Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros

Medicinas Complementares
Tipo de documento
Intervalo de ano de publicação
1.
F1000Res ; 8: 960, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31372216

RESUMO

Background: Although both genetic and environmental factors have been reported to influence the risk of isolated cleft lip with or without cleft palate (CL/P), the exact mechanisms behind CL/P are still largely unaccounted for. We recently developed new methods to identify parent-of-origin (PoO) interactions with environmental exposures (PoOxE) and applied them to families with children born with isolated cleft palate only. Here, we used the same genome-wide association study (GWAS) dataset and methodology to screen for PoOxE effects in the larger sample of CL/P triads. Methods: Genotypes from 1594 complete triads and 314 dyads (1908 nuclear families in total) with CL/P were available for the current analyses. Of these families, 1024 were Asian, 825 were European and 59 had other ancestries. After quality control, 341,191 SNPs remained from the original 569,244. The exposures were maternal cigarette smoking, use of alcohol, and use of vitamin supplements in the periconceptional period. The methodology applied in the analyses is implemented in the R-package Haplin. Results: Among Europeans, there was evidence of a PoOxSmoke effect for ANK3 with three SNPs (rs3793861, q=0.20, p=2.6e-6; rs7087489, q=0.20, p=3.1e-6; rs4310561, q=0.67, p=4.0e-5) and a PoOxAlcohol effect for ARHGEF10 with two SNPs (rs2294035, q=0.32, p=2.9e-6; rs4876274, q=0.76, p=1.3e-5). Conclusion: Our results indicate that the detected PoOxE effects have a plausible biological basis, and thus warrant replication in other independent cleft samples. Our demonstration of the feasibility of identifying complex interactions between relevant environmental exposures and PoO effects offers new avenues for future research aimed at unravelling  the complex etiology of cleft lip defects.


Assuntos
Consumo de Bebidas Alcoólicas , Anquirinas , Fenda Labial , Fissura Palatina , Fatores de Troca de Nucleotídeo Guanina Rho , Fumar , Anquirinas/genética , Criança , Fenda Labial/genética , Fissura Palatina/genética , Feminino , Estudo de Associação Genômica Ampla , Humanos , Gravidez , Efeitos Tardios da Exposição Pré-Natal
2.
J Nat Prod ; 81(7): 1610-1618, 2018 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-29969260

RESUMO

In an effort to find potent natural inhibitors of RhoA and p115 signaling G-proteins, a systematic in vitro evaluation using enzymatic and plasmonic resonance assays was undertaken on 11 317 plant extracts. The screening procedure led to the selection of the New Caledonian endemic species Meiogyne baillonii for a chemical investigation. Using a bioguided isolation procedure, three enediyne-γ-butyrolactones (1-3) and two enediyne-γ-butenolides (4 and 5), named sapranthins H-L, respectively, two enediyne carboxylic acid (6 and 7), two depsidones, stictic acid (8) and baillonic acid (9), aristolactams AIa and AIIa (10 and 11), and two aporphines, dehydroroemerine (12) and noraristolodione (13), were isolated from the ethyl acetate extract of the bark. The structures of the new compounds (1-6, 9, and 11) and their relative configurations were established by NMR spectroscopic analysis and by X-ray diffraction analysis for compound 9. Only stictic acid (8) exhibited a significant inhibiting activity of the RhoA-p115 complex, with an EC50 value of 0.19 ± 0.05 mM. This is the first time that a natural inhibitor of the complex RhoA-p115's activity was discovered from an HTS performed over a collection of higher plant extracts. Thus, stictic acid (8) could be used as the first reference compound inhibiting the interaction between RhoA and p115.


Assuntos
Annonaceae/química , Extratos Vegetais/farmacologia , Fatores de Troca de Nucleotídeo Guanina Rho/antagonistas & inibidores , Proteína rhoA de Ligação ao GTP/antagonistas & inibidores , Espectroscopia de Ressonância Magnética , Estrutura Molecular , Casca de Planta/química , Extratos Vegetais/química
3.
Int J Oncol ; 48(5): 2098-112, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26983669

RESUMO

Osteosarcoma is the most common primary malignant tumor of the bone. The long-term survivals continue to be unsatisfactory for patients with metastatic and recurrent disease. Metastasis is still a severe challenge in osteosarcoma treatment. Sinomenine, an alkaloid from traditional Chinese medicine, has been proved to possess potent antitumor and anti-invasion effect on various cancers. However, the effect of sinomenine on human osteosarcoma and the underlying mechanisms remains unknown. We report here that sinomenine inhibited proliferation by inducing S phase arrest and suppressing the clone formation. Significant inhibitory effects were found in invasion and metastasis in osteosarcoma, but little cytotoxicity was observed in tested concentrations. Exposure to sinomenine resulted in suppression of invasion and migration in osteosarcoma cells as well as tube formation ability in the human umbilical vein endothelial cells (HUVEC) and U2OS cells. Furthermore, it demonstrated that CXCR4 played a key role contributing to invasion in osteosarcoma which is considered to be a core target site in sinomenine treatment. Sinomenine inhibited invasion by suppressing CXCR4 and STAT3 phosphorylation then downregulating the expression of MMP-2, MMP-9, RANKL, VEGF downstream. In addition, then RANKL-mediated bone destruction stimulated by osteoclastogenesis and VEGF-related neovascularization were restrained. Importantly, in vivo, sinomenine suppressed proliferation, osteoclastogenesis and bone destruction. Through these various comprehensive means, sinomenine inhibits metastasis in osteosarcoma. Taken together, our results revealed that sinomenine caused S phase arrest, inhibited invasion and metastasis via suppressing the CXCR4-STAT3 pathway and then osteoclastogenesis-mediated bone destruction and neovascularization in osteosarcoma. Sinomenine is therefore a promising adjuvant agent for metastasis control in osteosarcoma.


Assuntos
Inibidores da Angiogênese/administração & dosagem , Neoplasias Ósseas/tratamento farmacológico , Morfinanos/administração & dosagem , Osteossarcoma/tratamento farmacológico , Receptores CXCR4/metabolismo , Pontos de Checagem da Fase S do Ciclo Celular/efeitos dos fármacos , Inibidores da Angiogênese/farmacologia , Animais , Neoplasias Ósseas/metabolismo , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana , Humanos , Camundongos , Morfinanos/farmacologia , Metástase Neoplásica , Osteossarcoma/metabolismo , Fatores de Troca de Nucleotídeo Guanina Rho/metabolismo , Transdução de Sinais/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Artigo em Inglês | WPRIM | ID: wpr-89593

RESUMO

PURPOSE: We investigated the protective effects of the herbal formulation KH-204 in the bladder of androgen-deprived rats. MATERIALS AND METHODS: Male rats aged eight weeks were randomly divided into four groups, containing eight rats each: sham operation only (normal control group), androgen-deprived only (androgen-deprived control group), and androgen-deprived followed by treatment with 200 mg/kg or 400 mg/kg of KH-204. After 0.5 mg/kg of leuprorelin was subcutaneously injected in the androgen-deprived groups, the oral administration of either distilled water in the two control groups or KH-204 in the treatment group was continued for four weeks. Serum testosterone levels, RhoGEF levels, nitric oxide (NO)-cyclic guanosine monophosphate (cGMP)-related parameters, oxidative stress, and histologic changes were evaluated after treatment. RESULTS: Treatment with the herbal formulation KH-204 (1) increased serum testosterone levels; (2) restored the expression of RhoGEFs, endothelial NO synthase, and neuronal NO synthase; (3) increased the expression of superoxide dismutase; and (4) decreased bladder fibrosis. CONCLUSIONS: Our results suggest that the positive effects of KH-204 on the urinary bladder may be attributed to its antioxidant effects or to an elevation in NO-cGMP activity.


Assuntos
Animais , Humanos , Masculino , Ratos , Administração Oral , Antioxidantes , Fibrose , Guanosina Monofosfato , Hipogonadismo , Leuprolida , Neurônios , Óxido Nítrico , Óxido Nítrico Sintase , Estresse Oxidativo , Fitoterapia , Fatores de Troca de Nucleotídeo Guanina Rho , Superóxido Dismutase , Testosterona , Bexiga Urinária , Água
5.
PLoS One ; 9(2): e88098, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24505392

RESUMO

The small GTPase cycles between the inactive GDP form and the activated GTP form, catalyzed by the upstream guanine exchange factors. The modulation of such process by small molecules has been proven to be a fruitful route for therapeutic intervention to prevent the over-activation of the small GTPase. The fragment based approach emerging in the past decade has demonstrated its paramount potential in the discovery of inhibitors targeting such novel and challenging protein-protein interactions. The details regarding the procedure of NMR fragment screening from scratch have been rarely disclosed comprehensively, thus restricts its wider applications. To achieve a consistent screening applicable to a number of targets, we developed a highly automated protocol to cover every aspect of NMR fragment screening as possible, including the construction of small but diverse libray, determination of the aqueous solubility by NMR, grouping compounds with mutual dispersity to a cocktail, and the automated processing and visualization of the ligand based screening spectra. We exemplified our streamlined screening in RhoA alone and the complex of the small GTPase RhoA and its upstream guanine exchange factor LARG. Two hits were confirmed from the primary screening in cocktail and secondary screening over individual hits for LARG/RhoA complex, while one of them was also identified from the screening for RhoA alone. HSQC titration of the two hits over RhoA and LARG alone, respectively, identified one compound binding to RhoA.GDP at a 0.11 mM affinity, and perturbed the residues at the switch II region of RhoA. This hit blocked the formation of the LARG/RhoA complex, validated by the native gel electrophoresis, and the titration of RhoA to ¹5N labeled LARG in the absence and presence the compound, respectively. It therefore provides us a starting point toward a more potent inhibitor to RhoA activation catalyzed by LARG.


Assuntos
Ressonância Magnética Nuclear Biomolecular/métodos , Fatores de Troca de Nucleotídeo Guanina Rho/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Avaliação Pré-Clínica de Medicamentos/métodos , Humanos , Modelos Moleculares , Mapas de Interação de Proteínas/efeitos dos fármacos , Estrutura Terciária de Proteína , Fatores de Troca de Nucleotídeo Guanina Rho/antagonistas & inibidores , Fatores de Troca de Nucleotídeo Guanina Rho/química , Solubilidade , Água/química , Proteína rhoA de Ligação ao GTP/antagonistas & inibidores , Proteína rhoA de Ligação ao GTP/química
6.
J Pharmacol Exp Ther ; 339(3): 746-56, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21875949

RESUMO

Leptin is a 16-kDa peptide primarily derived from white adipocytes and is typically elevated in plasma of obese individuals. Although leptin plays a critical role in appetite regulation, leptin receptors have been identified in numerous tissues including the heart and have been shown to directly mediate cardiac hypertrophy through RhoA/ROCK (Ras homolog gene family, member A/Rho-associated, coiled-coil containing protein kinase)-dependent p38 mitogen-activated protein kinase (MAPK) activation; however, the basis for RhoA stimulation is unknown. Rho guanine nucleotide exchange factors (GEFs) catalyze the exchange of GDP for GTP resulting in Rho activation and may be the potential upstream factors mediating leptin-induced RhoA activation and therefore a potential target for inhibition. We investigated the effects of North American ginseng (Panax quinquefolius), reported to reduce cardiac hypertrophy, on RhoA/ROCK and MAPK activation in ventricular cardiomyocytes exposed to leptin (50 ng/ml) and the possible role of p115RhoGEF and p63RhoGEF in these responses. Leptin produced a robust hypertrophic response that was associated with RhoA/ROCK activation resulting in a significant increase in cofilin-2 phosphorylation and actin polymerization, the latter evidenced by a reduction in the G/F actin ratio. These effects were prevented by ginseng (10 µg/ml). The stimulation of RhoA/ROCK by leptin was associated with significantly increased p115RhoGEF gene and protein expression and exchange activity, all of which were completely prevented by ginseng. The ability of ginseng to prevent leptin-induced activation of RhoA/ROCK was further associated with diminished p38 MAPK activation and nuclear translocation. These results demonstrate a potent inhibitory effect of ginseng against leptin-induced cardiac hypertrophy, an effect associated with prevention of p115RhoGEF-RhoA/ROCK-dependent p38 MAPK activation.


Assuntos
Cardiomegalia/tratamento farmacológico , Cardiotônicos/farmacologia , Leptina/fisiologia , Miócitos Cardíacos/efeitos dos fármacos , Panax , Fitoterapia , Extratos Vegetais/farmacologia , Actinas/efeitos dos fármacos , Actinas/fisiologia , Animais , Cardiomegalia/fisiopatologia , Cofilina 2/metabolismo , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Fatores de Troca do Nucleotídeo Guanina/genética , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Ventrículos do Coração/efeitos dos fármacos , Ventrículos do Coração/enzimologia , Ventrículos do Coração/fisiopatologia , Humanos , Leptina/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Miócitos Cardíacos/enzimologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Fosforilação/efeitos dos fármacos , Raízes de Plantas , Ratos , Ratos Sprague-Dawley , Receptores para Leptina/metabolismo , Fatores de Troca de Nucleotídeo Guanina Rho , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Quinases Associadas a rho/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo
7.
Blood ; 118(18): 4967-76, 2011 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-21715309

RESUMO

Genomewide association meta-analysis studies have identified > 100 independent genetic loci associated with blood cell indices, including volume and count of platelets and erythrocytes. Although several of these loci encode known regulators of hematopoiesis, the mechanism by which most sequence variants exert their effect on blood cell formation remains elusive. An example is the Rho guanine nucleotide exchange factor, ARHGEF3, which was previously implicated by genomewide association meta-analysis studies in bone cell biology. Here, we report on the unexpected role of ARHGEF3 in regulation of iron uptake and erythroid cell maturation. Although early erythroid differentiation progressed normally, silencing of arhgef3 in Danio rerio resulted in microcytic and hypochromic anemia. This was rescued by intracellular supplementation of iron, showing that arhgef3-depleted erythroid cells are fully capable of hemoglobinization. Disruption of the arhgef3 target, RhoA, also produced severe anemia, which was, again, corrected by iron injection. Moreover, silencing of ARHGEF3 in erythromyeloblastoid cells K562 showed that the uptake of transferrin was severely impaired. Taken together, this is the first study to provide evidence for ARHGEF3 being a regulator of transferrin uptake in erythroid cells, through activation of RHOA.


Assuntos
Inativação Gênica/fisiologia , Fatores de Troca do Nucleotídeo Guanina/genética , Fatores de Troca do Nucleotídeo Guanina/fisiologia , Ferro/metabolismo , Anemia Ferropriva/genética , Animais , Animais Geneticamente Modificados , Modelos Animais de Doenças , Embrião não Mamífero , Fatores de Troca do Nucleotídeo Guanina/antagonistas & inibidores , Humanos , Células K562 , Redes e Vias Metabólicas/genética , Receptores da Transferrina/genética , Receptores da Transferrina/metabolismo , Receptores da Transferrina/fisiologia , Fatores de Troca de Nucleotídeo Guanina Rho , Peixe-Zebra , Proteína rhoA de Ligação ao GTP/metabolismo
8.
J Biomol Screen ; 14(2): 161-72, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19196702

RESUMO

Guanine nucleotide exchange factors (GEFs) stimulate guanine nucleotide exchange and the subsequent activation of Rho-family proteins in response to extracellular stimuli acting upon cytokine, tyrosine kinase, adhesion, integrin, and G-protein-coupled receptors (GPCRs). Upon Rho activation, several downstream events occur, such as morphological and cytoskeletal changes, motility, growth, survival, and gene transcription. The leukemia-associated RhoGEF (LARG) is a member of the regulators of G-protein signaling homology domain (RH) family of GEFs originally identified as a result of chromosomal translocation in acute myeloid leukemia. Using a novel fluorescence polarization guanine nucleotide-binding assay using BODIPY-Texas Red-GTPgammaS (BODIPY-TR-GTPgammaS), the authors performed a 10,000-compound high-throughput screen for inhibitors of LARG-stimulated RhoA nucleotide binding. Five compounds identified from the high-throughput screen were confirmed in a nonfluorescent radioactive guanine nucleotide-binding assay measuring LARG-stimulated [( 35)S] GTPgammaS binding to RhoA, thus ruling out nonspecific fluorescent effects. All 5 compounds selectively inhibited LARG-stimulated RhoA [( 35)S] GTPgammaS binding but had little to no effect on RhoA or Galpha( o) [(35)S] GTPgammaS binding. Therefore, these 5 compounds should serve as promising starting points for the development of small-molecule inhibitors of LARG-mediated nucleotide exchange as both pharmacological tools and therapeutics. In addition, the fluorescence polarization guanine nucleotide-binding assay described here should serve as a useful approach for both high-throughput screening and general biological applications.


Assuntos
Avaliação Pré-Clínica de Medicamentos/métodos , Inibidores do Crescimento/isolamento & purificação , Fatores de Troca do Nucleotídeo Guanina/antagonistas & inibidores , Fatores de Troca do Nucleotídeo Guanina/fisiologia , Bibliotecas de Moléculas Pequenas/análise , Proteína rhoA de Ligação ao GTP/metabolismo , Algoritmos , Polarização de Fluorescência/métodos , Inibidores do Crescimento/farmacologia , Fatores de Troca do Nucleotídeo Guanina/química , Fatores de Troca do Nucleotídeo Guanina/isolamento & purificação , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/farmacocinética , Humanos , Modelos Biológicos , Ligação Proteica/efeitos dos fármacos , Fatores de Troca de Nucleotídeo Guanina Rho , Relação Estrutura-Atividade , Especificidade por Substrato , Proteína rhoA de Ligação ao GTP/antagonistas & inibidores
9.
J Biol Chem ; 282(14): 10210-22, 2007 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-17298951

RESUMO

This study investigated the involvement of Galpha(13) switch region I (SRI) in protease-activated receptor 1 (PAR1)-mediated platelet function and signaling. To this end, myristoylated peptides representing the Galpha(13) SRI (Myr-G(13)SRI(pep)) and its random counterpart were evaluated for their effects on PAR1 activation. Initial studies demonstrated that Myr-G(13)SRI(pep) and Myr-G(13)SRI(Random-pep) were equally taken up by human platelets and did not interfere with PAR1-ligand interaction. Subsequent experiments revealed that Myr-G(13)SRI(pep) specifically bound to platelet RhoA guanine nucleotide exchange factor (p115RhoGEF) and blocked PAR1-mediated RhoA activation in platelets and human embryonic kidney cells. These results suggest a direct interaction of Galpha(13) SRI with p115RhoGEF and a mechanism for Myr-G(13)SRI(pep) inhibition of RhoA activation. Platelet function studies demonstrated that Myr-G(13)SRI(pep) specifically inhibited PAR1-stimulated shape change, aggregation, and secretion in a dose-dependent manner but did not inhibit platelet activation induced by either ADP or A23187. It was also found that Myr-G(13)SRI(pep) inhibited low dose, but not high dose, thrombin-induced aggregation. Additional experiments showed that PAR1-mediated calcium mobilization was partially blocked by Myr-G(13)SRI(pep) but not by the Rho kinase inhibitor Y-27632. Finally, Myr-G(13)SRI(pep) effectively inhibited PAR1-induced stress fiber formation and cell contraction in endothelial cells. Collectively, these results suggest the following: 1) interaction of Galpha(13) SRI with p115RhoGEF is required for G(13)-mediated RhoA activation in platelets; 2) signaling through the G(13) pathway is critical for PAR1-mediated human platelet functional changes and low dose thrombin-induced aggregation; and 3) G(13) signaling elicits calcium mobilization in human platelets through a Rho kinase-independent mechanism.


Assuntos
Plaquetas/metabolismo , Sinalização do Cálcio/fisiologia , Forma Celular/fisiologia , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/metabolismo , Peptídeos/metabolismo , Agregação Plaquetária/fisiologia , Difosfato de Adenosina/farmacologia , Plaquetas/citologia , Calcimicina/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Células Cultivadas , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/farmacologia , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Hemostáticos/farmacologia , Humanos , Ionóforos/farmacologia , Rim/citologia , Rim/metabolismo , Peptídeos/farmacologia , Agregação Plaquetária/efeitos dos fármacos , Receptor PAR-1 , Fatores de Troca de Nucleotídeo Guanina Rho , Trombina/farmacologia , Proteína rhoA de Ligação ao GTP/metabolismo
10.
Synapse ; 59(7): 394-402, 2006 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-16485262

RESUMO

The excitatory amino acid transporters (EAATs) are a family of plasma membrane proteins that maintain synaptic glutamate concentration by removing glutamate from the synaptic cleft. EAATs are expressed by glia (EAAT1 and EAAT2) and neurons (EAAT3 and EAAT4) throughout the brain. Glutamate reuptake is regulated, in part, by EAAT-interacting proteins that modulate subcellular localization and glutamate transport activity of the EAATs. Several lines of investigation support the hypothesis of glutamatergic abnormalities in schizophrenia. Previous work in our laboratory demonstrated increased expression of EAAT1 and EAAT2 transcripts in the thalamus, suggesting that alterations in synaptic glutamate levels may contribute to the pathophysiology of schizophrenia. Since EAAT-interacting proteins regulate EAAT function, directly impacting glutamatergic neurotransmission, we hypothesized that expression of EAAT-interacting proteins may also be altered in schizophrenia. Using in situ hybridization in subjects with schizophrenia and a comparison group, we detected increased expression of JWA and KIAA0302, molecules that regulate EAAT3 and EAAT4, respectively, in the thalamus in schizophrenia. In contrast, we did not find changes in the expression of transcripts for the EAAT2 and EAAT4 regulatory proteins GPS-1 and ARHGEF11. To address prior antipsychotic treatment in our schizophrenic subjects, we treated rats with haloperidol and clozapine for 4 weeks, and found changes in transcript expression of the EAAT-interacting proteins in clozapine-, but not haloperidol-, treated rats. These findings suggest that proteins associated with the regulation of glutamate reuptake may be abnormal in this illness, supporting the hypothesis of altered thalamic glutamatergic neurotransmission in schizophrenia.


Assuntos
Proteínas de Transporte de Glutamato da Membrana Plasmática/biossíntese , Esquizofrenia/fisiopatologia , Tálamo/metabolismo , Idoso , Idoso de 80 Anos ou mais , Animais , Antipsicóticos/farmacologia , Complexo do Signalossomo COP9 , Clozapina/farmacologia , Feminino , Proteínas de Transporte de Glutamato da Membrana Plasmática/efeitos dos fármacos , Ácido Glutâmico/metabolismo , Fatores de Troca do Nucleotídeo Guanina/biossíntese , Haloperidol/farmacologia , Proteínas de Choque Térmico/biossíntese , Humanos , Processamento de Imagem Assistida por Computador , Hibridização In Situ , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Masculino , Proteínas de Membrana Transportadoras , Pessoa de Meia-Idade , RNA Mensageiro/análise , Ratos , Ratos Sprague-Dawley , Fatores de Troca de Nucleotídeo Guanina Rho , Esquizofrenia/metabolismo , Transcrição Gênica/efeitos dos fármacos
11.
Biochemistry ; 44(40): 13282-90, 2005 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-16201754

RESUMO

The Rho family GTPase Cdc42 functions as a molecular switch and controls many fundamental cellular processes such as cytoskeletal regulation, cell polarity, and vesicular trafficking. Guanine nucleotide exchange factors of the Dbl family activate Cdc42 and other Rho GTPases by catalyzing the removal of bound GDP, allowing for GTP loading, and subsequent effector recognition ultimately leading to downstream signaling events. Analysis of existing structural data reveals that the Dbl exchange factor intersectin engages a strictly conserved GTPase residue of Cdc42 (tyrosine 32) in a unique mode with respect to all other visualized exchange factor-Rho GTPase interfaces. To investigate this differential binding architecture, we analyzed the role of tyrosine 32 of Cdc42 in binding, and stimulation by Dbl family exchange factors. Deletion of the hydroxyl side chain of tyrosine 32 substantially increases the affinity of Cdc42 for intersectin, yet severely cripples interaction with Dbs, a normally potent exchange factor of Cdc42. Moreover, Cdc42(Y32F) is exclusively activated by intersectin, while virtually unresponsive to other Cdc42-activating exchange factors in vitro and in vivo. Further, the structural determinants unique to intersectin, which permit selective recognition and concomitant stimulation of Cdc42(Y32F), have been defined. Cdc42 and other individual Rho GTPases receive input stimulatory signals from a multitude of Dbl exchange factors, and therefore, Cdc42(Y32F) could act as a valuable reagent for understanding the specific influence of ITSN on Cdc42-mediated signaling phenomena.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/química , Mutação , Proteína cdc42 de Ligação ao GTP/genética , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Sequência de Aminoácidos , Animais , Catálise , Cristalografia por Raios X , Citoesqueleto/metabolismo , DNA Complementar/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Guanina/química , Fatores de Troca do Nucleotídeo Guanina/química , Guanosina Difosfato/química , Guanosina Trifosfato/química , Células HeLa , Humanos , Cinética , Camundongos , Microscopia de Fluorescência , Modelos Moleculares , Dados de Sequência Molecular , Plasmídeos/metabolismo , Mutação Puntual , Ligação Proteica , Conformação Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-vav/química , Fatores de Troca de Nucleotídeo Guanina Rho , Transdução de Sinais , Ressonância de Plasmônio de Superfície , Fatores de Tempo , Tirosina/química , Proteínas rho de Ligação ao GTP/química
13.
J Biol Chem ; 278(50): 50203-11, 2003 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-12975366

RESUMO

The betagamma subunits of G proteins modulate inwardly rectifying potassium (GIRK) channels through direct interactions. Although GIRK currents are stimulated by mammalian Gbetagamma subunits, we show that they were inhibited by the yeast Gbetagamma (Ste4/Ste18) subunits. A chimera between the yeast and the mammalian Gbeta1 subunits (ymbeta) stimulated or inhibited GIRK currents, depending on whether it was co-expressed with mammalian or yeast Ggamma subunits, respectively. This result underscores the critical functional influence of the Ggamma subunits on the effectiveness of the Gbetagamma complex. A series of chimeras between Ggamma2 and the yeast Ggamma revealed that the C-terminal half of the Ggamma2 subunit is required for channel activation by the Gbetagamma complex. Point mutations of Ggamma2 to the corresponding yeast Ggamma residues identified several amino acids that reduced significantly the ability of Gbetagamma to stimulate channel activity, an effect that was not due to improper association with Gbeta. Most of the identified critical Ggamma residues clustered together, forming an intricate network of interactions with the Gbeta subunit, defining an interaction surface of the Gbetagamma complex with GIRK channels. These results show for the first time a functional role for Ggamma in the effector role of Gbetagamma.


Assuntos
Subunidades beta da Proteína de Ligação ao GTP/química , Subunidades gama da Proteína de Ligação ao GTP/química , Canais de Potássio Corretores do Fluxo de Internalização , Canais de Potássio/química , Sequência de Aminoácidos , Aminoácidos/química , Animais , Membrana Celular/metabolismo , DNA Complementar/metabolismo , Dimerização , Eletrofisiologia , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G , Subunidades beta da Proteína de Ligação ao GTP/metabolismo , Subunidades gama da Proteína de Ligação ao GTP/metabolismo , Fatores de Troca do Nucleotídeo Guanina , Immunoblotting , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Oócitos/metabolismo , Plasmídeos/metabolismo , Canais de Potássio/metabolismo , Testes de Precipitina , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes/metabolismo , Fatores de Troca de Nucleotídeo Guanina Rho , Homologia de Sequência de Aminoácidos , Tripsina/farmacologia , Xenopus laevis
14.
J Biol Chem ; 278(32): 29420-34, 2003 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-12748184

RESUMO

In this study we have examined CD44 (a hyaluronan (HA) receptor) interaction with a RhoA-specific guanine nucleotide exchange factor (p115RhoGEF) in human metastatic breast tumor cells (MDA-MB-231 cell line). Immunoprecipitation and immunoblot analyses indicate that both CD44 and p115RhoGEF are expressed in MDA-MB-231 cells and that these two proteins are physically associated as a complex in vivo. The binding of HA to MDA-MB-231 cells stimulates p115RhoGEF-mediated RhoA signaling and Rho kinase (ROK) activity, which, in turn, increases serine/threonine phosphorylation of the adaptor protein, Gab-1 (Grb2-associated binder-1). Phosphorylated Gab-1 promotes PI 3-kinase recruitment to CD44v3. Subsequently, PI 3-kinase is activated (in particular, alpha, beta, gamma forms but not the delta form of the p110 catalytic subunit), AKT signaling occurs, the cytokine (macrophage-colony stimulating factor (M-CSF)) is produced, and tumor cell-specific phenotypes (e.g. tumor cell growth, survival and invasion) are up-regulated. Our results also demonstrate that HA/CD44-mediated oncogenic events (e.g. AKT activation, M-CSF production and breast tumor cell-specific phenotypes) can be effectively blocked by a PI 3-kinase inhibitor (LY294002). Finally, we have found that overexpression of a dominant-negative form of ROK (by transfection of MBA-MD-231 cells with the Rho-binding domain cDNA of ROK) not only inhibits HA/CD44-mediated RhoA-ROK activation and Gab-1 phosphorylation but also down-regulates oncogenic signaling events (e.g. Gab-1.PI 3-kinase-CD44v3 association, PI 3-kinase-mediated AKT activation, and M-CSF production) and tumor cell behaviors (e.g. cell growth, survival, and invasion). Taken together, these findings strongly suggest that CD44 interaction with p115RhoGEF and ROK plays a pivotal role in promoting Gab-1 phosphorylation leading to Gab-1.PI 3-kinase membrane localization, AKT signaling, and cytokine (M-CSF) production during HA-mediated breast cancer progression.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Fatores de Troca do Nucleotídeo Guanina/química , Receptores de Hialuronatos/química , Ácido Hialurônico/farmacologia , Fator Estimulador de Colônias de Macrófagos/biossíntese , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas/metabolismo , Trifosfato de Adenosina/metabolismo , Adjuvantes Imunológicos/farmacologia , Animais , Neoplasias da Mama/patologia , Células COS , Divisão Celular , Sobrevivência Celular , Clonagem Molecular , Citoplasma/metabolismo , DNA Complementar/metabolismo , Progressão da Doença , Relação Dose-Resposta a Droga , Regulação para Baixo , Inibidores Enzimáticos/farmacologia , Escherichia coli/metabolismo , Proteína Adaptadora GRB2 , Vetores Genéticos , Proteínas de Fluorescência Verde , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Humanos , Receptores de Hialuronatos/biossíntese , Receptores de Hialuronatos/metabolismo , Immunoblotting , Peptídeos e Proteínas de Sinalização Intracelular , Cinética , Proteínas Luminescentes/metabolismo , Fator Estimulador de Colônias de Macrófagos/metabolismo , Modelos Biológicos , Invasividade Neoplásica , Fenótipo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Fosforilação , Testes de Precipitina , Ligação Proteica , Estrutura Terciária de Proteína , Fatores de Troca de Nucleotídeo Guanina Rho , Transdução de Sinais , Fatores de Tempo , Transfecção , Células Tumorais Cultivadas , Regulação para Cima , Quinases Associadas a rho
15.
Gene ; 282(1-2): 237-46, 2002 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-11814696

RESUMO

Members of the Dbl family of guanine nucleotide exchange factors (GEFs) have important roles in the organization of actin-based cytoskeletal structures of a wide variety of cell types. Through the activation of members of the Rho family of GTP signaling molecules, these exchange factors elicit cytoskeletal alterations that allow cellular remodeling. As important regulators of RhoGTPase activity, members of this family are candidates for mediating the RhoGTPase activation and cytoskeletal changes that occur during cardiac development and during the myocardial response to hypertrophic stimuli. In this study, we characterize a novel human gene that is expressed in skeletal and cardiac muscle and has putative functional domains similar to those found in members of both the Dbl family of GEFs and the titin family of myosin light chain kinases (MLCK). The cDNA sequence of this gene, which has been designated Obscurin-myosin light chain kinase (Obscurin-MLCK), would be predicted to encode for at least 68 immunoglobulin domains, two fibronectin domains, one calcium/calmodulin binding domain, a RhoGTP exchange factor domain, and two serine-threonine kinase domains. The combination of the putative Rho GEF and two kinase domains has not been noted in any other members of the titin or Dbl families. Alternative splicing allows the generation of a number of unique Obscurin-MLCK isoforms that contain various combinations of the functional domains. One group of isoforms is comparable to Unc-89, a Caenorhabditis elegans sarcomere-associated protein, in that they contain a putative RhoGEF domain and multiple immunoglobulin repeats. Other isoforms more closely resemble MLCK, containing one or both of the putative carboxy-terminal serine-threonine kinase domains. The modular nature of the Obscurin-MLCK isoforms indicates that it may have an array of functions important to cardiac and skeletal muscle physiology.


Assuntos
Fatores de Troca do Nucleotídeo Guanina/genética , Proteínas Musculares/genética , Quinase de Cadeia Leve de Miosina/genética , Processamento Alternativo , Sequência de Aminoácidos , Sítios de Ligação/genética , Mapeamento Cromossômico , Cromossomos Humanos Par 1/genética , DNA Complementar/química , DNA Complementar/genética , Expressão Gênica , Genes/genética , Humanos , Dados de Sequência Molecular , Filogenia , Proteínas Serina-Treonina Quinases , Fatores de Troca de Nucleotídeo Guanina Rho , Alinhamento de Sequência , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos
16.
J Biol Chem ; 277(14): 12463-73, 2002 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-11799111

RESUMO

A recently identified family of guanine nucleotide exchange factors for Rho that includes PDZ-RhoGEF, LARG, and p115RhoGEF exhibits a unique structural feature consisting in the presence of area of similarity to regulators of G protein signaling (RGS). This RGS-like (RGL) domain provides a structural motif by which heterotrimeric G protein alpha subunits of the Galpha(12) family can bind and regulate the activity of RhoGEFs. Hence, these newly discovered RGL domain-containing RhoGEFs provide a direct link from Galpha(12) and Galpha(13) to Rho. Recently available data suggest, however, that tyrosine kinases can regulate the ability of G protein-coupled receptors (GPCRs) to stimulate Rho, although the underlying molecular mechanisms are still unknown. Here, we found that the activation of thrombin receptors endogenously expressed in HEK-293T cells leads to a remarkable increase in the levels of GTP-bound Rho within 1 min (11-fold) and a more limited but sustained activation (4-fold) thereafter, which lasts even for several hours. Interestingly, tyrosine kinase inhibitors did not affect the early phase of Rho activation, immediately after thrombin addition, but diminished the levels of GTP-bound Rho during the delayed phase. As thrombin receptors stimulate focal adhesion kinase (FAK) potently, we explored whether this non-receptor tyrosine kinase participates in the activation of Rho by GPCRs. We obtained evidence that FAK can be activated by thrombin, Galpha(12), Galpha(13), and Galpha(q) through both Rho-dependent and Rho-independent mechanisms and that PDZ-RhoGEF and LARG can in turn be tyrosine-phosphorylated through FAK in response to thrombin, thereby enhancing the activation of Rho in vivo. These data indicate that FAK may act as a component of a positive feedback loop that results in the sustained activation of Rho by GPCRs, thus providing evidence of the existence of a novel biochemical route by which tyrosine kinases may regulate the activity of Rho through the tyrosine phosphorylation of RGL-containing RhoGEFs.


Assuntos
Proteínas de Ligação ao GTP/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Proteínas do Tecido Nervoso , Tirosina/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Western Blotting , Linhagem Celular , DNA Complementar/metabolismo , Dimerização , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Genisteína/farmacologia , Humanos , Modelos Biológicos , Mutação , Fosforilação , Plasmídeos/metabolismo , Testes de Precipitina , Ligação Proteica , Estrutura Terciária de Proteína , Fatores de Troca de Nucleotídeo Guanina Rho , Trombina/metabolismo , Fatores de Tempo , Transfecção
17.
J Cell Biol ; 154(1): 123-36, 2001 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-11448995

RESUMO

Vertebrate-striated muscle is assumed to owe its remarkable order to the molecular ruler functions of the giant modular signaling proteins, titin and nebulin. It was believed that these two proteins represented unique results of protein evolution in vertebrate muscle. In this paper we report the identification of a third giant protein from vertebrate muscle, obscurin, encoded on chromosome 1q42. Obscurin is approximately 800 kD and is expressed specifically in skeletal and cardiac muscle. The complete cDNA sequence of obscurin reveals a modular architecture, consisting of >67 intracellular immunoglobulin (Ig)- or fibronectin-3-like domains with multiple splice variants. A large region of obscurin shows a modular architecture of tandem Ig domains reminiscent of the elastic region of titin. The COOH-terminal region of obscurin interacts via two specific Ig-like domains with the NH(2)-terminal Z-disk region of titin. Both proteins coassemble during myofibrillogenesis. During the progression of myofibrillogenesis, all obscurin epitopes become detectable at the M band. The presence of a calmodulin-binding IQ motif, and a Rho guanine nucleotide exchange factor domain in the COOH-terminal region suggest that obscurin is involved in Ca(2+)/calmodulin, as well as G protein-coupled signal transduction in the sarcomere.


Assuntos
Fatores de Troca do Nucleotídeo Guanina/química , Fatores de Troca do Nucleotídeo Guanina/fisiologia , Proteínas Musculares/química , Proteínas Musculares/fisiologia , Sarcômeros/química , Proteínas rho de Ligação ao GTP/química , Proteínas rho de Ligação ao GTP/fisiologia , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Western Blotting , Calmodulina/metabolismo , Adesão Celular , Células Cultivadas , Embrião de Galinha , Cromossomos Humanos Par 1 , Clonagem Molecular , DNA Complementar/metabolismo , Epitopos , Biblioteca Gênica , Humanos , Imunoglobulinas/metabolismo , Microscopia Confocal , Modelos Genéticos , Dados de Sequência Molecular , Músculo Esquelético/metabolismo , Miocárdio/metabolismo , Filogenia , Ligação Proteica , Proteínas Serina-Treonina Quinases , Estrutura Terciária de Proteína , Ratos , Ratos Wistar , Fatores de Troca de Nucleotídeo Guanina Rho , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Distribuição Tecidual , Transfecção
18.
Cell Mol Neurobiol ; 21(6): 783-97, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12043848

RESUMO

The mammalian acetylcholinesterase (ACHE) locus was investigated using computational predictive methods and experiments of reverse transcription polymerase chain reaction (RT-PCR). Computational analysis identified two genes downstream to ACHE, an inversely oriented arsenite resistance gene homologue (ARS), and a novel previously unidentified gene (PIX), co-oriented with ACHE. Experimental evidence shows coregulation of murine ACHE and ARS following confined swim, indicating coordinated locus response to stress, that is possibly mediated by altered cholinergic neurotransmission.


Assuntos
Acetilcolina/biossíntese , Acetilcolinesterase/genética , Encéfalo/enzimologia , Regulação Enzimológica da Expressão Gênica/genética , Neurônios/enzimologia , Estresse Fisiológico/genética , Transmissão Sináptica/genética , Acetilcolinesterase/metabolismo , Processamento Alternativo/genética , Animais , ATPases Transportadoras de Arsenito , Proteínas de Ciclo Celular/genética , Mapeamento Cromossômico , DNA Complementar/genética , DNA Complementar/isolamento & purificação , Genes/genética , Genoma , Fatores de Troca do Nucleotídeo Guanina/genética , Bombas de Íon/genética , Camundongos , Dados de Sequência Molecular , Complexos Multienzimáticos/genética , Isoformas de Proteínas/genética , RNA Mensageiro/genética , RNA Mensageiro/isolamento & purificação , Fatores de Troca de Nucleotídeo Guanina Rho , Homologia de Sequência de Aminoácidos , Homologia de Sequência do Ácido Nucleico , Estresse Fisiológico/metabolismo , Estresse Fisiológico/fisiopatologia
19.
FEBS Lett ; 474(1): 16-22, 2000 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-10828443

RESUMO

We have cloned a novel unconventional myosin gene myoM in Dictyostelium. Phylogenetic analysis of the motor domain indicated that MyoM does not belong to any known subclass of the myosin superfamily. Following the motor domain, two calmodulin-binding IQ motifs, a putative coiled-coil region, and a Pro, Ser and Thr-rich domain, lies a combination of dbl homology and pleckstrin homology domains. These are conserved in Rho GDP/GTP exchange factors (RhoGEFs). We have identified for the first time the RhoGEF domain in the myosin sequences. The growth and terminal developmental phenotype of Dictyostelium cells were not affected by the myoM(-) mutation. Green fluorescent protein-tagged MyoM, however, accumulated at crown-shaped projections and membranes of phase lucent vesicles in growing cells, suggesting its possible roles in macropinocytosis.


Assuntos
Dictyostelium/química , Fatores de Troca do Nucleotídeo Guanina/química , Miosinas/química , Proteínas de Protozoários , Sequência de Aminoácidos , Animais , Sequência de Bases , Sítios de Ligação , Calmodulina/metabolismo , Dictyostelium/genética , Dictyostelium/ultraestrutura , Proteínas de Fluorescência Verde , Humanos , Indicadores e Reagentes , Proteínas Luminescentes , Dados de Sequência Molecular , Miosinas/análise , Miosinas/genética , Filogenia , Fatores de Troca de Nucleotídeo Guanina Rho , Alinhamento de Sequência
20.
Proc Natl Acad Sci U S A ; 97(5): 2145-50, 2000 Feb 29.
Artigo em Inglês | MEDLINE | ID: mdl-10681437

RESUMO

We have identified a gene at 11q23, telomeric to MLL, that encodes a guanine nucleotide exchange factor (GEF). This gene is transcribed into a 9.5-kb mRNA containing a 4.6-kb ORF. By Northern analysis, it was found to be expressed in all human tissues examined including peripheral blood leukocytes, spleen, prostate, testis, ovary, small intestine, colon, and minimally in thymus. Analysis of the predicted protein sequence indicates that it has strong homology to several members of the family of Rho GEFs that includes such oncogenes as Dbl, Vav, Tiam, and Bcr. A patient with primary acute myeloid leukemia (AML) and a karyotype of 51,XY,+8,+19,+3mar was found to have the 5' end of MLL at exon 6 fused in-frame with the 3' end of almost the entire ORF of this gene, which we named LARG for leukemia-associated Rho GEF. Transcriptional orientation of both genes at 11q23 is from centromere to telomere, consistent with other data that suggest the MLL-LARG fusion resulted from an interstitial deletion rather than a balanced translocation. LARG does not appear to have any homology with other MLL partner genes reported thus far. Thus, LARG represents an additional member of the GEF family and a novel MLL fusion partner in acute myeloid leukemia.


Assuntos
Cromossomos Humanos Par 11 , Proteínas de Ligação a DNA/genética , Fatores de Troca do Nucleotídeo Guanina/genética , Leucemia Mielomonocítica Aguda/genética , Proto-Oncogenes , Fatores de Transcrição , Adulto , Sequência de Aminoácidos , Fusão Gênica Artificial , Sequência de Bases , Mapeamento Cromossômico , DNA Complementar , Histona-Lisina N-Metiltransferase , Humanos , Masculino , Dados de Sequência Molecular , Proteína de Leucina Linfoide-Mieloide , Fatores de Troca de Nucleotídeo Guanina Rho
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA