Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Mol Cell ; 84(2): 277-292.e9, 2024 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-38183983

RESUMO

iRhoms are pseudoprotease members of the rhomboid-like superfamily and are cardinal regulators of inflammatory and growth factor signaling; they function primarily by recognizing transmembrane domains of their clients. Here, we report a mechanistically distinct nuclear function of iRhoms, showing that both human and mouse iRhom2 are non-canonical substrates of signal peptidase complex (SPC), the protease that removes signal peptides from secreted proteins. Cleavage of iRhom2 generates an N-terminal fragment that enters the nucleus and modifies the transcriptome, in part by binding C-terminal binding proteins (CtBPs). The biological significance of nuclear iRhom2 is indicated by elevated levels in skin biopsies of patients with psoriasis, tylosis with oesophageal cancer (TOC), and non-epidermolytic palmoplantar keratoderma (NEPPK); increased iRhom2 cleavage in a keratinocyte model of psoriasis; and nuclear iRhom2 promoting proliferation of keratinocytes. Overall, this work identifies an unexpected SPC-dependent ER-to-nucleus signaling pathway and demonstrates that iRhoms can mediate nuclear signaling.


Assuntos
Psoríase , Transdução de Sinais , Animais , Humanos , Camundongos , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Psoríase/genética , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo
2.
Biochem Soc Trans ; 52(1): 137-150, 2024 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-38323651

RESUMO

Neuropilin-1 (NRP1) is a transmembrane glycoprotein expressed by several cell types including, neurons, endothelial cells (ECs), smooth muscle cells, cardiomyocytes and immune cells comprising macrophages, dendritic cells and T cell subsets. Since NRP1 discovery in 1987 as an adhesion molecule in the frog nervous system, more than 2300 publications on PubMed investigated the function of NRP1 in physiological and pathological contexts. NRP1 has been characterised as a coreceptor for class 3 semaphorins and several members of the vascular endothelial growth factor (VEGF) family. Because the VEGF family is the main regulator of blood and lymphatic vessel growth in addition to promoting neurogenesis, neuronal patterning, neuroprotection and glial growth, the role of NRP1 in these biological processes has been extensively investigated. It is now established that NRP1 promotes the physiological growth of new vessels from pre-existing ones in the process of angiogenesis. Furthermore, several studies have shown that NRP1 mediates signalling pathways regulating pathological vascular growth in ocular neovascular diseases and tumour development. Less defined are the roles of NRP1 in maintaining the function of the quiescent established vasculature in an adult organism. This review will focus on the opposite roles of NRP1 in regulating transforming growth factor ß signalling pathways in different cell types, and on the emerging role of endothelial NRP1 as an atheroprotective, anti-inflammatory factor involved in the response of ECs to shear stress.


Assuntos
Aterosclerose , Neuropilina-1 , Humanos , Adulto , Neuropilina-1/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Células Endoteliais/metabolismo , Angiogênese , Inflamação
4.
J Cell Sci ; 127(Pt 14): 3079-93, 2014 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-24777476

RESUMO

The protein iASPP (encoded by PPP1R13L) is an evolutionarily conserved p53 inhibitor, the expression of which is often upregulated in human cancers. We have recently shown that iASPP is a crucial regulator of epidermal homeostasis. Here, we report that iASPP also acts as autophagy inhibitor in keratinocytes. Our data show that depletion of iASPP protects keratinocytes from apoptosis by modulating the expression of Noxa (also known as PMAIP1). In our model, iASPP expression can affect the fission-fusion cycle, mass and shape of mitochondria. iASPP-silenced keratinocytes display disorganization of cytosolic compartments and increased metabolic stress caused by deregulation of mTORC1 signaling. Moreover, increased levels of lipidated LC3 protein confirmed the activation of autophagy in iASPP-depleted cells. We have identified a novel mechanism modulating autophagy in keratinocytes that relies upon iASPP expression specifically reducing the interaction of Atg5-Atg12 with Atg16L1, an interaction that is essential for autophagosome formation or maturation. Using organotypic culture, we further explored the link between autophagy and differentiation, and we showed that impairing autophagy affects epidermal terminal differentiation. Our data provide an alternative mechanism to explain how epithelial integrity is maintained against environmental stressors and might also improve the understanding of the etiology of skin diseases that are characterized by defects in differentiation and DNA damage responses.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Queratinócitos/citologia , Queratinócitos/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Apoptose/fisiologia , Autofagia/fisiologia , Proteína 5 Relacionada à Autofagia , Proteínas Relacionadas à Autofagia , Proteínas de Transporte/metabolismo , Diferenciação Celular/fisiologia , Células Epidérmicas , Epiderme/metabolismo , Humanos , Proteínas Associadas aos Microtúbulos/metabolismo
5.
EMBO J ; 30(20): 4261-73, 2011 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-21897369

RESUMO

iASPP, an inhibitory member of the ASPP (apoptosis stimulating protein of p53) family, is an evolutionarily conserved inhibitor of p53 which is frequently upregulated in human cancers. However, little is known about the role of iASPP under physiological conditions. Here, we report that iASPP is a critical regulator of epithelial development. We demonstrate a novel autoregulatory feedback loop which controls crucial physiological activities by linking iASPP to p63, via two previously unreported microRNAs, miR-574-3p and miR-720. By investigating its function in stratified epithelia, we show that iASPP participates in the p63-mediated epithelial integrity program by regulating the expression of genes essential for cell adhesion. Silencing of iASPP in keratinocytes by RNA interference promotes and accelerates a differentiation pathway, which also affects and slowdown cellular proliferation. Taken together, these data reveal iASPP as a key regulator of epithelial homeostasis.


Assuntos
Retroalimentação Fisiológica , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Membrana/metabolismo , Fosfoproteínas/metabolismo , Proteínas Repressoras/metabolismo , Transativadores/metabolismo , Animais , Adesão Celular , Diferenciação Celular , Linhagem Celular , Proliferação de Células , Células Cultivadas , Expressão Gênica , Células HEK293 , Humanos , Queratinócitos/metabolismo , Camundongos , MicroRNAs/metabolismo , Interferência de RNA , Pele/metabolismo
7.
J Cell Sci ; 125(Pt 13): 3153-63, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-22454520

RESUMO

3-Phosphoinositide-dependent protein kinase-1 (PDK1) and phospholipase C (PLC)γ1 are two key enzymes in signal transduction that control several intracellular processes. Despite the fact that PLCγ1 has been investigated for several years, the mechanisms of activation of this enzyme are still not completely clear. Similarly, although PDK1 has been mostly investigated for its role in activation of Akt, a crucial enzyme in regulation of several cellular processes, it has become evident recently that the role of PDK1 in physiological and pathological conditions is not limited to Akt activation. Here we demonstrate that PDK1 regulates PLCγ1 activation in a mechanism involving association of the two enzymes and modulation of PLCγ1 tyrosine phosphorylation. We further show that this novel PDK1-PLCγ1 pathway is important for cancer cell invasion. The identification of a PDK1-PLCγ1 pathway reveals the existence of a previously undetected link between two of the most important enzymes in signal transduction. This is likely to have profound consequences for our understanding of several cellular functions that are dependent on phosphoinositides and controlled by PDK1 and PLCγ1.


Assuntos
Regulação Neoplásica da Expressão Gênica , Fosfolipase C gama/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Proteínas Quinases Dependentes de 3-Fosfoinositídeo , Cálcio/metabolismo , Ativação Enzimática , Fator de Crescimento Epidérmico/farmacologia , Receptores ErbB/efeitos dos fármacos , Receptores ErbB/metabolismo , Citometria de Fluxo , Regulação Enzimológica da Expressão Gênica , Células HEK293 , Humanos , Indazóis/farmacologia , Invasividade Neoplásica/genética , Fosfolipase C gama/genética , Fosforilação , Mapeamento de Interação de Proteínas , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/genética , Pirimidinas/farmacologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transfecção
8.
Sci Signal ; 16(786): eabo4863, 2023 05 23.
Artigo em Inglês | MEDLINE | ID: mdl-37220183

RESUMO

Linear and disturbed flow differentially regulate gene expression, with disturbed flow priming endothelial cells (ECs) for a proinflammatory, atheroprone expression profile and phenotype. Here, we investigated the role of the transmembrane protein neuropilin-1 (NRP1) in ECs exposed to flow using cultured ECs, mice with an endothelium-specific knockout of NRP1, and a mouse model of atherosclerosis. We demonstrated that NRP1 was a constituent of adherens junctions that interacted with VE-cadherin and promoted its association with p120 catenin, stabilizing adherens junctions and inducing cytoskeletal remodeling in alignment with the direction of flow. We also showed that NRP1 interacted with transforming growth factor-ß (TGF-ß) receptor II (TGFBR2) and reduced the plasma membrane localization of TGFBR2 and TGF-ß signaling. NRP1 knockdown increased the abundance of proinflammatory cytokines and adhesion molecules, resulting in increased leukocyte rolling and atherosclerotic plaque size. These findings describe a role for NRP1 in promoting endothelial function and reveal a mechanism by which NRP1 reduction in ECs may contribute to vascular disease by modulating adherens junction signaling and promoting TGF-ß signaling and inflammation.


Assuntos
Células Endoteliais , Neuropilina-1 , Receptor do Fator de Crescimento Transformador beta Tipo II , Animais , Camundongos , Junções Aderentes , Endotélio , Caderinas
9.
iScience ; 11: 205-223, 2019 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-30623799

RESUMO

The transmembrane protein neuropilin-1 (NRP1) promotes vascular endothelial growth factor (VEGF) and extracellular matrix signaling in endothelial cells (ECs). Although it is established that NRP1 is essential for angiogenesis, little is known about its role in EC homeostasis. Here, we report that NRP1 promotes mitochondrial function in ECs by preventing iron accumulation and iron-induced oxidative stress through a VEGF-independent mechanism in non-angiogenic ECs. Furthermore, NRP1-deficient ECs have reduced growth and show the hallmarks of cellular senescence. We show that a subcellular pool of NRP1 localizes in mitochondria and interacts with the mitochondrial transporter ATP-binding cassette B8 (ABCB8). NRP1 loss reduces ABCB8 levels, resulting in iron accumulation, iron-induced mitochondrial superoxide production, and iron-dependent EC senescence. Treatment of NRP1-deficient ECs with the mitochondria-targeted antioxidant compound mitoTEMPO or with the iron chelator deferoxamine restores mitochondrial activity, inhibits superoxide production, and protects from cellular senescence. This finding identifies an unexpected role of NRP1 in EC homeostasis.

10.
Dev Biol ; 312(1): 331-43, 2007 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-17950267

RESUMO

The transduction pathways that branch out of fibroblast growth factor signaling are essential for the induction of the mesoderm and the specification of the vertebrate body plan. One of these pathways is thought to control remodeling of the actin cytoskeleton through the Ral binding protein (RLIP also known as RalBP1), an effector of the small G protein Ral. RLIP contains a region of homology with the GTPase-activating protein (GAP) domain involved in the regulation of GTPases of the Rho family. We demonstrate here that the GAP domain of RLIP is responsible for the stability of the actin cytoskeleton in Xenopus laevis embryos. We also demonstrate that the complete N-terminal domain of RLIP containing the mu2 binding domain (mu2BD) and the GAP domain induces disruption of the actin cytoskeleton when targeted to the plasma membrane. Neither domain, however, has any effect on the actin cytoskeleton when individually targeted to the plasma membrane. We also determined that Cdc42-GDP, but neither Rac-GDP nor Rho-GDP, rescues the effect of expression of the membrane-localized Xenopus RLIP on the actin cytoskeleton. We show that the GAP domain of RLIP interacts in vivo with Cdc42-GTP and Cdc42-GDP. Finally, a single mutation (K244A) in the GAP sequence prevented embryos from gastrulating. These results demonstrate that to participate in the control of the actin cytoskeleton, RLIP needs its complete N-terminal region coding for the mu2BD and the GAP domain. We suggest that RLIP, by coordinating two complementary mechanisms, the endocytosis of clathrin-coated pits and the remodeling of cortical actin, participates in the gastrulation process.


Assuntos
Actinas/metabolismo , Citoesqueleto/metabolismo , Gastrulação , Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Proteínas de Xenopus/química , Proteínas de Xenopus/metabolismo , Xenopus laevis/embriologia , Proteína cdc42 de Ligação ao GTP/metabolismo , Sequência de Aminoácidos , Animais , Movimento Celular , Embrião não Mamífero/citologia , Embrião não Mamífero/metabolismo , Gástrula/citologia , Gástrula/metabolismo , Genes Dominantes , Modelos Biológicos , Dados de Sequência Molecular , Ligação Proteica , Estrutura Terciária de Proteína , Deleção de Sequência , Relação Estrutura-Atividade , Xenopus laevis/metabolismo
11.
Nat Commun ; 9(1): 1021, 2018 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-29523849

RESUMO

Hyperproliferative keratinocytes induced by trauma, hyperkeratosis and/or inflammation display molecular signatures similar to those of palmoplantar epidermis. Inherited gain-of-function mutations in RHBDF2 (encoding iRHOM2) are associated with a hyperproliferative palmoplantar keratoderma and squamous oesophageal cancer syndrome (termed TOC). In contrast, genetic ablation of rhbdf2 in mice leads to a thinning of the mammalian footpad, and reduces keratinocyte hyperproliferation and migration. Here, we report that iRHOM2 is a novel target gene of p63 and that both p63 and iRHOM2 differentially regulate cellular stress-associated signalling pathways in normal and hyperproliferative keratinocytes. We demonstrate that p63-iRHOM2 regulates cell survival and response to oxidative stress via modulation of SURVIVIN and Cytoglobin, respectively. Furthermore, the antioxidant compound Sulforaphane downregulates p63-iRHOM2 expression, leading to reduced proliferation, inflammation, survival and ROS production. These findings elucidate a novel p63-associated pathway that identifies iRHOM2 modulation as a potential therapeutic target to treat hyperproliferative skin disease and neoplasia.


Assuntos
Proteínas de Transporte/metabolismo , Proliferação de Células/genética , Carcinoma de Células Escamosas do Esôfago/patologia , Queratinócitos/metabolismo , Estresse Oxidativo/genética , Fosfoproteínas/metabolismo , Transativadores/metabolismo , Animais , Apoptose/genética , Proteínas de Transporte/genética , Linhagem Celular , Sobrevivência Celular/genética , Citoglobina/biossíntese , Feminino , Células HEK293 , Humanos , Isotiocianatos/farmacologia , Camundongos , Camundongos Knockout , Fosfoproteínas/genética , Interferência de RNA , RNA Interferente Pequeno/genética , Espécies Reativas de Oxigênio/metabolismo , Dermatopatias/patologia , Sulfóxidos , Survivina/biossíntese , Transativadores/genética
12.
Nat Commun ; 8: 14174, 2017 01 27.
Artigo em Inglês | MEDLINE | ID: mdl-28128203

RESUMO

Keratin 16 (K16) is a cytoskeletal scaffolding protein highly expressed at pressure-bearing sites of the mammalian footpad. It can be induced in hyperproliferative states such as wound healing, inflammation and cancer. Here we show that the inactive rhomboid protease RHBDF2 (iRHOM2) regulates thickening of the footpad epidermis through its interaction with K16. K16 expression is absent in the thinned footpads of irhom2-/- mice compared with irhom2+/+mice, due to reduced keratinocyte proliferation. Gain-of-function mutations in iRHOM2 underlie Tylosis with oesophageal cancer (TOC), characterized by palmoplantar thickening, upregulate K16 with robust downregulation of its type II keratin binding partner, K6. By orchestrating the remodelling and turnover of K16, and uncoupling it from K6, iRHOM2 regulates the epithelial response to physical stress. These findings contribute to our understanding of the molecular mechanisms underlying hyperproliferation of the palmoplantar epidermis in both physiological and disease states, and how this 'stress' keratin is regulated.


Assuntos
Proteínas de Transporte/metabolismo , Epiderme/fisiologia , Queratina-16/metabolismo , Animais , Proteínas de Transporte/genética , Linhagem Celular , Proliferação de Células/fisiologia , Citoesqueleto/fisiologia , Regulação para Baixo , Células Epidérmicas , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patologia , Feminino , Fibroblastos , Mutação com Ganho de Função , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Queratina-6/metabolismo , Queratinócitos/fisiologia , Ceratodermia Palmar e Plantar/genética , Ceratodermia Palmar e Plantar/patologia , Masculino , Camundongos , Camundongos Knockout , Pressão , RNA Interferente Pequeno/metabolismo , Estresse Fisiológico/fisiologia , Técnicas de Cultura de Tecidos , Regulação para Cima , Cicatrização/fisiologia
13.
Biochem Pharmacol ; 72(11): 1417-22, 2006 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-16959223

RESUMO

The p53 family comprises three genes that encode for p53, p63 and p73. These genes have a significant degree of sequence homology, especially in the central sequence-specific DNA-binding domain. The high homology among the three DNA-binding domains indicates that these transcription factors have identical residues interacting with DNA, and thus potentially can recognize the same transcriptional targets. In this study, we demonstrate that PKCdelta is induced by p63 and p73 in Saos2 cells. The putative human PKCdelta promoter harbours three p53-like binding sites (RE I, RE II, RE III). In order to confirm the transactivation of PKCdelta by p53 family members, we performed transcription assays using the entire or selected regions of the promoter upstream of a luciferase reporter gene. The results obtained demonstrated that, at least in vitro, the p53 family members tested (TAp63alpha, TAp73alpha, DeltaNp63alpha, but not DeltaNp73alpha) were able to drive transcription from the PKCdelta promoter.


Assuntos
Proteínas de Ligação a DNA/genética , Proteínas Nucleares/genética , Proteína Quinase C-delta/genética , Ativação Transcricional/fisiologia , Proteína Supressora de Tumor p53/genética , Proteínas Supressoras de Tumor/genética , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/metabolismo , Humanos , Queratinócitos , Mutagênese Sítio-Dirigida , Proteínas Nucleares/metabolismo , Regiões Promotoras Genéticas , Proteína Quinase C-delta/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor/metabolismo
14.
J Invest Dermatol ; 136(7): 1460-1470, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27021405

RESUMO

Epidermal keratinocytes migrate through the epidermis up to the granular layer where, on terminal differentiation, they progressively lose organelles and convert into anucleate cells or corneocytes. Our report explores the role of autophagy in ensuring epidermal function providing the first comprehensive profile of autophagy marker expression in developing epidermis. We show that autophagy is constitutively active in the epidermal granular layer where by electron microscopy we identified double-membrane autophagosomes. We demonstrate that differentiating keratinocytes undergo a selective form of nucleophagy characterized by accumulation of microtubule-associated protein light chain 3/lysosomal-associated membrane protein 2/p62 positive autolysosomes. These perinuclear vesicles displayed positivity for histone interacting protein, heterochromatin protein 1α, and localize in proximity with Lamin A and B1 accumulation, whereas in newborn mice and adult human skin, we report LC3 puncta coincident with misshaped nuclei within the granular layer. This process relies on autophagy integrity as confirmed by lack of nucleophagy in differentiating keratinocytes depleted from WD repeat domain phosphoinositide interacting 1 or Unc-51 like autophagy activating kinase 1. Final validation into a skin disease model showed that impaired autophagy contributes to the pathogenesis of psoriasis. Lack of LC3 expression in psoriatic skin lesions correlates with parakeratosis and deregulated expression or location of most of the autophagic markers. Our findings may have implications and improve treatment options for patients with epidermal barrier defects.


Assuntos
Autofagia , Núcleo Celular/metabolismo , Epiderme/fisiologia , Queratinócitos/citologia , Proteínas Associadas aos Microtúbulos/metabolismo , Animais , Animais Recém-Nascidos , Diferenciação Celular , Células Cultivadas , Epiderme/embriologia , Humanos , Lamina Tipo A/metabolismo , Lamina Tipo B/metabolismo , Lisossomos/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Complexos Multiproteicos/metabolismo , Fagossomos/metabolismo , Psoríase/patologia , Pele/metabolismo , Serina-Treonina Quinases TOR/metabolismo
15.
Oncotarget ; 7(14): 18325-45, 2016 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-26934321

RESUMO

It is now well established that the enzymes phosphoinositide 3-kinases (PI3Ks) have a key role in the development and progression of many cancer types and indeed PI3Ks inhibitors are currently being tested in clinical trials. Although eight distinct PI3K isoforms exist, grouped into three classes, most of the evidence currently available are focused on one specific isoform with very little known about the potential role of the other members of this family in cancer. Here we demonstrate that the class II enzyme PI3K-C2ß is overexpressed in several human breast cancer cell lines and in human breast cancer specimens. Our data indicate that PI3K-C2ß regulates breast cancer cell growth in vitro and in vivo and that PI3K-C2ß expression in breast tissues is correlated with the proliferative status of the tumor. Specifically we show that downregulation of PI3K-C2ß in breast cancer cell lines reduces colony formation, induces cell cycle arrest and inhibits tumor growth, in particular in an estrogen-dependent in vivo xenograft. Investigation of the mechanism of the PI3K-C2ß-dependent regulation of cell cycle progression and cell growth revealed that PI3K-C2ß regulates cyclin B1 protein levels through modulation of microRNA miR-449a levels. Our data further demonstrate that downregulation of PI3K-C2ß inhibits breast cancer cell invasion in vitro and breast cancer metastasis in vivo. Consistent with this, PI3K-C2ß is highly expressed in lymph-nodes metastases compared to matching primary tumors. These data demonstrate that PI3K-C2ß plays a pivotal role in breast cancer progression and in metastasis development. Our data indicate that PI3K-C2ß may represent a key molecular switch that regulates a rate-limiting step in breast tumor progression and therefore it may be targeted to limit breast cancer spread.


Assuntos
Neoplasias da Mama/enzimologia , Classe II de Fosfatidilinositol 3-Quinases/metabolismo , Animais , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Classe II de Fosfatidilinositol 3-Quinases/genética , Progressão da Doença , Feminino , Xenoenxertos , Humanos , Células MCF-7 , Camundongos , Camundongos Nus , Transdução de Sinais
16.
J Exp Med ; 211(6): 1167-83, 2014 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-24863063

RESUMO

To enable new blood vessel growth, endothelial cells (ECs) express neuropilin 1 (NRP1), and NRP1 associates with the receptor tyrosine kinase VEGFR2 after binding the vascular endothelial growth factor A (VEGF) to enhance arteriogenesis. We report that NRP1 contributes to angiogenesis through a novel mechanism. In human and mouse ECs, the integrin ligand fibronectin (FN) stimulated actin remodeling and phosphorylation of the focal adhesion component paxillin (PXN) in a VEGF/VEGFR2-independent but NRP1-dependent manner. NRP1 formed a complex with ABL1 that was responsible for FN-dependent PXN activation and actin remodeling. This complex promoted EC motility in vitro and during angiogenesis on FN substrates in vivo. Accordingly, both physiological and pathological angiogenesis in the retina were inhibited by treatment with Imatinib, a small molecule inhibitor of ABL1 which is widely used to prevent the proliferation of tumor cells that express BCR-ABL fusion proteins. The finding that NRP1 regulates angiogenesis in a VEGF- and VEGFR2-independent fashion via ABL1 suggests that ABL1 inhibition provides a novel opportunity for anti-angiogenic therapy to complement VEGF or VEGFR2 blockade in eye disease or solid tumor growth.


Assuntos
Benzamidas/farmacologia , Células Endoteliais/efeitos dos fármacos , Neovascularização Fisiológica/efeitos dos fármacos , Neuropilina-1/metabolismo , Piperazinas/farmacologia , Proteínas Proto-Oncogênicas c-abl/metabolismo , Pirimidinas/farmacologia , Animais , Adesão Celular/genética , Movimento Celular/genética , Células Cultivadas , Células Endoteliais/metabolismo , Células Endoteliais/fisiologia , Ativação Enzimática/efeitos dos fármacos , Fibronectinas/metabolismo , Humanos , Mesilato de Imatinib , Immunoblotting , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Microscopia Confocal , Neuropilina-1/genética , Paxilina/metabolismo , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-abl/genética , Interferência de RNA , Neovascularização Retiniana/genética , Neovascularização Retiniana/fisiopatologia , Neovascularização Retiniana/prevenção & controle , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
17.
J Exp Med ; 210(3): 581-603, 2013 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-23420876

RESUMO

The role of apoptosis in melanoma pathogenesis and chemoresistance is poorly characterized. Mutations in TP53 occur infrequently, yet the TP53 apoptotic pathway is often abrogated. This may result from alterations in TP53 family members, including the TP53 homologue TP63. Here we demonstrate that TP63 has an antiapoptotic role in melanoma and is responsible for mediating chemoresistance. Although p63 was not expressed in primary melanocytes, up-regulation of p63 mRNA and protein was observed in melanoma cell lines and clinical samples, providing the first evidence of significant p63 expression in this lineage. Upon genotoxic stress, endogenous p63 isoforms were stabilized in both nuclear and mitochondrial subcellular compartments. Our data provide evidence of a physiological interaction between p63 with p53 whereby translocation of p63 to the mitochondria occurred through a codependent process with p53, whereas accumulation of p53 in the nucleus was prevented by p63. Using RNA interference technology, both isoforms of p63 (TA and ΔNp63) were demonstrated to confer chemoresistance, revealing a novel oncogenic role for p63 in melanoma cells. Furthermore, expression of p63 in both primary and metastatic melanoma clinical samples significantly correlated with melanoma-specific deaths in these patients. Ultimately, these observations provide a possible explanation for abrogation of the p53-mediated apoptotic pathway in melanoma, implicating novel approaches aimed at sensitizing melanoma to therapeutic agents.


Assuntos
Melanoma/tratamento farmacológico , Proteínas de Membrana/fisiologia , Neoplasias Cutâneas/tratamento farmacológico , Adulto , Idoso , Idoso de 80 Anos ou mais , Apoptose , Linhagem Celular Tumoral , Dano ao DNA , Resistencia a Medicamentos Antineoplásicos , Feminino , Citometria de Fluxo , Humanos , Masculino , Melanoma/patologia , Melanoma/secundário , Proteínas de Membrana/análise , Pessoa de Meia-Idade , Mitocôndrias/metabolismo , Prognóstico , Transporte Proteico , Proteínas Proto-Oncogênicas c-mdm2/fisiologia , Neoplasias Cutâneas/patologia , Proteína Supressora de Tumor p53/fisiologia
18.
Biochem Biophys Res Commun ; 361(1): 1-6, 2007 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-17632082

RESUMO

The epidermis is a multi-layered stratified epithelium continuously renewed by differentiating keratinocytes that develops by the action of p63, a member of the p53 family. The TP63 contains two promoters, resulting in the expression of different proteins, containing (TAp63) or not (DeltaNp63) an amino-terminal transactivation domain, which contribution in skin formation is not fully understood. We found that p63 binds and transactivate GATA-3 promoter, which in turn transactivate IKKalpha, two pivotal regulators of epithelial development. Indeed, GATA-3 is a regulator of cell lineage in skin and hair follicles formation. To further study the relationship between GATA-3 and p63 isoforms here we investigated their expression during keratinocyte differentiation, in human epidermis and hair follicle.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Epiderme/metabolismo , Fator de Transcrição GATA3/metabolismo , Folículo Piloso/metabolismo , Queratinócitos/metabolismo , Transativadores/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Anticorpos/imunologia , Diferenciação Celular , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/imunologia , Humanos , Queratinócitos/citologia , Transativadores/química , Transativadores/imunologia , Fatores de Transcrição , Proteínas Supressoras de Tumor/química , Proteínas Supressoras de Tumor/imunologia
19.
J Cell Sci ; 119(Pt 22): 4617-22, 2006 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-17093266

RESUMO

The epidermis, the outer layer of the skin composed of keratinocytes, develops following the action of the transcription factor p63. The mouse Trp63 gene contains two promoters, driving the production of distinct proteins, one with an N-terminal trans-activation domain (TAp63) and one without (DeltaNp63), although their relative contribution to epidermal development is not clearly established. To identify the relative role of p63 isoforms in relation to IKKalpha, also known to be essential for epithelial development, we performed both molecular and in vivo analyses using genetic complementation in mice. We found that the action of TAp63 is mediated at the molecular level by direct and indirect transactivation of IKKalpha and Ets-1, respectively. We also found that DeltaNp63 upregulates IKKalpha indirectly, through GATA-3. Our data are consistent with a role for p63 directly upstream of IKKalpha in epithelial development.


Assuntos
Epiderme/fisiologia , Quinase I-kappa B/metabolismo , Queratinócitos/metabolismo , Fosfoproteínas/metabolismo , Transativadores/metabolismo , Animais , Linhagem Celular Tumoral , Epiderme/metabolismo , Fator de Transcrição GATA3/metabolismo , Humanos , Camundongos , Camundongos Transgênicos , Fosfoproteínas/genética , Regiões Promotoras Genéticas , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estrutura Terciária de Proteína , Proteína Proto-Oncogênica c-ets-1/metabolismo , Transdução de Sinais , Transativadores/genética , Ativação Transcricional , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA