Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Chem Biol Drug Des ; 103(2): e14458, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-38383061

RESUMO

JNK3, a neuronal kinase activated by stress, plays a role in stress-induced apoptosis, leading to neuronal cell death following cerebral ischemia. This study investigates the neuroprotective effects of piceatannol (PCT) in SHSY-5Y neuroblastoma cells after hypoxic injury and its interaction with JNK3. We analyzed the crystal coordinates, interaction energies, and amino acid interactions to determine PCT's selectivity for JNK3. The electrostatic potential was computed using density functional theory, while molecular dynamics assessed the stability and structural consistency of the JNK3-PCT complex. We used SP600125 (SP6), a JNK3 inhibitor, as a reference compound. Additionally, we performed cell-free JNK 1, 2, and 3 kinase assays to evaluate the isoform selectivity of PCT. Cytotoxicity and cell viability were determined by an MTT test. To assess apoptosis, we used acridine orange/ethidium bromide dual fluorescent labeling and ANNEXIN A5-FITC flow cytometry. Western blot was used to evaluate the attenuation of JNK3 and apoptotic proteins. In silico studies revealed a stronger binding affinity between PCT and JNK3 compared to JNK1 and JNK2, which was further supported by the in vitro kinase assay. PCT-treated cells exhibited a decrease in Cyt-c and caspase-3 expression and an increase in Bcl-2 level, compared to hypoxic control (p < .001). PCT also demonstrated superior efficacy over SP6 in inhibiting JNK3 phosphorylation (p < .001). Furthermore, PCT significantly increased the expression of neuronal genes, including NgN1, neuroD2, and survivin (p < .001). In conclusion, PCT is a potential JNK3 inhibitor, since it inhibited phosphorylation and the Bcl-2/Cyt-C/caspase-3-dependent apoptotic pathway after ischemic/hypoxic insult.


Assuntos
Caspases , Oxigênio , Estilbenos , Caspase 3 , Caspases/farmacologia , Proteína Quinase 10 Ativada por Mitógeno/genética , Proteína Quinase 10 Ativada por Mitógeno/metabolismo , Apoptose , Linhagem Celular
2.
Pol J Vet Sci ; 24(3): 439-443, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34730307

RESUMO

Reticuloendotheliosis virus (REV) is an avian oncogenic retrovirus that causes atrophy of immune organs, such as the spleen, thymus, and bursa of Fabricius, leading to severe immunosuppression. However, there is limited information describing the genes or microRNAs (miRNAs) that play a role in replicating REV-spleen necrosis virus (SNV). Our previous miRNA and RNA sequencing data showed that the expression of gga-miR-222b-5p was significantly upregulated in REV-SNV-infected chicken spleens of 7, 14, and 21 dpi compared to non-infected chicken spleens, but mitogen-activated protein kinase 10 (MAPK10), which is related to innate immunity, had the opposite expression pattern. To understand chicken cellular miRNA function in the virus-host interactions during REV infection, we used quantitative reverse transcription PCR (qRT-PCR) to determine whether the expression of gga-miR-222b-5p and MAPK10 in the spleen of specific-pathogen-free chickens at 28, 35, and 42 dpi was consistent with the first 3 time points, and dual-luciferase reporter assay was used to determine the targeting relationship between gga-miR-222b-5p and MAPK10. Results show that MAPK10 was downregulated at all 3 time points; however, significant difference (p⟨0.01) was noted only at 35 dpi. Moreover, the expression of gga-miR-222b-5p was upregulated; however, significant difference (p⟨0.01) was observed only at 28 and 35 dpi. A dual-luciferase reporter assay showed that MAPK10 is a direct target of gga-miR-222b-5p. This study suggests that gga-miR-222b-5p may target MAPK10 to promote the REV-SNV-induced tumorigenesis via the RLRs signaling pathway.


Assuntos
MicroRNAs/metabolismo , Proteína Quinase 10 Ativada por Mitógeno/metabolismo , Infecções por Retroviridae/veterinária , Baço/metabolismo , Vírus do Infarto Esplênico do Pato de Trager/imunologia , Transcriptoma , Animais , Galinhas , Regulação para Baixo , Regulação da Expressão Gênica , Humanos , MicroRNAs/genética , Proteína Quinase 10 Ativada por Mitógeno/genética , Infecções por Retroviridae/virologia , Organismos Livres de Patógenos Específicos , Infecções Tumorais por Vírus/veterinária , Infecções Tumorais por Vírus/virologia
3.
Life Sci ; 275: 119396, 2021 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-33774030

RESUMO

AIMS: The mitogen-activated protein kinase (MAPK) cascades integrate various upstream signals to regulate many cellular functions, including proliferation, differentiation, and survival. Dysregulation of these pathways has been implicated in the occurrence and progression of a variety of cancers. MAIN METHODS: This study aimed to assess the association of 192 single nucleotide polymorphisms in 22 MAPK cascade genes with renal cell carcinoma (RCC) risk and survival in 312 patients and 318 controls. KEY FINDINGS: After multiple testing correction and multivariate analysis, the minor T allele of MAPK10 rs12648265 remained associated with a lower risk of RCC (adjusted odds ratio 0.64, 95% confidence interval 0.50-0.82, P = 0.000426) and metastasis (adjusted hazard ratio 0.50, 95% confidence interval 0.30-0.82, P = 0.006). Presence of the rs12648265 T allele demonstrated a trend towards being associated with increased MAPK10 expression, and meta-analysis of four RCC datasets indicated that high MAPK10 expression is associated with a favourable prognosis. Furthermore, activation of MAPK10 by the potent agonist anisomycin inhibited RCC cell growth in vitro, suggesting an involvement of MAPK10 in RCC progression. SIGNIFICANCE: In conclusion, MAPK10 may be a meaningful biomarker and a potential therapeutic target in RCC.


Assuntos
Carcinoma de Células Renais/genética , Predisposição Genética para Doença/genética , Neoplasias Renais/genética , Proteína Quinase 10 Ativada por Mitógeno/genética , Polimorfismo de Nucleotídeo Único/genética , Idoso , Western Blotting , Estudos de Casos e Controles , Linhagem Celular Tumoral , Feminino , Humanos , Masculino , Pessoa de Meia-Idade
4.
Cell Mol Life Sci ; 78(1): 287-298, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32189007

RESUMO

Unveiling the key pathways underlying postnatal beta-cell proliferation can be instrumental to decipher the mechanisms of beta-cell mass plasticity to increased physiological demand of insulin during weight gain and pregnancy. Using transcriptome and global Serine Threonine Kinase activity (STK) analyses of islets from newborn (10 days old) and adult rats, we found that highly proliferative neonatal rat islet cells display a substantially elevated activity of the mitogen activated protein 3 kinase 12, also called dual leucine zipper-bearing kinase (Dlk). As a key upstream component of the c-Jun amino terminal kinase (Jnk) pathway, Dlk overexpression was associated with increased Jnk3 activity and was mainly localized in the beta-cell cytoplasm. We provide the evidence that Dlk associates with and activates Jnk3, and that this cascade stimulates the expression of Ccnd1 and Ccnd2, two essential cyclins controlling postnatal beta-cell replication. Silencing of Dlk or of Jnk3 in neonatal islet cells dramatically hampered primary beta-cell replication and the expression of the two cyclins. Moreover, the expression of Dlk, Jnk3, Ccnd1 and Ccnd2 was induced in high replicative islet beta cells from ob/ob mice during weight gain, and from pregnant female rats. In human islets from non-diabetic obese individuals, DLK expression was also cytoplasmic and the rise of the mRNA level was associated with an increase of JNK3, CCND1 and CCND2 mRNA levels, when compared to islets from lean and obese patients with diabetes. In conclusion, we find that activation of Jnk3 signalling by Dlk could be a key mechanism for adapting islet beta-cell mass during postnatal development and weight gain.


Assuntos
Células Secretoras de Insulina/metabolismo , MAP Quinase Quinase Quinases/metabolismo , Proteína Quinase 10 Ativada por Mitógeno/metabolismo , Transdução de Sinais , Animais , Proliferação de Células/efeitos dos fármacos , Ciclina D1/genética , Ciclina D1/metabolismo , Ciclina D2/genética , Ciclina D2/metabolismo , Feminino , Glucose/farmacologia , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/citologia , MAP Quinase Quinase Quinases/antagonistas & inibidores , MAP Quinase Quinase Quinases/genética , Camundongos , Camundongos Endogâmicos C57BL , Proteína Quinase 10 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 10 Ativada por Mitógeno/genética , Obesidade/metabolismo , Obesidade/patologia , Pâncreas/crescimento & desenvolvimento , Pâncreas/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos
5.
Eur Rev Med Pharmacol Sci ; 24(11): 6014-6022, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32572915

RESUMO

OBJECTIVE: To explore the expression pattern and clinical significance of circ_0000515 in hepatocellular carcinoma (HCC), as well as the molecular mechanism. PATIENTS AND METHODS: Fifty HCC patients were recruited, and their cancer tissues and adjacent normal ones were collected for detecting the differential expression of circ_0000515. The relationship between circ_0000515 and clinical parameters in HCC patients was analyzed. Circ_0000515 knockdown model was generated by lentivirus transfection in Hep3B and MHCC88H cells that were highly expressed with circ_0000515. Regulatory effects of circ_0000515 on phenotypes of Hep3B and MHCC88H cells were examined by Cell Counting Kit-8 (CCK-8) and transwell assay. Target gene of circ_0000515 was verified by Dual-Luciferase reporter assay, and its involvement in HCC progression was detected by rescue experiments. In vivo xenograft model was generated in nude mice aiming to elucidate the role of circ_0000515 in regulating HCC growth. RESULTS: Circ_0000515 was highly expressed in HCC tissues and cell lines. High level of circ_0000515 predicted advanced stage, high incidence of lymphatic metastasis, and low disease-free survival and overall survival in HCC. Knockdown of circ_0000515 attenuated proliferative and migratory abilities in Hep3B and MHCC88H cells. MAPK10, as the target gene binding circ_0000515, was negatively regulated by circ_0000515. Rescue experiments and in vivo xenograft model both indicated that circ_0000515 aggravated the malignant progression of HCC by targeting MAPK10. CONCLUSIONS: Circ_0000515 is upregulated in HCC tissues and cell lines. It can be used for predicting tumor staging, lymphatic metastasis, and prognosis in HCC. Circ_0000515 aggravates the malignant progression of HCC by downregulating MAPK10.


Assuntos
Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , Proteína Quinase 10 Ativada por Mitógeno/metabolismo , RNA Circular/metabolismo , Carcinoma Hepatocelular/patologia , Linhagem Celular , Movimento Celular , Proliferação de Células , Feminino , Humanos , Neoplasias Hepáticas/patologia , Masculino , Pessoa de Meia-Idade , Proteína Quinase 10 Ativada por Mitógeno/genética , RNA Circular/genética
6.
Parasite Immunol ; 42(2): e12687, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31770453

RESUMO

AIMS: This study aimed at evaluating the DNA vaccination efficacy of Leishmania major-derived MAPK10 against Leishmania donovani infection. METHODS AND RESULTS: MAPK10 is one of the 15 mitogen-activated protein kinases (MAPKs) of Leishmania major. Herein, we expressed the gene through a mammalian vector and tested whether priming with this gene would offer protection against L donovani infection. We report that LmjMAPK10 DNA vaccination using a mammalian expression vector significantly reduces the parasite burden. The protection is accompanied by host-protective T-cell functions, TH 1-type cytokines and elevated leishmanial antigen-specific IgG2a isotype response. T-cell response to the L donovani/challenge infection is associated with increase in IL-12 and IFN-γ, but reduced IL-10 and IL-4 production. CONCLUSIONS: LmjMAPK10 is cross-protective against L donovani infection.


Assuntos
Leishmania donovani/imunologia , Vacinas contra Leishmaniose/imunologia , Leishmaniose Visceral/prevenção & controle , Proteína Quinase 10 Ativada por Mitógeno/imunologia , Proteínas de Protozoários/imunologia , Vacinas de DNA/imunologia , Animais , Anticorpos Antiprotozoários/imunologia , Humanos , Imunoglobulina G/imunologia , Interferon gama/biossíntese , Interleucina-10/biossíntese , Subunidade p35 da Interleucina-12/biossíntese , Interleucina-4/biossíntese , Leishmania donovani/genética , Leishmaniose Visceral/imunologia , Macrófagos/imunologia , Macrófagos/parasitologia , Camundongos , Camundongos Endogâmicos BALB C , Proteína Quinase 10 Ativada por Mitógeno/genética , Proteínas de Protozoários/administração & dosagem , Proteínas de Protozoários/genética , Células Th1/imunologia
7.
Nat Commun ; 10(1): 4223, 2019 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-31530804

RESUMO

Diseases related to impaired blood flow such as peripheral artery disease (PAD) impact nearly 10 million people in the United States alone, yet patients with clinical manifestations of PAD (e.g., claudication and limb ischemia) have limited treatment options. In ischemic tissues, stress kinases such as c-Jun N-terminal kinases (JNKs), are activated. Here, we show that inhibition of the JNK3 (Mapk10) in the neural compartment strikingly potentiates blood flow recovery from mouse hindlimb ischemia. JNK3 deficiency leads to upregulation of growth factors such as Vegfa, Pdgfb, Pgf, Hbegf and Tgfb3 in ischemic muscle by activation of the transcription factors Egr1/Creb1. JNK3 acts through Forkhead box O3 (Foxo3a) to suppress the activity of Egr1/Creb1 transcription regulators in vitro. In JNK3-deficient cells, Foxo3a is suppressed which leads to Egr1/Creb1 activation and upregulation of downstream growth factors. Collectively, these data suggest that the JNK3-Foxo3a-Egr1/Creb1 axis coordinates the vascular remodeling response in peripheral ischemia.


Assuntos
Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Membro Posterior/irrigação sanguínea , Isquemia/metabolismo , Proteína Quinase 10 Ativada por Mitógeno/metabolismo , Neurônios/metabolismo , Animais , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Proteína 1 de Resposta de Crescimento Precoce/genética , Proteína Forkhead Box O3/genética , Proteína Forkhead Box O3/metabolismo , Membro Posterior/inervação , Membro Posterior/metabolismo , Humanos , Isquemia/genética , Isquemia/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteína Quinase 10 Ativada por Mitógeno/genética , Músculo Esquelético/metabolismo , Fluxo Sanguíneo Regional , Transdução de Sinais
8.
Structure ; 27(7): 1162-1170.e3, 2019 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-31080119

RESUMO

Arrestins, in addition to desensitizing GPCR-induced G protein activation, also mediate G protein-independent signaling by interacting with various signaling proteins. Among these, arrestins regulate MAPK signal transduction by scaffolding mitogen-activated protein kinase (MAPK) signaling components such as MAPKKK, MAPKK, and MAPK. In this study, we investigated the binding mode and interfaces between arrestin-3 and JNK3 using hydrogen/deuterium exchange mass spectrometry, 19F-NMR, and tryptophan-induced Atto 655 fluorescence-quenching techniques. Results suggested that the ß1 strand of arrestin-3 is the major and potentially only interaction site with JNK3. The results also suggested that C-lobe regions near the activation loop of JNK3 form the potential binding interface, which is variable depending on the ATP binding status. Because the ß1 strand of arrestin-3 is buried by the C-terminal strand in its basal state, C-terminal truncation (i.e., pre-activation) of arrestin-3 facilitates the arrestin-3/JNK3 interaction.


Assuntos
Trifosfato de Adenosina/química , Arrestinas/química , Proteína Quinase 10 Ativada por Mitógeno/química , Trifosfato de Adenosina/metabolismo , Sequência de Aminoácidos , Animais , Arrestinas/genética , Arrestinas/metabolismo , Sítios de Ligação , Clonagem Molecular , Cristalografia por Raios X , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Humanos , Sistema de Sinalização das MAP Quinases/genética , Proteína Quinase 10 Ativada por Mitógeno/genética , Proteína Quinase 10 Ativada por Mitógeno/metabolismo , Modelos Moleculares , Fosforilação , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Domínios e Motivos de Interação entre Proteínas , Ratos , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
9.
Oncol Rep ; 37(5): 2679-2687, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28393229

RESUMO

miRNA-27a-3p is an important regulator of carcinogenesis and other pathological processes. However, its role in laryngeal carcinoma is still unknown. In our previous research, we found that miR-27a-3p expression was upregulated in nasopharyngeal carcinoma (NPC) using a microarray chip. In the present study, we identified miR-27a-3p as an endogenous promoter of metastatic invasion. The expression levels of miR-27a-3p were correlated with human metastatic progression outcomes and Kaplan-Meier survival. In silico database analyses revealed that Mapk10 is a potential target of miR-27a-3p, and luciferase reporter assay results revealed that miR-27a-3p directly inhibits the Mapk10 3' untranslated region (3'UTR). Real-time PCR and western blotting results ascertained that Mapk10 expression was regulated by miR­27a-3p. In addition, miR­27a-3p gain-of-function promoted cell proliferation, migration and invasion in 5-8 F NPC cells. These effects partially depended on Mapk10, and loss of miR­27a-3p function had the opposite effects.


Assuntos
Carcinoma/genética , Carcinoma/patologia , MicroRNAs/genética , Proteína Quinase 10 Ativada por Mitógeno/genética , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/patologia , Carcinoma/metabolismo , Carcinoma/mortalidade , Estudos de Casos e Controles , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Regulação para Baixo , Regulação Neoplásica da Expressão Gênica , Humanos , Estimativa de Kaplan-Meier , MicroRNAs/metabolismo , Proteína Quinase 10 Ativada por Mitógeno/metabolismo , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/metabolismo , Neoplasias Nasofaríngeas/mortalidade
10.
Toxicology ; 380: 30-37, 2017 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-28163110

RESUMO

Cadmium (Cd) is a heavy metal with a long biological half-life in humans and is recognized as a toxic pollutant. Cd is also a potential neurotoxicant and its exposure is associated with olfactory impairment in humans. However, the molecular and cellular mechanisms of Cd neurotoxicity are not well defined. Adult neurogenesis is a process that generates functional neurons from adult neural stem/progenitor cells (aNPCs). It occurs in specific regions of the adult brain including the subventricular zone (SVZ) along the lateral ventricles in mammals, a process that is critical for olfaction. Various external stimuli can modulate adult neurogenesis and the effect of neurotoxicants on adult neurogenesis is just beginning to be elucidated. Since Cd exposure can impair olfaction in humans, the goal of this study is to investigate the effects of Cd on SVZ adult neurogenesis and underlying mechanisms using primary cultured SVZ-aNPCs. In this study, we report that low-level Cd exposure decreases cell number, induces apoptosis, and inhibits cell proliferation in SVZ-aNPCs. Furthermore, Cd exposure significantly increases phosphorylation of c-Jun NH2-terminal kinase (JNK), and p38 MAP kinase in these cells, indicative of JNK and p38 activation. Pharmacological inhibition of JNK or p38 MAPK kinases attenuated Cd-induced cell loss and apoptosis. Cd treatment did not cause cell loss or apoptosis in SVZ-aNPCs prepared from transgenic mice null for the neural-specific JNK3 isoform. These data suggest a critical role for p38 and JNK3 MAP kinases in Cd neurotoxicity. These results are, to our knowledge, the first demonstration that Cd impairs SVZ adult neurogenesis in vitro, which may contribute to its neurotoxicity in olfaction.


Assuntos
Cádmio/toxicidade , Proliferação de Células/efeitos dos fármacos , Proteína Quinase 10 Ativada por Mitógeno/metabolismo , Células-Tronco Neurais/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Apoptose/efeitos dos fármacos , Células Cultivadas , Ventrículos Laterais/citologia , Ventrículos Laterais/efeitos dos fármacos , Ventrículos Laterais/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteína Quinase 10 Ativada por Mitógeno/genética , Células-Tronco Neurais/patologia , Fosforilação , Proteínas Quinases p38 Ativadas por Mitógeno/genética
11.
Acta Biomater ; 53: 59-69, 2017 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-28216301

RESUMO

In order to identify the mechanisms by which skeletal maturity alters the mechanosensitivity of mesenchymal stromal cells (MSCs) and, the implications for osteogenesis and angiogenesis during bone formation, we compared the response of MSCs derived from children and skeletally-mature healthy adults cultured on soft and stiff collagen-coated polyacrylamide substrates. MSCs from children were more mechanosensitive, showing enhanced angiogenesis and osteogenesis on stiff substrates as indicated by increased endothelial tubule formation, PGF production, nuclear-translocation of YAP, ALP activity and mineralisation. To examine these mechanisms in more detail, a customised PCR array identified an age-dependent, stiffness-induced upregulation of NOX1, VEGFR1, VEGFR2, WIF1 and, of particular interest, JNK3 in cells from children compared to adults. When JNK3 activity was inhibited, a reduction in stiffness-induced driven osteogenesis was observed - suggesting that JNK3 might serve as a novel target for recapitulating the enhanced regenerative potential of children in adults suffering from bone degeneration. STATEMENT OF SIGNIFICANCE: We investigated the age-associated changes in the capacity of MSCs for bone regeneration involving the mechanosensitive signalling pathways, which reduce the ability of adult cells to respond to biophysical cues in comparison to cells from children, who are still undergoing bone development. Our results offer new insights into the mechanobiology of MSCs and sheds new light on age-altered mechanosensitivity and, on why children have such an immense capacity to regenerate their skeletal system. We have identified the mechanisms by which skeletal maturity alters the mechanosensitivity of mesenchymal stromal cells and an age-dependent, stiffness-induced upregulation of a number of prominent genes including, most notably, JNK3 in children cells, thus suggesting its potential to promote enhanced bone repair.


Assuntos
Envelhecimento/fisiologia , Mecanotransdução Celular , Células-Tronco Mesenquimais/fisiologia , Neovascularização Fisiológica , Osteogênese , Transporte Ativo do Núcleo Celular , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Adulto , Envelhecimento/genética , Fenômenos Biomecânicos , Regeneração Óssea , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Células Cultivadas , Criança , Humanos , Sistema de Sinalização das MAP Quinases , Células-Tronco Mesenquimais/citologia , Proteína Quinase 10 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 10 Ativada por Mitógeno/genética , Fosfoproteínas/metabolismo , Fator de Crescimento Placentário/genética , Fator de Crescimento Placentário/metabolismo , Fatores de Transcrição , Regulação para Cima , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Proteínas de Sinalização YAP , Adulto Jovem
12.
EMBO Mol Med ; 9(2): 251-264, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28011860

RESUMO

Although remnant cardiomyocytes (CMs) possess a certain degree of proliferative ability, efficiency is too low for cardiac regeneration after injury. In this study, we identified a distinct stage within the initiation phase of CM reprogramming before the MET process, and microarray analysis revealed the strong up-regulation of several mitosis-related genes at this stage of reprogramming. Several candidate genes were selected and tested for their ability to induce CM proliferation. Delivering a cocktail of three genes, FoxM1, Id1, and Jnk3-shRNA (FIJs), induced CMs to re-enter the cell cycle and complete mitosis and cytokinesis in vitro More importantly, this gene cocktail increased CM proliferation in vivo and significantly improved cardiac function and reduced fibrosis after myocardial infarction. Collectively, our findings present a cocktail FIJs that may be useful in cardiac regeneration and also provide a practical strategy for probing reprogramming assays for regeneration of other tissues.


Assuntos
Proliferação de Células , Terapia Genética/métodos , Infarto do Miocárdio/terapia , Miócitos Cardíacos/fisiologia , Regeneração , Animais , Dependovirus/genética , Proteína Forkhead Box M1/genética , Vetores Genéticos , Proteína 1 Inibidora de Diferenciação/genética , Camundongos Endogâmicos C57BL , Proteína Quinase 10 Ativada por Mitógeno/genética , RNA Interferente Pequeno/genética , Medicina Regenerativa/métodos , Transdução Genética , Resultado do Tratamento
13.
PLoS Genet ; 12(11): e1006439, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27902697

RESUMO

Hirschsprung disease (HSCR) is characterized by absence of enteric neurons from the distal colon and severe intestinal dysmotility. To understand the pathophysiology and genetics of HSCR we developed a unique zebrafish model that allows combined genetic, developmental and in vivo physiological studies. We show that ret mutant zebrafish exhibit cellular, physiological and genetic features of HSCR, including absence of intestinal neurons, reduced peristalsis, and varying phenotype expressivity in the heterozygous state. We perform live imaging experiments using a UAS-GAL4 binary genetic system to drive fluorescent protein expression in ENS progenitors. We demonstrate that ENS progenitors migrate at reduced speed in ret heterozygous embryos, without changes in proliferation or survival, establishing this as a principal pathogenic mechanism for distal aganglionosis. We show, using live imaging of actual intestinal movements, that intestinal motility is severely compromised in ret mutants, and partially impaired in ret heterozygous larvae, and establish a clear correlation between neuron position and organised intestinal motility. We exploited the partially penetrant ret heterozygous phenotype as a sensitised background to test the influence of a candidate modifier gene. We generated mapk10 loss-of-function mutants, which show reduced numbers of enteric neurons. Significantly, we show that introduction of mapk10 mutations into ret heterozygotes enhanced the ENS deficit, supporting MAPK10 as a HSCR susceptibility locus. Our studies demonstrate that ret heterozygous zebrafish is a sensitized model, with many significant advantages over existing murine models, to explore the pathophysiology and complex genetics of HSCR.


Assuntos
Sistema Nervoso Entérico/metabolismo , Doença de Hirschsprung/genética , Proteína Quinase 10 Ativada por Mitógeno/genética , Proteínas Proto-Oncogênicas c-ret/genética , Peixe-Zebra/genética , Animais , Colo/inervação , Colo/patologia , Modelos Animais de Doenças , Sistema Nervoso Entérico/patologia , Doença de Hirschsprung/metabolismo , Doença de Hirschsprung/patologia , Humanos , Mutação , Neurônios/metabolismo , Neurônios/patologia , Fenótipo , Proteínas Proto-Oncogênicas c-ret/metabolismo
14.
Cell Physiol Biochem ; 39(4): 1433-43, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27606894

RESUMO

BACKGROUND/AIMS: The hepatitis B virus X protein (HBx) contributes to HBV-induced injury of renal tubular cells and induces apoptosis via Fas/FasL up-regulation. However, the mechanism of Fas/FasL activation is unknown. Recent studies indicated that HBx induction of apoptosis in hepatic cells depends on activating the MLK3-MKK7-JNKs signaling module, which then up-regulates FasL expression. In this study, we used NRK-52E cells transfected an HBx expression vector to examine the role of the MLK3-MKK7-JNKs signaling pathway on HBx-induced renal tubular cell injury. METHODS: NRK-52E cells were transfected with pc-DNA3.1(+)-HBx to establish an HBx over-expression model, and with pc-DNA3.1(+)-HBx and pSilencer3.1-shHBx to establish an HBx low expression model. One control group was not transfected and another control group was transfected with an empty plasmid. Cell proliferation was determined by the formazan dye method (Cell Counting Kit-8) and apoptosis was measured by flow cytometry and fluorescence microscopy. Western blotting was used to measure the expression of Fas, FasL, and MLK3-MKK7-JNKs signaling pathway-related proteins. The activity of caspase-8 was measured by spectrophotometry. RESULTS: Transfection of NRK-52E cells with pc-DNA3.1(+)-HBx inhibited cell proliferation and increased apoptosis and caspase-8 activity. The expression of Fas, FasL, and MLK3-MKK7-JNKs signaling pathway-related proteins were also greater in the pc-DNA3.1(+)-HBx group, but lower in RNAi group. Furthermore, the activity of MLK3-MKK7-JNKs signaling pathway, expression of Fas/FasL, and apoptosis were significantly lower in the pc-DNA3.1(+)-HBx group when treated with K252a, a known inhibitor of MLK3. CONCLUSIONS: Our results show that HBx induces apoptosis in NRK-52E cells and activates Fas/FasL via the MLK3-MKK7-JNK3-c-Jun signaling pathway.


Assuntos
Células Epiteliais/efeitos dos fármacos , Proteína Ligante Fas/agonistas , Vírus da Hepatite B/química , Transdução de Sinais/genética , Transativadores/farmacologia , Receptor fas/agonistas , Animais , Apoptose/efeitos dos fármacos , Carbazóis/farmacologia , Caspase 8/genética , Caspase 8/metabolismo , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Proteína Ligante Fas/genética , Proteína Ligante Fas/metabolismo , Regulação da Expressão Gênica , Alcaloides Indólicos/farmacologia , Túbulos Renais/citologia , Túbulos Renais/efeitos dos fármacos , Túbulos Renais/metabolismo , MAP Quinase Quinase 7/genética , MAP Quinase Quinase 7/metabolismo , MAP Quinase Quinase Quinases/genética , MAP Quinase Quinase Quinases/metabolismo , Proteína Quinase 10 Ativada por Mitógeno/genética , Proteína Quinase 10 Ativada por Mitógeno/metabolismo , Plasmídeos/química , Plasmídeos/metabolismo , Ratos , Transativadores/isolamento & purificação , Transfecção , Proteínas Virais Reguladoras e Acessórias , Receptor fas/genética , Receptor fas/metabolismo , MAP Quinase Quinase Quinase 11 Ativada por Mitógeno
15.
J Diabetes Res ; 2016: 1312705, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26962537

RESUMO

The relative contributions of the JNK subtypes in inflammatory ß-cell failure and apoptosis are unclear. The JNK protein family consists of JNK1, JNK2, and JNK3 subtypes, encompassing many different isoforms. INS-1 cells express JNK1α1, JNK1α2, JNK1ß1, JNK1ß2, JNK2α1, JNK2α2, JNK3α1, and JNK3α2 mRNA isoform transcripts translating into 46 and 54 kDa isoform JNK proteins. Utilizing Lentiviral mediated expression of shRNAs against JNK1, JNK2, or JNK3 in insulin-producing INS-1 cells, we investigated the role of individual JNK subtypes in IL-1ß-induced ß-cell apoptosis. JNK1 knockdown prevented IL-1ß-induced INS-1 cell apoptosis associated with decreased 46 kDa isoform JNK protein phosphorylation and attenuated Myc expression. Transient knockdown of Myc also prevented IL-1ß-induced apoptosis as well as caspase 3 cleavage. JNK2 shRNA potentiated IL-1ß-induced apoptosis and caspase 3 cleavage, whereas JNK3 shRNA did not affect IL-1ß-induced ß-cell death compared to nonsense shRNA expressing INS-1 cells. In conclusion, JNK1 mediates INS-1 cell death associated with increased Myc expression. These findings underline the importance of differentiated targeting of JNK subtypes in the development of inflammatory ß-cell failure and destruction.


Assuntos
Apoptose/efeitos dos fármacos , Células Secretoras de Insulina/efeitos dos fármacos , Insulina/metabolismo , Interleucina-1beta/farmacologia , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Animais , Caspase 3/metabolismo , Linhagem Celular Tumoral , Secreção de Insulina , Células Secretoras de Insulina/enzimologia , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Camundongos , Proteína Quinase 10 Ativada por Mitógeno/genética , Proteína Quinase 10 Ativada por Mitógeno/metabolismo , Proteína Quinase 8 Ativada por Mitógeno/genética , Proteína Quinase 9 Ativada por Mitógeno/genética , Proteína Quinase 9 Ativada por Mitógeno/metabolismo , Proteínas Proto-Oncogênicas c-myc/genética , Interferência de RNA , Ratos , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Transfecção
16.
Sci Rep ; 6: 21025, 2016 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-26868142

RESUMO

Three-kinase mitogen-activated protein kinase (MAPK) signaling cascades are present in virtually all eukaryotic cells. MAPK cascades are organized by scaffold proteins, which assemble cognate kinases into productive signaling complexes. Arrestin-3 facilitates JNK activation in cells, and a short 25-residue arrestin-3 peptide was identified as the critical JNK3-binding element. Here we demonstrate that this peptide also binds MKK4, MKK7, and ASK1, which are upstream JNK3-activating kinases. This peptide is sufficient to enhance JNK3 activity in cells. A homologous arrestin-2 peptide, which differs only in four positions, binds MKK4, but not MKK7 or JNK3, and is ineffective in cells at enhancing activation of JNK3. The arrestin-3 peptide is the smallest MAPK scaffold known. This peptide or its mimics can regulate MAPKs, affecting cellular decisions to live or die.


Assuntos
Ativadores de Enzimas , Proteína Quinase 10 Ativada por Mitógeno/metabolismo , Peptídeos , beta-Arrestina 1/química , beta-Arrestina 2/química , Animais , Células COS , Chlorocebus aethiops , Ativação Enzimática/efeitos dos fármacos , Ativadores de Enzimas/síntese química , Ativadores de Enzimas/química , Ativadores de Enzimas/farmacologia , Humanos , Proteína Quinase 10 Ativada por Mitógeno/genética , Peptídeos/síntese química , Peptídeos/química , Peptídeos/farmacologia
17.
J Immunol ; 194(8): 3852-60, 2015 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-25786685

RESUMO

Leishmania major is a parasite that resides and replicates in macrophages. We previously showed that the parasite enhanced CD40-induced Raf-MEK-ERK signaling but inhibited PI3K-MKK-p38MAPK signaling to proleishmanial effects. As Raf and PI3K have a Ras-binding domain but exert opposite effects on Leishmania infection, we examined whether Ras isoforms had differential roles in Leishmania infection. We observed that L. major enhanced N-Ras and H-Ras expression but inhibited K-Ras expression in macrophages. L. major infection enhanced N-Ras activity but inhibited H-Ras and K-Ras activity. TLR2 short hairpin RNA or anti-TLR2 or anti-lipophosphoglycan Abs reversed the L. major-altered N-Ras and K-Ras expressions. Pam3CSK4, a TLR2 ligand, enhanced N-Ras expression but reduced K-Ras expression, indicating TLR2-regulated Ras expression in L. major infection. Whereas N-Ras silencing reduced L. major infection, K-Ras and H-Ras silencing enhanced the infection both in macrophages in vitro and in C57BL/6 mice. BALB/c-derived macrophages transduced with lentivirally expressed N-Ras short hairpin RNA and pulsed with L. major-expressed MAPK10 enhanced MAPK10-specific Th1-type response. CD40-deficient mice primed with these macrophages had reduced L. major infection, accompanied by higher IFN-γ but less IL-4 production. As N-Ras is activated by Sos, a guanine nucleotide exchange factor, we modeled the N-Ras-Sos interaction and designed two peptides from their interface. Both the cell-permeable peptides reduced L. major infection in BALB/c mice but not in CD40-deficient mice. These data reveal the L. major-enhanced CD40-induced N-Ras activation as a novel immune evasion strategy and the potential for Ras isoform-targeted antileishmanial immunotherapy and immunoprophylaxis.


Assuntos
Antígenos CD40/imunologia , Regulação Enzimológica da Expressão Gênica/imunologia , Leishmania major/imunologia , Leishmaniose Cutânea/imunologia , Sistema de Sinalização das MAP Quinases/imunologia , Proteínas Monoméricas de Ligação ao GTP/imunologia , Animais , Antígenos CD40/genética , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/genética , Ativação Enzimática/imunologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Evasão da Resposta Imune/efeitos dos fármacos , Evasão da Resposta Imune/genética , Evasão da Resposta Imune/imunologia , Imunoterapia , Leishmaniose Cutânea/genética , Leishmaniose Cutânea/patologia , Leishmaniose Cutânea/prevenção & controle , Lipopeptídeos/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/genética , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Mutantes , Proteína Quinase 10 Ativada por Mitógeno/genética , Proteína Quinase 10 Ativada por Mitógeno/imunologia , Quinases de Proteína Quinase Ativadas por Mitógeno , Proteínas Monoméricas de Ligação ao GTP/genética , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/imunologia , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteínas Proto-Oncogênicas p21(ras)/imunologia , Proteína Son Of Sevenless de Drosófila/genética , Proteína Son Of Sevenless de Drosófila/imunologia , Células Th1/imunologia , Células Th1/patologia , Receptor 2 Toll-Like , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/imunologia
18.
Sci Rep ; 5: 8047, 2015 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-25623238

RESUMO

Three JNK isoforms, JNK1, JNK2, and JNK3 have been reported and unique biological function has been ascribed to each. It is unknown if selective inhibition of these isoforms would confer therapeutic or safety benefit. To probe JNK isoform function we designed JNK2/3 inhibitors that have >30-fold selectivity over JNK1. Utilizing site-directed mutagenesis and x-ray crystallography we identified L144 in JNK3 as a key residue for selectivity. To test whether JNK2/3 selective inhibitors protect human dopaminergic neurons against neurotoxin-induced mitochondrial dysfunction, we monitored reactive oxygen species (ROS) generation and mitochondrial membrane potential (MMP). The results showed that JNK2/3 selective inhibitors protected against 6-hydroxydopamine-induced ROS generation and MMP depolarization. These results suggest that it was possible to develop JNK2/3 selective inhibitors and that residues in hydrophobic pocket I were responsible for selectivity. Moreover, the findings also suggest that inhibition of JNK2/3 likely contributed to protecting mitochondrial function and prevented ultimate cell death.


Assuntos
Proteína Quinase 10 Ativada por Mitógeno/química , Proteína Quinase 9 Ativada por Mitógeno/química , Inibidores de Proteínas Quinases/química , Pirazóis/química , Sequência de Aminoácidos , Sítios de Ligação , Linhagem Celular Tumoral , Cristalografia por Raios X , Humanos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Proteína Quinase 10 Ativada por Mitógeno/genética , Proteína Quinase 10 Ativada por Mitógeno/metabolismo , Proteína Quinase 9 Ativada por Mitógeno/genética , Proteína Quinase 9 Ativada por Mitógeno/metabolismo , Simulação de Dinâmica Molecular , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Oxidopamina/farmacologia , Ligação Proteica , Isoformas de Proteínas/antagonistas & inibidores , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Inibidores de Proteínas Quinases/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Estrutura Terciária de Proteína , Pirazóis/metabolismo , Pirazóis/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/isolamento & purificação , Alinhamento de Sequência
19.
Proc Natl Acad Sci U S A ; 111(38): 13990-5, 2014 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-25201974

RESUMO

Although kainate receptors play important roles in ischemic stroke, the molecular mechanisms underlying postischemic regulation of kainate receptors remain unclear. In this study we demonstrate that Src family kinases contribute to the potentiation of kainate receptor function. Brain ischemia and reperfusion induce rapid and sustained phosphorylation of the kainate receptor subunit GluK2 by Src in the rat hippocampus, implicating a critical role for Src-mediated GluK2 phosphorylation in ischemic brain injury. The NMDA and kainate receptors are involved in the tyrosine phosphorylation of GluK2. GluK2 binds to Src, and the tyrosine residue at position 590 (Y590) on GluK2 is a major site of phosphorylation by Src kinases. GluK2 phosphorylation at Y590 is responsible for increases in whole-cell currents and calcium influx in response to transient kainate stimulation. In addition, GluK2 phosphorylation at Y590 facilitates the endocytosis of GluK2 subunits, and the activation of JNK3 and its substrate c-Jun after long-term kainate treatment. Thus, Src phosphorylation of GluK2 plays an important role in the opening of kainate receptor channels and downstream proapoptosis signaling after brain ischemia. The present study reveals an additional mechanism for the regulation of GluK2-containing kainate receptors by Src family kinases, which may be of pathological significance in ischemic stroke.


Assuntos
Isquemia Encefálica/metabolismo , Hipocampo/metabolismo , Receptores de Ácido Caínico/metabolismo , Transdução de Sinais/fisiologia , Acidente Vascular Cerebral/metabolismo , Regulação para Cima/fisiologia , Animais , Isquemia Encefálica/genética , Isquemia Encefálica/patologia , Células HEK293 , Hipocampo/patologia , Humanos , Masculino , Proteína Quinase 10 Ativada por Mitógeno/genética , Proteína Quinase 10 Ativada por Mitógeno/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-jun/genética , Proteínas Proto-Oncogênicas c-jun/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Ácido Caínico/genética , Acidente Vascular Cerebral/genética , Acidente Vascular Cerebral/patologia , Tirosina/genética , Tirosina/metabolismo , Quinases da Família src/genética , Quinases da Família src/metabolismo , Receptor de GluK2 Cainato
20.
Biochem Biophys Res Commun ; 452(3): 845-51, 2014 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-25218503

RESUMO

Exposure of human Jurkat T cells to JNK inhibitor IX (JNKi), targeting JNK2 and JNK3, caused apoptotic DNA fragmentation along with G2/M arrest, phosphorylation of Bcl-2, Mcl-1, and Bim, Δψm loss, and activation of Bak and caspase cascade. These JNKi-induced apoptotic events were abrogated by Bcl-2 overexpression, whereas G2/M arrest, cyclin B1 up-regulation, Cdk1 activation, and phosphorylation of Bcl-2 family proteins were sustained. In the concomitant presence of the G1/S blocking agent aphidicolin and JNKi, the cells underwent G1/S arrest and failed to induce all apoptotic events. The JNKi-induced phosphorylation of Bcl-2 family proteins and mitochondrial apoptotic events were suppressed by the Cdk1 inhibitor. Immunofluorescence microscopic analysis revealed that mitotic spindle defect and prometaphase arrest were the underlying factors for the G2/M arrest. These results demonstrate that JNKi-induced mitochondrial apoptosis was caused by microtubule damage-mediated prometaphase arrest, prolonged Cdk1 activation, and phosphorylation of Bcl-2 family proteins in Jurkat T cells.


Assuntos
Mitocôndrias/efeitos dos fármacos , Proteína Quinase 10 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 9 Ativada por Mitógeno/antagonistas & inibidores , Prometáfase/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Afidicolina/farmacologia , Apoptose/efeitos dos fármacos , Fragmentação do DNA/efeitos dos fármacos , Pontos de Checagem da Fase G1 do Ciclo Celular/efeitos dos fármacos , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Regulação da Expressão Gênica , Humanos , Células Jurkat , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Microtúbulos/efeitos dos fármacos , Mitocôndrias/metabolismo , Proteína Quinase 10 Ativada por Mitógeno/genética , Proteína Quinase 10 Ativada por Mitógeno/metabolismo , Proteína Quinase 9 Ativada por Mitógeno/genética , Proteína Quinase 9 Ativada por Mitógeno/metabolismo , Proteína de Sequência 1 de Leucemia de Células Mieloides/genética , Proteína de Sequência 1 de Leucemia de Células Mieloides/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA