Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Biomolecules ; 10(9)2020 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-32906765

RESUMO

Protein kinase Cs (PKCs) are activated by lipids in the plasma membrane and bind to a scaffold assembled on the epidermal growth factor (EGF) receptor (EGFR). Understanding how this complex is routed is important, because this determines whether EGFR is degraded, terminating signaling. Here, cells were preincubated in EGF-tagged gold nanoparticles, then allowed to internalize them in the presence or absence of a phorbol ester PKC activator. PKC colocalized with EGF-tagged nanoparticles within 5 min and migrated with EGFR-bearing vesicles into the cell. Two conformations of PKC-epsilon were distinguished by different primary antibodies. One, thought to be enzymatically active, was on endosomes and displayed a binding site for antibody RR (R&D). The other, recognized by Genetex green (GG), was soluble, on actin-rich structures, and loosely bound to vesicles. During a 15-min chase, EGF-tagged nanoparticles entered large, perinuclear structures. In phorbol ester-treated cells, vesicles bearing EGF-tagged nanoparticles tended to enter this endocytic recycling compartment (ERC) without the GG form. The correlation coefficient between the GG (inactive) and RR conformations on vesicles was also lower. Thus, active PKC has a Charon-like function, ferrying vesicles to the ERC, and inactivation counteracts this function. The advantage conferred on cells by aggregating vesicles in the ERC is unclear.


Assuntos
Receptores ErbB/metabolismo , Proteína Quinase C-épsilon/química , Proteína Quinase C-épsilon/metabolismo , Actinas/metabolismo , Animais , Linhagem Celular , Membrana Celular/metabolismo , Endossomos/metabolismo , Epitopos/metabolismo , Ouro , Mesotelina , Nanopartículas Metálicas , Transporte Proteico/fisiologia , Ratos
2.
BMC Bioinformatics ; 19(Suppl 13): 342, 2019 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-30717648

RESUMO

BACKGROUND: (-)-Balanol is an ATP-mimicking inhibitor that non-selectively targets protein kinase C (PKC) isozymes and cAMP-dependent protein kinase (PKA). While PKA constantly shows tumor promoting activities, PKC isozymes can ambiguously be tumor promoters or suppressors. In particular, PKCε is frequently implicated in tumorigenesis and a potential target for anticancer drugs. We recently reported that the C5(S)-fluorinated balanol analogue (balanoid 1c) had improved binding affinity and selectivity for PKCε but not to the other novel PKC isozymes, which share a highly similar ATP site. The underlying basis for this fluorine-based selectivity is not entirely comprehended and needs to be investigated further for the development of ATP mimic inhibitors specific for PKCε. RESULTS: Using molecular dynamics (MD) simulations assisted by homology modelling and sequence analysis, we have studied the fluorine-based selectivity in the highly similar ATP sites of novel PKC (nPKC) isozymes. The study suggests that every nPKC isozyme has different dynamics behaviour in both apo and 1c-bound forms. Interestingly, the apo form of PKCε, where 1c binds strongly, shows the highest degree of flexibility which dramatically decreases after binding 1c. CONCLUSIONS: For the first time to the best of our knowledge, we found that the origin of 1c selectivity for PKCε comes from the unique dynamics feature of each PKC isozyme. Fluorine conformational control in 1c can synergize with and lock down the dynamics of PKCε, which optimize binding interactions with the ATP site residues of the enzyme, particularly the invariant Lys437. This finding has implications for further rational design of balanol-based PKCε inhibitors for cancer drug development.


Assuntos
Azepinas/metabolismo , Halogenação , Hidroxibenzoatos/metabolismo , Proteína Quinase C-épsilon/metabolismo , Trifosfato de Adenosina/metabolismo , Azepinas/química , Análise por Conglomerados , Humanos , Hidroxibenzoatos/química , Isoenzimas/metabolismo , Simulação de Dinâmica Molecular , Fosforilação , Conformação Proteica , Proteína Quinase C-épsilon/química , Ribose/química , Alinhamento de Sequência , Eletricidade Estática , Especificidade por Substrato
3.
J Chem Inf Model ; 58(2): 511-519, 2018 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-29341608

RESUMO

(-)-Balanol is an adenosine triphosphate mimic that inhibits protein kinase C (PKC) isozymes and cAMP-dependent protein kinase (PKA) with limited selectivity. While PKA is known as a tumor promoter, PKC isozymes can be tumor promoters or suppressors. In particular, PKCε is frequently involved in tumorigenesis and a potential target for anticancer drugs. We recently reported that stereospecific fluorination of balanol yielded a balanoid with enhanced selectivity for PKCε over other PKC isozymes and PKA, although the global fluorine effect behind the selectivity enhancement is not fully understood. Interestingly, in contrast to PKA, PKCε is more sensitive to this fluorine effect. Here we investigate the global fluorine effect on the different binding responses of PKCε and PKA to balanoids using molecular dynamics (MD) simulations. For the first time to the best of our knowledge, we found that a structurally equivalent residue in each kinase, Thr184 in PKA and Ala549 in PKCε, is essential for the different binding responses. Furthermore, the study revealed that the invariant Lys, Lys73 in PKA and Lys437 in PKCε, already known to have a crucial role in the catalytic activity of kinases, serves as the main anchor for balanol binding. Overall, while Thr184 in PKA attenuates the effect of fluorination, Ala549 permits remote response of PKCε to fluorine substitution, with implications for rational design of future balanol-based PKCε inhibitors.


Assuntos
Azepinas/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/química , Flúor/química , Hidroxibenzoatos/metabolismo , Simulação de Dinâmica Molecular , Proteína Quinase C-épsilon/química , Sítios de Ligação , Humanos
4.
FEBS Lett ; 592(2): 179-189, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29266266

RESUMO

Targeting the interaction between PKC isoforms and their anchoring proteins can specifically regulate kinase activity. εV1-2 and pseudoεRACK peptides, derived from the PKCε C2 domain, modulate its association with receptor for activated C-kinase 2 (RACK2) and thus its function. Details of these interactions remain obscure, and we therefore investigated binding of these peptides using biophysical techniques. Surface plasmon resonance (SPR) indicated that the inhibitory εV1-2 peptide bound to RACK2, and inhibited PKCε binding as expected. In contrast, SPR and NMR demonstrated that the activating pseudoεRACK peptide and related sequences did not bind to PKCε, indicating that their mechanisms of action do not involve binding to the kinase as previously proposed. Our results clarify which interactions could be targeted in developing new therapeutics that inhibit PKCε-RACK2 interaction.


Assuntos
Peptídeos/farmacologia , Proteína Quinase C-épsilon/química , Proteína Quinase C-épsilon/metabolismo , Receptores de Quinase C Ativada/metabolismo , Animais , Sítios de Ligação , Espectroscopia de Ressonância Magnética , Modelos Moleculares , Peptídeos/química , Ligação Proteica/efeitos dos fármacos , Domínios Proteicos , Estrutura Terciária de Proteína , Ressonância de Plasmônio de Superfície
5.
Bioorg Med Chem ; 25(12): 2971-2980, 2017 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-28392275

RESUMO

C1 domain-containing proteins, such as protein kinase C (PKC), have a central role in cellular signal transduction. Their involvement in many diseases, including cancer, cardiovascular disease, and immunological and neurological disorders has been extensively demonstrated and has prompted a search for small molecules to modulate their activity. By employing a diacylglycerol (DAG)-lactone template, we have been able to develop ultra potent analogs of diacylglycerol with nanomolar binding affinities approaching those of complex natural products such as phorbol esters and bryostatins. One current challenge is the development of selective ligands capable of discriminating between different protein family members. Recently, structure-activity relationship studies have shown that the introduction of an indole ring as a DAG-lactone substituent yielded selective Ras guanine nucleotide-releasing protein (RasGRP1) activators when compared to PKCα and PKCε. In the present work, we examine the effects of ligand selectivity relative to the orientation of the indole ring and the nature of the DAG-lactone template itself. Our results show that the indole ring must be attached to the lactone moiety through the sn-2 position in order to achieve RasGRP1 selectivity.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Indóis/química , Indóis/farmacologia , Lactonas/química , Lactonas/farmacologia , Proteína Quinase C-alfa/metabolismo , Proteína Quinase C-épsilon/metabolismo , Proteínas de Ligação a DNA/química , Fatores de Troca do Nucleotídeo Guanina/química , Humanos , Simulação de Acoplamento Molecular , Ligação Proteica , Domínios Proteicos , Proteína Quinase C-alfa/química , Proteína Quinase C-épsilon/química , Relação Estrutura-Atividade
6.
J Biomed Sci ; 24(1): 3, 2017 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-28056995

RESUMO

BACKGROUND: Mitochondrial aldehyde dehydrogenase 2 (ALDH2) is a key enzyme for the metabolism of many toxic aldehydes such as acetaldehyde, derived from alcohol drinking, and 4HNE, an oxidative stress-derived lipid peroxidation aldehyde. Post-translational enhancement of ALDH2 activity can be achieved by serine/threonine phosphorylation by epsilon protein kinase C (εPKC). Elevated ALDH2 is beneficial in reducing injury following myocardial infarction, stroke and other oxidative stress and aldehyde toxicity-related diseases. We have previously identified three εPKC phosphorylation sites, threonine 185 (T185), serine 279 (S279) and threonine 412 (T412), on ALDH2. Here we further characterized the role and contribution of each phosphorylation site to the enhancement of enzymatic activity by εPKC. METHODS: Each individual phosphorylation site was mutated to a negatively charged amino acid, glutamate, to mimic a phosphorylation, or to a non-phosphorylatable amino acid, alanine. ALDH2 enzyme activities and protection against 4HNE inactivation were measured in the presence or absence of εPKC phosphorylation in vitro. Coevolution of ALDH2 and its εPKC phosphorylation sites was delineated by multiple sequence alignments among a diverse range of species and within the ALDH multigene family. RESULTS: We identified S279 as a critical εPKC phosphorylation site in the activation of ALDH2. The critical catalytic site, cysteine 302 (C302) of ALDH2 is susceptible to adduct formation by reactive aldehyde, 4HNE, which readily renders the enzyme inactive. We show that phosphomimetic mutations of T185E, S279E and T412E confer protection of ALDH2 against 4HNE-induced inactivation, indicating that phosphorylation on these three sites by εPKC likely also protects the enzyme against reactive aldehydes. Finally, we demonstrate that the three ALDH2 phosphorylation sites co-evolved with εPKC over a wide range of species. Alignment of 18 human ALDH isozymes, indicates that T185 and S279 are unique ALDH2, εPKC specific phosphorylation sites, while T412 is found in other ALDH isozymes. We further identified three highly conserved serine/threonine residues (T384, T433 and S471) in all 18 ALDH isozymes that may play an important phosphorylation-mediated regulatory role in this important family of detoxifying enzymes. CONCLUSION: εPKC phosphorylation and its coevolution with ALDH2 play an important role in the regulation and protection of ALDH2 enzyme activity.


Assuntos
Aldeído-Desidrogenase Mitocondrial/química , Evolução Molecular , Proteína Quinase C-épsilon/química , Aldeído-Desidrogenase Mitocondrial/genética , Aldeído-Desidrogenase Mitocondrial/metabolismo , Humanos , Fosforilação/fisiologia , Proteína Quinase C-épsilon/genética , Proteína Quinase C-épsilon/metabolismo
7.
Biochemistry ; 55(45): 6327-6336, 2016 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-27776404

RESUMO

Curcumin is a polyphenolic nutraceutical that acts on multiple biological targets, including protein kinase C (PKC). PKC is a family of serine/threonine kinases central to intracellular signal transduction. We have recently shown that curcumin selectively inhibits PKCα, but not PKCε, in CHO-K1 cells [Pany, S. (2016) Biochemistry 55, 2135-2143]. To understand which domain(s) of PKCα is responsible for curcumin binding and inhibitory activity, we made several domain-swapped mutants in which the C1 (combination of C1A and C1B) and C2 domains are swapped between PKCα and PKCε. Phorbol ester-induced membrane translocation studies using confocal microscopy and immunoblotting revealed that curcumin inhibited phorbol ester-induced membrane translocation of PKCε mutants, in which the εC1 domain was replaced with αC1, but not the PKCα mutant in which αC1 was replaced with the εC1 domain, suggesting that αC1 is a determinant for curcumin's inhibitory effect. In addition, curcumin inhibited membrane translocation of PKCε mutants, in which the εC1A and εC1B domains were replaced with the αC1A and αC1B domains, respectively, indicating the role of both αC1A and αC1B domains in curcumin's inhibitory effects. Phorbol 13-acetate inhibited the binding of curcumin to αC1A and αC1B with IC50 values of 6.27 and 4.47 µM, respectively. Molecular docking and molecular dynamics studies also supported the higher affinity of curcumin for αC1B than for αC1A. The C2 domain-swapped mutants were inactive in phorbol ester-induced membrane translocation. These results indicate that curcumin binds to the C1 domain of PKCα and highlight the importance of this domain in achieving PKC isoform selectivity.


Assuntos
Curcumina/química , Domínios Proteicos , Proteína Quinase C-alfa/química , Proteína Quinase C-épsilon/química , Sítios de Ligação/genética , Ligação Competitiva , Biocatálise/efeitos dos fármacos , Curcumina/metabolismo , Curcumina/farmacologia , Inibidores Enzimáticos/química , Inibidores Enzimáticos/metabolismo , Inibidores Enzimáticos/farmacologia , Células HEK293 , Humanos , Immunoblotting , Cinética , Microscopia Confocal , Simulação de Dinâmica Molecular , Mutação , Ésteres de Forbol/farmacologia , Ligação Proteica , Proteína Quinase C-alfa/genética , Proteína Quinase C-alfa/metabolismo , Proteína Quinase C-épsilon/genética , Proteína Quinase C-épsilon/metabolismo , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/genética , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
8.
Cell Transplant ; 23(7): 913-9, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-23562311

RESUMO

Transplantation of islets isolated from deceased donor pancreata is an attractive method of ß-cell replacement therapy for patients with type 1 diabetes (T1D). However, the loss of islet cell viability and function during the peritransplant period is a limiting factor to long-term islet engraftment. Activation of the isoenzyme PKCɛ may improve islet survival and function. The current study assesses the effects of PKCɛ activation on islet graft function in a syngeneic streptozotocin-induced diabetic mouse model. Islets were isolated from wild-type BALB/c mice preconditioned with either a PKCɛ activator (ψɛRACK) or a TAT carrier control peptide. Islets were further treated with the same agents during isolation, purification, and incubation prior to transplantation. Two hundred seventy-five islet equivalents were transplanted under the kidney capsule of streptozotocin-induced diabetic BALB/c mice. Islet function was assessed by measurement of blood glucose levels every 3 days for 42 days after transplant and through an intraperitoneal glucose tolerance test (IPGTT). The time for return to euglycemia in mice transplanted with islets treated with ψɛRACK was improved at 14 ± 6 days versus 21 ± 6 days with TAT-treated islets. The IPGTT showed a 50% reduction in the area under the curve associated with an improved insulin response in mice transplanted with ψɛRACK-treated islets compared to TAT-treated islets. A preconditioning regimen using PKCɛ agonist before pancreatic recovery and during islet isolation improves islet graft function and resistance to high glucose stress after transplantation.


Assuntos
Diabetes Mellitus Experimental/terapia , Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas/efeitos dos fármacos , Peptídeos/farmacologia , Proteína Quinase C-épsilon/química , Animais , Glicemia/análise , Modelos Animais de Doenças , Feminino , Teste de Tolerância a Glucose , Sobrevivência de Enxerto/efeitos dos fármacos , Insulina/metabolismo , Secreção de Insulina , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Proteína Quinase C-épsilon/metabolismo , Transplante Homólogo
9.
Biochim Biophys Acta ; 1828(2): 552-60, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23088913

RESUMO

The C2 domain of PKCε binds to negatively charged phospholipids but little is known so far about the docking orientation of this domain when it is bound. By using a FRET assay we have studied the binding of this domain to model membranes. We have also used ATR-Fourier transform infrared spectroscopy with polarized light (ATR-FTIR) to determine the docking mode by calculating the ß-sandwich orientation when the domain is bound to different types of model membranes. The vesicle lipid compositions were: POPC/POPE/POPA (22:36:42) imitating the inner leaflet of a plasma membrane, POPC/POPA (50:50) in which POPE has been eliminated with respect to the former composition and POPC/POPE/CL (43:36:21) imitating the inner mitochondrial membrane. Results show that the ß-sandwich of the PKCα-C2 domain is inclined at an angle α close to 45° to the membrane normal. Some differences were found with respect to the extent of binding as a function of phospholipid composition and small changes on secondary structure were only evident when the domain was bound to model membranes of POPC/POPA: in this case, the percentage of ß-sheet of the C2 domain increases if compared with the secondary structure of the domain in the absence of vesicles. With respect to the ß-sandwich orientation, when the domain is bound to POPC/POPE/CL membranes it forms an angle with the normal to the surface of the lipid bilayer (39°) smaller than that one observed when the domain interacts with vesicles of POPC/POPA (49°).


Assuntos
Transferência Ressonante de Energia de Fluorescência/métodos , Bicamadas Lipídicas/química , Proteína Quinase C-épsilon/química , Espectrofotometria Infravermelho/métodos , Adenosina/análogos & derivados , Adenosina/química , Cálcio/química , Glicerofosfolipídeos/química , Humanos , Lipídeos/química , Membranas Mitocondriais/metabolismo , Modelos Moleculares , Modelos Estatísticos , Conformação Molecular , Fosfatidilcolinas/química , Fosfatidiletanolaminas/química , Fosfolipídeos/química , Ligação Proteica , Conformação Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína
10.
Cell Physiol Biochem ; 30(3): 771-7, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22854271

RESUMO

BACKGROUND/AIMS: In our earlier study, indomethacin potentiated α7 acetylcholine (ACh) receptor responses by activating protein kinase C (PKC). The present study was conducted to gain further insight into the indomethacin action on PKC. METHODS: PKC activity was assayed in PC-12 cells or under the cell-free conditions. PKC-ε was knocked-down using the siRNA to silence the PKC-ε-targeted gene. A fluorescein-conjugated indomethacin was synthesized to examine the interaction of indomethacin with PKC-ε. RESULTS: In the in situ PKC assay, indomethacin activated PKC in PC-12 cells in a concentration (1-100 µM)-dependent manner, and the activation was suppressed by knocking-down PKC-ε. In the cell-free PKC assay, indomethacin (100 µM) activated PKC-ε in the absence of diacylglycerol, phosphatidylserine, and calcium, but other PKC isozymes such as α, ßΙ, ßΙΙ, γ, δ, ι, and ζ were not activated. In the indomethacin binding assay using a fluorescent-conjugated indomethacin on blue native-polyacrylamide gel electrophoresis (blue native-PAGE), a fluorescent signal was detected at the site consistent with PKC-ε protein and the signal was attenuated by adding non-conjugated indomethacin or eliminated by pretreatment with non-conjugated indomethacin. CONCLUSION: The results of the present study show that indomethacin has the potential to selectively activate PKC-ε through its direct binding, independently of cyclooxygenase (COX) inhibition.


Assuntos
Inibidores de Ciclo-Oxigenase/farmacologia , Ativação Enzimática/efeitos dos fármacos , Indometacina/farmacologia , Proteína Quinase C-épsilon/metabolismo , Animais , Fluoresceína/química , Células PC12 , Prostaglandina-Endoperóxido Sintases/química , Prostaglandina-Endoperóxido Sintases/metabolismo , Ligação Proteica , Isoformas de Proteínas/metabolismo , Proteína Quinase C-épsilon/química , Proteína Quinase C-épsilon/genética , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Ratos , Especificidade por Substrato
11.
PLoS One ; 7(4): e35630, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22536418

RESUMO

Dysregulation of PKCε is involved in several serious diseases such as cancer, type II diabetes and Alzheimer's disease. Therefore, specific activators and inhibitors of PKCε hold promise as future therapeutics, in addition to being useful in research into PKCε regulated pathways. We have previously described llama single chain antibodies (VHHs) that specifically activate (A10, C1 and D1) or inhibit (E6 and G8) human recombinant PKCε. Here we report a thorough kinetic analysis of these VHHs. The inhibiting VHHs act as non-competitive inhibitors of PKCε activity, whereas the activating VHHs have several different modes of action, either increasing V(max) and/or decreasing K(m) values. We also show that the binding of the VHHs to PKCε is conformation-dependent, rendering the determination of affinities difficult. Apparent affinities are in the micromolar range based on surface plasmon resonance studies. Furthermore, the VHHs have no effect on the activity of rat PKCε nor can they bind the rat form of the protein in immunoprecipitation studies despite the 98% identity between the human and rat PKCε proteins. Finally, we show for the first time that the VHHs can influence PKCε function also in cells, since an activating VHH increases the rate of PKCε translocation in response to PMA in HeLa cells, whereas an inhibiting VHH slows down the translocation. These results give insight into the mechanisms of PKCε activity modulation and highlight the importance of protein conformation on VHH binding.


Assuntos
Ativadores de Enzimas/farmacologia , Proteína Quinase C-épsilon/metabolismo , Anticorpos de Cadeia Única/farmacologia , Animais , Encéfalo/enzimologia , Ativação Enzimática , Proteínas de Fluorescência Verde/metabolismo , Células HeLa , Humanos , Cinética , Ligação Proteica , Proteína Quinase C-épsilon/antagonistas & inibidores , Proteína Quinase C-épsilon/química , Transporte Proteico , Ratos , Proteínas Recombinantes de Fusão/metabolismo , Anticorpos de Cadeia Única/biossíntese , Anticorpos de Cadeia Única/química , Especificidade da Espécie , Ressonância de Plasmônio de Superfície , Acetato de Tetradecanoilforbol/farmacologia
12.
PLoS One ; 7(12): e52888, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23285216

RESUMO

Resveratrol (1) is a naturally occurring phytoalexin that affects a variety of human disease models, including cardio- and neuroprotection, immune regulation, and cancer chemoprevention. One of the possible mechanisms by which resveratrol affects these disease states is by affecting the cellular signaling network involving protein kinase C (PKC). PKC is the family of serine/threonine kinases, whose activity is inhibited by resveratrol. To develop PKC isotype selective molecules on the resveratrol scaffold, several analogs (2-5) of resveratrol with a long aliphatic chain varying with number of unsaturated doubled bonds have been synthesized, their cytotoxic effects on CHO-K1 cells are measured and their effects on the membrane translocation properties of PKCα and PKCε have been determined. The analogs showed less cytotoxic effects on CHO-K1 cells. Analog 4 with three unsaturated double bonds in its aliphatic chain activated PKCα, but not PKCε. Analog 4 also activated ERK1/2, the downstream proteins in the PKC signaling pathway. Resveratrol analogs 2-5, however, did not show any inhibition of the phorbol ester-induced membrane translocation for either PKCα or PKCε. Molecular docking of 4 into the activator binding site of PKCα revealed that the resveratrol moiety formed hydrogen bonds with the activator binding residues and the aliphatic chain capped the activator binding loops making its surface hydrophobic to facilitate its interaction with the plasma membrane. The present study shows that subtle changes in the resveratrol structure can have profound impact on the translocation properties of PKCs. Therefore, resveratrol scaffold can be used to develop PKC selective modulators for regulating associated disease states.


Assuntos
Ácidos Graxos/química , Proteína Quinase C-alfa/metabolismo , Proteína Quinase C-épsilon/metabolismo , Estilbenos/química , Estilbenos/farmacologia , Animais , Células CHO , Sobrevivência Celular/efeitos dos fármacos , Cricetinae , Cricetulus , Ativação Enzimática/efeitos dos fármacos , Humanos , Isoenzimas/química , Isoenzimas/metabolismo , Modelos Biológicos , Modelos Moleculares , Simulação de Acoplamento Molecular , Proteína Quinase C-alfa/química , Proteína Quinase C-épsilon/química , Resveratrol , Transdução de Sinais/efeitos dos fármacos , Relação Estrutura-Atividade
13.
Mol Biol Cell ; 21(8): 1398-408, 2010 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-20164256

RESUMO

The C1 domains in protein kinase C (PKC) isozymes and other signaling molecules are responsible for binding the lipid second messenger diacylglycerol and phorbol esters, and for mediating translocation to membranes. Previous studies revealed that the C1 domain in alpha- and beta-chimaerins, diacylglycerol-regulated Rac-GAPs, interacts with the endoplasmic reticulum/Golgi protein p23/Tmp21. Here, we found that p23/Tmp21 acts as a C1 domain-docking protein that mediates perinuclear translocation of beta2-chimaerin. Glu227 and Leu248 in the beta2-chimaerin C1 domain are crucial for binding p23/Tmp21 and perinuclear targeting. Interestingly, isolated C1 domains from individual PKC isozymes differentially interact with p23/Tmp21. For PKCepsilon, it interacts with p23/Tmp21 specifically via its C1b domain; however, this association is lost in response to phorbol esters. These results demonstrate that p23/Tmp21 acts as an anchor that distinctively modulates compartmentalization of C1 domain-containing proteins, and it plays an essential role in beta2-chimaerin relocalization. Our study also highlights the relevance of C1 domains in protein-protein interactions in addition to their well-established lipid-binding properties.


Assuntos
Proteínas Ativadoras de GTPase/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Neoplasias/química , Proteínas de Neoplasias/metabolismo , Proteína Quinase C/química , Proteína Quinase C/metabolismo , Proteínas rac de Ligação ao GTP/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular , Núcleo Celular/metabolismo , Ácido Glutâmico/metabolismo , Humanos , Leucina/metabolismo , Dados de Sequência Molecular , Proteínas Mutantes/metabolismo , Proteínas de Transporte Nucleocitoplasmático , Ligação Proteica , Proteína Quinase C-épsilon/química , Proteína Quinase C-épsilon/metabolismo , Estrutura Terciária de Proteína , Transporte Proteico , Interferência de RNA , Relação Estrutura-Atividade
14.
Biophys J ; 96(9): 3638-47, 2009 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-19413969

RESUMO

The C1 domains of classical and novel PKCs mediate their diacylglycerol-dependent translocation. Using fluorescence resonance energy transfer, we studied the contribution of different negatively charged phospholipids and diacylglycerols to membrane binding. Three different C1B domains of PKCs were studied (the classical gamma, and the novel delta and epsilon), together with different lipid mixtures containing three types of acidic phospholipids and three types of activating diacylglycerols. The results show that C1Bgamma and C1Bepsilon exhibit a higher affinity to bind to vesicles containing 1-palmitoyl-2-oleoyl-sn-phosphatidic acid, 1-palmitoyl-2-oleoyl-sn-phoshatidylserine, or 1-palmitoyl-2-oleoyl-sn-phosphatidylglycerol, with C1Bepsilon being the most relevant case because its affinity for POPA-containing vesicles increased by almost two orders of magnitude. When the effect of the diacylglycerol fatty acid composition on membrane binding was studied, the C1Bepsilon domain showed the highest binding affinity to membranes containing 1-stearoyl-oleoyl-sn-glycerol or 1,2-sn-dioleoylglycerol with POPA as the acidic phospholipid. Of the three diacylglycerols used in this study, 1,2-sn-dioleoylglycerol and 1-stearoyl-oleoyl-sn-glycerol showed the highest affinities for each isoenzyme, whereas 1,2-sn-dipalmitoylglycerol; showed the lowest affinity. DSC experiments showed this to be a consequence of the nonfluid conditions of 1,2-sn-dipalmitoylglycerol;-containing systems.


Assuntos
Proteína Quinase C-delta/metabolismo , Proteína Quinase C-épsilon/metabolismo , Proteína Quinase C/metabolismo , Estrutura Terciária de Proteína , Lipossomas Unilamelares/metabolismo , Adenosina/análogos & derivados , Adenosina/metabolismo , Varredura Diferencial de Calorimetria , Linhagem Celular , Diglicerídeos/metabolismo , Transferência Ressonante de Energia de Fluorescência , Glicerofosfolipídeos/metabolismo , Humanos , Modelos Moleculares , Ácidos Fosfatídicos/metabolismo , Fosfatidilcolinas/metabolismo , Fosfatidilgliceróis/metabolismo , Fosfatidilserinas/metabolismo , Ligação Proteica , Proteína Quinase C/química , Proteína Quinase C-delta/química , Proteína Quinase C-épsilon/química , Temperatura
15.
Biochem J ; 421(3): 405-13, 2009 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-19432558

RESUMO

Alcohols regulate the expression and function of PKC (protein kinase C), and it has been proposed that an alcohol-binding site is present in PKC alpha in its C1 domain, which consists of two cysteine-rich subdomains, C1A and C1B. A PKC epsilon-knockout mouse showed a significant decrease in alcohol consumption compared with the wild-type. The aim of the present study was to investigate whether an alcohol-binding site could be present in PKC epsilon. Here we show that ethanol inhibited PKC epsilon activity in a concentration-dependent manner with an EC50 (equilibrium ligand concentration at half-maximum effect) of 43 mM. Ethanol, butanol and octanol increased the binding affinity of a fluorescent phorbol ester SAPD (sapintoxin-D) to PKC epsilon C1B in a concentration-dependent manner with EC50 values of 78 mM, 8 mM and 340 microM respectively, suggesting the presence of an allosteric alcohol-binding site in this subdomain. To identify this site, PKC epsilon C1B was photolabelled with 3-azibutanol and 3-azioctanol and analysed by MS. Whereas azibutanol preferentially labelled His236, Tyr238 was the preferred site for azioctanol. Inspection of the model structure of PKC epsilon C1B reveals that these residues are 3.46 A (1 A=0.1 nm) apart from each other and form a groove where His236 is surface-exposed and Tyr238 is buried inside. When these residues were replaced by alanine, it significantly decreased alcohol binding in terms of both photolabelling and alcohol-induced SAPD binding in the mutant H236A/Y238A. Whereas Tyr238 was labelled in mutant H236A, His236 was labelled in mutant Y238A. The present results provide direct evidence for the presence of an allosteric alcohol-binding site on protein kinase C epsilon and underscore the role of His236 and Tyr238 residues in alcohol binding.


Assuntos
Alcoolismo/metabolismo , Álcoois/metabolismo , Proteína Quinase C-épsilon/química , Proteína Quinase C-épsilon/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Cristalização , Humanos , Cinética , Conformação Molecular , Dados de Sequência Molecular , Mutação , Ligação Proteica , Conformação Proteica , Proteína Quinase C-épsilon/genética , Estrutura Terciária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
16.
J Biol Chem ; 282(44): 32288-97, 2007 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-17785460

RESUMO

Endothelial cell apoptosis is associated with vascular injury and predisposes to atherogenesis. Endothelial cells express anti-apoptotic genes including Bcl-2, Bcl-XL and survivin, which also contribute to angiogenesis and vascular remodeling. We report a central role for protein kinase Cepsilon (PKCepsilon) in the regulation of Bcl-2 expression and cytoprotection of human vascular endothelium against apoptosis. Using myristoylated inhibitory peptides, a predominant role for PKCepsilon in vascular endothelial growth factor-mediated endothelial resistance to apoptosis was revealed. Immunoblotting of endothelial cells infected with an adenovirus expressing a constitutively active form of PKCepsilon (Adv-PKCepsilon-CA) or control Adv-beta-galactosidase demonstrated a 3-fold, PKCepsilon-dependent increase in Bcl-2 expression, with no significant change in Bcl-XL, Bad, Bak, or Bax. The induction of Bcl-2 inhibited apoptosis induced by serum starvation or etoposide, and PKCepsilon activation attenuated etoposide-induced caspase-3 cleavage. The functional role of Bcl-2 was confirmed with Bcl-2 antagonist HA-14-1. Inhibition of phosphoinositide 3-kinase attenuated vascular endothelial growth factor-induced protection against apoptosis, and this was rescued by overexpression of constitutively active PKCepsilon, suggesting PKCepsilon acts downstream of phosphoinositide 3-kinase. Co-immunoprecipitation studies demonstrated a physical interaction between PKCepsilon and Akt, which resulted in formation of a signaling complex, leading to optimal induction of Bcl-2. This study reveals a pivotal role for PKCepsilon in endothelial cell cytoprotection against apoptosis. We demonstrate that PKCepsilon forms a signaling complex and acts co-operatively with Akt to protect human vascular endothelial cells against apoptosis through induction of the anti-apoptotic protein Bcl-2 and inhibition of caspase-3 cleavage.


Assuntos
Apoptose , Células Endoteliais/metabolismo , Proteína Quinase C-épsilon/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Transdução de Sinais , Células Endoteliais/citologia , Ativação Enzimática , Humanos , Proteína Quinase C-épsilon/química , Veias Umbilicais/citologia , Veias Umbilicais/metabolismo
17.
Cell Signal ; 19(9): 1830-43, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17561374

RESUMO

Phospholipase Cgamma1 (PLCgamma1) represents a major downstream signalling component of the epidermal growth factor (EGF) receptor (EGFR) and is activated by tyrosine phosphorylation. Here we show for the first time that cellular knockdown of protein kinase Cepsilon (PKCepsilon) leads to decreased activation of PLCgamma1 by EGF and that EGF induces tyrosine phosphorylation of PKCepsilon as well as association of PKCepsilon with both EGFR and PLCgamma1. Using several mutants, co-immunoprecipitation and phosphopeptide-based pull-down experiments we found that in dependency on c-Src and EGF-stimulation PKCepsilon may bind to the c-Src-specific phosphorylation site pY845-EGFR. Furthermore, we identified a single tyrosine residue, PKCepsilon-Y573, within a consensus binding sequence of the C-terminal SH2 domain of PLCgamma1 which is critical for both tyrosine phosphorylation of PKCepsilon and its association with PLCgamma1. Thus, in particular cells and independent of the kinase activity PKCepsilon may form a signalling module with EGFR and PLCgamma1. Thereby the tyrosine phosphorylation of PLCgamma1 via the EGFR may be facilitated. This is a novel function of PKCepsilon upstream of PLCgamma1 and a novel paradigm for the EGF-induced formation of multi-protein complexes.


Assuntos
Fator de Crescimento Epidérmico/farmacologia , Fosfolipase C gama/metabolismo , Proteína Quinase C-épsilon/metabolismo , Sequência de Aminoácidos , Animais , Células COS , Catálise/efeitos dos fármacos , Chlorocebus aethiops , Sequência Consenso , Ativação Enzimática/efeitos dos fármacos , Indução Enzimática/efeitos dos fármacos , Receptores ErbB/metabolismo , Humanos , Imunoprecipitação , Dados de Sequência Molecular , Proteínas Mutantes/metabolismo , Fosfolipase C gama/química , Fosfotirosina/metabolismo , Ligação Proteica/efeitos dos fármacos , Proteína Quinase C-épsilon/biossíntese , Proteína Quinase C-épsilon/química , Proteínas Proto-Oncogênicas pp60(c-src)/metabolismo , Interferência de RNA , Domínios de Homologia de src
18.
J Biol Chem ; 282(32): 23631-8, 2007 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-17569658

RESUMO

Protein kinase Cdelta (PKCdelta) is unusual among AGC kinases in that it does not require activation loop (Thr(505)) phosphorylation for catalytic competence. Nevertheless, Thr(505) phosphorylation has been implicated as a mechanism that influences PKCdelta activity. This study examines the controls of PKCdelta-Thr(505) phosphorylation in cardiomyocytes. We implicate phosphoinositide-dependent kinase-1 and PKCdelta autophosphorylation in the "priming" maturational PKCdelta-Thr(505) phosphorylation that accompanies de novo enzyme synthesis. In contrast, we show that PKCdelta-Thr(505) phosphorylation dynamically increases in cardiomyocytes treated with phorbol 12-myristate 13-acetate or the alpha(1)-adrenergic receptor agonist norepinephrine via a mechanism that requires novel PKC isoform activity and not phosphoinositide-dependent kinase-1. We used a PKCepsilon overexpression strategy as an initial approach to discriminate two possible novel PKC mechanisms, namely PKCdelta-Thr(505) autophosphorylation and PKCdelta-Thr(505) phosphorylation in trans by PKCepsilon. Our studies show that adenovirus-mediated PKCepsilon overexpression leads to an increase in PKCdelta-Thr(505) phosphorylation. However, this cannot be attributed to an effect of PKCepsilon to function as a direct PKCdelta-Thr(505) kinase, since the PKCepsilon-dependent increase in PKCdelta-Thr(505) phosphorylation is accompanied by (and dependent upon) increased PKCdelta phosphorylation at Tyr(311) and Tyr(332). Further studies implicate Src in this mechanism, showing that 1) PKCepsilon overexpression increases PKCdelta-Thr(505) phosphorylation in cardiomyocytes and Src(+) cells but not in SYF cells (that lack Src, Yes, and Fyn and exhibit a defect in PKCdelta-Tyr(311)/Tyr(332) phosphorylation), and 2) in vitro PKCdelta-Thr(505) autophosphorylation is augmented in assays performed with Src (which promotes PKCdelta-Tyr(311)/Tyr(332) phosphorylation). Collectively, these results identify a novel PKCdelta-Thr(505) autophosphorylation mechanism that is triggered by PKCepsilon overexpression and involves Src-dependent PKCdelta-Tyr(311)/Tyr(332) phosphorylation.


Assuntos
Ativação Enzimática , Miócitos Cardíacos/metabolismo , Proteína Quinase C-delta/química , Proteína Quinase C-delta/metabolismo , Proteína Quinase C-épsilon/química , Proteína Quinase C-épsilon/metabolismo , Proteínas Proto-Oncogênicas pp60(c-src)/fisiologia , Sítio Alostérico , Animais , Fibroblastos/metabolismo , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Miocárdio/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas pp60(c-src)/metabolismo , Ratos , Ratos Wistar
19.
Eur J Pharmacol ; 547(1-3): 174-83, 2006 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-16925996

RESUMO

We asked whether or not antiallergic drugs, azelastine hydrochloride and epinastine hydrochloride, inhibit IgE secretion from IgE-producing hybridoma FE-3 cells. FE-3 cells were cultured in the presence of azelastine or epinastine for 24 h, washed in phosphate-buffered saline , and then recultured in the medium in the absence of the antiallergic drugs. IgE levels in the cultured medium as well as those in the cytoplasm of FE-3 cells were measured by enzyme-linked immunosorbent assay. mRNA levels of Cepsilon, activation-induced cytidine deaminase (AID), XBP-1, and Bip were estimated by northern blot or reverse transcriptase polymerase chain reaction analysis. The activities of nuclear factor-kappa B (NF-kappaB) were analyzed by electrophoretic mobility shift assay (EMSA). Phosphorylation of I kappa B alpha (IkappaBalpha) was analyzed by immunoprecipitation followed by western blot analysis. IgE levels in the cultured medium and in the microsome fraction were lower on the treatment with 10(-5) M azelastine or epinastine than those on the treatment with vehicle. Cepsilon and AID mRNA levels were lower on the treatment with 10(-5) M azelastine than those on the treatment with vehicle, but were not decreased on the treatment with 10(-5) M epinastine. XBP-1 and Bip mRNA levels were not altered following treatment of the antiallergic drugs. Azelastine at 10(-5) M, but not epinastine, reduced DNA binding activity of NF-kappaB and also diminished IkappaBalpha phosphorylation, leading to sustaining IkappaBalpha protein levels. These findings suggest that azelastine exerts its inhibitory effect on the IgE secretion from FE-3 cells through the inhibition of Cepsilon mRNA expression, and that the inhibitory effect of epinastine is, at least in part, due to suppression of IgE synthesis at the post-transcriptional level.


Assuntos
Antialérgicos/farmacologia , Dibenzazepinas/farmacologia , Hibridomas/efeitos dos fármacos , Imidazóis/farmacologia , Imunoglobulina E/metabolismo , Ftalazinas/farmacologia , Animais , Fatores de Transcrição de Zíper de Leucina Básica/química , Fatores de Transcrição de Zíper de Leucina Básica/genética , Fatores de Transcrição de Zíper de Leucina Básica/metabolismo , Northern Blotting , Western Blotting , Citidina Desaminase/química , Citidina Desaminase/genética , Citidina Desaminase/metabolismo , Proteínas de Ligação a DNA , Relação Dose-Resposta a Droga , Ensaio de Desvio de Mobilidade Eletroforética , Expressão Gênica/efeitos dos fármacos , Proteínas de Choque Térmico/química , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Hibridomas/citologia , Hibridomas/metabolismo , Proteínas I-kappa B/metabolismo , Chaperonas Moleculares/química , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , NF-kappa B/genética , NF-kappa B/metabolismo , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Oligonucleotídeos/genética , Oligonucleotídeos/metabolismo , Fosforilação/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Dobramento de Proteína , Proteína Quinase C-épsilon/química , Proteína Quinase C-épsilon/genética , Proteína Quinase C-épsilon/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Fatores de Transcrição de Fator Regulador X , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição , Proteína 1 de Ligação a X-Box
20.
Biochem Biophys Res Commun ; 349(1): 91-8, 2006 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-16930532

RESUMO

We have shown that protein kinase C (PKC)epsilon, independently of the catalytic domain, induces outgrowth of cellular processes via its regulatory domain in both neural cells and fibroblasts. This was accompanied by stress fibre loss. Here, we have examined the role of the small GTPases, Rac1, and Cdc42, in these PKC-mediated morphological and cytoskeletal changes. Both constitutively active and dominant negative Rac1 and Cdc42 attenuated the PKC-mediated outgrowth of processes. The suppression was larger for Cdc42 than for Rac1. The PKC-mediated dismantling of the stress fibres in both HiB5 and fibroblasts was inhibited by the expression of the Cdc42 mutants whereas they had smaller effects on the stress fibre dismantling induced by the ROCK inhibitor, Y-27632, indicating a more crucial role for Cdc42 in the PKC-mediated pathway. We conclude that Cdc42 is an important downstream factor in the pathway through which PKC mediates morphological and cytoskeletal effects.


Assuntos
Neurônios/metabolismo , Proteína Quinase C-delta/química , Proteína Quinase C-épsilon/química , Proteína cdc42 de Ligação ao GTP/fisiologia , Amidas/farmacologia , Animais , Domínio Catalítico , Linhagem Celular Tumoral , Fibroblastos/metabolismo , Humanos , Camundongos , Células NIH 3T3 , Neuroblastoma/metabolismo , Piridinas/farmacologia , Proteína cdc42 de Ligação ao GTP/química , Proteínas rac1 de Ligação ao GTP/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA