Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.118
Filtrar
1.
PLoS Pathog ; 18(1): e1010245, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-35041707

RESUMO

Activation of the NF-κB signaling pathway by Protein Kinase C (PKC) agonists is a potent mechanism for human immunodeficiency virus (HIV) latency disruption in vitro. However, significant toxicity risks and the lack of evidence supporting their activity in vivo have limited further evaluation of PKC agonists as HIV latency-reversing agents (LRA) in cure strategies. Here we evaluated whether GSK445A, a stabilized ingenol-B derivative, can induce HIV/simian immunodeficiency virus (SIV) transcription and virus production in vitro and demonstrate pharmacological activity in nonhuman primates (NHP). CD4+ T cells from people living with HIV and from SIV+ rhesus macaques (RM) on antiretroviral therapy (ART) exposed in vitro to 25 nM of GSK445A produced cell-associated viral transcripts as well as viral particles at levels similar to those induced by PMA/Ionomycin, indicating that GSK445A can potently reverse HIV/SIV latency. Importantly, these concentrations of GSK445A did not impair the proliferation or survival of HIV-specific CD8+ T cells, but instead, increased their numbers and enhanced IFN-γ production in response to HIV peptides. In vivo, GSK445A tolerability was established in SIV-naïve RM at 15 µg/kg although tolerability was reduced in SIV-infected RM on ART. Increases in plasma viremia following GSK445A administration were suggestive of increased SIV transcription in vivo. Collectively, these results indicate that GSK445A is a potent HIV/SIV LRA in vitro and has a tolerable safety profile amenable for further evaluation in vivo in NHP models of HIV cure/remission.


Assuntos
Diterpenos/farmacologia , HIV , Proteína Quinase C/efeitos dos fármacos , Vírus da Imunodeficiência Símia , Ativação Viral/efeitos dos fármacos , Latência Viral/efeitos dos fármacos , Animais , Humanos , Macaca mulatta , Proteína Quinase C/metabolismo , RNA Viral/efeitos dos fármacos , Transcrição Gênica
2.
J Med Chem ; 64(23): 17304-17325, 2021 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-34847663

RESUMO

As a vital kinase in the glycolysis system, PKM2 is extensively expressed in colorectal cancer (CRC) to support the energy and biosynthetic needs. In this study, we designed a series of parthenolide (PTL) derivatives through a stepwise structure optimization, and an excellent derivate 29e showed good activity on PKM2 (AC50 = 86.29 nM) and displayed significant antiproliferative activity against HT29 (IC50 = 0.66 µM) and SW480 (IC50 = 0.22 µM) cells. 29e decreased the expression of total PKM2, prevented nucleus translocation of PKM2 dimer, and inhibited PKM2/STAT3 signaling pathway. 29e remarkably increased OCR and decreased the extracellular acidification rate (ECAR). The antiproliferative effect of 29e depended on PKM2, and the Cys424 of PKM2 was the key binding site. Furthermore, 29e significantly suppressed tumor growth in the HT29 xenograft model without obvious toxicity. These outcomes demonstrate that 29e is a promising drug candidate for the treatment of CRC.


Assuntos
Neoplasias Colorretais/patologia , Ativadores de Enzimas/farmacologia , Proteína Quinase C/efeitos dos fármacos , Sesquiterpenos/farmacologia , Animais , Sítios de Ligação , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Neoplasias Colorretais/enzimologia , Dimerização , Ativadores de Enzimas/química , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Fator de Transcrição STAT3/metabolismo , Sesquiterpenos/química , Transdução de Sinais/efeitos dos fármacos , Relação Estrutura-Atividade , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Mol Med Rep ; 23(6)2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33880589

RESUMO

Numerous human circular RNAs (circRNAs/circ) have been functionally characterized. However, the potential role of circ-protein kinase C iota (PRKCI) in hepatocellular carcinoma (HCC) remains unknown. The effects of each transfection and expression levels of circ-PRKCI, microRNA (miR)-1294, miR-186-5p and forkhead box K1 (FOXK1) in HCC cells were analyzed using reverse transcription-quantitative PCR analysis. The interactions between circ-PRKCI and miR-1294 or miR-186-5p, and miR-1294 or miR-186-5p and FOXK1 were validated using dual luciferase reporter assays. The viability, invasion and migration of HCC cells were determined using Cell Counting Kit-8, Transwell and wound healing assays, respectively. The expression levels of FOXK1, hexokinase-2 (HK2), glucose transporter 1 (GLUT1) and lactate dehydrogenase A (LDHA) in HCC cells were analyzed using western blotting. The levels of glucose and lactic acid in the cultured supernatant were detected using commercially available kits. The results of the present study revealed that miR-1294 and miR-186-5p expression levels were downregulated in the HCC cell line, HCCLM3, and were subsequently downregulated by circ-PRKCI overexpression and upregulated by the knockdown of circ-PRKCI. circ-PRKCI overexpression promoted the viability, invasion and migration of HCCLM3 cells, which was also reversed by the overexpression of miR-1294 and miR-186-5p. In addition, the overexpression of circ-PRKCI upregulated FOXK1 expression levels, while the overexpression of miR-1294 and miR-186-5p downregulated FOXK1 expression levels. Conversely, the knockdown of circ-PRKCI expression downregulated FOXK1 expression levels, while the knockdown of miR-1294 and miR-186-5p upregulated FOXK1 expression levels. Furthermore, circ-PRKCI was identified to target miR-1294 and miR-186-5p, and miR-1294 and miR-186-5p were subsequently found to target FOXK1. The overexpression of circ-PRKCI also increased glucose and lactic acid levels, while the knockdown of FOXK1 decreased glucose and lactic acid levels. The knockdown of circ-PRKCI decreased glucose and lactic acid levels, which were reversed by FOXK1 overexpression. In conclusion, the findings of the present study suggested that circ-PRKCI may promote the viability, invasion and migration of HCC cells by sponging miR-1294 and miR-186-5p to upregulate FOXK1 expression levels.


Assuntos
Carcinoma Hepatocelular/tratamento farmacológico , Regulação para Baixo , Fatores de Transcrição Forkhead/metabolismo , Glicólise/efeitos dos fármacos , Isoenzimas/metabolismo , Neoplasias Hepáticas/tratamento farmacológico , MicroRNAs/farmacologia , Proteína Quinase C/metabolismo , Carcinoma Hepatocelular/genética , Linhagem Celular Tumoral , Fatores de Transcrição Forkhead/genética , Regulação Neoplásica da Expressão Gênica , Transportador de Glucose Tipo 1/metabolismo , Hexoquinase/metabolismo , Humanos , Isoenzimas/efeitos dos fármacos , Isoenzimas/genética , L-Lactato Desidrogenase , Neoplasias Hepáticas/genética , MicroRNAs/metabolismo , Proteína Quinase C/efeitos dos fármacos , Proteína Quinase C/genética , RNA Circular , Regulação para Cima
4.
J Neurosci ; 41(1): 193-210, 2021 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-33172978

RESUMO

Oral squamous cell carcinoma (OSCC) is one of the most painful cancers, which interferes with orofacial function including talking and eating. We report that legumain (Lgmn) cleaves protease-activated receptor-2 (PAR2) in the acidic OSCC microenvironment to cause pain. Lgmn is a cysteine protease of late endosomes and lysosomes that can be secreted; it exhibits maximal activity in acidic environments. The role of Lgmn in PAR2-dependent cancer pain is unknown. We studied Lgmn activation in human oral cancers and oral cancer mouse models. Lgmn was activated in OSCC patient tumors, compared with matched normal oral tissue. After intraplantar, facial or lingual injection, Lgmn evoked nociception in wild-type (WT) female mice but not in female mice lacking PAR2 in NaV1.8-positive neurons (Par2Nav1.8), nor in female mice treated with a Lgmn inhibitor, LI-1. Inoculation of an OSCC cell line caused mechanical and thermal hyperalgesia that was reversed by LI-1. Par2Nav1.8 and Lgmn deletion attenuated mechanical allodynia in female mice with carcinogen-induced OSCC. Lgmn caused PAR2-dependent hyperexcitability of trigeminal neurons from WT female mice. Par2 deletion, LI-1, and inhibitors of adenylyl cyclase or protein kinase A (PKA) prevented the effects of Lgmn. Under acidified conditions, Lgmn cleaved within the extracellular N terminus of PAR2 at Asn30↓Arg31, proximal to the canonical trypsin activation site. Lgmn activated PAR2 by biased mechanisms in HEK293 cells to induce Ca2+ mobilization, cAMP formation, and PKA/protein kinase D (PKD) activation, but not ß-arrestin recruitment or PAR2 endocytosis. Thus, in the acidified OSCC microenvironment, Lgmn activates PAR2 by biased mechanisms that evoke cancer pain.SIGNIFICANCE STATEMENT Oral squamous cell carcinoma (OSCC) is one of the most painful cancers. We report that legumain (Lgmn), which exhibits maximal activity in acidic environments, cleaves protease-activated receptor-2 (PAR2) on neurons to produce OSCC pain. Active Lgmn was elevated in OSCC patient tumors, compared with matched normal oral tissue. Lgmn evokes pain-like behavior through PAR2 Exposure of pain-sensing neurons to Lgmn decreased the current required to generate an action potential through PAR2 Inhibitors of adenylyl cyclase and protein kinase A (PKA) prevented the effects of Lgmn. Lgmn activated PAR2 to induce calcium mobilization, cAMP formation, and activation of protein kinase D (PKD) and PKA, but not ß-arrestin recruitment or PAR2 endocytosis. Thus, Lgmn is a biased agonist of PAR2 that evokes cancer pain.


Assuntos
Dor do Câncer/induzido quimicamente , Carcinoma de Células Escamosas/complicações , Cisteína Endopeptidases , Neoplasias Bucais/complicações , Receptor PAR-2/agonistas , Idoso , Idoso de 80 Anos ou mais , Animais , Arrestina/metabolismo , Dor do Câncer/psicologia , Proteínas Quinases Dependentes de AMP Cíclico/efeitos dos fármacos , Cisteína Endopeptidases/administração & dosagem , Endocitose/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Proteína Quinase C/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Receptor PAR-2/genética , Microambiente Tumoral/efeitos dos fármacos
5.
Cereb Cortex ; 31(5): 2402-2415, 2021 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-33341872

RESUMO

Oxytocin (OXT) is a nonapeptide that serves as a neuromodulator in the brain and a hormone participating in parturition and lactation in the periphery. The subiculum is the major output region of the hippocampus and an integral component in the networks that process sensory and motor cues to form a cognitive map encoding spatial, contextual, and emotional information. Whilst the subiculum expresses the highest OXT-binding sites and is the first brain region to be activated by peripheral application of OXT, the precise actions of OXT in the subiculum have not been determined. Our results demonstrate that application of the selective OXT receptor (OXTR) agonist, [Thr4,Gly7]-oxytocin (TGOT), excited subicular neurons via activation of TRPV1 channels, and depression of K+ channels. The OXTR-mediated excitation of subicular neurons required the functions of phospholipase Cß, protein kinase C, and degradation of phosphatidylinositol 4,5-bisphosphate (PIP2). OXTR-elicited excitation of subicular neurons enhanced long-term potentiation via activation of TRPV1 channels. Our results provide a cellular and molecular mechanism to explain the physiological functions of OXT in the brain.


Assuntos
Hipocampo/metabolismo , Neurônios/metabolismo , Receptores de Ocitocina/metabolismo , Canais de Cátion TRPV/metabolismo , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Sinalização do Cálcio , Feminino , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Potenciação de Longa Duração/efeitos dos fármacos , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Neurônios/efeitos dos fármacos , Ocitocina/análogos & derivados , Ocitocina/farmacologia , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfolipase C beta/efeitos dos fármacos , Fosfolipase C beta/metabolismo , Canais de Potássio/efeitos dos fármacos , Canais de Potássio/metabolismo , Proteína Quinase C/efeitos dos fármacos , Proteína Quinase C/metabolismo , Ratos , Receptores de Ocitocina/agonistas , Transdução de Sinais , Canais de Cátion TRPV/efeitos dos fármacos
6.
Neuropharmacology ; 181: 108356, 2020 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-33069757

RESUMO

Endothelin-1 (ET-1), an endogenous vasoconstrictor, has been known as a pro-nociceptive agent involved in multitude of pain. ET-1 acts on endothelin receptors on vascular endothelial cells, sensitizes release of ATP, which then acts on P2X3 receptors on nociceptors and results in mechanical hyperalgesia. Both endothelin receptors and P2X3 receptors are present in primary sensory neuron, where it remains unclear whether there is an interaction between them. Herein, we reported that ET-1 potentiated the electrophysiological activity of P2X3 receptors in rat dorsal root ganglia (DRG) neurons. ET-1 concentration-dependently increased α,ß-methylene-ATP (α,ß-meATP)-evoked inward currents, which were mediated by P2X3 receptors. ET-1 shifted the α,ß-meATP concentration-response curve upwards, with an increase of 34.38 ± 4.72% in the maximal current response to α,ß-meATP in the presence of ET-1. ET-1 potentiation of α,ß-meATP-evoked currents was voltage-independent. ET-1 potentiated P2X3 receptor-mediated currents through endothelin-A receptors (ETAR), but not endothelin-B receptors (ETBR). ET-1 potentiation was supressed by blockade of intracellular G-protein or protein kinase C (PKC) signaling. Moreover, there is a synergistic effect on mechanical allodynia induced by intraplantar injection of ET-1 and α,ß-meATP in rats. Pharmacological blockade of P2X3 receptors also alleviated ET-1-induced mechanical allodynia. These results suggested that ET-1 sensitized P2X3 receptors in primary sensory neurons via an ETAR and PKC signaling pathway. Our data provide evidence that cutaneous ET-1 induced mechanical allodynia not only by increasing the release of ATP from vascular endothelial cells, but also by sensitizing P2X3 receptors on nociceptive DRG neurons.


Assuntos
Endotelina-1/farmacologia , Gânglios Espinais/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Agonistas do Receptor Purinérgico P2X/farmacologia , Receptores Purinérgicos P2X3/efeitos dos fármacos , Trifosfato de Adenosina/análogos & derivados , Animais , Comportamento Animal , Fenômenos Eletrofisiológicos , Proteínas de Ligação ao GTP/efeitos dos fármacos , Gânglios Espinais/citologia , Hiperalgesia/induzido quimicamente , Masculino , Proteína Quinase C/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptor de Endotelina A/efeitos dos fármacos , Células Receptoras Sensoriais/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
7.
Neuropharmacology ; 181: 108324, 2020 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-32976891

RESUMO

Kappa opioid receptor (KOR) agonists possess adverse dysphoric and psychotomimetic effects, thus limiting their applications as non-addictive anti-pruritic and analgesic agents. Here, we showed that protein kinase C (PKC) inhibition preserved the beneficial antinociceptive and antipruritic effects of KOR agonists, but attenuated the adverse condition placed aversion (CPA), sedation, and motor incoordination in mice. Using a large-scale mass spectrometry-based phosphoproteomics of KOR-mediated signaling in the mouse brain, we observed PKC-dependent modulation of G protein-coupled receptor kinases and Wnt pathways at 5 min; stress signaling, cytoskeleton, mTOR signaling and receptor phosphorylation, including cannabinoid receptor CB1 at 30 min. We further demonstrated that inhibition of CB1 attenuated KOR-mediated CPA. Our results demonstrated the feasibility of in vivo biochemical dissection of signaling pathways that lead to side effects.


Assuntos
Proteína Quinase C/genética , Receptores Opioides kappa/genética , Transdução de Sinais/efeitos dos fármacos , (trans)-Isômero de 3,4-dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclo-hexil)-benzenoacetamida/farmacologia , Animais , Aprendizagem da Esquiva , Quinases de Receptores Acoplados a Proteína G , Masculino , Camundongos , Atividade Motora/efeitos dos fármacos , Fosfoproteínas , Fosforilação , Proteína Quinase C/efeitos dos fármacos , Proteína Quinase C/efeitos da radiação , Inibidores de Proteínas Quinases , Proteômica , Receptor CB1 de Canabinoide/efeitos dos fármacos , Receptores Acoplados a Proteínas G/efeitos dos fármacos , Receptores Opioides kappa/efeitos dos fármacos , Receptores Opioides kappa/efeitos da radiação , Serina-Treonina Quinases TOR/efeitos dos fármacos , Via de Sinalização Wnt/efeitos dos fármacos
8.
Biomed Pharmacother ; 130: 110537, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32717630

RESUMO

Rhizoma Bletillae, the tubes of Bletilla striata, has been traditionally used in China as a hemostatic agent. In preliminary studies, the major active fraction responsible for its hemostatic effect have been confirmed to be Rhizoma Bletillae polysaccharide (RBp), but the hemostatic mechanism of action of RBp is still unknown.The main aim of this study was to clarify its mechanism of hemostatic effect. RBp was prepared by 80 % ethanol precipitation of the water extract of Rhizoma Bletillae followed by the Sevag method to remove proteins. The average molecular weight (Mw) of the crude RBp maintained at a range of 30.06-200 KDa. The hemostatic effects of RBp were evaluated by testing its effect on the platelet aggregation of rat platelet-rich plasma (PRP). PRP was dealt with different concentrations of RBp and platelet aggregation was measured by the turbidimetric method. The hemostatic mechanism of RBp was investigated by examining its effect on platelet shape, platelet secretion, and activation of related receptors (P2Y1, P2Y12 and TXA2) by electron microscopy and the turbidimetric method. RBp significantly enhanced the platelet aggregations at concentrations of 50-200 mg/L in a concentration-dependent manner. The inhibitory rate of platelet aggregation was significantly increased by apyrase and Ro31-8220 in a concentration-dependent manner, while RBp-induced platelet aggregation was completely inhibited by P2Y1, P2Y12 and the PKC receptor antagonists. However, the aggregation was not sensitive to TXA2. RBp, the active ingredients of Rhizoma Bletillae responsible for its hemostatic effect, could significantly accelerate the platelet aggregation and shape change. The hemostatic mechanism may involve activation of the P2Y1, P2Y12, and PKC receptors in the adenosine diphosphate (ADP) receptor signaling pathway.


Assuntos
Hemostáticos/farmacologia , Plasma Rico em Plaquetas/efeitos dos fármacos , Polissacarídeos/farmacologia , Receptores Purinérgicos P2/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Animais , Relação Dose-Resposta a Droga , Técnicas In Vitro , Peso Molecular , Extratos Vegetais/farmacologia , Tubérculos/química , Agregação Plaquetária/efeitos dos fármacos , Proteína Quinase C/efeitos dos fármacos , Ratos , Receptores Purinérgicos P2Y1/efeitos dos fármacos , Receptores Purinérgicos P2Y12/efeitos dos fármacos
9.
Auton Neurosci ; 227: 102688, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32502943

RESUMO

The central adrenergic and noradrenergic neurotransmitter systems diffusively affect the operation of the spinal neural network and dynamically gauge central sympathetic outflow. Using in vitro splanchnic nerve-thoracic spinal cord preparations as an experimental model, this study examined the intraspinal α1-adrenoceptor-meidated modulation of sympathetic firing behaviors. Several sympathetic single-fiber activities were simultaneously recorded. Application of phenylephrine (Phe, an α1-adrenoceptor agonist) increased, decreased or did not affect spontaneous firing. A log-log plot of the change ratios of the average firing rates (AFR) versus their basal AFR displays a linear data distribution. Thus, the heterogeneity in α1-adrenoceptor-mediated responses is well described by a power law function. Phe-induced power-law firing modulation (plFM) was sensitive to prazosin (Prz, an α1-adrenoceptor antagonist). Heparin (Hep, a competitive IP3 receptor blocker) and chelerythrine (Che, a protein kinase C inhibitor) also caused plFM. Phe-induced plFM persisted in the presence of Hep; however, it was occluded by Che pretreatment. Pair-wise analysis of single-fiber activities revealed synchronous sympathetic discharges. Application of Phe, Hep or Che suppressed synchronous discharges in fiber pairs with apparent correlated firing (ACF) and induced or potentiated synchronous discharges in those without or with minimal ACF. Thus, the basal activities of the sympathetic preganglionic neurons participate in determining the responses mediated by the activation of α1-adrenoceptors. This deterministic factor, which is intrinsic to spinal neural networks, helps the supraspinal adrenergic and noradrenergic systems differentially control their widely distributed neural targets.


Assuntos
Fenômenos Eletrofisiológicos/fisiologia , Rede Nervosa/metabolismo , Proteína Quinase C/metabolismo , Receptores Adrenérgicos alfa 1/metabolismo , Medula Espinal/metabolismo , Sistema Nervoso Simpático/metabolismo , Antagonistas de Receptores Adrenérgicos alfa 1/farmacologia , Animais , Animais Recém-Nascidos , Fenômenos Eletrofisiológicos/efeitos dos fármacos , Heparina/farmacologia , Receptores de Inositol 1,4,5-Trifosfato/antagonistas & inibidores , Prazosina/farmacologia , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptores Adrenérgicos alfa 1/efeitos dos fármacos
10.
Proc Natl Acad Sci U S A ; 117(20): 10688-10698, 2020 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-32371485

RESUMO

AIDS is a pandemic disease caused by HIV that affects 37 million people worldwide. Current antiretroviral therapy slows disease progression but does not eliminate latently infected cells, which resupply active virus, thus necessitating lifelong treatment with associated compliance, cost, and chemoexposure issues. Latency-reversing agents (LRAs) activate these cells, allowing for their potential clearance, thus presenting a strategy to eradicate the infection. Protein kinase C (PKC) modulators-including prostratin, ingenol esters, bryostatin, and their analogs-are potent LRAs in various stages of development for several clinical indications. While LRAs are promising, a major challenge associated with their clinical use is sustaining therapeutically meaningful levels of the active agent while minimizing side effects. Here we describe a strategy to address this problem based on LRA prodrugs, designed for controllable release of the active LRA after a single injection. As intended, these prodrugs exhibit comparable or superior in vitro activity relative to the parent compounds. Selected compounds induced higher in vivo expression of CD69, an activation biomarker, and, by releasing free agent over time, significantly improved tolerability when compared to the parent LRAs. More generally, selected prodrugs of PKC modulators avoid the bolus toxicities of the parent drug and exhibit greater efficacy and expanded tolerability, thereby addressing a longstanding objective for many clinical applications.


Assuntos
Fármacos Anti-HIV/farmacologia , Briostatinas/farmacologia , Infecções por HIV/virologia , HIV-1/efeitos dos fármacos , Pró-Fármacos/farmacologia , Proteína Quinase C/metabolismo , Latência Viral/efeitos dos fármacos , Animais , Fármacos Anti-HIV/síntese química , Fármacos Anti-HIV/uso terapêutico , Briostatinas/síntese química , Briostatinas/uso terapêutico , Linhagem Celular Tumoral , Células Cultivadas , Diterpenos/química , Infecções por HIV/tratamento farmacológico , HIV-1/fisiologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Ésteres de Forbol/química , Pró-Fármacos/síntese química , Pró-Fármacos/uso terapêutico , Proteína Quinase C/efeitos dos fármacos
11.
Respir Physiol Neurobiol ; 278: 103446, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32360368

RESUMO

Inflammation can increase the excitability of bronchopulmonary C-fibers leading to excessive sensations and reflexes (e.g. wheeze and cough). We have previously shown modulation of peripheral nerve terminal mitochondria by antimycin A causes hyperexcitability in TRPV1-expressing bronchopulmonary C-fibers through the activation of protein kinase C (PKC). Here, we have investigated the PKC isoform responsible for this signaling. We found PKCß1, PKCδ and PKCε were expressed by many vagal neurons, with PKCα and PKCß2 expressed by subsets of vagal neurons. In dissociated vagal neurons, antimycin A caused translocation of PKCα but not the other isoforms, and only in TRPV1-lineage neurons. In bronchopulmonary C-fiber recordings, antimycin A increased the number of action potentials evoked by α,ß-methylene ATP. Selective inhibition of PKCα, PKCß1 and PKCß2 with 50 nM bisindolylmaleimide I prevented the antimycin-induced bronchopulmonary C-fiber hyperexcitability, whereas selective inhibition of only PKCß1 and PKCß2 with 50 nM LY333531 had no effect. We therefore conclude that PKCα is required for antimycin-induced increases in bronchopulmonary C-fiber excitability.


Assuntos
Antimicina A/farmacologia , Brônquios/inervação , Fibras Nervosas Amielínicas/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Gânglio Nodoso/efeitos dos fármacos , Proteína Quinase C-alfa/efeitos dos fármacos , Nervo Vago , Animais , Pulmão/inervação , Camundongos , Fibras Nervosas Amielínicas/metabolismo , Neurônios/metabolismo , Gânglio Nodoso/citologia , Gânglio Nodoso/metabolismo , Isoformas de Proteínas/efeitos dos fármacos , Isoformas de Proteínas/metabolismo , Proteína Quinase C/efeitos dos fármacos , Proteína Quinase C/metabolismo , Proteína Quinase C-alfa/metabolismo , Canais de Cátion TRPV/metabolismo
12.
Pathol Res Pract ; 215(11): 152677, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31591052

RESUMO

Sanguinarine (SAG), a benzophenanthridine alkaloid extracted from Sanguinaria canadensis, exerts antioxidant, anti-inflammatory and antiproliferative activities in a variety of malignancies. However, the underlying mechanisms by which SAG affects the tumorigenesis of gastric cancer (GC) are unclear. The common targets of SAG and GC were identified by network pharmacology, and the association of thymocyte selection-associated high mobility group box (TOX) with the clinicopathological characteristics and prognosis of patients with GC was analyzed by using datasets from The Cancer Genome Atlas (TCGA). 3-(4,5-Dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide (MTT) assays, colony formation assays, flow cytometry analysis, and a xenograft tumor model were conducted to assess the effects of SAG on the growth of GC cells, and Quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot analysis were used to determine the effects of SAG on the TOX/DNA-PKcs/KU70/80 signaling pathway. We identified 9 collective targets of SAG and GC, of which TOX expression levels were dramatically downregulated in GC tissues compared with adjacent normal tissues, and a low expression of TOX served as an independent prognostic factor of poor survival in patients with GC. SAG suppressed cell viability, colony formation and in vivo tumorigenesis and induced cell apoptosis and cell cycle arrest. Furthermore, SAG increased the expression levels of TOX but decreased those of DNA-PKcs and KU70/80 in GC cells. Our findings indicate that SAG inhibits the tumorigenesis of GC cells by regulating TOX/DNA-PKcs/KU70/80 signaling and may provide therapeutic strategies for the treatment of GC.


Assuntos
Antineoplásicos/farmacologia , Benzofenantridinas/farmacologia , Carcinogênese/efeitos dos fármacos , Isoquinolinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Neoplasias Gástricas/metabolismo , Animais , Proliferação de Células/efeitos dos fármacos , Proteínas de Grupo de Alta Mobilidade/efeitos dos fármacos , Proteínas de Grupo de Alta Mobilidade/metabolismo , Humanos , Autoantígeno Ku/efeitos dos fármacos , Autoantígeno Ku/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Nus , Proteína Quinase C/efeitos dos fármacos , Proteína Quinase C/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Med Sci Monit ; 25: 4627-4638, 2019 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-31266934

RESUMO

BACKGROUND Subclinical epileptiform discharges (SEDs) are defined as epileptiform electroencephalographic (EEG) discharges without clinical signs of seizure in patients. The subthreshold convulsant discharge (SCD) is a frequently used model for SEDs. This study aimed to investigate the effect of levetiracetam (LEV), an anti-convulsant drug, on cognitive impairment of SCD model rats and to assess the associated mechanisms. MATERIAL AND METHODS A SCD rat model was established. Rats were divided into an SCD group, an SCD+ sodium valproate (VPA) group, and an SCD+ levetiracetam (LEV) group. The Morris water maze was used to evaluate the capacity of positioning navigation and space exploration. The field excitatory post-synaptic potentials (fEPSPs) were evaluated using a bipolar stimulation electrode. NCAM, GAP43, PS95, and CaMK II levels were detected using Western blot and RT-PCR, respectively. PKC activity was examined by a non-radioactive method. RESULTS LEV shortens the latency of platform seeking in SCD rats in positioning navigation. fEPSP slopes were significantly lower in the SCD group, and LEV treatment significantly enhanced the fEPSP slopes compared to the SCD group (P<0.05). The NCAM and GAP-43 levels were increased and PSD-95 levels were increased in SCD rats (P<0.05), which were improved by LEV treatment. The PKC activity and CaMK II levels were decreased in SCD rats and LEV treatment significantly enhanced PKC activity and increased CaMK II levels. CONCLUSIONS Cognitive impairment in of SCD model rats may be caused by decreased PKC activity, low expression of CaMK II, and inhibition of LTP formation. LEV can improve cognitive function by activating the PKC-GAP-43-CaMK signal transduction pathway.


Assuntos
Disfunção Cognitiva/metabolismo , Disfunção Cognitiva/prevenção & controle , Levetiracetam/farmacologia , Animais , Proteínas Quinases Dependentes de Cálcio-Calmodulina/efeitos dos fármacos , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Disfunção Cognitiva/tratamento farmacológico , Modelos Animais de Doenças , Eletroencefalografia , Proteína GAP-43/efeitos dos fármacos , Proteína GAP-43/metabolismo , Hipocampo/metabolismo , Levetiracetam/metabolismo , Masculino , Fosforilação , Proteína Quinase C/efeitos dos fármacos , Proteína Quinase C/metabolismo , Ratos , Ratos Sprague-Dawley , Convulsões/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos , Ácido Valproico/uso terapêutico
14.
Wei Sheng Yan Jiu ; 48(2): 269-278, 2019 Mar.
Artigo em Chinês | MEDLINE | ID: mdl-31133106

RESUMO

OBJECTIVE: To investigate the effects of resveratrol(Res) on N-methyl-D-aspartate receptor(NMDAR1)and protein kinase C(PKC)expressions in the hippocampus in Alzheimer's disease(AD) rats. METHODS: The model of AD was induced by ovariectomy combined intraperitoneal injection of D-galactose(100 mg/kg). Thirty-Six female Wistar rats were randomly divided into 6 groups inculding Sham control group, AD model group, Res low dose group(20 mg/kg), Res middle dose group(40 mg/kg), Res high dose group(80 mg/kg group)and estrogen replacement therapy(ERT) group. The genes of NMDAR1 and PKC were detected by real-time PCR. NMDAR1 total protein, p-NMDAR1 protein and PKC protein were checked by Western blot. RESULTS: Compared with the Sham control group, the gene expressions and the protein expressions of NMDAR1 and PKC in the model group were decreased(P<0. 05). Moreover, compared with the model group, genes of NMDAR1 and PKC in the 3 Res dose groups were significantly increased(P<0. 05 or P<0. 01). The elavated levels of genes of NMDAR1 and PKC in ERT group were similar to the Res 80 mg/kg group(P<0. 01). p-NMDAR1/NMDAR1 and the protein expressions of PKC were also significantly increased in Res 40 mg/kg group and Res 80 mg/kg group as well as in ERT group(P<0. 05 or P<0. 01). CONCLUSION: Up-regulating the gene and protein expressions of p-NMDAR1/NMDAR1 and PKC may be one of the mechanisms of improvement of Res on the memory in AD rats.


Assuntos
Doença de Alzheimer , Hipocampo/metabolismo , Proteína Quinase C/efeitos dos fármacos , Receptores de N-Metil-D-Aspartato/efeitos dos fármacos , Resveratrol/farmacologia , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Animais , Anti-Inflamatórios não Esteroides/administração & dosagem , Modelos Animais de Doenças , Feminino , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Humanos , Proteína Quinase C/metabolismo , Distribuição Aleatória , Ratos , Ratos Wistar , Receptores de N-Metil-D-Aspartato/metabolismo
15.
Acta Cir Bras ; 33(7): 556-564, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-30110057

RESUMO

PURPOSE: To investigate the effects of baicalin on inflammatory reaction, oxidative stress and protein kinase D1 (PKD1) and nuclear factor-kappa B (NF-κB) protein expressions in severe acute pancreatitis (SAP) rats. METHODS: Sixty rats were divided into sham operation, model, and low-, medium- and high-dose baicalin group. SAP model was established in later 4 groups. The later 3 groups were injected with 0.1, 0.2 and 0.4 ml/100 g 5% baicalin injection, respectively. At 12 h, the serum SAP related indexes and inflammatory factors, peripheral blood CD3 and γδT cell percentages, wet/dry ratio and pancreas ascites volume, oxidative stress indexes and PKD1 and NF-κB protein expressions in pancreatic tissue were determined. RESULTS: Compared with model group, in high-dose baicalin group the wet/dry ratio and ascites volume, serum amylase level, phospholipase A2 activity, TNF-α, IL-1 and IL-6 levels, and pancreatic malondialdehyde level and PKD1 and NF-κB protein expression were significantly decreased (P < 0.05), and peripheral blood CD3 and γδT cell percentages and pancreatic superoxide dismutase and glutathione peroxidase levels were significantly increased (P < 0.05). CONCLUSION: Baicalin can resist the inflammatory reaction and oxidative stress, and down-regulate protein kinase D1 and nuclear factor-kappa B protein expressions, thus exerting the protective effects on severe acute pancreatitis in rats.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Flavonoides/farmacologia , NF-kappa B/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Pancreatite/tratamento farmacológico , Proteína Quinase C/metabolismo , Amilases/sangue , Amilases/efeitos dos fármacos , Animais , Complexo CD3/sangue , Complexo CD3/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Glutationa Peroxidase/efeitos dos fármacos , Glutationa Peroxidase/metabolismo , Interleucina-1/sangue , Interleucina-6/sangue , Malondialdeído/metabolismo , NF-kappa B/efeitos dos fármacos , Pancreatite/metabolismo , Proteína Quinase C/efeitos dos fármacos , Distribuição Aleatória , Ratos Sprague-Dawley , Reprodutibilidade dos Testes , Superóxido Dismutase/efeitos dos fármacos , Superóxido Dismutase/metabolismo , Resultado do Tratamento , Fator de Necrose Tumoral alfa/sangue , Fator de Necrose Tumoral alfa/efeitos dos fármacos
16.
Acta cir. bras ; 33(7): 556-564, July 2018. tab, graf
Artigo em Inglês | LILACS | ID: biblio-949368

RESUMO

Abstract Purpose: To investigate the effects of baicalin on inflammatory reaction, oxidative stress and protein kinase D1 (PKD1) and nuclear factor-kappa B (NF-κB) protein expressions in severe acute pancreatitis (SAP) rats. Methods: Sixty rats were divided into sham operation, model, and low-, medium- and high-dose baicalin group. SAP model was established in later 4 groups. The later 3 groups were injected with 0.1, 0.2 and 0.4 ml/100 g 5% baicalin injection, respectively. At 12 h, the serum SAP related indexes and inflammatory factors, peripheral blood CD3 and γδT cell percentages, wet/dry ratio and pancreas ascites volume, oxidative stress indexes and PKD1 and NF-κB protein expressions in pancreatic tissue were determined. Results: Compared with model group, in high-dose baicalin group the wet/dry ratio and ascites volume, serum amylase level, phospholipase A2 activity, TNF-α, IL-1 and IL-6 levels, and pancreatic malondialdehyde level and PKD1 and NF-κB protein expression were significantly decreased (P < 0.05), and peripheral blood CD3 and γδT cell percentages and pancreatic superoxide dismutase and glutathione peroxidase levels were significantly increased (P < 0.05). Conclusion: Baicalin can resist the inflammatory reaction and oxidative stress, and down-regulate protein kinase D1 and nuclear factor-kappa B protein expressions, thus exerting the protective effects on severe acute pancreatitis in rats.


Assuntos
Animais , Pancreatite/tratamento farmacológico , Flavonoides/farmacologia , Proteína Quinase C/metabolismo , Anti-Inflamatórios não Esteroides/farmacologia , NF-kappa B/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Pancreatite/metabolismo , Superóxido Dismutase/efeitos dos fármacos , Proteína Quinase C/efeitos dos fármacos , Distribuição Aleatória , Regulação para Baixo/efeitos dos fármacos , Reprodutibilidade dos Testes , NF-kappa B/efeitos dos fármacos , Interleucina-6/sangue , Interleucina-1/sangue , Fator de Necrose Tumoral alfa/sangue , Resultado do Tratamento , Ratos Sprague-Dawley , Complexo CD3/efeitos dos fármacos , Complexo CD3/sangue , Glutationa Peroxidase/efeitos dos fármacos , Glutationa Peroxidase/metabolismo , Amilases/efeitos dos fármacos , Amilases/sangue , Malondialdeído/metabolismo
17.
Med Sci Monit ; 24: 1955-1961, 2018 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-29610452

RESUMO

BACKGROUND Like other human cancers, the malignancy of cervical cancer is also characterized by abilities of proliferation, migration, and invasion. Protein kinase C-zeta (PKCζ) has been highly correlated with several human cancers. Baicalin was proven to regulate PKC. This study aimed to investigate the anti-cancer effect and involved molecular mechanisms of baicalin on human cervical cancer. MATERIAL AND METHODS Baicalin at various concentrations was used to treat 2 human cervical cancer cell lines HeLa and SiHa. The proliferation was assessed by 3-(4,5-dimethylthiazol-2-yl)-2,5 diphenylterazolium bromide (MTT) assay. The apoptosis was detected by terminal transferase UTP nick end labeling (TUNEL) assay. Wound healing assay and Transwell assay were used to evaluate the migration and invasion respectively. Western blotting was performed to assess the protein expression levels. RESULTS Baicalin administration significantly reduced the viability by facilitating the apoptosis in HeLa and SiHa cells. Baicalin treatment also significantly reduced the wound closure and cell amount invaded as measured by Transwell assay. The expression levels of PKCζ, survivin, matrix metalloproteinase (MMP)2, MMP9 as well as the phosphorylation of signal transducer and activator of transcription (STAT) 3 were reduced in baicalin administrated cervical cancer cells. CONCLUSIONS Baicalin exerted anti-cancer effects on human cervical cancer cells by targeting STAT3 regulated signaling pathways.


Assuntos
Flavonoides/farmacologia , Proteína Quinase C/efeitos dos fármacos , Fator de Transcrição STAT3/metabolismo , Neoplasias do Colo do Útero/tratamento farmacológico , Neoplasias do Colo do Útero/metabolismo , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Feminino , Células HeLa , Humanos , Proteínas Inibidoras de Apoptose/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Fosforilação , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , Fator de Transcrição STAT3/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Survivina , Neoplasias do Colo do Útero/enzimologia , Neoplasias do Colo do Útero/patologia
18.
Arch Oral Biol ; 89: 49-54, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29471192

RESUMO

OBJECTIVE: Magnolol, a polyphenol compound from herbal medicines, was shown to alter physiology in various cell models. However, the effect of magnolol on Ca2+ homeostasis and its related physiology in oral cancer cells is unclear. This study examined whether magnolol altered Ca2+ signaling and cell viability in OC2 human oral cancer cells. METHODS: Cytosolic Ca2+ concentrations ([Ca2+]i) in suspended cells were measured by using the fluorescent Ca2+-sensitive dye fura-2. Cell viability was examined by 4-[3-[4-lodophenyl]-2-4(4-nitrophenyl)-2H-5-tetrazolio-1,3-benzene disulfonate] water soluble tetrazolium-1 (WST-1) assay. RESULTS: Magnolol at concentrations of 20-100 µM induced [Ca2+]i rises. Ca2+ removal reduced the signal by approximately 50%. Magnolol (100 µM) induced Mn2+ influx suggesting of Ca2+ entry. Magnolol-induced Ca2+ entry was partially suppressed by protein kinase C (PKC) regulators, and inhibitors of store-operated Ca2+ channels. In Ca2+-free medium, treatment with the endoplasmic reticulum Ca2+ pump inhibitor 2,5-di-tert-butylhydroquinone (BHQ) abolished magnolol-evoked [Ca2+]i rises. Conversely, treatment with magnolol abolished BHQ-evoked [Ca2+]i rises. Inhibition of phospholipase C (PLC) with U73122 partially inhibited magnolol-induced [Ca2+]i rises. Magnolol at 20-100 µM decreased cell viability, which was not reversed by pretreatment with the Ca2+ chelator 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid-acetoxymethyl ester (BAPTA/AM). CONCLUSIONS: Together, in OC2 cells, magnolol induced [Ca2+]i rises by evoking partially PLC-dependent Ca2+ release from the endoplasmic reticulum and Ca2+ entry via PKC-sensitive store-operated Ca2+ entry. Magnolol also caused Ca2+-independent cell death. Therefore, magnolol-induced cytotoxicity may not be involved in activation mechanisms associated with intracellular Ca2+ mobilization in oral cancer cells.


Assuntos
Compostos de Bifenilo/farmacologia , Cálcio/metabolismo , Homeostase/efeitos dos fármacos , Lignanas/farmacologia , Neoplasias Bucais/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Linhagem Celular Tumoral/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Citosol/metabolismo , Retículo Endoplasmático/metabolismo , Fura-2/farmacologia , Humanos , Manganês/metabolismo , Proteína Quinase C/efeitos dos fármacos , Proteína Quinase C/metabolismo , Sais de Tetrazólio , Fosfolipases Tipo C/efeitos dos fármacos
19.
Cell Mol Neurobiol ; 38(3): 691-701, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28779332

RESUMO

Critical and major operations are often accompanied by brain ischemic complications. Previous studies found that propofol post-conditioning provided neuroprotective functions through upregulating the expression of potassium chloride cotransporter 2 (KCC2) in gamma-aminobutyric acid (GABA) interneurons. Membrane expression and phosphorylation represents KCC2 activity, which were modulated by a protein kinase C (PKC)-dependent mechanism. However, the role of propofol in increasing KCC2 phosphorylation and the involvement of protein kinase Mζ (PKMζ), a major subtype of PKC, in the KCC2 pathway remained unclear. In this study, we established middle cerebral artery occlusion model in rats to evaluate the long-term recovery of brain functions using behavioral experiments. KCC2 and PKMζ were assessed via western blot. We used the selective inhibitor, zeta inhibitory peptide (ZIP), to investigate the relationship between KCC2 and PKMζ. Intracellular chloride concentration in the hippocampal CA1 area was measured to determine KCC2 activity. We found that propofol, infused at a speed of 20 mg kg-1 h-1 for 2 h at the onset of reperfusion, improved neurological deficits and cognitive dysfunction following ischemia/reperfusion injury. PKMζ expression was significantly upregulated, which improved KCC2 membrane expression and phosphorylation in the ischemic hippocampal CA1 area, and these effects could last up to 28 days. But ZIP inhibited this process. Ultimately, we showed that propofol increased KCC2 phosphorylation and PKMζ was the upstream of KCC2. Propofol led to long-term recovery of brain functions by upregulating the activity of the PKMζ/KCC2 pathway.


Assuntos
Neuroproteção/efeitos dos fármacos , Propofol/farmacologia , Proteína Quinase C/efeitos dos fármacos , Simportadores/efeitos dos fármacos , Animais , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Interneurônios/efeitos dos fármacos , Interneurônios/metabolismo , Masculino , Ratos Sprague-Dawley , Traumatismo por Reperfusão/tratamento farmacológico , Traumatismo por Reperfusão/metabolismo , Ácido gama-Aminobutírico/metabolismo , Ácido gama-Aminobutírico/farmacologia
20.
JCI Insight ; 2(21)2017 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-29093271

RESUMO

Advanced basal cell carcinomas (BCCs) circumvent Smoothened (SMO) inhibition by activating GLI transcription factors to sustain the high levels of Hedgehog (HH) signaling required for their survival. Unfortunately, there is a lack of efficacious therapies. We performed a gene expression-based drug repositioning screen in silico and identified the FDA-approved histone deacetylase (HDAC) inhibitor, vorinostat, as a top therapeutic candidate. We show that vorinostat only inhibits proliferation of BCC cells in vitro and BCC allografts in vivo at high dose, limiting its usefulness as a monotherapy. We leveraged this in silico approach to identify drug combinations that increase the therapeutic window of vorinostat and identified atypical PKC Ɩ/ʎ (aPKC) as a HDAC costimulator of HH signaling. We found that aPKC promotes GLI1-HDAC1 association in vitro, linking two positive feedback loops. Combination targeting of HDAC1 and aPKC robustly inhibited GLI1, lowering drug doses needed in vitro, in vivo, and ex vivo in patient-derived BCC explants. We identified a bioavailable and selective small-molecule aPKC inhibitor, bringing the pharmacological blockade of aPKC and HDAC1 into the realm of clinical possibility. Our findings provide a compelling rationale and candidate drugs for combined targeting of HDAC1 and aPKC in HH-dependent cancers.


Assuntos
Carcinoma Basocelular/tratamento farmacológico , Histona Desacetilase 1/efeitos dos fármacos , Inibidores de Histona Desacetilases/farmacologia , Isoenzimas/efeitos dos fármacos , Proteína Quinase C/efeitos dos fármacos , Neoplasias Cutâneas/tratamento farmacológico , Aloenxertos , Animais , Carcinoma Basocelular/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Biologia Computacional , Combinação de Medicamentos , Descoberta de Drogas , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Ouriços/genética , Ouriços/metabolismo , Histona Desacetilase 1/genética , Histona Desacetilase 1/metabolismo , Inibidores de Histona Desacetilases/química , Isoenzimas/metabolismo , Camundongos , Camundongos Knockout , Proteína Quinase C/metabolismo , Transdução de Sinais , Fatores de Transcrição/efeitos dos fármacos , Fatores de Transcrição/genética , Proteína GLI1 em Dedos de Zinco/genética , Proteína GLI1 em Dedos de Zinco/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...