Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
Biochimie ; 184: 8-17, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33556471

RESUMO

Acquired drug-resistance, often involving downregulation or mutations in the target protein, is a major caveat in precision medicine. Understanding mechanisms of resistance to therapeutic drugs may unravel strategies to overcome or prevent them. We previously identified phorbol ester (PE) compounds such as TPA that induce Protein Kinase δ (PKCδ), thereby suppressing leukemogenesis. Here we identified erythroleukemia cell lines that resist PEs and showed that reduced PKCδ protein expression underlies drug resistance. Reduced level of PKCδ in resistant cell lines was due to its phosphorylation followed by protein degradation. Indeed, proteasome inhibition prevented PE-induced loss of PKCδ. Accordingly, a combination of TPA and the proteasome inhibitor ALLN significantly suppressed leukemia in a mouse model of leukemia. PKCδ downregulation by TPA was independent of the downstream MAPK/ERK/P38/JNK pathway. Instead, expression of ubiquitin-associated and SH3 domain-containing protein b (Ubash3b) was induced by TPA, which leads to PKCδ protein dephosphorylation and degradation. This specific degradation was blocked by RNAi-mediated depletion of Ubash3b. In drug-sensitive leukemic cells, TPA did not induce Ubash3b, and consequently, PKCδ levels remained high. A PE-resistant cell line derived from PE-treated sensitive cells exhibited very low PKCδ expression. In these drug resistance cells, a Ubash3b independent mechanism led to PKCδ degradation. Thus, PE compounds in combination with proteasome or specific inhibitors for Ubash3b, or other factors can overcome resistance to TPA, leading to durable suppression of leukemic growth. These results identify Ubash3b as a potential target for drug development.


Assuntos
Carcinogênese/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Leucêmica da Expressão Gênica/efeitos dos fármacos , Leucemia/enzimologia , Proteínas de Neoplasias/metabolismo , Proteína Quinase C-delta/biossíntese , Proteínas Tirosina Fosfatases/metabolismo , Acetato de Tetradecanoilforbol/farmacologia , Carcinogênese/genética , Carcinogênese/metabolismo , Linhagem Celular Tumoral , Humanos , Leucemia/genética , Leucemia/patologia , Proteínas de Neoplasias/genética , Proteína Quinase C-delta/genética , Proteínas Tirosina Fosfatases/genética
2.
Med Sci Monit ; 23: 6057-6063, 2017 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-29272263

RESUMO

BACKGROUND This study was aimed to investigate the protective role of baicalin on vascular endothelium exposed to ischemia reperfusion injury and the involved molecular mechanisms. MATERIAL AND METHODS Cultured human arterial endothelial cells (HAECs) were exposed to hypoxia/deoxygenation (H/R). Cells were also treated with baicalin at serially diluted concentrations. Cells were also treated with PKC activator PEP005 or specific siRNA against protein kinase Cδ (PKCδ). MTT assay was used to evaluate the cell viabilities. Flow cytometry was used to detect cell apoptosis. The protein phosphorylation and expression levels were determined by Western blotting. RESULTS PKCδ-siRNA transfection increased cell viabilities and reduced cell apoptosis in HAECs exposed to H/R. Baicalin treatment preserved cell viabilities and reduced apoptosis of H/R-exposed HAECs in a concentration- dependent manner. Baicalin treatment reduced phosphorylation levels of PKCδ and p53, as well as the expression levels of active caspase3 and bax in HAECs exposed to H/R. The treatment of PKC activator PEP005 impaired the protective effects of baicalin in increasing cell viabilities and reducing apoptosis in HAECs exposed to H/R. CONCLUSIONS Baicalin exerts vascular a protective effect on HAECs exposed to H/R by reducing cell apoptosis. The PKCδ/p53 apoptotic signaling pathway was the pharmacological target of baicalin.


Assuntos
Hipóxia Celular/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Flavonoides/farmacologia , Proteína Quinase C-delta/antagonistas & inibidores , Proteína Supressora de Tumor p53/antagonistas & inibidores , Apoptose/efeitos dos fármacos , Linhagem Celular , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Diterpenos/metabolismo , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Humanos , Fosforilação , Proteína Quinase C-delta/biossíntese , Proteína Quinase C-delta/metabolismo , RNA Interferente Pequeno/metabolismo , Traumatismo por Reperfusão/tratamento farmacológico , Traumatismo por Reperfusão/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteína Supressora de Tumor p53/metabolismo
3.
Nat Neurosci ; 20(12): 1680-1685, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29184202

RESUMO

Experience-driven synaptic plasticity in the lateral amygdala is thought to underlie the formation of associations between sensory stimuli and an ensuing threat. However, how the central amygdala participates in such a learning process remains unclear. Here we show that PKC-δ-expressing central amygdala neurons are essential for the synaptic plasticity underlying learning in the lateral amygdala, as they convey information about the unconditioned stimulus to lateral amygdala neurons during fear conditioning.


Assuntos
Núcleo Central da Amígdala/fisiologia , Aprendizagem/fisiologia , Animais , Aprendizagem da Esquiva , Condicionamento Psicológico , Ingestão de Alimentos , Medo/psicologia , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Plasticidade Neuronal/fisiologia , Optogenética , Técnicas de Patch-Clamp , Proteína Quinase C-delta/biossíntese , Proteína Quinase C-delta/genética , Transmissão Sináptica/fisiologia , Tálamo/fisiologia
4.
Int J Oncol ; 50(5): 1590-1600, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28339053

RESUMO

Osteosarcoma is one of the most highly malignant types of cancer in adolescents and young adults with a high mortality rate. Despite advances in surgery, radiation therapy and chemotherapy, the prognosis for patients with osteosarcoma has not significantly improved over the past several decades. It is necessary to find new indicators of prognosis and therapeutic targets of osteosarcoma. Through the analysis of 40 osteosarcoma tissues, we found that the expression of miR­486 was low and the expression of PKC­Î´ was high in osteosarcoma. Median survival of patients with low expression of miR-486 (30 months) was shorter than the patients with higher expression of miR­486 (40 months). We further found that miR-486 can inhibit the targeting of PKC­Î´ signaling pathways, and this inhibition can inhibit the growth and invasion of osteosarcoma cells. After transfection of miR­486 for 24 h, the proliferation of osteosarcoma cells was inhibited by ~20%, and the migration was inhibited by ~15%. In the present investigation, we demonstrated that miR­486 is negatively associated with the expression of PKC-δ and could regulate the development of osteosarcoma. miR-486 may be a potential target for the treatment of osteosarcoma.


Assuntos
Proliferação de Células/genética , MicroRNAs/genética , Osteossarcoma/genética , Proteína Quinase C-delta/biossíntese , Adolescente , Adulto , Apoptose/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Metástase Neoplásica , Osteossarcoma/patologia , Prognóstico , Proteína Quinase C-delta/genética , Transfecção , Adulto Jovem
5.
Cancer Lett ; 374(1): 167-174, 2016 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-26884258

RESUMO

Genetic alterations and aberrant gene expression trigger malignant tumors. Tumor suppressor p53 is the most altered gene in human cancers. p53 induces apoptosis by promoting pro-apoptotic genes in response to DNA damage. Protein kinase C delta (PKCδ) also induces apoptosis via various mechanisms including modification of p53. The PKCδ-dependent apoptotic mechanism has been extensively studied; however, the transcriptional regulation of PKCδ remains obscure. The current study demonstrates the transcriptional regulation of PKCδ by p53 upon genotoxic stress. The p53-binding site in the promoter region of PKCδ was detected by the ChIP-sequencing assay. Notably, the expression of PKCδ was increased upon DNA damage, which is required for the stabilization of p53. More importantly, targeting single guide RNA-driven dead Cas9 to the p53-binding site of PKCδ disturbed p53-promoted PKCδ expression and suppressed apoptosis following DNA damage. Thus, our findings suggest that the transcriptional regulation of PKCδ is controlled by p53 in a positive feedback mechanism to induce apoptosis in response to DNA damage.


Assuntos
Dano ao DNA , Neoplasias/genética , Proteína Quinase C-delta/biossíntese , Proteína Quinase C-delta/genética , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Apoptose/genética , Sequência de Bases , Sítios de Ligação , Linhagem Celular Tumoral , Indução Enzimática , Regulação Neoplásica da Expressão Gênica , Células HCT116 , Células HEK293 , Humanos , Dados de Sequência Molecular , Neoplasias/enzimologia , Neoplasias/metabolismo , Neoplasias/patologia , Ativação Transcricional , Transfecção , Regulação para Cima
6.
Nat Neurosci ; 18(10): 1493-500, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26322928

RESUMO

Aversive experiences can lead to complex behavioral adaptations including increased levels of anxiety and fear generalization. The neuronal mechanisms underlying such maladaptive behavioral changes, however, are poorly understood. Here, using a combination of behavioral, physiological and optogenetic approaches in mouse, we identify a specific subpopulation of central amygdala neurons expressing protein kinase C δ (PKCδ) as key elements of the neuronal circuitry controlling anxiety. Moreover, we show that aversive experiences induce anxiety and fear generalization by regulating the activity of PKCδ(+) neurons via extrasynaptic inhibition mediated by α5 subunit-containing GABAA receptors. Our findings reveal that the neuronal circuits that mediate fear and anxiety overlap at the level of defined subpopulations of central amygdala neurons and demonstrate that persistent changes in the excitability of a single cell type can orchestrate complex behavioral changes.


Assuntos
Tonsila do Cerebelo/fisiopatologia , Ansiedade/fisiopatologia , Neurônios/fisiologia , Estresse Psicológico/fisiopatologia , Animais , Condicionamento Clássico , Modelos Animais de Doenças , Técnicas de Silenciamento de Genes , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Inibição Neural/fisiologia , Optogenética , Técnicas de Patch-Clamp , Proteína Quinase C-delta/biossíntese , Estresse Psicológico/psicologia
7.
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
8.
Sci Rep ; 4: 6082, 2014 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-25124193

RESUMO

Although proper tongue development is relevant to other structures in the craniofacial region, the molecular details of muscle development in tongue remain poorly understood. Here, we report that pregnant mice treated with retinoic acid (+RA) produce embryos with tongue malformation and a cleft palate. Histological analyses revealed that at E14.5, the tongues of +RA fetuses failed to descend and flatten. Ultrastructural analysis showed that at perinatal stage E18.5, the myofilaments failed to form normal structures of sarcomeres, and arranged disorderly in the genioglossus. The proliferation and levels of myogenic determination markers (Myf5 and MyoD) and myosin in the genioglossus were profoundly reduced. Wnt5a and Camk2d expressions were down-regulated, while levels of Tbx1, Ror2, and PKCδ were up-regulated in the tongues of +RA fetuses. In mock- and Wnt5a-transfected C2C12 (Wnt5a-C2C12) cells, Wnt5a overexpression impaired proliferation, and maintained Myf5 at a relative high level after RA treatment. Furthermore, Wnt5a overexpression positively correlated with levels of Camk2d and Ror2 in C2C12 cells after RA exposure. These data support the hypothesis that the Wnt5a/CaMKII pathway is directly involved in RA-induced hypoplasia and disorder of tongue muscles.


Assuntos
Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Desenvolvimento Muscular/efeitos dos fármacos , Língua/anormalidades , Tretinoína/efeitos adversos , Proteínas Wnt/metabolismo , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/biossíntese , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/genética , Linhagem Celular , Proliferação de Células , Fissura Palatina/induzido quimicamente , Embrião de Mamíferos/embriologia , Feminino , Camundongos , Camundongos Endogâmicos ICR , Proteína MyoD/metabolismo , Fator Regulador Miogênico 5/metabolismo , Miosinas/metabolismo , Gravidez , Proteína Quinase C-delta/biossíntese , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/biossíntese , Sarcômeros/patologia , Proteínas com Domínio T/biossíntese , Língua/embriologia , Tretinoína/farmacologia , Proteínas Wnt/biossíntese , Proteínas Wnt/genética , Proteína Wnt-5a
9.
Int J Biochem Cell Biol ; 53: 55-65, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24814288

RESUMO

Multidrug-resistant (MDR) cancer is a major clinical problem in chemotherapy of cancer patients. We have noted inappropriate PKCδ hypomethylation and overexpression of genes in the PKCδ/AP-1 pathway in the human uterus sarcoma drug-resistant cell line, MES-SA/Dx5 cells, which also overexpress p-glycoprotein (ABCB1). Recent studies have indicated that FZD1 is overexpressed in both multidrug-resistant cancer cell lines and in clinical tumor samples. These data have led us to hypothesize that the FZD1-mediated PKCδ signal-transduction pathway may play an important role in drug resistance in MES-SA/Dx5 cells. In this work, the PKCδ inhibitor Rottlerin was found to reduce ABCB1 expression and to inhibit the MDR drug pumping ability in the MES-SA/Dx5 cells when compared with the doxorubicin-sensitive parental cell line, MES-SA. PKCδ was up-regulated with concurrent up-regulation of the mRNA levels of the AP-1-related factors, c-JUN and c-FOS. Activation of AP-1 also correlated with up-regulation of the AP-1 downstream genes HGF and EGR1. Furthermore, AP-1 activities were reduced and the AP-1 downstream genes were down-regulated in Rottlerin-treated or PKCδ shRNA-transfected cells. MES-SA/Dx5 cells were resensitized to doxorubicin-induced toxicity by co-treatment with doxorubicin and Rottlerin or PKCδ shRNA. In addition, cell viability and drug pump-out ability were significantly reduced in the FZD1 inhibitor curcumin-treated and FZD1 shRNA-knockdown MES-SA/Dx5 cells, indicating involvement of PKCδ in FZD1-modulated ABCB1 expression pathway. Taken together, our data demonstrate that FZD1 regulates PKCδ, and the PKCδ/AP-1 signalling transduction pathway plays an important role in drug resistance in MES-SA/Dx5 cells.


Assuntos
Receptores Frizzled/genética , Proteína Quinase C-delta/biossíntese , Sarcoma/genética , Neoplasias Uterinas/genética , Subfamília B de Transportador de Cassetes de Ligação de ATP/biossíntese , Subfamília B de Transportador de Cassetes de Ligação de ATP/genética , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Doxorrubicina/administração & dosagem , Resistencia a Medicamentos Antineoplásicos/genética , Feminino , Receptores Frizzled/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Sarcoma/tratamento farmacológico , Sarcoma/patologia , Transdução de Sinais , Neoplasias Uterinas/tratamento farmacológico , Neoplasias Uterinas/patologia
10.
Neuromolecular Med ; 16(1): 25-37, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23896721

RESUMO

A balance between cell survival and apoptosis is crucial to avoid neurodegeneration. Here, we analyzed whether the pro-apoptotic protein PKCδ, and the pro-survival PKCα and ßII, were dysregulated in the brain of R6/1 mouse model of Huntington's disease (HD). Protein levels of the three PKCs examined were reduced in all the brain regions analyzed being PKCδ the most affected isoform. Interestingly, PKCδ protein levels were also decreased in the striatum and cortex of R6/2 and Hdh(Q111/Q111) mice, and in the putamen of HD patients. Nuclear PKCδ induces apoptosis, but we detected reduced PKCδ in both cytoplasmic and nuclear enriched fractions from R6/1 mouse striatum, cortex and hippocampus. In addition, we show that phosphorylation and ubiquitination of PKCδ are increased in 30-week-old R6/1 mouse brain. All together these results suggest a pro-survival role of reduced PKCδ levels in response to mutant huntingtin-induced toxicity. In fact, we show that over-expression of PKCδ increases mutant huntingtin-induced cell death in vitro, whereas over-expression of a PKCδ dominant negative form or silencing of endogenous PKCδ partially blocks mutant huntingtin-induced cell death. Finally, we show that the analysis of lamin B protein levels could be a good marker of PKCδ activity, but it is not involved in PKCδ-mediated cell death in mutant huntingtin-expressing cells. In conclusion, our results suggest that neurons increase the degradation of PKCδ as a compensatory pro-survival mechanism in response to mutant huntingtin-induced toxicity that can help to understand why cell death appears late in the disease.


Assuntos
Doença de Huntington/enzimologia , Proteínas do Tecido Nervoso/fisiologia , Proteína Quinase C-delta/fisiologia , Animais , Apoptose , Núcleo Celular/enzimologia , Córtex Cerebral/enzimologia , Corpo Estriado/enzimologia , Citoplasma/enzimologia , Modelos Animais de Doenças , Progressão da Doença , Regulação para Baixo , Feminino , Perfilação da Expressão Gênica , Hipocampo/enzimologia , Humanos , Proteína Huntingtina , Doença de Huntington/patologia , Lamina Tipo B/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/enzimologia , Proteínas Nucleares/metabolismo , Fosforilação , Proteína Quinase C beta/análise , Proteína Quinase C-alfa/análise , Proteína Quinase C-delta/biossíntese , Proteína Quinase C-delta/genética , Processamento de Proteína Pós-Traducional , Putamen/enzimologia , RNA Interferente Pequeno/farmacologia , Proteínas Recombinantes de Fusão/metabolismo , Ubiquitinação
11.
Mol Neurobiol ; 49(2): 658-72, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24018979

RESUMO

Among matrix metalloproteinases (MMPs), MMP-9 has been observed in patients with brain inflammatory diseases and may contribute to the pathology of brain diseases. Thrombin has been known as a regulator of MMP-9 expression and cells migration. However, the mechanisms underlying thrombin-induced MMP-9 expression in rat brain astrocytes (RBA-1 cells) were not completely understood. Here, we demonstrated that thrombin induced the expression of pro-form MMP-9 in RBA-1 cells and cells migration which were attenuated by pretreatment with the inhibitor of receptor tyrosine kinase (Genistein), c-Src (PP1), Jak2 (AG490), PDGFR (AG1296), PI3K (LY294002), Akt (SH-5), PKCs (Ro318220), PKCδ (Rottlerin), or NF-κB (Bay11-7082) and transfection with siRNA of c-Src, PDGFR, Akt, PKCδ, ATF2, p65, IKKα, or IKKß. In addition, thrombin-stimulated c-Src, Jak2, or PDGFR phosphorylation was inhibited by a thrombin inhibitor (PPACK), PP1, AG490, or AG1296. Thrombin further stimulated c-Src and PDGFR complex formation in RBA-1 cells. Thrombin also stimulated Akt and PKCδ phosphorylation and PKCδ translocation which were reduced by PPACK, PP1, AG490, AG1296, or LY294002. We further observed that thrombin markedly stimulated ATF2 or IκBα phosphorylation and NF-κB p65 translocation which were inhibited by Rottlerin or LY294002. Finally, thrombin stimulated in vivo binding of p65 to the MMP-9 promoter, which was reduced by pretreatment with Rottlerin or LY294002. These results concluded that in RBA-1 cells, thrombin activated a c-Src/Jak2/PDGFR/PI3K/Akt/PKCδ pathway, which in turn triggered ATF2 and NF-κB activation and ultimately induced MMP-9 expression associated with cell migration.


Assuntos
Astrócitos/metabolismo , Genes src/fisiologia , Janus Quinase 2/biossíntese , Metaloproteinase 9 da Matriz/biossíntese , Proteína Quinase C-delta/biossíntese , Receptores do Fator de Crescimento Derivado de Plaquetas/biossíntese , Animais , Astrócitos/efeitos dos fármacos , Encéfalo/citologia , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Células Cultivadas , Indução Enzimática/efeitos dos fármacos , Indução Enzimática/fisiologia , Genes src/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Trombina/farmacologia
12.
Biochem Biophys Res Commun ; 437(3): 380-5, 2013 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-23827392

RESUMO

Our previous works have shown that the (NADPH) oxidase (Nox) enzyme, in particular Nox1, plays an important role in oxidative stress and subsequent dopaminergic cell death elicited by paraquat (PQ). In non-neuronal and glial cells, protein kinase C δ (PKCδ) shows the ability to regulate the activity of the Nox system. Herein we aimed to investigate if also in dopaminergic neurons exposed to PQ, PKCδ can regulate Nox1 expression. The chemical inhibitor, rottlerin, and short interference RNA (siRNA) were used to inhibit or selectively knockdown PKCδ, respectively. The studies were performed using the immortalized rat mesencephalic dopaminergic cell line (N27 cells) exposed to PQ, after pre-incubation with rottlerin or transfected with PKCδ-siRNA. We observed that inhibition or knockdown of PKCδ significantly reduced PQ induced Nox1 transcript and protein levels, ROS generation and subsequent dopaminergic cell death. The results suggest that PKCδ plays a role in the regulation of Nox1-mediated oxidative stress elicited by PQ and could have a role in the pathogenesis of Parkinson's disease.


Assuntos
Dopamina/fisiologia , NADH NADPH Oxirredutases/biossíntese , Neurônios/enzimologia , Paraquat/farmacologia , Proteína Quinase C-delta/fisiologia , Acetofenonas/farmacologia , Animais , Benzopiranos/farmacologia , Morte Celular/efeitos dos fármacos , Morte Celular/fisiologia , Linhagem Celular Transformada , Indução Enzimática/efeitos dos fármacos , Indução Enzimática/genética , Técnicas de Silenciamento de Genes , NADH NADPH Oxirredutases/genética , NADPH Oxidase 1 , Neurônios/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Proteína Quinase C-delta/antagonistas & inibidores , Proteína Quinase C-delta/biossíntese , Ratos , Espécies Reativas de Oxigênio/metabolismo
13.
J Biomol Screen ; 18(7): 797-806, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23580666

RESUMO

There is a growing need in drug discovery and basic research to measure multiple second-messenger components of cell signaling pathways in real time and in relevant tissues and cell types. Many G-protein-coupled receptors activate the heterotrimeric protein, Gq, which in turn activates phospholipase C (PLC). PLC cleaves phosphatidylinositol 4,5-bisphosphate (PIP2) to produce two second messengers: diacylglycerol (DAG), which remains in the plasma membrane, and inositol triphosphate (IP3), which diffuses through the cytosol to release stores of intracellular calcium ions (Ca(2+)). Our goal was to create a series of multiplex sensors that would make it possible to simultaneously measure two different components of the Gq pathway in living cells. Here we describe new fluorescent sensors for DAG and PIP2 that produce robust changes in green or red fluorescence and can be combined with one another, or with existing Ca(2+) sensors, in a live-cell assay. These assays can detect multiple components of Gq signaling, simultaneously in real time, on standard fluorescent plate readers or live-cell imaging systems.


Assuntos
Bioensaio , Sinalização do Cálcio , Receptores Acoplados a Proteínas G/metabolismo , Trifosfato de Adenosina/fisiologia , Técnicas Biossensoriais , Diglicerídeos/metabolismo , Proteínas de Fluorescência Verde/biossíntese , Células HEK293 , Humanos , Proteínas Luminescentes/biossíntese , Fosfatidilinositol 4,5-Difosfato/metabolismo , Proteína Quinase C-delta/biossíntese , Proteínas Recombinantes de Fusão/biossíntese , Sistemas do Segundo Mensageiro , Espectrometria de Fluorescência , Fosfolipases Tipo C/metabolismo , Proteína Vermelha Fluorescente
14.
J Neurophysiol ; 108(12): 3196-205, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22972957

RESUMO

Recent findings implicate the central lateral amygdala (CeL) in conditioned fear. Indeed, CeL contains neurons exhibiting positive (CeL-On) or negative (CeL-Off) responses to fear-inducing conditioned stimuli (CSs). In mice, these cells differ in their expression of protein kinase Cδ (PKCδ) and physiological properties. CeL-Off cells are PKCδ(+) and late firing (LF), whereas CeL-On cells are PKCδ(-) and express a regular-spiking (RS) or low-threshold bursting (LTB) phenotype. However, the scarcity of LF cells in rats raises questions about the correspondence between the organization of CeL in mice and rats. Therefore, we studied the PKCδ expression, morphological properties, synaptic responsiveness, and fear conditioning-induced plasticity of rat CeL neurons. No PKCδ(+) LF cells were encountered, but ≈20-25% of RS and LTB neurons were PKCδ(+). Compared with RS neurons, a higher proportion of LTB cells projected to central medial amygdala (CeM) and they had fewer primary dendritic branches, yet the amplitude of excitatory postsynaptic potentials (EPSPs) evoked by lateral amygdala (LA) stimulation was similar in RS and LTB cells. In contrast, LA-evoked inhibitory postsynaptic potentials (IPSPs) had a higher amplitude in LTB than RS neurons. Finally, fear conditioning did not induce plasticity at LA inputs to RS or LTB neurons. These findings point to major species differences in the organization of CeL. Since rat LTB cells are subjected to stronger feedforward inhibition, they are more likely to exhibit inhibitory CS responses than RS cells. This is expected to cause a disinhibition of CeM fear output neurons and therefore an increase in fear expression.


Assuntos
Tonsila do Cerebelo/citologia , Tonsila do Cerebelo/enzimologia , Regulação Enzimológica da Expressão Gênica , Neurônios/enzimologia , Proteína Quinase C-delta/biossíntese , Potenciais Sinápticos/fisiologia , Animais , Masculino , Plasticidade Neuronal/fisiologia , Ratos , Ratos Sprague-Dawley , Sinapses/enzimologia
15.
Anticancer Res ; 32(9): 3707-13, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22993309

RESUMO

Photodynamic therapy is becoming a widely accepted form of cancer treatment using a photosensitizing agent and light. Our previous study has demonstrated that photoactivated aloe-emodin induced anoikis and changes in cell morphology, which were in part mediated through its effect on cytoskeleton in lung carcinoma H460 cells. However, the molecular mechanisms of these photoactivated aloe-emodin-induced changes remain unknown. The present study demonstrated that the expression of protein kinase Cδ (PKCδ) was triggered by aloe-emodin and irradiation in H460 cells. Furthermore, the photoactivated aloe-emodin-induced cell death and translocation of PKCδ from the cytosol to the nucleus was found to be significantly inhibited by rottlerin, a PKCδ-selective inhibitor. Western blot analysis demonstrated that rottlerin also reversed the decrease in protein expression of cytoskeleton-related proteins, such as rat sarcoma (RAS), ras homolog gene family member A (RHO), p38, heat shock protein 27 (HSP27), focal adhesion kinase (FAK), α-actinin and tubulin, induced by photoactivated aloe-emodin. Our findings suggest that the regulation of cytoskeleton-related proteins mediated by PKCδ may be the mechanisms for the protective effects of rottlerin against the photoactivated aloe-emodin induced H460 cell death.


Assuntos
Antraquinonas/farmacologia , Citoesqueleto/efeitos dos fármacos , Neoplasias Pulmonares/tratamento farmacológico , Fotoquimioterapia/métodos , Proteína Quinase C-delta/metabolismo , Acetofenonas/farmacologia , Citoesqueleto de Actina/efeitos dos fármacos , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Apoptose/efeitos dos fármacos , Benzopiranos/farmacologia , Morte Celular , Linhagem Celular Tumoral , Citoesqueleto/enzimologia , Citoesqueleto/patologia , Humanos , Neoplasias Pulmonares/enzimologia , Neoplasias Pulmonares/patologia , Proteína Quinase C-delta/biossíntese
16.
Am J Physiol Lung Cell Mol Physiol ; 303(10): L880-8, 2012 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-22983354

RESUMO

The pathogenesis of acute lung injury and acute respiratory distress syndrome is characterized by sequestration of leukocytes in lung tissue, disruption of capillary integrity, and pulmonary edema. PKCδ plays a critical role in RhoA-mediated endothelial barrier function and inflammatory responses. We used mice with genetic deletion of PKCδ (PKCδ(-/-)) to assess the role of PKCδ in susceptibility to LPS-induced lung injury and pulmonary edema. Under baseline conditions or in settings of increased capillary hydrostatic pressures, no differences were noted in the filtration coefficients (k(f)) or wet-to-dry weight ratios between PKCδ(+/+) and PKCδ(-/-) mice. However, at 24 h after exposure to LPS, the k(f) values were significantly higher in lungs isolated from PKCδ(+/+) than PKCδ(-/-) mice. In addition, bronchoalveolar lavage fluid obtained from LPS-exposed PKCδ(+/+) mice displayed increased protein and cell content compared with LPS-exposed PKCδ(-/-) mice, but similar changes in inflammatory cytokines were measured. Histology indicated elevated LPS-induced cellularity and inflammation within PKCδ(+/+) mouse lung parenchyma relative to PKCδ(-/-) mouse lungs. Transient overexpression of catalytically inactive PKCδ cDNA in the endothelium significantly attenuated LPS-induced endothelial barrier dysfunction in vitro and increased k(f) lung values in PKCδ(+/+) mice. However, transient overexpression of wild-type PKCδ cDNA in PKCδ(-/-) mouse lung vasculature did not alter the protective effects of PKCδ deficiency against LPS-induced acute lung injury. We conclude that PKCδ plays a role in the pathological progression of endotoxin-induced lung injury, likely mediated through modulation of inflammatory signaling and pulmonary vascular barrier function.


Assuntos
Lesão Pulmonar Aguda/enzimologia , Barreira Alveolocapilar/enzimologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Lipopolissacarídeos/toxicidade , Proteína Quinase C-delta/biossíntese , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/genética , Lesão Pulmonar Aguda/patologia , Animais , Barreira Alveolocapilar/patologia , Citocinas/genética , Citocinas/metabolismo , Mediadores da Inflamação/metabolismo , Camundongos , Camundongos Knockout , Proteína Quinase C-delta/genética , Edema Pulmonar/induzido quimicamente , Edema Pulmonar/enzimologia , Edema Pulmonar/genética , Edema Pulmonar/patologia , Síndrome do Desconforto Respiratório
17.
J Invest Dermatol ; 132(8): 1988-97, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22475757

RESUMO

Protein kinase C (PKC) isoforms have crucial roles in cutaneous signaling. Interestingly, we lack information about their involvement in human sebaceous gland biology. Therefore, in this current study, we investigated the functions of the PKC system in human immortalized SZ95 sebocytes. Using molecular biological approaches, imaging, and functional assays, we report that SZ95 sebocytes express the conventional cPKCα; the novel nPKCδ, ɛ, and η; and the atypical aPKCζ. Activation of the PKC system by phorbol 12-myristate 13-acetate (PMA) stimulated lipid synthesis (a hallmark of differentiation) and resulted in translocation and then downregulation of cPKCα and nPKCδ. In good accord with these findings, the effect of PMA was effectively abrogated by inhibitors and short interfering RNA-mediated "silencing" of cPKCα and nPKCδ. Of further importance, molecular or pharmacological inhibition of nPKCδ also prevented the lipogenic and apoptosis-promoting action of arachidonic acid. Finally, we also found that "knockdown" of the endogenous aPKCζ activity markedly increased basal lipid synthesis and apoptosis, suggesting its constitutive activity in suppressing these processes. Collectively, our findings strongly argue for the fact that certain PKCs have pivotal, isoform-specific, differential, and antagonistic roles in the regulation of human sebaceous gland-derived sebocyte biology.


Assuntos
Regulação Enzimológica da Expressão Gênica , Proteína Quinase C-alfa/biossíntese , Proteína Quinase C-delta/biossíntese , Proteína Quinase C-épsilon/biossíntese , Proteína Quinase C/biossíntese , Glândulas Sebáceas/citologia , Apoptose , Diferenciação Celular , Regulação para Baixo , Inativação Gênica , Humanos , Lipídeos/química , Necrose , Isoformas de Proteínas , RNA Interferente Pequeno/metabolismo , Acetato de Tetradecanoilforbol/farmacologia
18.
J Biol Chem ; 287(10): 7313-23, 2012 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-22199349

RESUMO

PKCδ is a key regulator of keratinocyte differentiation that activates p38δ phosphorylation leading to increased differentiation as measured by an increased expression of the structural protein involucrin. Our previous studies suggest that p38δ exists in association with protein partners. A major goal is to identify these partners and understand their role in regulating keratinocyte differentiation. In this study we use affinity purification and mass spectrometry to identify protein arginine methyltransferase 5 (PRMT5) as part of the p38δ signaling complex. PRMT5 is an arginine methyltransferase that symmetrically dimethylates arginine residues on target proteins to alter target protein function. We show that PRMT5 knockdown is associated with increased p38δ phosphorylation, suggesting that PRMT5 impacts the p38δ signaling complex. At a functional level we show that PRMT5 inhibits the PKCδ- or 12-O-tetradecanoylphorbol-13-acetate-dependent increase in human involucrin expression, and PRMT5 dimethylates proteins in the p38δ complex. Moreover, PKCδ expression reduces the PRMT5 level, suggesting that PKCδ activates differentiation in part by reducing PRMT5 level. These studies indicate antagonism between the PKCδ and PRMT5 signaling in control of keratinocyte differentiation.


Assuntos
Diferenciação Celular/fisiologia , Regulação Enzimológica da Expressão Gênica/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Proteína Quinase C-delta/biossíntese , Proteínas Metiltransferases/biossíntese , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Carcinógenos/farmacologia , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Complexos Multienzimáticos/metabolismo , Proteína Quinase C-delta/antagonistas & inibidores , Proteínas Metiltransferases/antagonistas & inibidores , Precursores de Proteínas/biossíntese , Proteína-Arginina N-Metiltransferases , Acetato de Tetradecanoilforbol/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
19.
Oncol Rep ; 26(5): 1221-6, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21822543

RESUMO

Protein kinase Cδ (PKCδ), an isoform of PKC, has been shown to act as a critical mediator of tumor progression and apoptosis; however, its role in musculoskeletal tumors is still unknown. In the current study, we examined the expression of PKCδ in human musculoskeletal tumor tissue samples, and investigated the effects of siRNA downregulation of PKCδ on human malignant fibrous histiocytoma (MFH) cell proliferation, migration, and apoptosis, to elucidate its functional roles in musculoskeletal tumorigenesis. Of note, real-time PCR analysis revealed that mRNA expression of PKCδ in high-grade musculoskeletal MFH tumors was significantly lower than that in benign schwannomas. siRNA downregulation of PKCδ significantly increased human MFH cell proliferation and migration, and markedly suppressed apoptosis. These findings suggest that PKCδ has a negative effect on tumorigenesis and/or acts as a pro-apoptotic kinase in human MFH cells. The data presented here could be applied in the development of new therapeutic avenues, with the elevation of PKCδ expression being one potential strategy to prevent MFH progression. Thus, PKCδ may be a potent therapeutic target for human MFH.


Assuntos
Transformação Celular Neoplásica/metabolismo , Histiocitoma Fibroso Maligno/enzimologia , Proteína Quinase C-delta/biossíntese , Apoptose/fisiologia , Processos de Crescimento Celular/fisiologia , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Transformação Celular Neoplásica/patologia , Regulação para Baixo , Histiocitoma Fibroso Maligno/patologia , Humanos , Proteína Quinase C-delta/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase em Tempo Real , Transfecção , Células Tumorais Cultivadas
20.
Mol Cell Biochem ; 357(1-2): 163-9, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21625957

RESUMO

Disruption to the sensitive balance of long-chain fatty acids and glucose in the heart could cause cardiovascular diseases. Searching for a possible role of novel protein kinase C (nPKC) in heart with disrupted energy balance, we compared the insulin-resistant spontaneously hypertensive rats (SHR), which carry a nonfunctional variant of the fatty acid transporter FAT/CD36, with the less insulin-resistant congenic strain SHR-4 that is genetically identical except for a segment on chromosome 4 including a wild-type gene for a functional FAT/CD36. We analyzed expression of the nPKC-δ and -ε isoforms plus triacylglycerols (TAG) content in the myocardium of both FAT/CD36 strains and after a high sucrose diet (HSD). Two weeks before killing, males of both strains were randomly divided into two groups and fed either a standard laboratory chow or an HSD. PKC was determined by Western blotting in particulate and cytosolic fractions from left ventricles. The SHR-4 rats exhibited lower serum levels of insulin and free fatty acids than did SHR rats and higher amounts of PKC-ε in the heart particulate fraction. HSD caused accumulation of heart TAG in SHR but not in SHR-4. HSD increased PKC-δ and decreased PKC-ε expression in particulate fraction from left ventricles of SHR-4 while having no effects in SHR. These results demonstrate that reduced insulin resistance in SHR-4 rats with wild-type FAT/CD36 is associated with the insulin signaling pathway involving nPKCs.


Assuntos
Antígenos CD36/metabolismo , Ventrículos do Coração/metabolismo , Resistência à Insulina/genética , Proteína Quinase C-delta/biossíntese , Proteína Quinase C-épsilon/biossíntese , Animais , Glicemia/análise , Glicemia/metabolismo , Antígenos CD36/genética , Citosol/metabolismo , Ativação Enzimática , Ácidos Graxos não Esterificados/sangue , Ácidos Graxos não Esterificados/metabolismo , Regulação da Expressão Gênica , Insulina/sangue , Insulina/metabolismo , Resistência à Insulina/fisiologia , Masculino , Miocárdio/metabolismo , Ratos , Ratos Endogâmicos SHR , Transdução de Sinais , Sacarose/metabolismo , Triglicerídeos/sangue , Triglicerídeos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...