Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
J Clin Lab Anal ; 36(1): e24111, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34811816

RESUMO

BACKGROUND: Dual specificity phosphatase 22 (DUSP22), also named as Jun N-terminal kinase pathway associated phosphatase recently, is reported to be closely engaged in immune and inflammation regulation. This study aimed to investigate the interaction between synovium DUSP22 and serum DUSP22 levels and to explore their correlation with rheumatoid arthritis (RA) risk, inflammation, and disease activity. METHODS: Synovium and serum samples from 42 RA patients with knee involvement underwent arthroscopy, and 20 knee trauma patients were collected. Besides, serum samples from 40 healthy controls were also obtained. Synovium DUSP22 expression was detected by reverse transcription quantitative polymerase chain reaction, while serum DUSP22 level was detected by enzyme-linked immunosorbent assay. RESULTS: Synovium DUSP22 level was greatly decreased in RA patients compared to trauma controls (p < 0.001), and it was negatively correlated with tender joint count (TJC) (r = -0.318, p = 0.040), C-reactive protein (CRP) (r = -0.330, p = 0.033), and Lysholm score (r = -0.423, p = 0.005) in RA patients. Serum DUSP22 level was lowest in RA patients, followed by trauma controls, then highest in healthy controls (p < 0.001). Serum DUSP22 level was negatively associated with TJC (r = -0.438, p = 0.004), swollen joint count (SJC) (r = -0.372, p = 0.015), CRP (r = -0.391, p = 0.011), and disease activity score in 28 joints (DAS28ESR ) score (r = -0.406, p = 0.008), and it increased after treatment (p = 0.001) in RA patients. In addition, serum DUSP22 level positively related to synovium DUSP22 level in RA patients (r = 0.394, p = 0.010). CONCLUSION: Synovium and serum DUSP22 are intercorrelated and insufficiently expressed in RA patients; meanwhile, their deficiency correlates with increased systemic inflammation, disease activity, and joint dysfunction.


Assuntos
Artrite Reumatoide , Fosfatases de Especificidade Dupla/análise , Fosfatases da Proteína Quinase Ativada por Mitógeno/análise , Membrana Sinovial/química , Idoso , Artrite Reumatoide/sangue , Artrite Reumatoide/diagnóstico , Artrite Reumatoide/metabolismo , Biomarcadores/análise , Biomarcadores/sangue , Fosfatases de Especificidade Dupla/sangue , Fosfatases de Especificidade Dupla/metabolismo , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Fosfatases da Proteína Quinase Ativada por Mitógeno/sangue , Fosfatases da Proteína Quinase Ativada por Mitógeno/metabolismo
2.
Nat Commun ; 12(1): 2284, 2021 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-33863904

RESUMO

Drug resistance is a major obstacle to the treatment of most human tumors. In this study, we find that dual-specificity phosphatase 16 (DUSP16) regulates resistance to chemotherapy in nasopharyngeal carcinoma, colorectal cancer, gastric and breast cancer. Cancer cells expressing higher DUSP16 are intrinsically more resistant to chemotherapy-induced cell death than cells with lower DUSP16 expression. Overexpression of DUSP16 in cancer cells leads to increased resistance to cell death upon chemotherapy treatment. In contrast, knockdown of DUSP16 in cancer cells increases their sensitivity to treatment. Mechanistically, DUSP16 inhibits JNK and p38 activation, thereby reducing BAX accumulation in mitochondria to reduce apoptosis. Analysis of patient survival in head & neck cancer and breast cancer patient cohorts supports DUSP16 as a marker for sensitivity to chemotherapy and therapeutic outcome. This study therefore identifies DUSP16 as a prognostic marker for the efficacy of chemotherapy, and as a therapeutic target for overcoming chemoresistance in cancer.


Assuntos
Biomarcadores Tumorais/metabolismo , Fosfatases de Especificidade Dupla/metabolismo , Mitocôndrias/efeitos dos fármacos , Fosfatases da Proteína Quinase Ativada por Mitógeno/metabolismo , Neoplasias/terapia , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Biomarcadores Tumorais/análise , Biomarcadores Tumorais/genética , Fracionamento Celular , Linhagem Celular Tumoral , Quimioterapia Adjuvante , Cisplatino/farmacologia , Cisplatino/uso terapêutico , Intervalo Livre de Doença , Resistencia a Medicamentos Antineoplásicos , Fosfatases de Especificidade Dupla/análise , Feminino , Técnicas de Silenciamento de Genes , Técnicas de Inativação de Genes , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Camundongos , Pessoa de Meia-Idade , Mitocôndrias/metabolismo , Fosfatases da Proteína Quinase Ativada por Mitógeno/análise , Neoplasias/mortalidade , Neoplasias/patologia , Ensaios Antitumorais Modelo de Xenoenxerto , Proteína X Associada a bcl-2/metabolismo
3.
Int J Mol Sci ; 22(2)2021 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-33466673

RESUMO

Dual specificity phosphatases (DUSPs) play a crucial role in the regulation of intracellular signalling pathways, which in turn influence a broad range of physiological processes. DUSP malfunction is increasingly observed in a broad range of human diseases due to deregulation of key pathways, most notably the MAP kinase (MAPK) cascades. Dual specificity phosphatase 26 (DUSP26) is an atypical DUSP with a range of physiological substrates including the MAPKs. The residues that govern DUSP26 substrate specificity are yet to be determined; however, recent evidence suggests that interactions with a binding partner may be required for DUSP26 catalytic activity. DUSP26 is heavily implicated in cancer where, akin to other DUSPs, it displays both tumour-suppressive and -promoting properties, depending on the context. Here we review DUSP26 by evaluating its transcriptional patterns, protein crystallographic structure and substrate binding, as well as its physiological role(s) and binding partners, its role in human disease and the development of DUSP26 inhibitors.


Assuntos
Fosfatases de Especificidade Dupla/metabolismo , Fosfatases da Proteína Quinase Ativada por Mitógeno/metabolismo , Animais , Fosfatases de Especificidade Dupla/análise , Fosfatases de Especificidade Dupla/genética , Humanos , Fosfatases da Proteína Quinase Ativada por Mitógeno/análise , Fosfatases da Proteína Quinase Ativada por Mitógeno/genética , Modelos Moleculares , Neoplasias/genética , Neoplasias/metabolismo , Conformação Proteica , Mapas de Interação de Proteínas , Especificidade por Substrato , Ativação Transcricional
4.
Medicina (Kaunas) ; 55(5)2019 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-31121943

RESUMO

Background and objectives: Alzheimer's disease (AD) is a progressive neurodegenerative disease that results in severe dementia. Having ischemic strokes (IS) is one of the risk factors of the AD, but the molecular mechanisms that underlie IS and AD are not well understood. We thus aimed to identify common molecular biomarkers and pathways in IS and AD that can help predict the progression of these diseases and provide clues to important pathological mechanisms. Materials and Methods: We have analyzed the microarray gene expression datasets of IS and AD. To obtain robust results, combinatorial statistical methods were used to analyze the datasets and 26 transcripts (22 unique genes) were identified that were abnormally expressed in both IS and AD. Results: Gene Ontology (GO) and KEGG pathway analyses indicated that these 26 common dysregulated genes identified several altered molecular pathways: Alcoholism, MAPK signaling, glycine metabolism, serine metabolism, and threonine metabolism. Further protein-protein interactions (PPI) analysis revealed pathway hub proteins PDE9A, GNAO1, DUSP16, NTRK2, PGAM2, MAG, and TXLNA. Transcriptional and post-transcriptional components were then identified, and significant transcription factors (SPIB, SMAD3, and SOX2) found. Conclusions: Protein-drug interaction analysis revealed PDE9A has interaction with drugs caffeine, γ-glutamyl glycine, and 3-isobutyl-1-methyl-7H-xanthine. Thus, we identified novel putative links between pathological processes in IS and AD at transcripts levels, and identified possible mechanistic and gene expression links between IS and AD.


Assuntos
Doença de Alzheimer/sangue , Biomarcadores/sangue , Isquemia Encefálica/sangue , 3',5'-AMP Cíclico Fosfodiesterases/análise , 3',5'-AMP Cíclico Fosfodiesterases/sangue , Doença de Alzheimer/complicações , Biomarcadores/análise , Isquemia Encefálica/complicações , Fosfatases de Especificidade Dupla/análise , Fosfatases de Especificidade Dupla/sangue , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/análise , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/sangue , Humanos , Glicoproteínas de Membrana/análise , Glicoproteínas de Membrana/sangue , Fosfatases da Proteína Quinase Ativada por Mitógeno/análise , Fosfatases da Proteína Quinase Ativada por Mitógeno/sangue , Glicoproteína Associada a Mielina/análise , Glicoproteína Associada a Mielina/sangue , Receptor trkB/análise , Receptor trkB/sangue , Transdução de Sinais/fisiologia , Acidente Vascular Cerebral/sangue , Acidente Vascular Cerebral/complicações , Proteínas de Transporte Vesicular/análise , Proteínas de Transporte Vesicular/sangue
5.
Bioconjug Chem ; 28(6): 1677-1683, 2017 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-28449575

RESUMO

The RAS-RAF-MEK-ERK pathway has been intensively studied in oncology, with RAS known to be mutated in ∼30% of all human cancers. The recent emergence of ERK1/2 inhibitors and their ongoing clinical investigation demands a better understanding of ERK1/2 behavior following small-molecule inhibition. Although fluorescent fusion proteins and fluorescent antibodies are well-established methods of visualizing proteins, we show that ERK1/2 can be visualized via a less-invasive approach based on a two-step process using inverse electron demand Diels-Alder cycloaddition. Our previously reported trans-cyclooctene-tagged covalent ERK1/2 inhibitor was used in a series of imaging experiments following a click reaction with a tetrazine-tagged fluorescent dye. Although limitations were encountered with this approach, endogenous ERK1/2 was successfully imaged in cells, and "on-target" staining was confirmed by over-expressing DUSP5, a nuclear ERK1/2 phosphatase that anchors ERK1/2 in the nucleus.


Assuntos
Sistema de Sinalização das MAP Quinases , Proteína Quinase 1 Ativada por Mitógeno/análise , Sondas Moleculares/química , Linhagem Celular , Reação de Cicloadição , Fosfatases de Especificidade Dupla/análise , Corantes Fluorescentes , Humanos , Inibidores de Proteínas Quinases
6.
Sci Rep ; 7: 42074, 2017 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-28181547

RESUMO

Medullary thyroid cancer (MTC) can be caused by germline mutations of the RET proto-oncogene or occurs as a sporadic form. It is well known that RET mutations affecting the cysteine-rich region of the protein (MEN2A-like mutations) are correlated with different phenotypes than those in the kinase domain (MEN2B-like mutations). Our aim was to analyse the whole-gene expression profile of MTC with regard to the type of RET gene mutation and the cancer genetic background (hereditary vs sporadic). We studied 86 MTC samples. We demonstrated that there were no distinct differences in the gene expression profiles of hereditary and sporadic MTCs. This suggests a homogeneous nature of MTC. We also noticed that the site of the RET gene mutation slightly influenced the gene expression profile of MTC. We found a significant association between the localization of RET mutations and the expression of three genes: NNAT (suggested to be a tumour suppressor gene), CDC14B (involved in cell cycle control) and NTRK3 (tyrosine receptor kinase that undergoes rearrangement in papillary thyroid cancer). This study suggests that these genes are significantly deregulated in tumours with MEN2A-like and MEN2B-like mutations; however, further investigations are necessary to demonstrate any clinical impact of these findings.


Assuntos
Carcinoma Neuroendócrino/genética , Receptor com Domínio Discoidina 2/análise , Fosfatases de Especificidade Dupla/análise , Perfilação da Expressão Gênica , Proteínas de Membrana/análise , Mutação , Proteínas do Tecido Nervoso/análise , Proteínas Proto-Oncogênicas c-ret/genética , Neoplasias da Glândula Tireoide/genética , Adulto , Idoso , Carcinoma Neuroendócrino/patologia , Receptor com Domínio Discoidina 2/genética , Fosfatases de Especificidade Dupla/genética , Feminino , Humanos , Masculino , Proteínas de Membrana/genética , Pessoa de Meia-Idade , Proteínas do Tecido Nervoso/genética , Proto-Oncogene Mas , Neoplasias da Glândula Tireoide/patologia
7.
Methods Mol Biol ; 1447: 197-215, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27514808

RESUMO

The spatiotemporal regulation of the Ras/ERK pathway is critical in determining the physiological and pathophysiological outcome of signaling. Dual-specificity mitogen-activated protein kinase (MAPK) phosphatases (DUSPs or MKPs) are key regulators of pathway activity and may also localize ERK to distinct subcellular locations. Here we present methods largely based on the use of high content microscopy to both visualize and quantitate the subcellular distribution of activated (p-ERK) and total ERK in populations of mouse embryonic fibroblasts derived from mice lacking DUSP5, a nuclear ERK-specific MKP. Such methods in combination with rescue experiments using adenoviral vectors encoding wild-type and mutant forms of DUSP5 have allowed us to visualize specific defects in ERK regulation in these cells thus confirming the role of this phosphatase as both a nuclear regulator of ERK activity and localization.


Assuntos
Fosfatases de Especificidade Dupla/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Microscopia de Fluorescência/métodos , Transdução de Sinais , Proteínas ras/metabolismo , Animais , Células Cultivadas , Fosfatases de Especificidade Dupla/análise , Fosfatases de Especificidade Dupla/genética , MAP Quinases Reguladas por Sinal Extracelular/análise , Fibroblastos/metabolismo , Fibroblastos/ultraestrutura , Imunofluorescência/métodos , Deleção de Genes , Células HEK293 , Humanos , Immunoblotting/métodos , Sistema de Sinalização das MAP Quinases , Camundongos , Proteínas ras/análise
8.
Biochim Biophys Acta ; 1862(6): 1074-83, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26976331

RESUMO

Lafora disease (LD, OMIM 254780) is a fatal rare disorder characterized by epilepsy and neurodegeneration. Although in recent years a lot of information has been gained on the molecular basis of the neurodegeneration that accompanies LD, the molecular basis of epilepsy is poorly understood. Here, we present evidence indicating that the homeostasis of glutamate transporter GLT-1 (EAAT2) is compromised in mouse models of LD. Our results indicate that primary astrocytes from LD mice have reduced capacity of glutamate transport, probably because they present a reduction in the levels of the glutamate transporter at the plasma membrane. On the other hand, the overexpression in cellular models of laforin and malin, the two proteins related to LD, results in an accumulation of GLT-1 (EAAT2) at the plasma membrane and in a severe reduction of the ubiquitination of the transporter. All these results suggest that the laforin/malin complex slows down the endocytic recycling of the GLT-1 (EAAT2) transporter. Since, defects in the function of this transporter lead to excitotoxicity and epilepsy, we suggest that the epilepsy that accompanies LD could be due, at least in part, to deficiencies in the function of the GLT-1 (EAAT2) transporter.


Assuntos
Astrócitos/metabolismo , Transportador 2 de Aminoácido Excitatório/metabolismo , Doença de Lafora/metabolismo , Animais , Astrócitos/patologia , Linhagem Celular , Células Cultivadas , Modelos Animais de Doenças , Fosfatases de Especificidade Dupla/análise , Fosfatases de Especificidade Dupla/metabolismo , Endocitose , Transportador 2 de Aminoácido Excitatório/análise , Homeostase , Humanos , Doença de Lafora/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Tirosina Fosfatases não Receptoras , Ubiquitinação
9.
Breast Cancer Res Treat ; 148(1): 211-20, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25281216

RESUMO

Triple-negative breast cancer (TNBC) is an aggressive cancer with limited treatment options. Dual specificity phosphatase 4 (DUSP4) has recently been suggested as a potential marker of chemotherapy resistance for TNBC. DUSP4 gene expression levels were measured in breast cancer tissue from 469 TNBC patients aged 20-75 years who participated in the Shanghai Breast Cancer Survival Study, and their association with recurrence/breast cancer mortality and total mortality was evaluated. Information on breast cancer diagnosis, treatment, and disease progression was collected via medical chart review and multiple in-person follow-up surveys. A Cox regression model was applied in the data analyses. Over a median follow-up of 5.3 years (range: 0.7-8.9 years), 100 deaths and 92 recurrences/breast cancer deaths were documented. Expression levels of transcript variant 1 (NM_001394) and transcript variant 2 (NM_057158) of the DUSP4 gene were studied and were highly correlated (r = 0.76). Low DUSP4 expression levels, particularly of variant 1, were associated with both increased recurrence/breast cancer mortality and increased overall mortality. Hazard ratios with adjustment for age at diagnosis and TNM stage associated with below versus above the median expression level were 1.97 (95 % confidence interval (CI): 1.27-3.05) for recurrence/breast cancer mortality and 2.09 (95 % CI: 1.38-3.17) for overall mortality. Additional adjustment for expression levels of MKI67 and TP53, common treatment types, breast cancer subtype, and grade did not materially alter the observed associations. Low DUSP4 expression levels predict recurrence and mortality in TNBC patients independently from known clinical and molecular predictors.


Assuntos
Biomarcadores Tumorais/genética , Fosfatases de Especificidade Dupla/biossíntese , Fosfatases da Proteína Quinase Ativada por Mitógeno/biossíntese , Recidiva Local de Neoplasia/genética , Neoplasias de Mama Triplo Negativas/genética , Adulto , Idoso , Fosfatases de Especificidade Dupla/análise , Fosfatases de Especificidade Dupla/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Pessoa de Meia-Idade , Fosfatases da Proteína Quinase Ativada por Mitógeno/análise , Fosfatases da Proteína Quinase Ativada por Mitógeno/genética , Recidiva Local de Neoplasia/patologia , Modelos de Riscos Proporcionais , Transcriptoma , Neoplasias de Mama Triplo Negativas/patologia , Adulto Jovem
10.
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
11.
J Cell Biol ; 201(7): 997-1012, 2013 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-23775190

RESUMO

Mitosis is controlled by a network of kinases and phosphatases. We screened a library of small interfering RNAs against a genome-wide set of phosphatases to comprehensively evaluate the role of human phosphatases in mitosis. We found four candidate spindle checkpoint phosphatases, including the tumor suppressor CDKN3. We show that CDKN3 is essential for normal mitosis and G1/S transition. We demonstrate that subcellular localization of CDKN3 changes throughout the cell cycle. We show that CDKN3 dephosphorylates threonine-161 of CDC2 during mitotic exit and we visualize CDC2(pThr-161) at kinetochores and centrosomes in early mitosis. We performed a phosphokinome-wide mass spectrometry screen to find effectors of the CDKN3-CDC2 signaling axis. We found that one of the identified downstream phosphotargets, CKß phosphorylated at serine 209, localizes to mitotic centrosomes and controls the spindle checkpoint. Finally, we show that CDKN3 protein is down-regulated in brain tumors. Our findings indicate that CDKN3 controls mitosis through the CDC2 signaling axis. These results have implications for targeted anticancer therapeutics.


Assuntos
Proteínas Inibidoras de Quinase Dependente de Ciclina/fisiologia , Fosfatases de Especificidade Dupla/fisiologia , Mitose/fisiologia , Proteína Quinase CDC2 , Centrossomo/metabolismo , Centrossomo/ultraestrutura , Ciclina B/metabolismo , Proteínas Inibidoras de Quinase Dependente de Ciclina/análise , Proteínas Inibidoras de Quinase Dependente de Ciclina/metabolismo , Quinases Ciclina-Dependentes , Fosfatases de Especificidade Dupla/análise , Fosfatases de Especificidade Dupla/metabolismo , Células HeLa , Humanos , Cinetocoros/metabolismo , Cinetocoros/ultraestrutura , Espectrometria de Massas , Mitose/genética , Fosforilação , Interferência de RNA , Transdução de Sinais
12.
Hum Mol Genet ; 21(7): 1521-33, 2012 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-22186026

RESUMO

Lafora disease (LD), a fatal neurodegenerative disorder characterized by the presence of intracellular inclusions called Lafora bodies (LBs), is caused by loss-of-function mutations in laforin or malin. Previous studies suggested a role of these proteins in the regulation of glycogen biosynthesis, in glycogen dephosphorylation and in the modulation of the intracellular proteolytic systems. However, the contribution of each of these processes to LD pathogenesis is unclear. We have generated a malin-deficient (Epm2b-/-) mouse with a phenotype similar to that of LD patients. By 3-6 months of age, Epm2b-/- mice present neurological and behavioral abnormalities that correlate with a massive presence of LBs in the cortex, hippocampus and cerebellum. Sixteen-day-old Epm2b-/- mice, without detectable LBs, show an impairment of macroautophagy (hereafter called autophagy), which remains compromised in adult animals. These data demonstrate similarities between the Epm2a-/- and Epm2b-/- mice that provide further insights into LD pathogenesis. They illustrate that the dysfunction of autophagy is a consequence of the lack of laforin-malin complexes and a common feature of both mouse models of LD. Because this dysfunction precedes other pathological manifestations, we propose that decreased autophagy plays a primary role in the formation of LBs and it is critical in LD pathogenesis.


Assuntos
Autofagia , Doença de Lafora/patologia , Ubiquitina-Proteína Ligases/genética , Animais , Comportamento Animal , Encéfalo/metabolismo , Encéfalo/patologia , Fosfatases de Especificidade Dupla/análise , Fosfatases de Especificidade Dupla/metabolismo , Glucanos/química , Doença de Lafora/genética , Doença de Lafora/fisiopatologia , Camundongos , Camundongos Knockout , Transtornos das Habilidades Motoras/genética , Miocárdio/ultraestrutura , Proteínas Tirosina Fosfatases não Receptoras , Ubiquitina/análise , Ubiquitina-Proteína Ligases/deficiência
13.
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
15.
Cell Cycle ; 8(23): 3904-13, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19923902

RESUMO

Following fertilization the transition from a highly differentiated oocyte to a totipotent 2-cell embryo requires two unique mitotic cell cycles. The first cell cycle is characterized by a prolonged G(1) phase, DNA replication (S phase) that occurs separately in the female and male pronuclei, and a short G(2) phase that occur in the absence of cell growth. During the second cell cycle, G(1) is short whereas G(2) is prolonged and occurs concurrently with zygotic genome activation, which is essential for progression past the 2-cell stage. CDC14B, a dual specificity phosphatase that counteracts cyclin dependent kinase 1 (CDK1/CDC2A) action, regulates mitosis in somatic cells and prevents premature meiotic resumption in mouse oocytes. It is not known if CDC14B plays a role during the unique mitotic cell cycles of preimplantation development. We report that CDC14B is present in mouse embryos and localizes to mitotic centrosomes and spindles. Overexpressing CDC14B in 1-cell embryos results in 40% and 60% of the embryos arresting at the 1- and 2-cell stages, respectively. Embryos arrested at the 1-cell stage contained reduced CDC2A activity, whereas embryos arrested at the 2-cell stage were in G(2) and failed to activate the zygotic genome. In contrast, overexpressing CDC14B in meiotically-incompetent oocytes, which are arrested in a G(2)-like state and are transcriptionally active, does not repress global transcription. These data suggest that CDC14B is a negative regulator of the 1-to-2-cell transition and of zygotic genome activation in mouse embryogenesis.


Assuntos
Fosfatases de Especificidade Dupla/metabolismo , Transferência Embrionária , Genoma , Mitose , Zigoto/citologia , Zigoto/enzimologia , Animais , Blastocisto , Proteína Quinase CDC2/metabolismo , Centrossomo , Fosfatases de Especificidade Dupla/análise , Feminino , Fase G1 , Fase G2 , Masculino , Camundongos , Oócitos/citologia , Fuso Acromático
16.
Acta Crystallogr D Biol Crystallogr ; 65(Pt 10): 1013-20, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19770498

RESUMO

Dual-specificity phosphatases (DUSPs) are enzymes that participate in the regulation of biological processes such as cell growth, differentiation, transcription and metabolism. A number of DUSPs are able to dephosphorylate phosphorylated serine, threonine and tyrosine residues on mitogen-activated protein kinases (MAPKs) and thus are also classified as MAPK phosphatases (MKPs). As an increasing number of DUSPs are being identified and characterized, there is a growing need to understand their biological activities at the molecular level. There is also significant interest in identifying DUSPs that could be potential targets for drugs that modulate MAPK-dependent signaling and immune responses, which have been implicated in a variety of maladies including cancer, infectious diseases and inflammatory disorders. Here, the overproduction, purification and crystal structure at 1.88 A resolution of human dual-specificity phosphatase 14, DUSP14 (MKP6), are reported. This structural information should accelerate the study of DUSP14 at the molecular level and may also accelerate the discovery and development of novel therapeutic agents.


Assuntos
Fosfatases de Especificidade Dupla/análise , Fosfatases da Proteína Quinase Ativada por Mitógeno/análise , Sequência de Aminoácidos , Domínio Catalítico , Clonagem Molecular , Cristalografia por Raios X , Fosfatases de Especificidade Dupla/genética , Fosfatases de Especificidade Dupla/isolamento & purificação , Humanos , Fosfatases da Proteína Quinase Ativada por Mitógeno/genética , Fosfatases da Proteína Quinase Ativada por Mitógeno/isolamento & purificação , Conformação Molecular , Dados de Sequência Molecular , Alinhamento de Sequência
17.
Cell Signal ; 21(12): 1794-805, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19666109

RESUMO

DUSP5 is an inducible, nuclear, dual-specificity phosphatase, which specifically interacts with and inactivates the ERK1/2 MAP kinases in mammalian cells. In addition, expression of DUSP5 causes nuclear translocation of ERK2 indicating that it may act as a nuclear anchor for the inactive kinase. Here we show that induction of DUSP5 mRNA and protein in response to growth factors is dependent on ERK1/2 activation and that the accumulation of DUSP5 protein is regulated by rapid proteasomal degradation. DUSP5 is phosphorylated by ERK1/2 both in vitro and in vivo on three sites (Thr321, Ser346 and Ser376) within its C-terminal domain. DUSP5 phosphorylation is absolutely dependent on the conserved kinase interaction motif (KIM) within the amino-terminal domain of DUSP5, indicating that the same protein-protein contacts are required for both the inactivation of ERK2 by DUSP5 and for DUSP5 to act as a substrate for this MAPK. Using a combination of pharmacological inhibitors and phospho-site mutants we can find no evidence that phosphorylation of DUSP5 by ERK2 significantly affects either the half-life of the DUSP5 protein or its ability to bind to, inactivate or anchor ERK2 in the nucleus. However, co-expression of ERK2 results in significant stabilisation of DUSP5, which is accompanied by reduced levels of DUSP5 ubiquitination. These changes are independent of ERK2 kinase activity but absolutely depend on the ability of ERK2 to bind to DUSP5. We conclude that DUSP5 is stabilised by complex formation with its physiological substrate and that this may reinforce its activity as both a phosphatase and nuclear anchor for ERK2.


Assuntos
Fosfatases de Especificidade Dupla/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Animais , Células COS , Chlorocebus aethiops , Fosfatases de Especificidade Dupla/análise , Fosfatases de Especificidade Dupla/genética , Regulação da Expressão Gênica , Sistema de Sinalização das MAP Quinases , Camundongos , Células NIH 3T3 , Estabilidade Proteica , RNA Mensageiro/genética
18.
Methods Enzymol ; 457: 275-87, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19426873

RESUMO

Dual specificity phosphatase (DSP) 18 and 21 are members of a poorly understood subfamily of protein tyrosine phosphatases (PTP) that are unique in their ability to dephosphorylate both phosphotyrosine and phosphoserine/threonine residues in vitro. Because of the difficulty in identifying substrate specificity, determining subcellular localization can help to resolve biological function of these phosphatases. DSP18 and DSP21 are targeted to mitochondria by internal localization signals. Surprisingly, DSP18 and DSP21 are both peripherally associated with the mitochondrial inner membrane, however, DSP18 is oriented toward the intermembrane space while DSP21 is facing the matrix compartment. This chapter describes methodology for purification of recombinant protein and demonstration of phosphatase activity, for mitochondrial purification and subfractionation of mitochondria to determine submitochondrial localization and for determining membrane orientation and strength of membrane association.


Assuntos
Fosfatases de Especificidade Dupla/análise , Fosfatases de Especificidade Dupla/genética , Membranas Mitocondriais/enzimologia , Animais , Fracionamento Celular , Fosfatases de Especificidade Dupla/isolamento & purificação , Fosfatases de Especificidade Dupla/metabolismo , Rim/enzimologia , Mitocôndrias/enzimologia , Mutagênese , Fosfotirosina/análogos & derivados , Fosfotirosina/metabolismo , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/análise , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Tripsina/metabolismo
19.
PLoS Biol ; 6(6): e140, 2008 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-18547142

RESUMO

Gene duplication was prevalent during hominoid evolution, yet little is known about the functional fate of new ape gene copies. We characterized the CDC14B cell cycle gene and the functional evolution of its hominoid-specific daughter gene, CDC14Bretro. We found that CDC14B encodes four different splice isoforms that show different subcellular localizations (nucleus or microtubule-associated) and functional properties. A microtubular CDC14B variant spawned CDC14Bretro through retroposition in the hominoid ancestor 18-25 million years ago (Mya). CDC14Bretro evolved brain-/testis-specific expression after the duplication event and experienced a short period of intense positive selection in the African ape ancestor 7-12 Mya. Using resurrected ancestral protein variants, we demonstrate that by virtue of amino acid substitutions in distinct protein regions during this time, the subcellular localization of CDC14Bretro progressively shifted from the association with microtubules (stabilizing them) to an association with the endoplasmic reticulum. CDC14Bretro evolution represents a paradigm example of rapid, selectively driven subcellular relocalization, thus revealing a novel mode for the emergence of new gene function.


Assuntos
Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Fosfatases de Especificidade Dupla/genética , Fosfatases de Especificidade Dupla/metabolismo , Evolução Molecular , Sequência de Aminoácidos , Animais , Encéfalo/metabolismo , Proteínas de Ciclo Celular/análise , Linhagem Celular , Fosfatases de Especificidade Dupla/análise , Duplicação Gênica , Genes Duplicados , Hominidae/fisiologia , Humanos , Dados de Sequência Molecular , Isoformas de Proteínas/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA