Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Artigo em Inglês | MEDLINE | ID: mdl-30069819

RESUMO

Protein tyrosine kinases (PTK), discovered in the 1970s, have been considered master regulators of biological processes with high clinical significance as targets for human diseases. Their actions are countered by protein tyrosine phosphatases (PTP), enzymes yet underrepresented as drug targets because of the high homology of their catalytic domains and high charge of their catalytic pocket. This scenario is still worse for some PTP subclasses, for example, for the atypical dual-specificity phosphatases (ADUSPs), whose biological functions are not even completely known. In this sense, the present work focuses on the dual-specificity phosphatase 3 (DUSP3), also known as VH1-related phosphatase (VHR), an uncommon regulator of mitogen-activated protein kinase (MAPK) phosphorylation. DUSP3 expression and activities are suggestive of a tumor suppressor or tumor-promoting enzyme in different types of human cancers. Furthermore, DUSP3 has other biological functions involving immune response mediation, thrombosis, hemostasis, angiogenesis, and genomic stability that occur through either MAPK-dependent or MAPK-independent mechanisms. This broad spectrum of actions is likely due to the large substrate diversity and molecular mechanisms that are still under scrutiny. The growing advances in characterizing new DUSP3 substrates will allow the development of pharmacological inhibitors relevant for possible future clinical trials. This review covers all aspects of DUSP3, since its gene cloning and crystallographic structure resolution, in addition to its classical and novel substrates and the biological processes involved, followed by an update of what is currently known about the DUSP3/VHR-inhibiting compounds that might be considered potential drugs to treat human diseases.


Assuntos
Fosfatase 3 de Especificidade Dupla/genética , Fosfatase 3 de Especificidade Dupla/fisiologia , Fosfatase 3 de Especificidade Dupla/antagonistas & inibidores , Humanos , Proteínas Quinases Ativadas por Mitógeno , Neoplasias/enzimologia , Neovascularização Patológica , Fosforilação , Proteínas Tirosina Fosfatases , Proteínas Tirosina Quinases
2.
Clinics (Sao Paulo) ; 73(suppl 1): e466s, 2018 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-30208163

RESUMO

Protein tyrosine phosphatases have long been considered key regulators of biological processes and are therefore implicated in the origins of various human diseases. Heterozygosity, mutations, deletions, and the complete loss of some of these enzymes have been reported to cause neurodegenerative diseases, autoimmune syndromes, genetic disorders, metabolic diseases, cancers, and many other physiological imbalances. Vaccinia H1-related phosphatase, also known as dual-specificity phosphatase 3, is a protein tyrosine phosphatase enzyme that regulates the phosphorylation of the mitogen-activated protein kinase signaling pathway, a central mediator of a diversity of biological responses. It has been suggested that vaccinia H1-related phosphatase can act as a tumor suppressor or tumor-promoting phosphatase in different cancers. Furthermore, emerging evidence suggests that this enzyme has many other biological functions, such as roles in immune responses, thrombosis, hemostasis, angiogenesis, and genomic stability, and this broad spectrum of vaccinia H1-related phosphatase activity is likely the result of its diversity of substrates. Hence, fully identifying and characterizing these substrate-phosphatase interactions will facilitate the identification of pharmacological inhibitors of vaccinia H1-related phosphatase that can be evaluated in clinical trials. In this review, we describe the biological processes mediated by vaccinia H1-related phosphatase, especially those related to genomic stability. We also focus on validated substrates and signaling circuitry with clinical relevance in human diseases, particularly oncogenesis.


Assuntos
Fosfatase 3 de Especificidade Dupla/fisiologia , Neoplasias/enzimologia , Humanos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Neoplasias/mortalidade , Transdução de Sinais , Análise de Sobrevida
3.
Clinics ; 73(supl.1): e466s, 2018. graf
Artigo em Inglês | LILACS | ID: biblio-952823

RESUMO

Protein tyrosine phosphatases have long been considered key regulators of biological processes and are therefore implicated in the origins of various human diseases. Heterozygosity, mutations, deletions, and the complete loss of some of these enzymes have been reported to cause neurodegenerative diseases, autoimmune syndromes, genetic disorders, metabolic diseases, cancers, and many other physiological imbalances. Vaccinia H1-related phosphatase, also known as dual-specificity phosphatase 3, is a protein tyrosine phosphatase enzyme that regulates the phosphorylation of the mitogen-activated protein kinase signaling pathway, a central mediator of a diversity of biological responses. It has been suggested that vaccinia H1-related phosphatase can act as a tumor suppressor or tumor-promoting phosphatase in different cancers. Furthermore, emerging evidence suggests that this enzyme has many other biological functions, such as roles in immune responses, thrombosis, hemostasis, angiogenesis, and genomic stability, and this broad spectrum of vaccinia H1-related phosphatase activity is likely the result of its diversity of substrates. Hence, fully identifying and characterizing these substrate-phosphatase interactions will facilitate the identification of pharmacological inhibitors of vaccinia H1-related phosphatase that can be evaluated in clinical trials. In this review, we describe the biological processes mediated by vaccinia H1-related phosphatase, especially those related to genomic stability. We also focus on validated substrates and signaling circuitry with clinical relevance in human diseases, particularly oncogenesis.


Assuntos
Humanos , Fosfatase 3 de Especificidade Dupla/fisiologia , Neoplasias/enzimologia , Transdução de Sinais , Análise de Sobrevida , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Neoplasias/mortalidade
4.
Oncogene ; 36(47): 6509-6517, 2017 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-28759036

RESUMO

Vaccinia H1-related phosphatase (VHR/DUSP3) is a member of the dual-specificity phosphatase family. Deregulation of VHR is observed in various malignant diseases. We identified focal adhesion kinase (FAK) as a VHR-interacting molecule. Over-expression of VHR decreased tyrosine phosphorylation of FAK and decreasing VHR promoted FAK tyrosine phosphorylation. In vitro assays proved that recombinant VHR directly dephosphorylated FAK and paxillin. VHR-knockout mice did not have obvious abnormality; however, VHR-knockout cells showed decreased expression of integrins and FAK but stronger FAK and paxillin phosphorylation upon attachment to fibronectin. Additionally, VHR-knockout fibroblast and lung epithelial cells had elevated ligand-induced epidermal growth factor receptor (EGFR) phosphorylation. Inducible expression of VHR suppressed directional cell migration, and VHR deficiency resulted in a higher cell migratory ability. VHR-knockout cells have stronger FAK phosphorylation in cell adhesions, long-lasting trailing ends and slower turnover of focal adhesions. These collective data indicate that VHR is a FAK phosphatase and participates in regulating the formation and disassembly of focal adhesions.


Assuntos
Adesão Celular , Movimento Celular , Fosfatase 3 de Especificidade Dupla/fisiologia , Quinase 1 de Adesão Focal/metabolismo , Animais , Linhagem Celular Tumoral , Receptores ErbB/metabolismo , Adesões Focais/metabolismo , Técnicas de Inativação de Genes , Humanos , Integrinas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Paxilina/metabolismo , Fosforilação/fisiologia , Tirosina/metabolismo
5.
Biochim Biophys Acta Gen Subj ; 1861(7): 1879-1894, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28389334

RESUMO

BACKGROUND: Radiotherapy causes the regression of many human tumors by increasing DNA damage, and the novel molecular mechanisms underlying the genomic instability leading to cancer progression and metastasis must be elucidated. Atypical dual-specificity phosphatase 3 (DUSP3) has been shown to down-regulate mitogen-activated protein kinases (MAPKs) to control the proliferation and apoptosis of human cancer cells. We have recently identified novel molecular targets of DUSP3 that function in DNA damage response and repair; however, whether DUSP3 affects these processes remains unknown. METHODS: Tumor cell lines in which DUSP3 activity was suppressed by pharmacological inhibitors or a targeted siRNA were exposed to gamma radiation, and proliferation, survival, DNA strand breaks and recombination repair pathways were sequentially analyzed. RESULTS: The combination of reduced DUSP3 activity and gamma irradiation resulted in decreased cellular proliferation and survival and increased cellular senescence compared with the effects of radiation exposure alone. Gamma radiation-induced DNA damage was increased by the loss of DUSP3 activity and correlated with increased levels of phospho-H2AX protein and numbers of ionizing radiation-induced γ-H2AX foci, which were reflected in diminished efficiencies of homologous recombination (HR) and non-homologous end-joining (NHEJ) repair. Similar results were obtained in ATM-deficient cells, in which reduced DUSP3 activity increased radiosensitivity, independent of increased MAPK phosphorylation. CONCLUSION: The loss of DUSP3 activity markedly increases gamma radiation-induced DNA strand breaks, suggesting a potential novel role for DUSP3 in DNA repair. GENERAL SIGNIFICANCE: The radioresistance of tumor cells is effectively reduced by a combination of approaches through the inhibition of DUSPs.


Assuntos
Reparo do DNA , Fosfatase 3 de Especificidade Dupla/fisiologia , Neoplasias/radioterapia , Tolerância a Radiação , Proteínas Mutadas de Ataxia Telangiectasia/fisiologia , Linhagem Celular Tumoral , Dano ao DNA , Fosfatase 3 de Especificidade Dupla/antagonistas & inibidores , Raios gama , Histonas/análise , Humanos , Proteínas Quinases Ativadas por Mitógeno/metabolismo
6.
J Clin Invest ; 123(12): 5231-46, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24200691

RESUMO

Epigenetic dysregulation has emerged as a major contributor to tumorigenesis. Histone methylation is a well-established mechanism of epigenetic regulation that is dynamically modulated by histone methyltransferases and demethylases. The pathogenic role of histone methylation modifiers in non-small cell lung cancer (NSCLC), which is the leading cause of cancer deaths worldwide, remains largely unknown. Here, we found that the histone H3 lysine 36 (H3K36) demethylase KDM2A (also called FBXL11 and JHDM1A) is frequently overexpressed in NSCLC tumors and cell lines. KDM2A and its catalytic activity were required for in vitro proliferation and invasion of KDM2A-overexpressing NSCLC cells. KDM2A overexpression in NSCLC cells with low KDM2A levels increased cell proliferation and invasiveness. KDM2A knockdown abrogated tumor growth and invasive abilities of NSCLC cells in mouse xenograft models. We identified dual-specificity phosphatase 3 (DUSP3) as a key KDM2A target gene and found that DUSP3 dephosphorylates ERK1/2 in NSCLC cells. KDM2A activated ERK1/2 through epigenetic repression of DUSP3 expression via demethylation of dimethylated H3K36 at the DUSP3 locus. High KDM2A levels correlated with poor prognosis in NSCLC patients. These findings uncover an unexpected role for a histone methylation modifier in activating ERK1/2 in lung tumorigenesis and metastasis, suggesting that KDM2A may be a promising therapeutic target in NSCLC.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/patologia , Epigênese Genética/fisiologia , Proteínas F-Box/fisiologia , Regulação Neoplásica da Expressão Gênica/fisiologia , Histonas/metabolismo , Histona Desmetilases com o Domínio Jumonji/fisiologia , Neoplasias Pulmonares/patologia , Sistema de Sinalização das MAP Quinases , Proteínas de Neoplasias/fisiologia , Processamento de Proteína Pós-Traducional/fisiologia , Animais , Carcinoma Pulmonar de Células não Pequenas/genética , Divisão Celular , Linhagem Celular Tumoral , Fosfatase 3 de Especificidade Dupla/biossíntese , Fosfatase 3 de Especificidade Dupla/genética , Fosfatase 3 de Especificidade Dupla/fisiologia , Epigênese Genética/genética , Proteínas F-Box/antagonistas & inibidores , Proteínas F-Box/biossíntese , Proteínas F-Box/genética , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Xenoenxertos , Humanos , Histona Desmetilases com o Domínio Jumonji/antagonistas & inibidores , Histona Desmetilases com o Domínio Jumonji/biossíntese , Histona Desmetilases com o Domínio Jumonji/genética , Neoplasias Pulmonares/genética , Masculino , Metilação , Camundongos , Camundongos Nus , Invasividade Neoplásica , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Prognóstico , Regiões Promotoras Genéticas , Processamento de Proteína Pós-Traducional/genética , Interferência de RNA , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , RNA Neoplásico/biossíntese , RNA Neoplásico/genética
7.
J Biol Chem ; 284(15): 10129-37, 2009 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-19211553

RESUMO

The Vaccinia virus H1 gene product, VH1, is a dual specificity phosphatase that down-regulates the cellular antiviral response by dephosphorylating STAT1. The crystal structure of VH1, determined at 1.32 A resolution, reveals a novel dimeric quaternary structure, which exposes two active sites spaced approximately 39 A away from each other. VH1 forms a stable dimer via an extensive domain swap of the N-terminal helix (residues 1-20). In vitro, VH1 can dephosphorylate activated STAT1, in a reaction that is competed by the nuclear transport adapter importin alpha5. Interestingly, VH1 is inactive with respect to STAT1 bound to DNA, suggesting that the viral phosphatase acts predominantly on the cytoplasmic pool of activated STAT1. We propose that the dimeric quaternary structure of VH1 is essential for specific recognition of activated STAT1, which prevents its nuclear translocation, thus blocking interferon-gamma signal transduction and antiviral response.


Assuntos
Fosfatase 3 de Especificidade Dupla/química , Fosfatase 3 de Especificidade Dupla/fisiologia , Fator de Transcrição STAT1/metabolismo , Vaccinia virus/enzimologia , Transporte Ativo do Núcleo Celular , Domínio Catalítico , Dicroísmo Circular , DNA/química , Dimerização , Humanos , Interferon gama/metabolismo , Modelos Moleculares , Conformação Proteica , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Transdução de Sinais
8.
Cancer Res ; 68(22): 9255-64, 2008 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-19010898

RESUMO

Androgen ablation during the initial stages of prostate cancer causes regression of the tumor due to an increase in apoptosis and reduced cellular proliferation. However, prostate cancer invariably progresses to an androgen-independent state for poorly understood reasons. Previous studies showed that c-Jun NH(2) terminal kinase (JNK) is required for 12-O-tetradecanoylphorbol-13-acetate (TPA)- and thapsigargin (TG)-induced apoptosis in the androgen-responsive prostate cancer cell line LNCaP. Androgens protect LNCaP cells from TPA-induced or TG-induced apoptosis via down-regulation of JNK activation. However, the molecular mechanisms of this inhibition are not clear. Here, we systematically investigated the possible regulation of mitogen-activated protein kinase phosphatases/dual-specificity phosphatases during apoptosis of LNCaP cells and found that Vaccinia H1-related protein (VHR/DUSP3) is up-regulated by androgens during inhibition of apoptosis in LNCaP cells, but not in androgen-independent DU145 cells. Ectopic expression of wild-type VHR, but not a catalytically inactive mutant, interfered with TPA- and TG-induced apoptosis. Consistently, small interfering RNA-mediated knockdown of endogenous VHR increased apoptosis in response to TPA or TG in the presence of androgens. Furthermore, COS7 cells stably expressing wild-type VHR, but not a mutant, had a decrease in JNK phosphorylation. In vivo, VHR expression decreased in the androgen-dependent human prostate cancer xenograft CWR22 upon androgen withdrawal and was inversely correlated to JNK phosphorylation. Expression analysis in human prostate cancer specimens showed that VHR is increased in prostate cancer compared with normal prostate. These data show that VHR has a direct role in the inhibition of JNK-dependent apoptosis in LNCaP cells and may therefore have a role in prostate cancer progression.


Assuntos
Apoptose , Fosfatase 3 de Especificidade Dupla/fisiologia , Neoplasias da Próstata/patologia , Androgênios/fisiologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Fosfatase 1 de Especificidade Dupla/fisiologia , Fosfatase 3 de Especificidade Dupla/genética , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Sistema de Sinalização das MAP Quinases , Masculino , Metribolona/farmacologia , Fosforilação , RNA Mensageiro/análise , Acetato de Tetradecanoilforbol/farmacologia , Tapsigargina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA