Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Exp Neurol ; 341: 113688, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33713655

RESUMO

Increasing numbers of patients with spontaneous subarachnoid hemorrhage(SAH) who recover from surgery and intensive care management still live with cognitive impairment after discharge, indicating the importance of white matter injury at the acute stage of SAH. In the present study, standard endovascular perforation was employed to establish an SAH mouse model, and a microRNA (miRNA) chip was used to analyze the changes in gene expression in white matter tissue after SAH. The data indicate that 17 miRNAs were downregulated, including miR-706, miR-669a-5p, miR-669p-5p, miR-7116-5p and miR-195a-3p, while 13 miRNAs were upregulated, including miR-6907-5p, miR-5135, miR-6982-5p, miR-668-5p, miR-8119. Strikingly, miR-706 was significantly downregulated with the highest fold change. Further experiments confirmed that miR-706 could alleviate white matter injury and improve neurological behavior, at least partially by inhibiting the PKCα/MST1/NF-κB pathway and the release of inflammatory cytokines. These results might provide a deeper understanding of the pathophysiological processes in white matter after SAH, as well as potential therapeutic strategies for the translational research.


Assuntos
Fator de Crescimento de Hepatócito/antagonistas & inibidores , MicroRNAs/biossíntese , NF-kappa B/antagonistas & inibidores , Proteína Quinase C-alfa/antagonistas & inibidores , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Hemorragia Subaracnóidea/metabolismo , Substância Branca/metabolismo , Animais , Regulação para Baixo/fisiologia , Fator de Crescimento de Hepatócito/biossíntese , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/biossíntese , Proteína Quinase C-alfa/biossíntese , Proteínas Proto-Oncogênicas/biossíntese , Transdução de Sinais/fisiologia , Hemorragia Subaracnóidea/patologia , Hemorragia Subaracnóidea/prevenção & controle , Substância Branca/lesões , Substância Branca/patologia
2.
Biomed Pharmacother ; 127: 110117, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32244197

RESUMO

Anorectal malformations (ARMs) is one of the most common gastrointestinal anomalies. Previous research revealed that miR-92a-2-5p was upregulated in ARMs. However, the underlying roles remains unknown. The current study was to further investigate the spatiotemporal expression patterns of miR-92a-2-5p and its target gene protein kinase C alpha (PRKCA) predicted by bioinformatic method, and to explore their potential functions in anorectal malformations (ARMs). Rat models with ethylenethiourea-induced ARMs were made for subsequent experiments. Direct target relationship between miR-92a-2-5p and PRKCA was validated using a luciferase reporter assay. The spatiotemporal expression pattern of miR-92a-2-5p was evaluated using fluorescence in situ hybridization (FISH), while the expression of PRKCA was revealed by immunohistochemical staining and western blotting. IEC-6 cells were transfected with mimics/mimics NC (Negative control)/inhibitor/inhibitor NC of miR-92a-2-5p or si-PRKCA/si-PRKCA NC, respectively. Then the downstream molecules of miR-92a-2-5p, PRKCA and ß-catenin, were subsequently detected. Meanwhile, apoptosis and viability assays were measured. Dual luciferase assay confirmed the direct regulatory relationship between miR-92a-2-5p and PRKCA. FISH revealed that miR-92a-2-5p was expressed with a higher level in ARMs fetuses. Further analyses of PRKCA showed lower protein expression level in ARMs group, which was opposite to miR-92a-2-5p. In vitro experiments revealed that overexpression of miR-92a-2-5p or knockdown of PRKCA can down-regulate PRKCA, up-regulate and facilitate nuclear localization of ß-catenin, increase apoptosis and decrease proliferation of IEC-6. Taken together, these findings suggest that aberrantly high expression of miR-92a-2-5p potentially contribute to ARMs by inhibiting proliferation and enhancing apoptosis of intestinal cells via negatively regulating PRKCA/ß-catenin.


Assuntos
Malformações Anorretais/fisiopatologia , Apoptose/fisiologia , Proliferação de Células/fisiologia , MicroRNAs/fisiologia , beta Catenina/biossíntese , Animais , Malformações Anorretais/induzido quimicamente , Etilenotioureia , Feminino , MicroRNAs/biossíntese , Proteína Quinase C-alfa/biossíntese , Ratos , Regulação para Cima
3.
Clin Exp Pharmacol Physiol ; 47(3): 478-484, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31730233

RESUMO

The right ventricle (RV) enlargement and pulmonary fibrosis are involved in cor pulmonale. The role of miR-200b in cor pulmonale is less well understood. This study was designed to evaluate the regulatory roles of miR-200b in cor pulmonale. Cor pulmonary mouse model was built via monocrotaline injection of monocrotaline (MCT). The expression of miR-200b in the lungs, RV and left ventricle (LV) are using real-time polymerase chain reaction. The transthoracic echocardiography was employed to determine the effects of miR-200b mimics and Gö6976 injection on MCT mice. The protein levels of protein kinase C α (PKCα), collagen, and fibronectin in the lung, RV, and LV in the mice with and without miR-200b mimics and Gö6976 injection were evaluated using western blot. The expression of miR-200b decreased in MCT mice, while there was no difference in LV. Both the miR-200b mimics and Gö6976 injection reversed the muscularization in the pulmonary artery, reversed RV hypertrophy, reduced RV systolic pressure, wall thickness and pulmonary fibrosis. The injection of miR-200b can reduce the PKCα expression in the lung, RV, and LV. This study confirmed the down-regulation of miR-200b in cor pulmonale. The reverse effects of miR-200b in the present study may provide a potential tool for cor pulmonary treatment.


Assuntos
Modelos Animais de Doenças , Hipertensão Pulmonar/metabolismo , MicroRNAs/metabolismo , Proteína Quinase C-alfa/biossíntese , Doença Cardiopulmonar/metabolismo , Transdução de Sinais/fisiologia , Animais , Hipertensão Pulmonar/induzido quimicamente , Hipertensão Pulmonar/prevenção & controle , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Monocrotalina/toxicidade , Proteína Quinase C-alfa/antagonistas & inibidores , Doença Cardiopulmonar/induzido quimicamente , Doença Cardiopulmonar/prevenção & controle , Transdução de Sinais/efeitos dos fármacos
4.
J Cell Physiol ; 234(7): 10101-10110, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30422318

RESUMO

MicroRNAs (miRNAs) are closely related with the posttranscriptional regulation of gene expression. Our past study showed that let-7g-5p decreased during pregnancy and lactation in mouse mammary gland, what suggested the prospective regulating role of let-7g-5p in mammary epithelial cells. In this study, to unravel the role of let-7g-5p, we found PRKCA (PKC-alpha, PKC-α) might serve as potential targets of let-7g-5p by bioinformatics. Then let-7g-5p was knocked down by an antisense oligonucleotide in mouse primary mammary epithelial cells. As predicted, PRKCA gene expression and mammary epithelial cells growth were increased by anti-let-7g-5p regulation in vitro. By using reporter constructs, it showed that the let-7g-5p could direct binding with 3'-the untranslated regions (3'-UTR) of PRKCA mRNA. Furthermore, ectopic overexpression of let-7g-5p in vivo reduced PRKCA and ß-casein protein expression as well as inhibited mammary gland growth. These results suggested that let-7g-5p could inhibit the mammary epithelial cells differentiation and ß-casein protein synthesis and expression through suppression of PRKCA on the cycle of mammary cell differentiation and development and that let-7g-5p may be a novel important regulated target in mammary cells function.


Assuntos
Diferenciação Celular/genética , Regulação da Expressão Gênica/fisiologia , Glândulas Mamárias Animais/citologia , MicroRNAs/metabolismo , Proteína Quinase C-alfa/biossíntese , Animais , Feminino , Glândulas Mamárias Animais/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , MicroRNAs/genética , Proteína Quinase C-alfa/genética
5.
Biomed Pharmacother ; 100: 467-477, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29477910

RESUMO

A series of cardiovascular complications associated with hyperglycemia is a critical threat to the diabetic population. Here we elucidate the link between hyperglycemia and cardiovascular diseases onset, focusing on oxidative stress and associated cardiac dysfunctions. The contribution of advanced glycation end products (AGE) and protein kinase C (PKC) signaling is extensively studied. For induction of hyperglycemia, H9c2 cells were incubated with 33 mM glucose for 48 h to simulate the diabetic condition in in vitro system. Development of cardiac dysfunction was confirmed with the significant increase of lactate dehydrogenase (LDH) release to the medium and associated decrease in cell viability. Various parameters like free radical generation, alteration in innate antioxidant system, lipid peroxidation, AGE production and PKC α -ERK axis were investigated during hyperglycemia and with chlorogenic acid. Hyperglycemia has significantly enhanced reactive oxygen species (ROS- 4 fold) generation, depleted SOD activity (1.3 fold) and expression of enzymes particularly CuZnSOD (SOD1) and MnSOD (SOD2), increased production of AGE (2.18 fold). Besides, PKC α dependent ERK signaling pathway was found activated (1.43 fold) leading to cardiac dysfunction during hyperglycemia. Chlorogenic acid (CA) was found beneficial against hyperglycemia most probably through its antioxidant mediated activity. The outcome of this preliminary study reveals the importance of integrated approach emphasizing redox status, glycation and signaling pathways like PKC α - ERK axis for control and management of diabetic cardiomyopathy (DCM) and potential of bioactives like CA.


Assuntos
Antioxidantes/farmacologia , Ácido Clorogênico/farmacologia , Glucose/toxicidade , Proteína Quinase C-alfa/biossíntese , Regulação para Cima/efeitos dos fármacos , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Glicosilação/efeitos dos fármacos , Proteína Quinase C-alfa/antagonistas & inibidores , Ratos , Espécies Reativas de Oxigênio/metabolismo , Regulação para Cima/fisiologia
6.
Br J Pharmacol ; 175(3): 456-468, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29139546

RESUMO

BACKGROUND AND PURPOSE: Haem oxygenase-1 (HO-1) could provide cytoprotection against various inflammatory diseases. However, the mechanisms underlying the protective effect of CO-releasing molecule-2 (CORM-2)-induced HO-1 expression against TNF-α-induced inflammatory responses in human pulmonary alveolar epithelial cells (HPAEpiCs) remain unknown. EXPERIMENTAL APPROACH: CORM-2-induced HO-1 protein and mRNA expression, and signalling pathways were determined by Western blot and real-time PCR, coupled with respective pharmacological inhibitors or transfection with siRNAs. The effect of CORM-2 on TNF-α-induced increase in leukocyte counts in BAL fluid and VCAM-1 expression in lung was determined by cell counting and Western blot analysis. KEY RESULTS: CORM-2 attenuated the TNF-α-induced pulmonary haematoma, VCAM-1 expression and increase in leukocytes through an up-regulation of HO-1 in mice; this effect of CORM-2 was reversed by the HO-1 inhibitor zinc protoporphyrin IX. Furthermore, CORM-2 increased HO-1 protein and mRNA expression as well as the phosphorylation of PYK2, PKCα and ERK1/2 (p44/p42 MAPK) in HPAEpiCs; these effects were attenuated by their respective pharmacological inhibitors or transfection with siRNAs. Inhibition of PKCα by Gö6976 or Gö6983 attenuated CORM-2-induced stimulation of PKCα and ERK1/2 phosphorylation but had no effect on PYK2 phosphorylation. Moreover, inhibition of PYK2 by PF431396 reduced the phosphorylation of all three protein kinases. Finally, PYK2/PKCα/ERK1/2-mediated stimulation of activator protein 1 was shown to play a key role in CORM-2-induced HO-1 expression via an up-regulation of c-Fos mRNA. CONCLUSIONS AND IMPLICATIONS: CORM-2 activates a PYK2/PKCα/ERK1/2/AP-1 pathway leading to HO-1 expression in HPAEpiCs. This HO-1/CO system might have potential as a therapeutic target in pulmonary inflammation.


Assuntos
Quinase 2 de Adesão Focal/biossíntese , Heme Oxigenase-1/biossíntese , Compostos Organometálicos/farmacologia , Pneumonia/metabolismo , Proteína Quinase C-alfa/biossíntese , Fator de Necrose Tumoral alfa/toxicidade , Animais , Linhagem Celular , Humanos , Masculino , Camundongos , Camundongos Endogâmicos ICR , Compostos Organometálicos/uso terapêutico , Pneumonia/induzido quimicamente , Pneumonia/prevenção & controle , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Regulação para Cima/fisiologia
7.
Mol Cell Biochem ; 425(1-2): 103-112, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27837432

RESUMO

Orexin-A, which is an endogenous neuropeptide, is reported to have a protective role in ischemic stroke. High-concentration glutamic acid (Glu) induced by hypoxia injury in ischemic stroke can be inhibited by glial glutamate transporter GLT-1 which is only expressed in astroglia cells. A previous study reported that Orexin-A may regulate GLT-1 expression. However, the role of orexin-A in the regulation of GLT-1 in ischemic stroke still remains unclear. In this study, we aimed to investigate the effect and the underlying mechanism of orexin-A on Glu uptake in astrocytes in vitro and this effect on protecting the neurons from anoxia/hypoglycemic injury. The expression of GLT-1 significantly increased in the astrocytes with orexin-A treatment under anoxia/hypoglycemic conditions, promoting the uptake of Glu and inhibiting the apoptosis of co-cultured cells of astrocytes and neurons. However, these effects were significantly weakened by treatment with orexin-A receptor 1 (OX1R) antagonist. Orexin-A significantly up-regulated the expressions of PKCα and ERK1/2 under anoxia/hypoglycemic conditions in astrocytes, whereas the OX1R antagonist markedly reversed the effect. Furthermore, PKCα or ERK1/2 inhibitor significantly constrained the GLT-1 expression in astrocytes and facilitated the apoptosis of co-cultured cells, and GLT-1 overexpression could reverse those effects of PKCα or ERK1/2 inhibitor. Taken together, orexin-A promoted the GLT-1 expression via OX1R/PKCα/ERK1/2 pathway in astrocytes and protected co-cultured cells against anoxia/hypoglycemic injury.


Assuntos
Apoptose/efeitos dos fármacos , Astrócitos/metabolismo , Transportador 2 de Aminoácido Excitatório/biossíntese , Ácido Glutâmico/metabolismo , Hipoglicemia/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteína Quinase 3 Ativada por Mitógeno/sangue , Neurônios/metabolismo , Receptores de Orexina/metabolismo , Orexinas/farmacologia , Proteína Quinase C-alfa/biossíntese , Animais , Hipóxia Celular/efeitos dos fármacos , Células Cultivadas , Técnicas de Cocultura , Regulação da Expressão Gênica/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley
8.
Sci Rep ; 6: 33717, 2016 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-27644318

RESUMO

Chemoresistant mechanisms in T-cell acute lymphoblastic leukemia (T-ALL) patients are not clarified. The apoptotic signaling mediated by receptor of activated C kinase 1 (Rack1), protein kinase C (PKC) and FEM1 homolog b (FEM1b) was investigated in two T-ALL-derived cell lines (Jurkat and CCRF-CEM) following treatment with chemotherapy drugs vincristine and prednisone. Serum starvation or chemotherapeutic drugs significantly reduced Rack1 level and PKC activation, while promoted cellular apoptosis in both cell lines. Rack1 overexpression protected T-ALL cell against starvation or chemotherapeutic drug-induced apoptosis. Moreover, Rack1 overexpression reduced the level of cytochrome c and active caspase 3 as well as FEM1b and apoptotic protease activating factor-1 (Apaf-1), and inhibited induction of cellular apoptosis in chemotherapeutic drug-treated Jurkat cell. Interaction of Rack1 and PKCα, not PKCß, was detected in both cell lines. Of note, Rack1 overexpression abrogated reduction of PKC kinase activity in chemotherapeutic drug-treated T-ALL cell. PKC kinase inhibitor Go6976 or siPKCα inhibited downregulation of FEM1b and/or Apaf-1, and thus increased cellular apoptosis in Rack1-overexpressed T-ALL cell receiving chemotherapeutic drugs. Accordingly, our data provided evidence that increased Rack1-mediated upregulation of PKC kinase activity may be responsible for the development of chemoresistance in T-ALL-derived cell line potentially by reducing FEM1b and Apaf-1 level.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Regulação Enzimológica da Expressão Gênica , Regulação Leucêmica da Expressão Gênica , Proteínas de Neoplasias/biossíntese , Leucemia-Linfoma Linfoblástico de Células T Precursoras/enzimologia , Proteína Quinase C-alfa/biossíntese , Receptores de Quinase C Ativada/biossíntese , Fator Apoptótico 1 Ativador de Proteases/genética , Fator Apoptótico 1 Ativador de Proteases/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Humanos , Células Jurkat , Proteínas de Neoplasias/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/tratamento farmacológico , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patologia , Prednisolona/farmacologia , Proteína Quinase C-alfa/genética , Receptores de Quinase C Ativada/genética , Vincristina/farmacologia
9.
Oncogene ; 35(14): 1785-96, 2016 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-26234676

RESUMO

Accumulating evidence suggests that changes of the protein synthesis machinery alter translation of specific mRNAs and participate in malignant transformation. Here we show that protein kinase C α (PKCα) interacts with TRM61, the catalytic subunit of the TRM6/61 tRNA methyltransferase. The TRM6/61 complex is known to methylate the adenosine 58 of the initiator methionine tRNA (tRNAi(Met)), a nuclear post-transcriptional modification associated with the stabilization of this crucial component of the translation-initiation process. Depletion of TRM6/61 reduced proliferation and increased death of C6 glioma cells, effects that can be partially rescued by overexpression of tRNAi(Met). In contrast, elevated TRM6/61 expression regulated the translation of a subset of mRNAs encoding proteins involved in the tumorigenic process and increased the ability of C6 cells to form colonies in soft agar or spheres when grown in suspension. In TRM6/61/tRNAi(Met)-overexpressing cells, PKCα overexpression decreased tRNAi(Met) expression and both colony- and sphere-forming potentials. A concomitant increase in TRM6/TRM61 mRNA and tRNAi(Met) expression with decreased expression of PKCα mRNA was detected in highly aggressive glioblastoma multiforme as compared with Grade II/III glioblastomas, highlighting the clinical relevance of our findings. Altogether, we suggest that PKCα tightly controls TRM6/61 activity to prevent translation deregulation that would favor neoplastic development.


Assuntos
Biomarcadores Tumorais/biossíntese , Glioblastoma/genética , Proteína Quinase C-alfa/genética , tRNA Metiltransferases/biossíntese , Apoptose/genética , Biomarcadores Tumorais/genética , Carcinogênese/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica , Glioblastoma/patologia , Humanos , Metionina/genética , Proteína Quinase C-alfa/biossíntese , RNA de Transferência/genética , tRNA Metiltransferases/genética
10.
Int J Oncol ; 47(5): 1839-44, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26398575

RESUMO

There is an ongoing search for plant-derived sesquiterpenes, particularly for those with anticancer activity against human cancer cells. The sesquiterpene ineupatorolide B (InB), isolated from the Inula cappa, showed potent growth-inhibitory activity against HeLa cells but less activity against MM1-CB melanoma cells. Staining by terminal deoxynucleotidyl transferase dUTP nick-end labeling method revealed that this activity was, at least in part, due to the induction of apoptosis. The activities of major transcription factors were examined by using a luciferase reporter assay. The results showed that the transactivation ability of nuclear factor of activated T-cell (NFAT) was enhanced. The activation of NFAT by InB was largely suppressed by preincubation with protein kinase C (PKC) inhibitors such as staurosporine and K252a. Western blot analysis revealed that the the levels of phosphorylated PKCα, but not other subtypes, increased after treatment with InB. Knockdown of PKCα using siRNA attenuated the cytotoxic activity of InB. Thus, InB may exhibit growth-inhibitory activity through the activation of PKCα, followed by an increase in NFAT transactivation ability.


Assuntos
Fatores de Transcrição NFATC/genética , Proteína Quinase C-alfa/biossíntese , Sesquiterpenos/administração & dosagem , Neoplasias do Colo do Útero/genética , Apoptose/efeitos dos fármacos , Feminino , Células HeLa , Humanos , Inula/química , Fatores de Transcrição NFATC/biossíntese , Fosforilação , Proteína Quinase C-alfa/genética , RNA Interferente Pequeno , Neoplasias do Colo do Útero/patologia
11.
Invest Ophthalmol Vis Sci ; 56(8): 4961-74, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26230760

RESUMO

PURPOSE: Protein kinase C α (PKCα) is abundantly expressed in rod bipolar cells (RBCs) in the retina, yet the physiological function of PKCα in these cells is not well understood. To elucidate the role of PKCα in visual processing in the eye, we examined the effect of genetic deletion of PKCα on the ERG and on RBC light responses in the mouse. METHODS: Immunofluorescent labeling was performed on wild-type (WT), TRPM1 knockout, and PKCα knockout (PKC-KO) retina. Scotopic and photopic ERGs were recorded from WT and PKC-KO mice. Light responses of RBCs were measured using whole-cell recordings in retinal slices from WT and PKC-KO mice. RESULTS: Protein kinase C alpha expression in RBCs is correlated with the activity state of the cell. Rod bipolar cells dendrites are a major site of PKCα phosphorylation. Electroretinogram recordings indicated that loss of PKCα affects the scotopic b-wave, including a larger peak amplitude, longer implicit time, and broader width of the b-wave. There were no differences in the ERG a- or c-wave between PKCα KO and WT mice, indicating no measurable effect of PKCα in photoreceptors or the RPE. The photopic ERG was unaffected consistent with the lack of detectable PKCα in cone bipolar cells. Whole-cell recordings from RBCs in PKC-KO retinal slices revealed that, compared with WT, RBC light responses in the PKC-KO retina are delayed and of longer duration. CONCLUSIONS: Protein kinase C alpha plays an important modulatory role in RBCs, regulating both the peak amplitude and temporal properties of the RBC light response in the rod visual pathway.


Assuntos
DNA/genética , Regulação da Expressão Gênica , Proteína Quinase C-alfa/genética , Células Bipolares da Retina/enzimologia , Doenças Retinianas/genética , Células Fotorreceptoras Retinianas Bastonetes/enzimologia , Vias Visuais/enzimologia , Animais , Western Blotting , Modelos Animais de Doenças , Eletrorretinografia , Terapia Genética/métodos , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Técnicas de Patch-Clamp , Proteína Quinase C-alfa/biossíntese , Células Bipolares da Retina/patologia , Doenças Retinianas/enzimologia , Doenças Retinianas/fisiopatologia , Células Fotorreceptoras Retinianas Bastonetes/patologia , Vias Visuais/fisiopatologia
12.
Tumour Biol ; 36(11): 8913-24, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26076811

RESUMO

Cancer cells are characterized by increased production of reactive oxygen species (ROS) and an altered redox environment as compared to normal cells. Continuous accumulation of ROS triggers oxidative stress leading to hyper-activation of signaling pathways that promote cell proliferation, survival, and metabolic adaptation to the tumor microenvironment. Therefore, antioxidants are proposed to contribute to cancer prevention. Protein kinase C (PKC) is a crucial regulator of diverse cellular processes and contributes to cancer progression. The activation of PKC is partially dependent on ROS signaling. In the present study, cancer preventive activity of natural flavonoid quercetin is analyzed in ascite cells of Dalton's lymphoma-bearing mice. The total ROS level and activity of PKC were downregulated after quercetin treatment in lymphoma-bearing mice. Quercetin modulates the expression of almost all isozymes of classical, novel, and atypical PKC as well as downregulates the level and expression of PKCα. Further, quercetin improves apoptotic potential, as observed by the levels of caspase 3, caspase 9, PARP, PKCδ, and nuclear condensation. Additionally, quercetin reduces cell survival and promotes death receptor-mediated apoptosis via differential localization of the TNFR1 level in ascite cells. The overall result suggests the cancer preventive activity of quercetin via the induction of apoptosis and modulates PKC signaling with the reduction of oxidative stress in ascite cells of lymphoma-bearing mice.


Assuntos
Linfoma/tratamento farmacológico , Proteína Quinase C-alfa/biossíntese , Quercetina/administração & dosagem , Receptores Tipo I de Fatores de Necrose Tumoral/biossíntese , Animais , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica , Humanos , Isoenzimas/biossíntese , Linfoma/genética , Linfoma/patologia , Camundongos , Estresse Oxidativo/efeitos dos fármacos , Proteína Quinase C-alfa/genética , Espécies Reativas de Oxigênio/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Transdução de Sinais/efeitos dos fármacos
13.
PLoS One ; 10(5): e0127420, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26010542

RESUMO

In this study, the molecular mechanism of protein kinase C alpha (PKCα) gene regulation in hepatocellular carcinoma (HCC) involving Ets-like protein-1 (Elk-1) and myeloid zinc finger-1 (MZF-1) was investigated. The luciferase reporter assay results revealed that the presence of both MZF-1 and Elk-1 significantly contributed to the upregulation of PKCα gene transcription activity, and the transcriptional activity decreased when the transfection included a DNA-binding-deficient (∆DBD) gene vector of either MZF-1 or Elk-1 DNA-binding deficiency (MZF-1∆DBD or Elk-1∆DBD), thereby indicating that the enhanced expression of PKCα was caused by the binding of MZF-1 and/or Elk-1 with the PKCα promoter. We investigated MZF-1 and Elk-1 to determine whether they bind to each other. The results of immunoprecipitation (IP), Co-IP, chromatin IP (ChIP), and Re-ChIP analyses indicated that Elk-1 can directly bind to the N-terminal region of MZF-1 and MZF-1 can directly bind to the C-terminal region of Elk-1 to form a complex before attaching to the PKCα promoter. Furthermore, when MZF-1∆DBD or Elk-1∆DBD was added to the cells, PKCα expression decreased, and cell proliferation, migration, invasion, and tumorigenicity also decreased. These findings suggest that PKCα expression in HCC could be stimulated by the formation of MZF-1/Elk-1 complex, which directly binds to the PKCα promoter.


Assuntos
Carcinoma Hepatocelular/metabolismo , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Fatores de Transcrição Kruppel-Like/metabolismo , Neoplasias Hepáticas/metabolismo , Proteína Quinase C-alfa/biossíntese , Elementos de Resposta , Proteínas Elk-1 do Domínio ets/metabolismo , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Células Hep G2 , Humanos , Fatores de Transcrição Kruppel-Like/genética , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Proteína Quinase C-alfa/genética , Estrutura Terciária de Proteína , Transcrição Gênica , Regulação para Cima , Proteínas Elk-1 do Domínio ets/genética
14.
BMC Cancer ; 15: 326, 2015 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-25924946

RESUMO

BACKGROUND: The local invasion of tumor cells into the surrounding tissue is the first and most critical step of the metastatic cascade. Cells can invade either collectively, or individually. Individual cancer cell invasion can occur in the mesenchymal or amoeboid mode, which are mutually interchangeable. This plasticity of individual cancer cell invasiveness may represent an escape mechanism for invading cancer cells from anti-metastatic treatment. METHODS: To identify new signaling proteins involved in the plasticity of cancer cell invasiveness, we performed proteomic analysis of the amoeboid to mesenchymal transition with A375m2 melanoma cells in a 3D Matrigel matrix. RESULTS: In this screen we identified PKCα as an important protein for the maintenance of amoeboid morphology. We found that the activation of PKCα resulted in the mesenchymal-amoeboid transition of mesenchymal K2 and MDA-MB-231 cell lines. Consistently, PKCα inhibition led to the amoeboid-mesenchymal transition of amoeboid A375m2 cells. Next, we showed that PKCα inhibition resulted in a considerable decrease in the invading abilities of all analyzed cancer cell lines. CONCLUSIONS: Our results suggest that PKCα is an important protein for maintenance of the amoeboid morphology of cancer cells, and that downregulation of PKCα results in the amoeboid to mesenchymal transition. Our data also suggest that PKCα is important for both mesenchymal and amoeboid invasiveness, making it an attractive target for anti-metastatic therapies.


Assuntos
Melanoma/genética , Mesoderma/metabolismo , Invasividade Neoplásica/genética , Proteína Quinase C-alfa/biossíntese , Linhagem Celular Tumoral , Movimento Celular/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Melanoma/patologia , Mesoderma/patologia , Invasividade Neoplásica/patologia , Proteína Quinase C-alfa/genética , Proteômica , Transdução de Sinais
15.
Protein Expr Purif ; 110: 14-21, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25582765

RESUMO

We report the protocol for heterologous expression and purification of the N-terminal regulatory region of two Protein Kinase C (PKC)(1) isozymes, one conventional and one novel. Previous studies of these domains relied almost exclusively on the fusion constructs with high-molecular-weight solubility fusion partners such as GST and MBP. We developed experimental procedures that enabled us to overcome challenges associated with the amphiphilic character of the regulatory domain and generate sufficient quantities of fusion partner-free proteins for biophysical work. The key features of the protocol are the identity of the cleavable fusion partner, expression conditions, growth medium additives, introduction of mutation/solubility tags, and incorporation of osmolytes. The protein yields are sufficient for cost-effective production of isotopically enriched proteins for NMR work and biophysical studies in general. Our work opens up an avenue for the structural studies of these challenging proteins with high amphiphilic character.


Assuntos
Proteína Quinase C-alfa/isolamento & purificação , Proteína Quinase C-delta/isolamento & purificação , Proteína Quinase C/isolamento & purificação , Sequência de Aminoácidos , Animais , Clonagem Molecular , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Ressonância Magnética Nuclear Biomolecular , Concentração Osmolar , Plasmídeos/química , Plasmídeos/metabolismo , Proteína Quinase C/biossíntese , Proteína Quinase C/genética , Proteína Quinase C-alfa/biossíntese , Proteína Quinase C-alfa/genética , Proteína Quinase C-delta/biossíntese , Proteína Quinase C-delta/genética , Estrutura Terciária de Proteína , Ratos , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/isolamento & purificação , Alinhamento de Sequência
16.
PLoS One ; 9(10): e108963, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25330241

RESUMO

Chronic hyperglycemia induces insulin resistance by mechanisms that are incompletely understood. One model of hyperglycemia-induced insulin resistance involves chronic preincubation of adipocytes in the presence of high glucose and low insulin concentrations. We have previously shown that the mTOR complex 1 (mTORC1) plays a partial role in the development of insulin resistance in this model. Here, we demonstrate that treatment with Go-6976, a widely used "specific" inhibitor of cPKCs, alleviates hyperglycemia-induced insulin resistance. However, the effects of mTOR inhibitor, rapamycin and Go-6976 were not additive and only rapamycin restored impaired insulin-stimulated AKT activation. Although, PKCα, (but not -ß) was abundantly expressed in these adipocytes, our studies indicate cPKCs do not play a major role in causing insulin-resistance in this model. There was no evidence of changes in the expression or phosphorylation of PKCα, and PKCα knock-down did not prevent the reduction of insulin-stimulated glucose transport. This was also consistent with lack of IRS-1 phosphorylation on Ser-24 in hyperglycemia-induced insulin-resistant adipocytes. Treatment with Go-6976 did inhibit a component of the mTORC1 pathway, as evidenced by decreased phosphorylation of S6 ribosomal protein. Raptor knock-down enhanced the effect of insulin on glucose transport in insulin resistant adipocytes. Go-6976 had the same effect in control cells, but was ineffective in cells with Raptor knock-down. Taken together these findings suggest that Go-6976 exerts its effect in alleviating hyperglycemia-induced insulin-resistance independently of cPKC inhibition and may target components of the mTORC1 signaling pathway.


Assuntos
Adipócitos/efeitos dos fármacos , Carbazóis/administração & dosagem , Hiperglicemia/genética , Resistência à Insulina/genética , Insulina/metabolismo , Células 3T3-L1 , Adipócitos/enzimologia , Animais , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Hiperglicemia/patologia , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Complexos Multiproteicos/genética , Proteína Quinase C-alfa/biossíntese , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/genética
17.
Oncotarget ; 5(14): 5234-45, 2014 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-25026300

RESUMO

Pediatric T-cell Acute Lymphoblastic Leukemia (T-ALL) outcome has improved in the last decades, yet one patient in every four still relapses. Except treatment response and immunophenotype, few markers are reliably prognostic in pediatric T-ALL patients. Aiming to improve T-ALL risk stratification, we investigated a new candidate biomarker with potential prognostic relevance. A phosphoproteomic screening of 98 pediatric T-ALL samples at diagnosis had been performed using the high-throughput Reverse Phase Protein Arrays technique, which led to the identification of PKCαS657 as an activated protein with a broad variation among T-ALL samples. To evaluate PKCα potential as a prognostic biomarker, PKCα expression was analyzed using RQ-PCR in a cohort of 173 patients, representative of ALL2000-ALLR2006 AIEOP study. A threshold of PKCα expression with the highest discrimination for incidence of relapse was identified. Patients with PKCα down-regulation, compared to patients with PKCα levels above the threshold, presented a markedly increased cumulative incidence of relapse (43.8% vs. 10.9%, P<0.001), as well as a worse 4-year overall survival (66% vs. 87.9%, P=0.002) and event-free survival (53.1% vs. 85.2%, P=0.002). In particular, low PKCα expression identified cases with extremely poor outcome within the high-risk minimal residual disease (MRD) stratum, their incidence of relapse being of 69% vs. 15% in the high PKCα levels group. In a multivariate analysis adjusting for main prognostic features, PKCα proved to be an independent prognostic factor related to incidence of relapse. Very high risk patients within the high-risk MRD stratum, identified by PKCα expression, could be proposed for experimental therapeutic protocols.


Assuntos
Leucemia-Linfoma Linfoblástico de Células T Precursoras/enzimologia , Proteína Quinase C-alfa/biossíntese , Adolescente , Biomarcadores Tumorais/genética , Criança , Pré-Escolar , Feminino , Humanos , Lactente , Masculino , Neoplasia Residual , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Prognóstico , Proteína Quinase C-alfa/genética , Proteína Quinase C-alfa/metabolismo , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Fatores de Risco , Transdução de Sinais , Taxa de Sobrevida , Resultado do Tratamento
18.
Biochim Biophys Acta ; 1843(4): 735-45, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24412308

RESUMO

In the present study, we report that somatostatin receptor 2 (SSTR2) plays a crucial role in modulation of ß1AR and ß2AR mediated signaling pathways that are associated with increased intracellular Ca(2+) and cardiac complications. In H9c2 cells, SSTR2 colocalizes with ß1AR or ß2AR in receptor specific manner. SSTR2 selective agonist inhibits isoproterenol and formoterol stimulated cAMP formation and PKA phosphorylation in concentration dependent manner. In the presence of SSTR2 agonist, the expression of PKCα and PKCß was comparable to the basal condition, however SSTR2 agonist inhibits isoproterenol or formoterol induced PKCα and PKCß expression, respectively. Furthermore, the activation of SSTR2 not only inhibits calcineurin expression and its activity, but also blocks NFAT dephosphorylation and its nuclear translocation. SSTR2 selective agonist abrogates isoproterenol mediated increase in cell size and protein content (an index of hypertrophy). Taken together, the results described here provide direct evidence in support of cardiac protective role of SSTR2 via modulation of Ca(2+) associated signaling pathways attributed to cardiac hypertrophy.


Assuntos
Cálcio/metabolismo , AMP Cíclico/metabolismo , Fosforilação/efeitos dos fármacos , Receptores Adrenérgicos beta 1/metabolismo , Receptores de Somatostatina/metabolismo , Animais , Linhagem Celular , Etanolaminas/farmacologia , Fumarato de Formoterol , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Isoproterenol/farmacologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Proteína Quinase C beta/biossíntese , Proteína Quinase C beta/metabolismo , Proteína Quinase C-alfa/biossíntese , Proteína Quinase C-alfa/metabolismo , Ratos , Receptores Adrenérgicos beta 2/metabolismo , Receptores de Somatostatina/agonistas , Transdução de Sinais/efeitos dos fármacos , Somatostatina/agonistas , Somatostatina/metabolismo
19.
Cell Signal ; 26(3): 580-93, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24316392

RESUMO

Although PLCε has been verified to enhance bladder cancer cell invasion, the signaling pathways responsible for this remain elusive. Protein kinase C (PKCα/ß), which is involved in cancer development and progression, has been demonstrated to be activated by PLCε. However, the roles of PKCα/ß in PLCε-mediated bladder carcinoma cell invasion and migration have not been clearly identified. In this study, to determine what role PKCα/ß plays in PLCε-mediated bladder cancer cell invasion and migration, we silenced PLCε gene by adenovirus-shPLCε in T24 and BIU-87 cells and then revealed that it significantly inhibited cell migration and invasion. Further research indicated that cell bio-function of PLCε-regulated was related with PKCα/ß activity. These in vitro findings were supported by data from bladder carcinoma patient samples. In 35 case bladder cancer tumor samples, PLCε-overexpressing tumors showed significantly higher positive rates of PKCα/ß membrane immunohistochemistry staining than PLCε-low-expressing tumors. Mechanistically, study further showed that PLCε knockdown gene induced E-cadherin expression and decreased TBX3 expression, both of which were dependent on PKCα/ß activity. In addition, we demonstrated that treatment cells with TBX3-specific shorting hairpin RNA (shRNA) up-regulated E-cadherin expression and inhibited cell invasion/migration. Moreover, in in vivo experiment, immunohistochemistry analysis of Ad-shPLCε-infected tumor tissue showed low expression levels of phospho-PKCα/ß and TBX3 and high expression levels of E-cadherin compared with those of the control group. In summary, our findings uncover that PKCα/ß is critical for PLCε-mediated cancer cell invasion and migration and provide valuable insights for current and future Ad-shPLCε and PKCα/ß clinical trials.


Assuntos
Fosfoinositídeo Fosfolipase C/genética , Proteína Quinase C beta/metabolismo , Proteína Quinase C-alfa/metabolismo , Proteínas com Domínio T/biossíntese , Animais , Caderinas/biossíntese , Carbazóis/farmacologia , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células , Humanos , Camundongos , Camundongos Nus , Invasividade Neoplásica/genética , Transplante de Neoplasias , Proteína Quinase C beta/antagonistas & inibidores , Proteína Quinase C beta/biossíntese , Proteína Quinase C-alfa/antagonistas & inibidores , Proteína Quinase C-alfa/biossíntese , Interferência de RNA , RNA Interferente Pequeno , Proteínas com Domínio T/genética , Transplante Heterólogo
20.
Endocrinology ; 154(9): 3197-208, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23766132

RESUMO

Corticosterone (CORT) and other glucocorticoids cause peripheral insulin resistance and compensatory increases in ß-cell mass. A prolonged high-fat diet (HFD) induces insulin resistance and impairs ß-cell insulin secretion. This study examined islet adaptive capacity in rats treated with CORT and a HFD. Male Sprague-Dawley rats (age ∼6 weeks) were given exogenous CORT (400 mg/rat) or wax (placebo) implants and placed on a HFD (60% calories from fat) or standard diet (SD) for 2 weeks (N = 10 per group). CORT-HFD rats developed fasting hyperglycemia (>11 mM) and hyperinsulinemia (∼5-fold higher than controls) and were 15-fold more insulin resistant than placebo-SD rats by the end of ∼2 weeks (Homeostatic Model Assessment for Insulin Resistance [HOMA-IR] levels, 15.08 ± 1.64 vs 1.0 ± 0.12, P < .05). Pancreatic ß-cell function, as measured by HOMA-ß, was lower in the CORT-HFD group as compared to the CORT-SD group (1.64 ± 0.22 vs 3.72 ± 0.64, P < .001) as well as acute insulin response (0.25 ± 0.22 vs 1.68 ± 0.41, P < .05). Moreover, ß- and α-cell mass were 2.6- and 1.6-fold higher, respectively, in CORT-HFD animals compared to controls (both P < .05). CORT treatment increased p-protein kinase C-α content in SD but not HFD-fed rats, suggesting that a HFD may lower insulin secretory capacity via impaired glucose sensing. Isolated islets from CORT-HFD animals secreted more insulin in both low and high glucose conditions; however, total insulin content was relatively depleted after glucose challenge. Thus, CORT and HFD, synergistically not independently, act to promote severe insulin resistance, which overwhelms islet adaptive capacity, thereby resulting in overt hyperglycemia.


Assuntos
Diabetes Mellitus Tipo 2/etiologia , Dieta Hiperlipídica/efeitos adversos , Glucocorticoides/efeitos adversos , Hiperglicemia/etiologia , Hiperinsulinismo/etiologia , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Animais , Ritmo Circadiano/efeitos dos fármacos , Corticosterona/administração & dosagem , Corticosterona/efeitos adversos , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Diabetes Mellitus Tipo 2/fisiopatologia , Modelos Animais de Doenças , Implantes de Medicamento , Indução Enzimática/efeitos dos fármacos , Glucocorticoides/administração & dosagem , Imuno-Histoquímica , Insulina/sangue , Resistência à Insulina , Secreção de Insulina , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/patologia , Masculino , Fosforilação/efeitos dos fármacos , Proteína Quinase C-alfa/biossíntese , Proteína Quinase C-alfa/metabolismo , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Índice de Gravidade de Doença
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA