Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Arch Biochem Biophys ; 705: 108910, 2021 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-33991498

RESUMEN

The C2 domain of novel protein kinases C (nPKC) binds to membranes in a Ca2+-independent way contributing to the activation of these enzymes. We have studied the C2 domain of one of these nPKCs, namely PKCε, and confirmed that it establishes a strong interaction with POPA, which is clearly visible through changes in chemical shifts detected through 31P-MAS-NMR and the protection that it exerts on the domain against thermal denaturation seen through DSC and FT-IR. In this study, using two-dimensional correlation analysis (2D-COS) applied to infrared spectra, we determined the sequence of events that occur during the thermal unfolding of the domain and highlighted some differences when phosphatidic acid or cardiolipin are present. Finally, by means of FRET and DLS experiments, we wanted to determine the effect of membrane curvature on the domain/membrane interaction by using lysophosphatidylcholine to introduce positive curvature as a control and we observed that the effect of these phospholipids on the protein binding is not exerted through the change of membrane curvature.


Asunto(s)
Membrana Celular/metabolismo , Proteína Quinasa C-epsilon/química , Proteína Quinasa C-epsilon/metabolismo , Desplegamiento Proteico , Cardiolipinas/metabolismo , Membrana Celular/química , Modelos Moleculares , Ácidos Fosfatidicos/metabolismo , Unión Proteica , Dominios Proteicos
2.
Biomolecules ; 10(9)2020 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-32906765

RESUMEN

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.


Asunto(s)
Receptores ErbB/metabolismo , Proteína Quinasa C-epsilon/química , Proteína Quinasa C-epsilon/metabolismo , Actinas/metabolismo , Animales , Línea Celular , Membrana Celular/metabolismo , Endosomas/metabolismo , Epítopos/metabolismo , Oro , Mesotelina , Nanopartículas del Metal , Transporte de Proteínas/fisiología , Ratas
3.
BMC Bioinformatics ; 19(Suppl 13): 342, 2019 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-30717648

RESUMEN

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.


Asunto(s)
Azepinas/metabolismo , Halogenación , Hidroxibenzoatos/metabolismo , Proteína Quinasa C-epsilon/metabolismo , Adenosina Trifosfato/metabolismo , Azepinas/química , Análisis por Conglomerados , Humanos , Hidroxibenzoatos/química , Isoenzimas/metabolismo , Simulación de Dinámica Molecular , Fosforilación , Conformación Proteica , Proteína Quinasa C-epsilon/química , Ribosa/química , Alineación de Secuencia , Electricidad Estática , Especificidad por Sustrato
4.
Cell Rep ; 25(9): 2470-2483.e8, 2018 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-30485813

RESUMEN

Protein kinase C (PKC)-dependent mechanisms promote synaptic function in the mature brain. However, the roles of PKC signaling during synapse development remain largely unknown. Investigating each brain-enriched PKC isoform in early neuronal development, we show that PKCε acutely and specifically reduces the number of dendritic spines, sites of eventual synapse formation on developing dendrites. This PKCε-mediated spine suppression is temporally restricted to immature neurons and mediated through the phosphorylation and activation of Ephexin5, a RhoA guanine nucleotide exchange factor (GEF) and inhibitor of hippocampal synapse formation. Our data suggest that PKCε acts as an early developmental inhibitor of dendritic spine formation, in contrast to its emerging pro-synaptic roles in mature brain function. Moreover, we identify a substrate of PKCε, Ephexin5, whose early-elevated expression in developing neurons may in part explain the mechanism by which PKCε plays seemingly opposing roles that depend on neuronal maturity.


Asunto(s)
Espinas Dendríticas/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Proteína Quinasa C-epsilon/metabolismo , Secuencia de Aminoácidos , Animales , Animales Recién Nacidos , Encéfalo/metabolismo , Diferenciación Celular , Activación Enzimática , Células HEK293 , Hipocampo/metabolismo , Humanos , Isoenzimas/metabolismo , Ratones Endogámicos C57BL , Fosforilación , Fosfoserina/metabolismo , Proteína Quinasa C-alfa/metabolismo , Proteína Quinasa C-epsilon/química , Transducción de Señal , Proteína de Unión al GTP rhoA/metabolismo
5.
J Chem Inf Model ; 58(2): 511-519, 2018 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-29341608

RESUMEN

(-)-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.


Asunto(s)
Azepinas/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/química , Flúor/química , Hidroxibenzoatos/metabolismo , Simulación de Dinámica Molecular , Proteína Quinasa C-epsilon/química , Sitios de Unión , Humanos
6.
FEBS Lett ; 592(2): 179-189, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29266266

RESUMEN

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.


Asunto(s)
Péptidos/farmacología , Proteína Quinasa C-epsilon/química , Proteína Quinasa C-epsilon/metabolismo , Receptores de Cinasa C Activada/metabolismo , Animales , Sitios de Unión , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Péptidos/química , Unión Proteica/efectos de los fármacos , Dominios Proteicos , Estructura Terciaria de Proteína , Resonancia por Plasmón de Superficie
7.
Bioorg Med Chem ; 25(12): 2971-2980, 2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-28392275

RESUMEN

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.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Indoles/química , Indoles/farmacología , Lactonas/química , Lactonas/farmacología , Proteína Quinasa C-alfa/metabolismo , Proteína Quinasa C-epsilon/metabolismo , Proteínas de Unión al ADN/química , Factores de Intercambio de Guanina Nucleótido/química , Humanos , Simulación del Acoplamiento Molecular , Unión Proteica , Dominios Proteicos , Proteína Quinasa C-alfa/química , Proteína Quinasa C-epsilon/química , Relación Estructura-Actividad
8.
J Biomed Sci ; 24(1): 3, 2017 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-28056995

RESUMEN

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.


Asunto(s)
Aldehído Deshidrogenasa Mitocondrial/química , Evolución Molecular , Proteína Quinasa C-epsilon/química , Aldehído Deshidrogenasa Mitocondrial/genética , Aldehído Deshidrogenasa Mitocondrial/metabolismo , Humanos , Fosforilación/fisiología , Proteína Quinasa C-epsilon/genética , Proteína Quinasa C-epsilon/metabolismo
9.
Biochemistry ; 55(45): 6327-6336, 2016 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-27776404

RESUMEN

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.


Asunto(s)
Curcumina/química , Dominios Proteicos , Proteína Quinasa C-alfa/química , Proteína Quinasa C-epsilon/química , Sitios de Unión/genética , Unión Competitiva , Biocatálisis/efectos de los fármacos , Curcumina/metabolismo , Curcumina/farmacología , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacología , Células HEK293 , Humanos , Immunoblotting , Cinética , Microscopía Confocal , Simulación de Dinámica Molecular , Mutación , Ésteres del Forbol/farmacología , Unión Proteica , Proteína Quinasa C-alfa/genética , Proteína Quinasa C-alfa/metabolismo , Proteína Quinasa C-epsilon/genética , Proteína Quinasa C-epsilon/metabolismo , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/genética , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
10.
Biochemistry ; 55(14): 2135-43, 2016 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-26983836

RESUMEN

Members of the protein kinase C (PKC) family of serine/threonine kinases regulate various cellular functions, including cell growth, differentiation, metabolism, and apoptosis. Modulation of isoform-selective activity of PKC by curcumin (1), the active constituent of Curcuma L., is poorly understood, and the literature data are inconsistent and obscure. The effect of curcumin (1) and its analogues, 4-[(2Z,6E)-3-hydroxy-7-(4-hydroxy-3-methoxyphenyl)-5-oxohepta-2,6-dien-1-yl]-2-methoxyphenyl oleate (2), (9Z,12Z)-4-[(2Z,6E)-3-hydroxy-7-(4-hydroxy-3-methoxyphenyl)-5-oxohepta-2,6-dien-1-yl]-2-methoxyphenyl octadeca-9,12-dienoate (3), (9Z,12Z,15Z)-4-[(2Z,6E)-3-hydroxy-7-(4-hydroxy-3-methoxyphenyl)-5-oxohepta-2,6-dien-1-yl]-2-methoxyphenyl octadeca-9,12,15-trienoate (4), and (1E,6E)-1-[4-(hexadecyloxy)-3-methoxyphenyl]-7-(4-hydroxy-3-methoxyphenyl)hepta-1,6-diene-3,5-dione (5), and didemethylcurcumin (6) on the membrane translocation of PKCα, a conventional PKC, and PKCε, a novel PKC, has been studied in CHO-K1 cells, in which these PKC isoforms are endogenously expressed. Translocation of PKC from the cytosol to the membrane was measured using immunoblotting and confocal microscopy. 1 and 6 inhibited the TPA-induced membrane translocation of PKCα but not of PKCε. Modification of the hydroxyl group of curcumin with a long aliphatic chain containing unsaturated double bonds in 2-4 completely abolished this inhibition property. Instead, 2-4 showed significant translocation of PKCα but not of PKCε to the membrane. No membrane translocation was observed with 1, 6, or the analogue 5 having a saturated long chain for either PKCα or PKCε. 1 and 6 inhibited TPA-induced activation of ERK1/2, and 2-4 activated it. ERK1/2 is the downstream readout of PKC. These results show that the hydroxyl group of curcumin is important for PKC activity and the curcumin template can be useful in developing isoform specific PKC modulators for regulating a particular disease state.


Asunto(s)
Antioxidantes/farmacología , Curcumina/análogos & derivados , Diseño de Fármacos , Proteína Quinasa C-alfa/metabolismo , Proteína Quinasa C-epsilon/metabolismo , Animales , Antioxidantes/efectos adversos , Antioxidantes/química , Células CHO , Membrana Celular/efectos de los fármacos , Membrana Celular/enzimología , Supervivencia Celular/efectos de los fármacos , Cricetulus , Curcumina/efectos adversos , Curcumina/química , Curcumina/farmacología , Activación Enzimática/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Cinética , Lipoilación , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Metilación , Microscopía Confocal , Fosforilación/efectos de los fármacos , Proteína Quinasa C-alfa/antagonistas & inhibidores , Proteína Quinasa C-alfa/química , Proteína Quinasa C-epsilon/antagonistas & inhibidores , Proteína Quinasa C-epsilon/química , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos
11.
Biochim Biophys Acta ; 1850(11): 2368-76, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26210390

RESUMEN

BACKGROUND: Alcohol regulates the expression and function of protein kinase C epsilon (PKCε). In a previous study we identified an alcohol binding site in the C1B, one of the twin C1 subdomains of PKCε (Das et al., Biochem. J., 421, 405-13, 2009). METHODS: In this study, we investigated alcohol binding in the entire C1 domain (combined C1A and C1B) of PKCε. Fluorescent phorbol ester, SAPD and fluorescent diacylglycerol (DAG) analog, dansyl-DAG were used to study the effect of ethanol, butanol, and octanol on the ligand binding using fluorescence resonance energy transfer (FRET). To identify alcohol binding site(s), PKCεC1 was photolabeled with 3-azibutanol and 3-azioctanol, and analyzed by mass spectrometry. The effects of alcohols and the azialcohols on PKCε were studied in NG108-15 cells. RESULTS: In the presence of alcohol, SAPD and dansyl-DAG showed different extent of FRET, indicating differential effects of alcohol on the C1A and C1B subdomains. Effects of alcohols and azialcohols on PKCε in NG108-15 cells were comparable. Azialcohols labeled Tyr-176 of C1A and Tyr-250 of C1B. Inspection of the model structure of PKCεC1 reveals that these residues are 40Šapart from each other indicating that these residues form two different alcohol binding sites. CONCLUSIONS: The present results provide evidence for the presence of multiple alcohol-binding sites on PKCε and underscore the importance of targeting this PKC isoform in developing alcohol antagonists.


Asunto(s)
Alcoholes/farmacología , Proteína Quinasa C-epsilon/química , Secuencia de Aminoácidos , Sitios de Unión , Transferencia Resonante de Energía de Fluorescencia , Datos de Secuencia Molecular , Estructura Terciaria de Proteína
12.
Mol Med Rep ; 11(6): 4720-6, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25625661

RESUMEN

Mitochondrial connexin 43 (Cx43) is important in cardioprotection by ischemic preconditioning; however, whether mitochondrial Cx43 is involved in mitochondrial dysfunction in the pathogenesis of dilated cardiomyopathy (DCM) remains to be elucidated. The present study was performed to investigate the changes in expression and the phosphorylation state of mitochondrial Cx43 in a rat model of DCM, and to determine whether the altered phosphorylation state of mitochondrial Cx43 was involved in mitochondrial dysfunction. A rat model of DCM was generated by daily oral administration of furazolidone (FZD) for 30 weeks. Reverse transcription polymerase chain reaction and western blot analysis revealed a decrease in the overall expression of Cx43, accompanied by reduced levels of serine 368­phosphorylated­Cx43 immunoreactivity in the myocardium and myocardial mitochondria. In addition, the mitochondrial membrane potential and the activities of cytochrome c oxidase, succinate dehydrogenase and protein kinase C (PKC) Îµ were all significantly reduced compared with those of the control group. Phorbol­12­myristate­13­acetate (PMA), a specific PKC activator, partially reversed the FZD­induced mitochondrial Cx43 dephosphorylation at serine 368 and mitochondrial dysfunction in the cardiomyocytes. However, pretreatment with 18ß­glycerrhetinic acid, a connexin channel inhibitor, eliminated the mitochondrial protective effect of PMA in the cardiomyocytes sparsely plated without cell to cell contact. These results suggested that dephosphorylation of mitochondrial Cx43 at serine 368, due to the suppression of PKCε activity, may be a novel mechanism for mitochondrial dysfunction in the pathogenesis of DCM.


Asunto(s)
Cardiomiopatía Dilatada/patología , Conexina 43/metabolismo , Mitocondrias Cardíacas/metabolismo , Proteína Quinasa C-epsilon/metabolismo , Administración Oral , Animales , Cardiomiopatía Dilatada/tratamiento farmacológico , Cardiomiopatía Dilatada/metabolismo , Células Cultivadas , Conexina 43/química , Modelos Animales de Enfermedad , Complejo IV de Transporte de Electrones/metabolismo , Furazolidona/uso terapéutico , Ácido Glicirretínico/análogos & derivados , Ácido Glicirretínico/farmacología , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Miocardio/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Fosforilación/efectos de los fármacos , Proteína Quinasa C-epsilon/química , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Serina/metabolismo , Succinato Deshidrogenasa/metabolismo , Acetato de Tetradecanoilforbol/farmacología , Transcripción Genética/efectos de los fármacos
13.
J Med Chem ; 57(8): 3235-46, 2014 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-24712764

RESUMEN

Ten protein kinase C (PKC) isozymes play divergent roles in signal transduction. Because of sequence similarities, it is particularly difficult to generate isozyme-selective small molecule inhibitors. In order to identify such a selective binder, we derived a pharmacophore model from the peptide EAVSLKPT, a fragment of PKCε that inhibits the interaction of PKCε and receptor for activated C-kinase 2 (RACK2). A database of 330 000 molecules was screened in silico, leading to the discovery of a series of thienoquinolines that disrupt the interaction of PKCε with RACK2 in vitro. The most active molecule, N-(3-acetylphenyl)-9-amino-2,3-dihydro-1,4-dioxino[2,3-g]thieno[2,3-b]quinoline-8-carboxamide (8), inhibited this interaction with a measured IC50 of 5.9 µM and the phosphorylation of downstream target Elk-1 in HeLa cells with an IC50 of 11.2 µM. Compound 8 interfered with MARCKS phosphorylation and TPA-induced translocation of PKCε (but not that of PKCδ) from the cytosol to the membrane. The compound reduced the migration of HeLa cells into a gap, reduced invasion through a reconstituted basement membrane matrix, and inhibited angiogenesis in a chicken egg assay.


Asunto(s)
Proteína Quinasa C-epsilon/antagonistas & inhibidores , Quinolinas/farmacología , Receptores de Superficie Celular/antagonistas & inhibidores , Animales , Proliferación Celular/efectos de los fármacos , Embrión de Pollo , Descubrimiento de Drogas , Células HeLa , Humanos , Modelos Moleculares , Fosforilación , Unión Proteica , Proteína Quinasa C-epsilon/química , Inhibidores de Proteínas Quinasas/farmacología , Transporte de Proteínas/efectos de los fármacos , Receptores de Cinasa C Activada , Receptores de Superficie Celular/química , Relación Estructura-Actividad , Proteína Elk-1 con Dominio ets/metabolismo
14.
Cell Transplant ; 23(7): 913-9, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-23562311

RESUMEN

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.


Asunto(s)
Diabetes Mellitus Experimental/terapia , Trasplante de Islotes Pancreáticos , Islotes Pancreáticos/efectos de los fármacos , Péptidos/farmacología , Proteína Quinasa C-epsilon/química , Animales , Glucemia/análisis , Modelos Animales de Enfermedad , Femenino , Prueba de Tolerancia a la Glucosa , Supervivencia de Injerto/efectos de los fármacos , Insulina/metabolismo , Secreción de Insulina , Islotes Pancreáticos/citología , Islotes Pancreáticos/metabolismo , Ratones , Ratones Endogámicos BALB C , Proteína Quinasa C-epsilon/metabolismo , Trasplante Homólogo
15.
Artículo en Inglés | MEDLINE | ID: mdl-24291721

RESUMEN

Bryostatin 1, a potential anti-Alzheimer drug, is effective at subnanomolar concentrations. Measurement is complicated by the formation of low m/z degradation products and the formation of adducts with various cations, which make accurate quantitation difficult. Adduct formation caused the sample matrix or mobile phase to partition bryostatin 1 into products of different mass. Degradation of the 927 [M+Na](+) ion to a 869m/z product was strongly influenced by ionization conditions. We validated a bryostatin 1 assay in biological tissues using capillary column HPLC with nanospray ionization (NSI) in a triple-quadrupole mass spectrometer in selected reaction monitoring (SRM) mode. Adduct formation was controlled by adding 1mM acetic acid and 0.1mM sodium acetate to the HPLC buffer, maximizing the formation of the [M+Na](+) ion. Efficient removal of contaminating cholesterol from the sample during solvent extraction was also critical. The increased sensitivity provided by NSI and capillary-bore columns and the elimination of signal partitioning due to adduct formation and degradation in the ionization source enabled a detection limit of 1×10(-18)mol of bryostatin 1 and a LLOQ of 3×10(-18)mol from 1µl of sample. Bryostatin 1 at low pmol/l concentrations enabled measurement in brain and other tissues without the use of radioactive labels. Despite bryostatin 1's high molecular weight, considerable brain access was observed, with peak brain concentrations exceeding 8% of the peak blood plasma concentrations. Bryostatin 1 readily crosses the blood-brain barrier, reaching peak concentrations of 0.2nM, and specifically activates and translocates brain PKCɛ.


Asunto(s)
Brioestatinas/análisis , Brioestatinas/sangre , Cromatografía Liquida/métodos , Espectrometría de Masas/métodos , Animales , Química Encefálica , Brioestatinas/química , Brioestatinas/farmacología , Bovinos , Estabilidad de Medicamentos , Humanos , Límite de Detección , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína Quinasa C-epsilon/análisis , Proteína Quinasa C-epsilon/química , Proteína Quinasa C-epsilon/metabolismo , Reproducibilidad de los Resultados
16.
Biochim Biophys Acta ; 1828(2): 552-60, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23088913

RESUMEN

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°).


Asunto(s)
Transferencia Resonante de Energía de Fluorescencia/métodos , Membrana Dobles de Lípidos/química , Proteína Quinasa C-epsilon/química , Espectrofotometría Infrarroja/métodos , Adenosina/análogos & derivados , Adenosina/química , Calcio/química , Glicerofosfolípidos/química , Humanos , Lípidos/química , Membranas Mitocondriales/metabolismo , Modelos Moleculares , Modelos Estadísticos , Conformación Molecular , Fosfatidilcolinas/química , Fosfatidiletanolaminas/química , Fosfolípidos/química , Unión Proteica , Conformación Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína
17.
Cell Physiol Biochem ; 30(3): 771-7, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22854271

RESUMEN

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.


Asunto(s)
Inhibidores de la Ciclooxigenasa/farmacología , Activación Enzimática/efectos de los fármacos , Indometacina/farmacología , Proteína Quinasa C-epsilon/metabolismo , Animales , Fluoresceína/química , Células PC12 , Prostaglandina-Endoperóxido Sintasas/química , Prostaglandina-Endoperóxido Sintasas/metabolismo , Unión Proteica , Isoformas de Proteínas/metabolismo , Proteína Quinasa C-epsilon/química , Proteína Quinasa C-epsilon/genética , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Ratas , Especificidad por Sustrato
18.
PLoS One ; 7(4): e35630, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22536418

RESUMEN

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.


Asunto(s)
Activadores de Enzimas/farmacología , Proteína Quinasa C-epsilon/metabolismo , Anticuerpos de Cadena Única/farmacología , Animales , Encéfalo/enzimología , Activación Enzimática , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Humanos , Cinética , Unión Proteica , Proteína Quinasa C-epsilon/antagonistas & inhibidores , Proteína Quinasa C-epsilon/química , Transporte de Proteínas , Ratas , Proteínas Recombinantes de Fusión/metabolismo , Anticuerpos de Cadena Única/biosíntesis , Anticuerpos de Cadena Única/química , Especificidad de la Especie , Resonancia por Plasmón de Superficie , Acetato de Tetradecanoilforbol/farmacología
19.
PLoS One ; 7(12): e52888, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23285216

RESUMEN

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.


Asunto(s)
Ácidos Grasos/química , Proteína Quinasa C-alfa/metabolismo , Proteína Quinasa C-epsilon/metabolismo , Estilbenos/química , Estilbenos/farmacología , Animales , Células CHO , Supervivencia Celular/efectos de los fármacos , Cricetinae , Cricetulus , Activación Enzimática/efectos de los fármacos , Humanos , Isoenzimas/química , Isoenzimas/metabolismo , Modelos Biológicos , Modelos Moleculares , Simulación del Acoplamiento Molecular , Proteína Quinasa C-alfa/química , Proteína Quinasa C-epsilon/química , Resveratrol , Transducción de Señal/efectos de los fármacos , Relación Estructura-Actividad
20.
Phytomedicine ; 19(2): 99-110, 2012 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-21899994

RESUMEN

Danshen-Gegen (DG) Decoction, an herbal formulation containing Radix Salviae miltiorrhizae and Radix Puerariae lobatae, has been used for the treatment of coronary artery disease in Chinese medicine. In the present study, the involvement of ERK- and PKCε-mediated pathways in the cytoprotection against apoptosis afforded by DG pretreatment was investigated in H9c2 cardiomyocytes. Pretreatment with a methanol extract of aqueous DG decoction protected against hypoxia/reoxygenation-induced apoptosis in H9c2 cardiomyocytes. The cytoprotection was associated the enhancement of cellular reduced glutathione and a reduced sensitivity to Ca(2+)-induced mitochondrial permeability transition. DG extract increased the production of cytochrome P-450 (CYP)-dependent reactive oxygen species (ROS) in H9c2 cardiomyocytes, which was accompanied by the concomitant activation of ERK1/2 and PKCε. The DG-induced ERK1/2 activation was followed by the translocation of Nrf2 from the cytosol to the mitochondria accompanied by an increase in the expression of glutathione-related antioxidant proteins. In addition, the increased expression of hemeoxygenase-1 was associated with the activation of Akt and BAD, indicative of anti-apoptotic activity. In conclusion, DG treatment activated both ERK/Nrf2 and PKCε pathways, presumably by ROS arising from CYP-catalyzed processes, with resultant inhibition of hypoxia/reoxygenation-induced apoptosis immediately after DG treatment or even after an extended time interval following DG treatment.


Asunto(s)
Apoptosis , Medicamentos Herbarios Chinos/farmacología , Mitocondrias Cardíacas/química , Miocitos Cardíacos/efectos de los fármacos , Animales , Antioxidantes/química , Western Blotting , Calcio/química , Hipoxia de la Célula , Línea Celular , Sistema Enzimático del Citocromo P-450/química , Citoprotección , Citosol/química , Sistema de Señalización de MAP Quinasas , Metanol/química , Mitocondrias Cardíacas/efectos de los fármacos , Miocitos Cardíacos/química , Factor 2 Relacionado con NF-E2/química , Oxidación-Reducción , Permeabilidad , Proteína Quinasa C-epsilon/química , Transporte de Proteínas , Pueraria/química , Ratas , Especies Reactivas de Oxígeno/química , Salvia miltiorrhiza/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA