Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Oncogene ; 36(30): 4370-4378, 2017 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-28368426

RESUMO

Recent studies suggest that the presence of a KRAS mutation may be insufficient for defining a clinically homogenous molecular group, as many KRAS mutant tumors lose reliance on K-Ras for survival. Identifying pathways that support K-Ras dependency may define clinically relevant KRAS subgroups and lead to the identification of new drug targets. We have analyzed a panel of 17 KRAS mutant lung cancer cell lines classified as K-Ras-dependent or -independent for co-dependency on protein kinase C δ (PKCδ). We show that functional dependency on K-Ras and PKCδ co-segregate, and that dependency correlates with a more epithelial-like phenotype. Furthermore, we show that the pro-apoptotic and pro-tumorigenic functions of PKCδ also segregate based on K-Ras dependency, as K-Ras-independent cells are more sensitive to topoisomerase inhibitors, and depletion of PKCδ in this subgroup suppresses apoptosis through increased activation of extracellular signal-regulated kinase (ERK). In contrast, K-Ras-dependent lung cancer cells are largely insensitive to topoisomerase inhibitors, and depletion of PKCδ can increase apoptosis and decrease activation of ERK in this subgroup. We have previously shown that nuclear translocation of PKCδ is necessary and sufficient for pro-apoptotic signaling. Our current studies show that K-Ras-dependent cells are refractive to PKCδ-driven apoptosis. Analysis of this subgroup showed increased PKCδ expression and an increase in the nuclear:cytoplasmic ratio of PKCδ. In addition, targeting PKCδ to the nucleus induces apoptosis in K-Ras-independent, but not K-Ras-dependent non-small-cell lung cancer (NSCLC) cells. Our studies provide tools for identification of the subset of patients with KRAS mutant tumors most amenable to targeting of the K-Ras pathway, and identify PKCδ as a potential target in this tumor population. These subgroups are likely to be of clinical relevance, as high PKCδ expression correlates with increased overall survival and a more epithelial tumor phenotype in patients with KRAS mutant lung adenocarcinomas.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Proteína Quinase C-delta/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Fragmentação do DNA , Resistencia a Medicamentos Antineoplásicos , Técnicas de Silenciamento de Genes , Humanos , Immunoblotting , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Mutação , Proteínas Proto-Oncogênicas p21(ras)/genética , Reação em Cadeia da Polimerase em Tempo Real
2.
Oncogenesis ; 6(1): e287, 2017 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-28092369

RESUMO

A subset of clear cell renal cell carcinoma (ccRCC) tumors exhibit a HIF1A gene mutation, yielding two ccRCC tumor types, H1H2 type expressing both HIF1α and HIF2α, and H2 type expressing HIF2α, but not functional HIF1α protein. However, it is unclear how the H1H2 type ccRCC tumors escape HIF1's tumor-suppressive activity. The polybromo-1 (PBRM1) gene coding for the BAF180 protein, a component of the SWItch/Sucrose Non-Fermentable (SWI/SNF) chromatin remodeling complex, is inactivated in 40% ccRCCs, the function and mechanism of BAF180 mutation is unknown. Our previous study indicates that BAF180-containing SWI/SNF chromatin remodeling complex is a co-activator for transcription factor HIF to induce HIF target genes. Thus, our questions are if BAF180 is involved in HIF-mediated hypoxia response and if PBRM1/BAF180 mutation has any association with the HIF1A retention in H1H2 type ccRCC. We report here that BAF180 is mutated in H1H2 ccRCC cell lines and tumors, and BAF180 re-expression in H1H2 ccRCC cell lines reduced cell proliferation/survival, indicating that BAF180 has tumor-suppressive role in these cells. However, BAF180 is expressed in HIF1-deficient H2 ccRCC cell lines and tumors, and BAF180 knockdown in H2 type ccRCC cell lines reduced cell proliferation/survival, indicating that BAF180 has tumor-promoting activity in these cells. In addition, our data show that BAF180 functions as co-activator for HIF1- and HIF2-mediated transcriptional response, and BAF180's tumor-suppressive and -promoting activity in ccRCC cell lines depends on co-expression of HIF1 and HIF2, respectively. Thus, our studies reveal that BAF180 function in ccRCC is context dependent, and that mutation of PBRM1/BAF180 serves as an alternative strategy for ccRCC tumors to reduce HIF1 tumor-suppressive activity in H1H2 ccRCC tumors. Our studies define distinct functional subgroups of ccRCCs based on expression of BAF180, and suggest that BAF180 inhibition may be a novel therapeutic target for patients with H2, but not H1H2, ccRCC tumors.

3.
Oncogene ; 33(10): 1306-15, 2014 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-23474764

RESUMO

Protein kinase C δ (PKCδ) regulates apoptosis in the mammary gland, however, the functional contribution of PKCδ to the development or progression of breast cancer has yet to be determined. Meta-analysis of ErbB2-positive breast cancers shows increased PKCδ expression, and a negative correlation between PKCδ expression and prognosis. Here, we present in-vivo evidence that PKCδ is essential for the development of mammary gland tumors in a ErbB2-overexpressing transgenic mouse model, and in-vitro evidence that PKCδ is required for proliferative signaling downstream of the ErbB2 receptor. Mouse mammary tumor virus (MMTV)-ErbB2 mice lacking PKCδ (δKO) have increased tumor latency compared with MMTV-ErbB2 wild-type (δWT) mice, and the tumors show a dramatic decrease in Ki-67 staining. To explore the relationship between PKCδ and ErbB2-driven proliferation more directly, we used MCF-10A cells engineered to express a synthetic ligand-inducible form of the ErbB2 receptor. Depletion of PKCδ with short hairpin RNA inhibited ligand-induced growth in both two-dimensional (2D) (plastic) and three-dimensional (3D) (Matrigel) culture, and correlated with decreased phosphorylation of the ErbB2 receptor and reduced activation of Src and MAPK/ERK pathways. Similarly, in human breast cancer cell lines in which ErbB2 is overexpressed, depletion of PKCδ suppresses proliferation, Src and ERK activation. PKCδ appears to drive proliferation through the formation of an active ErbB2/PKCδ/Src signaling complex, as depletion of PKCδ disrupts association of Src with the ErbB2 receptor. Taken together, our studies present the first evidence that PKCδ is a critical regulator of ErbB2-mediated tumorigenesis, and suggest further investigation of PKCδ as a target in ErbB2-positive breast cancer.


Assuntos
Neoplasias da Mama/enzimologia , Carcinogênese/metabolismo , Neoplasias Mamárias Experimentais/enzimologia , Proteína Quinase C-delta/fisiologia , Receptor ErbB-2/fisiologia , Animais , Neoplasias da Mama/mortalidade , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proliferação de Células , Intervalo Livre de Doença , Feminino , Humanos , Estimativa de Kaplan-Meier , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Knockout , Prognóstico , Transdução de Sinais
4.
Cell Death Dis ; 4: e520, 2013 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-23449456

RESUMO

During atherogenesis, excess amounts of low-density lipoproteins (LDL) accumulate in the subendothelial space where they undergo oxidative modifications. Oxidized LDL (oxLDL) alter the fragile balance between survival and death of vascular smooth muscle cells (VSMC) thereby leading to plaque instability and finally to atherothrombotic events. As protein kinase C δ (PKCδ) is pro-apoptotic in many cell types, we investigated its potential role in the regulation of VSMC apoptosis induced by oxLDL. We found that human VSMC silenced for PKCδ exhibited a protection towards oxLDL-induced apoptosis. OxLDL triggered the activation of PKCδ as shown by its phosphorylation and nuclear translocation. PKCδ activation was dependent on the reactive oxygen species generated by oxLDL. Moreover, we demonstrated that PKCδ participates in oxLDL-induced endoplasmic reticulum (ER) stress-dependent apoptotic signaling mainly through the IRE1α/JNK pathway. Finally, the role of PKCδ in the development of atherosclerosis was supported by immunohistological analyses showing the colocalization of activated PKCδ with ER stress and lipid peroxidation markers in human atherosclerotic lesions. These findings highlight a role for PKCδ as a key regulator of oxLDL-induced ER stress-mediated apoptosis in VSMC, which may contribute to atherosclerotic plaque instability and rupture.


Assuntos
Apoptose , Estresse do Retículo Endoplasmático , Lipoproteínas LDL/metabolismo , Músculo Liso Vascular/metabolismo , Proteína Quinase C-delta/metabolismo , Animais , Núcleo Celular/metabolismo , Células Cultivadas , Endorribonucleases/metabolismo , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Camundongos , Músculo Liso Vascular/citologia , Fosforilação , Proteína Quinase C-delta/antagonistas & inibidores , Proteína Quinase C-delta/genética , Proteínas Serina-Treonina Quinases/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais
5.
Oral Dis ; 17(6): 601-9, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21702866

RESUMO

OBJECTIVES: Chronic infiltration of lymphocytes into the salivary and lacrimal glands of patients with Sjögren's syndrome (SS) leads to destruction of acinar cells and loss of exocrine function. Protein kinase C-delta (PKCδ) is known to play a critical role in B-cell maintenance. Mice in which the PKCδ gene has been disrupted have a loss of B-cell tolerance, multiple organ lymphocytic infiltration, and altered apoptosis. To determine whether PKCδ contributes to the pathogenesis of SS, we quantified changes in indicators of SS in PKCδ-/- mice as a function of age. Salivary gland histology, function, the presence of autoantibodies, and cytokine expression were examined. MATERIALS AND METHODS: Submandibular glands were examined for the presence of lymphocytic infiltrates, and the type of infiltrating lymphocyte and cytokine deposition was evaluated by immunohistochemistry. Serum samples were tested by autoantibody screening, which was graded by its staining pattern and intensity. Salivary gland function was determined by saliva collection at various ages. RESULTS: PKCδ-/- mice have reduced salivary gland function, B220+ B-cell infiltration, anti-nuclear antibody production, and elevated IFN-γ in the salivary glands as compared to PKCδ+/+ littermates. CONCLUSIONS: PKCδ-/- mice have exocrine gland tissue damage indicative of a SS-like phenotype.


Assuntos
Proteína Quinase C-delta/imunologia , Síndrome de Sjogren/imunologia , Doenças da Glândula Submandibular/imunologia , Animais , Anticorpos Antinucleares/análise , Apoptose/genética , Autoanticorpos/análise , Autoanticorpos/sangue , Linfócitos B/imunologia , Movimento Celular/imunologia , Proliferação de Células , Modelos Animais de Doenças , Feminino , Centro Germinativo/patologia , Interferon gama/análise , Interleucina-4/análise , Antígeno Ki-67/análise , Antígenos Comuns de Leucócito/análise , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Knockout , Proteína Quinase C-delta/genética , Ductos Salivares/imunologia , Ductos Salivares/patologia , Taxa Secretória/fisiologia , Tolerância a Antígenos Próprios/imunologia , Glândula Submandibular/metabolismo , Glândula Submandibular/patologia , Doenças da Glândula Submandibular/fisiopatologia
6.
Neuroscience ; 170(4): 1065-79, 2010 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-20691763

RESUMO

PKC-theta (PKC-θ), a member of the novel protein kinase C family (nPKC), regulates a wide variety of functions in the periphery. However, its presence and role in the CNS has remained largely unknown. Recently, we demonstrated the presence of PKC-θ in the arcuate hypothalamic nucleus (ARC) and knockdown of PKC-θ from the ARC protected mice from developing diet-induced obesity. Another isoform of the nPKC group, PKC-delta (PKC-δ), is expressed in several non-hypothalamic brain sites including the thalamus and hippocampus. Although PKC-δ has been implicated in regulating hypothalamic glucose homeostasis, its distribution in the hypothalamus has not previously been described. In the current study, we used immunohistochemistry to examine the distribution of PKC-θ and -δ immunoreactivity in rat and mouse hypothalamus. We found PKC-θ immunoreactive neurons in several hypothalamic nuclei including the ARC, lateral hypothalamic area, perifornical area and tuberomammillary nucleus. PKC-δ immunoreactive neurons were found in the paraventricular and supraoptic nuclei. Double-label immunohistochemisty in mice expressing green fluorescent protein either with the long form of leptin receptor (LepR-b) or in orexin (ORX) neurons indicated that PKC-θ is highly colocalized in lateral hypothalamic ORX neurons but not in lateral hypothalamic LepR-b neurons. Double-label immunohistochemistry in oxytocin-enhanced yellow fluorescent protein mice or arginine vasopressin-enhanced green fluorescent protein (AVP-EGFP) transgenic rats revealed a high degree of colocalization of PKC-δ within paraventricular and supraoptic oxytocin neurons but not the vasopressinergic neurons. We conclude that PKC-θ and -δ are expressed in different hypothalamic neuronal populations.


Assuntos
Hipotálamo/enzimologia , Isoenzimas/metabolismo , Proteína Quinase C-delta/metabolismo , Proteína Quinase C/metabolismo , Animais , Arginina Vasopressina/metabolismo , Histidina Descarboxilase/metabolismo , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/metabolismo , Ocitocina/metabolismo , Proteína Quinase C-theta , Ratos , Ratos Long-Evans , Receptores para Leptina/metabolismo
7.
Cell Death Dis ; 1: e17, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-21364618

RESUMO

As apoptotic pathways are commonly deregulated in breast cancer, exploring how mammary gland cell death is regulated is critical for understanding human disease. We show that primary mammary epithelial cells from protein kinase C delta (PKCδ) -/- mice have a suppressed response to apoptotic agents in vitro. In the mammary gland in vivo, apoptosis is critical for ductal morphogenesis during puberty and involution following lactation. We have explored mammary gland development in the PKCδ -/- mouse during these two critical windows. Branching morphogenesis was altered in 4- to 6-week-old PKCδ -/- mice as indicated by reduced ductal branching; however, apoptosis and proliferation in the terminal end buds was unaltered. Conversely, activation of caspase-3 during involution was delayed in PKCδ -/- mice, but involution proceeded normally. The thymus also undergoes apoptosis in response to physiological signals. A dramatic suppression of caspase-3 activation was observed in the thymus of PKCδ -/- mice treated with irradiation, but not mice treated with dexamethasone, suggesting that there are both target- and tissue-dependent differences in the execution of apoptotic pathways in vivo. These findings highlight a role for PKCδ in both apoptotic and nonapoptotic processes in the mammary gland and underscore the redundancy of apoptotic pathways in vivo.


Assuntos
Apoptose , Glândulas Mamárias Animais/crescimento & desenvolvimento , Proteína Quinase C-delta/fisiologia , Animais , Caspase 3/metabolismo , Células Cultivadas , Dexametasona/farmacologia , Feminino , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/metabolismo , Camundongos , Camundongos Knockout , Proteína Quinase C-delta/genética , Proteína Quinase C-delta/metabolismo , Timo/metabolismo , Timo/efeitos da radiação
8.
Oncogene ; 27(21): 3045-53, 2008 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-18059334

RESUMO

PKCdelta is essential for apoptosis, but regulation of the proapoptotic function of this ubiquitous kinase is not well understood. Nuclear translocation of PKCdelta is necessary and sufficient to induce apoptosis and is mediated via a C-terminal bipartite nuclear localization sequence. However, PKCdelta is found predominantly in the cytoplasm of nonapoptotic cells, and the apoptotic signal that activates its nuclear translocation is not known. We show that in salivary epithelial cells, phosphorylation at specific tyrosine residues in the N-terminal regulatory domain directs PKCdelta to the nucleus where it induces apoptosis. Analysis of each tyrosine residue in PKCdelta by site-directed mutagenesis identified two residues, Y64 and Y155, as essential for nuclear translocation. Suppression of apoptosis correlated with suppressed nuclear localization of the Y --> F mutant proteins. Moreover, a phosphomimetic PKCdelta Y64D/Y155D mutant accumulated in the nucleus in the absence of an apoptotic signal. Forced nuclear accumulation of PKCdelta-Y64F and Y155F mutant proteins, by attachment of an SV40 nuclear localization sequence, fully reconstituted their ability to induce apoptosis, indicating that tyrosine phosphorylation per se is not required for apoptosis, but for targeting PKCdelta to the nucleus. We propose that phosphorylation/dephosphorylation of PKCdelta in the regulatory domain functions as a switch to promote cell survival or cell death.


Assuntos
Núcleo Celular/metabolismo , Proteína Quinase C-delta/metabolismo , Tirosina/metabolismo , Animais , Sequência de Bases , Linhagem Celular , Núcleo Celular/enzimologia , Primers do DNA , Marcação In Situ das Extremidades Cortadas , Camundongos , Mutagênese Sítio-Dirigida , Fosforilação , Proteína Quinase C-delta/química , Proteína Quinase C-delta/genética , Transporte Proteico
9.
Biochem Soc Trans ; 35(Pt 5): 1001-4, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17956263

RESUMO

The PKC (protein kinase C) family regulates diverse cellular functions and specific isoforms have been shown to be critical regulators of cell proliferation and survival. In particular, PKCdelta is known to be a critical pro-apoptotic signal in many cell types. Work in our laboratory has focused on understanding the molecular mechanisms through which PKCdelta regulates apoptosis and on how the pro-apoptotic activity of this ubiquitous kinase is regulated such that cells only activate the apoptotic cascade when appropriate. We have identified multiple regulatory steps that activate the pro-apoptotic function of PKCdelta in response to genotoxins. Our studies show that apoptotic signals induce rapid post-translational modification of PKCdelta in the regulatory domain, which facilitates translocation of the kinase from the cytoplasm to the nucleus. Active caspase 3 also accumulates in the nucleus under these conditions, resulting in caspase cleavage of PKCdelta and generation of a constitutively activated form of PKCdelta [deltaCF (PKCdelta catalytic fragment)]. In contrast with PKCdelta, deltaCF is constitutively present in the nucleus, and this nuclear accumulation of PKCdelta is essential for apoptosis. Thus our studies suggest that tight regulation of nuclear import and of PKCdelta is critical for cell survival and that caspase cleavage of PKCdelta in the nucleus signals an irreversible commitment to apoptosis.


Assuntos
Apoptose , Proteína Quinase C-delta/metabolismo , Caspases/metabolismo , Domínio Catalítico , Núcleo Celular/metabolismo , Ativação Enzimática , Homeostase , Fosforilação , Proteína Quinase C-delta/química , Processamento de Proteína Pós-Traducional , Transdução de Sinais , Tirosina/metabolismo
10.
J Mol Endocrinol ; 32(2): 507-17, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15072555

RESUMO

The steroidogenic acute regulatory (StAR) protein promotes intramitochondrial delivery of cholesterol to the cholesterol side-chain cleavage system, which catalyzes the first enzymatic step in all steroid synthesis. Intriguingly, substrate cholesterol derived from lipoprotein can upregulate StAR gene expression. Moreover, substrate oxysterols have been suggested to also play a role. To investigate whether oxysterols can regulate StAR expression, two steroidogenic cell lines, mouse Y1 adrenocortical and MA-10 Leydig tumor cells, were treated with various oxysterols and steroids, including 25-hydroxycholesterol (25 OHC), 22(R)OHC and 20alphaOHC. The majority of these compounds rapidly increased StAR protein levels within as little as 1 h. The most potent oxysterols were 20alphaOHC for Y1 and 25 OHC for MA-10 cells. After 8 h, StAR mRNA abundance also increased whereas there were no detected changes in promoter activity. Thus, in contrast to lipoprotein, oxysterols acutely increase StAR protein levels independently of mRNA abundance, and later increase mRNA levels independently of new gene transcription. Therefore, we propose that oxysterols modulate steroidogenesis at two levels. First, oxysterols may be important in post-transcriptional regulation of StAR activity and production of steroids for paracrine action. Secondly, through direct conversion to steroid, oxysterols may account in part for StAR-independent steroid production in the body.


Assuntos
Fosfoproteínas/metabolismo , Esteróis/metabolismo , Neoplasias do Córtex Suprarrenal/tratamento farmacológico , Neoplasias do Córtex Suprarrenal/metabolismo , Animais , Regulação da Expressão Gênica/efeitos dos fármacos , Hidroxicolesteróis/metabolismo , Hidroxicolesteróis/farmacologia , Cetocolesteróis/metabolismo , Cetocolesteróis/farmacologia , Tumor de Células de Leydig , Camundongos , Fosfoproteínas/efeitos dos fármacos , Fosfoproteínas/genética , Regiões Promotoras Genéticas , Esteróis/farmacologia , Transcrição Gênica , Células Tumorais Cultivadas
11.
Cell Death Differ ; 10(3): 269-77, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12700627

RESUMO

We have used expression of a kinase dead mutant of PKCalpha (PKCalphaKD) to explore the role of this isoform in salivary epithelial cell apoptosis. Expression of PKCalphaKD by adenovirus-mediated transduction results in a dose-dependent induction of apoptosis in salivary epithelial cells as measured by the accumulation of sub-G1 DNA, activation of caspase-3, and cleavage of PKCdelta and PKCzeta, known caspase substrates. Induction of apoptosis is accompanied by nine-fold activation of c-Jun-N-terminal kinase, and an approximately two to three-fold increase in activated mitogen-activated protein kinase (MAPK) as well as total MAPK protein. Previous studies from our laboratory have shown that PKCdelta activity is essential for the apoptotic response of salivary epithelial cells to a variety of cell toxins. To explore the contribution of PKCdelta to PKCalphaKD-induced apoptosis, salivary epithelial cells were cotransduced with PKCalphaKD and PKCdeltaKD expression vectors. Inhibition of endogenous PKCdelta blocked the ability of PKCalphaKD to induce apoptosis as indicated by cell morphology, DNA fragmentation, and caspase-3 activation, indicating that PKCdelta activity is required for the apoptotic program induced under conditions where PKCalpha is inhibited. These findings indicate that PKCalpha functions as a survival factor in salivary epithelial cells, while PKCdelta functions to regulate entry into the apoptotic pathway.


Assuntos
Apoptose , Células Epiteliais/metabolismo , Proteína Quinase C/fisiologia , Glândulas Salivares/metabolismo , Adenoviridae/genética , Animais , Caspase 3 , Caspases/metabolismo , Linhagem Celular , Separação Celular , Células Cultivadas , DNA/metabolismo , Relação Dose-Resposta a Droga , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Citometria de Fluxo , Fase G1 , Glutationa Transferase/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Glândula Parótida/citologia , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , Proteína Quinase C-alfa , Proteína Quinase C-delta , Ratos , Fatores de Tempo
12.
Exp Neurol ; 170(2): 258-69, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11476591

RESUMO

Transplantation of fetal ventral mesencephalic (VM) tissue shows great promise as an experimental therapy for patients with Parkinson's disease. However, cell survival in brain tissue grafts is poor, with survival rates of only 5-15%. We have utilized a combination of the caspase inhibitor bocaspartyl (OMe)-fluoromethylketone (BOC-ASP-CH2F) and glial cell line-derived neurotrophic factor (GDNF) to enhance survival of grafted dopamine neurons. The VM tissue was dissected from embryonic day 13-15 rat fetuses, incubated in different doses of BOC-ASP-CH2F and GDNF, and transplanted to the anterior chamber of the eye of adult rats. Growth of the tissue was assessed through the translucent cornea. Doses of 50 and 100 micromolar of the general caspase inhibitor appeared to have detrimental effects on mesencephalic tissue, while 20 micromolar had beneficial effects on overall transplant growth. A combination of the caspase inhibitor and GDNF appeared to have more prominent effects on cell survival as well as dopaminergic fiber density than either agent by itself. The transplants doubled in size when they were treated with a combination of BOC-ASP-CH2F and GDNF, and cell death markers were significantly reduced at both 48 h and 4-6 days postgrafting. This is, to our knowledge, the first combined approach using apoptotic blockers with trophic factors, and demonstrates a viable strategy for protection of developing neurons, since several different aspects of graft function may be addressed simultaneously.


Assuntos
Ácido Aspártico/análogos & derivados , Ácido Aspártico/farmacologia , Transplante de Tecido Encefálico/fisiologia , Sobrevivência de Enxerto/efeitos dos fármacos , Fatores de Crescimento Neural , Proteínas do Tecido Nervoso/farmacologia , Neurônios/citologia , Fármacos Neuroprotetores , Substância Negra/citologia , Substância Negra/transplante , Animais , Apoptose/efeitos dos fármacos , Inibidores de Caspase , Dopamina/fisiologia , Inibidores Enzimáticos/farmacologia , Transplante de Tecido Fetal/fisiologia , Feto , Idade Gestacional , Fator Neurotrófico Derivado de Linhagem de Célula Glial , Sobrevivência de Enxerto/fisiologia , Marcação In Situ das Extremidades Cortadas , Neurônios/fisiologia , Ratos , Ratos Endogâmicos F344 , Fatores de Tempo , Transplante Heterotópico , Tirosina 3-Mono-Oxigenase/análise
13.
J Biol Chem ; 276(32): 29719-28, 2001 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-11369761

RESUMO

We report here that the novel protein kinase C isoform, PKCdelta, is required at or prior to the level of the mitochondria for apoptosis induced by a diverse group of cell toxins. We have used adenoviral expression of a kinase-dead (KD) mutant of PKCdelta to explore the requirement for PKCdelta in the mitochondrial-dependent apoptotic pathway. Expression of PKCdeltaKD, but not PKCalphaKD, in salivary epithelial cells resulted in a dose-dependent inhibition of apoptosis induced by etoposide, UV-irradiation, brefeldin A, and paclitaxel. DNA fragmentation was blocked up to 71% in parotid C5 cells infected with the PKCdeltaKD adenovirus, whereas caspase-3 activity was inhibited up to 65%. The activation of caspase-9-like proteases by all agents was also inhibited in parotid C5 cells expressing PKCdeltaKD. The ability of PKCdeltaKD to block the loss of mitochondrial membrane potential was similarly determined. Expression of PKCdeltaKD blocked the decrease in mitochondrial membrane potential observed in cells treated with etoposide, UV, brefeldin A, or paclitaxel in a dose-dependent manner. In contrast to the protective function of PKCdeltaKD, expression of PKCdeltaWT resulted in a potent induction of apoptosis, which could be inhibited by co-infection with PKCdeltaKD. These results suggest that PKCdelta is a common intermediate in mitochondrial-dependent apoptosis in salivary epithelial cells.


Assuntos
Apoptose , Células Epiteliais/metabolismo , Isoenzimas/química , Isoenzimas/fisiologia , Mitocôndrias/metabolismo , Proteína Quinase C/química , Proteína Quinase C/fisiologia , Glândulas Salivares/metabolismo , Adenoviridae/genética , Animais , Western Blotting , Brefeldina A/farmacologia , Caspase 3 , Caspase 9 , Caspases/metabolismo , Linhagem Celular , Separação Celular , Fragmentação do DNA , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Etoposídeo/farmacologia , Citometria de Fluxo , Genes Dominantes , Óperon Lac , Mutação , Inibidores da Síntese de Ácido Nucleico/farmacologia , Paclitaxel/farmacologia , Isoformas de Proteínas , Proteína Quinase C-delta , Inibidores da Síntese de Proteínas/farmacologia , Radiossensibilizantes/farmacologia , Ratos , Fatores de Tempo , Raios Ultravioleta
14.
J Biol Chem ; 275(51): 40620-7, 2000 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-11016947

RESUMO

Atypical protein kinase C zeta (PKCzeta) is known to transduce signals that influence cell proliferation and survival. Here we show that recombinant human caspases can process PKCzeta at three sites in the hinge region between the regulatory and catalytic domains. Caspase-3, -6, -7, and -8 chiefly cleaved human PKCzeta at EETD downward arrowG, and caspase-3 and -7 also cleaved PKCzeta at DGMD downward arrowG and DSED downward arrowL, respectively. Processing of PKCzeta expressed in transfected cells occurred chiefly at EETD downward arrowG and DGMD downward arrowG and produced carboxyl-terminal polypeptides that contained the catalytic domain. Epitope-tagged PKCzeta that lacked the regulatory domain was catalytically active following expression in HeLa cells. Induction of apoptosis in HeLa cells by tumor necrosis factor alpha plus cycloheximide evoked the conversion of full-length epitope-tagged PKCzeta to two catalytic domain polypeptides and increased PKCzeta activity. A caspase inhibitor, zVAD-fmk, prevented epitope-tagged PKCzeta processing and activation following the induction of apoptosis. Induction of apoptosis in rat parotid C5 cells produced catalytic domain polypeptides of endogenous PKCzeta and increased PKCzeta activity. Caspase inhibitors prevented the increase in PKCzeta activity and production of the catalytic domain polypeptides. Treatment with lactacystin, a selective inhibitor of the proteasome, caused polyubiquitin-PKCzeta conjugates to accumulate in cells transfected with the catalytic domain or full-length PKCzeta, or with a PKCzeta mutant that was resistant to caspase processing. We conclude that caspases process PKCzeta to carboxyl-terminal fragments that are catalytically active and that are degraded by the ubiquitin-proteasome pathway.


Assuntos
Caspases/metabolismo , Cisteína Endopeptidases/metabolismo , Complexos Multienzimáticos/metabolismo , Proteína Quinase C/metabolismo , Ubiquitinas/metabolismo , Animais , Apoptose/efeitos dos fármacos , Sequência de Bases , Domínio Catalítico , Linhagem Celular , Cricetinae , Cicloeximida/farmacologia , Primers do DNA , Ativação Enzimática , Células HeLa , Humanos , Hidrólise , Complexo de Endopeptidases do Proteassoma , Proteína Quinase C/química , Processamento de Proteína Pós-Traducional , Ratos , Radioisótopos de Enxofre , Fator de Necrose Tumoral alfa/farmacologia
15.
J Biol Chem ; 275(47): 36637-44, 2000 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-10960482

RESUMO

The steroidogenic acute regulatory protein (StAR) is required for the movement of cholesterol from the outer to the inner mitochondrial membrane, the site of cholesterol side chain cleavage. Here we describe a novel form of regulation of StAR gene expression in steroidogenic cells. Treatment of Y-1 BS1 adrenocortical cells with either low density lipoprotein (LDL) or high density lipoprotein (HDL) increases expression of endogenous StAR mRNA and protein in a dose-dependent manner. Induction of StAR mRNA by lipoprotein requires basal cAMP-dependent protein kinase, since the inhibitor, R(p)-8-Br-cAMP, inhibited induction of StAR protein by LDL. Likewise, basal StAR expression or LDL induction of StAR protein was not detectable in Y-1 kin-8 cells which are deficient in cAMP-dependent protein kinase. Aminoglutethimide and ketoconazole were used to determine if side chain cleavage of lipoprotein-derived cholesterol is required for induction of StAR mRNA. Treatment with either drug alone induced StAR mRNA expression 1.5-3-fold, while induction of StAR in cells treated with either drug plus LDL, was equal to, or greater than, induction seen with either agent alone, suggesting that lipoprotein does not regulate StAR via generation of an oxysterol intermediate. Both LDL and HDL increased expression of a mouse -966 StAR promoter-reporter construct 1.5-2.5-fold, indicating that regulation occurs at the level of transcription. In contrast, neither lipoprotein was able to induce transcription from a -966 StAR promoter in which the steroidogenic factor-1 site at -135 was abolished, indicating that regulation of StAR transcription by lipoproteins requires steroidogenic factor-1. The regulation of StAR gene expression by lipoproteins may represent a positive feedback circuit which links cholesterol availability with steroidogenic output.


Assuntos
Córtex Suprarrenal/metabolismo , Regulação da Expressão Gênica , Lipoproteínas/fisiologia , Fosfoproteínas/genética , Aminoglutetimida/farmacologia , Animais , Células Cultivadas , Enzima de Clivagem da Cadeia Lateral do Colesterol/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Eletroforese em Gel de Poliacrilamida , Cetoconazol/farmacologia , Lipoproteínas HDL/metabolismo , Lipoproteínas LDL/metabolismo , Camundongos , Fosfoproteínas/biossíntese , RNA Mensageiro/metabolismo
16.
Am J Physiol Lung Cell Mol Physiol ; 279(3): L429-38, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10956616

RESUMO

Individual protein kinase C (PKC) isozymes have been implicated in many cellular responses important in lung health and disease, including permeability, contraction, migration, hypertrophy, proliferation, apoptosis, and secretion. New ideas on mechanisms that regulate PKC activity, including the identification of a novel PKC kinase, 3-phosphoinositide-dependent kinase-1 (PDK-1), that regulates phosphorylation of PKC, have been advanced. The importance of targeted translocation of PKC and isozyme-specific binding proteins (like receptors for activated C-kinase and caveolins) is well established. Phosphorylation state and localization are now thought to be key determinants of isozyme activity and specificity. New concepts on the role of individual PKC isozymes in proliferation and apoptosis are emerging. Opposing roles for selected isozymes in the same cell system have been defined. Coupling to the Wnt signaling pathway has been described. Phenotypes for PKC knockout mice have recently been reported. More specific approaches for studying PKC isozymes and their role in cell responses have been developed. Strengths and weaknesses of different experimental strategies are reviewed. Future directions for investigation are identified.


Assuntos
Isoenzimas/fisiologia , Pulmão/fisiologia , Proteína Quinase C/fisiologia , Animais , Apoptose/fisiologia , Divisão Celular/fisiologia , Humanos , Isquemia/patologia , Isquemia/fisiopatologia , Pulmão/citologia , Circulação Pulmonar
17.
Cell Death Differ ; 7(12): 1200-9, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11175257

RESUMO

Accumulating evidence suggests that specific isoforms of PKC may function to promote apoptosis. We show here that activation of the conventional and novel isoforms of PKC with 12-O-tetradecanoyl phorbol-13- ester (TPA) induces apoptosis in salivary acinar cells as indicated by DNA fragmentation and activation of caspase-3. TPA-induced DNA fragmentation, caspase-3 activation, and morphologic indicators of apoptosis, can be enhanced by pretreatment of cells with the calpain inhibitor, calpeptin, prior to the addition of TPA. Analysis of PKC isoform expression by immunoblot shows that TPA-induced downregulation of PKC alpha and PKC delta is delayed in cells pre-treated with calpeptin, and that this correlates with an increase of these isoforms in the membrane fraction of cells. TPA-induced apoptosis is accompanied by biphasic activation of the c-jun-N-terminal kinase (JNK) pathway and inactivation of the extracellular regulated kinase (ERK) pathway. Expression of constitutively activated PKC alpha or PKC delta, but not kinase negative mutants of these isoforms, or constitutively activated PKC epsilon, induces apoptosis in salivary acinar cells, suggesting a role for these isoforms in TPA-induced apoptosis. These studies demonstrate that activation of PKC is sufficient for initiation of an apoptotic program in salivary acinar cells. Cell Death and Differentiation (2000) 7, 1200 - 1209.


Assuntos
Apoptose/fisiologia , Proteína Quinase C/metabolismo , Glândulas Salivares/enzimologia , Animais , Apoptose/efeitos dos fármacos , Carcinógenos/farmacologia , Caspase 3 , Caspases/efeitos dos fármacos , Caspases/metabolismo , Células Cultivadas , Inibidores de Cisteína Proteinase/farmacologia , Fragmentação do DNA/efeitos dos fármacos , Fragmentação do DNA/fisiologia , Dipeptídeos/farmacologia , Humanos , Proteína Quinase 8 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/efeitos dos fármacos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Glândula Parótida/citologia , Glândula Parótida/efeitos dos fármacos , Glândula Parótida/enzimologia , Isoformas de Proteínas/efeitos dos fármacos , Isoformas de Proteínas/metabolismo , Proteína Quinase C/efeitos dos fármacos , Proteína Quinase C/genética , Glândulas Salivares/citologia , Glândulas Salivares/efeitos dos fármacos , Acetato de Tetradecanoilforbol/farmacologia
18.
Am J Respir Cell Mol Biol ; 22(1): 15-25, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10615061

RESUMO

Enhanced proliferation of adventitial fibroblasts is a major contributor to the structural remodeling of the pulmonary artery (PA) that occurs during hypoxia-induced pulmonary hypertension. The mechanisms responsible for the exuberant growth of fibroblasts are unknown; however, protein kinase C (PKC) isozymes have previously been shown to be important in the enhanced growth properties of immature PA fibroblasts. We tested the hypotheses that PA adventitial fibroblasts from neonatal calves exposed chronically to hypoxia after birth would express augmented growth responses compared with fibroblasts from the control adventitia and that these properties would be associated with selective changes in expression of PKC isozymes. We studied the effects of serum, purified mitogens, and hypoxia on the growth of aggregate populations of fibroblasts isolated from the PA of neonatal control calves (Neo-C) and calves chronically exposed to hypoxia for 2 wk beginning on Day 1 of life (Neo-Hyp). Neo-Hyp fibroblasts demonstrated higher proliferative capabilities than did Neo-C cells in response to all the stimuli tested. Importantly, hypoxia was found to act synergistically with peptide mitogens (platelet-derived growth factor, basic fibroblast growth factor, insulin-like growth factor-I) to stimulate growth in Neo-Hyp but not in Neo-C cells. Using PKC-isozyme nonselective and selective inhibitors and immunoblot analysis, we found differences in utilization of PKC isozymes in Neo-Hyp and Neo-C fibroblasts and have identified PKC-betaI and -zeta as key contributors to the augmented growth of Neo-Hyp fibroblasts. Although the activity of PKC-betaI and -zeta isozymes was increased by hypoxia in serum-deprived Neo-C and Neo-Hyp fibroblasts, under normoxia, quiescent Neo-Hyp fibroblasts had higher PKC-zeta-specific activity than did Neo-C cells. These results suggest that neonatal PA adventitial fibroblasts acquire new growth properties in the setting of hypoxia- induced pulmonary hypertension and that the augmented proliferative characteristics of the Neo-Hyp fibroblasts might be associated with changes in specifc PKC isozyme expression and activation patterns.


Assuntos
Fibroblastos/enzimologia , Fibroblastos/patologia , Hipóxia/enzimologia , Hipóxia/patologia , Proteína Quinase C/fisiologia , Artéria Pulmonar/enzimologia , Artéria Pulmonar/patologia , Animais , Animais Recém-Nascidos , Bovinos , Divisão Celular , Células Cultivadas , Doença Crônica , Inibidores Enzimáticos/farmacologia , Hipertensão Pulmonar/enzimologia , Hipertensão Pulmonar/patologia , Hipertensão Pulmonar/fisiopatologia , Indóis/farmacologia , Isoenzimas/antagonistas & inibidores , Isoenzimas/metabolismo , Isoenzimas/fisiologia , Maleimidas/farmacologia , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , Proteína Quinase C beta , Artéria Pulmonar/fisiopatologia
19.
J Lipid Res ; 40(8): 1440-52, 1999 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10428980

RESUMO

SW-13 adrenal tumor cells that lack detectable intermediate filaments (IF-free) exhibit an impaired capacity to esterify lipoprotein-derived cholesterol compared with cells that contain vimentin filaments. IF-free cells were found to synthesize and secrete significant amounts of apoE, while apoE secretion was nearly undetectable in cell lines that spontaneously express vimentin. However, stable transfectants that express a mouse vimentin cDNA exhibited elevated levels of cholesterol esterification and apoE secretion compared with untransfected IF-free cells, indicating that apoE secretion is not directly related to the capacity of these cells to esterify cholesterol. Some of the cell lines that differed in the level of apoE synthesis and secretion had similar levels of apoE mRNA, suggesting that the differences in expression involve a post-transcriptional mechanism. Treatment of these cells with forskolin and IBMX, 8br-cAMP, or TPA had no effect on apoE secretion. The level of sterol carrier protein-2 (SCP(2)) synthesis and the distribution of SCP(2) between membrane and soluble cellular fractions was not observably different in cells that contained or lacked vimentin. SW-13 cell lines contained little or no detectable caveolin-1 or caveolin-2. These studies demonstrate that the difference in the capacity of these adrenal tumor cells that contain or lack vimentin filaments to esterify low density lipoprotein-cholesterol is not obviously associated with the level of expression or distribution of apoE, SCP(2), or caveolins.


Assuntos
Neoplasias das Glândulas Suprarrenais/metabolismo , Caveolinas , Ésteres do Colesterol/biossíntese , LDL-Colesterol/metabolismo , Filamentos Intermediários/metabolismo , Proteínas de Plantas , Vimentina/metabolismo , 1-Metil-3-Isobutilxantina/farmacologia , 8-Bromo Monofosfato de Adenosina Cíclica/farmacologia , Animais , Apolipoproteínas E/metabolismo , Proteínas de Transporte/metabolismo , Caveolina 1 , Caveolina 2 , Compartimento Celular , Esterificação , Imunofluorescência , Regulação Neoplásica da Expressão Gênica , Humanos , Proteínas de Membrana/metabolismo , Camundongos , Proteínas Quinases/efeitos dos fármacos , Acetato de Tetradecanoilforbol , Células Tumorais Cultivadas
20.
J Biol Chem ; 274(27): 19115-23, 1999 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-10383415

RESUMO

We have previously shown that parotid C5 salivary acinar cells undergo apoptosis in response to etoposide treatment as indicated by alterations in cell morphology, caspase-3 activation, DNA fragmentation, sustained activation of c-Jun N-terminal kinase, and inactivation of extracellular regulated kinases 1 and 2. Here we report that apoptosis results in the caspase-dependent cleavage of protein kinase C-delta (PKCdelta) to a 40-kDa fragment, the appearance of which correlates with a 9-fold increase in PKCdelta activity. To understand the function of activated PKCdelta in apoptosis, we have used the PKCdelta-specific inhibitor, rottlerin. Pretreatment of parotid C5 cells with rottlerin prior to the addition of etoposide blocks the appearance of the apoptotic morphology, the sustained activation of c-Jun N-terminal kinase, and inactivation of extracellular regulated kinases 1 and 2. Inhibition of PKCdelta also partially inhibits caspase-3 activation and DNA fragmentation. Immunoblot analysis shows that the PKCdelta cleavage product does not accumulate in parotid C5 cells treated with rottlerin and etoposide together, suggesting that the catalytic activity of PKCdelta may be required for cleavage. PKCalpha and PKCbeta1 activities also increase during etoposide-induced apoptosis. Inhibition of these two isoforms with Gö6976 slightly suppresses the apoptotic morphology, caspase-3 activation, and DNA fragmentation, but has no effect on the sustained activation of c-Jun N-terminal kinase or inactivation of extracellular regulated kinase 1 and 2. These data demonstrate that activation of PKCdelta is an integral and essential part of the apoptotic program in parotid C5 cells and that specific activated isoforms of PKC may have distinct functions in cell death.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Apoptose/efeitos dos fármacos , Etoposídeo/farmacologia , Isoenzimas/metabolismo , Proteína Quinase C/metabolismo , Glândulas Salivares/enzimologia , Acetofenonas/farmacologia , Benzopiranos/farmacologia , Proteínas Quinases Dependentes de Cálcio-Calmodulina/antagonistas & inibidores , Caspase 3 , Caspases/metabolismo , Células Cultivadas , Fragmentação do DNA , Eletroforese em Gel de Poliacrilamida , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Humanos , Proteína Quinase C beta , Proteína Quinase C-delta , Glândulas Salivares/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...