Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Toxicol Appl Pharmacol ; 487: 116954, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38705402

RESUMO

Dual-specificity phosphatase 26 (DUSP26) acts as a pivotal player in the transduction of signalling cascades with its dephosphorylating activity. Currently, DUSP26 attracts extensive attention due to its particular function in several pathological conditions. However, whether DUSP26 plays a role in kidney ischaemia-reperfusion (IR) injury is unknown. Aims of the current work were to explore the relevance of DUSP26 in kidney IR damage. DUSP26 levels were found to be decreased in renal tubular epithelial cells following hypoxia-reoxygenation (HR) and kidney samples subjected to IR treatments. DUSP26-overexpressed renal tubular epithelial cells exhibited protection against HR-caused apoptosis and inflammation, while DUSP26-depleted renal tubular epithelial cells were more sensitive to HR damage. Upregulation of DUSP26 in rat kidneys by infecting adenovirus expressing DUSP26 markedly ameliorated kidney injury caused by IR, while also effectively reducing apoptosis and inflammation. The mechanistic studies showed that the activation of transforming growth factor-ß-activated kinase 1 (TAK1)-JNK/p38 MAPK, contributing to kidney injury under HR or IR conditions, was restrained by increasing DUSP26 expression. Pharmacological restraint of TAK1 markedly diminished DUSP26-depletion-exacebated effects on JNK/p38 activation and HR injury of renal tubular cells. The work reported a renal-protective function of DUSP26, which protects against IR-related kidney damage via the intervention effects on the TAK1-JNK/p38 axis. The findings laid a foundation for understanding the molecular pathogenesis of kidney IR injury and provide a prospective target for treating this condition.


Assuntos
Apoptose , Células Epiteliais , Túbulos Renais , MAP Quinase Quinase Quinases , Ratos Sprague-Dawley , Traumatismo por Reperfusão , Proteínas Quinases p38 Ativadas por Mitógeno , Animais , Traumatismo por Reperfusão/patologia , MAP Quinase Quinase Quinases/metabolismo , MAP Quinase Quinase Quinases/genética , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Masculino , Túbulos Renais/patologia , Túbulos Renais/metabolismo , Ratos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Fosfatases de Especificidade Dupla/metabolismo , Fosfatases de Especificidade Dupla/genética , Linhagem Celular , Injúria Renal Aguda/patologia , Injúria Renal Aguda/metabolismo , Inflamação/patologia , Inflamação/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Fosfatases da Proteína Quinase Ativada por Mitógeno/metabolismo , Fosfatases da Proteína Quinase Ativada por Mitógeno/genética , Transdução de Sinais/fisiologia
2.
Int Immunopharmacol ; 117: 109980, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-37012870

RESUMO

Dual-specificity phosphatase 26 (DUSP26) is linked to a broad range of human disorders as it affects numerous signaling cascades. However, the involvement of DUSP26 in ischemic stroke has not been explored. Here, we investigated DUSP26 as a key mediator of oxygen-glucose deprivation/reoxygenation (OGD/R)-associated neuronal injury, an in vitro model for investigating ischemic stroke. A decline in DUSP26 occurred in neurons suffering from OGD/R. A deficiency in DUSP26 rendered neurons more susceptible to OGD/R by aggravating neuronal apoptosis and inflammation, while the overexpression of DUSP26 blocked OGD/R-evoked neuronal apoptosis and inflammation. Mechanistically, enhanced phosphorylation of transforming growth factor-ß-activated kinase 1 (TAK1), c-Jun N-terminal kinase (JNK) and P38 mitogen-activated protein kinase (MAPK) was evidenced in DUSP26-deficient neurons suffering from OGD/R, whereas the opposite effects were observed in DUSP26-overexpressed neurons. Moreover, the inhibition of TAK1 abolished the DUSP26-deficiency-elicited activation of JNK and P38 MAPK and exhibited anti-OGD/R injury effects in DUSP26-deficiency neurons. Results from these experiments show that DUSP26 is essential for neurons in defending against OGD/R insult, while neuroprotection is achieved by restraining the TAK1-mediated JNK/P38 MAPK pathway. Therefore, DUSP26 may serve as a therapeutic target for the management of ischemic stroke.


Assuntos
AVC Isquêmico , Proteína Quinase 14 Ativada por Mitógeno , Humanos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Oxigênio/metabolismo , Glucose/metabolismo , Apoptose , Proteína Quinase 14 Ativada por Mitógeno/metabolismo , Neurônios , AVC Isquêmico/metabolismo , Fosfatases de Especificidade Dupla/genética , Fosfatases de Especificidade Dupla/metabolismo
3.
Mol Biol Rep ; 49(11): 10195-10204, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36053282

RESUMO

BACKGROUND: Transforming Growth Factor ß (TGFß) proteins are potent inducers of the epithelial-mesenchymal transition (EMT) in tumor cells. Although mitogen-activated protein kinase (MAPK) family has been shown to be involved in TGFß-induced EMT, role of Dual Specificity Phosphatases (DUSP), key regulators of MAPK activity, in TGFß-induced EMT is largely unkonwn. METHODS AND RESULTS: Real-time qPCR analyses were performed to determine the effect of TGFß1 on expression of EMT genes and DUSP proteins in the non-small cell lung cancer model A549 and pancreatic adenocarcinoma model PANC1 cells. Western blot analyses were conducted to study the changes in protein levels of EMT proteins and select DUSP proteins, as well as phosphorylations of MAPK proteins upon TGFß1 stimulation. Small interfering RNA (siRNA) was utilized to reduce expressions of DUSP genes. We observed that the EMT phenotype coincided with increases in phosphorylations of the MAPK proteins ERK1/2, p38MAPK, and JNK upon TGFß1 stimulation. Real-time qPCR analysis showed increases in DUSP15 and DUSP26 mRNA levels and Western blot analysis confirmed the increase in DUSP26 protein levels in both A549 and PANC1 cells treated with TGFß1 relative to control. Silencing of DUSP26 expression by siRNA markedly suppressed the effect of TGFß1 on E-cadherin and mesenchymal genes in the cells. CONCLUSIONS: Data provided suggest that TGFß1 modulates the expression of DUSP genes and that upregulation of DUSP26 may be required for TGFß1-promoted EMT in A549 and PANC1 cells. Further studies should be carried out to elucidate the requirement of individual DUSPs in TGFß1-associated EMT in tumor cells.


Assuntos
Adenocarcinoma , Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Neoplasias Pancreáticas , Humanos , Fator de Crescimento Transformador beta1/farmacologia , Fator de Crescimento Transformador beta1/metabolismo , Transição Epitelial-Mesenquimal/genética , Regulação para Cima , RNA Interferente Pequeno/farmacologia , Neoplasias Pulmonares/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Fosfatases de Especificidade Dupla/genética , Fosfatases de Especificidade Dupla/metabolismo , Fosfatases de Especificidade Dupla/farmacologia , Células A549 , Linhagem Celular Tumoral
4.
Cell Mol Life Sci ; 79(4): 198, 2022 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-35313355

RESUMO

The dual specificity protein phosphatases (Dusps) control dephosphorylation of mitogen-activated protein kinases (MAPKs) as well as other substrates. Here, we report that Dusp26, which is highly expressed in neuroblastoma cells and primary neurons is targeted to the mitochondrial outer membrane via its NH2-terminal mitochondrial targeting sequence. Loss of Dusp26 has a significant impact on mitochondrial function that is associated with increased levels of reactive oxygen species (ROS), reduction in ATP generation, reduction in mitochondria motility and release of mitochondrial HtrA2 protease into the cytoplasm. The mitochondrial dysregulation in dusp26-deficient neuroblastoma cells leads to the inhibition of cell proliferation and cell death. In vivo, Dusp26 is highly expressed in neurons in different brain regions, including cortex and midbrain (MB). Ablation of Dusp26 in mouse model leads to dopaminergic (DA) neuronal cell loss in the substantia nigra par compacta (SNpc), inflammatory response in MB and striatum, and phenotypes that are normally associated with Neurodegenerative diseases. Consistent with the data from our mouse model, Dusp26 expressing cells are significantly reduced in the SNpc of Parkinson's Disease patients. The underlying mechanism of DA neuronal death is that loss of Dusp26 in neurons increases mitochondrial ROS and concurrent activation of MAPK/p38 signaling pathway and inflammatory response. Our results suggest that regulation of mitochondrial-associated protein phosphorylation is essential for the maintenance of mitochondrial homeostasis and dysregulation of this process may contribute to the initiation and development of neurodegenerative diseases.


Assuntos
Neurônios Dopaminérgicos/fisiologia , Fosfatases de Especificidade Dupla/fisiologia , Mitocôndrias/metabolismo , Fosfatases da Proteína Quinase Ativada por Mitógeno/fisiologia , Animais , Morte Celular/genética , Respiração Celular/genética , Células Cultivadas , Citoproteção/genética , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Knockout , Mitocôndrias/genética , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Estresse Oxidativo/genética , Doença de Parkinson/genética , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia
5.
Free Radic Biol Med ; 183: 106-124, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35278641

RESUMO

Non-small-cell lung cancer (NSCLC) is one of the most commonly diagnosed cancers worldwide with limited effective therapies. Cisplatin (DDP), as the first-line treatment, is always served as a mainstay of chemotherapeutic agents in combination with other drugs for NSCLC treatment. Nevertheless, DDP-based therapy is limited due to the frequent development of chemoresistance and adverse effects. Herein, it is necessary to find a more effective therapeutic approach with less toxicity. Falnidamol (FLD) is a pyrimido-pyrimidine compound and exerts anti-cancer activity. In the present study, we found that FLD could strongly promote the cytotoxicity of DDP and markedly reduce the IC50 values to restrain the proliferation of NSCLC cells. Furthermore, combination of FLD and DDP remarkably induced G2/M cell cycle arrest, DNA damage and mitochondrial apoptosis, which was largely through the induction of reactive oxygen species (ROS). Additionally, FLD/DDP in combination greatly triggered ferroptosis, along with free iron accumulation and enhanced lipid peroxidation. Epithelial to mesenchymal transition (EMT) and epidermal growth factor receptor (EGFR) phosphorylation were also considerably restrained in NSCLC cells co-treated with FLD/DDP. Mechanistically, the combinative treatment significantly reduced DUSP26 expression in NSCLC cells. More studies showed that DUSP26 was strongly up-regulated in human NSCLC samples compared with the paired normal tissues, and high DUSP26 predicted poor overall survival rate among patients. Importantly, we found that DUSP26 suppression intensively reduced the proliferation, EMT process and pEGFR expression in NSCLC cells, whereas facilitated ROS production, DNA damage and cell death; however, opposite phenotype was observed in NSCLC cells over-expressing DUSP26. More importantly, DUSP26 over-expression completely abolished the anti-cancer function of FLD/DDP in NSCLC cells. Animal studies finally confirmed that FLD/DDP in combination efficiently reduced tumor growth and lung metastasis in mice with ameliorated side effects. In conclusion, all these data illustrated that FLD and DDP combinational treatment effectively restrained NSCLC progression, and thus can be served as a promising therapeutic strategy.


Assuntos
Antineoplásicos , Carcinoma Pulmonar de Células não Pequenas , Fosfatases de Especificidade Dupla , Neoplasias Pulmonares , Fosfatases da Proteína Quinase Ativada por Mitógeno , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Proliferação de Células , Cisplatino/farmacologia , Cisplatino/uso terapêutico , Resistencia a Medicamentos Antineoplásicos/genética , Fosfatases de Especificidade Dupla/genética , Fosfatases de Especificidade Dupla/metabolismo , Transição Epitelial-Mesenquimal , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Camundongos , Fosfatases da Proteína Quinase Ativada por Mitógeno/genética , Fosfatases da Proteína Quinase Ativada por Mitógeno/metabolismo , Transdução de Sinais
6.
J Am Heart Assoc ; 10(4): e014311, 2021 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-33522247

RESUMO

Background Heart pathological hypertrophy has been recognized as a predisposing risk factor for heart failure and arrhythmia. DUSP (dual-specificity phosphatase) 26 is a member of the DUSP family of proteins, which has a significant effect on nonalcoholic fatty liver disease, neuroblastoma, glioma, and so on. However, the involvement of DUSP26 in cardiac hypertrophy remains unclear. Methods and Results Our study showed that DUSP26 expression was significantly increased in mouse hearts in response to pressure overload as well as in angiotensin II-treated cardiomyocytes. Cardiac-specific overexpression of DUSP26 mice showed attenuated cardiac hypertrophy and fibrosis, while deficiency of DUSP26 in mouse hearts resulted in increased cardiac hypertrophy and deteriorated cardiac function. Similar effects were also observed in cellular hypertrophy induced by angiotensin II. Importantly, we showed that DUSP26 bound to transforming growth factor-ß activated kinase 1 and inhibited transforming growth factor-ß activated kinase 1 phosphorylation, which led to suppression of the mitogen-activated protein kinase signaling pathway. In addition, transforming growth factor-ß activated kinase 1-specific inhibitor inhibited cardiomyocyte hypertrophy induced by angiotensin II and attenuated the exaggerated hypertrophic response in DUSP26 conditional knockout mice. Conclusions Taken together, DUSP26 was induced in cardiac hypertrophy and protected against pressure overload induced cardiac hypertrophy by modulating transforming growth factor-ß activated kinase 1-p38/ c-Jun N-terminal kinase-signaling axis. Therefore, DUSP26 may provide a therapeutic target for treatment of cardiac hypertrophy and heart failure.


Assuntos
Cardiomegalia/tratamento farmacológico , Fosfatases de Especificidade Dupla/farmacologia , Regulação da Expressão Gênica , MAP Quinase Quinase Quinases/genética , Fosfatases da Proteína Quinase Ativada por Mitógeno/farmacologia , Miócitos Cardíacos/metabolismo , Animais , Cardiomegalia/genética , Cardiomegalia/metabolismo , Modelos Animais de Doenças , Ecocardiografia , MAP Quinase Quinase Quinases/biossíntese , Camundongos , Camundongos Knockout , Miócitos Cardíacos/patologia , RNA/genética , Transdução de Sinais
7.
Biochem Biophys Res Commun ; 515(3): 410-416, 2019 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-31155289

RESUMO

Diabetic nephropathy (DN) is a leading cause of renal failure worldwide. Unfortunately, the pathogenetic mechanism of DN is far from to be understood. Dual-specificity phosphatase 26 (DUSP26) is a member of the Dusp protein family, and is suggested to be involved in divers biological and pathological processes, such as cell growth, differentiation, inflammation and apoptosis. However, its role in the development of DN is still vague. In this study, we found that DUSP26 expression was increased in kidney of DN patients. Then, the wild type (DUSP26+/+) and gene knockout (DUSP26-/-) mice were used to further explore the effects of DUSP26 on DN development induced by streptozotocin (STZ). DUSP26 deficiency accelerated renal injury and dysfunction, as evidenced by the elevated glomerulosclerosis, reduced expression of Nephrin and promoted glomerular basement membrane thickness. In addition, STZ treatment resulted in reactive oxygen species (ROS) accumulation, H2O2 overproduction and superoxide dismutase (SOD) reduction in renal cortex or glomeruli of mice. The ROS production caused the activation of mitogen-activated protein kinase (MAPKs) signaling in kidney glomeruli of STZ-induced mice. These in vivo pathological processes were further confirmed in the differentiated podocytes stimulated by glucose (GLU). Intriguingly, we found that STZ-induced DN as mentioned above was further accelerated by DUSP26-/- in mice following STZ injection. Moreover, STZ-induced fibrosis in kidney glomeruli of DN mice was markedly prolonged in DUSP26-knockout mice through potentiating transforming growth factor-ß1 (TGF-ß1) expression. More importantly, reducing ROS generation could significantly abolish DUSP26 knockdown-exacerbated TGF-ß1 expression and MAPKs activation, thereby protecting podocytes from GLU-induced podocyte injury. Thus, DUSP26-regulated DN development was largely dependent on ROS generation. Taken together, we concluded that DUSP26 might be a promising therapeutic target for developing effective treatments against DN progression.


Assuntos
Nefropatias Diabéticas/patologia , Fosfatases de Especificidade Dupla/metabolismo , Fosfatases da Proteína Quinase Ativada por Mitógeno/metabolismo , Estresse Oxidativo , Podócitos/metabolismo , Podócitos/patologia , Espécies Reativas de Oxigênio/metabolismo , Animais , Nefropatias Diabéticas/genética , Modelos Animais de Doenças , Regulação para Baixo/genética , Ativação Enzimática , Fibrose , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Podócitos/ultraestrutura
8.
Biomed Pharmacother ; 89: 376-385, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28249240

RESUMO

Dual-specificity phosphatase 26 (DUSP26) has recently emerged as a target for treatment of human cancers. However, only two small-molecule inhibitors of DUSP26 are known so far, namely NSC-87877 and ethyl-3, 4-dephostatin. DUSP26 contains an N-terminal region (residues 1-60) and a conserved C-terminal catalytic domain (residues 61-211, DUSP26-C). The crystal structure of DUSP26-C, showing a catalytically inactive conformation of the active site, was reported in a previous study. However, the detailed catalytic mechanism of DUSP26 cannot be described based on that structure. In this study, the 3D structure of DUSP26 (residues 42-211) adopting catalytically active conformation, was built by homology modeling, and the established 3D structure was validated using enzyme kinetic assays. Pharmacophore modeling based on the validated 3D structure of human DUSP26 was carried out. The established pharmacophore model was considered as a 3D query for retrieving novel DUSP26 inhibitors from the chemical databases "Diversity Libraries" (129,087 compounds). Next, a docking study was performed to refine the obtained hit compounds. Then a total of 100 compounds were selected based on the ranking order and visual examination, which were then evaluated by an enzyme-based assay. Eight compounds were found to have inhibitory activities against DUSP26, and the most potent compound was assigned No. F1063-0967 with an IC50 value of 11.62µM. The inhibitory activity of F1063-0967 against DUSP26 is higher than that of NCS87877 (IC50 value: 16.67±2.89µM), but lower than that of ethyl-3, 4-dephostatin (IC50 value: 6.8±0.41µM). MTT assay results revealed that F1063-0967 can induce apoptosis in IMR-32 cell line with an IC50 value of 4.13µM. These results suggest that F1063-0967 should be investigated further for other pharmacological properties.


Assuntos
Simulação por Computador , Avaliação Pré-Clínica de Medicamentos , Fosfatases de Especificidade Dupla/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Fosfatases da Proteína Quinase Ativada por Mitógeno/antagonistas & inibidores , Modelos Químicos , Simulação de Acoplamento Molecular/métodos , Sequência de Aminoácidos , Sítios de Ligação , Fosfatases de Especificidade Dupla/metabolismo , Inibidores Enzimáticos/química , Fosfatases da Proteína Quinase Ativada por Mitógeno/metabolismo , Modelos Moleculares , Conformação Proteica
9.
Acta Neuropathol Commun ; 4(1): 96, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27586084

RESUMO

The infiltrative behavior of diffuse gliomas severely reduces therapeutic potential of surgical resection and radiotherapy, and urges for the identification of new drug-targets affecting glioma growth and migration. To address the potential role of protein tyrosine phosphatases (PTPs), we performed mRNA expression profiling for 91 of the 109 known human PTP genes on a series of clinical diffuse glioma samples of different grades and compared our findings with in silico knowledge from REMBRANDT and TCGA databases. Overall PTP family expression levels appeared independent of characteristic genetic aberrations associated with lower grade or high grade gliomas. Notably, seven PTP genes (DUSP26, MTMR4, PTEN, PTPRM, PTPRN2, PTPRT and PTPRZ1) were differentially expressed between grade II-III gliomas and (grade IV) glioblastomas. For DUSP26, PTEN, PTPRM and PTPRT, lower expression levels correlated with poor prognosis, and overexpression of DUSP26 or PTPRT in E98 glioblastoma cells reduced tumorigenicity. Our study represents the first in-depth analysis of PTP family expression in diffuse glioma subtypes and warrants further investigations into PTP-dependent signaling events as new entry points for improved therapy.


Assuntos
Neoplasias Encefálicas/enzimologia , Glioma/enzimologia , Proteínas Tirosina Fosfatases/metabolismo , Neoplasias Encefálicas/patologia , Sistemas CRISPR-Cas , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Progressão da Doença , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Glioma/genética , Glioma/patologia , Células HEK293 , Humanos , Imuno-Histoquímica , Isocitrato Desidrogenase/genética , Isocitrato Desidrogenase/metabolismo , Estimativa de Kaplan-Meier , Gradação de Tumores , PTEN Fosfo-Hidrolase/deficiência , PTEN Fosfo-Hidrolase/genética , Proteínas Tirosina Fosfatases/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
10.
J Neurochem ; 137(5): 770-81, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26924229

RESUMO

Amyloid beta peptide (Aß) is a pathological hallmark of Alzheimer's disease (AD) and is generated through the sequential cleavage of amyloid precursor protein (APP) by ß- and γ-secretases. Hypoxia is a known risk factor for AD and stimulates Aß generation by γ-secretase; however, the underlying mechanisms remain unclear. In this study, we showed that dual-specificity phosphatase 26 (DUSP26) regulates Aß generation through changes in subcellular localization of the γ-secretase complex and its substrate C99 under hypoxic conditions. DUSP26 was identified as a novel γ-secretase regulator from a genome-wide functional screen using a cDNA expression library. The phosphatase activity of DUSP26 was required for the increase in Aß42 generation through γ-secretase, but this regulation did not affect the amount of the γ-secretase complex. Interestingly, DUSP26 induced the accumulation of C99 in the axons by stimulating anterograde transport of C99-positive vesicles. Additionally, DUSP26 induced c-Jun N-terminal kinase (JNK) activation for APP processing and axonal transport of C99. Under hypoxic conditions, DUSP26 expression levels were elevated together with JNK activation, and treatment with JNK inhibitor SP600125, or the DUSP26 inhibitor NSC-87877, reduced hypoxia-induced Aß generation by diminishing vesicle trafficking of C99 to the axons. Finally, we observed enhanced DUSP26 expression and JNK activation in the hippocampus of AD patients. Our results suggest that DUSP26 mediates hypoxia-induced Aß generation through JNK activation, revealing a new regulator of γ-secretase-mediated APP processing under hypoxic conditions. We propose the role of phosphatase dual-specificity phosphatase 26 (DUSP26) in the selective regulation of Aß42 production in neuronal cells under hypoxic stress. Induction of DUSP26 causes JNK-dependent shift in the subcellular localization of γ-secretase and C99 from the cell body to axons for Aß42 generation. These findings provide a new strategy for developing new therapeutic targets to arrest AD progression.


Assuntos
Peptídeos beta-Amiloides/biossíntese , Precursor de Proteína beta-Amiloide/metabolismo , Transporte Axonal/fisiologia , Fosfatases de Especificidade Dupla/biossíntese , Fosfatases de Especificidade Dupla/farmacologia , Fosfatases da Proteína Quinase Ativada por Mitógeno/biossíntese , Fosfatases da Proteína Quinase Ativada por Mitógeno/farmacologia , Fragmentos de Peptídeos/biossíntese , Doença de Alzheimer/metabolismo , Transporte Axonal/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Hipóxia Celular/efeitos dos fármacos , Hipóxia Celular/fisiologia , Linhagem Celular Tumoral , Células HEK293 , Humanos , Técnicas de Cultura de Órgãos
11.
FEBS J ; 282(10): 1871-90, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25757426

RESUMO

Vaccinia H1-related (VHR) phosphatase, also known as dual-specificity phosphatase (DUSP) 3, is a small member of the DUSP (also called DSP) family of phosphatases. VHR has a preference for phospho-tyrosine substrates, and has important roles in cellular signaling ranging from cell-cycle regulation and the DNA damage response to MAPK signaling, platelet activation and angiogenesis. VHR/DUSP3 has been implicated in several human cancers, where its tumor-suppressing and -promoting properties have been described. We give a detailed overview of VHR/DUSP3 phosphatase and compare it with its most closely related phosphatases DUSP13B, DUSP26 and DUSP27.


Assuntos
Fosfatase 3 de Especificidade Dupla/metabolismo , Animais , Humanos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Neoplasias/enzimologia , Neoplasias/metabolismo , Transdução de Sinais/fisiologia
12.
Acta Crystallogr D Biol Crystallogr ; 69(Pt 6): 1160-70, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23695260

RESUMO

Dual-specificity phosphatases (DUSPs) play an important role in regulating cellular signalling pathways governing cell growth, differentiation and apoptosis. Human DUSP26 inhibits the apoptosis of cancer cells by dephosphorylating substrates such as p38 and p53. High-resolution crystal structures of the DUSP26 catalytic domain (DUSP26-C) and its C152S mutant [DUSP26-C (C152S)] have been determined at 1.67 and 2.20 Å resolution, respectively. The structure of DUSP26-C showed a novel type of domain-swapped dimer formed by extensive crossover of the C-terminal α7 helix. Taken together with the results of a phosphatase-activity assay, structural comparison with other DUSPs revealed that DUSP26-C adopts a catalytically inactive conformation of the protein tyrosine phosphate-binding loop which significantly deviates from that of canonical DUSP structures. In particular, a noticeable difference exists between DUSP26-C and the active forms of other DUSPs at the hinge region of a swapped C-terminal domain. Additionally, two significant gaps were identified between the catalytic core and its surrounding loops in DUSP26-C, which can be exploited as additional binding sites for allosteric enzyme regulation. The high-resolution structure of DUSP26-C may thus provide structural insights into the rational design of DUSP26-targeted anticancer drugs.


Assuntos
Fosfatases de Especificidade Dupla/química , Fosfatases da Proteína Quinase Ativada por Mitógeno/química , Proteínas Mutantes/química , Calorimetria , Domínio Catalítico , Fosfatases de Especificidade Dupla/genética , Fosfatases de Especificidade Dupla/metabolismo , Humanos , Fosfatases da Proteína Quinase Ativada por Mitógeno/genética , Fosfatases da Proteína Quinase Ativada por Mitógeno/metabolismo , Mutagênese , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Difração de Raios X
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA