Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Neurosci ; 41(35): 7340-7349, 2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-34290083

RESUMO

Alzheimer's disease (AD) is a progressive neurodegenerative disease marked by the accumulation of amyloid-ß (Aß) plaques and neurofibrillary tangles. Aß oligomers cause synaptic dysfunction early in AD by enhancing long-term depression (LTD; a paradigm for forgetfulness) via metabotropic glutamate receptor (mGluR)-dependent regulation of striatal-enriched tyrosine phosphatase (STEP61). Reelin is a neuromodulator that signals through ApoE (apolipoprotein E) receptors to protect the synapse against Aß toxicity (Durakoglugil et al., 2009) Reelin signaling is impaired by ApoE4, the most important genetic risk factor for AD, and Aß-oligomers activate metabotropic glutamate receptors (Renner et al., 2010). We therefore asked whether Reelin might also affect mGluR-LTD. To this end, we induced chemical mGluR-LTD using DHPG (Dihydroxyphenylglycine), a selective mGluR5 agonist. We found that exogenous Reelin reduces the DHPG-induced increase in STEP61, prevents the dephosphorylation of GluA2, and concomitantly blocks mGluR-mediated LTD. By contrast, Reelin deficiency increased expression of Ca2+-permeable GluA2-lacking AMPA receptors along with higher STEP61 levels, resulting in occlusion of DHPG-induced LTD in hippocampal CA1 neurons. We propose a model in which Reelin modulates local protein synthesis as well as AMPA receptor subunit composition through modulation of mGluR-mediated signaling with implications for memory consolidation or neurodegeneration.SIGNIFICANCE STATEMENT Reelin is an important neuromodulator, which in the adult brain controls synaptic plasticity and protects against neurodegeneration. Amyloid-ß has been shown to use mGluRs to induce synaptic depression through endocytosis of NMDA and AMPA receptors, a mechanism referred to as LTD, a paradigm of forgetfulness. Our results show that Reelin regulates the phosphatase STEP, which plays an important role in neurodegeneration, as well as the expression of calcium-permeable AMPA receptors, which play a role in memory formation. These data suggest that Reelin uses mGluR LTD pathways to regulate memory formation as well as neurodegeneration.


Assuntos
Depressão Sináptica de Longo Prazo/fisiologia , Neurônios/fisiologia , Proteínas Tirosina Fosfatases não Receptoras/fisiologia , Receptores de Glutamato Metabotrópico/fisiologia , Proteína Reelina/fisiologia , 2-Amino-5-fosfonovalerato/farmacologia , Animais , Região CA1 Hipocampal/citologia , Região CA1 Hipocampal/efeitos dos fármacos , Cálcio/fisiologia , Células Cultivadas , Córtex Cerebral/citologia , Indução Enzimática/efeitos dos fármacos , Depressão Sináptica de Longo Prazo/efeitos dos fármacos , Memória/fisiologia , Metoxi-Hidroxifenilglicol/análogos & derivados , Metoxi-Hidroxifenilglicol/farmacologia , Camundongos , Degeneração Neural/fisiopatologia , Neurônios/efeitos dos fármacos , Técnicas de Patch-Clamp , Fosforilação/efeitos dos fármacos , Picrotoxina/farmacologia , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptores de AMPA/metabolismo , Receptores de Glutamato Metabotrópico/agonistas , Proteínas Recombinantes/metabolismo , Proteína Reelina/deficiência , Proteína Reelina/genética
2.
Behav Brain Res ; 391: 112713, 2020 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-32461127

RESUMO

Autism spectrum disorders (ASDs) are highly prevalent childhood illnesses characterized by impairments in communication, social behavior, and repetitive behaviors. Studies have found aberrant synaptic plasticity and neuronal connectivity during the early stages of brain development and have suggested that these contribute to an increased risk for ASD. STEP is a protein tyrosine phosphatase that regulates synaptic plasticity and is implicated in several cognitive disorders. Here we test the hypothesis that STEP may contribute to some of the aberrant behaviors present in the VPA-induced mouse model of ASD. In utero VPA exposure of pregnant dams results in autistic-like behavior in the pups, which is associated with a significant increase in the STEP expression in the prefrontal cortex. The elevated STEP protein levels are correlated with increased dephosphorylation of STEP substrates GluN2B, Pyk2 and ERK, suggesting upregulated STEP activity. Moreover, pharmacological inhibition of STEP rescues the sociability, repetitive and abnormal anxiety phenotypes commonly associated with ASD. These data suggest that STEP may play a role in the VPA model of ASD and STEP inhibition may have a potential therapeutic benefit in this model.


Assuntos
Transtorno do Espectro Autista/fisiopatologia , Proteínas Tirosina Fosfatases não Receptoras/metabolismo , Animais , Transtorno do Espectro Autista/tratamento farmacológico , Transtorno Autístico/tratamento farmacológico , Comportamento Animal , Modelos Animais de Doenças , Comportamento Exploratório/fisiologia , Feminino , Inibição Psicológica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Plasticidade Neuronal/fisiologia , Córtex Pré-Frontal , Gravidez , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Proteínas Tirosina Fosfatases/metabolismo , Proteínas Tirosina Fosfatases/fisiologia , Proteínas Tirosina Fosfatases não Receptoras/fisiologia , Comportamento Social , Comportamento Estereotipado/fisiologia , Ácido Valproico/efeitos adversos
3.
Cell Signal ; 69: 109554, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32027948

RESUMO

The formation of erythroid progenitor cells depends sharply upon erythropoietin (EPO), its cell surface receptor (erythropoietin receptor, EPOR), and Janus kinase 2 (JAK2). Clinically, recombinant human EPO (rhEPO) additionally is an important anti-anemia agent for chronic kidney disease (CKD), myelodysplastic syndrome (MDS) and chemotherapy, but induces hypertension, and can exert certain pro-tumorigenic effects. Cellular signals transduced by EPOR/JAK2 complexes, and the nature of EPO-modulated signal transduction factors, therefore are of significant interest. By employing phospho-tyrosine post-translational modification (p-Y PTM) proteomics and human EPO- dependent UT7epo cells, we have identified 22 novel kinases and phosphatases as novel EPO targets, together with their specific sites of p-Y modification. New kinases modified due to EPO include membrane palmitoylated protein 1 (MPP1) and guanylate kinase 1 (GUK1) guanylate kinases, together with the cytoskeleton remodeling kinases, pseudopodium enriched atypical kinase 1 (PEAK1) and AP2 associated kinase 1 (AAK1). Novel EPO- modified phosphatases include protein tyrosine phosphatase receptor type A (PTPRA), phosphohistidine phosphatase 1 (PHPT1), tensin 2 (TENC1), ubiquitin associated and SH3 domain containing B (UBASH3B) and protein tyrosine phosphatase non-receptor type 18 (PTPN18). Based on PTPN18's high expression in hematopoietic progenitors, its novel connection to JAK kinase signaling, and a unique EPO- regulated PTPN18-pY389 motif which is modulated by JAK2 inhibitors, PTPN18's actions in UT7epo cells were investigated. Upon ectopic expression, wt-PTPN18 promoted EPO dose-dependent cell proliferation, and survival. Mechanistically, PTPN18 sustained the EPO- induced activation of not only mitogen-activated protein kinases 1 and 3 (ERK1/2), AKT serine/threonine kinase 1-3 (AKT), and signal transducer and activator of transcription 5A and 5B (STAT5), but also JAK2. Each effect further proved to depend upon PTPN18's EPO- modulated (p)Y389 site. In analyses of the EPOR and the associated adaptor protein RHEX (regulator of hemoglobinization and erythroid cell expansion), wt-PTPN18 increased high molecular weight EPOR forms, while sharply inhibiting the EPO-induced phosphorylation of RHEX-pY141. Each effect likewise depended upon PTPN18-Y389. PTPN18 thus promotes signals for EPO-dependent hematopoietic cell growth, and may represent a new druggable target for myeloproliferative neoplasms.


Assuntos
Eritropoese , Eritropoetina/metabolismo , Janus Quinase 2/metabolismo , Fragmentos de Peptídeos/metabolismo , Proteínas Tirosina Fosfatases não Receptoras/fisiologia , Receptores da Eritropoetina/metabolismo , Linhagem Celular , Humanos , Proteômica , Transdução de Sinais
4.
Ann Rheum Dis ; 78(5): 600-609, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30808624

RESUMO

OBJECTIVE: We aimed to understand the role of the tyrosine phosphatase PTPN14-which in cancer cells modulates the Hippo pathway by retaining YAP in the cytosol-in fibroblast-like synoviocytes (FLS) from patients with rheumatoid arthritis (RA). METHODS: Gene/protein expression levels were measured by quantitative PCR and/or Western blotting. Gene knockdown in RA FLS was achieved using antisense oligonucleotides. The interaction between PTPN14 and YAP was assessed by immunoprecipitation. The cellular localisation of YAP and SMAD3 was examined via immunofluorescence. SMAD reporter studies were carried out in HEK293T cells. The RA FLS/cartilage coimplantation and passive K/BxN models were used to examine the role of YAP in arthritis. RESULTS: RA FLS displayed overexpression of PTPN14 when compared with FLS from patients with osteoarthritis (OA). PTPN14 knockdown in RA FLS impaired TGFß-dependent expression of MMP13 and potentiation of TNF signalling. In RA FLS, PTPN14 formed a complex with YAP. Expression of PTPN14 or nuclear YAP-but not of a non-YAP-interacting PTPN14 mutant-enhanced SMAD reporter activity. YAP promoted TGFß-dependent SMAD3 nuclear localisation in RA FLS. Differences in epigenetic marks within Hippo pathway genes, including YAP, were found between RA FLS and OA FLS. Inhibition of YAP reduced RA FLS pathogenic behaviour and ameliorated arthritis severity. CONCLUSION: In RA FLS, PTPN14 and YAP promote nuclear localisation of SMAD3. YAP enhances a range of RA FLS pathogenic behaviours which, together with epigenetic evidence, points to the Hippo pathway as an important regulator of RA FLS behaviour.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Proteínas Tirosina Fosfatases não Receptoras/fisiologia , Transdução de Sinais/fisiologia , Sinoviócitos/metabolismo , Fatores de Transcrição/fisiologia , Fator de Crescimento Transformador beta/fisiologia , Animais , Artrite Reumatoide/metabolismo , Proteínas de Ciclo Celular/fisiologia , Humanos , Camundongos , Proteínas de Sinalização YAP
5.
BMB Rep ; 50(11): 584-589, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29065968

RESUMO

Intercellular adhesion molecule-1 (ICAM-1), which is induced by tumor necrosis factor (TNF)-α, contributes to the entry of immune cells into the site of inflammation in the skin. Here, we show that protein tyrosine phosphatase non-receptor type 21 (PTPN21) negatively regulates ICAM-1 expression in human keratinocytes. PTPN21 expression was transiently induced after stimulation with TNF-α. When overexpressed, PTPN21 inhibited the expression of ICAM-1 in HaCaT cells but PTPN21 C1108S, a phosphatase activity-inactive mutant, failed to inhibit ICAM-1 expression. Nuclear factor-κB (NF-κB), a key transcription factor of ICAM-1 gene expression, was inhibited by PTPN21, but not by PTPN21 C1108S. PTPN21 directly dephosphorylated phospho-inhibitor of κB (IκB)-kinase ß (IKKß) at Ser177/181. This dephosphorylation led to the stabilization of IκBα and inhibition of NF-κB activity. Taken together, our results suggest that PTPN21 could be a valuable molecular target for regulation of inflammation in the skin by dephosphorylating p-IKKß and inhibiting NF-κB signaling. [BMB Reports 2017; 50(11): 584-589].


Assuntos
Molécula 1 de Adesão Intercelular/metabolismo , Proteínas Tirosina Fosfatases não Receptoras/metabolismo , Proteínas Tirosina Fosfatases não Receptoras/fisiologia , Adesão Celular/efeitos dos fármacos , Linhagem Celular , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Quinase I-kappa B/metabolismo , Proteínas I-kappa B/metabolismo , Queratinócitos/metabolismo , Queratinócitos/patologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Inibidor de NF-kappaB alfa/metabolismo , NF-kappa B/metabolismo , Fosforilação , Proteínas Tirosina Fosfatases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo
6.
Cancer Cell ; 32(4): 460-473.e6, 2017 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-29017057

RESUMO

The p53 transcription factor is a critical barrier to pancreatic cancer progression. To unravel mechanisms of p53-mediated tumor suppression, which have remained elusive, we analyzed pancreatic cancer development in mice expressing p53 transcriptional activation domain (TAD) mutants. Surprisingly, the p5353,54 TAD2 mutant behaves as a "super-tumor suppressor," with an enhanced capacity to both suppress pancreatic cancer and transactivate select p53 target genes, including Ptpn14. Ptpn14 encodes a negative regulator of the Yap oncoprotein and is necessary and sufficient for pancreatic cancer suppression, like p53. We show that p53 deficiency promotes Yap signaling and that PTPN14 and TP53 mutations are mutually exclusive in human cancers. These studies uncover a p53-Ptpn14-Yap pathway that is integral to p53-mediated tumor suppression.


Assuntos
Proteínas Nucleares/fisiologia , Neoplasias Pancreáticas/genética , Proteínas Tirosina Fosfatases não Receptoras/fisiologia , Fatores de Transcrição/fisiologia , Proteína Supressora de Tumor p53/fisiologia , Animais , Proteínas de Ciclo Celular , Proliferação de Células , Transformação Celular Neoplásica , Perfilação da Expressão Gênica , Humanos , Camundongos , Mutação , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/prevenção & controle , Transdução de Sinais
7.
Neurobiol Aging ; 41: 25-38, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-27103516

RESUMO

The STriatal-Enriched tyrosine Phosphatase (STEP) is involved in the etiology of several age-associated neurologic disorders linked to oxidative stress and is also known to play a role in neuroprotection by modulating glutamatergic transmission. However, the possible effect of aging on STEP level and activity in the brain is still unclear. In this study, using young (1 month), adult (4 months), and aged (18 months) rats, we show that aging is associated with increase in dimerization and loss of activity of STEP. Increased dimerization of STEP is primarily observed in the cortex and hippocampus and is associated with depletion of both reduced and total glutathione levels, suggesting an increase in oxidative stress. Consistent with this interpretation, studies in cell culture models of glutathione depletion and oxidative stress also demonstrate formation of dimers and higher order oligomers of STEP that involve intermolecular disulfide bond formation between multiple cysteine residues. Conversely, administration of N-acetyl cysteine, a major antioxidant that enhances glutathione biosynthesis, attenuates STEP dimerization both in the cortex and hippocampus. The findings indicate that loss of this intrinsic protective response pathway with age-dependent increase in oxidative stress may be a contributing factor for the susceptibility of the brain to age-associated neurologic disorders.


Assuntos
Envelhecimento/genética , Envelhecimento/metabolismo , Doenças do Sistema Nervoso/etiologia , Multimerização Proteica , Proteínas Tirosina Fosfatases não Receptoras/fisiologia , Acetilcisteína/farmacologia , Animais , Antioxidantes/farmacologia , Encéfalo/enzimologia , Encéfalo/metabolismo , Células Cultivadas , Ativação Enzimática , Glutamatos/fisiologia , Glutationa/metabolismo , Neuroproteção , Estresse Oxidativo/fisiologia , Proteínas Tirosina Fosfatases não Receptoras/metabolismo , Ratos Sprague-Dawley , Transmissão Sináptica/genética
8.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 31(9): 1251-4, 2015 Sep.
Artigo em Chinês | MEDLINE | ID: mdl-26359106

RESUMO

OBJECTIVE: To investigate the expression level of protein tyrosine phosphatase non-receptor type 14 (PTPN14), and analyze the relationship between PTPN14 and clinical pathological features and prognosis of patients with cholangiocarcinoma. METHODS: Expression of PTPN14 protein was detected by immunohistochemistry (IHC) in 57 cholangiocarcinoma tissues and corresponding adjacent normal tissues. The relationship between PTPN14 protein level and the clinical-pathological features of cholangiocarcinoma was analyzed using IBM SPSS 20.0 statistical software. The relationship between PTPN14 protein expression and 5-year overall survival of cholangiocarcinoma patients was investigated by survival curves. RESULTS: IHC revealed that positive rates of PTPN14 protein were 49.1% and 75.4% in cholangiocarcinoma tissues and adjacent tissues, respectively. The expression of PTPN14 protein was significantly associated with TNM I, II, and differentiation degree of cholangiocarcinoma patients, but not significantly associated with age and gender of cholangiocarcinoma patients. The 5-year overall survival rate was higher in the PTPN14-positive patients than the PTPN14-negative ones. CONCLUSION: PTPN14 was down-regulated in cholangiocarcinoma, and negatively correlated with better clinical-pathological features and 5-year overall survival rate of cholangiocarcinoma.


Assuntos
Neoplasias dos Ductos Biliares/patologia , Colangiocarcinoma/patologia , Proteínas Tirosina Fosfatases não Receptoras/fisiologia , Adulto , Idoso , Neoplasias dos Ductos Biliares/mortalidade , Colangiocarcinoma/mortalidade , Feminino , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Proteínas Tirosina Fosfatases não Receptoras/análise , Taxa de Sobrevida
9.
Mol Cell ; 57(2): 261-72, 2015 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-25544560

RESUMO

Glycogen is the major mammalian glucose storage cache and is critical for energy homeostasis. Glycogen synthesis in neurons must be tightly controlled due to neuronal sensitivity to perturbations in glycogen metabolism. Lafora disease (LD) is a fatal, congenital, neurodegenerative epilepsy. Mutations in the gene encoding the glycogen phosphatase laforin result in hyperphosphorylated glycogen that forms water-insoluble inclusions called Lafora bodies (LBs). LBs induce neuronal apoptosis and are the causative agent of LD. The mechanism of glycogen dephosphorylation by laforin and dysfunction in LD is unknown. We report the crystal structure of laforin bound to phosphoglucan product, revealing its unique integrated tertiary and quaternary structure. Structure-guided mutagenesis combined with biophysical and biochemical analyses reveal the basis for normal function of laforin in glycogen metabolism. Analyses of LD patient mutations define the mechanism by which subsets of mutations disrupt laforin function. These data provide fundamental insights connecting glycogen metabolism to neurodegenerative disease.


Assuntos
Glicogênio/metabolismo , Doença de Lafora/metabolismo , Proteínas Tirosina Fosfatases não Receptoras/química , Domínio Catalítico , Cristalografia por Raios X , Humanos , Modelos Moleculares , Oligossacarídeos/química , Fosfatos/química , Fosforilação , Ligação Proteica , Multimerização Proteica , Estrutura Secundária de Proteína , Proteínas Tirosina Fosfatases não Receptoras/fisiologia
10.
Br J Cancer ; 111(11): 2091-102, 2014 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-25349971

RESUMO

BACKGROUND: Gastrointestinal stromal tumour (GIST) is mainly initialised by receptor tyrosine kinase gene mutations. Although the tyrosine kinase inhibitor imatinib mesylate considerably improved the outcome of patients, imatinib resistance still remains a major therapeutic challenge in GIST therapy. Herein we evaluated the clinical impact of microRNAs in imatinib-treated GISTs. METHODS: The expression levels of microRNAs were quantified using microarray and RT-qPCR in GIST specimens from patients treated with neoadjuvant imatinib. The functional roles of miR-125a-5p and PTPN18 were evaluated in GIST cells. PTPN18 expression was quantified by western blotting in GIST samples. RESULTS: We showed that overexpression levels of miR-125a-5p and miR-107 were associated with imatinib resistance in GIST specimens. Functionally, miR-125a-5p expression modulated imatinib sensitivity in GIST882 cells with a homozygous KIT mutation but not in GIST48 cells with double KIT mutations. Overexpression of miR-125a-5p suppressed PTPN18 expression, and silencing of PTPN18 expression increased cell viability in GIST882 cells upon imatinib treatment. PTPN18 protein levels were significantly lower in the imatinib-resistant GISTs and inversely correlated with miR-125a-5p. Furthermore, several microRNAs were significantly associated with metastasis, KIT mutational status and survival. CONCLUSIONS: Our findings highlight a novel functional role of miR-125a-5p on imatinib response through PTPN18 regulation in GIST.


Assuntos
Antineoplásicos/uso terapêutico , Benzamidas/uso terapêutico , Neoplasias Gastrointestinais/tratamento farmacológico , Tumores do Estroma Gastrointestinal/tratamento farmacológico , MicroRNAs/fisiologia , Piperazinas/uso terapêutico , Pirimidinas/uso terapêutico , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Neoplasias Gastrointestinais/genética , Neoplasias Gastrointestinais/mortalidade , Tumores do Estroma Gastrointestinal/genética , Tumores do Estroma Gastrointestinal/mortalidade , Humanos , Mesilato de Imatinib , Mutação , Proteínas Tirosina Fosfatases não Receptoras/genética , Proteínas Tirosina Fosfatases não Receptoras/fisiologia , Proteínas Proto-Oncogênicas c-kit/genética , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética
11.
J Cell Sci ; 127(Pt 12): 2761-70, 2014 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-24727614

RESUMO

Protein tyrosine phosphatases (PTPs) are involved in hematopoiesis, but the function of many PTPs is not well characterized in vivo. Here, we have identified Ptpn9a, an ortholog of human PTPN9, as a crucial regulator of erythroid cell development in zebrafish embryos. ptpn9a, but not ptpn9b, was expressed in the posterior lateral plate mesoderm and intermediate cell mass - two primitive hematopoietic sites during zebrafish embryogenesis. Morpholino-mediated knockdown of ptpn9a caused erythrocytes to be depleted by inhibiting erythroid cell maturation without affecting erythroid proliferation and apoptosis. Consistently, both dominant-negative PTPN9 (with mutation C515S) and siRNA against PTPN9 inhibited erythroid differentiation in human K562 cells. Mechanistically, depletion of ptpn9 in zebrafish embryos in vivo or in K562 cells in vitro increased phosphorylated STAT3, and the hyper-phosphorylated STAT3 entrapped and prevented the transcription factors GATA1 and ZBP-89 (also known as ZNF148) from regulating erythroid gene expression. These findings imply that PTPN9 plays an important role in erythropoiesis by disrupting an inhibitory complex of phosphorylated STAT3, GATA1 and ZBP-89, providing new cellular and molecular insights into the role of ptpn9a in developmental hematopoiesis.


Assuntos
Células Eritroides/enzimologia , Processamento de Proteína Pós-Traducional , Proteínas Tirosina Fosfatases não Receptoras/fisiologia , Fator de Transcrição STAT3/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/fisiologia , Peixe-Zebra/fisiologia , Animais , Embrião não Mamífero/citologia , Embrião não Mamífero/enzimologia , Eritropoese , Fator de Transcrição GATA1/metabolismo , Gastrulação , Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Células K562 , Fosforilação , Fatores de Transcrição/metabolismo , Peixe-Zebra/embriologia
12.
Mol Genet Genomics ; 289(5): 795-806, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24752400

RESUMO

PTP1B is an important negative regulator of insulin and other signaling pathways in mammals. However, the role of PTP1B in the regulation of RAS-MAPK signaling remains open to deliberation, due to conflicting evidence from different experimental systems. The Drosophila orthologue of mammalian PTP1B, PTP61F, has until recently remained largely uncharacterized. To establish the potential role of PTP61F in the regulation of signaling pathways in Drosophila and particularly to help resolve its fundamental function in RAS-MAPK signaling, we generated a new allele of Ptp61F as well as employed both RNA interference and overexpression alleles. Our results validate recent data showing that the activity of insulin and Abl kinase signaling is increased in Ptp61F mutants and RNA interference lines. Importantly, we establish negative regulation of the RAS/MAPK pathway by Ptp61F activity in whole animals. Of particular interest, our results document the modulation of hyperactive MAP kinase activity by Ptp61F alleles, showing that the phosphatase intervenes to directly or indirectly regulate MAP kinase itself.


Assuntos
Proteínas de Drosophila/fisiologia , Drosophila melanogaster/enzimologia , Sistema de Sinalização das MAP Quinases , Proteína Tirosina Fosfatase não Receptora Tipo 1/fisiologia , Proteínas Tirosina Fosfatases não Receptoras/fisiologia , Animais , Olho Composto de Artrópodes/enzimologia , Olho Composto de Artrópodes/crescimento & desenvolvimento , Drosophila melanogaster/crescimento & desenvolvimento , Epistasia Genética , Receptores ErbB/metabolismo , Feminino , Estudos de Associação Genética , Masculino , Dados de Sequência Molecular , Asas de Animais/enzimologia , Asas de Animais/crescimento & desenvolvimento
13.
Biol Psychiatry ; 74(11): 817-26, 2013 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-24012328

RESUMO

BACKGROUND: Striatal-enriched protein tyrosine phosphatase (STEP) is a brain-specific protein tyrosine phosphatase that opposes the development of synaptic strengthening and the consolidation of fear memories. In contrast, stress facilitates fear memory formation, potentially by activating corticotrophin releasing factor (CRF) neurons in the anterolateral cell group of the bed nucleus of the stria terminalis (BNSTALG). METHODS: Here, using dual-immunofluorescence, single-cell reverse transcriptase polymerase chain reaction, quantitative reverse transcriptase polymerase chain reaction, Western blot, and whole-cell patch-clamp electrophysiology, we examined the expression and role of STEP in regulating synaptic plasticity in rat BNSTALG neurons and its modulation by stress. RESULTS: Striatal-enriched protein tyrosine phosphatase was selectively expressed in CRF neurons in the oval nucleus of the BNSTALG. Following repeated restraint stress (RRS), animals displayed a significant increase in anxiety-like behavior, which was associated with a downregulation of STEP messenger RNA and protein expression in the BNSTALG, as well as selectively enhancing the magnitude of long-term potentiation (LTP) induced in Type III, putative CRF neurons. To determine if the changes in STEP expression following RRS were mechanistically related to LTP facilitation, we examined the effects of intracellular application of STEP on the induction of LTP. STEP completely blocked the RRS-induced facilitation of LTP in BNSTALG neurons. CONCLUSIONS: Hence, STEP acts to buffer CRF neurons against excessive activation, while downregulation of STEP after chronic stress may result in pathologic activation of CRF neurons in the BNSTALG and contribute to prolonged states of anxiety. Thus, targeted manipulations of STEP activity might represent a novel treatment strategy for stress-induced anxiety disorders.


Assuntos
Hormônio Liberador da Corticotropina/metabolismo , Neurônios/enzimologia , Proteínas Tirosina Fosfatases não Receptoras/metabolismo , Núcleos Septais/enzimologia , Estresse Fisiológico , Animais , Masculino , Proteínas Tirosina Fosfatases não Receptoras/fisiologia , Ratos , Ratos Sprague-Dawley , Transdução de Sinais
14.
Curr Top Microbiol Immunol ; 362: 209-33, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23086420

RESUMO

The level and turnover of phosphoinositides (PIs) are tightly controlled by a large set of PI-specific enzymes (PI kinases and phosphatases). Mammalian PI phosphatases are conserved through evolution and among this large family the dual-specificity phosphatase (PTP/DSP) are metal-independent enzymes displaying the amino acid signature Cys-X5-Arg-Thr/Ser (CX5RT/S) in their active site. Such catalytic site characterizes the myotubularin 3-phosphatases that dephosphorylate PtdIns3P and PtdIns(3,5)P2 and produce PtdIns5P. Substrates of myotubularins have been implicated in endocytosis and membrane trafficking while PtdIns5P may have a role in signal transduction. As a paradox, 6 of the 14 members of the myotubularin family lack enzymatic activity and are considered as dead phosphatases. Several myotubularins have been genetically linked to human diseases: MTM1 is mutated in the congenital myopathy X-linked centronuclear or myotubular myopathy (XLCNM) and MTMR14 (JUMPY) has been linked to an autosomal form of such disease, while MTMR2 and MTMR13 are mutated in Charcot-Marie-Tooth (CMT) neuropathies. Furthermore, recent evidences from genetic association studies revealed that several other myotubularins could be associated to chronic disorders such as cancer and obesity, highlighting their importance for human health. Here, we discuss cellular and physiological roles of myotubularins and their implication in human diseases, and we present potential pathological mechanisms affecting specific tissues in myotubularin-associated diseases.


Assuntos
Proteínas Tirosina Fosfatases não Receptoras/fisiologia , Animais , Doença de Charcot-Marie-Tooth/etiologia , Humanos , Miopatias Congênitas Estruturais/etiologia , Fosfatidilinositóis/metabolismo
15.
Proc Natl Acad Sci U S A ; 109(24): 9539-44, 2012 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-22647598

RESUMO

The myotubularins are a large family of inositol polyphosphate 3-phosphatases that, despite having common substrates, subsume unique functions in cells that are disparate. The myotubularin family consists of 16 different proteins, 9 members of which possess catalytic activity, dephosphorylating phosphatidylinositol 3-phosphate [PtdIns(3)P] and phosphatidylinositol 3,5-bisphosphate [PtdIns(3,5)P(2)] at the D-3 position. Seven members are inactive because they lack the conserved cysteine residue in the CX(5)R motif required for activity. We studied a subfamily of homologous myotubularins, including myotubularin-related protein 6 (MTMR6), MTMR7, and MTMR8, all of which dimerize with the catalytically inactive MTMR9. Complex formation between the active myotubularins and MTMR9 increases their catalytic activity and alters their substrate specificity, wherein the MTMR6/R9 complex prefers PtdIns(3,5)P(2) as substrate; the MTMR8/R9 complex prefers PtdIns(3)P. MTMR9 increased the enzymatic activity of MTMR6 toward PtdIns(3,5)P(2) by over 30-fold, and enhanced the activity toward PtdIns(3)P by only 2-fold. In contrast, MTMR9 increased the activity of MTMR8 by 1.4-fold and 4-fold toward PtdIns(3,5)P(2) and PtdIns(3)P, respectively. In cells, the MTMR6/R9 complex significantly increases the cellular levels of PtdIns(5)P, the product of PI(3,5)P(2) dephosphorylation, whereas the MTMR8/R9 complex reduces cellular PtdIns(3)P levels. Consequentially, the MTMR6/R9 complex serves to inhibit stress-induced apoptosis and the MTMR8/R9 complex inhibits autophagy.


Assuntos
Autofagia/fisiologia , Proteínas Tirosina Fosfatases não Receptoras/fisiologia , Catálise , Células HeLa , Humanos , Microscopia de Fluorescência , Proteínas Tirosina Fosfatases não Receptoras/metabolismo , Especificidade por Substrato
16.
Artigo em Inglês | MEDLINE | ID: mdl-22369717

RESUMO

Lafora disease (LD) is a fatal autosomal recessive form of progressive myoclonus epilepsy. Patients manifest myoclonus and tonic-clonic seizures, visual hallucinations, intellectual, and progressive neurologic deterioration beginning in adolescence. The two genes known to be involved in Lafora disease are EPM2A and NHLRC1 (EPM2B). The EPM2A gene encodes laforin, a dual-specificity protein phosphatase, and the NHLRC1 gene encodes malin, an E3-ubiquitin ligase. The two proteins interact with each other and, as a complex, are thought to regulate glycogen synthesis. It may also be considered as a disorder of carbohydrate metabolism because of the formation of polyglucosan inclusion bodies in neural and other tissues due to abnormalities of the proteins laforin or malin. The review also outlines important patents related to Lafora disease.


Assuntos
Proteínas de Transporte/fisiologia , Doença de Lafora/fisiopatologia , Proteínas Tirosina Fosfatases não Receptoras/fisiologia , Metabolismo dos Carboidratos , Proteínas de Transporte/genética , Glicogênio/biossíntese , Humanos , Doença de Lafora/genética , Neurônios/patologia , Proteínas Tirosina Fosfatases não Receptoras/genética , Ubiquitina-Proteína Ligases
17.
Prog Mol Biol Transl Sci ; 106: 343-79, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22340724

RESUMO

Alzheimer's Disease (AD) is characterized by progressive loss of cognitive function, linked to marked neuronal loss. Pathological hallmarks of the disease are the accumulation of the amyloid-ß (Aß) peptide in the form of amyloid plaques and the intracellular formation of neurofibrillary tangles (NFTs). Accumulating evidence supports a key role for protein phosphorylation in both the normal and pathological actions of Aß as well as the formation of NFTs. NFTs contain hyperphosphorylated forms of the microtubule-binding protein tau, and phosphorylation of tau by several different kinases leads to its aggregation. The protein kinases involved in the generation and/or actions of tau or Aß are viable drug targets to prevent or alleviate AD pathology. However, it has also been recognized that the protein phosphatases that reverse the actions of these protein kinases are equally important. Here, we review recent advances in our understanding of serine/threonine and tyrosine protein phosphatases in the pathology of AD.


Assuntos
Doença de Alzheimer/enzimologia , Proteínas do Tecido Nervoso/fisiologia , Fosfoproteínas Fosfatases/fisiologia , Processamento de Proteína Pós-Traducional , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/fisiopatologia , Peptídeos beta-Amiloides/metabolismo , Animais , Calcineurina/fisiologia , Inibidores de Calcineurina , Modelos Animais de Doenças , Neurônios Dopaminérgicos/enzimologia , Neurônios Dopaminérgicos/fisiologia , Inibidores Enzimáticos/uso terapêutico , Humanos , Camundongos , Camundongos Transgênicos , Proteínas Nucleares/fisiologia , Fosfoproteínas/metabolismo , Fosforilação , Proteína Fosfatase 1/fisiologia , Proteína Fosfatase 2/fisiologia , Proteínas Tirosina Fosfatases não Receptoras/fisiologia , Tauopatias/enzimologia , Proteínas tau/metabolismo
18.
Nat Commun ; 3: 616, 2012 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-22233626

RESUMO

Hereditary haemorrhagic telangiectasia (HHT) [corrected] is a vascular dysplasia syndrome caused by mutations in transforming growth factor-ß/bone morphogenetic protein pathway genes, ENG and ACVRL1. HHT [corrected] shows considerable variation in clinical manifestations, suggesting environmental and/or genetic modifier effects. Strain-specific penetrance of the vascular phenotypes of Eng(+/-) and Tgfb1(-/-) mice provides further support for genetic modification of transforming growth factor-ß pathway deficits. We previously identified variant genomic loci, including Tgfbm2, which suppress prenatal vascular lethality of Tgfb1(-/-) mice. Here we show that human polymorphic variants of PTPN14 within the orthologous TGFBM2 locus influence clinical severity of HHT, [corrected] as assessed by development of pulmonary arteriovenous malformation. We also show that PTPN14, ACVRL1 and EFNB2, encoding EphrinB2, show interdependent expression in primary arterial endothelial cells in vitro. This suggests an involvement of PTPN14 in angiogenesis and/or arteriovenous fate, acting via EphrinB2 and ACVRL1/activin receptor-like kinase 1. These findings contribute to a deeper understanding of the molecular pathology of HHT [corrected] in particular and to angiogenesis in general.


Assuntos
Proteínas Tirosina Fosfatases não Receptoras/fisiologia , Telangiectasia Hemorrágica Hereditária/genética , Receptores de Ativinas Tipo I/metabolismo , Receptores de Activinas Tipo II/metabolismo , Animais , Mapeamento Cromossômico , Efrina-B2/metabolismo , Éxons , Feminino , Variação Genética , Haplótipos , Humanos , Masculino , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Genéticos , Mutação , Fenótipo , Proteínas Tirosina Fosfatases não Receptoras/genética , Especificidade da Espécie , Fator de Crescimento Transformador beta/metabolismo
19.
Pharmacol Rev ; 64(1): 65-87, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22090472

RESUMO

Striatal-enriched protein tyrosine phosphatase (STEP) is a brain-specific phosphatase that modulates key signaling molecules involved in synaptic plasticity and neuronal function. Targets include extracellular-regulated kinase 1 and 2 (ERK1/2), stress-activated protein kinase p38 (p38), the Src family tyrosine kinase Fyn, N-methyl-D-aspartate receptors (NMDARs), and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs). STEP-mediated dephosphorylation of ERK1/2, p38, and Fyn leads to inactivation of these enzymes, whereas STEP-mediated dephosphorylation of surface NMDARs and AMPARs promotes their endocytosis. Accordingly, the current model of STEP function posits that it opposes long-term potentiation and promotes long-term depression. Phosphorylation, cleavage, dimerization, ubiquitination, and local translation all converge to maintain an appropriate balance of STEP in the central nervous system. Accumulating evidence over the past decade indicates that STEP dysregulation contributes to the pathophysiology of several neuropsychiatric disorders, including Alzheimer's disease, schizophrenia, fragile X syndrome, epileptogenesis, alcohol-induced memory loss, Huntington's disease, drug abuse, stroke/ischemia, and inflammatory pain. This comprehensive review discusses STEP expression and regulation and highlights how disrupted STEP function contributes to the pathophysiology of diverse neuropsychiatric disorders.


Assuntos
Transtornos Mentais , Doenças do Sistema Nervoso , Proteínas Tirosina Fosfatases não Receptoras , Encéfalo/metabolismo , Dimerização , Humanos , Transtornos Mentais/tratamento farmacológico , Transtornos Mentais/etiologia , Transtornos Mentais/metabolismo , Doenças do Sistema Nervoso/tratamento farmacológico , Doenças do Sistema Nervoso/etiologia , Doenças do Sistema Nervoso/metabolismo , Fosforilação , Conformação Proteica , Proteínas Tirosina Fosfatases não Receptoras/genética , Proteínas Tirosina Fosfatases não Receptoras/metabolismo , Proteínas Tirosina Fosfatases não Receptoras/fisiologia , Especificidade por Substrato
20.
Structure ; 19(10): 1485-95, 2011 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-21889351

RESUMO

Alix and cellular paralogs HD-PTP and Brox contain N-terminal Bro1 domains that bind ESCRT-III CHMP4. In contrast to HD-PTP and Brox, expression of the Bro1 domain of Alix alleviates HIV-1 release defects that result from interrupted access to ESCRT. In an attempt to elucidate this functional discrepancy, we solved the crystal structures of the Bro1 domains of HD-PTP and Brox. They revealed typical "boomerang" folds they share with the Bro1 Alix domain. However, they each contain unique structural features that may be relevant to their specific function(s). In particular, phenylalanine residue in position 105 (Phe105) of Alix belongs to a long loop that is unique to its Bro1 domain. Concurrently, mutation of Phe105 and surrounding residues at the tip of the loop compromise the function of Alix in HIV-1 budding without affecting its interactions with Gag or CHMP4. These studies identify a new functional determinant in the Bro1 domain of Alix.


Assuntos
Proteínas de Ligação ao Cálcio/química , Proteínas de Ciclo Celular/química , Complexos Endossomais de Distribuição Requeridos para Transporte/química , HIV-1/química , Fenilalanina/química , Liberação de Vírus , Clonagem Molecular , Complexos Endossomais de Distribuição Requeridos para Transporte/fisiologia , Escherichia coli/genética , Escherichia coli/metabolismo , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Células HEK293 , HIV-1/fisiologia , Humanos , Imunoprecipitação , Mutagênese Sítio-Dirigida , Dobramento de Proteína , Mapeamento de Interação de Proteínas , Estrutura Secundária de Proteína , Proteínas Tirosina Fosfatases não Receptoras/química , Proteínas Tirosina Fosfatases não Receptoras/fisiologia , Relação Estrutura-Atividade , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...