Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
1.
Molecules ; 27(17)2022 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-36080217

RESUMO

BACKGROUND: The dysregulation of melanin production causes skin-disfiguring ultraviolet (UV)-associated hyperpigmented spots. Previously, we found that the activation of c-Jun N-terminal kinase (JNK), a mitogen-activated protein kinase (MAPK), inhibited melanogenesis. METHODS: We selected BCI-215 as it may modify MAPK expression via a known function of a dual-specificity phosphatase (DUSP) 1/6 inhibitor. B16F10 melanoma cells, Mel-ab cells, human melanocytes, and a coculture were used to assess the anti-melanogenic activity of BCI-215. The molecular mechanisms were deciphered by assaying the melanin content and cellular tyrosinase activity via immunoblotting and RT-PCR. RESULTS: BCI-215 was found to suppress basal and cAMP-stimulated melanin production and cellular tyrosinase activity in vitro through the downregulation of microphthalmia-associated transcription factor (MITF) protein and its downstream enzymes. The reduction in MITF expression caused by BCI-215 was found to be due to all three types of MAPK activation, including extracellular signal-regulated kinase (ERK), JNK, and p38. The degree of activation was greater in ERK. A phosphorylation of the ß-catenin pathway was also demonstrated. The melanin index, expression of MITF, and downstream enzymes were well-reduced in UVB-irradiated ex vivo human skin by BCI-215. CONCLUSIONS: As BCI-215 potently inhibits UV-stimulated melanogenesis, small molecules of DUSP-related signaling modulators may provide therapeutic benefits against pigmentation disorders.


Assuntos
Interfaces Cérebro-Computador , Fosfatases de Especificidade Dupla , Hiperpigmentação , Linhagem Celular Tumoral , Fosfatases de Especificidade Dupla/antagonistas & inibidores , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Hiperpigmentação/metabolismo , Melaninas , Melanócitos/metabolismo , Monofenol Mono-Oxigenase , Pigmentação
2.
Theranostics ; 11(10): 4790-4808, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33754028

RESUMO

Rationale: Doxorubicin is a widely used anticancer drug. However, its major side effect, cardiotoxicity, results from cardiomyocyte loss that causes left ventricle (LV) wall thinning, chronic LV dysfunction and heart failure. Cardiomyocyte number expansion by thyroid hormone (T3) during preadolescence is suppressed by the developmental induction of an ERK1/2-specific dual specificity phosphatase 5 (DUSP5). Here, we sought to determine if a brief course of combined DUSP5 suppression plus T3 therapy replaces cardiomyocytes lost due to preexisting doxorubicin injury and reverses heart failure. Methods: We used in vivo-jetPEI to deliver DUSP5 or scrambled siRNA to ~5-week-old C57BL6 mice followed by 5 daily injections of T3 (2 ng/µg body weight). Genetic lineage tracing using Myh6-MerCreMer::Rosa26fs-Confetti mice and direct cardiomyocyte number counting, along with cell cycle inhibition (danusertib), was used to test if this treatment leads to de novo cardiomyocyte generation and improves LV contractile function. Three doses of doxorubicin (20 µg/g) given at 2-weekly intervals, starting at 5-weeks of age in C57BL6 mice, caused severe heart failure, as evident by a decrease in LV ejection fraction. Mice with an ~40 percentage point decrease in LVEF post-doxorubicin injury were randomized to receive either DUSP5 siRNA plus T3, or scrambled siRNA plus vehicle for T3. Age-matched mice without doxorubicin injury served as controls. Results: In uninjured adult mice, transient therapy with DUSP5 siRNA and T3 increases cardiomyocyte numbers, which is required for the associated increase in LV contractile function, since both are blocked by danusertib. In mice with chronic doxorubicin injury, DUSP5 siRNA plus T3 therapy rebuilds LV muscle by increasing cardiomyocyte numbers, which reverses LV dysfunction and prevents progressive chamber dilatation. Conclusion: RNA therapies are showing great potential. Importantly, a GMP compliant in vivo-jetPEI system for delivery of siRNA is already in use in humans, as is T3. Given these considerations, our findings provide a potentially highly translatable strategy for addressing doxorubicin cardiomyopathy, a currently untreatable condition.


Assuntos
Fosfatases de Especificidade Dupla/genética , Contração Miocárdica/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Tri-Iodotironina/farmacologia , Função Ventricular Esquerda/efeitos dos fármacos , Animais , Antibióticos Antineoplásicos/toxicidade , Benzamidas/farmacologia , Cardiotoxicidade/etiologia , Contagem de Células , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Doxorrubicina/toxicidade , Fosfatases de Especificidade Dupla/antagonistas & inibidores , Fator de Crescimento Insulin-Like I/metabolismo , Camundongos , Contração Miocárdica/genética , Miócitos Cardíacos/citologia , Inibidores de Proteínas Quinases/farmacologia , Pirazóis/farmacologia , RNA Interferente Pequeno , Disfunção Ventricular Esquerda/induzido quimicamente , Função Ventricular Esquerda/genética , Remodelação Ventricular/efeitos dos fármacos , Remodelação Ventricular/genética
3.
Vet Res ; 52(1): 7, 2021 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-33431056

RESUMO

Elucidating virus-cell interactions is fundamental to understanding viral replication and identifying targets for therapeutic control of viral infection. The extracellular signal-regulated kinase (ERK) pathway has been shown to regulate pathogenesis during many viral infections, but its role during coronavirus infection is undetermined. Infectious bronchitis virus is the representative strain of Gammacoronavirus, which causes acute and highly contagious diseases in the poultry farm. In this study, we investigated the role of ERK1/2 signaling pathway in IBV infection. We found that IBV infection activated ERK1/2 signaling and the up-regulation of phosphatase DUSP6 formed a negative regulation loop. Pharmacological inhibition of MEK1/2-ERK1/2 signaling suppressed the expression of DUSP6, promoted cell death, and restricted virus replication. In contrast, suppression of DUSP6 by chemical inhibitor or siRNA increased the phosphorylation of ERK1/2, protected cells from apoptosis, and facilitated IBV replication. Overexpression of DUSP6 decreased the level of phospho-ERK1/2, promoted apoptosis, while dominant negative mutant DUSP6-DN lost the regulation function on ERK1/2 signaling and apoptosis. In conclusion, these data suggest that MEK-ERK1/2 signaling pathway facilitates IBV infection, probably by promoting cell survival; meanwhile, induction of DUSP6 forms a negative regulation loop to restrict ERK1/2 signaling, correlated with increased apoptosis and reduced viral load. Consequently, components of the ERK pathway, such as MEK1/2 and DUSP6, represent excellent targets for the development of antiviral drugs.


Assuntos
Apoptose/fisiologia , Fosfatases de Especificidade Dupla/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Vírus da Bronquite Infecciosa/fisiologia , Fosfatases da Proteína Quinase Ativada por Mitógeno/metabolismo , Animais , Butadienos/farmacologia , Linhagem Celular , Galinhas , Chlorocebus aethiops , Fosfatases de Especificidade Dupla/antagonistas & inibidores , Fosfatases de Especificidade Dupla/genética , MAP Quinases Reguladas por Sinal Extracelular/genética , Fosfatases da Proteína Quinase Ativada por Mitógeno/antagonistas & inibidores , Fosfatases da Proteína Quinase Ativada por Mitógeno/genética , Nitrilas/farmacologia , Regulação para Cima , Replicação Viral
4.
Mol Neurobiol ; 58(1): 391-407, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32959171

RESUMO

In peripheral neuropathies, axonal degeneration (AxD) impairs the prognosis for recovery. Here, we describe a role for dual specificity phosphatases (DUSPs; MAP kinase phosphatases, MKPs), in supporting autonomous axon plasticity and viability. Both DUSPs 1 and 4 were identified within intact or axotomized sensory neurons. Knockdown of DUSP 1 or 4 independently or combined impaired neurite outgrowth in adult dissociated sensory neurons. Furthermore, adult sensory neurons with DUSP knockdown were rendered sensitive to axonopathy in vitro following exposure to low, subtoxic TrpV1 (transient receptor potential cation channel subfamily V member 1) activation by capsaicin, an intervention normally supportive of growth. This was not prevented by concurrent DLK (dual leucine zipper kinase) knockdown. Ex vivo neurofilament dissolution was heightened by DUSP inhibition within explanted nerves. In vivo DUSP knockdown or inhibition was associated with more rapid loss of motor axon excitability. The addition of SARM1 (sterile alpha and TIR motif containing 1) siRNA abrogated DUSP1 and 4 mediated loss of excitability. DUSP knockdown accelerated neurofilament breakdown and there was earlier morphological evidence of myelinated axon degeneration distal to axotomy. Taken together, the findings identify a key role for DUSPs in supporting axon plasticity and survival.


Assuntos
Axônios/enzimologia , Axônios/patologia , Fosfatases de Especificidade Dupla/metabolismo , Plasticidade Neuronal , Animais , Axotomia , Sobrevivência Celular , Fosfatases de Especificidade Dupla/antagonistas & inibidores , Gânglios Espinais/metabolismo , Filamentos Intermediários/metabolismo , MAP Quinase Quinase Quinases/metabolismo , Masculino , Degeneração Neural/patologia , Crescimento Neuronal , Neuroproteção , Ratos Sprague-Dawley , Células Receptoras Sensoriais/metabolismo
5.
Pigment Cell Melanoma Res ; 34(1): 122-131, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32767816

RESUMO

Targeting the MAPK signaling pathway has transformed the treatment of metastatic melanoma. CRISPR-Cas9 genetic screens provide a genome-wide approach to uncover novel genetic dependencies that might serve as therapeutic targets. Here, we analyzed recently reported CRISPR-Cas9 screens comparing data from 28 melanoma cell lines and 313 cell lines of other tumor types in order to identify fitness genes related to melanoma. We found an average of 1,494 fitness genes in each melanoma cell line. We identified 33 genes, inactivation of which specifically reduced the fitness of melanoma. This set of tumor type-specific genes includes established melanoma fitness genes as well as many genes that have not previously been associated with melanoma growth. Several genes encode proteins that can be targeted using available inhibitors. We verified that genetic inactivation of DUSP4 and PPP2R2A reduces the proliferation of melanoma cells. DUSP4 encodes an inhibitor of ERK, suggesting that further activation of MAPK signaling activity through its loss is selectively deleterious to melanoma cells. Collectively, these data present a resource of genetic dependencies in melanoma that may be explored as potential therapeutic targets.


Assuntos
Sistemas CRISPR-Cas , Fosfatases de Especificidade Dupla/antagonistas & inibidores , Técnicas de Inativação de Genes/métodos , Genoma Humano , Melanoma/patologia , Fosfatases da Proteína Quinase Ativada por Mitógeno/antagonistas & inibidores , Proteína Fosfatase 2/antagonistas & inibidores , Proliferação de Células , Fosfatases de Especificidade Dupla/genética , Fosfatases de Especificidade Dupla/metabolismo , Humanos , Melanoma/genética , Melanoma/metabolismo , Fosfatases da Proteína Quinase Ativada por Mitógeno/genética , Fosfatases da Proteína Quinase Ativada por Mitógeno/metabolismo , Proteína Fosfatase 2/genética , Proteína Fosfatase 2/metabolismo , Células Tumorais Cultivadas
6.
Sci Signal ; 13(646)2020 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-32843541

RESUMO

The mitogen-activated protein kinase (MAPK) phosphatases (MKPs) have been considered "undruggable," but their position as regulators of the MAPKs makes them promising therapeutic targets. MKP5 has been suggested as a potential target for the treatment of dystrophic muscle disease. Here, we identified an inhibitor of MKP5 using a p38α MAPK-derived, phosphopeptide-based small-molecule screen. We solved the structure of MKP5 in complex with this inhibitor, which revealed a previously undescribed allosteric binding pocket. Binding of the inhibitor to this pocket collapsed the MKP5 active site and was predicted to limit MAPK binding. Treatment with the inhibitor recapitulated the phenotype of MKP5 deficiency, resulting in activation of p38 MAPK and JNK. We demonstrated that MKP5 was required for TGF-ß1 signaling in muscle and that the inhibitor blocked TGF-ß1-mediated Smad2 phosphorylation. TGF-ß1 pathway antagonism has been proposed for the treatment of dystrophic muscle disease. Thus, allosteric inhibition of MKP5 represents a therapeutic strategy against dystrophic muscle disease.


Assuntos
Fosfatases de Especificidade Dupla/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Ensaios de Triagem em Larga Escala/métodos , Fosfatases da Proteína Quinase Ativada por Mitógeno/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/farmacologia , Sítio Alostérico/genética , Sequência de Aminoácidos , Animais , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Fosfatases de Especificidade Dupla/química , Fosfatases de Especificidade Dupla/metabolismo , Inibidores Enzimáticos/química , Inibidores Enzimáticos/metabolismo , Feminino , Humanos , Cinética , Camundongos , Camundongos Knockout , Fosfatases da Proteína Quinase Ativada por Mitógeno/química , Fosfatases da Proteína Quinase Ativada por Mitógeno/metabolismo , Mioblastos/citologia , Mioblastos/efeitos dos fármacos , Mioblastos/metabolismo , Ligação Proteica/efeitos dos fármacos , Homologia de Sequência de Aminoácidos , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/metabolismo
7.
Nat Commun ; 11(1): 1268, 2020 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-32152317

RESUMO

Regulation of mitosis secures cellular integrity and its failure critically contributes to the development, maintenance, and treatment resistance of cancer. In yeast, the dual phosphatase Cdc14 controls mitotic progression by antagonizing Cdk1-mediated protein phosphorylation. By contrast, specific mitotic functions of the mammalian Cdc14 orthologue CDC14B have remained largely elusive. Here, we find that CDC14B antagonizes CDK1-mediated activating mitotic phosphorylation of the deubiquitinase USP9X at serine residue 2563, which we show to be essential for USP9X to mediate mitotic survival. Starting from an unbiased proteome-wide screening approach, we specify Wilms' tumor protein 1 (WT1) as the relevant substrate that becomes deubiquitylated and stabilized by serine 2563-phosphorylated USP9X in mitosis. We further demonstrate that WT1 functions as a mitotic transcription factor and specify CXCL8/IL-8 as a target gene of WT1 that conveys mitotic survival. Together, we describe a ubiquitin-dependent signaling pathway that directs a mitosis-specific transcription program to regulate mitotic survival.


Assuntos
Proteína Quinase CDC2/antagonistas & inibidores , Fosfatases de Especificidade Dupla/antagonistas & inibidores , Mitose/fisiologia , Ubiquitina Tiolesterase/efeitos dos fármacos , Ubiquitina Tiolesterase/metabolismo , Proteínas WT1/metabolismo , Células A549 , Apoptose , Técnicas de Silenciamento de Genes , Células HEK293 , Células HeLa , Humanos , Interleucina-8/metabolismo , Fosforilação , Fatores de Transcrição , Ubiquitina Tiolesterase/genética , Proteínas WT1/genética
8.
Eur Rev Med Pharmacol Sci ; 24(3): 1116-1125, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-32096171

RESUMO

OBJECTIVE: The purpose of this study was to explore the possible role of ROR1-AS1 in the pathogenesis of colon cancer and the underlying mechanism. PATIENTS AND METHODS: The expression levels of ROR1-AS1 in 75 colon cancer tissue samples and adjacent ones, as well as in cell lines were examined by quantitative Polymerase Chain Reaction (qPCR). Then, ROR1-AS1 overexpression plasmid and siRNA were transfected into colon cancer cells using liposome method. After that, Cell Counting Kit-8 (CCK-8) and plate colony formation assays were conducted to analyze cell proliferation, while flow cytometry was applied for the analysis of cell cycle and apoptosis. At last, the mechanism of action of ROR1-AS1 was further explored by nuclear separation, RNA binding protein immunoprecipitation (RIP) and chromatin immunoprecipitation (CHIP) assays. RESULTS: ROR1-AS1 level in colon cancer tissues was remarkably higher than that in normal tissues, and the expression in tumors of stage III and IV was remarkably higher than those of stage I and II. Meanwhile, tumors with diameters more than 5 cm had a higher ROR1-AS1 expression than those less than 5 cm. After transfection with ROR1-AS1 overexpression plasmid, the cell proliferation ability was enhanced, the G0/G1 phase time of cell cycle was shortened, and the apoptosis was suppressed. However, the opposite result was observed after ROR1-AS1 was downregulated. Furthermore, RIP showed that ROR1-AS1 can bind to enhancer of zeste homolog 2 (EZH2) and inhibit the expression of DUSP5, and thus be engaged in the proliferation and apoptosis of colon cancer cells. CONCLUSIONS: ROR1-AS1 is highly expressed either in colon cancer tissues or in cell lines, which is able to enhance cell proliferation, accelerate cell cycle, and inhibit cell apoptosis. The mechanism of ROR1-AS1 to participate in the development of colon cancer may be the downregulation of DUSP5 via combination with EZH2.


Assuntos
Proliferação de Células/fisiologia , Neoplasias do Colo/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Fosfatases de Especificidade Dupla/metabolismo , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/biossíntese , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Inibidor de Quinase Dependente de Ciclina p21/antagonistas & inibidores , Inibidor de Quinase Dependente de Ciclina p21/genética , Fosfatases de Especificidade Dupla/antagonistas & inibidores , Fosfatases de Especificidade Dupla/genética , Regulação Neoplásica da Expressão Gênica , Células HCT116 , Humanos , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/genética
9.
Neurol Res ; 42(1): 31-38, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31813339

RESUMO

Objective: Spinal cord injury (SCI) is a common injury that seriously threatens human health. NF-κB may be involved in the secondary injury of SCI that is mediated by inflammation and aggravates damage. Our study was aimed to investigate the role of NF-κB signaling in DUSP19-mediated cleaved Caspase-3 expression and the release of inflammatory factors in vivo and in vitro.Materials and Methods: DUSP19 mRNA expression and the content of IL-6 and IL-8 in patients with traumatic SCI (TSCI) were measured by real-time PCR and ELISA, respectively. The levels of p-NF-κBp65, NF-κBp65 and cleaved Caspase-3 expression and the concentrations of IL-6 and IL-8 were measured by western blotting and ELISA, respectively.Results: Patients with TSCI showed lower DUSP19 expression and higher concentration of IL-6 and IL-8 compared with healthy controls. DUSP19 overexpression inhibited p-NF-κBp65 level, cleaved Caspase-3 expression, and production of IL-8 and IL-6 in the mice induced by TSCI. DUSP19 silencing increased p-NF-κBp65 level, cleaved Caspase-3 expression, and concentration of IL-6 and IL-8 in mouse primary microglia cells. DUSP19 overexpression had an inverse effect. Importantly, DUSP19 silencing and overexpression mediated p-NF-κBp65 level, cleaved Caspase-3 expression, and concentration of IL-6 and IL-8 in mouse primary microglia cells were reversed by NF-κB inhibitor pyrrolidine dithiocarbamate (PDTC) and NF-κB activator 12-myristate 13-acetate (PMA), respectively.Conclusion: These results suggested that DUSP19-mediated SCI-induced apoptosis and inflammation via NF-κB signaling and might therefore serve as a potential therapeutic target for SCI.


Assuntos
Apoptose/fisiologia , Fosfatases de Especificidade Dupla/biossíntese , Microglia/metabolismo , NF-kappa B/metabolismo , Traumatismos da Medula Espinal/metabolismo , Animais , Células Cultivadas , Fosfatases de Especificidade Dupla/antagonistas & inibidores , Fosfatases de Especificidade Dupla/genética , Humanos , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microglia/patologia , Estudos Prospectivos , Distribuição Aleatória , Transdução de Sinais/fisiologia , Traumatismos da Medula Espinal/genética , Traumatismos da Medula Espinal/patologia
10.
Phys Chem Chem Phys ; 21(42): 23501-23513, 2019 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-31617551

RESUMO

YIV-906 (formally PHY906, KD018) is a four-herb formulation that is currently being developed to improve the therapeutic index and ameliorate the side effects of many chemotherapeutic drugs including sorafenib, irinotecan, and capecitabine. However, as a promising anti-cancer adjuvant, the molecular mechanism of action of YIV-906 remains unrevealed due to its multi-component and multi-target features. Since YIV-906 has been shown to induce apoptosis and autophagy in cancer cells through modulating the negative regulators of ERK1/2, namely DUSPs, it is of great interest to elucidate the key components that cause the therapeutic effect of YIV-906. In this work, we investigated the mechanism of YIV-906 inhibiting DUSPs, using a broad spectrum of molecular modelling techniques, including molecular docking, molecular dynamics (MD) simulations, and binding free energy calculations. In total, MD simulations and binding free energy calculations were performed for 99 DUSP-ligand complexes. We found that some herbal components or their metabolites could inhibit DUSPs. Based on the docking scores and binding free energies, the sulfation and glucuronidation metabolites of the S ingredient in YIV-906 play a leading role in inhibiting DUSPs, although several original herbal chemicals with carboxyl groups from the P and Z ingredients also make contributions to this inhibitory effect. It is not a surprise that the electrostatic interaction plays the dominant role in the ligand binding process, given the fact that several charged residues reside in the binding pockets of DUSPs. Our MD simulation results demonstrate that the sulfate moieties and carboxyl moieties of the advantageous ligands from YIV-906 can occupy the enzymes' catalytic sites, mimicking the endogenous phosphate substrates of DUSPs. As such, the ligand binding can inhibit the association of DUSPs and ERK1/2, which in turn reduces the dephosphorylation of ERK1/2 and causes cell cycle arrest in the tumor. Our modelling study provides useful insights into the rational design of highly potent anti-cancer drugs targeting DUSPs. Finally, we have demonstrated that multi-scale molecular modelling techniques are able to elucidate molecular mechanisms involving complex molecular systems.


Assuntos
Antineoplásicos Fitogênicos/química , Medicamentos de Ervas Chinesas/química , Antineoplásicos Fitogênicos/metabolismo , Sítios de Ligação , Domínio Catalítico , Medicamentos de Ervas Chinesas/metabolismo , Fosfatases de Especificidade Dupla/antagonistas & inibidores , Fosfatases de Especificidade Dupla/metabolismo , Humanos , Ligantes , Proteína Quinase 3 Ativada por Mitógeno/química , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Termodinâmica
11.
Aging Cell ; 18(6): e13022, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31400088

RESUMO

Intracerebral haemorrhage (ICH) is a severe neurological disorder caused by bleeding within the brain tissue. Inflammation has been implicated in ICH pathogenesis and is a potential therapeutic target for ICH. Haemin, an activator of haem oxygenase-1 (HO-1), rapidly increases HO-1 protein expression and activity and has been shown to distinctly affect anti-inflammatory functions after central nervous system (CNS) injury. However, less is known about the mechanisms that underlie the anti-inflammatory effects of haemin in aged rats post-ICH. Here, we performed microarray analysis to identify miRNAs that respond strongly to HO-1 regulation in ICH rats and found that miR-21-5p induced the most significant change. Using Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment and Gene Ontology (GO) analysis, we focused on dual-specificity phosphatase 8 (DUSP8) from the predicted miR-21-5p targets. Luciferase reporter assays confirmed that miR-21-5p bound directly to DUSP8. MiR-21-5p upregulation in vitro downregulated DUSP8 expression. Importantly, intracerebroventricularly injecting antagomir for miR-21-5p (A-miR-21-5p), which was used to inhibit miR-21-5p in aged ICH rats, significantly reduced the neurological defects, repaired cognitive impairment, alleviated blood-brain barrier (BBB) permeability, inhibited neuronal apoptosis posthaemorrhage and accelerated haematoma absorption. In addition, serum miR-21-5p levels were notably elevated in patients relative to healthy individuals and were correlated with National Institutes of Health Stroke Scale (NIHSS) scores and clinical outcomes. In summary, A-miR-21-5p increased HO-1 expression in cerebral haematomas, thus eliciting the DUSP8-modulated perifocal neuroprotective effect of haemin. MiR-21-5p with haemin therapy may be a potential therapy post-ICH.


Assuntos
Anti-Inflamatórios/farmacologia , Hemorragia Cerebral/metabolismo , Fosfatases de Especificidade Dupla/antagonistas & inibidores , Heme Oxigenase-1/antagonistas & inibidores , Hemina/farmacologia , MicroRNAs/metabolismo , Envelhecimento/efeitos dos fármacos , Animais , Antagomirs/farmacologia , Células Cultivadas , Hemorragia Cerebral/tratamento farmacológico , Fosfatases de Especificidade Dupla/metabolismo , Células HEK293 , Heme Oxigenase-1/metabolismo , Humanos , Masculino , MicroRNAs/antagonistas & inibidores , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos
12.
Comput Biol Chem ; 80: 138-146, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30952040

RESUMO

BACKGROUND: Breast cancer remains the most lethal type of cancer for women. A significant proportion of breast cancer cases are characterised by overexpression of the human epidermal growth factor receptor 2 protein (HER2). These cancers are commonly treated by Herceptin (Trastuzumab), but resistance to drug treatment frequently develops in tumour cells. Dual-specificity phosphatases (DUSPs) are thought to play a role in the mechanism of resistance, since some of them were reported to be overexpressed in tumours resistant to Herceptin. RESULTS: We used a systems biology approach to investigate how DUSP overexpression could favour cell proliferation and to predict how this mechanism could be reversed by targeted inhibition of selected DUSPs. We measured the expression of 20 DUSP genes in two breast cancer cell lines following long-term (6 months) exposure to Herceptin, after confirming that these cells had become resistant to the drug. We constructed several Boolean models including specific substrates of each DUSP, and showed that our models correctly account for resistance when overexpressed DUSPs were kept activated. We then simulated inhibition of both individual and combinations of DUSPs, and determined conditions under which the resistance could be reversed. CONCLUSIONS: These results show how a combination of experimental analysis and modelling help to understand cell survival mechanisms in breast cancer tumours, and crucially enable us to generate testable predictions potentially leading to new treatments of resistant tumours.


Assuntos
Antineoplásicos/efeitos adversos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Fosfatases de Especificidade Dupla/metabolismo , Biologia de Sistemas/métodos , Trastuzumab/efeitos adversos , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Fosfatases de Especificidade Dupla/antagonistas & inibidores , Fosfatases de Especificidade Dupla/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Modelos Biológicos , Regulação para Cima/efeitos dos fármacos
13.
Am J Physiol Lung Cell Mol Physiol ; 316(5): L918-L933, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-30628487

RESUMO

The involvement of several microRNAs (miRs) in the initiation and development of tumors through the suppression of the target gene expression has been highlighted. The aberrant expression of miR-181d-5p and cyclin-dependent kinase inhibitor 3 (CDKN3) in non-small-cell lung cancer (NSCLC) was then screened by microarray analysis. In the present study, we performed a series of in vivo and in vitro experiments for the purpose of investigating their roles in NSCLC and the underlying mechanism. There was a high expression of CDKN3, whereas miR-181d-5p was downregulated in NSCLC. Quantitative RT-PCR, Western blot analysis, and dual-luciferase reporter gene assay further identified that CDKN3 could be negatively regulated by miR-181d-5p. Moreover, the upregulation of miR-181d-5p or silencing of CDKN3 could inactivate the Akt signaling pathway. A549 with the lowest miR-181d-5p and H1975 with the highest CDKN3 among the five NSCLC cell lines (H1299, A549, H1975, NCI-H157, and GLC-82) were adopted for in vitro experiments, in which expression of miR-181d-5p and CDKN3 was altered by transfection of miR-181d-5p mimic/inhibitor or siRNA-targeting CDKN3. Afterwards, cell proliferation, apoptosis, invasion, migration, and angiogenesis, as well as epithelial-mesenchymal transition (EMT), were evaluated, and tumorigenicity was assessed. In addition, an elevation in miR-181d-5p or depletion in CDKN3 led to significant reductions in proliferation, invasion, migration, angiogenesis, EMT, and tumorigenicity of NSCLC cells, coupling with increased cell apoptosis. In conclusion, this study highlights the tumor-suppressive effects of miR-181d-5p on NSCLC via Akt signaling pathway inactivation by suppressing CDKN3, thus providing a promising therapeutic strategy for the treatment of NSCLC.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Proteínas Inibidoras de Quinase Dependente de Ciclina/metabolismo , Fosfatases de Especificidade Dupla/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , MicroRNAs/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Apoptose/genética , Apoptose/fisiologia , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Proliferação de Células/genética , Proliferação de Células/fisiologia , Proteínas Inibidoras de Quinase Dependente de Ciclina/antagonistas & inibidores , Proteínas Inibidoras de Quinase Dependente de Ciclina/genética , Fosfatases de Especificidade Dupla/antagonistas & inibidores , Fosfatases de Especificidade Dupla/genética , Transição Epitelial-Mesenquimal/genética , Transição Epitelial-Mesenquimal/fisiologia , Regulação Neoplásica da Expressão Gênica , Genes Supressores de Tumor , Xenoenxertos , Humanos , Neoplasias Pulmonares/patologia , Masculino , Camundongos , Camundongos Nus , MicroRNAs/antagonistas & inibidores , MicroRNAs/metabolismo , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética , Transdução de Sinais
14.
Bioorg Chem ; 81: 264-269, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30153591

RESUMO

Several protein tyrosine phosphatases (PTPs) that disrupt the insulin-signaling pathway were investigated by siRNAs to identify potential antidiabetic targets. Individual knockdown of PTPN9 and DUSP9 in 3T3-L1 preadipocytes increased AMPK phosphorylation, respectively, and furthermore, concurrent knockdown of both PTPN9 and DUSP9 synergistically increased AMPK phosphorylation. Next, 658 natural products were screened to identify dual inhibitors of both PTPN9 and DUSP9. Based on the selectivity and inhibition potency of the compounds, ginkgolic acid (GA) was selected for further study as a potential antidiabetic drug candidate. GA inhibited the enzymatic activity of PTPN9 (Ki = 53 µM) and DUSP9 (Ki = 2.5 µM) in vitro and resulted in a significant increase of glucose-uptake in differentiated C2C12 muscle cells and 3T3-L1 adipocytes. In addition, GA increased phosphorylation of AMPK in 3T3L1 adipocytes. In this study, GA as a dual targeting inhibitor of PTPN9 and DUSP9 increased glucose uptake in 3T3L1 and C2C12 cells by activating the AMPK signaling pathway. These results strongly suggest GA could be used as a therapeutic candidate for type 2 diabetes.


Assuntos
Fosfatases de Especificidade Dupla/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Hipoglicemiantes/farmacologia , Fosfatases da Proteína Quinase Ativada por Mitógeno/antagonistas & inibidores , Proteínas Tirosina Fosfatases não Receptoras/antagonistas & inibidores , Salicilatos/farmacologia , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Linhagem Celular , Fosfatases de Especificidade Dupla/genética , Técnicas de Silenciamento de Genes , Glucose/metabolismo , Camundongos , Fosfatases da Proteína Quinase Ativada por Mitógeno/genética , Fosforilação/efeitos dos fármacos , Fosforilação/genética , Proteínas Tirosina Fosfatases não Receptoras/genética
15.
J Med Chem ; 60(22): 9299-9319, 2017 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-29116812

RESUMO

Excessive activity of striatal-enriched protein tyrosine phosphatase (STEP) in the brain has been detected in numerous neuropsychiatric disorders including Alzheimer's disease. Notably, knockdown of STEP in an Alzheimer mouse model effected an increase in the phosphorylation levels of downstream STEP substrates and a significant reversal in the observed cognitive and memory deficits. These data point to the promising potential of STEP as a target for drug discovery in Alzheimer's treatment. We previously reported a substrate-based approach to the development of low molecular weight STEP inhibitors with Ki values as low as 7.8 µM. Herein, we disclose the first X-ray crystal structures of inhibitors bound to STEP and the surprising finding that they occupy noncoincident binding sites. Moreover, we utilize this structural information to optimize the inhibitor structure to achieve a Ki of 110 nM, with 15-60-fold selectivity across a series of phosphatases.


Assuntos
Organofosfonatos/química , Proteínas Tirosina Fosfatases não Receptoras/antagonistas & inibidores , Sulfonamidas/química , Doença de Alzheimer/tratamento farmacológico , Animais , Domínio Catalítico , Cristalografia por Raios X , Descoberta de Drogas , Estabilidade de Medicamentos , Fosfatases de Especificidade Dupla/antagonistas & inibidores , Microssomos Hepáticos/metabolismo , Organofosfonatos/síntese química , Organofosfonatos/metabolismo , Proteínas Tirosina Fosfatases não Receptoras/química , Ratos , Sulfonamidas/síntese química , Sulfonamidas/metabolismo
16.
Compr Physiol ; 7(4): 1449-1461, 2017 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-28915331

RESUMO

The mammalian genome contains approximately 200 phosphatases that are responsible for catalytically removing phosphate groups from proteins. In this review, we discuss dual specificity phosphatase 5 (DUSP5). DUSP5 belongs to the dual specificity phosphatase (DUSP) family, so named after the family members' abilities to remove phosphate groups from serine/threonine and tyrosine residues. We provide a comparison of DUSP5's structure to other DUSPs and, using molecular modeling studies, provide an explanation for DUSP5's mechanistic interaction and specificity toward phospho-extracellular regulated kinase, its only known substrate. We also discuss new insights from molecular modeling studies that will influence our current thinking of mitogen-activated protein kinase signaling. Finally, we discuss the lessons learned from identifying small molecules that target DUSP5, which might benefit targeting efforts for other phosphatases. © 2017 American Physiological Society. Compr Physiol 7:1449-1461, 2017.


Assuntos
Fosfatases de Especificidade Dupla/metabolismo , Inibidores Enzimáticos/farmacologia , Simulação de Acoplamento Molecular , Animais , Sítios de Ligação , Fosfatases de Especificidade Dupla/antagonistas & inibidores , Fosfatases de Especificidade Dupla/química , Fosfatases de Especificidade Dupla/genética , Inibidores Enzimáticos/química , Humanos , Sistema de Sinalização das MAP Quinases , Ligação Proteica
17.
BMC Biochem ; 18(1): 10, 2017 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-28569147

RESUMO

BACKGROUND: Protein tyrosine phosphatases (PTPs) like dual specificity phosphatase 5 (DUSP5) and protein tyrosine phosphatase 1B (PTP1B) are drug targets for diseases that include cancer, diabetes, and vascular disorders such as hemangiomas. The PTPs are also known to be notoriously difficult targets for designing inihibitors that become viable drug leads. Therefore, the pipeline for approved drugs in this class is minimal. Furthermore, drug screening for targets like PTPs often produce false positive and false negative results. RESULTS: Studies presented herein provide important insights into: (a) how to detect such artifacts, (b) the importance of compound re-synthesis and verification, and (c) how in situ chemical reactivity of compounds, when diagnosed and characterized, can actually lead to serendipitous discovery of valuable new lead molecules. Initial docking of compounds from the National Cancer Institute (NCI), followed by experimental testing in enzyme inhibition assays, identified an inhibitor of DUSP5. Subsequent control experiments revealed that this compound demonstrated time-dependent inhibition, and also a time-dependent change in color of the inhibitor that correlated with potency of inhibition. In addition, the compound activity varied depending on vendor source. We hypothesized, and then confirmed by synthesis of the compound, that the actual inhibitor of DUSP5 was a dimeric form of the original inhibitor compound, formed upon exposure to light and oxygen. This compound has an IC50 of 36 µM for DUSP5, and is a competitive inhibitor. Testing against PTP1B, for selectivity, demonstrated the dimeric compound was actually a more potent inhibitor of PTP1B, with an IC50 of 2.1 µM. The compound, an azo-bridged dimer of sulfonated naphthol rings, resembles previously reported PTP inhibitors, but with 18-fold selectivity for PTP1B versus DUSP5. CONCLUSION: We report the identification of a potent PTP1B inhibitor that was initially identified in a screen for DUSP5, implying common mechanism of inhibitory action for these scaffolds.


Assuntos
Naftóis/farmacologia , Proteína Tirosina Fosfatase não Receptora Tipo 1/antagonistas & inibidores , Dimerização , Avaliação Pré-Clínica de Medicamentos/métodos , Fosfatases de Especificidade Dupla/antagonistas & inibidores , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/farmacologia , Humanos , Simulação de Acoplamento Molecular , Naftóis/síntese química , Naftóis/química
18.
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
19.
Cell Stress Chaperones ; 22(2): 307-315, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27975203

RESUMO

Heat stress to a cell leads to the activation of heat shock response, which is required for the management of misfolded and unfolded proteins. Macroautophagy and proteasome-mediated degradation are the two cellular processes that degrade polyubiquitinated, misfolded proteins. Contrasting pieces of evidence exist on the effect of heat stress on the activation of the above-mentioned degradative pathways. Laforin phosphatase and malin E3 ubiquitin ligase, the two proteins defective in Lafora neurodegenerative disorder, are involved in cellular stress response pathways and are required for the activation of heat shock transcription factor - the heat shock factor 1 (HSF1) - and, consequently, for cellular protection under heat shock. While the role of laforin and malin in the proteolytic pathways is well established, their role in cellular recovery from heat shock was not explored. To address this, we investigated autophagic flux, proteasomal activity, and the level of polyubiquitinated proteins in Neuro2a cells partially silenced for laforin or malin protein and exposed to heat shock. We found that heat shock was able to induce autophagic flux, proteasomal activity and reduce the polyubiquitinated proteins load in the laforin-silenced cells but not in the malin-deficient cells. Loss of malin leads to reduced proteasomal activity in the heat-shocked cells. Taken together, our results suggest a distinct mode of action for laforin and malin in the heat shock-induced proteolytic processes.


Assuntos
Autofagia , Fosfatases de Especificidade Dupla/metabolismo , Resposta ao Choque Térmico/fisiologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Linhagem Celular Tumoral , Fosfatases de Especificidade Dupla/antagonistas & inibidores , Fosfatases de Especificidade Dupla/genética , Fatores de Transcrição de Choque Térmico/metabolismo , Leupeptinas/farmacologia , Macrolídeos/toxicidade , Camundongos , Proteínas Associadas aos Microtúbulos/metabolismo , Complexo de Endopeptidases do Proteassoma/química , Proteínas Tirosina Fosfatases não Receptoras , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Temperatura , Ubiquitina-Proteína Ligases/antagonistas & inibidores , Ubiquitina-Proteína Ligases/genética , Ubiquitinação
20.
Pharmazie ; 71(4): 196-200, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27209699

RESUMO

Protein tyrosine phosphatases (PTPs) regulate protein function by dephosphorylating phosphorylated proteins in many signaling cascades and some of them have been targets for drug development against many human diseases. There have been many reports that some chemical inhibitors could regulate particular phosphatases. However, there was no extensive study on specificity of inhibitors towardss phosphatases. We investigated the effects of ethyl-3,4-dephostatin, a potent inhibitor of five PTPs including PTP-1B and Src homology-2-containing protein tyrosine phosphatase-1 (SHP-1), on thirteen other PTPs using in vitro phosphatase assays. Of them, dual-specificity protein phosphatase 26 (DUSP26), which inhibits mitogen-activated protein kinase (MAPK) and p53 tumor suppressor and is known to be overexpressed in anaplastic thyroid carcinoma, was inhibited by ethyl-3,4-dephostatin in a concentration-dependent manner. Kinetic studies with ethyl-3,4-dephostatin and DUSP26 revealed competitive inhibition, suggesting that ethyl-3,4-dephostatin binds to the catalytic site of DUSP26 like other substrate PTPs. Moreover, ethyl-3,4-dephostatin protects DUSP26-mediated dephosphorylation of p38, a member of the MAPK family, and p53. Taken together, these results suggest that ethyl-3,4-dephostatin functions as a multiphosphatase inhibitor and is useful as a therapeutic agent for cancers overexpressing DUSP26.


Assuntos
Fosfatases de Especificidade Dupla/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Hidroquinonas/farmacologia , Fosfatases da Proteína Quinase Ativada por Mitógeno/antagonistas & inibidores , Ligação Competitiva/efeitos dos fármacos , Domínio Catalítico , Fosfatases de Especificidade Dupla/genética , Células HEK293 , Humanos , Cinética , Fosfatases da Proteína Quinase Ativada por Mitógeno/genética , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Plasmídeos/genética , Proteína Tirosina Fosfatase não Receptora Tipo 1/biossíntese , Proteína Tirosina Fosfatase não Receptora Tipo 6/biossíntese , Proteína Supressora de Tumor p53/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA