Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Cell Rep ; 24(3): 655-669, 2018 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-30021163

RESUMO

Protein kinase C (PKC) isozymes are commonly recognized as oncoproteins based on their activation by tumor-promoting phorbol esters. However, accumulating evidence indicates that PKCs can be inhibitory in some cancers, with recent findings propelling a shift in focus to understanding tumor suppressive functions of these enzymes. Here, we report that PKCα acts as a tumor suppressor in PI3K/AKT-driven endometrial cancer. Transcriptional suppression of PKCα is observed in human endometrial tumors in association with aggressive disease and poor prognosis. In murine models, loss of PKCα is rate limiting for endometrial tumor initiation. PKCα tumor suppression involves PP2A-family-dependent inactivation of AKT, which can occur even in the context of genetic hyperactivation of PI3K/AKT signaling by coincident mutations in PTEN, PIK3CA, and/or PIK3R1. Together, our data point to PKCα as a crucial tumor suppressor in the endometrium, with deregulation of a PKCα→PP2A/PP2A-like phosphatase signaling axis contributing to robust AKT activation and enhanced endometrial tumorigenesis.


Assuntos
Neoplasias do Endométrio/enzimologia , Neoplasias do Endométrio/patologia , Endométrio/enzimologia , Endométrio/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteína Quinase C-alfa/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Animais , Carcinogênese/metabolismo , Carcinogênese/patologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Ativação Enzimática , Feminino , Células HEK293 , Humanos , Camundongos , Gradação de Tumores , Proteínas Nucleares/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Fosforilação , Proteína Quinase C-alfa/deficiência , Proteína Fosfatase 2/metabolismo
2.
Am J Physiol Renal Physiol ; 315(1): F7-F15, 2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-29412702

RESUMO

α-Ketoglutarate (α-KG) is a citric acid cycle intermediate and a glutamine catabolism product. It is also the natural ligand of 2-oxoglutarate receptor 1 (OXGR1), a Gq protein-coupled receptor expressed on the apical membrane of intercalated cells. In the cortical collecting duct (CCD), Cl-/[Formula: see text] exchange increases upon α-KG binding to the OXGR1. To determine the signaling pathway(s) by which α-KG stimulates Cl- absorption, we examined α-KG-stimulated Cl- absorption in isolated perfused mouse CCDs. α-KG increased electroneutral Cl- absorption in CCDs from wild-type mice but had no effect on Cl- absorption in pendrin knockout mice. Because Gq protein-coupled receptors activate PKC, we hypothesized that α-KG stimulates Cl- absorption through PKC. If so, PKC agonists should mimic, whereas PKC inhibitors should abolish, α-KG-stimulated Cl- absorption. Like α-KG, PKC agonist (phorbol-12,13-dibutyrate, 500 nM) application increased Cl- absorption in wild-type but not in pendrin null CCDs. Moreover, PKC inhibitors (2.5 mM GF109203X and 20 nM calphostin C), Ca2+ chelators (BAPTA, 10-20 µM), or PKC-α or -δ gene ablation eliminated α-KG-stimulated Cl- absorption. We have shown that STE20/SPS-1-related proline-alanine-rich protein kinase (SPAK) gene ablation increases urinary α-KG excretion, renal pendrin abundance, and CCD Cl- absorption. However, in SPAK null CCDs, Cl- absorption was not activated further by luminal α-KG application nor was Cl- absorption reduced with the PKC inhibitor GF109203 . Thus SPAK gene ablation likely acts through a PKC-independent pathway to produce a chronic adaptive increase in pendrin function. In conclusion, α-KG stimulates pendrin-dependent Cl-/[Formula: see text] exchange through a mechanism dependent on PKC and Ca2+ that involves PKC-α and PKC-δ.


Assuntos
Cloretos/metabolismo , Ácidos Cetoglutáricos/farmacologia , Túbulos Renais Coletores/efeitos dos fármacos , Proteína Quinase C-alfa/metabolismo , Proteína Quinase C-delta/metabolismo , Reabsorção Renal/efeitos dos fármacos , Transportadores de Sulfato/metabolismo , Animais , Cálcio/metabolismo , Técnicas In Vitro , Ácidos Cetoglutáricos/metabolismo , Túbulos Renais Coletores/metabolismo , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Proteína Quinase C-alfa/deficiência , Proteína Quinase C-alfa/genética , Proteína Quinase C-delta/deficiência , Proteína Quinase C-delta/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Receptores Purinérgicos P2/efeitos dos fármacos , Receptores Purinérgicos P2/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transportadores de Sulfato/deficiência , Transportadores de Sulfato/genética
3.
Antiviral Res ; 140: 1-12, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28077314

RESUMO

Although hepatitis E has emerged as a global health issue, there is limited knowledge of its infection biology and no FDA-approved medication is available. Aiming to investigate the role of protein kinases in hepatitis E virus (HEV) infection and to identify potential antiviral targets, we screened a library of pharmacological kinase inhibitors in a cell culture model, a subgenomic HEV replicon containing luciferase reporter. We identified protein kinase C alpha (PKCα) as an essential cell host factor restricting HEV replication. Both specific inhibitor and shRNA-mediated knockdown of PKCα enhanced HEV replication. Conversely, over-expression of the activated form of PKCα or treatment with its pharmacological activator strongly inhibited HEV replication. Interestingly, upon the stimulation by its activator, PKCα efficiently activates its downstream Activator Protein 1 (AP-1) pathway, leading to the induction of antiviral interferon-stimulated genes (ISGs). This process is independent of the JAK-STAT machinery and interferon production. However, PKCα induced HEV inhibition appears independent of the AP1 cascade. The discovery that activated PKCα restricts HEV replication reveals new insight of HEV-host interactions and provides new target for antiviral drug development.


Assuntos
Antivirais/farmacologia , Vírus da Hepatite E/fisiologia , Proteína Quinase C-alfa/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Replicação Viral/efeitos dos fármacos , Antivirais/isolamento & purificação , Técnicas de Cultura de Células , Replicação do DNA , Ativação Enzimática , Hepatite E/tratamento farmacológico , Hepatócitos/virologia , Interações Hospedeiro-Patógeno , Humanos , Proteína Quinase C-alfa/antagonistas & inibidores , Proteína Quinase C-alfa/deficiência , Proteína Quinase C-alfa/genética , Inibidores de Proteínas Quinases/isolamento & purificação , Replicon , Transdução de Sinais , Acetato de Tetradecanoilforbol/farmacologia , Fator de Transcrição AP-1/genética , Fator de Transcrição AP-1/metabolismo , Ativação Transcricional
4.
J Vasc Res ; 52(4): 279-88, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26890419

RESUMO

Pulmonary vascular hyperresponsiveness is a main characteristic of pulmonary arterial hypertension (PAH). In PAH patients, elevated levels of the vasoconstrictors thromboxane A2 (TXA2), endothelin (ET)-1 and serotonin further contribute to pulmonary hypertension. Protein kinase C (PKC) isozyme alpha (PKCα) is a known modulator of smooth muscle cell contraction. However, the effects of PKCα deficiency on pulmonary vasoconstriction have not yet been investigated. Thus, the role of PKCα in pulmonary vascular responsiveness to the TXA2 analog U46619, ET-1, serotonin and acute hypoxia was investigated in isolated lungs of PKCα-/- mice and corresponding wild-type mice, with or without prior administration of the PKC inhibitor bisindolylmaleimide I or Gö6976. mRNA was quantified from microdissected intrapulmonary arteries. We found that broad-spectrum PKC inhibition reduced pulmonary vascular responsiveness to ET-1 and acute hypoxia and, by trend, to U46619. Analogously, selective inhibition of conventional PKC isozymes or PKCα deficiency reduced ET-1-evoked pulmonary vasoconstriction. The pulmonary vasopressor response to serotonin was unaffected by either broad PKC inhibition or PKCα deficiency. Surprisingly, PKCα-/- mice showed pulmonary vascular hyperresponsiveness to U46619 and increased TXA2 receptor (TP receptor) expression in the intrapulmonary arteries. To conclude, PKCα regulates ET-1-induced pulmonary vasoconstriction. However, PKCα deficiency leads to pulmonary vascular hyperresponsiveness to TXA2, possibly via increased pulmonary arterial TP receptor expression.


Assuntos
Proteína Quinase C-alfa/deficiência , Artéria Pulmonar/efeitos dos fármacos , Receptores de Tromboxano A2 e Prostaglandina H2/agonistas , Tromboxano A2/farmacologia , Vasoconstrição/efeitos dos fármacos , Vasoconstritores/farmacologia , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacologia , Animais , Relação Dose-Resposta a Droga , Endotelina-1/farmacologia , Feminino , Genótipo , Camundongos da Linhagem 129 , Camundongos Knockout , Fenótipo , Proteína Quinase C-alfa/antagonistas & inibidores , Proteína Quinase C-alfa/genética , Inibidores de Proteínas Quinases/farmacologia , Artéria Pulmonar/enzimologia , Receptores de Tromboxano A2 e Prostaglandina H2/genética , Receptores de Tromboxano A2 e Prostaglandina H2/metabolismo , Serotonina/farmacologia , Regulação para Cima
5.
Elife ; 3: e03011, 2014 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-25097249

RESUMO

In presynaptic boutons, calcium (Ca(2+)) triggers both neurotransmitter release and short-term synaptic plasticity. Whereas synaptotagmins are known to mediate vesicle fusion through binding of high local Ca(2+) to their C2 domains, the proteins that sense smaller global Ca(2+) increases to produce short-term plasticity have remained elusive. Here, we identify a Ca(2+) sensor for post-tetanic potentiation (PTP), a form of plasticity thought to underlie short-term memory. We find that at the functionally mature calyx of Held synapse the Ca(2+)-dependent protein kinase C isoforms α and ß are necessary for PTP, and the expression of PKCß in PKCαß double knockout mice rescues PTP. Disruption of Ca(2+) binding to the PKCß C2 domain specifically prevents PTP without impairing other PKCß-dependent forms of synaptic enhancement. We conclude that different C2-domain-containing presynaptic proteins are engaged by different Ca(2+) signals, and that Ca(2+) increases evoked by tetanic stimulation are sensed by PKCß to produce PTP.DOI: http://dx.doi.org/10.7554/eLife.03011.001.


Assuntos
Sinalização do Cálcio/fisiologia , Plasticidade Neuronal/fisiologia , Proteína Quinase C beta/metabolismo , Proteína Quinase C-alfa/metabolismo , Animais , Tronco Encefálico/metabolismo , Feminino , Masculino , Memória de Curto Prazo/fisiologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Modelos Neurológicos , Terminações Pré-Sinápticas/metabolismo , Proteína Quinase C beta/deficiência , Proteína Quinase C beta/genética , Proteína Quinase C-alfa/deficiência , Proteína Quinase C-alfa/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
6.
Ann Anat ; 196(5): 365-71, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24862691

RESUMO

Megaesophagus in mice has been associated with several genetic defects. In the present study we expand the range of genes associated with esophageal function and morphology by protein kinase C alpha (PKCα). PKCα-deficient mice showed a six times increased prevalence of megaesophagus at the age of 9-10 weeks compared to wild-type animals. In contrast, in a restricted number of 14-month-old animals of both genotypes a similar prevalence of megaesophagus was found. Megaesophagus was associated with an increased portion of the distal esophagus lined by smooth muscle cells. Achalasia-like degeneration or loss of neuronal cells, inflammation or fibrosis was not present in any of the animals. The results of the study therefore suggest that PKCα expression is associated with a delayed replacement of embryonic smooth muscle by skeletal muscle at the distal esophagus and consecutive megaesophagus in young mice, which, however, is not present at the same prevalence at an advanced age.


Assuntos
Acalasia Esofágica/genética , Acalasia Esofágica/patologia , Esôfago/patologia , Miócitos de Músculo Liso/patologia , Proteína Quinase C-alfa/deficiência , Actinas/metabolismo , Animais , Esfíncter Esofágico Inferior/patologia , Esôfago/crescimento & desenvolvimento , Camundongos , Camundongos Knockout
7.
Am J Physiol Renal Physiol ; 306(3): F309-20, 2014 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-24338818

RESUMO

The epithelial Na channel (ENaC) is negatively regulated by protein kinase C (PKC) as shown using PKC activators in a cell culture model. To determine whether PKCα influences ENaC activity in vivo, we examined the regulation of ENaC in renal tubules from PKCα⁻/⁻ mice. Cortical collecting ducts were dissected and split open, and the exposed principal cells were subjected to cell-attached patch clamp. In the absence of PKCα, the open probability (P0) of ENaC was increased three-fold vs. wild-type SV129 mice (0.52 ± 0.04 vs. 0.17 ± 0.02). The number of channels per patch was also increased. Using confocal microscopy, we observed an increase in membrane localization of α-, ß-, and γ-subunits of ENaC in principal cells in the cortical collecting ducts of PKCα⁻/⁻ mice compared with wild-type mice. To confirm this increase, one kidney from each animal was perfused with biotin, and membrane protein was pulled down with streptavidin. The nonbiotinylated kidney was used to assess total protein. While total ENaC protein did not change in PKCα⁻/⁻ mice, membrane localization of all the ENaC subunits was increased. The increase in membrane ENaC could be explained by the observation that ERK1/2 phosphorylation was decreased in the knockout mice. These results imply a reduction in ENaC membrane accumulation and P0 by PKCα in vivo. The PKC-mediated increase in ENaC activity was associated with an increase in blood pressure in knockout mice fed a high-salt diet.


Assuntos
Canais Epiteliais de Sódio/metabolismo , Túbulos Renais Coletores/citologia , Proteína Quinase C-alfa/deficiência , Aldosterona/sangue , Animais , Aquaporina 2/metabolismo , Pressão Sanguínea/efeitos dos fármacos , Túbulos Renais Coletores/fisiologia , Camundongos , Camundongos Knockout , Microscopia Confocal , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Técnicas de Patch-Clamp , Fosforilação , Proteína Quinase C-alfa/metabolismo , Cloreto de Sódio na Dieta/administração & dosagem
9.
J Biol Chem ; 288(3): 1439-47, 2013 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-23184957

RESUMO

Discovering proteins that modulate Akt signaling has become a critical task, given the oncogenic role of Akt in a wide variety of cancers. We have discovered a novel diacylglycerol signaling pathway that promotes dephosphorylation of Akt. This pathway is regulated by diacylglycerol kinase δ (DGKδ). In DGKδ-deficient cells, we found reduced Akt phosphorylation downstream of three receptor tyrosine kinases. Phosphorylation upstream of Akt was not affected. Our data indicate that PKCα, which is excessively active in DGKδ-deficient cells, promotes dephosphorylation of Akt through pleckstrin homology domain leucine-rich repeats protein phosphatase (PHLPP) 2. Depletion of either PKCα or PHLPP2 rescued Akt phosphorylation in DGKδ-deficient cells. In contrast, depletion of PHLPP1, another Akt phosphatase, failed to rescue Akt phosphorylation. Other PHLPP substrates were not affected by DGKδ deficiency, suggesting mechanisms allowing specific modulation of Akt dephosphorylation. We found that ß-arrestin 1 acted as a scaffold for PHLPP2 and Akt1, providing a mechanism for specificity. Because of its ability to reduce Akt phosphorylation, we tested whether depletion of DGKδ could attenuate tumorigenic properties of cultured cells and found that DGKδ deficiency reduced cell proliferation and migration and enhanced apoptosis. We have, thus, discovered a novel pathway in which diacylglycerol signaling negatively regulates Akt activity. Our collective data indicate that DGKδ is a pertinent cancer target, and our studies could lay the groundwork for development of novel cancer therapeutics.


Assuntos
Diacilglicerol Quinase/metabolismo , Diglicerídeos/metabolismo , Fosfoproteínas Fosfatases/deficiência , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/genética , Arrestinas/genética , Arrestinas/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Transformação Celular Neoplásica , Diacilglicerol Quinase/genética , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Vetores Genéticos , Humanos , Lentivirus/genética , Proteínas Nucleares/deficiência , Proteínas Nucleares/genética , Fosfoproteínas Fosfatases/genética , Fosforilação , Proteína Quinase C-alfa/deficiência , Proteína Quinase C-alfa/genética , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-akt/genética , Especificidade por Substrato , beta-Arrestina 1 , beta-Arrestinas
10.
J Neurosci ; 32(40): 13796-804, 2012 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-23035091

RESUMO

Depolarization of presynaptic terminals that arises from activation of presynaptic ionotropic receptors, or somatic depolarization, can enhance neurotransmitter release; however, the molecular mechanisms mediating this plasticity are not known. Here we investigate the mechanism of this enhancement at the calyx of Held synapse, in which presynaptic glycine receptors depolarize presynaptic terminals, elevate resting calcium levels, and potentiate release. Using knock-out mice of the calcium-sensitive PKC isoforms (PKC(Ca)), we find that enhancement of evoked but not spontaneous synaptic transmission by glycine is mediated primarily by PKC(Ca). Measurements of calcium at the calyx of Held indicate that deficits in synaptic modulation in PKC(Ca) knock-out mice occur downstream of presynaptic calcium increases. Glycine enhances synaptic transmission primarily by increasing the effective size of the pool of readily releasable vesicles. Our results reveal that PKC(Ca) can enhance evoked neurotransmitter release in response to calcium increases caused by small presynaptic depolarizations.


Assuntos
Sinalização do Cálcio/fisiologia , Núcleo Coclear/enzimologia , Glicina/farmacologia , Potenciação de Longa Duração/efeitos dos fármacos , Proteínas do Tecido Nervoso/fisiologia , Proteína Quinase C-alfa/fisiologia , Proteína Quinase C/fisiologia , Sinapses/enzimologia , Animais , Sinalização do Cálcio/efeitos dos fármacos , Núcleo Coclear/fisiologia , Núcleo Coclear/ultraestrutura , Estimulação Elétrica , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Feminino , Masculino , Camundongos , Camundongos Knockout , Terminações Pré-Sinápticas/efeitos dos fármacos , Terminações Pré-Sinápticas/fisiologia , Proteína Quinase C/deficiência , Proteína Quinase C/genética , Proteína Quinase C beta , Proteína Quinase C-alfa/deficiência , Proteína Quinase C-alfa/genética , Estricnina/farmacologia , Sinapses/efeitos dos fármacos , Sinapses/fisiologia
11.
J Neurosci ; 32(38): 13004-9, 2012 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-22993418

RESUMO

Posttetanic potentiation (PTP) is a transient, calcium-dependent increase in the efficacy of synaptic transmission following elevated presynaptic activity. The calcium-dependent protein kinase C (PKC(Ca)) isoforms PKCα and PKCß mediate PTP at the calyx of Held synapse, with PKCß contributing significantly more than PKCα. It is not known whether PKC(Ca) isoforms play a conserved role in PTP at other synapses. We examined this question at the parallel fiber → Purkinje cell (PF→PC) synapse, where PKC inhibitors suppress PTP. We found that PTP is preserved when single PKC(Ca) isoforms are knocked out and in PKCα/ß double knock-out (dko) mice, even though in the latter all PKC(Ca) isoforms are eliminated from granule cells. However, in contrast to wild-type and single knock-out animals, PTP in PKCα/ß dko animals is not suppressed by PKC inhibitors. These results indicate that PKC(Ca) isoforms mediate PTP at the PF→PC synapse in wild-type and single knock-out animals. However, unlike the calyx of Held, at the PF→PC synapse either PKCα or PKCß alone is sufficient to mediate PTP, and if both isoforms are eliminated a compensatory PKC-independent mechanism preserves the plasticity. These results suggest that a feedback mechanism allows granule cells to maintain the normal properties of short-term synaptic plasticity even when the mechanism that mediates PTP in wild-type mice is eliminated.


Assuntos
Adaptação Fisiológica/fisiologia , Cálcio/metabolismo , Cerebelo/citologia , Potenciais Pós-Sinápticos Excitadores/fisiologia , Neurônios/fisiologia , Proteína Quinase C-alfa/metabolismo , Proteína Quinase C/metabolismo , Adaptação Fisiológica/genética , Análise de Variância , Animais , Animais Recém-Nascidos , Biofísica , Estimulação Elétrica , Inibidores Enzimáticos/farmacologia , Potenciais Pós-Sinápticos Excitadores/genética , Feminino , GABAérgicos/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Técnicas In Vitro , Masculino , Camundongos , Camundongos Knockout , Técnicas de Patch-Clamp , Piperidinas/farmacologia , Terminações Pré-Sinápticas/efeitos dos fármacos , Terminações Pré-Sinápticas/fisiologia , Proteína Quinase C/deficiência , Proteína Quinase C beta , Proteína Quinase C-alfa/deficiência , Pirazóis/farmacologia
12.
Dev Biol ; 369(2): 286-97, 2012 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-22819675

RESUMO

During embryonic development tissues remain malleable to participate in morphogenetic movements but on completion of morphogenesis they must acquire the toughness essential for independent adult life. Desmosomes are cell-cell junctions that maintain tissue integrity especially where resistance to mechanical stress is required. Desmosomes in adult tissues are termed hyper-adhesive because they adhere strongly and are experimentally resistant to extracellular calcium chelation. Wounding results in weakening of desmosomal adhesion to a calcium-dependent state, presumably to facilitate cell migration and wound closure. Since desmosomes appear early in mouse tissue development we hypothesised that initial weak adhesion would be followed by acquisition of hyper-adhesion, the opposite of what happens on wounding. We show that epidermal desmosomes are calcium-dependent until embryonic day 12 (E12) and become hyper-adhesive by E14. Similarly, trophectodermal desmosomes change from calcium-dependence to hyper-adhesiveness as blastocyst development proceeds from E3 to E4.5. In both, development of hyper-adhesion is accompanied by the appearance of a midline between the plasma membranes supporting previous evidence that hyper-adhesiveness depends on the organised arrangement of desmosomal cadherins. By contrast, adherens junctions remain calcium-dependent throughout but tight junctions become calcium-independent as desmosomes mature. Using protein kinase C (PKC) activation and PKCα-/- mice, we provide evidence suggesting that conventional PKC isoforms are involved in developmental progression to hyper-adhesiveness. We demonstrate that modulation of desmosomal adhesion by PKC can regulate migration of trophectoderm. It appears that tissue stabilisation is one of several roles played by desmosomes in animal development.


Assuntos
Adesão Celular/fisiologia , Desmossomos/fisiologia , Desenvolvimento Embrionário/fisiologia , Animais , Sequência de Bases , Blastocisto/fisiologia , Blastocisto/ultraestrutura , Cálcio/metabolismo , Movimento Celular/fisiologia , Primers do DNA/genética , Desmossomos/ultraestrutura , Ectoderma/embriologia , Ectoderma/fisiologia , Ectoderma/ultraestrutura , Feminino , Idade Gestacional , Camundongos , Camundongos Knockout , Microscopia Eletrônica de Transmissão , Gravidez , Proteína Quinase C-alfa/deficiência , Proteína Quinase C-alfa/genética , Proteína Quinase C-alfa/fisiologia , Junções Íntimas/fisiologia , Junções Íntimas/ultraestrutura , Trofoblastos/fisiologia , Trofoblastos/ultraestrutura
13.
J Pathol ; 227(3): 346-56, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22407785

RESUMO

Non-healing wounds cause considerable patient morbidity and represent a significant economic burden. Central to wound repair is re-epithelialization, a crucial process involving the modulation of cell adhesion to allow keratinocyte migration to cover the exposed underlying tissues. The cellular mechanisms regulating the earliest stages of re-epithelialization are unclear. We present the first direct evidence that protein kinase Cα (PKCα) plays an important role in regulating wound re-epithelialization. In PKCα(-/-) mice re-epithelialization is delayed, while in novel bitransgenic mice over-expressing constitutively active PKCα it is accelerated. These effects are not due to changes in keratinocyte proliferation, apoptosis or intrinsic cell motility. Instead, they correlate with changes in desmosomal adhesiveness, delay being preceded by retained desmosomal hyper-adhesiveness and acceleration with a rapid switch to desmosomal Ca(2+) -dependence. We demonstrate mechanistic conservation in acute human wounds where PKCα localizes to wound edge desmosomes, which become Ca(2+) -dependent. However, in chronic wounds PKCα remains cytoplasmic and desmosomes fail to switch from the hyper-adhesive state. These results throw new mechanistic light on the earliest stages of wound re-epithelialization and suggest activation of PKCα as a new therapeutic strategy for non-healing wounds.


Assuntos
Adesão Celular , Desmossomos/enzimologia , Queratinócitos/enzimologia , Proteína Quinase C-alfa/metabolismo , Cicatrização , Animais , Apoptose , Cálcio/metabolismo , Adesão Celular/efeitos dos fármacos , Linhagem Celular , Movimento Celular , Proliferação de Células , Desmossomos/efeitos dos fármacos , Desmossomos/patologia , Relação Dose-Resposta a Droga , Ativação Enzimática , Genótipo , Humanos , Queratinócitos/efeitos dos fármacos , Queratinócitos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Fenótipo , Mutação Puntual , Proteína Quinase C-alfa/antagonistas & inibidores , Proteína Quinase C-alfa/deficiência , Proteína Quinase C-alfa/genética , Inibidores de Proteínas Quinases/farmacologia , Transdução de Sinais , Fatores de Tempo , Cicatrização/efeitos dos fármacos
14.
Neuron ; 70(5): 1005-19, 2011 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-21658591

RESUMO

High-frequency stimulation leads to a transient increase in the amplitude of evoked synaptic transmission that is known as posttetanic potentiation (PTP). Here we examine the roles of the calcium-dependent protein kinase C isoforms PKCα and PKCß in PTP at the calyx of Held synapse. In PKCα/ß double knockouts, 80% of PTP is eliminated, whereas basal synaptic properties are unaffected. PKCα and PKCß produce PTP by increasing the size of the readily releasable pool of vesicles evoked by high-frequency stimulation and by increasing the fraction of this pool released by the first stimulus. PKCα and PKCß do not facilitate presynaptic calcium currents. The small PTP remaining in double knockouts is mediated partly by an increase in miniature excitatory postsynaptic current amplitude and partly by a mechanism involving myosin light chain kinase. These experiments establish that PKCα and PKCß are crucial for PTP and suggest that long-lasting presynaptic calcium increases produced by tetanic stimulation may activate these isoforms to produce PTP.


Assuntos
Cálcio/metabolismo , Potenciais Pós-Sinápticos Excitadores/fisiologia , Proteína Quinase C-alfa/metabolismo , Proteína Quinase C/metabolismo , Sinapses/fisiologia , Teto do Mesencéfalo/citologia , Animais , Azepinas/farmacologia , Biofísica , Estimulação Elétrica , Potenciais Pós-Sinápticos Excitadores/genética , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Técnicas de Patch-Clamp , Ésteres de Forbol/farmacologia , Terminações Pré-Sinápticas/fisiologia , Proteína Quinase C/deficiência , Proteína Quinase C beta , Proteína Quinase C-alfa/deficiência , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/genética , Teprotida/farmacologia , Proteína Vesicular 1 de Transporte de Glutamato/metabolismo
15.
Cell Physiol Biochem ; 24(5-6): 627-34, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19910703

RESUMO

BACKGROUND: Progressive loss of podocytes has been documented as an early lesion in the development of glomerular disease. In a variety of glomerular diseases, including diabetic nephropathy the activation of transforming growth factor-beta (TGF-beta) has been demonstrated to promote podocyte death and the development of glomerulosclerosis. In this manuscript we analyzed the role of PKC-alpha (PKCalpha) on TGF-beta1 induced apoptosis in podocytes. METHODS: To accomplish this we generated stable murine PKCalpha deficient podocyte cell lines and examined survival- and pro-apoptotic signaling signatures as well as caspase activation after stimulation with TGF-beta. RESULTS: After stimulation with TGF-beta we can demonstrate an enhanced and prolonged activation of PI3K/AKT and ERK1/2 in PKCalpha-knockout (PKCalpha-/-) podocytes compared to PKCalpha-wildtype (PKCalpha+/ +) podocytes, whereas proapoptotic signaling via p38MAPK is significantly reduced. Interestingly, activation of the Smad-pathway is also prolonged in the PKCalpha-/-podocytes. When we analyzed the underlying mechanisms we found a TGF-beta inducible interaction of PKCalpha with the TGF-beta-type-I-receptor (TGFbetaRI). Moreover, endocytosis assays showed that the TGFbetaRI is less internalized in PKCalpha-/- podocytes. CONCLUSION: Since we can demonstrate a key role for PKCalpha in the signaling response after stimulation with TGF-beta we conclude that PKCalpha might be an interesting target molecule as a "podocyte protective" therapy.


Assuntos
Podócitos/enzimologia , Proteína Quinase C-alfa/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Animais , Apoptose , Linhagem Celular , Endocitose , Técnicas de Inativação de Genes , Camundongos , Camundongos Knockout , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteína Quinase C-alfa/deficiência , Proteína Quinase C-alfa/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Transdução de Sinais , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
16.
Circ Res ; 105(2): 194-200, 2009 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-19556521

RESUMO

Protein kinase (PK)Calpha, PKCbeta, and PKCgamma comprise the conventional PKC isoform subfamily, which is thought to regulate cardiac disease responsiveness. Indeed, mice lacking the gene for PKCalpha show enhanced cardiac contractility and reduced susceptibility to heart failure. Recent data also suggest that inhibition of conventional PKC isoforms with Ro-32-0432 or Ro-31-8220 enhances heart function and antagonizes failure, although the isoform responsible for these effects is unknown. Here, we investigated mice lacking PKCalpha, PKCbeta, and PKCgamma for effects on cardiac contractility and heart failure susceptibility. PKCalpha(-/-) mice, but not PKCbetagamma(-/-) mice, showed increased cardiac contractility, myocyte cellular contractility, Ca(2+) transients, and sarcoplasmic reticulum Ca(2+) load. PKCalpha(-/-) mice were less susceptible to heart failure following long-term pressure-overload stimulation or 4 weeks after myocardial infarction injury, whereas PKCbetagamma(-/-) mice showed more severe failure. Infusion of ruboxistaurin (LY333531), an orally available PKCalpha/beta/gamma inhibitor, increased cardiac contractility in wild-type and PKCbetagamma(-/-) mice, but not in PKCalpha(-/-) mice. More importantly, ruboxistaurin prevented death in wild-type mice throughout 10 weeks of pressure-overload stimulation, reduced ventricular dilation, enhanced ventricular performance, reduced fibrosis, and reduced pulmonary edema comparable to or better than metoprolol treatment. Ruboxistaurin was also administered to PKCbetagamma(-/-) mice subjected to pressure overload, resulting in less death and heart failure, implicating PKCalpha as the primary target of this drug in mitigating heart disease. As an aside, PKCalphabetagamma triple-null mice showed no defect in cardiac hypertrophy following pressure-overload stimulation. In conclusion, PKCalpha functions distinctly from PKCbeta and PKCgamma in regulating cardiac contractility and heart failure, and broad-acting PKC inhibitors such as ruboxistaurin could represent a novel therapeutic approach in treating human heart failure.


Assuntos
Insuficiência Cardíaca/prevenção & controle , Indóis/farmacologia , Maleimidas/farmacologia , Contração Miocárdica/efeitos dos fármacos , Infarto do Miocárdio/tratamento farmacológico , Miocárdio/enzimologia , Proteína Quinase C-alfa/metabolismo , Proteína Quinase C/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Antagonistas Adrenérgicos beta/farmacologia , Animais , Sinalização do Cálcio/efeitos dos fármacos , Cardiomegalia/enzimologia , Cardiomegalia/etiologia , Cardiomegalia/fisiopatologia , Cardiomegalia/prevenção & controle , Modelos Animais de Doenças , Fibrose , Insuficiência Cardíaca/enzimologia , Insuficiência Cardíaca/etiologia , Insuficiência Cardíaca/fisiopatologia , Metoprolol/farmacologia , Camundongos , Camundongos Knockout , Infarto do Miocárdio/complicações , Infarto do Miocárdio/enzimologia , Infarto do Miocárdio/fisiopatologia , Miocárdio/patologia , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/deficiência , Proteína Quinase C/genética , Proteína Quinase C beta , Proteína Quinase C-alfa/antagonistas & inibidores , Proteína Quinase C-alfa/deficiência , Proteína Quinase C-alfa/genética , Edema Pulmonar/enzimologia , Edema Pulmonar/etiologia , Edema Pulmonar/fisiopatologia , Edema Pulmonar/prevenção & controle , Fatores de Tempo
17.
Mol Immunol ; 46(10): 2071-9, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19356803

RESUMO

The physiological roles of PKC alpha and PKC theta were defined in T cell immune functions downstream of the antigen receptor. To investigate the hypothesis that both PKC isotypes may have overlapping functions, we generated mice lacking both genes. We find that PKC alpha(-/-)/theta(-/-) animals have additive T cell response defects in comparison to animals carrying single mutations in these genes. Our studies demonstrate that the activities of PKC alpha and PKC theta converge to regulate both IL-2 cytokine responses and T cell intrinsic alloreactivity in vivo. Mechanistically, this PKC alpha/theta crosstalk primarily affects the NFAT transactivation pathway in T lymphocytes, as observed by decreased phosphorylation of Ser-9 on GSK3 beta, reduced nuclear translocation and DNA binding of NFAT in isolated PKC alpha(-/-)/theta(-/-) CD3(+) T cells. This additive defect proved to be of physiological relevance, because PKC alpha(-/-)/theta(-/-) mice demonstrated significantly prolonged allograft survival in heart transplantation experiments, whereas both PKC alpha(-/-) and PKC theta(-/-) mice showed only minimal graft prolongation when compared to wild type controls. While PKC theta appears to be the rate-limiting PKC isotype mediating T lymphocyte activation, we here provide genetic evidence that PKC alpha and PKC theta have overlapping functions in alloimmunoreactivity in vivo and both PKC theta and PKC alpha isotypes must be targeted to prevent organ allograft rejection.


Assuntos
Isoenzimas/metabolismo , Proteína Quinase C-alfa/metabolismo , Proteína Quinase C/metabolismo , Linfócitos T/enzimologia , Linfócitos T/imunologia , Transferência Adotiva , Animais , Complexo CD3/metabolismo , Morte Celular , Proliferação de Células , Ativação Enzimática , Citometria de Fluxo , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Transplante de Coração , Interleucina-2/metabolismo , Isoenzimas/deficiência , Subpopulações de Linfócitos/enzimologia , Camundongos , Fatores de Transcrição NFATC/metabolismo , Proteína Quinase C/deficiência , Proteína Quinase C-alfa/deficiência , Proteína Quinase C-theta , Linfócitos T/citologia , Ativação Transcricional , Transplante Homólogo
18.
J Cell Biochem ; 105(1): 194-207, 2008 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-18523985

RESUMO

Ultraviolet (UV) irradiation is a major environmental factor responsible for a high incidence of premature skin aging, referred to as photoaging, as well as skin cancer and melanoma. UVA irradiation represents 90% of the solar UV light reaching the earth's surface, and yet the mechanisms by which it exerts its biological effects are not clear. UVA penetrates into the skin tissue, reaching the basal layers of the active dividing cells and, therefore, the contribution of UVA to skin damage may be significant. The majority of UVA energy is absorbed by unidentified photosensitizers in the cells which are postulated to generate reactive oxygen species (ROS). It has been believed that both chronological aging and photoaging share the same molecular features and, as such, it is very common to utilize UV irradiation for induction of skin aging. To determine the involvement of protein kinase isoforms in chronological aging and photoaging, we utilized in vitro aging model systems of primary murine fibroblasts and primary fibroblasts isolated from PKC null mice. We show for the first time distinct involvement of PKC isoforms PKCdelta and PKCalpha in photoaging versus cellular senescence. While chronological aging is accompanied by overexpression and activation of PKCalpha, UV irradiation and ROS production are associated with photoaging accompanied by PKCdelta downregulation and nuclear translocation.


Assuntos
Proteína Quinase C-delta/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos da radiação , Animais , Apoptose/efeitos da radiação , Catalase/metabolismo , Forma Celular , Células Cultivadas , Senescência Celular , Fibroblastos , Isoenzimas/metabolismo , Camundongos , Camundongos Knockout , Proteína Quinase C-alfa/deficiência , Proteína Quinase C-alfa/genética , Proteína Quinase C-alfa/metabolismo , Proteína Quinase C-delta/deficiência , Proteína Quinase C-delta/genética , Transporte Proteico , Superóxido Dismutase/metabolismo
19.
Am J Physiol Heart Circ Physiol ; 293(6): H3768-71, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17921332

RESUMO

Mice null for the gene encoding protein kinase Calpha (Prkca), or mice treated with pharmacologic inhibitors of the PKCalpha/beta/gamma isoforms, show an augmentation in cardiac contractility that appears to be cardioprotective. However, it remains uncertain if PKCalpha itself functions in a myocyte autonomous manner to affect cardioprotection in vivo. Here we generated cardiac myocyte-specific transgenic mice using a tetracycline-inducible system to permit controlled expression of dominant negative PKCalpha in the heart. Consistent with the proposed function of PKCalpha, induction of dominant negative PKCalpha expression in the adult heart enhanced baseline cardiac contractility. This increase in cardiac contractility was associated with a partial protection from long-term decompensation and secondary dilated cardiomyopathy after myocardial infarction injury. Similarly, Prkca null mice were also partially protected from infarction-induced heart failure, although the area of infarction injury was identical to controls. Thus, myocyte autonomous inhibition of PKCalpha protects the adult heart from decompensation and dilated cardiomyopathy after infarction injury in association with a primary enhancement in contractility.


Assuntos
Cardiomegalia/prevenção & controle , Insuficiência Cardíaca/prevenção & controle , Contração Miocárdica , Infarto do Miocárdio/fisiopatologia , Miocárdio/enzimologia , Proteína Quinase C-alfa/metabolismo , Disfunção Ventricular/prevenção & controle , Animais , Cardiomegalia/enzimologia , Cardiomegalia/etiologia , Cardiomegalia/fisiopatologia , Modelos Animais de Doenças , Insuficiência Cardíaca/enzimologia , Insuficiência Cardíaca/etiologia , Insuficiência Cardíaca/fisiopatologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Contração Miocárdica/genética , Infarto do Miocárdio/complicações , Infarto do Miocárdio/enzimologia , Infarto do Miocárdio/patologia , Miocárdio/patologia , Proteína Quinase C-alfa/deficiência , Proteína Quinase C-alfa/genética , Fatores de Tempo , Disfunção Ventricular/enzimologia , Disfunção Ventricular/etiologia , Disfunção Ventricular/fisiopatologia
20.
J Cell Biol ; 177(3): 527-38, 2007 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-17485492

RESUMO

Cell migration in wound healing and disease is critically dependent on integration with the extracellular matrix, but the receptors that couple matrix topography to migratory behavior remain obscure. Using nano-engineered fibronectin surfaces and cell-derived matrices, we identify syndecan-4 as a key signaling receptor determining directional migration. In wild-type fibroblasts, syndecan-4 mediates the matrix-induced protein kinase Calpha (PKCalpha)-dependent activation of Rac1 and localizes Rac1 activity and membrane protrusion to the leading edge of the cell, resulting in persistent migration. In contrast, syndecan-4-null fibroblasts migrate randomly as a result of high delocalized Rac1 activity, whereas cells expressing a syndecan-4 cytodomain mutant deficient in PKCalpha regulation fail to localize active Rac1 to points of matrix engagement and consequently fail to recognize and respond to topographical changes in the matrix.


Assuntos
Movimento Celular , Matriz Extracelular , Fibroblastos/metabolismo , Neuropeptídeos/metabolismo , Proteína Quinase C-alfa/metabolismo , Sindecana-4/metabolismo , Proteínas rac de Ligação ao GTP/metabolismo , Animais , Movimento Celular/genética , Células Cultivadas , Ativação Enzimática/genética , Fibroblastos/citologia , Fibronectinas , Camundongos , Camundongos Knockout , Mutação , Neuropeptídeos/genética , Proteína Quinase C-alfa/deficiência , Estrutura Terciária de Proteína/genética , Sindecana-4/genética , Proteínas rac de Ligação ao GTP/genética , Proteínas rac1 de Ligação ao GTP
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA