogenic actions of vascular endothelial growth factor (VEGF). VEGF signaling to eNOS is principally mediated by an Akt-dependent phosphorylation of eNOS and by increased association of eNOS to the molecular chaperone, heat-shock protein 90 kDa (Hsp90). Herein, we report that VEGFR-2 activation induces tyrosine phosphorylation of VEGF receptor 2 (VEGFR-2)-associated Hsp90beta. Tyrosine phosphorylation of Hsp90beta in response to VEGF is dependent on internalization of the VEGFR-2 and on Src kinase activation. Furthermore, we demonstrate that c-Src directly phosphorylates Hsp90 on tyrosine 300 residue and that this event is essential for VEGF-stimulated eNOS association to Hsp90 and thus NO release from endothelial cells. Our work identifies Y300 phosphorylation of Hsp90 as a novel regulated posttranslational modification of the chaperone and demonstrates its importance in the proangiogenic actions of VEGF, namely by regulating NO release from endothelial cells.


Assuntos
Proteínas de Choque Térmico HSP90/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Proteína Oncogênica pp60(v-src)/metabolismo , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Bovinos , Células Cultivadas , Chlorocebus aethiops , Lisina/genética , Lisina/metabolismo , Mutação/genética , Óxido Nítrico/metabolismo , Fosforilação , Fosfotirosina/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo
20.
Cells ; 9(1)2020 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-31936361

RESUMO

Angiopoietin-1 (Ang-1) is an important proangiogenic factor also involved in the maintenance of endothelial-barrier integrity. The small GTPase Rap1 is involved in the regulation of adherens junctions through VE-cadherin-mediated adhesion, and in endothelial permeability. While many studies established that Rap1 activation is critical for endothelial cell-cell adhesions, its roles in the antipermeability effects of Ang-1 are ill-defined. Thus, we determined the contribution of Rap1 to Ang-1-stimulated angiogenic effects on endothelial cells (ECs). We found that Rap1 is activated following Ang-1 stimulation and is required for the antipermeability effects of Ang-1 on EC monolayers. Our results also revealed that Rap1 is necessary for EC sprouting stimulated by Ang-1 but had no significant effect on Ang-1-induced EC migration and adhesion. In contrast, downregulation of VE-cadherin markedly increased the adhesiveness of ECs to the substratum, which resulted in inhibition of Ang-1-stimulated migration. These results revealed that Rap1 is central to the effects of Ang-1 at intercellular junctions of ECs, whereas VE-cadherin is also involved in the adhesion of ECs to the extracellular matrix.


Assuntos
Angiopoietina-1/farmacologia , Aorta/fisiologia , Adesão Celular , Comunicação Celular , Endotélio Vascular/fisiologia , Proteínas rap1 de Ligação ao GTP/metabolismo , Animais , Aorta/citologia , Aorta/efeitos dos fármacos , Bovinos , Células Cultivadas , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Transdução de Sinais , Proteínas rap1 de Ligação ao GTP/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA