Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Clin Immunol ; 238: 109020, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35462050

RESUMO

Proliferation of endothelial cells (EC) and smooth muscle cells (SMC) is a critical process in atherosclerosis. Here, we investigated the involvement of sublytic C5b-9 effector Response Gene to Complement 32 (RGC-32) in cell cycle activation, phenotypic switch, and production of extracellular matrix (ECM) in SMC. Overexpression of RGC-32 augmented C5b-9-induced cell cycle activation and proliferation of SMC in an ERK1-dependent manner and silencing of RGC-32 inhibited C5b-9-induced cell cycle activation. C5b-9-induced cell cycle activation also required phosphorylation of RGC-32 at threonine 91. We found that ECM components fibronectin and collagens I-V were expressed by SMC in human aortic atherosclerotic tissue. Silencing of RGC-32 in cultured SMC was followed by a significant reduction in TGF-ß-induced expression of SMC differentiation markers myocardin, SM22 and α-SMA, and that of collagens I, IV and V. These data suggest that RGC-32 participates in both sublytic C5b-9-induced cell cycle activation and TGF-ß-induced ECM production.


Assuntos
Aterosclerose , Proteínas de Ciclo Celular , Complexo de Ataque à Membrana do Sistema Complemento , Proteínas Musculares , Proteínas do Tecido Nervoso , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Complexo de Ataque à Membrana do Sistema Complemento/metabolismo , Proteínas do Sistema Complemento , Células Endoteliais , Humanos , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Miócitos de Músculo Liso/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Fator de Crescimento Transformador beta
2.
Exp Mol Pathol ; 101(2): 221-230, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27619159

RESUMO

The complement system is an important player in the development of atherosclerosis. Previously reported as a cell cycle regulator, RGC-32 is an essential effector of the terminal complement complex, C5b-9. In this study, our aims were to determine the expression of RGC-32 in the human atherosclerotic arterial wall and to delineate the mechanisms through which RGC-32 affects C5b-9-induced endothelial cell proliferation and migration. We now demonstrate that RGC-32 is expressed in human aortic atherosclerotic wall and that RGC-32 expression increases with the progression of atherosclerosis. Furthermore, silencing of RGC-32 expression abolished C5b-9-induced human aortic endothelial cell (HAEC) proliferation and migration. Of the 279 genes differentially expressed in HAECs after RGC-32 silencing, the genes involved in cell adhesion and cell cycle activation were significantly regulated by RGC-32. RGC-32 silencing caused a significant reduction in the expression of cyclin D1, cyclin D3, Akt, ROCK1, Rho GDP dissociation inhibitor alpha and profilin. These data suggest that RGC-32 mediates HAEC migration through the regulation of RhoA and ROCK1 expression and is involved in actin cytoskeletal organization. Thus, RGC-32 has promising therapeutic potential with regard to angiogenesis and atherosclerosis.


Assuntos
Aorta/patologia , Aterosclerose/patologia , Proteínas de Ciclo Celular/metabolismo , Movimento Celular , Complexo de Ataque à Membrana do Sistema Complemento/metabolismo , Proteínas Musculares/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Idoso , Idoso de 80 Anos ou mais , Aorta/metabolismo , Aterosclerose/genética , Western Blotting , Proliferação de Células , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Feminino , Inativação Gênica , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Mitose , Miócitos de Músculo Liso/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Transcrição Gênica
4.
Proc Natl Acad Sci U S A ; 107(12): 5459-64, 2010 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-20212108

RESUMO

The signaling pathway mediated by JNK transduces different types of signals, such as stress stimuli and cytokines, into functional responses that mediate apoptosis, as well as proliferation, differentiation, and inflammation. To better characterize the dynamic information flow and signal processing of this pathway in the cellular context, a genetically encoded, fluorescent protein-based biosensor was engineered to detect endogenous JNK activity. This biosensor, named JNKAR1 (for JNK activity reporter), specifically detects stress- (ribotoxic and osmotic) and cytokine- (TNF-alpha) induced JNK activity in living cells with a 15 to 30% increase in the yellow-to-cyan emission ratio because of a phosphorylation-dependent increase in FRET between two fluorescent proteins. JNK activity was detected not only in the cytoplasm, but also in the nucleus, mitochondria, and plasma membrane with similar kinetics after induction of ribotoxic stress by anisomycin, suggesting relatively rapid signal propagation to the nuclear, mitochondrial, and plasma membrane compartments. Furthermore, quantitative single-cell analysis revealed that anisomycin-induced JNK activity exhibited ultrasensitivity, sustainability, and bimodality, features that are consistent with behaviors of bistable systems. The development of JNKAR1, therefore, laid a foundation for evaluating the signaling properties and behaviors of the JNK cascade in single living cells.


Assuntos
Técnicas Biossensoriais/métodos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Sequência de Aminoácidos , Transferência Ressonante de Energia de Fluorescência , Corantes Fluorescentes/química , Genes Reporter , Células HeLa , Humanos , Proteínas Luminescentes/química , Proteínas Luminescentes/genética , Sistema de Sinalização das MAP Quinases , Modelos Moleculares , Dados de Sequência Molecular , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Estresse Fisiológico , Frações Subcelulares/enzimologia , Fator de Necrose Tumoral alfa/farmacologia
5.
Exp Mol Pathol ; 88(1): 67-76, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19883641

RESUMO

First described as a cell cycle activator, RGC-32 is both an activator and a substrate for CDC2. Deregulation of RGC-32 expression has been detected in a wide variety of human cancers. We have now shown that RGC-32 is expressed in precancerous states, and its expression is significantly higher in adenomas than in normal colon tissue. The expression of RGC-32 was higher in advanced stages of colon cancer than in precancerous states or the initial stages of colon cancer. In order to identify the genes that are regulated by RGC-32, we used gene array analysis to investigate the effect of RGC-32 knockdown on gene expression in the SW480 colon cancer cell line. Of the 230 genes that were differentially regulated after RGC-32 knockdown, a group of genes involved in chromatin assembly were the most significantly regulated in these cells: RGC-32 knockdown induced an increase in acetylation of histones H2B lysine 5 (H2BK5), H2BK15, H3K9, H3K18, and H4K8. RGC-32 silencing was also associated with decreased expression of SIRT1 and decreased trimethylation of histone H3K27 (H3K27me3). In addition, RGC-32 knockdown caused a significantly higher percentage of SW480 cells to enter S phase and subsequently G2/M. These data suggest that RGC-32 may contribute to the development of colon cancer by regulating chromatin assembly.


Assuntos
Adenocarcinoma/genética , Adenoma/genética , Proteínas de Ciclo Celular/genética , Neoplasias Colorretais/genética , Epigênese Genética , Proteínas Musculares/genética , Proteínas do Tecido Nervoso/genética , Lesões Pré-Cancerosas/genética , Acetilação , Adenocarcinoma/metabolismo , Adenoma/metabolismo , Linhagem Celular Tumoral , Montagem e Desmontagem da Cromatina/genética , Neoplasias Colorretais/metabolismo , Metilação de DNA , Técnica Indireta de Fluorescência para Anticorpo , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Histonas/genética , Histonas/metabolismo , Humanos , Técnicas Imunoenzimáticas , Lesões Pré-Cancerosas/metabolismo , Análise Serial de Tecidos
6.
Exp Mol Pathol ; 86(2): 87-94, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19162005

RESUMO

Proliferation of vascular endothelial cells (EC) and smooth muscle cells (SMC) is a critical event in angiogenesis and atherosclerosis. We previously showed that the C5b-9 assembly during complement activation induces cell cycle in human aortic EC (AEC) and SMC. C5b-9 can induce the expression of Response Gene to Complement (RGC)-32 and over expression of this gene leads to cell cycle activation. Therefore, the present study was carried out to test the requirement of endogenous RGC-32 for the cell cycle activation induced by C5b-9 by knocking-down its expression using siRNA. We identified two RGC-32 siRNAs that can markedly reduce the expression of RGC-32 mRNA in AEC. RGC-32 silencing in these cells abolished DNA synthesis induced by C5b-9 and serum growth factors, indicating the requirement of RGC-32 activity for S-phase entry. RGC-32 siRNA knockdown also significantly reduced the C5b-9 induced CDC2 activation and Akt phosphorylation. CDC2 does not play a role in G1/S transition in HeLa cells stably overexpressing RGC-32. RGC-32 was found to physically associate with Akt and was phosphorylated by Akt in vitro. Mutation of RGC-32 protein at Ser 45 and Ser 47 prevented Akt mediated phosphorylation. In addition, RGC-32 was found to regulate the release of growth factors from AEC. All these data together suggest that cell cycle induction by C5b-9 in AEC is RGC-32 dependent and this is in part through regulation of Akt and growth factor release.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Ciclo Celular/efeitos dos fármacos , Complexo de Ataque à Membrana do Sistema Complemento/farmacologia , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Proteínas Musculares/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Adulto , Indutores da Angiogênese/metabolismo , Proteína Quinase CDC2/metabolismo , Proteínas de Ciclo Celular/genética , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Células Endoteliais/enzimologia , Ativação Enzimática/efeitos dos fármacos , Células HeLa , Humanos , Proteínas de Membrana/metabolismo , Proteínas Musculares/genética , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/metabolismo , Fosforilação/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo
7.
Arch Immunol Ther Exp (Warsz) ; 56(2): 115-22, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18373239

RESUMO

The role of response gene to complement (RGC)-32 as a cell cycle regulator has been attributed to its ability to activate cdc2 kinases and to induce S-phase entry and mitosis. However, recent studies revealed novel functions for RGC-32 in diverse processes such as cellular differentiation, inflammation, and fibrosis. Besides responding to C5b-9 stimulation, RGC-32 expression is also induced by growth factors, hormones, and cytokines. Transforming growth factor beta activates RGC-32 through Smad and RhoA signaling, thus initiating smooth muscle cell differentiation. Accumulating evidence has drawn attention to the deregulated expression of RGC-32 in human malignancies, hyper-immunoglobulin E syndrome, and fibrosis. RCG-32 expression is up-regulated in cutaneous T cell lymphoma and colon, ovarian, and breast cancer, but down-regulated in invasive prostate cancer, multiple myeloma, and drug-resistant glioblastoma. A better understanding of the mechanism by which RGC-32 contributes to the pathogenesis of these diseases will provide new insights into its therapeutic potential. In this review we provide an overview of this field and discuss the most recent research on RGC-32.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Proteínas Musculares/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Animais , Proteínas de Ciclo Celular/genética , Diferenciação Celular , Proliferação de Células , Corticosterona/farmacologia , Estradiol/farmacologia , Fibrose , Humanos , Imunidade , Proteínas Musculares/genética , Neoplasias/etiologia , Proteínas do Tecido Nervoso/genética , Regeneração
8.
J Biol Chem ; 282(50): 36634-41, 2007 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-17928291

RESUMO

Activation of the serine/threonine protein kinase Akt/PKB is a multi-step process involving membrane recruitment, phosphorylation, and membrane detachment. To investigate this process in the cellular context, we employed a live-cell fluorescence imaging approach to examine conformational changes of Akt and its membrane association. A fluorescence resonance energy transfer-based reporter of Akt action (ReAktion) reveals a conformational change that is critically dependent on the existence of a phosphorylatable threonine 308 in the activation loop, because mutations to either aspartate or alanine abolished the change. Furthermore, a mutant carrying a phosphorylation mimic at this position showed diminished membrane association, suggesting that this phosphorylation plays an important role of promoting the dissociation of activated Akt from the membrane. In addition, the membrane-associating pleckstrin homology domain was found to associate with the catalytic domain when Thr308 is phosphorylated, suggesting such an interdomain interaction as a mechanism by which phosphorylation within the catalytic domain can affect membrane association. These studies uncover new regulatory roles of this critical phosphorylation event of Akt for ensuring its proper activation and function.


Assuntos
Membrana Celular/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Membrana Celular/genética , Ativação Enzimática/fisiologia , Transferência Ressonante de Energia de Fluorescência , Humanos , Camundongos , Microscopia de Fluorescência , Mutação , Células NIH 3T3 , Fosforilação , Estrutura Terciária de Proteína/fisiologia , Proteínas Proto-Oncogênicas c-akt/genética
9.
J Biol Chem ; 281(28): 19009-18, 2006 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-16670089

RESUMO

Migration and proliferation of aortic endothelial cells (AEC) are critical processes involved in angiogenesis, atherosclerosis, and postangioplasty restenosis. Activation of complement and assembly of the C5b-9 complement complex have been implicated in the pre-lesional stage of atherogenesis and progression of the atherosclerotic lesion. We have shown that C5b-9 induces proliferation and activates phosphatidylinositol 3-kinase (PI3K), but it is unknown whether this can lead to activation of Akt in AEC, a major downstream target of PI3K, or if C5b-9 can induce the migration of AEC, a critical step in angiogenesis. In this study, we show that C5b-9 induces AEC proliferation and migration and also activates the PI3K/Akt pathway. C5b-9 activates Akt as shown by in vitro kinase assay and phosphorylation of Ser-473. C5b-9-induced cell cycle activation was inhibited by pretreatment with LY294002 (PI3K inhibitor), SH-5 (Akt inhibitor), or transfection with Akt siRNA. These data suggests that the PI3K/Akt pathway is required for C5b-9-induced cell cycle activation. FOXO1, a member of forkhead transcription factor family, was phosphorylated at Ser-256 and inactivated after C5b-9 stimulation as shown by a decrease in DNA binding and cytoplasmic relocalization. Cytoplasmic relocalization was significantly reduced after pretreatment with LY294002, SH-5, or transfection with Akt siRNA. Silencing FOXO1 expression using siRNA stimulated AEC proliferation and regulated angiogenic factor release. Our data indicate that C5b-9 regulation of the cell cycle activation in AEC through Akt pathway is dependent on inactivation of FOXO1.


Assuntos
Aorta/citologia , Complexo de Ataque à Membrana do Sistema Complemento/fisiologia , Endotélio Vascular/citologia , Fatores de Transcrição Forkhead/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Angioplastia , Aterosclerose/patologia , Movimento Celular , Proliferação de Células , Progressão da Doença , Células Endoteliais/metabolismo , Proteína Forkhead Box O1 , Humanos , Neovascularização Patológica
10.
J Peripher Nerv Syst ; 11(1): 77-87, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16519786

RESUMO

The protein zero (P0) glycoprotein is an important component of compact peripheral nerve myelin produced by the glial cells of the mammalian peripheral nervous system. P0 mRNA expression is reduced following exposure of Schwann cells to sublytic C5b-9, the terminal activation complex of the complement cascade. Sublytic complement treatment decreased P0 mRNA by 81% within 6 h and required C5b-9 assembly. C5b-9 induced a threefold increase in both JNK1 activity and c-jun mRNA within 20 and 30 min, respectively, compared with cells treated with either human serum depleted of complement component C7 (C7dHS) or medium alone. Sublytic C5b-9 stimulation, in the presence of the transcription inhibitor Actinomycin D, decreased P0 mRNA expression by 52%, indicating that mRNA was selectively destabilized. This effect was prevented by pretreatment with L-JNK inhibitor 1 (L-JNKI1). To study a potential inhibition of P0 gene transcription, we transfected Schwann cells with a P0 promoter-firefly luciferase construct. Sublytic C5b-9 stimulation of the transfected cells decreased luciferase activity by 82% at 6 h, and this effect was prevented by pretreatment with L-JNKI1 inhibitor. Our results indicate that the ability of C5b-9 in vitro to affect P0 gene expression is mediated via JNK1 activation that leads to enhanced mRNA decay and transcriptional repression of P0.


Assuntos
Complexo de Ataque à Membrana do Sistema Complemento/metabolismo , Ativação Enzimática/fisiologia , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Proteína P0 da Mielina/metabolismo , Células de Schwann/metabolismo , Animais , Northern Blotting , Complexo de Ataque à Membrana do Sistema Complemento/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Expressão Gênica/fisiologia , Proteína Quinase 8 Ativada por Mitógeno/efeitos dos fármacos , Proteína P0 da Mielina/efeitos dos fármacos , Proteína P0 da Mielina/genética , Estabilidade de RNA/fisiologia , RNA Mensageiro , Ratos , Ratos Sprague-Dawley , Células de Schwann/efeitos dos fármacos , Transcrição Gênica
11.
J Immunol ; 176(5): 3173-80, 2006 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-16493077

RESUMO

Activation of the terminal complement cascade involving C5 to C9 proteins has a beneficial role for oligodendrocytes (OLG) in experimental allergic encephalomyelitis, an animal model of multiple sclerosis, by protecting them from apoptotic cell death. We have previously shown that sublytic C5b-9 complexes, through posttranslational regulation of Bad, inhibit the mitochondrial pathway of apoptosis induced by serum deprivation. In the present study, we examined the possible involvement of the caspase-8 and Fas pathway in OLG apoptosis and the role of C5b-9 in this process. In a serum-free defined medium, OLG undergo apoptosis and differentiation concomitantly. Under this condition, we found that caspase-8 processing was increased in association with Bid cleavage and markedly reduced expression of cellular FLIP long isoform protein. The caspase-8 inhibitor Z-IETD-FMK inhibited cell death associated with differentiation in a dose-dependent manner. Exposure to C5b-9 induced an inhibition of caspase-8 activation, Bid cleavage, and a significant increase in expression of cellular FLIP long isoform. These C5b-9 effects were reversed by PI3K inhibitor LY294002. C5b-9 also down-regulated the expression of FasL and the Fas-induced apoptosis. These data suggest that C5b-9 through PI3K signaling can rescue OLG from Fas-mediated apoptosis by regulating caspase-8 processing.


Assuntos
Apoptose/imunologia , Inibidores de Caspase , Caspases/metabolismo , Complexo de Ataque à Membrana do Sistema Complemento/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Oligodendroglia/imunologia , Processamento de Proteína Pós-Traducional/imunologia , Regulação para Cima/imunologia , Animais , Animais Recém-Nascidos , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3/antagonistas & inibidores , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3/metabolismo , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD , Caspase 8 , Células Cultivadas , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte , Regulação para Baixo/fisiologia , Proteína Ligante Fas , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Glicoproteínas de Membrana/biossíntese , Glicoproteínas de Membrana/genética , Oligodendroglia/citologia , Oligodendroglia/enzimologia , Inibidores de Fosfoinositídeo-3 Quinase , Ratos , Ratos Sprague-Dawley , Peptídeos e Proteínas Associados a Receptores de Fatores de Necrose Tumoral/metabolismo , Fatores de Necrose Tumoral/biossíntese , Fatores de Necrose Tumoral/genética
12.
J Biol Chem ; 281(11): 7118-28, 2006 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-16407259

RESUMO

RUNX2 is a member of the runt family of DNA-binding transcription factors. RUNX2 mediates endothelial cell migration and invasion during tumor angiogenesis and is expressed in metastatic breast and prostate tumors. Our published studies showed that RUNX2 DNA-binding activity is low during growth arrest, but elevated in proliferating endothelial cells. To investigate its role in cell proliferation and cell cycle regulation, RUNX2 was depleted in human bone marrow endothelial cells using RNA interference. Specific RUNX2 depletion inhibited DNA-binding activity as measured by electrophoretic mobility shift assay resulting in inhibition of cell proliferation. Cells were synchronized at the G(1)/S boundary with excess thymidine or in mitosis (M phase) with nocodazole. Endogenous or ectopic RUNX2 activity was maximal at late G(2) and during M phase. Inhibition of RUNX2 expression by RNA interference delayed entry into and exit out of the G(2)/M phases of the cell cycle. RUNX2 was coimmunoprecipitated with cyclin B1 in mitotic cells, which further supported a role for RUNX2 in cell cycle progression. Moreover, in vitro kinase assays using recombinant cdc2 kinase showed that RUNX2 was phosphorylated at Ser(451). The cdc2 inhibitor roscovitine dose dependently inhibited in vivo RUNX2 DNA-binding activity during mitosis and the RUNX2 mutant S451A exhibited lower DNA-binding activity and reduced stimulation of anchorage-independent growth relative to wild type RUNX2. These results suggest for the first time that RUNX2 phosphorylation by cdc2 may facilitate cell cycle progression possibly through regulation of G(2) and M phases, thus promoting endothelial cell proliferation required for tumor angiogenesis.


Assuntos
Proteína Quinase CDC2/metabolismo , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Células Endoteliais/metabolismo , Regulação Neoplásica da Expressão Gênica , Fatores de Transcrição/metabolismo , Animais , Sequência de Bases , Western Blotting , Células da Medula Óssea/metabolismo , Ciclo Celular , Divisão Celular , Linhagem Celular , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Proliferação de Células , Separação Celular , Ciclina B/metabolismo , Ciclina B1 , DNA/metabolismo , Regulação para Baixo , Endotélio Vascular/metabolismo , Epitopos , Citometria de Fluxo , Fase G1 , Fase G2 , Humanos , Imunoprecipitação , Camundongos , Mitose , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Células NIH 3T3 , Metástase Neoplásica , Fosforilação , Ligação Proteica , Interferência de RNA , Fase S , Timidina/farmacologia , Fatores de Tempo , Transfecção
13.
Exp Mol Pathol ; 78(2): 116-22, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15713436

RESUMO

Tumors often exhibit deregulation of the cell cycle and overexpression of cyclins and cyclin-dependent kinases (CDKs). Response gene to complement (RGC)-32 is a substrate and regulator of CDC2 and its overexpression induces cell cycle activation. We investigated RGC-32 mRNA and protein expression in tumors with special emphasis in colon carcinoma. By using an expression array technique we found that 19% of tumor tissues showed increased RGC-32 mRNA expression over the levels of corresponding normal tissues. On the other hand, an increased RGC-32 protein was found in 70% of colon adenocarcinoma samples tested. In colon carcinomas, two major patterns of RGC-32 immunoreactivity were seen: staining of malignant epithelial cells only in some tumors and RGC-32 reactivity of both malignant epithelia as well as cells in the interstitium in others. Colonic epithelium obtained from normal individuals was consistently negative for RGC-32 protein. Overexpression of RGC-32 protein was found in other tumors including prostate, bladder, breast, lung, and other digestive tract tumors. RGC-32 expression was present in the same malignant epithelial cells that also expressed the proliferation marker Ki-67. Our data suggest that RGC-32 overexpression might be part of the deregulation of the cell cycle that is required for the growth of tumor cells.


Assuntos
Biomarcadores Tumorais/análise , Proteínas de Ciclo Celular/biossíntese , Neoplasias do Colo/metabolismo , Proteínas Musculares/biossíntese , Neoplasias/metabolismo , Proteínas do Tecido Nervoso/biossíntese , Adenocarcinoma/metabolismo , Northern Blotting , Western Blotting , Carcinoma/metabolismo , Linhagem Celular Tumoral , Humanos , Imuno-Histoquímica , Imunoprecipitação , Antígeno Ki-67/metabolismo , Reação em Cadeia da Polimerase , RNA Mensageiro/análise , Regulação para Cima
14.
Immunol Res ; 31(1): 37-46, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15591621

RESUMO

Activation of the complement system plays an important role in innate and acquired immunity. Activation of complement and subsequent formation of C5b-9 channels on the surface of cellular membranes leads to cell lysis. When the number of channels assembled on the surface of nucleated cells is limited, C5b-9 does not cause lysis, but instead can induce cell-cycle progression by activating signal transduction pathways, transcription factors, and key components of the cell-cycle machinery. Cell-cycle induction by C5b-9 is dependent on the activation of phosphatidylinositol 3-kinase and the ERK1 pathway in a Gi protein-dependent manner. Cell-cycle activation is regulated, in part, by activation of proto-oncogene c-jun and AP1 DNA binding activity. C5b-9 induces sequential activation of CDK4 and CDK2, leading to G1/S-phase transition and cellular proliferation. RGC-32 is a novel gene whose expression is induced by C5b-9. RGC-32 may play a key role in cell-cycle activation by increasing cyclin B1-CDC2 activity. C5b-9-mediated cell-cycle activation plays an important role in cellular proliferation and protection from apoptosis.


Assuntos
Ciclo Celular/fisiologia , Complexo de Ataque à Membrana do Sistema Complemento/fisiologia , Transcrição Gênica/fisiologia , Animais , Regulação da Expressão Gênica/fisiologia , Humanos , Proto-Oncogene Mas , Proto-Oncogenes/fisiologia , Transdução de Sinais/fisiologia
15.
J Biol Chem ; 277(1): 502-8, 2002 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-11687586

RESUMO

Proliferation of aortic smooth muscle cells contributes to atherogenesis and neointima formation. Sublytic activation of complement, particularly C5b-9, induces cell cycle progression in aortic smooth muscle cells. RGC-32 is a novel protein that may promote cell cycle progression in response to complement activation. We cloned human RGC-32 cDNA from a human fetal brain cDNA library. The human RGC-32 cDNA encodes a 117-amino acid protein with 92% similarity to the rat and mouse protein. Human RGC-32 maps to chromosome 13 and is expressed in most tissues. Sublytic complement activation enhanced RGC-32 mRNA expression in human aortic smooth muscle cells and induced nuclear translocation of the protein. RGC-32 was physically associated with cyclin-dependent kinase p34CDC2 and increased the kinase activity in vivo and in vitro. In addition, RGC-32 was phosphorylated by p34CDC2-cyclin B1 in vitro. Mutation of RGC-32 protein at Thr-91 prevented the p34CDC2-mediated phosphorylation and resulted in loss of p34CDC2 kinase enhancing activity. Overexpression of RGC-32 induced quiescent aortic smooth muscle cells to enter S-phase. These data indicate that cell cycle activation by C5b-9 may involve p34CDC2 activity through RGC-32. RGC-32 appears to be a cell cycle regulatory factor that mediates cell proliferation, both as an activator and substrate of p34CDC2.


Assuntos
Aorta/citologia , Proteína Quinase CDC2/biossíntese , Proteínas de Ciclo Celular , Proteínas Musculares , Músculo Liso Vascular/citologia , Proteínas do Tecido Nervoso/fisiologia , Transporte Ativo do Núcleo Celular , Sequência de Aminoácidos , Animais , Sequência de Bases , Mapeamento Cromossômico , Clonagem Molecular , Proteínas do Sistema Complemento/fisiologia , Humanos , Camundongos , Dados de Sequência Molecular , Proteínas do Tecido Nervoso/genética , Fosforilação , RNA Mensageiro/análise , Fase S
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...