Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
EMBO J ; 40(22): e108125, 2021 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-34617326

RESUMO

Mutations in VAV1, a gene that encodes a multifunctional protein important for lymphocytes, are found at different frequencies in peripheral T-cell lymphoma (PTCL), non-small cell lung cancer, and other tumors. However, their pathobiological significance remains unsettled. After cataloguing 51 cancer-associated VAV1 mutations, we show here that they can be classified in five subtypes according to functional impact on the three main VAV1 signaling branches, GEF-dependent activation of RAC1, GEF-independent adaptor-like, and tumor suppressor functions. These mutations target new and previously established regulatory layers of the protein, leading to quantitative and qualitative changes in VAV1 signaling output. We also demonstrate that the most frequent VAV1 mutant subtype drives PTCL formation in mice. This process requires the concurrent engagement of two downstream signaling branches that promote the chronic activation and transformation of follicular helper T cells. Collectively, these data reveal the genetic constraints associated with the lymphomagenic potential of VAV1 mutant subsets, similarities with other PTCL driver genes, and potential therapeutic vulnerabilities.


Assuntos
Linfócitos T CD4-Positivos/patologia , Linfoma de Células T Periférico/genética , Mutação , Proteínas Proto-Oncogênicas c-vav/genética , Animais , Linfócitos T CD4-Positivos/fisiologia , Linfócitos T CD4-Positivos/transplante , Células COS , Proliferação de Células/genética , Chlorocebus aethiops , Humanos , Células Jurkat , Linfoma de Células T Periférico/patologia , Camundongos Transgênicos , Proteínas Proto-Oncogênicas c-vav/química , Proteínas Proto-Oncogênicas c-vav/metabolismo , Transdução de Sinais , Proteínas rac1 de Ligação ao GTP/genética , Proteínas rac1 de Ligação ao GTP/metabolismo
2.
Biochem J ; 477(19): 3791-3801, 2020 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-32897354

RESUMO

Vav2 is a ubiquitous guanine nucleotide exchange factor (GEF) for Rho family GTPases that is involved in regulating a wide range of biological processes. It interacts with several tyrosine-phosphorylated cell surface receptors, including the Eph family receptors, through its SH2 domain. The interaction of Vav2 with EphA2 is crucial for EphA2-mediated tumor angiogenesis. Here we show that Vav2-SH2 domain is a lipid-binding module that can recognize PI(4,5)P2 and PI(3,4,5)P3 lipids weakly but specifically. The specific lipid-binding site in Vav2-SH2 domain was identified by NMR chemical shift perturbation experiments using the head groups of PI(4,5)P2 and PI(3,4,5)P3, both of which bind to Vav2-SH2 with millimolar binding affinities. In addition, the interaction between Vav2-SH2 and the phosphorylated juxtamembrane region (JM) of EphA2 (Y594 phosphorylated) was investigated using NMR techniques. Furthermore, by using a nickel-lipid containing peptide-based nanodiscs system, we studied the binding of Vav2-SH2 to the phosphorylated JM region of EphA2 on lipid membrane and uncovered a role of membrane environment in modulating this protein-protein recognition.


Assuntos
Efrina-A2/química , Membranas Artificiais , Fosfatidilinositol 4,5-Difosfato/química , Fosfatos de Fosfatidilinositol/química , Proteínas Proto-Oncogênicas c-vav/química , Efrina-A2/metabolismo , Humanos , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Proteínas Proto-Oncogênicas c-vav/metabolismo , Receptor EphA2 , Domínios de Homologia de src
3.
Oncogene ; 39(28): 5098-5111, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32528129

RESUMO

The current paradigm holds that the inhibition of Rho guanosine nucleotide exchange factors (GEFs), the enzymes that stimulate Rho GTPases, can be a valuable therapeutic strategy to treat Rho-dependent tumors. However, formal validation of this idea using in vivo models is still missing. In this context, it is worth remembering that many Rho GEFs can mediate both catalysis-dependent and independent responses, thus raising the possibility that the inhibition of their catalytic activities might not be sufficient per se to block tumorigenic processes. On the other hand, the inhibition of these enzymes can trigger collateral side effects that could preclude the practical implementation of anti-GEF therapies. To address those issues, we have generated mouse models to mimic the effect of the systemic application of an inhibitor for the catalytic activity of the Rho GEF Vav2 at the organismal level. Our results indicate that lowering the catalytic activity of Vav2 below specific thresholds is sufficient to block skin tumor initiation, promotion, and progression. They also reveal that the negative side effects typically induced by the loss of Vav2 can be bypassed depending on the overall level of Vav2 inhibition achieved in vivo. These data underscore the pros and cons of anti-Rho GEF therapies for cancer treatment. They also support the idea that Vav2 could represent a viable drug target.


Assuntos
Proteínas Proto-Oncogênicas c-vav/genética , Fatores de Troca de Nucleotídeo Guanina Rho/genética , Proteínas rac1 de Ligação ao GTP/genética , Proteínas rho de Ligação ao GTP/genética , Animais , Biocatálise , Células COS , Carcinogênese/genética , Chlorocebus aethiops , Cricetinae , Humanos , Camundongos Knockout , Camundongos Transgênicos , Mutação , Proteínas Proto-Oncogênicas c-vav/química , Proteínas Proto-Oncogênicas c-vav/metabolismo , Fatores de Troca de Nucleotídeo Guanina Rho/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo
4.
Cell Signal ; 65: 109438, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31654719

RESUMO

Vav1 is physiologically active as a GDP/GTP nucleotide exchange factor (GEF) in the hematopoietic system. Its wild-type form was recently implicated in mammalian malignancies of hematologic and non-hematologic tissue origins. Moreover, it was recently identified as a mutated gene in human cancers of various origins. In this review we focus on the functional activities of several of the Vav1 mutants analyzed for their tumorigenic properties. We also discuss the relationship of the tested biochemical properties of Vav1 mutants, E59K, D517E and L801P, to their computer-based predicted properties. These comparisons further enhance the need for integration of computation-based structural analyses with experimental data in order to fully appreciate the activity of mutant proteins. Our comprehensive evaluation supports the classification of Vav1 as a bona fide oncogene in human cancers.


Assuntos
Mutação/genética , Oncogenes , Proteínas Proto-Oncogênicas c-vav/genética , Animais , Carcinogênese/genética , Carcinogênese/patologia , Humanos , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Neoplasias/genética , Proteínas Proto-Oncogênicas c-vav/química
5.
J Lipid Res ; 60(12): 2006-2019, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31570505

RESUMO

During foam cell formation and atherosclerosis development, the scavenger receptor CD36 plays critical roles in lipid uptake and triggering of atherogenicity via the activation of Vav molecules. The Vav family includes three highly conserved members known as Vav1, Vav2, and Vav3. As Vav1 and Vav3 were found to exert function in atherosclerosis development, it remains thus to decipher whether Vav2 also plays a role in the development of atherosclerosis. In this study we found that Vav2 deficiency in RAW264.7 macrophages significantly diminished oxidized LDL uptake and CD36 signaling, demonstrating that each Vav protein family member was required for foam cell formation. Genetic disruption of Vav2 in ApoE-deficient C57BL/6 mice significantly inhibited the severity of atherosclerosis. Strikingly, we further found that the genetic deletion of each member of the Vav protein family by CRISPR/Cas9 resulted in a similar alteration of transcriptomic profiles of macrophages. The three members of the Vav proteins were found to form complexes, and genetic ablation of each single Vav molecule was sufficient to prevent endocytosis of CD36. The functional interdependence of the three Vav family members in foam cell formation was due to their indispensable roles in transcriptomic programing, lipid uptake, and activation of the JNK kinase in macrophages.


Assuntos
Aterosclerose/metabolismo , Células Espumosas/citologia , Multimerização Proteica , Proteínas Proto-Oncogênicas c-vav/química , Proteínas Proto-Oncogênicas c-vav/metabolismo , Animais , Apolipoproteínas E/deficiência , Aterosclerose/genética , Sequência de Bases , Antígenos CD36/metabolismo , Diferenciação Celular , Técnicas de Inativação de Genes , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo , Estrutura Quaternária de Proteína , Transporte Proteico , Proteínas Proto-Oncogênicas c-vav/deficiência , Proteínas Proto-Oncogênicas c-vav/genética , Células RAW 264.7
6.
Cells ; 8(5)2019 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-31100928

RESUMO

Vav proteins play roles as guanosine nucleotide exchange factors for Rho GTPases and signaling adaptors downstream of protein tyrosine kinases. The recent sequencing of the genomes of many species has revealed that this protein family originated in choanozoans, a group of unicellular organisms from which animal metazoans are believed to have originated from. Since then, the Vav family underwent expansions and reductions in its members during the evolutionary transitions that originated the agnates, chondrichthyes, some teleost fish, and some neoaves. Exotic members of the family harboring atypical structural domains can be also found in some invertebrate species. In this review, we will provide a phylogenetic perspective of the evolution of the Vav family. We will also pay attention to the structure, signaling properties, regulatory layers, and functions of Vav proteins in both invertebrate and vertebrate species.


Assuntos
Evolução Molecular , Filogenia , Proteínas Proto-Oncogênicas c-vav/genética , Proteínas Proto-Oncogênicas c-vav/metabolismo , Animais , Coanoflagelados/metabolismo , Cordados/metabolismo , Humanos , Estrutura Molecular , Fosforilação , Proteínas Proto-Oncogênicas c-vav/química , Transdução de Sinais , Proteínas rho de Ligação ao GTP/metabolismo
7.
Nat Commun ; 9(1): 4042, 2018 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-30279442

RESUMO

Controlling protein activity with chemogenetics and optogenetics has proven to be powerful for testing hypotheses regarding protein function in rapid biological processes. Controlling proteins by splitting them and then rescuing their activity through inducible reassembly offers great potential to control diverse protein activities. Building split proteins has been difficult due to spontaneous assembly, difficulty in identifying appropriate split sites, and inefficient induction of effective reassembly. Here we present an automated approach to design effective split proteins regulated by a ligand or by light (SPELL). We develop a scoring function together with an engineered domain to enable reassembly of protein halves with high efficiency and with reduced spontaneous assembly. We demonstrate SPELL by applying it to proteins of various shapes and sizes in living cells. The SPELL server (spell.dokhlab.org) offers an automated prediction of split sites.


Assuntos
Optogenética , Engenharia de Proteínas/métodos , Algoritmos , Automação , Inibidores de Dissociação do Nucleotídeo Guanina/química , Proteínas Proto-Oncogênicas c-vav/química , Proteína 1A de Ligação a Tacrolimo/química , Quinases da Família src/química
8.
Zhejiang Da Xue Xue Bao Yi Xue Ban ; 47(1): 75-81, 2018 01 25.
Artigo em Chinês | MEDLINE | ID: mdl-30146815

RESUMO

Vav1, as a key downstream signaling molecule of T cell receptor, includes a catalytic core DH-PH-ZF domain with the function as guanine nucleotide exchange factor (GEF), and a SH3-SH2-SH3 domain with the function as adaptor protein. These two structures of Vav1 play different roles in the development, activation, proliferation and function of T cells, and thereby exert the different regulatory effect on the occurrence and development of autoimmune disease, graft rejection, cancer and other clinical conditions, implicating that Vav1 might be a potential therapeutic target for these diseases. This paper reviews the role of Vav1 in T cells and the occurrence of related diseases.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Proteínas Proto-Oncogênicas c-vav , Linfócitos T , Animais , Doenças Autoimunes/genética , Doenças Autoimunes/fisiopatologia , Humanos , Neoplasias/genética , Neoplasias/fisiopatologia , Proteínas Proto-Oncogênicas c-vav/química , Proteínas Proto-Oncogênicas c-vav/imunologia , Proteínas Proto-Oncogênicas c-vav/metabolismo
9.
Cell Signal ; 40: 133-142, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28927664

RESUMO

Vav1/2/3 comprise a protein family with guanyl nucleotide exchange activity for Rho and Rac as well as with motifs conferring adapter activity. Biologically, Vav1 plays a critical role in hematologic cell signaling, whereas Vav2/3 have a wider tissue distribution, but all 3 Vav proteins are implicated in cancer development. A structural feature of Vav1/2/3 is the presence of an atypical C1 domain, which possesses close structural homology to the typical C1 domains of protein kinase C but which fails to bind the second messenger diacylglycerol or the potent analogs, the phorbol esters. Previously, we have shown that five residues in the Vav1 C1 domain are responsible for its lack of phorbol ester binding. Here, we show that the lack of phorbol ester binding of Vav3 has a similar basis. We then explore the consequences of phorbol ester binding to a modified Vav3 in which the C1 domain has been altered to allow phorbol ester binding. We find both disruption of the guanyl nucleotide exchange activity of the modified Vav 3 as well as a shift in localization to the membrane upon phorbol ester treatment. This change in localization is associated with altered interactions with other signaling proteins. The studies provide a first step in assessing the potential for the design of custom C1 domain targeted molecules selective for the atypical C1 domains of Vav family proteins.


Assuntos
Neoplasias/genética , Domínios Proteicos/genética , Proteína Quinase C/genética , Proteínas Proto-Oncogênicas c-vav/genética , Sequência de Aminoácidos , Humanos , Terapia de Alvo Molecular , Neoplasias/patologia , Neoplasias/terapia , Ésteres de Forbol/química , Proteína Quinase C/química , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-vav/química , Transdução de Sinais/genética
10.
Mol Cancer Res ; 15(11): 1469-1480, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28811363

RESUMO

Castration-resistant prostate cancer (CRPC) progresses rapidly and is incurable. Constitutively active androgen receptor splice variants (AR-Vs) represent a well-established mechanism of therapeutic resistance and disease progression. These variants lack the AR ligand-binding domain and, as such, are not inhibited by androgen deprivation therapy (ADT), which is the standard systemic approach for advanced prostate cancer. Signaling by AR-Vs, including the clinically relevant AR-V7, is augmented by Vav3, an established AR coactivator in CRPC. Using mutational and biochemical studies, we demonstrated that the Vav3 Diffuse B-cell lymphoma homology (DH) domain interacted with the N-terminal region of AR-V7 (and full length AR). Expression of the Vav3 DH domain disrupted Vav3 interaction with and enhancement of AR-V7 activity. The Vav3 DH domain also disrupted AR-V7 interaction with other AR coactivators: Src1 and Vav2, which are overexpressed in PC. This Vav3 domain was used in proof-of-concept studies to evaluate the effects of disrupting the interaction between AR-V7 and its coactivators on CRPC cells. This disruption decreased CRPC cell proliferation and anchorage-independent growth, caused increased apoptosis, decreased migration, and resulted in the acquisition of morphological changes associated with a less aggressive phenotype. While disrupting the interaction between FL-AR and its coactivators decreased N-C terminal interaction, disrupting the interaction of AR-V7 with its coactivators decreased AR-V7 nuclear levels.Implications: This study demonstrates the potential therapeutic utility of inhibiting constitutively active AR-V signaling by disrupting coactivator binding. Such an approach is significant, as AR-Vs are emerging as important drivers of CRPC that are particularly recalcitrant to current therapies. Mol Cancer Res; 15(11); 1469-80. ©2017 AACR.


Assuntos
Neoplasias de Próstata Resistentes à Castração/metabolismo , Proteínas Proto-Oncogênicas c-vav/genética , Proteínas Proto-Oncogênicas c-vav/metabolismo , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Processamento Alternativo , Sítios de Ligação , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Sobrevivência Celular , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Mutação , Neoplasias de Próstata Resistentes à Castração/genética , Neoplasias de Próstata Resistentes à Castração/terapia , Ligação Proteica , Proteínas Proto-Oncogênicas c-vav/química , Receptores Androgênicos/química , Transdução de Sinais , Regulação para Cima
11.
Sci Rep ; 7(1): 1284, 2017 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-28455498

RESUMO

Src homology domain containing leukocyte protein of 65 kDa (SLP65), the growth factor receptor binding protein 2 (Grb2), and the guanine nucleotide exchange factor for the Rho family GTPases (Vav), self associate in unstimulated B cells as components of the preformed B cell receptor transducer module, in an SH3-dependent manner. The complex enables the B cell to promptly respond to BCR aggregation, resulting in signal amplification. It also facilitates Vav translocation to the membrane rafts, for activation. Here we uncover the molecular mechanism by which the complex may be formed in the B cell. The C-terminal SH3 domain (SH3C) of Grb2 bivalently interacts with the atypical non-PxxP proline rich region of SLP65, and the N-terminal SH3 domain (SH3N) of Vav, both the interactions crucial for the proper functioning of the B cell. Most surprisingly, the two ligands bind the same ligand binding site on the surface of Grb2 SH3C. Addition of SLP65 peptide to the Grb2-Vav complex abrogates the interaction completely, displacing Vav. However, the addition of Vav SH3N to the SLP65-Grb2 binary complex, results in a trimeric complex. Extrapolating these results to the in vivo conditions, Grb2 should bind the SLP65 transducer module first, and then Vav should associate.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/química , Proteína Adaptadora GRB2/química , Proteínas Proto-Oncogênicas c-vav/química , Domínios de Homologia de src , Animais , Ligantes , Camundongos , Domínios Proteicos Ricos em Prolina , Ligação Proteica , Sistemas de Translocação de Proteínas/química
12.
Science ; 354(6318): 1441-1444, 2016 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-27980211

RESUMO

Optogenetic and chemogenetic control of proteins has revealed otherwise inaccessible facets of signaling dynamics. Here, we use light- or ligand-sensitive domains to modulate the structural disorder of diverse proteins, thereby generating robust allosteric switches. Sensory domains were inserted into nonconserved, surface-exposed loops that were tight and identified computationally as allosterically coupled to active sites. Allosteric switches introduced into motility signaling proteins (kinases, guanosine triphosphatases, and guanine exchange factors) controlled conversion between conformations closely resembling natural active and inactive states, as well as modulated the morphodynamics of living cells. Our results illustrate a broadly applicable approach to design physiological protein switches.


Assuntos
Luz , Engenharia de Proteínas , Quinases da Família src , Regulação Alostérica/genética , Regulação Alostérica/efeitos da radiação , Sítio Alostérico , Domínio Catalítico , Ativação Enzimática/genética , Ativação Enzimática/efeitos da radiação , GTP Fosfo-Hidrolases/antagonistas & inibidores , GTP Fosfo-Hidrolases/química , GTP Fosfo-Hidrolases/genética , GTP Fosfo-Hidrolases/efeitos da radiação , Fatores de Troca do Nucleotídeo Guanina/antagonistas & inibidores , Fatores de Troca do Nucleotídeo Guanina/química , Fatores de Troca do Nucleotídeo Guanina/genética , Células HEK293 , Humanos , Ligantes , Optogenética , Domínios Proteicos/efeitos da radiação , Proteínas Proto-Oncogênicas c-vav/química , Transdução de Sinais , Quinases da Família src/antagonistas & inibidores , Quinases da Família src/química , Quinases da Família src/genética , Quinases da Família src/efeitos da radiação
13.
Nat Methods ; 13(9): 755-8, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27427858

RESUMO

LOVTRAP is an optogenetic approach for reversible light-induced protein dissociation using protein A fragments that bind to the LOV domain only in the dark, with tunable kinetics and a >150-fold change in the dissociation constant (Kd). By reversibly sequestering proteins at mitochondria, we precisely modulated the proteins' access to the cell edge, demonstrating a naturally occurring 3-mHz cell-edge oscillation driven by interactions of Vav2, Rac1, and PI3K proteins.


Assuntos
Luz , Optogenética/métodos , Fosfatidilinositol 3-Quinase/química , Fotorreceptores de Plantas , Proteínas Proto-Oncogênicas c-vav/química , Proteínas rac1 de Ligação ao GTP/química , Avena/metabolismo , Células HeLa , Humanos , Cinética , Fosfatidilinositol 3-Quinase/genética , Fosfatidilinositol 3-Quinase/efeitos da radiação , Fotorreceptores de Plantas/química , Fotorreceptores de Plantas/genética , Fotorreceptores de Plantas/efeitos da radiação , Mapeamento de Interação de Proteínas , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-vav/genética , Proteínas Proto-Oncogênicas c-vav/efeitos da radiação , Proteínas Recombinantes de Fusão , Proteínas rac1 de Ligação ao GTP/genética , Proteínas rac1 de Ligação ao GTP/efeitos da radiação
14.
FEBS Lett ; 590(6): 857-65, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26919541

RESUMO

Thioredoxin-interacting protein (TXNIP) is a multifunctional protein involved in diverse cellular processes such as cell proliferation and apoptosis. TXNIP stability is controlled by the ubiquitin-proteasome pathway, and the E3 ubiquitin ligase Itch directly interacts with TXNIP via PPxY motifs of TXNIP. In a previously published study, we have shown that phosphorylation of the PPxY tyrosyl residue switches TXNIP selectivity between different binding partners. Here, we describe that tyrosine-phosphorylated PPxY motifs also bind to SH2 domains of Vav2 and Src with dissociation constants around 10 µm and that phosphorylation is indispensable for these interactions as well. The crystal structure of the complex between a phosphorylated PPxY motif, and the SH2 domain of Vav2 reveals a conserved recognition mechanism.


Assuntos
Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Proteínas Proto-Oncogênicas c-vav/química , Proteínas Proto-Oncogênicas c-vav/metabolismo , Sequência de Aminoácidos , Proteínas de Transporte/genética , Cristalografia por Raios X , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Fosforilação , Domínios e Motivos de Interação entre Proteínas , Proteínas Proto-Oncogênicas c-vav/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Repressoras/metabolismo , Homologia de Sequência de Aminoácidos , Eletricidade Estática , Tirosina/química , Ubiquitina-Proteína Ligases/metabolismo , Domínios de Homologia de src
15.
J Biol Chem ; 290(20): 12595-602, 2015 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-25795782

RESUMO

In a case-control association study with 3716 North Americans of Hispanic descent and 4867 North Americans of European descent, we show that the associations of rs17849502 (NCF2 His-389 → Gln) and rs13306575 (NCF2 Arg-395 → Trp) with systemic lupus erythematosus are independent. We have shown that His-389 → Gln disrupts the binding of NCF2 to the ZF domain of VAV1, resulting in decreased NADPH oxidase activity. With respect to Arg-395 → Trp, using protein docking and structure analyses, we provide a model for the involvement of this mutation in the structure and function of the NADPH oxidase complex. This model assigns a central role to Arg-395 in the structure and stability of the quaternary NCF2/NCF4/VAV1/RAC1 NADPH oxidase complex. Arg-395 stabilizes the C-terminal tail of NCF4 and the conformation of NCF2 loop 395-402, which in turn stabilize the evolutionarily conserved interactions of NCF2/NCF4 with the DH domain of VAV1 and RAC1 region 120-137. Our findings are consistent with the high levels of conservation of all of the residues involved in these interactions.


Assuntos
Lúpus Eritematoso Sistêmico , Simulação de Acoplamento Molecular , Mutação de Sentido Incorreto , NADPH Oxidases/química , Substituição de Aminoácidos , Estabilidade Enzimática/genética , Feminino , Hispânico ou Latino , Humanos , Masculino , NADPH Oxidases/genética , NADPH Oxidases/metabolismo , Fosfoproteínas/química , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-vav/química , Proteínas Proto-Oncogênicas c-vav/genética , Proteínas Proto-Oncogênicas c-vav/metabolismo , População Branca , Proteínas rac1 de Ligação ao GTP/química , Proteínas rac1 de Ligação ao GTP/genética , Proteínas rac1 de Ligação ao GTP/metabolismo
16.
Small GTPases ; 5(2): 9, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25483299

RESUMO

The Vav family is a group of tyrosine phosphorylation-regulated signal transduction molecules hierarchically located downstream of protein tyrosine kinases. The main function of these proteins is to work as guanosine nucleotide exchange factors (GEFs) for members of the Rho GTPase family. In addition, they can exhibit a variety of catalysis-independent roles in specific signaling contexts. Vav proteins play essential signaling roles for both the development and/or effector functions of a large variety of cell lineages, including those belonging to the immune, nervous, and cardiovascular systems. They also contribute to pathological states such as cancer, immune-related dysfunctions, and atherosclerosis. Here, I will provide an integrated view about the evolution, regulation, and effector properties of these signaling molecules. In addition, I will discuss the pros and cons for their potential consideration as therapeutic targets.


Assuntos
Proteínas Proto-Oncogênicas c-vav/metabolismo , Transdução de Sinais , Sequência de Aminoácidos , Animais , Humanos , Dados de Sequência Molecular , Proteínas Proto-Oncogênicas c-vav/química , Proteínas Proto-Oncogênicas c-vav/genética , Proteínas rho de Ligação ao GTP/metabolismo
17.
Sci Signal ; 7(321): ra35, 2014 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-24736456

RESUMO

Vav proteins are phosphorylation-dependent guanine nucleotide exchange factors (GEFs) that catalyze the activation of members of the Rho family of guanosine triphosphatases (GTPases). The current regulatory model holds that the nonphosphorylated, catalytically inactive state of these GEFs is maintained by intramolecular interactions among the amino-terminal domains and the central catalytic core, which block the binding of Vav proteins to GTPases. We showed that this autoinhibition is mechanistically more complex, also involving the bivalent association of the carboxyl-terminal Src homology 3 (SH3) region of Vav with its catalytic and pleckstrin homology (PH) domains. Such interactions occurred through proline-rich region-independent mechanisms. Full release from this double-locked state required synergistic weakening effects from multiple phosphorylated tyrosine residues, thus providing an optimized system to generate gradients of Vav GEF activity depending on upstream signaling inputs. This mechanism is shared by mammalian and Drosophila melanogaster Vav proteins, suggesting that it may be a common regulatory feature for this protein family.


Assuntos
Proteínas Proto-Oncogênicas c-vav/química , Domínios de Homologia de src , Animais , Células COS , Domínio Catalítico , Chlorocebus aethiops , Drosophila melanogaster/metabolismo , Escherichia coli/metabolismo , GTP Fosfo-Hidrolases/metabolismo , Humanos , Imageamento Tridimensional , Células Jurkat , Camundongos , Família Multigênica , Células NIH 3T3 , Fosforilação , Prolina/química , Proteínas Proto-Oncogênicas c-vav/metabolismo , Transdução de Sinais , Tirosina/química
18.
J Cell Sci ; 125(Pt 22): 5302-14, 2012 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-22956543

RESUMO

The antigen-specific binding of T cells to antigen presenting cells results in recruitment of signalling proteins to microclusters at the cell-cell interface known as the immunological synapse (IS). The Vav1 guanine nucleotide exchange factor plays a critical role in T cell antigen receptor (TCR) signalling, leading to the activation of multiple pathways. We now show that it is recruited to microclusters and to the IS in primary CD4(+) and CD8(+) T cells. Furthermore, we show that this recruitment depends on the SH2 and C-terminal SH3 (SH3(B)) domains of Vav1, and on phosphotyrosines 112 and 128 of the SLP76 adaptor protein. Biophysical measurements show that Vav1 binds directly to these residues on SLP76 and that efficient binding depends on the SH2 and SH3(B) domains of Vav1. Finally, we show that the same two domains are critical for the phosphorylation of Vav1 and its signalling function in TCR-induced calcium flux. We propose that Vav1 is recruited to the IS by binding to SLP76 and that this interaction is critical for the transduction of signals leading to calcium flux.


Assuntos
Proteínas Proto-Oncogênicas c-vav/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Transdução de Sinais/imunologia , Animais , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Cálcio/metabolismo , Células Cultivadas , Humanos , Sinapses Imunológicas/metabolismo , Camundongos , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Fosforilação , Transporte Proteico/imunologia , Proteínas Proto-Oncogênicas c-vav/química , Domínios de Homologia de src
19.
J Struct Biol ; 180(1): 84-95, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22750419

RESUMO

Vav2 is a ubiquitous guanine nucleotide exchange factor (GEF) for the small GTPase Rac1. It regulates processes including cell migration, neuronal development and phagocytosis through interactions with different proteins. In this study, Arap3, a dual GTPase-activating protein (GAP) for RhoA and Arf6, was first identified to be a novel interaction partner for Vav2 both in vitro and in vivo. ITC and NMR chemical shift perturbation experiments demonstrated that Vav2 SH2 domain was able to interact directly with phosphorylated Y1403 and Y1408 within the C-terminal region of Arap3 with high affinities, with the dissociation constants (Kd) of ≈ 0.27 and ≈ 1.40 µM, respectively. In addition, using different phosphotyrosine peptides, the pY +3 specificity of Vav2 SH2 domain was discovered. The solution structures of Vav2 SH2 domain in free and in complex with the phosphotyrosine peptide pY1408 were therefore determined to understand the structural basis of this recognition specificity. Structural analysis revealed that the presence of a Phe residue in the BG loop (BG6) leads to the formation of a shallow hydrophobic pY +3 pocket on the surface of Vav2 SH2 domain, which determines the pY +3 specificity of Vav2 SH2 domain.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Simulação de Dinâmica Molecular , Proteínas Proto-Oncogênicas c-vav/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/química , Sítios de Ligação , Proteínas Ativadoras de GTPase/química , Células HEK293 , Células HeLa , Humanos , Interações Hidrofóbicas e Hidrofílicas , Fragmentos de Peptídeos/química , Fosfoproteínas/química , Fosforilação , Domínios e Motivos de Interação entre Proteínas , Mapeamento de Interação de Proteínas , Processamento de Proteína Pós-Traducional , Estrutura Secundária de Proteína , Proteínas Proto-Oncogênicas c-vav/química , Propriedades de Superfície , Termodinâmica
20.
J Biol Chem ; 287(16): 13137-58, 2012 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-22351766

RESUMO

C1 domains, the recognition motif of the second messenger diacylglycerol and of the phorbol esters, are classified as typical (ligand-responsive) or atypical (not ligand-responsive). The C1 domain of Vav1, a guanine nucleotide exchange factor, plays a critical role in regulation of Vav activity through stabilization of the Dbl homology domain, which is responsible for exchange activity of Vav. Although the C1 domain of Vav1 is classified as atypical, it retains a binding pocket geometry homologous to that of the typical C1 domains of PKCs. This study clarifies the basis for its failure to bind ligands. Substituting Vav1-specific residues into the C1b domain of PKCδ, we identified five crucial residues (Glu(9), Glu(10), Thr(11), Thr(24), and Tyr(26)) along the rim of the binding cleft that weaken binding potency in a cumulative fashion. Reciprocally, replacing these incompatible residues in the Vav1 C1 domain with the corresponding residues from PKCδ C1b (δC1b) conferred high potency for phorbol ester binding. Computer modeling predicts that these unique residues in Vav1 increase the hydrophilicity of the rim of the binding pocket, impairing membrane association and thereby preventing formation of the ternary C1-ligand-membrane binding complex. The initial design of diacylglycerol-lactones to exploit these Vav1 unique residues showed enhanced selectivity for C1 domains incorporating these residues, suggesting a strategy for the development of ligands targeting Vav1.


Assuntos
Diglicerídeos/metabolismo , Ésteres de Forbol/metabolismo , Proteínas Proto-Oncogênicas c-vav/química , Proteínas Proto-Oncogênicas c-vav/metabolismo , Sequência de Aminoácidos , Linhagem Celular Tumoral , Humanos , Lactonas/metabolismo , Ligantes , Masculino , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Fosfolipídeos/metabolismo , Neoplasias da Próstata , Ligação Proteica/fisiologia , Proteína Quinase C-delta/metabolismo , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-vav/genética , Transdução de Sinais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA