Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Eur J Med Chem ; 78: 294-303, 2014 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-24686016

RESUMO

In our drug discovery program, a series of 2-thioxo-pyrazolo[1,5-a][1,3,5]triazin-4-ones were designed, synthesized and evaluated for their TP inhibitory potential. All the synthesized analogues conferred a varying degree of TP inhibitory activity, comparable or better than positive control, 7-deazaxanthine (7-DX, 2) (IC50 value = 42.63 µM). A systematic approach to the lead optimization identified compounds 3c and 4a as the most promising TP inhibitors, exhibiting mixed mode of enzyme inhibition. Moreover, selected compounds demonstrated the ability to attenuate the expression of the angiogenic markers (viz. MMP-9 and VEGF) in MDA-MB-231 cells at sublethal concentrations. In addition, molecular docking studies revealed the plausible binding orientation of these inhibitors towards TP, which was in accordance with the experimental results. Taken as a whole, these compounds would constitute a new direction for the design of novel TP inhibitors with promising antiangiogenic properties.


Assuntos
Inibidores da Angiogênese/farmacologia , Descoberta de Drogas , Inibidores Enzimáticos/farmacologia , Neovascularização Patológica/tratamento farmacológico , Pirazóis/farmacologia , Compostos de Enxofre/farmacologia , Timidina Fosforilase/antagonistas & inibidores , Triazinas/farmacologia , Inibidores da Angiogênese/síntese química , Inibidores da Angiogênese/química , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Humanos , Metaloproteinase 9 da Matriz/biossíntese , Modelos Moleculares , Estrutura Molecular , Pirazóis/síntese química , Pirazóis/química , Relação Estrutura-Atividade , Compostos de Enxofre/síntese química , Compostos de Enxofre/química , Timidina Fosforilase/metabolismo , Triazinas/síntese química , Triazinas/química , Fator A de Crescimento do Endotélio Vascular/biossíntese
2.
Eur J Med Chem ; 67: 325-34, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23871912

RESUMO

Thirty-three 1,2,4-triazolo[1,5-a][1,3,5]triazin-5,7-dione and its 5-thioxo analogues were designed and synthesized which contained different substituents at meta- and/or para-positions of 2-phenyl or 2-benzyl ring attached to the fused ring structure. The preliminary pharmacological evaluation demonstrated that the 5-thioxo analogues of 1,2,4-triazolo[1,5-a][1,3,5]triazine exhibited a varying degree of inhibitory activity towards thymidine phosphorylase, comparable or better than reference compound, 7-Deazaxanthine (7-DX, 2) (IC50 value = 42.63 µM). Moreover, compounds 5q and 6i displayed a mixed-type of inhibitory mechanism in the presence of variable concentrations of thymidine (dThd). In addition, selected compounds were found to have a noticeable inhibitory effect on the expression of angiogenesis markers, including VEGF and MMP-9 in MDA-MB-231 breast cancer cells.


Assuntos
Inibidores da Angiogênese/farmacologia , Biomarcadores Tumorais/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Metaloproteinase 9 da Matriz/metabolismo , Timidina Fosforilase/antagonistas & inibidores , Triazinas/farmacologia , Triazóis/farmacologia , Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Inibidores da Angiogênese/síntese química , Inibidores da Angiogênese/química , Biomarcadores Tumorais/biossíntese , Biomarcadores Tumorais/metabolismo , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Humanos , Cinética , Metaloproteinase 9 da Matriz/biossíntese , Estrutura Molecular , Relação Estrutura-Atividade , Timidina Fosforilase/metabolismo , Triazinas/síntese química , Triazinas/química , Triazóis/síntese química , Triazóis/química , Fatores de Crescimento do Endotélio Vascular/biossíntese , Fatores de Crescimento do Endotélio Vascular/metabolismo
3.
Int J Oncol ; 42(5): 1605-12, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23467622

RESUMO

Since its isolation from Tripterygium wilfordii in 1972, triptolide has been shown to possess potent anticancer activity against a variety of cancers, and has entered phase I clinical trial. It is a diterpenoid triepoxide that acts through multiple molecular targets and signaling pathways. The mitogen-activated protein kinases are well known for their modulation of cell survival and proliferation. In particular, the ERK pathway has a dual role in cell proliferation and cell death. Thus far, data on the effect of triptolide on ERK signaling remain limited. In our current study, we have shown for the first time that ERK activation rather than inhibition occurred in a dose- and time-dependent manner following triptolide treatment in MDA-MB-231 breast cancer cells. ERK activation was crucial in mediating triptolide-induced caspase-dependent apoptosis. Tritpolide-induced ERK activation modulated the expression of the Bcl-2 protein family member Bax but was not involved in the downregulation of Bcl-xL expression. Signals acted upstream of ERK activation included generation of reactive oxygen species (ROS) and endoplasmic reticulum stress predominantly via the PERK­eIF2α pathway, as the MEK inhibitor U0126 did not inhibit the phosphorylation of PERK and eIF2α or the generation of ROS.


Assuntos
Apoptose/efeitos dos fármacos , Neoplasias da Mama/genética , Diterpenos/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Fenantrenos/farmacologia , Neoplasias da Mama/metabolismo , Butadienos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Ensaios Clínicos como Assunto , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Estresse do Retículo Endoplasmático/genética , Compostos de Epóxi/farmacologia , Fator de Iniciação 2 em Eucariotos/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Humanos , MAP Quinase Quinase Quinases/antagonistas & inibidores , MAP Quinase Quinase Quinases/metabolismo , Nitrilas/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/genética , Espécies Reativas de Oxigênio , Transdução de Sinais/efeitos dos fármacos , eIF-2 Quinase/metabolismo
4.
J Control Release ; 172(3): 852-61, 2013 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-24459693

RESUMO

Liposome co-encapsulation of synergistic anti-cancer drug combination is an emerging area that has demonstrated therapeutic benefit in clinical trials. Remote loading of two or more drugs into a single liposome constitutes a new challenge that calls for a re-examination of drug loading strategies to allow the loading of the drug combination efficiently and with high drug content. In this study, the Mn(2+) gradient coupled with A23187 ionophore was applied in the sequential co-encapsulation of doxorubicin and irinotecan, as this drug loading method is capable of remotely loading drugs by apparently two different mechanisms, namely, coordination complexation and pH gradient. Doxorubicin and irinotecan could be co-encapsulated into liposomes in a wide range of drug-to-drug ratios, with encapsulation efficiencies of > 80%. The total encapsulated drug content was non-linearly correlated with increases in the intraliposomal Mn(2+) concentration, with a maximum total drug-to-lipid molar ratio of 0.8:1 achieved with 600 mM Mn(2+). This high encapsulated drug content did not affect the stability of the co-encapsulated liposomes upon storage for six months. Regardless of the encapsulated drug amount, the liposomes did not exhibit the fiber bundle precipitate morphology but rather an undefined structural organization in the aqueous core. The co-encapsulated liposome formulation was further tested in an intraperitoneally grown, human ovarian tumor xenograft model, and was shown to significantly improve the survival of the tumor-bearing animals. The improvement in therapeutic efficacy was possibly due to the increase in systemic drug exposure, with the maintenance of the synergistic molar drug ratio of 1:1 in circulation.


Assuntos
Antineoplásicos/administração & dosagem , Camptotecina/análogos & derivados , Doxorrubicina/administração & dosagem , Neoplasias Ovarianas/tratamento farmacológico , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Camptotecina/administração & dosagem , Camptotecina/farmacologia , Camptotecina/uso terapêutico , Linhagem Celular Tumoral , Doxorrubicina/farmacologia , Doxorrubicina/uso terapêutico , Sinergismo Farmacológico , Feminino , Humanos , Irinotecano , Lipossomos/química , Manganês/química , Camundongos , Camundongos SCID , Neoplasias Ovarianas/patologia , Ovário/efeitos dos fármacos , Ovário/patologia
5.
Int J Nanomedicine ; 7: 651-61, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22334787

RESUMO

BACKGROUND: Realizing the therapeutic benefits of quercetin is mostly hampered by its low water solubility and poor absorption. In light of the advantages of nanovehicles in the delivery of flavanoids, we aimed to deliver quercetin perorally with nanomicelles made from the diblock copolymer, polyethylene glycol (PEG)-derivatized phosphatidylethanolamine (PE). METHODS: Quercetin-loaded nanomicelles were prepared by using the film casting method, and were evaluated in terms of drug incorporation efficiency, micelle size, interaction with Caco-2 cells, and anticancer activity in the A549 lung cancer cell line and murine xenograft model. RESULTS: The incorporation efficiency into the nanomicelles was ≥88.9% when the content of quercetin was up to 4% w/w, with sizes of 15.4-18.5 nm and polydispersity indices of <0.250. Solubilization of quercetin by the nanomicelles increased its aqueous concentration by 110-fold. The quercetin nanomicelles were stable when tested in simulated gastric (pH 1.2) and intestinal (pH 7.4) fluids, and were non-toxic to the Caco-2 cells as reflected by reversible reduction in transepithelial electrical resistance and ≤25% lactose dehydrogenase release. The anticancer activity of quercetin could be significantly improved over the free drug through the nanomicellar formulation when tested using the A549 cancer cell line and murine xenograft model. The nanomicellar quercetin formulation was well tolerated by the tumor-bearing animals, with no significant weight loss observed at the end of the 10-week study period. CONCLUSION: A stable PEG-PE nanomicellar formulation of quercetin was developed with enhanced peroral anticancer activity and no apparent toxicity to the intestinal epithelium.


Assuntos
Antineoplásicos/administração & dosagem , Neoplasias Pulmonares/tratamento farmacológico , Micelas , Nanopartículas/química , Quercetina/administração & dosagem , Quercetina/química , Administração Oral , Análise de Variância , Animais , Antineoplásicos/química , Antioxidantes/administração & dosagem , Antioxidantes/química , Células CACO-2 , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Portadores de Fármacos/administração & dosagem , Portadores de Fármacos/química , Estabilidade de Medicamentos , Feminino , Humanos , Concentração de Íons de Hidrogênio , Neoplasias Pulmonares/patologia , Camundongos , Nanopartículas/administração & dosagem , Tamanho da Partícula , Fosfatidiletanolaminas/administração & dosagem , Fosfatidiletanolaminas/química , Polietilenoglicóis/administração & dosagem , Polietilenoglicóis/química , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Int J Nanomedicine ; 6: 2253-63, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22125408

RESUMO

Although carbon nanomaterials (CNMs) have been increasingly studied for their biomedical applications, there is limited research on these novel materials for oral drug delivery. As such, this study aimed to explore the potential of CNMs in oral drug delivery, and the objectives were to evaluate CNM cytotoxicity and their abilities to modulate paracellular transport and the P-glycoprotein (P-gp) efflux pump. Three types of functionalized CNMs were studied, including polyhydroxy small-gap fullerenes (OH-fullerenes), carboxylic acid functionalized single-walled carbon nanotubes (f SWCNT-COOH) and poly(ethylene glycol) functionalized single-walled carbon nanotubes (f SWCNT-PEG), using the well-established Caco-2 cell monolayer to represent the intestinal epithelium. All three CNMs had minimum cytotoxicity on Caco-2 cells, as demonstrated through lactose dehydrogenase release and 3-(4,5-dimethyliazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assays. Of the three CNMs, f SWCNT-COOH significantly reduced transepithelial electrical resistance and enhanced transport of Lucifer Yellow across the Caco-2 monolayer. Confocal fluorescence microscopy showed that f SWCNT-COOH treated cells had the highest perturbation in the distribution of ZO-1, a protein marker of tight junction, suggesting that f SWCNT-COOH could enhance paracellular permeability via disruption of tight junctions. This modulating effect of f SWCNT-COOH can be reversed over time. Furthermore, cellular accumulation of the P-gp substrate, rhodamine-123, was significantly increased in cells treated with f SWCNT-COOH, suggestive of P-gp inhibition. Of note, f SWCNT-PEG could increase rhodamine-123 accumulation without modifying the tight junction. Collectively, these results suggest that the functionalized CNMs could be useful as modulators for oral drug delivery, and the differential effects on the intestinal epithelium imparted by different types of CNMs would create unique opportunities for drug-specific oral delivery applications.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Fulerenos/administração & dosagem , Fulerenos/química , Nanotubos de Carbono/química , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Administração Oral , Análise de Variância , Transporte Biológico , Células CACO-2 , Permeabilidade da Membrana Celular , Sobrevivência Celular/efeitos dos fármacos , Impedância Elétrica , Fulerenos/farmacocinética , Humanos , Mucosa Intestinal , Isoquinolinas/farmacocinética , Rodaminas/administração & dosagem , Rodaminas/farmacocinética , Junções Íntimas/efeitos dos fármacos , Junções Íntimas/metabolismo
7.
Oncol Rep ; 26(5): 1315-21, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21805042

RESUMO

Triptolide, a diterpene from Tripterygium wilfordii, has been shown to have potent anticancer activity, exerting its effects through multiple molecular targets and signaling pathways. Yet, its effect on focal adhesion kinase (FAK), a non-receptor tyrosine kinase overexpressed in breast cancer that regulates cellular adhesion and survival, has not been reported. The current study is the first to report on the effect of triptolide on FAK expression, cell adhesion and survival using MCF-7 breast cancer cells. Triptolide significantly reduced MCF-7 anchorage-independent growth in a concentration-dependent manner. Cell rounding and detachment from culture plates were observed as early as 8 h, with significant cell detachment observed after 24 h of triptolide treatment. The adhesion potential of triptolide-treated MCF-7 cells to Matrigel was also compromised. Triptolide induced concentration- and time-dependent cleavage of FAK and PARP, which was dependent on caspase activation. The pan-caspase inhibitor, zVAD-fmk, was the only inhibitor that could significantly reduce FAK and PARP cleavage and cell detachment. However, the presence of zVAD-fmk failed to significantly reverse triptolide-induced cell death. Finally, triptolide-induced FAK cleavage was specific to MCF-7 cells, as no cleaved FAK was observed in MDA-MB-231 cells. In conclusion, our data present the first evidence of triptolide-mediated induction of FAK cleavage that correlates with cell detachment and loss of adhesion potential to the extracellular matrix.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Diterpenos/farmacologia , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Fenantrenos/farmacologia , Antineoplásicos Alquilantes/farmacologia , Neoplasias da Mama/enzimologia , Neoplasias da Mama/patologia , Adesão Celular/efeitos dos fármacos , Processos de Crescimento Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Compostos de Epóxi/farmacologia , Feminino , Proteína-Tirosina Quinases de Adesão Focal/antagonistas & inibidores , Humanos
8.
Int J Oncol ; 37(1): 211-8, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20514413

RESUMO

In the management of metastatic breast cancer, fewer recognized therapeutic standards are available as compared to the early stages of the disease. Thus, it is pertinent to search for new, effective therapy to improve survival, tolerability and quality of life of patients. In this study, a liposomal formulation was developed for a novel dihydrofolate reductase (DHFR) inhibitor, M-V-05. Drug encapsulation into liposomes was achieved by the citrate-based, pH gradient loading technique, with a final drug-to-lipid weight ratio of 0.1:1. The liposome formulation exhibited a sustained release profile of the encapsulated drug that followed first order release kinetics. Liposomal M-V-05 was found to be more effective than the standard DHFR inhibitor, methotrexate, and its activity was comparable to liposomal doxorubicin, with IC50 values of 37 and 59 microM achieved in MDA-MB-231 and JIMT-1 cells, respectively. Similar cytotoxicity was observed in primary patient samples of invasive ductal carcinoma of the breast. The combination of liposomal M-V-05 and liposomal doxorubicin in fixed molar ratio of 3:1 was additive in cytotoxicity, allowing the concentrations of liposomal doxorubicin and liposomal M-V-05 to be reduced by 62 and 46%, respectively. Taken together, liposomal M-V-05 represents a promising agent and offers a potential new adjuvant therapy for breast cancer treatment.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Carcinoma/tratamento farmacológico , Antagonistas do Ácido Fólico/administração & dosagem , Lipossomos/síntese química , Lipossomos/uso terapêutico , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacocinética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Carcinoma/metabolismo , Carcinoma/patologia , Sobrevivência Celular/efeitos dos fármacos , Química Farmacêutica , Relação Dose-Resposta a Droga , Composição de Medicamentos/métodos , Avaliação Pré-Clínica de Medicamentos , Feminino , Antagonistas do Ácido Fólico/farmacocinética , Humanos , Concentração de Íons de Hidrogênio , Lipossomos/metabolismo , Modelos Biológicos , Compostos de Espiro/administração & dosagem , Compostos de Espiro/síntese química , Compostos de Espiro/uso terapêutico , Tetra-Hidrofolato Desidrogenase/metabolismo , Fatores de Tempo , Triazinas/administração & dosagem , Triazinas/síntese química , Triazinas/uso terapêutico , Células Tumorais Cultivadas
9.
Curr Drug Metab ; 10(8): 861-74, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20214582

RESUMO

The use of drug cocktails has become a widely adopted strategy in clinical cancer therapy. Cytotoxic drug cocktails are often administered based on maximum tolerated dose (MTD) of each agent, with the belief of achieving maximum cell kill through tolerable toxicity level. Yet, MTD administration may not have fully captured the therapeutic synergism that exists among the individual agents in the drug cocktail, as the response to a cocktail regimen, that is, whether the effect is synergistic or not, could be highly sensitive to the concentration ratios of the individual drugs at the site of action. It is important to realize that the inherently different pharmacokinetic profiles of the individual agents could have significant influence on the response to an anti-cancer drug cocktail by dictating the amount of the individual agents reaching the tumor site and therefore the concentration ratios. Furthermore, the individual agents may have unfavorable pharmacokinetic interactions that add to the difficulty in determining the therapeutic and/or toxicological effects of the drug cocktail. In this review, we will focus on how lipid-based nanoparticulate systems could address the above issues associated with anti-cancer drug cocktails. Specifically, we will highlight the use of liposome systems as the means to control and coordinate the delivery of various anti-cancer drug cocktails, encompassing conventional chemotherapeutics, chemosensitizing agents and molecularly targeted agents.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Drogas em Investigação/uso terapêutico , Lipossomos/uso terapêutico , Nanopartículas/uso terapêutico , Neoplasias/tratamento farmacológico , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/química , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Portadores de Fármacos/química , Portadores de Fármacos/farmacocinética , Portadores de Fármacos/uso terapêutico , Sistemas de Liberação de Medicamentos/métodos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Drogas em Investigação/efeitos adversos , Drogas em Investigação/química , Drogas em Investigação/farmacocinética , Humanos , Lipossomos/química , Modelos Biológicos , Nanopartículas/química
10.
Arch Biochem Biophys ; 479(2): 170-8, 2008 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-18835241

RESUMO

PRK2/PKNgamma is a Rho effector and a member of the protein kinase C superfamily of serine/threonine kinases. Here, we explore the structure-function relationship between various motifs in the C-terminal half of PRK2 and its kinase activity and regulation. We report that two threonine residues at conserved phosphoacceptor position in the activation loop and the turn motif are essential for the catalytic activity of PRK2, but the phosphomimetic Asp-978 at hydrophobic motif is dispensable for kinase catalytic competence. Moreover, the PRK2-Delta958 mutant with the turn motif truncated still interacts with 3-phosphoinositide-dependent kinase-1 (PDK-1). Thus, both the intact hydrophobic motif and the turn motif in PRK2 are dispensable for the binding of PDK-1. We also found that while the last seven amino acid residues at the C-terminus of PRK2 are not required for the activation of the kinase by RhoA in vitro, however, the extreme C-terminal segment is critical for the full activation of PRK2 by RhoA in cells in a GTP-dependent manner. Our data suggest that the extreme C-terminus of PRK2 may represent a potential drug target for effector-specific pharmacological intervention of Rho-medicated biological processes.


Assuntos
Proteína Quinase C/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Motivos de Aminoácidos/fisiologia , Animais , Células COS , Chlorocebus aethiops , Ativação Enzimática/fisiologia , Humanos , Interações Hidrofóbicas e Hidrofílicas , Proteína Quinase C/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Estrutura Terciária de Proteína/fisiologia , Piruvato Desidrogenase Quinase de Transferência de Acetil , Proteína rhoA de Ligação ao GTP/genética
11.
J Biol Chem ; 281(41): 30768-81, 2006 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-16895917

RESUMO

The segment C-terminal to the hydrophobic motif at the V5 domain of protein kinase C (PKC) is the least conserved both in length and in amino acid identity among all PKC isozymes. By generating serial truncation mutants followed by biochemical and functional analyses, we show here that the very C terminus of PKCalpha is critical in conferring the full catalytic competence to the kinase and for transducing signals in cells. Deletion of one C-terminal amino acid residue caused the loss of approximately 60% of the catalytic activity of the mutant PKCalpha, whereas deletion of 10 C-terminal amino acid residues abrogated the catalytic activity of PKCalpha in immune complex kinase assays. The PKCalpha C-terminal truncation mutants were found to lose their ability to activate mitogen-activated protein kinase, to rescue apoptosis induced by the inhibition of endogenous PKC in COS cells, and to augment melatonin-stimulated neurite outgrowth. Furthermore, molecular dynamics simulations revealed that the deletion of 1 or 10 C-terminal residues results in the deformation of the V5 domain and the ATP-binding pocket, respectively. Finally, PKCalpha immunoprecipitated using an antibody against its C terminus had only marginal catalytic activity compared with that of the PKCalpha immunoprecipitated by an antibody against its N terminus. Therefore, the very C-terminal tail of PKCalpha is a novel determinant of the catalytic activity of PKC and a promising target for selective modulation of PKCalpha function. Molecules that bind preferentially to the very C terminus of distinct PKC isozymes and suppress their catalytic activity may constitute a new class of selective inhibitors of PKC.


Assuntos
Proteína Quinase C-alfa/química , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Células COS , Domínio Catalítico , Bovinos , Chlorocebus aethiops , Dados de Sequência Molecular , Neurônios/metabolismo , Proteína Quinase C-alfa/metabolismo , Estrutura Terciária de Proteína , Ratos , Homologia de Sequência de Aminoácidos , Transdução de Sinais
12.
Cell Signal ; 18(9): 1473-81, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16427251

RESUMO

PRK1 is a lipid- and Rho GTPase-activated serine/threonine protein kinase implicated in the regulation of receptor trafficking, cytoskeletal dynamics and tumorigenesis. Although Rho binding has been mapped to the HR1 region in the regulatory domain of PRK1, the mechanism involved in the control of PRK1 activation following Rho binding is poorly understood. We now provide the first evidence that the very C-terminus beyond the hydrophobic motif in PRK1 is essential for the activation of this kinase by RhoA. Deletion of the HR1 region did not completely abolish the binding of PRK1-DeltaHR1 to GTPgammaS-RhoA nor the activation of this mutant by GTPgammaS-RhoA in vitro. In contrast, removing of the last six amino acid residues from the C-terminus of PRK1 or truncating of a single C-terminal residue from PRK1-DeltaHR1 completely abrogated the activation of these mutants by RhoA both in vitro and in vivo. The critical dependence of the very C-terminus of PRK1 on the signaling downstream of RhoA was further demonstrated by the failure of the PRK1 mutant lacking its six C-terminal residues to augment lisophosphatidic acid-elicited neurite retraction in neuronal cells. Thus, we show that the HR1 region is necessary but not sufficient in eliciting a full activation of PRK1 upon binding of RhoA. Instead, such activation is controlled by the very C-terminus of PRK1. Our results also suggest that the very C-terminus of PRK1, which is the least conserved among members of the protein kinase C superfamily, is a potential drug target for pharmacological intervention of RhoA-mediated signaling pathways.


Assuntos
Proteína Quinase C/metabolismo , Transdução de Sinais/fisiologia , Proteína rhoA de Ligação ao GTP/metabolismo , Animais , Linhagem Celular , Ativação Enzimática , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Humanos , Lisofosfolipídeos/metabolismo , Camundongos , Contração Muscular/fisiologia , Mutação , Proteína Quinase C/química , Proteína Quinase C/genética , Estrutura Terciária de Proteína , Ratos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteína rhoA de Ligação ao GTP/genética
13.
Cell Signal ; 18(6): 807-18, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16137858

RESUMO

In this article, we explore the role of the C-terminus (V5 domain) of PKCepsilon plays in the catalytic competence of the kinase using serial truncations followed by immune-complex kinase assays. Surprisingly, removal of the last seven amino acid residues at the C-terminus of PKCepsilon resulted in a PKCepsilon-Delta731 mutant with greatly reduced intrinsic catalytic activity while truncation of eight amino acid residues at the C-terminus resulted in a catalytically inactive PKCepsilon mutant. Computer modeling and molecular dynamics simulations showed that the last seven and/or eight amino acid residues of PKCepsilon were involved in interactions with residues in the catalytic core. Further truncation analyses revealed that the hydrophobic phosphorylation motif was dispensable for the physical interaction between PKCepsilon and 3-phosphoinositide-dependent kinase-1 (PDK-1) as the PKCepsilon mutant lacking both the turn and the hydrophobic motifs could still be co-immunoprecipitated with PDK-1. These results provide fresh insights into the biochemical and structural basis underlying the isozyme-specific regulation of PKC and suggest that the very C-termini of PKCs constitute a promising new target for the development of novel isozyme-specific inhibitors of PKC.


Assuntos
Proteína Quinase C-épsilon/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Quinases Dependentes de 3-Fosfoinositídeo , Motivos de Aminoácidos/fisiologia , Sequência de Aminoácidos , Animais , Catálise , Domínio Catalítico , Simulação por Computador , Deleção de Genes , Interações Hidrofóbicas e Hidrofílicas , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Fosforilação , Ligação Proteica , Proteína Quinase C-épsilon/genética
14.
Cell Signal ; 17(9): 1084-97, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15993750

RESUMO

PRK1/PKN is a member of the protein kinase C (PKC) superfamily of serine/threonine protein kinases. Despite its important role as a RhoA effector, limited information is available regarding how this kinase is regulated. We show here that the last seven amino acid residues at the C-terminus is dispensable for the catalytic activity of PRK1 but is critical for the in vivo stability of this kinase. Surprisingly, the intact hydrophobic motif in PRK1 is dispensable for 3-phosphoinositide-dependent kinase-1 (PDK-1) binding and phosphorylation of the activation loop, as the PRK1-Delta940 mutant lacking the last two residues of the hydrophobic motif and the last 5 residues at the C-terminus interacts with PDK-1 in vivo and has a similar specific activity as the wild-type protein. We also found that the last four amino acid residues at the C-terminus of PRK1 is critical for the full lipid responsiveness as the PRK1-Delta942 deletion mutant is no longer activated by arachidonic acid. Our data suggest that the very C-terminus in PRK1 is critically involved in the control of the catalytic activity and activation by lipids. Since this very C-terminal segment is the least conserved among members of the PKC superfamily, it would be a promising target for isozyme-specific pharmaceutical interventions.


Assuntos
Proteína Quinase C/química , Proteína Quinase C/metabolismo , Proteínas Quinases Dependentes de 3-Fosfoinositídeo , Sequência de Aminoácidos , Animais , Ácido Araquidônico/metabolismo , Ácido Aspártico/química , Células COS , Catálise , Chlorocebus aethiops , Ativação Enzimática , Estabilidade Enzimática , Interações Hidrofóbicas e Hidrofílicas , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Fenilalanina/química , Proteína Quinase C/genética , Proteínas Serina-Treonina Quinases/metabolismo , Estrutura Terciária de Proteína , Ratos , Alinhamento de Sequência
15.
Cell Signal ; 17(9): 1125-36, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15993753

RESUMO

Protein kinase C (PKC) is a family of serine/threonine protein kinases that are pivotal in cellular regulation. Since its discovery in 1977, PKCs have been known as cytosolic and peripheral membrane proteins. However, there are reports that PKC can insert into phospholipids vesicles in vitro. Given the intimate relationship between the plasma membrane and the activation of PKC, it is important to determine whether such "membrane-inserted" form of PKC exists in mammalian cells or tissues. Here, we report the identification of an integral plasma membrane pool for all the 10 PKC isozymes in vivo by their ability to partition into the detergent-rich phase in Triton X-114 phase partitioning, and by their resistance to extractions with 0.2 M sodium carbonate (pH 11.5), 2 M urea and 2 M sodium chloride. The endogenous integral membrane pool of PKC in mouse fibroblasts is found to be acutely regulated by phorbol ester or diacylglycerol, suggesting that this pool of PKC may participate in cellular processes known to be regulated by PKC. At least for PKC(alpha), the C2-V3 region at the regulatory domain of the kinase is responsible for membrane integration. Further exploration of the function of this novel integral plasma membrane pool of PKC will not only shed new light on molecular mechanisms underlying its cellular functions but also provide new strategies for pharmaceutical modulation of this important group of kinases.


Assuntos
Membrana Celular/enzimologia , Proteína Quinase C/análise , Animais , Células COS , Chlorocebus aethiops , Diglicerídeos/farmacologia , Isoenzimas/análise , Isoenzimas/metabolismo , Metabolismo dos Lipídeos , Camundongos , Proteína Quinase C/química , Proteína Quinase C/metabolismo , Proteína Quinase C-alfa , Estrutura Terciária de Proteína , Acetato de Tetradecanoilforbol/farmacologia
16.
Cancer Res ; 65(11): 4520-4, 2005 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-15930268

RESUMO

Protein kinase C (PKC) is a key regulator of cell proliferation, differentiation, and apoptosis and is one of the drug targets of anticancer therapy. Recently, a single point mutation (D294G) in PKCalpha has been found in pituitary and thyroid tumors with more invasive phenotype. Although the PKCalpha-D294G mutant is implicated in the progression of endocrine tumors, no apparent biochemical/cell biological abnormalities underlying tumorigenesis with this mutant have been found. We report here that the PKCalpha-D294G mutant is unable to bind to cellular membranes tightly despite the fact that it translocates to the membrane as efficiently as the wild-type PKCalpha upon treatment of phorbol ester. The impaired membrane binding is associated with this mutant's inability to transduce several antitumorigenic signals as it fails to mediate phorbol ester-stimulated translocation of myristoylated alanine-rich protein kinase C substrate (MARCKS), to activate mitogen-activated protein kinase and to augment melatonin-stimulated neurite outgrowth. Thus, the PKCalpha-D294G is a loss-of-function mutation. We propose that the wild-type PKCalpha may play important antitumorigenic roles in the progression of endocrine tumors. Therefore, developing selective activators instead of inhibitors of PKCalpha might provide effective pharmacological interventions for the treatment of certain endocrine tumors.


Assuntos
Neoplasias Hipofisárias/enzimologia , Mutação Puntual , Proteína Quinase C/genética , Neoplasias da Glândula Tireoide/enzimologia , Animais , Células CHO , Bovinos , Membrana Celular/enzimologia , Cricetinae , Citoplasma/enzimologia , Ativação Enzimática/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Melatonina/farmacologia , Proteínas de Membrana/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Substrato Quinase C Rico em Alanina Miristoilada , Neuritos/efeitos dos fármacos , Neuritos/enzimologia , Fosforilação/efeitos dos fármacos , Neoplasias Hipofisárias/genética , Proteína Quinase C/metabolismo , Proteína Quinase C-alfa , Transdução de Sinais/fisiologia , Acetato de Tetradecanoilforbol/farmacologia , Neoplasias da Glândula Tireoide/genética , Transfecção
17.
FASEB J ; 18(14): 1722-4, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15375078

RESUMO

Mammalian serine/threonine protein kinases, except for TGF-beta receptor kinase family, are intracellular proteins. PRK1/PKN is a member of the protein kinase C superfamily of serine/threonine kinases and is one of the first identified effectors for RhoA GTPase. However, the role of PRK1 in mediating signaling downstream of activated RhoA is largely unknown. Here, we present evidence that identifies a novel plasma membrane pool of PRK1. This integral membrane form of PRK1 is catalytically active. The phosphorylation of serine377 of PRK1 is required for its integration into membranes. This integration is essential for PRK1 to function as a Rho effector as only the integral plasma membrane PRK1 is able to initiate RhoA-mediated and ligand-dependent transcriptional activation of the androgen receptor in human epithelial cells and to mediate RhoA-induced neurite retraction in mouse neuronal cells. These results indicate that RhoA signals via the integral membrane pool of its effectors in its immediate vicinity at the plasma membrane, thus establishing a new paradigm in mammalian cell signaling.


Assuntos
Membrana Celular/enzimologia , Proteína Quinase C/metabolismo , Transdução de Sinais , Proteína rhoA de Ligação ao GTP/metabolismo , Substituição de Aminoácidos , Animais , Linhagem Celular , Cricetinae , Humanos , Lisofosfolipídeos/farmacologia , Camundongos , Neurônios/enzimologia , Proteína Quinase C/genética , Ratos , Receptores Androgênicos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...