Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Neurochem ; 154(6): 618-634, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32068252

RESUMO

The SynGAP protein is a major regulator of synapse biology and neural circuit function. Genetic variants linked to epilepsy and intellectual disability disrupt synaptic function and neural excitability. SynGAP has been involved in multiple signaling pathways and can regulate small GTPases with very different roles. Yet, the molecular bases behind this pleiotropy are poorly understood. We hypothesize that different SynGAP isoforms will mediate different sets of functions and that deciphering their spatio-temporal expression and subcellular localization will accelerate understanding their multiple functions. Using isoform-specific antibodies recognizing SynGAP in mouse and human samples we found distinctive developmental expression patterns for all SynGAP isoforms in five mouse brain areas. Particularly noticeable was the delayed expression of SynGAP-α1 isoforms, which directly bind to postsynaptic density-95, in cortex and hippocampus during the first 2 weeks of postnatal development. Suggesting that during this period other isoforms would have a more prominent role. Furthermore, we observed subcellular localization differences between isoforms, particularly throughout postnatal development. Consistent with previous reports, SynGAP was enriched in the postsynaptic density in the mature forebrain. However, SynGAP was predominantly found in non-synaptic locations in a period of early postnatal development highly sensitive to SynGAP levels. While, α1 isoforms were always found enriched in the postsynaptic density, α2 isoforms changed from a non-synaptic to a mostly postsynaptic density localization with age and ß isoforms were always found enriched in non-synaptic locations. The differential expression and subcellular distribution of SynGAP isoforms may contribute to isoform-specific regulation of small GTPases, explaining SynGAP pleiotropy.


Assuntos
Encéfalo/crescimento & desenvolvimento , Proteínas Ativadoras de ras GTPase/genética , Animais , Córtex Cerebral/crescimento & desenvolvimento , Córtex Cerebral/metabolismo , Simulação por Computador , Regulação da Expressão Gênica no Desenvolvimento/genética , Hipocampo/crescimento & desenvolvimento , Hipocampo/metabolismo , Humanos , Isomerismo , Camundongos , Camundongos Endogâmicos C57BL , Isoformas de Proteínas/biossíntese , Isoformas de Proteínas/genética , Proteômica , Frações Subcelulares/metabolismo , Proteínas Ativadoras de ras GTPase/biossíntese
2.
Arterioscler Thromb Vasc Biol ; 40(1): 112-127, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31619063

RESUMO

OBJECTIVE: Vascular endothelial cells (ECs) normally maintain vascular homeostasis and are regulated by proinflammatory cytokines and reactive oxygen species. A human genome-wide association study identified that AIP1 (ASK1 [apoptosis signal-regulating kinase 1]-interacting protein-1; also identified as DAB2IP) gene variants confer susceptibility to cardiovascular disease, but the underlying mechanism is unknown. Approach and Results: We detected a normal AIP1 form (named AIP1A) in the healthy aorta, but a shorter form of AIP1 (named AIP1B) was found in diseased aortae that contained atherosclerotic plaques and graft arteriosclerosis. AIP1B transcription in resting ECs was suppressed through epigenetic inhibition by RIF1 (Rap1 [ras-related protein 1]-interacting factor 1)/H3K9 (histone H3 lysine 9) methyltransferase-mediated H3K9 trimethylation, and this inhibition was released by proinflammatory cytokines. AIP1A, but not AIP1B, was downregulated by proteolytic degradation through a Smurf1 (SMAD [suppressor of mothers against decapentaplegic miscellaneous] ubiquitylation regulatory factor 1)-dependent pathway in ECs under inflammation. Therefore, AIP1B was the major form present during inflammatory conditions. AIP1B, which lacks the N-terminal pleckstrin homology domain of AIP1A, localized to the mitochondria and augmented TNFα (tumor necrosis factor alpha)-induced mitochondrial reactive oxygen species generation and EC activation. AIP1B-ECTG (EC-specific AIP1B transgenic) mice exhibited augmented reactive oxygen species production, EC activation, and neointima formation in vascular remodeling models. CONCLUSIONS: Our current study suggests that a shift from anti-inflammatory AIP1A to proinflammatory AIP1B during chronic inflammation plays a key role in inflammatory vascular diseases.


Assuntos
Arteriosclerose/genética , Endotélio Vascular/metabolismo , Regulação da Expressão Gênica , Estudo de Associação Genômica Ampla/métodos , Mitocôndrias/metabolismo , Proteínas Ativadoras de ras GTPase/genética , Animais , Aorta Torácica/metabolismo , Aorta Torácica/patologia , Apoptose , Arteriosclerose/metabolismo , Arteriosclerose/patologia , Western Blotting , Células Cultivadas , DNA/genética , Modelos Animais de Doenças , Endotélio Vascular/patologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia de Fluorescência , Mitocôndrias/patologia , Transdução de Sinais , Proteínas Ativadoras de ras GTPase/biossíntese
3.
J Cell Biochem ; 119(5): 3864-3872, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29125886

RESUMO

Nasopharyngeal carcinoma (NPC) is frequently seen in Chinese, especially the population that resides in southeast China. Metastasis-associated protein 1 (MTA1) is a chromatin modifier and plays a role in tumor cell metastasis. IQGAP1 is a ubiquitously expressed protein that contributes to cytoskeleton remodeling. This study aimed to investigate the role of MTA1 and IQGAP1 in NPC malignant transformation. MTA1 and IQGAP1 expression in NPC (n = 43) and control tissues (n = 31) were detected using qRT-PCR, immunoblot, and immunohistochemistry. MTA1 was overexpressed in CNE-1 and CNE-2 cell line by pcDNA3.1/MTA1 transfection. Dominant-negative p53 was transfected to inhibit p53 activity. si-IQGAP1 or dominant-negative IQGAP1 (IQGAP1ΔGRD) was used to suppress IQGAP1 activity. Cell proliferation was measured by CKK-8 assay. Cell migration was evaluated by Transwell assay. The results showed that MTA1 and IQGAP1 were highly expressed in NPC tissues compared with the controls. Forced expression of MTA1 accelerated cell proliferation and migration and upregulated IQGAP1 expression in a p53-independent way. Knockdown of IQGAP1 or transfection of dominant-negative IQGAP1 impeded tumor cell proliferation and migration as well as PI3K/Akt signaling induced by MTA1. In conclusion, MTA1 participates in NPC malignant transformation via regulating IQGAP1 expression and PI3K/Akt signaling pathway.


Assuntos
Movimento Celular , Regulação Neoplásica da Expressão Gênica , Histona Desacetilases/metabolismo , Carcinoma Nasofaríngeo/metabolismo , Neoplasias Nasofaríngeas/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Repressoras/metabolismo , Transdução de Sinais , Proteínas Ativadoras de ras GTPase/biossíntese , Adulto , Sobrevivência Celular , Feminino , Histona Desacetilases/genética , Humanos , Masculino , Pessoa de Meia-Idade , Carcinoma Nasofaríngeo/genética , Carcinoma Nasofaríngeo/patologia , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/patologia , Proteínas de Neoplasias/genética , Proteínas Repressoras/genética , Transativadores , Proteínas Ativadoras de ras GTPase/genética
4.
Genes Brain Behav ; 16(4): 462-471, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28058793

RESUMO

Previous studies on changes in murine brain gene expression associated with the selection for ethanol preference have used F2 intercross or heterogeneous stock (HS) founders, derived from standard laboratory strains. However, these populations represent only a small proportion of the genetic variance available in Mus musculus. To investigate a wider range of genetic diversity, we selected mice for ethanol preference using an HS derived from the eight strains of the collaborative cross. These HS mice were selectively bred (four generations) for high and low ethanol preference. The nucleus accumbens shell of naive S4 mice was interrogated using RNA sequencing (RNA-Seq). Gene networks were constructed using the weighted gene coexpression network analysis assessing both coexpression and cosplicing. Selection targeted one of the network coexpression modules (greenyellow) that was significantly enriched in genes associated with receptor signaling activity including Chrna7, Grin2a, Htr2a and Oprd1. Connectivity in the module as measured by changes in the hub nodes was significantly reduced in the low preference line. Of particular interest was the observation that selection had marked effects on a large number of cell adhesion molecules, including cadherins and protocadherins. In addition, the coexpression data showed that selection had marked effects on long non-coding RNA hub nodes. Analysis of the cosplicing network data showed a significant effect of selection on a large cluster of Ras GTPase-binding genes including Cdkl5, Cyfip1, Ndrg1, Sod1 and Stxbp5. These data in part support the earlier observation that preference is linked to Ras/Mapk pathways.


Assuntos
Consumo de Bebidas Alcoólicas/genética , Núcleo Accumbens/fisiologia , Animais , Etanol , Feminino , Expressão Gênica , Regulação da Expressão Gênica , Redes Reguladoras de Genes , Variação Genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/metabolismo , Análise de Sequência de RNA/métodos , Proteínas Ativadoras de ras GTPase/biossíntese , Proteínas Ativadoras de ras GTPase/genética
5.
Oncotarget ; 7(14): 18424-39, 2016 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-26919099

RESUMO

HER2-positive advanced gastric cancer patients frequently develop resistance to trastuzumab through mechanisms still poorly understood. In breast cancer, other members of the HER-family are known to be involved in trastuzumab-resistance, as is overexpression of the scaffold protein IQGAP1. In the present work, we investigated acquired resistance to trastuzumab in gastric cancer experimental models. Trastuzumab-resistant (HR) subclones derived from 3 HER2-overexpressing gastric cancer cells were generated and characterized for alterations in HER2-signaling mechanisms by next-generation sequencing, immunohistochemical, western blot and qRT-PCR techniques, and molecular modeling analysis. All subclones showed a reduced growth rate with respect to parental cell lines but each had a different resistance mechanism. In NCI N87 HR cells, characterized by a marked increase in HER2-signaling pathways with respect to the parental cell line, trastuzumab sensitivity was restored when IQGAP1 expression was silenced. AKG HR subclone showed higher HER3 protein expression than the parental line. High nuclear HER4 levels were observed in KKP HR cells. In conclusion, our study revealed that high IQGAP1 expression leads to resistance to trastuzumab in gastric cancer. Furthermore, 2 new mutations of the HER2 gene that may be involved in acquired resistance were identified in AKG HR and KKP HR subclones.


Assuntos
Neoplasias Gástricas/tratamento farmacológico , Trastuzumab/farmacologia , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos , Humanos , Receptor ErbB-2/biossíntese , Receptor ErbB-2/genética , Transdução de Sinais , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia , Proteínas Ativadoras de ras GTPase/biossíntese , Proteínas Ativadoras de ras GTPase/genética
6.
Oncogene ; 35(35): 4663-74, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-26876207

RESUMO

Targeted therapies using small-molecule inhibitors (SMIs) are commonly used in metastatic renal cell cancer (mRCC) patients; patients often develop drug resistance and eventually succumb to disease. Currently, understanding of mechanisms leading to SMIs resistance and any identifiable predictive marker(s) are still lacking. We discovered that DAB2IP, a novel Ras-GTPase-activating protein, was frequently epigenetically silenced in RCC, and DAB2IP loss was correlated with the overall survival of RCC patients. Loss of DAB2IP in RCC cells enhances their sensitivities to growth factor stimulation and resistances to SMI (such as mammalian target of rapamycin (mTOR) inhibitors). Mechanistically, loss of DAB2IP results in the activation of extracellular signal-regulated kinase/RSK1 and phosphoinositide-3 kinase/mTOR pathway, which synergizes the induction of hypoxia-inducible factor (HIF)-2α expression. Consequently, elevated HIF-2α suppresses p21/WAF1 expression that is associated with resistance to mTOR inhibitors. Thus combinatorial targeting both pathways resulted in a synergistic tumor inhibition. DAB2IP appears to be a new prognostic/predictive marker for mRCC patients, and its function provides a new insight into the molecular mechanisms of drug resistance to mTOR inhibitors, which also can be used to develop new strategies to overcome drug-resistant mRCC.


Assuntos
Carcinoma de Células Renais/genética , Proliferação de Células/genética , Resistencia a Medicamentos Antineoplásicos/genética , Serina-Treonina Quinases TOR/genética , Proteínas Ativadoras de ras GTPase/genética , Adulto , Idoso , Animais , Carcinoma de Células Renais/tratamento farmacológico , Carcinoma de Células Renais/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Pessoa de Meia-Idade , Terapia de Alvo Molecular , Inibidores de Proteínas Quinases/administração & dosagem , Transdução de Sinais/genética , Sirolimo/administração & dosagem , Serina-Treonina Quinases TOR/antagonistas & inibidores , Proteínas Ativadoras de ras GTPase/biossíntese
7.
J Neurosci ; 35(45): 15073-81, 2015 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-26558778

RESUMO

Previous studies have hypothesized that diverse genetic causes of intellectual disability (ID) and autism spectrum disorders (ASDs) converge on common cellular pathways. Testing this hypothesis requires detailed phenotypic analyses of animal models with genetic mutations that accurately reflect those seen in the human condition (i.e., have structural validity) and which produce phenotypes that mirror ID/ASDs (i.e., have face validity). We show that SynGAP haploinsufficiency, which causes ID with co-occurring ASD in humans, mimics and occludes the synaptic pathophysiology associated with deletion of the Fmr1 gene. Syngap(+/-) and Fmr1(-/y) mice show increases in basal protein synthesis and metabotropic glutamate receptor (mGluR)-dependent long-term depression that, unlike in their wild-type controls, is independent of new protein synthesis. Basal levels of phosphorylated ERK1/2 are also elevated in Syngap(+/-) hippocampal slices. Super-resolution microscopy reveals that Syngap(+/-) and Fmr1(-/y) mice show nanoscale alterations in dendritic spine morphology that predict an increase in biochemical compartmentalization. Finally, increased basal protein synthesis is rescued by negative regulators of the mGlu subtype 5 receptor and the Ras-ERK1/2 pathway, indicating that therapeutic interventions for fragile X syndrome may benefit patients with SYNGAP1 haploinsufficiency. SIGNIFICANCE STATEMENT: As the genetics of intellectual disability (ID) and autism spectrum disorders (ASDs) are unraveled, a key issue is whether genetically divergent forms of these disorders converge on common biochemical/cellular pathways and hence may be amenable to common therapeutic interventions. This study compares the pathophysiology associated with the loss of fragile X mental retardation protein (FMRP) and haploinsufficiency of synaptic GTPase-activating protein (SynGAP), two prevalent monogenic forms of ID. We show that Syngap(+/-) mice phenocopy Fmr1(-/y) mice in the alterations in mGluR-dependent long-term depression, basal protein synthesis, and dendritic spine morphology. Deficits in basal protein synthesis can be rescued by pharmacological interventions that reduce the mGlu5 receptor-ERK1/2 signaling pathway, which also rescues the same deficit in Fmr1(-/y) mice. Our findings support the hypothesis that phenotypes associated with genetically diverse forms of ID/ASDs result from alterations in common cellular/biochemical pathways.


Assuntos
Proteína do X Frágil da Deficiência Intelectual/biossíntese , Hipocampo/metabolismo , Hipocampo/fisiopatologia , Proteínas Ativadoras de ras GTPase/biossíntese , Animais , Espinhas Dendríticas/metabolismo , Espinhas Dendríticas/patologia , Potenciais Pós-Sinápticos Excitadores/fisiologia , Proteína do X Frágil da Deficiência Intelectual/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Técnicas de Cultura de Órgãos , Proteínas Ativadoras de ras GTPase/genética
8.
Int J Clin Exp Pathol ; 8(8): 8821-31, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26464624

RESUMO

IQGAPs genes play critical role in either induction or suppression of cancer and its progression, however the relationship between Ras genes and these genes are still unclear. In this study, we tried to understand the mechanistic action of IQGAPs genes and its correlation with Ras genes in mouse hepatic cancer model. The genetic expressions of IQGAP1, IQGAP2, IQGAP3, Hras, Kras, Nras, Mras, Caspase3, and BAX were followed in both hepatocellular carcinoma and normal liver cells of Balbc mice. Genotoxic agent diethylnitrosamine (DEN)-induced hepatic cancer model was induced in male mice and recorded the occurrence of hepatocellular carcinoma by morphological and histological changes in the liver. It was observed that mRNA expressions of IQGAP1, Hras, Kras, Nras, Mras, Caspase3, and BAX genes were highly elevated in hepatocellular carcinoma cells when compared with normal liver cells, additionally their expressions increased by concentrating the dose of DEN. While, the expressions of IQGAP2 and IQGAP3 were significantly decreased in hepatocellular carcinoma cells when compared with normal liver cells, as well as their expressions decreased more with increasing the dose of DEN. It was concluded from this study that IQGAP1 has a strong signaling relationship with Ras genes in induction of cancer and it is considered as a key gene for induction or suppression of the hepatocellular carcinoma.


Assuntos
Carcinoma Hepatocelular/patologia , Neoplasias Hepáticas/patologia , Proteínas Ativadoras de ras GTPase/biossíntese , Animais , Carcinoma Hepatocelular/genética , Modelos Animais de Doenças , Neoplasias Hepáticas/genética , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Reação em Cadeia da Polimerase em Tempo Real , Transcriptoma
9.
Int J Oncol ; 47(5): 1963-70, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26398880

RESUMO

MicroRNA (miRNA or miR)-506 is a novel miRNA related to the survival of breast cancer patients. However, the mechanism underlying miRNA-506 involvement in breast carcinogenesis remains unclear. In the present study, we found that miR-506 was downregulated in human breast malignant tissues and breast cancer cell lines by RT-qPCR analysis, and the expression level of miR-506 was decreased with the increasing of tumor stage. Subsequently, gain-of-function and loss-of-function experiments were performed in vitro, and the results from MTT assay, Transwell-Matrigel invasion assay and cell adhesion assay revealed that miR-506 suppresses cell proliferation, invasion and adhesion of breast cancer cells. Luciferase reporter assay revealed that IQ motif containing GTPase activating protein 1 (IQGAP1) is a direct target of miR-506. miR-506 represses the expression of IQGAP1 and its downstream extracellular signal regulated kinase (ERK) mitogen-activated protein kinase (MAPK) signaling pathways, as demonstrated by the RT-qPCR and western blot analysis. Furthermore, we found that IQGAP1 rescues the effect of miR-506 on cell proliferation, invasion, adhesion, and the activation of ERK MAPK signaling. In conclusion, the present study is the first to provide evidence that miR-506 acts as a tumor suppressor, at least partially, by directly downregulating IQGAP1 in breast cancer cells. The miR-506/IQGAP1/ERK pathway may be a novel therapeutic target in breast cancer.


Assuntos
Neoplasias da Mama/genética , MicroRNAs/biossíntese , Proteínas Ativadoras de ras GTPase/biossíntese , Neoplasias da Mama/patologia , Adesão Celular/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Sistema de Sinalização das MAP Quinases/genética , MicroRNAs/genética , Invasividade Neoplásica/genética , Metástase Neoplásica , Proteínas Ativadoras de ras GTPase/genética
10.
FEBS Lett ; 589(10): 1127-35, 2015 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-25841337

RESUMO

MicroRNAs have been reported to play critical roles in various cancers, but there has been no study on the role of miR-889 in cancers. Here, we report that over-expression of miR-889 leads to rapid proliferation of EC109 and EC9706 cells in vitro and in vivo by inducing cells into S-phase. Using bioinformatics methods, DAB2IP was further confirmed to be a direct target of miR-889. In addition, the expression of DAB2IP, which was negatively correlated with that of miR-889, was significantly associated with clinicopathological features of ESCC patients. In conclusion, miR-889 is an important regulator in ESCC and both miR-889 and DAB2IP may serve as promising biomarkers and therapeutic targets in patients with ESCC.


Assuntos
Biomarcadores Tumorais/biossíntese , Carcinoma de Células Escamosas/metabolismo , Proliferação de Células , Neoplasias Esofágicas/metabolismo , Regulação Neoplásica da Expressão Gênica , MicroRNAs/metabolismo , Proteínas de Neoplasias/biossíntese , RNA Neoplásico/metabolismo , Proteínas Ativadoras de ras GTPase/biossíntese , Biomarcadores Tumorais/genética , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patologia , Carcinoma de Células Escamosas/terapia , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patologia , Neoplasias Esofágicas/terapia , Células HeLa , Humanos , MicroRNAs/genética , Proteínas de Neoplasias/genética , RNA Neoplásico/genética , Proteínas Ativadoras de ras GTPase/genética
11.
Mol Med Rep ; 12(1): 442-8, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25684348

RESUMO

IQ motif containing guanosine triphosphatase activating protein 1 (IQGAP1) is associated with idiopathic pulmonary fibrogenesis (IPF); however, characterization of the expression of IQGAP1 in lung fibroblasts has remained elusive. The present study therefore evaluated IQGAP1 expression in mouse and human lung fibroblasts under fibrotic conditions via western blot analysis. It was revealed that IQGAP1 expression levels were significantly decreased in lung fibroblasts isolated from bleomycin-challenged mice than in those of control mice. Transforming growth factor-ß (TGF-ß) induced differentiation, as well as decreased expression of IQGAP1 in WI-38 cells human lung fibroblasts. Furthermore, inhibition of nuclear factor (NF)-κB activation restored the TGF-ß-induced inhibition of IQGAP1 expression in WI-38 cells. In lysophosphatidic acid (LPA)-challenged WI-38 cells, the expression of IQGAP1 was also decreased, while neutralized anti-TGF-ß antibody treatment restored the LPA-induced inhibition of IQGAP1 expression. These data indicated that TGF-ß inhibited IQGAP1 expression in lung fibroblasts via the NF-κB signaling pathway, presenting a potential novel therapeutic target for the treatment of IPF.


Assuntos
Fibrose Pulmonar Idiopática/genética , NF-kappa B/genética , Fator de Crescimento Transformador beta/genética , Proteínas Ativadoras de ras GTPase/biossíntese , Animais , Bleomicina/administração & dosagem , Diferenciação Celular/efeitos dos fármacos , Fibroblastos/metabolismo , Fibroblastos/patologia , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Fibrose Pulmonar Idiopática/patologia , Pulmão/metabolismo , Pulmão/patologia , Lisofosfolipídeos/administração & dosagem , Camundongos , Transdução de Sinais/efeitos dos fármacos , Fator de Crescimento Transformador beta/metabolismo , Proteínas Ativadoras de ras GTPase/genética
12.
Cancer Sci ; 105(6): 704-12, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24684735

RESUMO

The DOC-2/DAB2 interactive protein (DAB2IP) is a member of the Ras GTPase-activating protein family. It has been shown to be often downregulated and a poor prognostic factor in several human malignancies. In this study, we analyzed the clinicopathological features and outcomes of DAB2IP expression in 135 patients with urothelial carcinoma of the bladder (UCB) treated by radical cystectomy plus bilateral lymph node dissection, and evaluated the effect of DAB2IP knockdown in vitro using the MTT method, colony formation assay, cell cycle assay, and cell migration and invasive assay. We found low expression of DAB2IP was significantly associated with high pathological stage (P = 0.002), high pathological grade (P = 0.02), tumor size more than 3 cm (P = 0.04), and presence of histological variants (P = 0.01). DAB2IP was an independent prognostic factor of disease recurrence (hazard ratio, 2.67; P = 0.034) and cancer-specific survival (hazard ratio, 2.79; P = 0.038). Knockdown of DAB2IP could promote cell proliferation, migration, and invasion. Downregulation of DAB2IP could activate the ERK and Akt pathways and was correlated with the expression of epithelial-mesenchymal transition markers, such as E-cadherin and vimentin. In conclusion, downregulation of DAB2IP is associated with features of biologically aggressive UCB and results in cell proliferation, migration, and invasion of bladder cancer. DAB2IP may serve as a promising biomarker in patients with UCB treated by radical cystectomy and bilateral lymph node dissection.


Assuntos
Proliferação de Células , Neoplasias da Bexiga Urinária/patologia , Proteínas Ativadoras de ras GTPase/biossíntese , Caderinas/biossíntese , Movimento Celular/genética , Cistectomia , Regulação para Baixo , Ativação Enzimática , Transição Epitelial-Mesenquimal/genética , MAP Quinases Reguladas por Sinal Extracelular/genética , Feminino , Humanos , Excisão de Linfonodo , Metástase Linfática , Masculino , Pessoa de Meia-Idade , Invasividade Neoplásica , Recidiva Local de Neoplasia/genética , Proteínas Proto-Oncogênicas c-akt/genética , Interferência de RNA , RNA Interferente Pequeno , Resultado do Tratamento , Neoplasias da Bexiga Urinária/mortalidade , Neoplasias da Bexiga Urinária/cirurgia , Urotélio/patologia , Urotélio/cirurgia , Vimentina/biossíntese , Proteínas Ativadoras de ras GTPase/genética
13.
Cell Rep ; 5(4): 1060-9, 2013 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-24268772

RESUMO

Elevated bile acid levels increase hepatocellular carcinoma by unknown mechanisms. Here, we show that mice with a severe defect in bile acid homeostasis due to the loss of the nuclear receptors FXR and SHP have enlarged livers, progenitor cell proliferation, and Yes-associated protein (YAP) activation and develop spontaneous liver tumorigenesis. This phenotype mirrors mice with loss of hippo kinases or overexpression of their downstream target, YAP. Bile acids act as upstream regulators of YAP via a pathway dependent on the induction of the scaffold protein IQGAP1. Patients with diverse biliary dysfunctions exhibit enhanced IQGAP1 and nuclear YAP expression. Our findings reveal an unexpected mechanism for bile acid regulation of liver growth and tumorigenesis via the Hippo pathway.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Ácidos e Sais Biliares/metabolismo , Carcinogênese , Carcinoma Hepatocelular/patologia , Neoplasias Hepáticas/patologia , Fosfoproteínas/metabolismo , Proteínas Ativadoras de ras GTPase/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Adolescente , Animais , Carcinoma Hepatocelular/genética , Proteínas de Ciclo Celular , Células Cultivadas , Criança , Pré-Escolar , Ativação Enzimática/genética , Fator de Crescimento de Hepatócito/genética , Via de Sinalização Hippo , Humanos , Lactente , Recém-Nascido , Neoplasias Hepáticas/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosfoproteínas/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas/genética , Receptores Citoplasmáticos e Nucleares/genética , Serina-Treonina Quinase 3 , Fatores de Transcrição , Proteínas de Sinalização YAP , Proteínas Ativadoras de ras GTPase/biossíntese , Proteínas Ativadoras de ras GTPase/genética
14.
Laryngoscope ; 122(12): 2640-6, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22833392

RESUMO

OBJECTIVES/HYPOTHESIS: The scaffold protein IQGAP1 is a ubiquitously expressed 190 kDa protein that participates in protein-protein interactions, integrating diverse signaling pathways. In this study, we examined the expression of IQGAP1 in sinonasal inverted papillomas (IP) and sinonasal squamous cell carcinomas (SCC), which include IP with SCC (IPcSCC) and SCC alone. We also tried to elucidate whether the amount of IQGAP1 expression is related with any clinicopathologic features of sinonasal SCCs. STUDY DESIGN: This was a retrospective study. METHODS: The expression of IQGAP1 was analyzed by means of immunohistochemistry. The degrees of IQGAP1 expression and subcellular localization (rim formation) were analyzed with respect to the following clinicopathologic variables: age, gender, T stage, histologic differentiation, local recurrence, and distant metastasis. RESULTS: The expression of IQGAP1 was significantly higher in patients with IPcSCC (3.62 ± 0.14), and SCC (2.93 ± 0.34) compared with IP (1.43 ± 0.26). The rim formation scale was significantly higher in IPcSCC (2.39 ± 0.24) compared with IP (1.68 ± 0.15). Distant metastasis in patients with SCC significantly correlated with an increased IQGAP1 rim formation (p = 0.011). Histologic differentiation was also significantly correlated with a positive staining scale for IQGAP1 in IPcSCCs (p = 0.038). CONCLUSION: These results indicated that IQGAP1 expression is associated with both the histologic differentiation of IPcSCCs and is present in SCC cases where distant metastasis has occurred.


Assuntos
Carcinoma de Células Escamosas/metabolismo , Neoplasias Nasais/metabolismo , Papiloma Invertido/metabolismo , Neoplasias dos Seios Paranasais/metabolismo , Proteínas Ativadoras de ras GTPase/biossíntese , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores Tumorais/biossíntese , Carcinoma de Células Escamosas/patologia , Diagnóstico Diferencial , Feminino , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Neoplasias Nasais/patologia , Papiloma Invertido/patologia , Neoplasias dos Seios Paranasais/patologia , Estudos Retrospectivos , Adulto Jovem
15.
Cancer Genet ; 205(3): 113-23, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22469510

RESUMO

MicroRNAs (miRNAs) are a class of 21-23 nucleotide RNA molecules that play critical roles in the regulation of various cancers, including human lung cancer. Among them, miR-26a has been identified as a tumor-related regulator in several cancers, but its pathophysiologic properties and correlation with the development of human lung cancer remain unclear. In this study, it was determined that miR-26a expression is clearly down-regulated in human lung cancer tissues relative to normal tissues. Meanwhile, the overexpression of miR-26a in the A549 human lung cancer cell line dramatically inhibited cell proliferation, blocked G1/S phase transition, induced apoptosis, and inhibited cell metastasis and invasion in vitro. In contrast, a miR-26a inhibitor was used to transfect A549 cells, and the inhibition of endogenous miR-26a promoted cell metastasis and invasion. In addition, miR-26a expression inhibited the expression of enhancer of zeste homolog 2 (EZH2) and transactivated downstream target genes, including disabled homolog 2 (Drosophila) interacting protein gene (DAB2IP) and human Runt-related transcription factor 3 (RUNX3), which suggests that EZH2 is a potential target of miR-26a as previously reported. In conclusion, miR-26a plays an important role as an anti-oncogene in the molecular mechanism of human lung cancer and could potentially be used for the treatment of lung cancer.


Assuntos
Adenocarcinoma/genética , Proteínas de Ligação a DNA/metabolismo , Genes Supressores de Tumor , Neoplasias Pulmonares/genética , MicroRNAs/genética , MicroRNAs/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/biossíntese , Proteínas Adaptadoras de Transdução de Sinal/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Adenocarcinoma de Pulmão , Apoptose/genética , Proteínas Reguladoras de Apoptose , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Pontos de Checagem do Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células , Subunidade alfa 3 de Fator de Ligação ao Core/biossíntese , Subunidade alfa 3 de Fator de Ligação ao Core/genética , Proteínas de Ligação a DNA/genética , Proteína Potenciadora do Homólogo 2 de Zeste , Regulação Neoplásica da Expressão Gênica , Humanos , Pulmão/metabolismo , Pulmão/patologia , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Invasividade Neoplásica/genética , Metástase Neoplásica/genética , Complexo Repressor Polycomb 2 , Fatores de Transcrição/genética , Proteínas Supressoras de Tumor , Proteínas Ativadoras de ras GTPase/biossíntese , Proteínas Ativadoras de ras GTPase/genética
16.
Free Radic Res ; 45(10): 1124-35, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21740309

RESUMO

Reactive oxygen species (ROS) are important mediators for VEGF receptor 2 (VEGFR2) signalling involved in angiogenesis. The initial product of Cys oxidation, cysteine sulfenic acid (Cys-OH), is a key intermediate in redox signal transduction; however, its role in VEGF signalling is unknown. We have previously demonstrated IQGAP1 as a VEGFR2 binding scaffold protein involved in ROS-dependent EC migration and post-ischemic angiogenesis. Using a biotin-labelled Cys-OH trapping reagent, we show that VEGF increases protein-Cys-OH formation at the lamellipodial leading edge where it co-localizes with NADPH oxidase and IQGAP1 in migrating ECs, which is prevented by IQGAP1 siRNA or trapping of Cys-OH with dimedone. VEGF increases IQGAP1-Cys-OH formation, which is prevented by N-acetyl cysteine or dimedone, which inhibits VEGF-induced EC migration and capillary network formation. In vivo, hindlimb ischemia in mice increases Cys-OH formation in small vessels and IQGAP1 in ischemic tissues. In summary, VEGF stimulates localized formation of Cys-OH-IQGAP1 at the leading edge, thereby promoting directional EC migration, which may contribute to post-natal angiogenesis in vivo. Thus, targeting Cys-oxidized proteins at specific compartments may be the potential therapeutic strategy for various angiogenesis-dependent diseases.


Assuntos
Movimento Celular/fisiologia , Cisteína/análogos & derivados , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Actinas/metabolismo , Animais , Movimento Celular/efeitos dos fármacos , Cisteína/biossíntese , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , NADPH Oxidases/metabolismo , Neovascularização Fisiológica/efeitos dos fármacos , Neovascularização Fisiológica/fisiologia , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Ácidos Sulfênicos , Transfecção , Fator A de Crescimento do Endotélio Vascular/farmacologia , Proteínas Ativadoras de ras GTPase/biossíntese , Proteínas Ativadoras de ras GTPase/genética , Proteínas Ativadoras de ras GTPase/metabolismo
17.
Biol Psychiatry ; 70(7): 646-54, 2011 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-21549355

RESUMO

BACKGROUND: Impairment of glutamate neurons that relay sensory and cognitive information from the medial dorsal thalamus to the dorsolateral prefrontal cortex and other cortical regions may contribute to the pathophysiology of schizophrenia. In this study, we have assessed the cell-specific expression of glutamatergic transcripts in the medial dorsal thalamus. METHODS: We used laser capture microdissection to harvest two populations of medial dorsal thalamic cells, one enriched with glutamatergic relay neurons and the other with gamma-aminobutyric acidergic neurons and astroglia, from postmortem brains of subjects with schizophrenia (n = 14) and a comparison group (n = 20). Quantitative polymerase chain reaction of extracted RNA was used to assay gene expression in the different cell populations. RESULTS: The transcripts encoding the ionotropic glutamate receptor subunits NR2D, GluR3, GluR6, GluR7, and the intracellular proteins GRIP1 and SynGAP1 were significantly decreased in relay neurons but not in the mixed glial and interneuron population in schizophrenia. CONCLUSIONS: Our data suggest that reduced ionotropic glutamatergic expression occurs selectively in neurons, which give rise to the cortical projections of the medial dorsal thalamus in schizophrenia, rather than in thalamic cells that function locally. Our findings indicate that glutamatergic innervation is dysfunctional in the circuitry between the medial dorsal thalamus and cortex.


Assuntos
Córtex Cerebral/metabolismo , Receptores de Glutamato/biossíntese , Esquizofrenia/metabolismo , Tálamo/metabolismo , Proteínas de Transporte/biossíntese , Neurônios GABAérgicos/metabolismo , Expressão Gênica , Humanos , Interneurônios/metabolismo , Microdissecção e Captura a Laser/métodos , Proteínas do Tecido Nervoso/biossíntese , Vias Neurais/metabolismo , Neuroglia/metabolismo , Neurônios/metabolismo , Proteínas Ativadoras de ras GTPase/biossíntese
18.
J Neurophysiol ; 106(1): 309-18, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21525372

RESUMO

Synaptic GTPase-activating protein (SynGAP) is a neuronal-specific Ras/Rap-GAP that increases the hydrolysis rate of GTP to GDP, converting Ras/Rap from the active into the inactive form. The Ras protein family modulates a wide range of cellular pathways including those involved in sensitization of sensory neurons. Since GAPs regulate Ras activity, SynGAP might be an important regulator of peripheral sensitization and pain. Therefore, we evaluated excitability, stimulus-evoked release of the neuropeptide calcitonin gene-related peptide (CGRP), and nociception from wild-type (WT) mice and those with a heterozygous mutation of the SynGAP gene (SynGAP(+/-)). Our results demonstrate that SynGAP is expressed in primary afferent sensory neurons and that the capsaicin-stimulated CGRP release from spinal cord slices was two-fold higher from SynGAP(+/-) mice than that observed from WT mouse tissue, consistent with an increase in expression of the capsaicin receptor, transient receptor potential cation channel subfamily V member 1 (TRPV1), in SynGAP(+/-) dorsal root ganglia. However, there was no difference between the two genotypes in potassium-stimulated release of CGRP, the number of action potentials generated by a ramp of depolarizing current, or mechanical hypernociception elicited by intraplantar injection of capsaicin. In contrast, capsaicin-induced thermal hypernociception occurred at lower doses of capsaicin and had a longer duration in SynGAP(+/-) mice than WT mice. These results provide the first evidence that SynGAP is an important regulator of neuropeptide release from primary sensory neurons and can modulate capsaicin-induced hypernociception, demonstrating the importance of GAP regulation in signaling pathways that play a role in peripheral sensitization.


Assuntos
Capsaicina/farmacologia , Fármacos do Sistema Sensorial/farmacologia , Proteínas Ativadoras de ras GTPase/biossíntese , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Células Cultivadas , Estimulação Elétrica , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dor/induzido quimicamente , Potássio/fisiologia , Células Receptoras Sensoriais/efeitos dos fármacos , Células Receptoras Sensoriais/fisiologia , Canais de Cátion TRPV/fisiologia , Proteínas Ativadoras de ras GTPase/genética , Proteínas Ativadoras de ras GTPase/fisiologia
19.
Mol Cell Proteomics ; 10(4): M110.003178, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21263009

RESUMO

Class I phosphoinositide 3-kinases exert important cellular effects through their two primary lipid products, phosphatidylinositol 3,4,5-trisphosphate and phosphatidylinositol 3,4-bisphosphate (PtdIns(3,4)P(2)). As few molecular targets for PtdIns(3,4)P(2) have yet been identified, a screen for PI 3-kinase-responsive proteins that is selective for these is described. This features a tertiary approach incorporating a unique, primary recruitment of target proteins in intact cells to membranes selectively enriched in PtdIns(3,4)P(2). A secondary purification of these proteins, optimized using tandem pleckstrin homology domain containing protein-1 (TAPP-1), an established PtdIns(3,4)P(2) selective ligand, yields a fraction enriched in proteins of potentially similar lipid binding character that are identified by liquid chromatography-tandem MS. Thirdly, this approach is coupled to stable isotope labeling with amino acids in cell culture using differential isotope labeling of cells stimulated in the absence and presence of the PI 3-kinase inhibitor wortmannin. This provides a ratio-metric readout that distinguishes authentically responsive components from copurifying background proteins. Enriched fractions thus obtained from astrocytoma cells revealed a subset of proteins that exhibited ratios indicative of their initial, cellular responsiveness to PI 3-kinase activation. The inclusion among these of tandem pleckstrin homology domain containing protein-1, three isoforms of Akt, switch associated protein-70, early endosome antigen-1 and of additional proteins expressing recognized lipid binding domains demonstrates the utility of this strategy and lends credibility to the novel candidate proteins identified. The latter encompass a broad set of proteins that include the gene product of TBC1D2A, a putative Rab guanine nucleotide triphosphatase activating protein (GAP) and IQ motif containing GAP1, a potential tumor promoter. A sequence comparison of the former protein indicates the presence of a pleckstrin homology domain whose lipid binding character remains to be established. IQ motif containing GAP1 lacks known lipid interacting components and a preliminary analysis here indicates that this may exemplify a novel class of atypical phosphoinositide (aPI) binding domain.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Membrana/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais , Linhagem Celular Tumoral , Estudos de Viabilidade , Humanos , Fragmentos de Peptídeos/biossíntese , Fosfatos de Fosfatidilinositol/metabolismo , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Mapeamento de Interação de Proteínas , Proteínas Recombinantes de Fusão/biossíntese , Ressonância de Plasmônio de Superfície , Proteínas Ativadoras de ras GTPase/biossíntese
20.
BMC Gastroenterol ; 10: 125, 2010 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-20977743

RESUMO

BACKGROUND: IQGAP1 and IQGAP2 are homologous members of the IQGAP family of scaffold proteins. Accumulating evidence implicates IQGAPs in tumorigenesis. We recently reported that IQGAP2 deficiency leads to the development of hepatocellular carcinoma (HCC) in mice. In the current study we extend these findings, and investigate IQGAP1 and IQGAP2 expression in human HCC. METHODS: IQGAP1 and IQGAP2 protein expression was assessed by Western blotting and immunohistochemistry. IQGAP mRNA was measured by quantitative RT-PCR. The methylation status of the Iqgap2 promoter was determined by pyrosequencing of bisulfite-treated genomic DNA. RESULTS: IQGAP1 and IQGAP2 expression was reciprocally altered in 6/6 liver cancer cell lines. Similarly, immunohistochemical staining of 82 HCC samples showed that IQGAP2 protein expression was reduced in 64/82 (78.0%), while IQGAP1 was present in 69/82 (84.1%). No IQGAP1 staining was detected in 23/28 (82.1%) normal livers, 4/4 (100.0%) hepatic adenomas and 23/23 (100.0%) cirrhosis cases, while IQGAP2 was increased in 22/28 (78.6%), 4/4 (100.0%) and 23/23 (100.0%), respectively. Although the Iqgap2 promoter was not hypermethylated in HCC at any of the 25 CpG sites studied (N = 17), IQGAP2 mRNA levels were significantly lower in HCC specimens (N = 23) than normal livers (N = 6). CONCLUSIONS: We conclude that increased IQGAP1 and/or decreased IQGAP2 contribute to the pathogenesis of human HCC. Furthermore, downregulation of IQGAP2 in HCC occurs independently of hypermethylation of the Iqgap2 promoter. Immunostaining of IQGAP1 and IQGAP2 may aid in the diagnosis of HCC, and their pharmacologic modulation may represent a novel therapeutic strategy for the treatment of liver cancer.


Assuntos
Carcinoma Hepatocelular/genética , DNA de Neoplasias/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Hepáticas/genética , Proteínas Ativadoras de ras GTPase/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Biópsia , Western Blotting , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Feminino , Predisposição Genética para Doença , Humanos , Imuno-Histoquímica , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Masculino , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase , Células Tumorais Cultivadas , Proteínas Ativadoras de ras GTPase/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...