Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Cell Physiol Biochem ; 46(6): 2335-2346, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29742497

RESUMO

BACKGROUND/AIMS: During the occurrence and progression of hepatocellular carcinoma (HCC), phosphotyrosine phosphatases (PTPs) are usually described as tumor suppressors or proto-oncogenes, and to some degree are correlated with the prognosis of HCC. METHODS: A total of 321 patients from the Cancer Genome Atlas (TCGA) database and 180 patients from our validated cohort with hepatocellular carcinoma were recruited in this study. Kaplan-Meier, univariate and multivariate Cox proportional hazards model were used to evaluate the risk factors for survival. Quantitative real-time PCR (qRT-PCR) and immunohistochemistry (IHC) were applied to detect the expression levels of PTP genes. RESULTS: After screening the data of TCGA, we identified five PTPs as HCC overall survival related PTP genes, among which only three (PTPN12, PTPRN, PTPN18) exhibited differential expression levels in our 180 paired HCC and adjacent tissues (P< 0.001). Further analysis revealed that expression of PTPN18 was positively, but PTPRN was negatively associated with prognosis of HCC both in TCGA cohort and our own cohort. As to PTPN12, results turned out to be opposite according to HBV status. In detail, higher expression of PTPN12 was associated with better outcome in HBV group but worse prognosis in Non-HBV group. CONCLUSION: Our results suggested that PTPN12, PTPRN and PTPN18 were independent prognostic factors in HCC.


Assuntos
Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/genética , Proteína Tirosina Fosfatase não Receptora Tipo 12/genética , Proteínas Tirosina Fosfatases não Receptoras/genética , Proteínas Tirosina Fosfatases Classe 8 Semelhantes a Receptores/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Carcinoma Hepatocelular/diagnóstico , Estudos de Coortes , Feminino , Regulação da Expressão Gênica , Humanos , Estimativa de Kaplan-Meier , Neoplasias Hepáticas/diagnóstico , Masculino , Pessoa de Meia-Idade , Prognóstico , Proteína Tirosina Fosfatase não Receptora Tipo 12/análise , Proteínas Tirosina Fosfatases não Receptoras/análise , Proteínas Tirosina Fosfatases Classe 8 Semelhantes a Receptores/análise
2.
Proc Natl Acad Sci U S A ; 113(32): E4736-44, 2016 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-27457929

RESUMO

Phosphorylation regulates surface and synaptic expression of NMDA receptors (NMDARs). Both the tyrosine kinase Fyn and the tyrosine phosphatase striatal-enriched protein tyrosine phosphatase (STEP) are known to target the NMDA receptor subunit GluN2B on tyrosine 1472, which is a critical residue that mediates NMDAR endocytosis. STEP reduces the surface expression of NMDARs by promoting dephosphorylation of GluN2B Y1472, whereas the synaptic scaffolding protein postsynaptic density protein 95 (PSD-95) stabilizes the surface expression of NMDARs. However, nothing is known about a potential functional interaction between STEP and PSD-95. We now report that STEP61 binds to PSD-95 but not to other PSD-95 family members. We find that PSD-95 expression destabilizes STEP61 via ubiquitination and degradation by the proteasome. Using subcellular fractionation, we detect low amounts of STEP61 in the PSD fraction. However, STEP61 expression in the PSD is increased upon knockdown of PSD-95 or in vivo as detected in PSD-95-KO mice, demonstrating that PSD-95 excludes STEP61 from the PSD. Importantly, only extrasynaptic NMDAR expression and currents were increased upon STEP knockdown, as is consistent with low STEP61 localization in the PSD. Our findings support a dual role for PSD-95 in stabilizing synaptic NMDARs by binding directly to GluN2B but also by promoting synaptic exclusion and degradation of the negative regulator STEP61.


Assuntos
Proteína 4 Homóloga a Disks-Large/fisiologia , Proteínas Tirosina Fosfatases não Receptoras/metabolismo , Receptores de N-Metil-D-Aspartato/fisiologia , Animais , Feminino , Células HEK293 , Humanos , Camundongos , Complexo de Endopeptidases do Proteassoma/fisiologia , Proteínas Tirosina Fosfatases não Receptoras/análise , Ratos , Ratos Sprague-Dawley , Receptores de N-Metil-D-Aspartato/metabolismo , Ubiquitinação
3.
Biochem Biophys Res Commun ; 476(3): 167-73, 2016 07 29.
Artigo em Inglês | MEDLINE | ID: mdl-27155155

RESUMO

Phosphoinositides play a key role in the spatiotemporal control of central intracellular processes and several specific kinases and phosphatases regulating the level of these lipids are implicated in human diseases. Myotubularins are a family of 3-phosphatases acting specifically on phosphatidylinositol 3-monophosphate and phosphatidylinositol 3,5 bisphosphate. Members of this family are mutated in genetic diseases including myotubularin 1 (MTM1) and myotubularin-related protein 2 (MTMR2) which mutations are responsible of X-linked centronuclear myopathy and Charcot-Marie-Tooth neuropathy, respectively. Here we show that MTM1 is expressed in blood platelets and that hundred microliters of blood is sufficient to detect the protein by western blotting. Since the most severe cases of pathogenic mutations of MTM1 lead to loss of expression of the protein, we propose that a minimal amount of blood can allow a rapid diagnostic test of X-linked myotubular myopathy, which is currently based on histopathology of muscle biopsy and molecular genetic testing. In platelets, MTM1 is a highly active 3-phosphatase mainly associated to membranes and found on the dense granules and to a lesser extent on alpha-granules. However, deletion of MTM1 in mouse had no significant effect on platelet count and on platelet secretion and aggregation induced by thrombin or collagen stimulation. Potential compensation by other members of the myotubularin family is conceivable since MTMR2 was easily detectable by western blotting and the mRNA of several members of the family increased during in vitro differentiation of human megakaryocytes and MEG-01 cells. In conclusion, we show the presence of several myotubularins in platelets and propose that minimal amounts of blood can be used to develop a rapid diagnostic test for genetic pathologies linked to loss of expression of these phosphatases.


Assuntos
Plaquetas/patologia , Miopatias Congênitas Estruturais/diagnóstico , Proteínas Tirosina Fosfatases não Receptoras/análise , Animais , Plaquetas/citologia , Plaquetas/metabolismo , Linhagem Celular , Regulação da Expressão Gênica , Humanos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miopatias Congênitas Estruturais/sangue , Miopatias Congênitas Estruturais/genética , Agregação Plaquetária , Proteínas Tirosina Fosfatases não Receptoras/sangue , Proteínas Tirosina Fosfatases não Receptoras/genética , RNA Mensageiro/genética
4.
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
5.
Mol Neurobiol ; 49(2): 645-57, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24068615

RESUMO

In Lafora disease (LD), the deficiency of either EPM2A or NHLRC1, the genes encoding the phosphatase laforin and E3 ligase, respectively, causes massive accumulation of less-branched glycogen inclusions, known as Lafora bodies, also called polyglucosan bodies (PBs), in several types of cells including neurons. The biochemical mechanism underlying the PB accumulation, however, remains undefined. We recently demonstrated that laforin is a phosphatase of muscle glycogen synthase (GS1) in PBs, and that laforin recruits malin, together reducing PBs. We show here that accomplishment of PB degradation requires a protein assembly consisting of at least four key enzymes: laforin and malin in a complex, and the glycogenolytic enzymes, glycogen debranching enzyme 1 (AGL1) and brain isoform glycogen phosphorylase (GPBB). Once GS1-synthesized polyglucosan accumulates into PBs, laforin recruits malin to the PBs where laforin dephosphorylates, and malin degrades the GS1 in concert with GPBB and AGL1, resulting in a breakdown of polyglucosan. Without fountional laforin-malin complex assembled on PBs, GPBB and AGL1 together are unable to efficiently breakdown polyglucosan. All these events take place on PBs and in cytoplasm. Deficiency of each of the four enzymes causes PB accumulation in the cytoplasm of affected cells. Demonstration of the molecular mechanisms underlying PB degradation lays a substantial biochemical foundation that may lead to understanding how PB metabolizes and why mutations of either EPM2A or NHLRC1 in humans cause LD. Mutations in AGL1 or GPBB may cause diseases related to PB accumulation.


Assuntos
Encéfalo/enzimologia , Proteínas de Transporte/metabolismo , Glucanos/metabolismo , Sistema da Enzima Desramificadora do Glicogênio/metabolismo , Glicogênio Fosforilase/metabolismo , Proteínas Tirosina Fosfatases não Receptoras/metabolismo , Animais , Proteínas de Transporte/análise , Linhagem Celular Tumoral , Glucanos/análise , Sistema da Enzima Desramificadora do Glicogênio/análise , Glicogênio Fosforilase/análise , Células HEK293 , Humanos , Isoenzimas/análise , Isoenzimas/metabolismo , Doença de Lafora/metabolismo , Doença de Lafora/patologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Tirosina Fosfatases não Receptoras/análise , Ubiquitina-Proteína Ligases
6.
Clin Biochem ; 46(18): 1869-76, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24012855

RESUMO

OBJECTIVES: Lafora disease is a rare yet invariably fatal form of progressive neurodegenerative epilepsy resulting from mutations in the phosphatase laforin. Several therapeutic options for Lafora disease patients are currently being explored, and these therapies would benefit from a biochemical means of assessing functional laforin activity following treatment. To date, only clinical outcomes such as decreases in seizure frequency and severity have been used to indicate success of epilepsy treatment. However, these qualitative measures exhibit variability and must be assessed over long periods of time. In this work, we detail a simple and sensitive bioassay that can be used for the detection of functional endogenous laforin from human and mouse tissue. DESIGN AND METHODS: We generated antibodies capable of detecting and immunoprecipitating endogenous laforin. Following laforin immunoprecipitation, laforin activity was assessed via phosphatase assays using para-nitrophenylphosphate (pNPP) and a malachite green-based assay specific for glucan phosphatase activity. RESULTS: We found that antibody binding to laforin does not impede laforin activity. Furthermore, the malachite green-based glucan phosphatase assay used in conjunction with a rabbit polyclonal laforin antibody was capable of detecting endogenous laforin activity from human and mouse tissues. Importantly, this assay discriminated between laforin activity and other phosphatases. CONCLUSIONS: The bioassay that we have developed utilizing laforin antibodies and an assay specific for glucan phosphatase activity could prove valuable in the rapid detection of functional laforin in patients to which novel Lafora disease therapies have been administered.


Assuntos
Bioensaio/métodos , Fosfatases de Especificidade Dupla/análise , Doença de Lafora/metabolismo , Proteínas Tirosina Fosfatases não Receptoras/análise , Animais , Fosfatases de Especificidade Dupla/imunologia , Fosfatases de Especificidade Dupla/metabolismo , Células Hep G2 , Humanos , Imunoprecipitação , Camundongos , Camundongos Endogâmicos C57BL , Músculo Esquelético/metabolismo , Nitrofenóis/química , Compostos Organofosforados/química , Proteínas Tirosina Fosfatases não Receptoras/imunologia , Proteínas Tirosina Fosfatases não Receptoras/metabolismo , Coelhos , Corantes de Rosanilina/química , Pele/metabolismo
7.
Hum Mol Genet ; 21(4): 811-25, 2012 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-22068590

RESUMO

X-linked myotubular myopathy (MTM) is a severe neuromuscular disease of infancy caused by mutations of MTM1, which encodes the phosphoinositide lipid phosphatase, myotubularin. The Mtm1 knockout (KO) mouse has a severe phenotype and its short lifespan (8 weeks) makes it a challenge to use as a model in the testing of certain preclinical therapeutics. Many MTM patients succumb early in life, but some have a more favorable prognosis. We used human genotype-phenotype correlation data to develop a myotubularin-deficient mouse model with a less severe phenotype than is seen in Mtm1 KO mice. We modeled the human c.205C>T point mutation in Mtm1 exon 4, which is predicted to introduce the p.R69C missense change in myotubularin. Hemizygous male Mtm1 p.R69C mice develop early muscle atrophy prior to the onset of weakness at 2 months. The median survival period is 66 weeks. Histopathology shows small myofibers with centrally placed nuclei. Myotubularin protein is undetectably low because the introduced c.205C>T base change induced exon 4 skipping in most mRNAs, leading to premature termination of myotubularin translation. Some full-length Mtm1 mRNA bearing the mutation is present, which provides enough myotubularin activity to account for the relatively mild phenotype, as Mtm1 KO and Mtm1 p.R69C mice have similar muscle phosphatidylinositol 3-phosphate levels. These data explain the basis for phenotypic variability among human patients with MTM1 p.R69C mutations and establish the Mtm1 p.R69C mouse as a valuable model for the disease, as its less severe phenotype will expand the scope of testable preclinical therapies.


Assuntos
Modelos Animais de Doenças , Éxons/genética , Estudos de Associação Genética , Miopatias Congênitas Estruturais/genética , Miopatias Congênitas Estruturais/patologia , Mutação Puntual/genética , Proteínas Tirosina Fosfatases não Receptoras/genética , Animais , Cálcio/metabolismo , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Esquelético/metabolismo , Mutação de Sentido Incorreto/genética , Miopatias Congênitas Estruturais/fisiopatologia , Fenótipo , Fosfatos de Fosfatidilinositol/metabolismo , Proteínas Tirosina Fosfatases não Receptoras/análise , Proteínas Tirosina Fosfatases não Receptoras/biossíntese , Proteínas Tirosina Fosfatases não Receptoras/metabolismo
8.
PLoS One ; 6(8): e24040, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21887368

RESUMO

Lafora Disease (LD) is a fatal neurodegenerative epileptic disorder that presents as a neurological deterioration with the accumulation of insoluble, intracellular, hyperphosphorylated carbohydrates called Lafora bodies (LBs). LD is caused by mutations in either the gene encoding laforin or malin. Laforin contains a dual specificity phosphatase domain and a carbohydrate-binding module, and is a member of the recently described family of glucan phosphatases. In the current study, we investigated the functional and physiological relevance of laforin dimerization. We purified recombinant human laforin and subjected the monomer and dimer fractions to denaturing gel electrophoresis, mass spectrometry, phosphatase assays, protein-protein interaction assays, and glucan binding assays. Our results demonstrate that laforin prevalently exists as a monomer with a small dimer fraction both in vitro and in vivo. Of mechanistic importance, laforin monomer and dimer possess equal phosphatase activity, and they both associate with malin and bind glucans to a similar extent. However, we found differences between the two states' ability to interact simultaneously with malin and carbohydrates. Furthermore, we tested other members of the glucan phosphatase family. Cumulatively, our data suggest that laforin monomer is the dominant form of the protein and that it contains phosphatase activity.


Assuntos
Fosfatases de Especificidade Dupla/análise , Doença de Lafora/enzimologia , Proteínas Tirosina Fosfatases não Receptoras/metabolismo , Carboidratos , Proteínas de Transporte/metabolismo , Fosfatases de Especificidade Dupla/metabolismo , Humanos , Ligação Proteica , Multimerização Proteica , Proteínas Tirosina Fosfatases não Receptoras/análise , Proteínas Tirosina Fosfatases não Receptoras/química , Ubiquitina-Proteína Ligases
9.
J Assist Reprod Genet ; 28(9): 851-61, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21701840

RESUMO

PURPOSE: To understand the molecular basis of sperm-motility and to identify related novel motility biomarkers. METHODS: Two-dimensional electrophoresis (2DE) followed by Reverse-phase-nano-high-performance liquid chromatography-electrospray ionization tandem mass spectrometry (RP-nano-HPLC-ESI-MS/MS) were applied to establish the human sperm proteome. Then the sperm proteome of moderate-motile human sperm fraction and that of good-motile human sperm fraction from pooled spermatozoa of forty normozoospermic donors (Group 1 subjects) were compared to identify the dysregulated proteins. Among these down-regulated proteins, Protein tyrosine phosphatase non-receptor type 14 (PTPN14) was chosen to reconfirm by Western blotting and semi-quantitative reverse transcription polymerase chain reaction. For clinical application, Western blotting and real-time reverse transcription polymerase chain reaction was performed to compare the expression level of PTPN14 in (Group 2 subjects) nine normozoospermic controls and thirty-three asthenozoospermic patients (including 21 mild asthenozoospermic cases and 12 severe cases). Finally, bioinformatic tools prediction and immunofluorescence assay were performed to elucidate the potential localization of PTPN14. RESULTS: The expression levels of three proteins were observed to be lower in the moderate-motile sperm fraction than in good-motile sperm of group 1 subjects. Among three proteins with persistent down-regulation in the moderate-motile sperm, we reconfirmed that the expression level of PTPN14 was significantly lower in both mRNA and protein levels from the moderate-motile sperm fraction. Further, down-regulation of PTPN14 was found at the translational and transcriptional level in the asthenozoospermic men. Finally, Bioinformatic tools prediction and immunofluorescence assay showed that PTPN14 maybe predominantly localized at the mitochondria in the midpiece of human ejaculated sperm. CONCLUSIONS: Proteomics tools were applied to identify three possible sperm motility-related proteins. Among these proteins, PTPN14 was highly likely a novel sperm-motility biomarker and a potential mitochondrial protein.


Assuntos
Proteínas Tirosina Fosfatases não Receptoras/metabolismo , Motilidade dos Espermatozoides , Espermatozoides/metabolismo , Adulto , Biomarcadores/análise , Biomarcadores/metabolismo , Cromatografia Líquida , Cromatografia de Fase Reversa , Eletroforese em Gel Bidimensional , Humanos , Masculino , Mitocôndrias/metabolismo , Proteínas Tirosina Fosfatases não Receptoras/análise , Proteínas Tirosina Fosfatases não Receptoras/genética , RNA Mensageiro/metabolismo , Peça Intermédia do Espermatozoide/metabolismo , Espermatozoides/fisiologia , Espectrometria de Massas em Tandem
10.
Traffic ; 11(4): 468-78, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20059746

RESUMO

Autophagy is a catabolic process that delivers cytoplasmic material to the lysosome for degradation. The mechanisms regulating autophagosome formation and size remain unclear. Here, we show that autophagosome formation was triggered by the overexpression of a dominant-negative inactive mutant of Myotubularin-related phosphatase 3 (MTMR3). Mutant MTMR3 partially localized to autophagosomes, and PtdIns3P and two autophagy-related PtdIns3P-binding proteins, GFP-DFCP1 and GFP-WIPI-1alpha (WIPI49/Atg18), accumulated at sites of autophagosome formation. Knock-down of MTMR3 increased autophagosome formation, and overexpression of wild-type MTMR3 led to significantly smaller nascent autophagosomes and a net reduction in autophagic activity. These results indicate that autophagy initiation depends on the balance between PI 3-kinase and PI 3-phosphatase activity. Local levels of PtdIns3P at the site of autophagosome formation determine autophagy initiation and the size of the autophagosome membrane structure.


Assuntos
Autofagia/fisiologia , Fosfatos de Fosfatidilinositol/fisiologia , Proteínas Tirosina Fosfatases não Receptoras/fisiologia , Autofagia/genética , Linhagem Celular , Humanos , Lisossomos/enzimologia , Lisossomos/fisiologia , Fagossomos/enzimologia , Fagossomos/fisiologia , Fosfatidilinositol 3-Quinases/análise , Fosfatidilinositol 3-Quinases/fisiologia , Fosfatos de Fosfatidilinositol/análise , Proteínas Tirosina Fosfatases não Receptoras/análise , Proteínas Tirosina Fosfatases não Receptoras/genética
11.
PLoS One ; 4(3): e4979, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19325702

RESUMO

BACKGROUND: It has been shown that mutations in at least four myotubularin family genes (MTM1, MTMR1, 2 and 13) are causative for human neuromuscular disorders. However, the pathway and regulative mechanism remain unknown. METHODOLOGY/PRINCIPAL FINDINGS: Here, we reported a new role for Mtmr8 in neuromuscular development of zebrafish. Firstly, we cloned and characterized zebrafish Mtmr8, and revealed the expression pattern predominantly in the eye field and somites during early somitogenesis. Using morpholino knockdown, then, we observed that loss-of-function of Mtmr8 led to defects in somitogenesis. Subsequently, the possible underlying mechanism and signal pathway were examined. We first checked the Akt phosphorylation, and observed an increase of Akt phosphorylation in the morphant embryos. Furthermore, we studied the PH/G domain function within Mtmr8. Although the PH/G domain deletion by itself did not result in embryonic defect, addition of PI3K inhibitor LY294002 did give a defective phenotype in the PH/G deletion morphants, indicating that the PH/G domain was essential for Mtmr8's function. Moreover, we investigated the cooperation of Mtmr8 with PI3K in actin filament modeling and muscle development, and found that both Mtmr8-MO1 and Mtmr8-MO2+LY294002 led to the disorganization of the actin cytoskeleton. In addition, we revealed a possible participation of Mtmr8 in the Hedgehog pathway, and cell transplantation experiments showed that Mtmr8 worked in a non-cell autonomous manner in actin modeling. CONCLUSION/SIGNIFICANCE: The above data indicate that a conserved functional cooperation of Mtmr8 with PI3K regulates actin filament modeling and muscle development in zebrafish, and reveal a possible participation of Mtmr8 in the Hedgehog pathway. Therefore, this work provides a new clue to study the physiological function of MTM family members.


Assuntos
Citoesqueleto de Actina/metabolismo , Desenvolvimento Muscular , Fosfatidilinositol 3-Quinases/fisiologia , Proteínas Tirosina Fosfatases não Receptoras/fisiologia , Proteínas de Peixe-Zebra/fisiologia , Animais , Embrião não Mamífero , Olho/química , Proteínas Hedgehog , Organogênese , Monoéster Fosfórico Hidrolases/fisiologia , Fosforilação , Proteínas Tirosina Fosfatases não Receptoras/análise , Proteínas Tirosina Fosfatases não Receptoras/genética , Somitos/química , Distribuição Tecidual , Peixe-Zebra , Proteínas de Peixe-Zebra/análise , Proteínas de Peixe-Zebra/genética
12.
Methods Mol Biol ; 462: 265-78, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19160676

RESUMO

Phosphoinositides (PPIn) are important regulators of cellular processes like intracellular protein transport, cellular proliferation, apoptosis, and cytoskeletal organization. The amount and localization of these membrane-bound second messengers are regulated through a set of specific phospholipases, lipid kinases, and phosphatases. The elucidation of PPIn-phosphatases and their cellular function has gained much attention because phosphatase dysregulation is often associated with human genetic diseases. Our laboratory has identified the 3'-PPIn-phosphatase myotubularin 1 (MTM1) mutated in X-linked myotubular myopathy (XLMTM). In addition, a whole family of myotubularin-related proteins (MTMR1-MTMR13) has been discovered. Some of them display phosphatase activity, whereas for other family members no enzymatic activity could be detected. Nevertheless, these "dead phosphatases" myotubularins are conserved throughout evolution and probably exert regulatory function by heteromeric interaction with active phosphatase members. It was shown that MTM1 and related phosphatases act on PtdIns3P and PtdIns(3,5)P2; both PPIn species are important regulators of endocytic pathways. We describe two methods to determine phosphatase activity and substrate specificity of myotubularins. One is an immunoprecipitation-phosphatase assay, testing the activity of myotubularin immunoprecipitated from overexpressing cells on artificial PPIn. The other method analyzes phosphatase activity indirectly ex vivo in transiently transfected mammalian cells. The presence and subcellular localization of the myotubularin substrate PtdIns3P were determined using a specific binding domain (2xFYVE) produced recombinantly as a biosensor.


Assuntos
Fosfatidilinositóis/metabolismo , Proteínas Tirosina Fosfatases não Receptoras/análise , Proteínas Tirosina Fosfatases não Receptoras/metabolismo , Animais , Técnicas Biossensoriais , Biotinilação , Extratos Celulares , Linhagem Celular , Humanos , Imunoprecipitação , Microscopia Confocal , Proteínas Tirosina Fosfatases não Receptoras/isolamento & purificação , Especificidade por Substrato , Transfecção
13.
Hum Mol Genet ; 17(19): 3010-20, 2008 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-18617530

RESUMO

The EPM2A gene, encoding the dual-phosphatase laforin, is mutated in a fatal form of progressive myoclonus epilepsy known as Lafora disease (LD). The EPM2A gene, by differential splicing of its transcripts, is known to encode two laforin isoforms having distinct carboxyl termini; a major isoform localized in the cytoplasm (laf331), and a minor isoform that is targeted to the nucleus as well (laf317). We show here that the two laforin isoforms interact with each other and form homo and heterodimers. The homodimer of laf331 display robust phosphatase activity, whereas the laf317 homodimer and the laf331-laf317 heterodimer lack phosphatase activity. Laf331 binds to glycogen only as a monomeric form. Laf317, on the other hand, was unable to bind to glycogen as a homodimer or as a heterodimer. Similar to laf331, laf317 interacts with and functions as a substrate for the malin ubiquitin ligase--a product of another gene defective in LD. Malin, however, shows higher affinity towards laf331 when compared with laf317. We have also tested the effect of LD-associated mutations, whose effects are restricted to the laf331 isoform, on laf331-laf317 interaction. Two such mutations are known and both abolish the interactions between laf317 and laf331 and their heterodimerization, but not the homodimerization property of laf331. Thus, laf317 could function as a dominant-negative regulator of laf331, and laf331-specific mutations might affect laf317 functions as well. Thus, our findings reveal a novel mechanism for the EPM2A gene function, regulated by alternative splicing, in normal as well as disease conditions.


Assuntos
Processamento Alternativo , Doença de Lafora/metabolismo , Proteínas Tirosina Fosfatases não Receptoras/genética , Proteínas Tirosina Fosfatases não Receptoras/metabolismo , Animais , Células COS , Proteínas de Transporte/metabolismo , Chlorocebus aethiops , Glicogênio/metabolismo , Humanos , Doença de Lafora/genética , Mutação , Ligação Proteica , Isoformas de Proteínas/análise , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transporte Proteico , Proteínas Tirosina Fosfatases não Receptoras/análise , Ubiquitina-Proteína Ligases
14.
Hum Mol Genet ; 17(14): 2132-43, 2008 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-18434328

RESUMO

Myotubular myopathy (XLMTM, OMIM 310400) is a severe congenital muscular disease due to mutations in the myotubularin gene (MTM1) and characterized by the presence of small myofibers with frequent occurrence of central nuclei. Myotubularin is a ubiquitously expressed phosphoinositide phosphatase with a muscle-specific role in man and mouse that is poorly understood. No specific treatment exists to date for patients with myotubular myopathy. We have constructed an adeno-associated virus (AAV) vector expressing myotubularin in order to test its therapeutic potential in a XLMTM mouse model. We show that a single intramuscular injection of this vector in symptomatic Mtm1-deficient mice ameliorates the pathological phenotype in the targeted muscle. Myotubularin replacement in mice largely corrects nuclei and mitochondria positioning in myofibers and leads to a strong increase in muscle volume and recovery of the contractile force. In addition, we used this AAV vector to overexpress myotubularin in wild-type skeletal muscle and get insight into its localization and function. We show that a substantial proportion of myotubularin associates with the sarcolemma and I band, including triads. Myotubularin overexpression in muscle induces the accumulation of packed membrane saccules and presence of vacuoles that contain markers of sarcolemma and T-tubules, suggesting that myotubularin is involved in plasma membrane homeostasis of myofibers. This study provides a proof-of-principle that local delivery of an AAV vector expressing myotubularin can improve the motor capacities of XLMTM muscle and represents a novel approach to study myotubularin function in skeletal muscle.


Assuntos
Membrana Celular/metabolismo , Terapia Genética , Músculo Esquelético/metabolismo , Miopatias Congênitas Estruturais/genética , Miopatias Congênitas Estruturais/terapia , Proteínas Tirosina Fosfatases não Receptoras/genética , Animais , Linhagem Celular , Membrana Celular/química , Membrana Celular/genética , Membrana Celular/patologia , Dependovirus/genética , Dependovirus/metabolismo , Feminino , Vetores Genéticos/genética , Homeostase , Injeções Intramusculares , Masculino , Camundongos , Músculo Esquelético/química , Músculo Esquelético/patologia , Músculo Esquelético/fisiopatologia , Miopatias Congênitas Estruturais/metabolismo , Miopatias Congênitas Estruturais/fisiopatologia , Fenótipo , Proteínas Tirosina Fosfatases não Receptoras/administração & dosagem , Proteínas Tirosina Fosfatases não Receptoras/análise , Proteínas Tirosina Fosfatases não Receptoras/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...