Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Blood ; 127(11): 1438-48, 2016 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-26729899

RESUMO

Aberrant expression of the oncogenic transcription factor forkhead box protein 1 (FOXP1) is a common feature of diffuse large B-cell lymphoma (DLBCL). We have combined chromatin immunoprecipitation and gene expression profiling after FOXP1 depletion with functional screening to identify targets of FOXP1 contributing to tumor cell survival. We find that the sphingosine-1-phosphate receptor 2 (S1PR2) is repressed by FOXP1 in activated B-cell (ABC) and germinal center B-cell (GCB) DLBCL cell lines with aberrantly high FOXP1 levels; S1PR2 expression is further inversely correlated with FOXP1 expression in 3 patient cohorts. Ectopic expression of wild-type S1PR2, but not a point mutant incapable of activating downstream signaling pathways, induces apoptosis in DLBCL cells and restricts tumor growth in subcutaneous and orthotopic models of the disease. The proapoptotic effects of S1PR2 are phenocopied by ectopic expression of the small G protein Gα13 but are independent of AKT signaling. We further show that low S1PR2 expression is a strong negative prognosticator of patient survival, alone and especially in combination with high FOXP1 expression. The S1PR2 locus has previously been demonstrated to be recurrently mutated in GCB DLBCL; the transcriptional silencing of S1PR2 by FOXP1 represents an alternative mechanism leading to inactivation of this important hematopoietic tumor suppressor.


Assuntos
Fatores de Transcrição Forkhead/fisiologia , Linfoma Difuso de Grandes Células B/patologia , Proteínas de Neoplasias/fisiologia , Receptores de Lisoesfingolipídeo/fisiologia , Proteínas Repressoras/fisiologia , Transdução de Sinais/fisiologia , Animais , Apoptose/fisiologia , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Fatores de Transcrição Forkhead/genética , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/biossíntese , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/genética , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Centro Germinativo/patologia , Xenoenxertos , Humanos , Estimativa de Kaplan-Meier , Linfoma Difuso de Grandes Células B/classificação , Linfoma Difuso de Grandes Células B/genética , Linfoma Difuso de Grandes Células B/mortalidade , Camundongos , Transplante de Neoplasias , Prognóstico , Proteínas Proto-Oncogênicas c-akt/análise , Interferência de RNA , RNA Interferente Pequeno/genética , Receptores de Lisoesfingolipídeo/biossíntese , Receptores de Lisoesfingolipídeo/deficiência , Receptores de Lisoesfingolipídeo/genética , Proteínas Repressoras/genética , Receptores de Esfingosina-1-Fosfato
2.
Oncotarget ; 6(22): 19055-69, 2015 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-25965999

RESUMO

MicroRNA-122 (miR-122) is implicated as a regulator of physiological and pathophysiological processes in the liver. Overexpression of Gα12 is associated with overall survival in patients with hepatocellular carcinoma (HCC). Array-based miRNA profiling was performed on Huh7 stably transfected with activated Gα12 to find miRNAs regulated by the Gα12 pathway; among them, miR-122 was most greatly repressed. miR-122 directly inhibits c-Met expression, playing a role in HCC progression. Gα12 destabilized HNF4α by accelerating ubiquitination, impeding constitutive expression of miR-122. miR-122 mimic transfection diminished the ability of Gα12 to increase c-Met and to activate ERK, STAT3, and Akt/mTOR, suppressing cell proliferation with augmented apoptosis. Consistently, miR-122 transfection prohibited tumor cell colony formation and endothelial tube formation. In a xenograft model, Gα12 knockdown attenuated c-Met expression by restoring HNF4α levels, and elicited tumor cell apoptosis but diminished Ki67 intensities. In human HCC samples, Gα12 levels correlated to c-Met and were inversely associated with miR-122. Both miR-122 and c-Met expression significantly changed in tumor node metastasis (TNM) stage II/III tumors. Moreover, changes in Gα12 and miR-122 levels discriminated recurrence-free and overall survival rates of HCC patients. Collectively, Gα12 overexpression in HCC inhibits MIR122 transactivation by inactivating HNF4α, which causes c-Met induction, contributing to cancer aggressiveness.


Assuntos
Carcinoma Hepatocelular/genética , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/biossíntese , Fator 4 Nuclear de Hepatócito/metabolismo , Neoplasias Hepáticas/genética , MicroRNAs/biossíntese , Proteínas Proto-Oncogênicas c-met/biossíntese , Animais , Carcinoma Hepatocelular/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/genética , Indução Enzimática , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/genética , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/metabolismo , Células Hep G2 , Fator 4 Nuclear de Hepatócito/genética , Xenoenxertos , Humanos , Neoplasias Hepáticas/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , MicroRNAs/genética , Proteínas Proto-Oncogênicas c-met/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-met/genética , Proteínas Proto-Oncogênicas c-met/metabolismo , Transdução de Sinais , Transfecção
3.
Cell Signal ; 26(7): 1456-65, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24631529

RESUMO

The high mortality rate of hepatocellular carcinoma (HCC) is associated with its fast-growing malignancy. In tumor microenvironments, certain GPCRs are coupled to Gα12 for signal transduction. Given the role of forkhead box O1 (FOXO1) in the inhibition of various tumors, this study investigated whether increase of Gα12 in HCC causes FOXO1 repression, and if so, whether this event occurs through microRNA dysregulation. Overexpression of an active mutant of Gα12 (Gα12QL) decreased FOXO1 levels, whereas knockdown of Gα12 had the opposite effect. Of the microRNAs targeting FOXO1, miR-135b levels were markedly increased by Gα12 signaling, which led to FOXO1 repression as shown by the experiments using mimic, antisense oligonucleotide or siRNA. Gα12QL increased the primary form of miR-135b by activating JunB (or c-Jun)/AP-1. Consistently, knockdown of JunB (or c-Jun) decreased miR-135b levels, thereby increasing FOXO1. Moreover, Gα12QL induced MDM2, the deficiency of which facilitated FOXO1 accumulation. In addition, Gα12QL repressed miR-194 cluster gene products (194/192/215), which contributed to MDM2-mediated FOXO1 repression. In functional assays, Gα12QL facilitated tumor cell growth with alterations in cell cycle-associated protein levels, which was antagonized by enforced expression of FOXO1. In human HCCs, FOXO1 levels were decreased as compared with the surrounding liver tissue. Moreover, decrease of FOXO1 or miR-194 was statistically significant between stages T1 and T2, whereas increase of miR-135b discriminated tumor stage T3a versus T1/T2. In conclusion, Gα12gep oncogene inhibits FOXO1, which may result from the inhibition of FOXO1 de novo synthesis by miR-135b in conjunction with MDM2-mediated destabilization of FOXO1.


Assuntos
Carcinoma Hepatocelular/metabolismo , Fatores de Transcrição Forkhead/antagonistas & inibidores , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/metabolismo , MicroRNAs/genética , Animais , Carcinoma Hepatocelular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Fibroblastos , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/biossíntese , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/biossíntese , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/genética , Regulação Neoplásica da Expressão Gênica , Células Hep G2 , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/biossíntese , Proteínas Quinases JNK Ativadas por Mitógeno/genética , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Camundongos , MicroRNAs/biossíntese , Proteínas Proto-Oncogênicas c-mdm2/biossíntese , Proteínas Proto-Oncogênicas c-mdm2/genética , Interferência de RNA , RNA Interferente Pequeno , Transdução de Sinais , Fator de Transcrição AP-1/biossíntese , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética
4.
Pharmacol Rep ; 64(2): 256-65, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22661174

RESUMO

BACKGROUND: SB 269970, a 5-HT(7) receptor antagonist may produce a faster antidepressant-like effect in animal models, than do antidepressant drugs, e.g., imipramine. The present work was aimed at examining the effect of single and repeated (14 days) administration of SB 269970 on the 5-HT(7) receptor in the hippocampus. METHODS: The reactivity of 5-HT(7) receptors was determined using 5-carboxamidotryptamine (5-CT), which increased the bursting frequency of spontaneous epileptiform activity in hippocampal slices. Additionally, the effects of SB 269970 administration on the affinity and density of 5-HT(7) receptors were investigated using [(3)H]-SB 269970 and the influence of SB 269970 and imipramine on mRNA expression levels of Gα(s) and Gα(12) mRNA were studied using RT-qPCR. RESULTS: Acute and repeated treatment with SB 269970 led to attenuation of the excitatory effects of activation of 5-HT(7) receptors. Neither single nor repeated administration of SB 269970 changed the mean affinity of 5-HT(7) receptors for [(3)H]-SB 269970. Repeated, but not single, administration of SB 269970 decreased the maximum density of [(3)H]-SB 269970 binding sites. While administration of imipramine did not change the expression of mRNAs for Gα(s) and Gα(12) proteins after both single and repeated administration of SB 269970, a reduction in Gα(s) and Gα(12) mRNA expression levels was evident. CONCLUSIONS: These findings indicate that even single administration of SB269970 induces functional desensitization of the 5-HT(7) receptor system, which precedes changes in the receptor density. This mechanism may be responsible for the rapid antidepressant-like effect of the 5-HT(7) antagonist in animal models.


Assuntos
Hipocampo/efeitos dos fármacos , Fenóis/farmacologia , Receptores de Serotonina/metabolismo , Antagonistas da Serotonina/farmacologia , Sulfonamidas/farmacologia , Animais , Antidepressivos Tricíclicos/farmacologia , Sítios de Ligação , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Relação Dose-Resposta a Droga , Esquema de Medicação , Fenômenos Eletrofisiológicos , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/biossíntese , Subunidades alfa Gs de Proteínas de Ligação ao GTP/biossíntese , Hipocampo/metabolismo , Imipramina/farmacologia , Masculino , Fenóis/administração & dosagem , Ligação Proteica , RNA Mensageiro/biossíntese , Ratos , Ratos Wistar , Serotonina/análogos & derivados , Serotonina/farmacologia , Antagonistas da Serotonina/administração & dosagem , Sulfonamidas/administração & dosagem
5.
J Neurosci ; 32(9): 2915-30, 2012 Feb 29.
Artigo em Inglês | MEDLINE | ID: mdl-22378867

RESUMO

The common neurotransmitter serotonin controls different aspects of early neuronal differentiation, although the underlying mechanisms are poorly understood. Here we report that activation of the serotonin 5-HT(7) receptor promotes synaptogenesis and enhances synaptic activity in hippocampal neurons at early postnatal stages. An analysis of Gα(12)-deficient mice reveals a critical role of G(12)-protein for 5-HT(7) receptor-mediated effects in neurons. In organotypic preparations from the hippocampus of juvenile mice, stimulation of 5-HT(7)R/G(12) signaling potentiates formation of dendritic spines, increases neuronal excitability, and modulates synaptic plasticity. In contrast, in older neuronal preparations, morphogenetic and synaptogenic effects of 5-HT(7)/G(12) signaling are abolished. Moreover, inhibition of 5-HT(7) receptor had no effect on synaptic plasticity in hippocampus of adult animals. Expression analysis reveals that the production of 5-HT(7) and Gα(12)-proteins in the hippocampus undergoes strong regulation with a pronounced transient increase during early postnatal stages. Thus, regulated expression of 5-HT(7) receptor and Gα(12)-protein may represent a molecular mechanism by which serotonin specifically modulates formation of initial neuronal networks during early postnatal development.


Assuntos
Envelhecimento/genética , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/fisiologia , Hipocampo/citologia , Hipocampo/fisiologia , Neurogênese/genética , Neurônios/fisiologia , Receptores de Serotonina/fisiologia , Transdução de Sinais/genética , Animais , Animais Recém-Nascidos , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/biossíntese , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/genética , Hipocampo/crescimento & desenvolvimento , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Técnicas de Cultura de Órgãos , Receptores de Serotonina/biossíntese , Receptores de Serotonina/genética , Sinapses/genética
6.
Cancer Res ; 69(15): 6122-30, 2009 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-19602597

RESUMO

The molecular mechanisms behind the aggressiveness of nasopharyngeal carcinoma (NPC), a highly invasive and metastatic head and neck malignancy, have not been made clear. In this study investigating these mechanisms, guanine nucleotide-binding protein alpha(12) subunit (G(alpha)(12)) signaling was found by microarray analysis to be increased in primary NPC cells and NPC-derived cell lines. Using small interfering RNA to knock down G(alpha)(12) in NPC cells resulted in a reduction in cell migration and invasion as well as a reversal in fibroblastoid morphology. Using microarray analysis, we also found a reduction in expression of key actin dynamics regulators and several epithelial-to-mesenchymal transition-related genes in G(alpha)(12)-depleted NPC cells. Knocking down one of those genes, IQ motif containing GTPase activating protein 1, reduced the migration and formation of adherens junctions and reversed the fibroblastoid morphology of NPC cells, as knocking down G(alpha)(12) was found to do. Immunohistochemical analysis found NPC tumors to have significantly greater levels of G(alpha)(12) protein than the normal basal epithelial cells. Quantitative real-time PCR analysis revealed a significant correlation between G(alpha)(12) mRNA levels and NPC lymph node metastasis. Together, our findings support a model in which activation of G(alpha)(12) signaling promotes tumorigenesis and progression of NPC by modulating actin cytoskeleton reorganization and expression of epithelial-to-mesenchymal transition-related genes. =


Assuntos
Actinas/metabolismo , Citoesqueleto/metabolismo , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/metabolismo , Neoplasias Nasofaríngeas/metabolismo , Movimento Celular/fisiologia , Citoesqueleto/patologia , Regulação para Baixo , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/biossíntese , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/genética , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Imuno-Histoquímica , Metástase Linfática , Análise em Microsséries , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/patologia , Invasividade Neoplásica , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Transdução de Sinais , Transfecção , Células Tumorais Cultivadas , Proteínas Ativadoras de ras GTPase/biossíntese , Proteínas Ativadoras de ras GTPase/genética , Proteínas Ativadoras de ras GTPase/metabolismo
7.
Sheng Wu Gong Cheng Xue Bao ; 25(2): 235-41, 2009 Feb.
Artigo em Chinês | MEDLINE | ID: mdl-19459329

RESUMO

The G13 domain derived from granulysin shows high antimicrobial activities against Gram-positive and Gram-negative bacteria but does not lyse Jurkat cells or liposomes. To explore a new approach for high expression of the G13 domain, we fused the sequence encoding G13 to thioredoxin (Trx) gene to construct the recombinant expression vector (pThioHisA-G13). A cyanogen bromide (CNBr) cleavage site was introduced between the Trx and G13 to facilitate final release of the recombinant G13. The recombinant expression vector, pThioHisA-G13, was transformed into E. coli BL21 (DE3). Upon induction by IPTG Trx-G13 fusion protein was expressed and took the form of inclusion bodies counting 58% (W/W) of total cellular proteins. The inclusion body was solved by urea (8 mol/L) and then cleaved by CNBr. We purified the recombinant peptide G13 by one-step cation exchange chromatography. Results of agarose diffuse assay analysis indicated that the recombinant G13 exhibited antibacterial activity. The procedure described in this study will provide a reliable and simple method for highly efficient production of some cationic antimicrobial peptides.


Assuntos
Antígenos de Diferenciação de Linfócitos T/genética , Escherichia coli/metabolismo , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/biossíntese , Proteínas Recombinantes de Fusão/biossíntese , Anti-Infecciosos/metabolismo , Brometo de Cianogênio/farmacologia , Escherichia coli/genética , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/genética , Corpos de Inclusão/metabolismo , Estrutura Terciária de Proteína/genética , Proteínas Recombinantes de Fusão/genética , Tiorredoxinas/genética , Transfecção
8.
J Pharmacol Exp Ther ; 315(1): 248-55, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15987828

RESUMO

A growing body of literature indicates that chronic morphine exposure alters the expression and function of cytoskeletal proteins in addition to the well established interactions between mu opioid receptors and G proteins. In the present study, we hypothesized that chronic morphine alters the expression and functional effects of G alpha12, a G protein that regulates downstream cytoskeletal proteins via its control of RhoA. Our results showed that chronic morphine treatment decreased the expression of G alpha i2 (64%) and G alpha i3 (60%), had no effect of G alpha o, and increased G alpha12 (66%) expression in Chinese hamster ovary (CHO) cells expressing the cloned human mu opioid receptors (hMOR-CHO cells) but not in cells expressing a mutant mu opioid receptor that do not develop morphine tolerance and dependence (T394A-CHO cells). Morphine treatment had no significant effect on PAR-1 thrombin receptor-activated G protein activity, as measured by thrombin-stimulated guanosine 5'-O-(3-[35S]thio)triphosphate binding. Chronic morphine treatment significantly enhanced thrombin-stimulated RhoA activity and thrombin-stimulated expression of alpha-actinin, a cytoskeletal anchoring protein, in hMOR-CHO cells. Proteomic analysis of two-dimensional gel spots prepared from hMOR-CHO cells showed that morphine treatment affected the expression of a number of proteins associated with morphological changes. Up-regulation of G alpha12 and alpha-actinin by chronic morphine was also observed in mouse brain. Viewed collectively, these findings indicate, for the first time, that chronic morphine enhances the G alpha12-associated signaling system, which is involved in regulating cellular morphology and growth, supporting other findings that chronic morphine may alter cellular morphology, in addition to cellular function.


Assuntos
Proteínas do Citoesqueleto/biossíntese , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/biossíntese , Morfina/farmacologia , Receptores Opioides mu/fisiologia , Sequência de Aminoácidos , Animais , Células CHO , Cricetinae , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos ICR , Dados de Sequência Molecular , Proteômica , Receptores de Trombina/fisiologia , Regulação para Cima , Proteína rhoA de Ligação ao GTP/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...