Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 254
Filtrar
1.
Epigenetics ; 19(1): 2299044, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38154055

RESUMO

Early detection of hepatocellular carcinoma (HCC) can greatly improve the survival rate of patients. We aimed to develop a novel marker panel based on cell-free DNA (cfDNA) methylation for the detection of HCC. The differentially methylated CpG sites (DMCs) specific for HCC blood diagnosis were selected from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases, then validated by the whole genome bisulphite sequencing (WGBS) of 12 paired HCC and paracancerous tissues. The clinical performance of the panel was evaluated using tissue samples [32 HCC, chronic liver disease (CLD), and healthy individuals] and plasma cohorts (173 HCC, 199 CLD, and 98 healthy individuals). The combination of G protein subunit beta 4 (GNB4) and Riplet had the optimal area under the curve (AUC) in seven candidates through TCGA, GEO, and WGBS analyses. In tissue validation, the GNB4 and Riplet showed an AUC of 100% with a sensitivity and specificity of 100% for detecting any-stage HCC. In plasma, it demonstrated a high sensitivity of 84.39% at 91.92% specificity, with an AUC of 92.51% for detecting any-stage HCC. The dual-marker panel had a higher sensitivity of 78.26% for stage I HCC than alpha-fetoprotein (AFP) of 47.83%, and a high sensitivity of 70.27% for detecting a single tumour (size ≤3 cm). In conclusion, we developed a novel dual-marker panel that demonstrates high accuracy in detecting HCC, surpassing the performance of AFP testing.


Assuntos
Carcinoma Hepatocelular , Subunidades beta da Proteína de Ligação ao GTP , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/diagnóstico , Carcinoma Hepatocelular/genética , alfa-Fetoproteínas/análise , alfa-Fetoproteínas/genética , alfa-Fetoproteínas/metabolismo , Neoplasias Hepáticas/diagnóstico , Neoplasias Hepáticas/genética , Biomarcadores Tumorais/metabolismo , Metilação de DNA , Subunidades beta da Proteína de Ligação ao GTP/genética , Subunidades beta da Proteína de Ligação ao GTP/metabolismo
2.
BMC Med ; 21(1): 134, 2023 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-37016382

RESUMO

BACKGROUND: Helicobacter pylori (H. pylori) infection causes aberrant DNA methylation and contributes to the risk of gastric cancer (GC). Guanine nucleotide-binding protein subunit beta-4 (GNB4) is involved in various tumorigenic processes. We found an aberrant methylation level of GNB4 in H. pylori-induced GC in our previous bioinformatic analysis; however, its expression and underlying molecular mechanisms are poorly understood. METHODS: The expression, underlying signaling pathways, and clinical significance of GNB4 were analyzed in a local cohort of 107 patients with GC and several public databases. H. pylori infection was induced in in vitro and in vivo models. Methylation-specific PCR, pyrosequencing, and mass spectrometry analysis were used to detect changes in methylation levels. GNB4, TET1, and YAP1 were overexpressed or knocked down in GC cell lines. We performed gain- and loss-of-function experiments, including CCK-8, EdU, colony formation, transwell migration, and invasion assays. Nude mice were injected with genetically manipulated GC cells, and the growth of xenograft tumors and metastases was measured. Real-time quantitative PCR, western blotting, immunofluorescence, immunohistochemistry, chromatin immunoprecipitation, and co-immunoprecipitation experiments were performed to elucidate the underlying molecular mechanisms. RESULTS: GNB4 expression was significantly upregulated in GC and correlated with aggressive clinical characteristics and poor prognosis. Increased levels of GNB4 were associated with shorter survival times. Infection with H. pylori strains 26695 and SS1 induced GNB4 mRNA and protein expression in GC cell lines and mice. Additionally, silencing of GNB4 blocked the pro-proliferative, metastatic, and invasive ability of H. pylori in GC cells. H. pylori infection remarkably decreased the methylation level of the GNB4 promoter region, particularly at the CpG#5 site (chr3:179451746-179451745). H. pylori infection upregulated TET1 expression via activation of the NF-κB. TET binds to the GNB4 promoter region which undergoes demethylation modification. Functionally, we identified that GNB4 induced oncogenic behaviors of tumors via the Hippo-YAP1 pathway in both in vitro and in vivo models. CONCLUSIONS: Our findings demonstrate that H. pylori infection activates the NF-κB-TET1-GNB4 demethylation-YAP1 axis, which may be a potential therapeutic target for GC.


Assuntos
Subunidades beta da Proteína de Ligação ao GTP , Helicobacter pylori , Neoplasias Gástricas , Humanos , Camundongos , Animais , NF-kappa B/genética , NF-kappa B/metabolismo , Helicobacter pylori/metabolismo , Camundongos Nus , Carcinogênese/genética , Neoplasias Gástricas/genética , Desmetilação , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Oxigenases de Função Mista/genética , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Subunidades beta da Proteína de Ligação ao GTP/genética , Subunidades beta da Proteína de Ligação ao GTP/metabolismo
3.
Science ; 379(6639): 1352-1358, 2023 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-36996198

RESUMO

Glycine is a major neurotransmitter involved in several fundamental neuronal processes. The identity of the metabotropic receptor mediating slow neuromodulatory effects of glycine is unknown. We identified an orphan G protein-coupled receptor, GPR158, as a metabotropic glycine receptor (mGlyR). Glycine and a related modulator, taurine, directly bind to a Cache domain of GPR158, and this event inhibits the activity of the intracellular signaling complex regulator of G protein signaling 7-G protein ß5 (RGS7-Gß5), which is associated with the receptor. Glycine signals through mGlyR to inhibit production of the second messenger adenosine 3',5'-monophosphate. We further show that glycine, but not taurine, acts through mGlyR to regulate neuronal excitability in cortical neurons. These results identify a major neuromodulatory system involved in mediating metabotropic effects of glycine, with implications for understanding cognition and affective states.


Assuntos
Glicina , Receptores Acoplados a Proteínas G , Receptores de Glicina , Glicina/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Glicina/química , Receptores de Glicina/genética , Receptores de Glicina/metabolismo , Transdução de Sinais , Humanos , Células HEK293 , Subunidades beta da Proteína de Ligação ao GTP/metabolismo , Proteínas RGS/metabolismo , Domínios Proteicos
4.
J Biol Chem ; 299(3): 102924, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36736897

RESUMO

G protein-coupled receptors (GPCRs) initiate an array of intracellular signaling programs by activating heterotrimeric G proteins (Gα and Gßγ subunits). Therefore, G protein modifiers are well positioned to shape GPCR pharmacology. A few members of the potassium channel tetramerization domain (KCTD) protein family have been found to adjust G protein signaling through interaction with Gßγ. However, comprehensive details on the KCTD interaction with Gßγ remain unresolved. Here, we report that nearly all the 25 KCTD proteins interact with Gßγ. In this study, we screened Gßγ interaction capacity across the entire KCTD family using two parallel approaches. In a live cell bioluminescence resonance energy transfer-based assay, we find that roughly half of KCTD proteins interact with Gßγ in an agonist-induced fashion, whereas all KCTD proteins except two were found to interact through coimmunoprecipitation. We observed that the interaction was dependent on an amino acid hot spot in the C terminus of KCTD2, KCTD5, and KCTD17. While KCTD2 and KCTD5 require both the Bric-à-brac, Tramtrack, Broad complex domain and C-terminal regions for Gßγ interaction, we uncovered that the KCTD17 C terminus is sufficient for Gßγ interaction. Finally, we demonstrated the functional consequence of the KCTD-Gßγ interaction by examining sensitization of the adenylyl cyclase-cAMP pathway in live cells. We found that Gßγ-mediated sensitization of adenylyl cyclase 5 was blunted by KCTD. We conclude that the KCTD family broadly engages Gßγ to shape GPCR signal transmission.


Assuntos
AMP Cíclico , Subunidades beta da Proteína de Ligação ao GTP , Subunidades gama da Proteína de Ligação ao GTP , Canais de Potássio , Adenilil Ciclases/metabolismo , Subunidades beta da Proteína de Ligação ao GTP/genética , Subunidades beta da Proteína de Ligação ao GTP/metabolismo , Subunidades gama da Proteína de Ligação ao GTP/genética , Subunidades gama da Proteína de Ligação ao GTP/metabolismo , Canais de Potássio/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais , AMP Cíclico/metabolismo
5.
J Biol Chem ; 299(2): 102880, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36626984

RESUMO

Heterotrimeric G protein stimulation via G protein-coupled receptors promotes downstream proliferative signaling. Mutations can occur in Gα proteins which prevent GTP hydrolysis; this allows the G proteins to signal independently of G protein-coupled receptors and can result in various cancers, such as uveal melanoma (UM). Most UM cases harbor Q209L, Q209P, or R183C mutations in Gαq/11 proteins, rendering the proteins constitutively active (CA). Although it is generally thought that active, GTP-bound Gα subunits are dissociated from and signal independently of Gßγ, accumulating evidence indicates that some CA Gα mutants, such as Gαq/11, retain binding to Gßγ, and this interaction is necessary for signaling. Here, we demonstrate that disrupting the interaction between Gßγ and Gαq is sufficient to inhibit aberrant signaling driven by CA Gαq. Introduction of the I25A point mutation in the N-terminal α helical domain of CA Gαq to inhibit Gßγ binding, overexpression of the G protein Gαo to sequester Gßγ, and siRNA depletion of Gß subunits inhibited or abolished CA Gαq signaling to the MAPK and YAP pathways. Moreover, in HEK 293 cells and in UM cell lines, we show that Gαq-Q209P and Gαq-R183C are more sensitive to the loss of Gßγ interaction than Gαq-Q209L. Our study challenges the idea that CA Gαq/11 signals independently of Gßγ and demonstrates differential sensitivity between the Gαq-Q209L, Gαq-Q209P, and Gαq-R183C mutants.


Assuntos
Proteínas Heterotriméricas de Ligação ao GTP , Transdução de Sinais , Humanos , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/genética , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Subunidades beta da Proteína de Ligação ao GTP/genética , Subunidades beta da Proteína de Ligação ao GTP/metabolismo , Subunidades gama da Proteína de Ligação ao GTP/genética , Subunidades gama da Proteína de Ligação ao GTP/metabolismo , Guanosina Trifosfato/metabolismo , Células HEK293 , Proteínas Heterotriméricas de Ligação ao GTP/genética , Proteínas Heterotriméricas de Ligação ao GTP/metabolismo , Mutação , Transdução de Sinais/genética
6.
Cancer Sci ; 114(5): 2001-2013, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-36718954

RESUMO

G-proteins are intracellular partners of G-protein-coupled receptors. As a member of the G-protein family, GNB1 has been shown to play a pro-cancer role in lung cancer and breast cancer. However, the biological function and detailed mechanisms of GNB1 in hepatocellular carcinoma progression are unclear. In this study, we investigated the effects of GNB1 and its possible mechanism of action in hepatocellular carcinoma (HCC). The clinical significance of GNB1 was evaluated in a large cohort of HCC patients, showing that GNB1 was overexpressed in HCC compared to adjacent normal liver tissues, and increased GNB1 expression was associated with poor prognosis. We also demonstrated that GNB1 enhances cell proliferation, colony formation, and cell migration and invasion in vitro and promotes the epithelial-to-mesenchymal transition process in HCC cells. Tumor xenograft model assay confirmed the oncogenic role of GNB1 in tumorigenicity in nude mice. Activation of P38 signaling was found in the GNB1 overexpressed HCC cells. Further intervention of P38 confirmed it as an important signaling pathway for the oncogenic role of GNB1 in HCC. Moreover, co-immunoprecipitation followed by liquid chromatograph-mass spectrometry identified that GNB1 exerted oncogenic functions via the interaction of BAG2 and activated P38 signaling pathway. Together, our results reveal that GNB1 plays a pivotal oncogenic role in HCC by promoting the P38 pathway via cooperating with BAG2. GNB1 may serve as a prognostic biomarker for patients with HCC.


Assuntos
Carcinoma Hepatocelular , Subunidades beta da Proteína de Ligação ao GTP , Neoplasias Hepáticas , Animais , Camundongos , Humanos , Carcinoma Hepatocelular/patologia , Neoplasias Hepáticas/patologia , Camundongos Nus , Linhagem Celular Tumoral , Sistema de Sinalização das MAP Quinases , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica , Movimento Celular/genética , Prognóstico , Subunidades beta da Proteína de Ligação ao GTP/genética , Subunidades beta da Proteína de Ligação ao GTP/metabolismo , Subunidades beta da Proteína de Ligação ao GTP/farmacologia , Chaperonas Moleculares/metabolismo
7.
Trends Pharmacol Sci ; 44(2): 98-111, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36494204

RESUMO

G protein Gßγ subunits are key mediators of G protein-coupled receptor (GPCR) signaling under physiological and pathological conditions; their inhibitors have been tested for the treatment of human disease. Conventional wisdom is that the Gßγ complex is activated and subsequently exerts its functions at the plasma membrane (PM). Recent studies have revealed non-canonical activation of Gßγ at intracellular organelles, where the Golgi apparatus is a major locale, via translocation or local activation. Golgi-localized Gßγ activates specific signaling cascades and regulates fundamental cell processes such as membrane trafficking, proliferation, and migration. More recent studies have shown that inhibiting Golgi-compartmentalized Gßγ signaling attenuates cardiomyocyte hypertrophy and prostate tumorigenesis, indicating new therapeutic targets. We review novel activation mechanisms and non-canonical functions of Gßγ at the Golgi, and discuss potential therapeutic interventions by targeting Golgi-biased Gßγ-directed signaling.


Assuntos
Subunidades beta da Proteína de Ligação ao GTP , Subunidades gama da Proteína de Ligação ao GTP , Humanos , Subunidades beta da Proteína de Ligação ao GTP/metabolismo , Subunidades gama da Proteína de Ligação ao GTP/metabolismo , Transdução de Sinais , Complexo de Golgi/metabolismo , Membrana Celular/metabolismo
8.
J Biol Chem ; 298(12): 102618, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36272647

RESUMO

Heterotrimeric G proteins (αßγ subunits) that are activated by G protein-coupled receptors (GPCRs) mediate the biological responses of eukaryotic cells to extracellular signals. The α subunits and the tightly bound ßγ subunit complex of G proteins have been extensively studied and shown to control the activity of effector molecules. In contrast, the potential roles of the large family of γ subunits have been less studied. In this review, we focus on present knowledge about these proteins. Induced loss of individual γ subunit types in animal and plant models result in strikingly distinct phenotypes indicating that γ subtypes play important and specific roles. Consistent with these findings, downregulation or upregulation of particular γ subunit types result in various types of cancers. Clues about the mechanistic basis of γ subunit function have emerged from imaging the dynamic behavior of G protein subunits in living cells. This shows that in the basal state, G proteins are not constrained to the plasma membrane but shuttle between membranes and on receptor activation ßγ complexes translocate reversibly to internal membranes. The translocation kinetics of ßγ complexes varies widely and is determined by the membrane affinity of the associated γ subtype. On translocating, some ßγ complexes act on effectors in internal membranes. The variation in translocation kinetics determines differential sensitivity and adaptation of cells to external signals. Membrane affinity of γ subunits is thus a parsimonious and elegant mechanism that controls information flow to internal cell membranes while modulating signaling responses.


Assuntos
Subunidades beta da Proteína de Ligação ao GTP , Subunidades gama da Proteína de Ligação ao GTP , Receptores Acoplados a Proteínas G , Animais , Membrana Celular/metabolismo , Subunidades beta da Proteína de Ligação ao GTP/genética , Subunidades beta da Proteína de Ligação ao GTP/metabolismo , Subunidades gama da Proteína de Ligação ao GTP/genética , Subunidades gama da Proteína de Ligação ao GTP/metabolismo , Transdução de Sinais , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Humanos
9.
New Phytol ; 236(2): 447-463, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35766993

RESUMO

The plant hormone abscisic acid (ABA) plays crucial roles in regulation of stress responses and growth modulation. Heterotrimeric G-proteins are key mediators of ABA responses. Both ABA and G-proteins have also been implicated in intracellular redox regulation; however, the extent to which reversible protein oxidation manipulates ABA and/or G-protein signaling remains uncharacterized. To probe the role of reversible protein oxidation in plant stress response and its dependence on G-proteins, we determined the ABA-dependent reversible redoxome of wild-type and Gß-protein null mutant agb1 of Arabidopsis. We quantified 6891 uniquely oxidized cysteine-containing peptides, 923 of which show significant changes in oxidation following ABA treatment. The majority of these changes required the presence of G-proteins. Divergent pathways including primary metabolism, reactive oxygen species response, translation and photosynthesis exhibited both ABA- and G-protein-dependent redox changes, many of which occurred on proteins not previously linked to them. We report the most comprehensive ABA-dependent plant redoxome and uncover a complex network of reversible oxidations that allow ABA and G-proteins to rapidly adjust cellular signaling to adapt to changing environments. Physiological validation of a subset of these observations suggests that functional G-proteins are required to maintain intracellular redox homeostasis and fully execute plant stress responses.


Assuntos
Proteínas de Arabidopsis , Arabidopsis , Subunidades beta da Proteína de Ligação ao GTP , Proteínas Heterotriméricas de Ligação ao GTP , Ácido Abscísico/metabolismo , Arabidopsis/metabolismo , Proteínas de Arabidopsis/metabolismo , Cisteína/metabolismo , Subunidades beta da Proteína de Ligação ao GTP/genética , Subunidades beta da Proteína de Ligação ao GTP/metabolismo , Regulação da Expressão Gênica de Plantas , Proteínas Heterotriméricas de Ligação ao GTP/genética , Proteínas Heterotriméricas de Ligação ao GTP/metabolismo , Oxirredução , Reguladores de Crescimento de Plantas/metabolismo , Proteoma/metabolismo , Espécies Reativas de Oxigênio/metabolismo
10.
Science ; 375(6576): 86-91, 2022 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-34793198

RESUMO

GPR158 is an orphan G protein­coupled receptor (GPCR) highly expressed in the brain, where it controls synapse formation and function. GPR158 has also been implicated in depression, carcinogenesis, and cognition. However, the structural organization and signaling mechanisms of GPR158 are largely unknown. We used single-particle cryo­electron microscopy (cryo-EM) to determine the structures of human GPR158 alone and bound to an RGS signaling complex. The structures reveal a homodimeric organization stabilized by a pair of phospholipids and the presence of an extracellular Cache domain, an unusual ligand-binding domain in GPCRs. We further demonstrate the structural basis of GPR158 coupling to RGS7-Gß5. Together, these results provide insights into the unusual biology of orphan receptors and the formation of GPCR-RGS complexes.


Assuntos
Subunidades beta da Proteína de Ligação ao GTP/química , Proteínas RGS/química , Receptores Acoplados a Proteínas G/química , Sítios de Ligação , Microscopia Crioeletrônica , Subunidades beta da Proteína de Ligação ao GTP/metabolismo , Humanos , Ligantes , Modelos Moleculares , Fosfolipídeos/química , Ligação Proteica , Conformação Proteica , Conformação Proteica em alfa-Hélice , Domínios Proteicos , Multimerização Proteica , Subunidades Proteicas/química , Proteínas RGS/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais
11.
J Biol Chem ; 298(1): 101440, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34808208

RESUMO

Metastatic lung cancer is a major cause of death worldwide. Dissemination of cancer cells can be facilitated by various agonists within the tumor microenvironment, including by lysophosphatidic acid (LPA). We postulate that Rho guanine nucleotide exchange factors (RhoGEFs), which integrate signaling cues driving cell migration, are critical effectors in metastatic cancer. Specifically, we addressed the hypothetical role of ARHGEF17, a RhoGEF, as a potential effector of Gßγ in metastatic lung cancer cells responding to LPA. Here, we show that ARHGEF17, originally identified as a tumor endothelial marker, is involved in tumor growth and metastatic dissemination of lung cancer cells in an immunocompetent murine model. Gene expression-based analysis of lung cancer datasets showed that increased levels of ARHGEF17 correlated with reduced survival of patients with advanced-stage tumors. Cellular assays also revealed that this RhoGEF participates in the invasive and migratory responses elicited by Gi protein-coupled LPA receptors via the Gßγ subunit complex. We demonstrate that this signaling heterodimer promoted ARHGEF17 recruitment to the cell periphery and actin fibers. Moreover, Gßγ allosterically activates ARHGEF17 by the removal of inhibitory intramolecular restrictions. Taken together, our results indicate that ARHGEF17 may be a valid potential target in the treatment of metastatic lung cancer.


Assuntos
Subunidades beta da Proteína de Ligação ao GTP , Subunidades gama da Proteína de Ligação ao GTP , Neoplasias Pulmonares , Fatores de Troca de Nucleotídeo Guanina Rho , Transdução de Sinais , Animais , Movimento Celular , Progressão da Doença , Subunidades beta da Proteína de Ligação ao GTP/metabolismo , Subunidades gama da Proteína de Ligação ao GTP/metabolismo , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Camundongos , Metástase Neoplásica , Receptores de Ácidos Lisofosfatídicos/genética , Receptores de Ácidos Lisofosfatídicos/metabolismo , Fatores de Troca de Nucleotídeo Guanina Rho/genética , Fatores de Troca de Nucleotídeo Guanina Rho/metabolismo , Transdução de Sinais/fisiologia , Microambiente Tumoral
12.
Med Mol Morphol ; 54(4): 356-367, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34398348

RESUMO

Data mining on a public domain detected eight potential transcripts which were upregulated in advanced UBUCs, suggesting that they may take part in UC development or/and progression. Retrospectively, immunohistochemistry along with H-score recording was carried out to evaluate the GNB4 protein levels on tissues from UC patients. Correlations between GNB4 H-score and imperative clinicopathological factors, as well as the implication of GNB4 protein level on disease-specific and metastasis-free survivals were assessed. In UTUCs (n = 340) and UBUCs (n = 295), 170 (50.0%) and 148 (50.0%) cases, respectively, were identified to be of high GNB4 expression. The GNB4 protein levels were correlated to numerous clinicopathological features and patients' survivals. Upregulation of the GNB4 protein was significantly associated with primary tumor, nodal metastasis, histological grade, vascular invasion and mitotic rate. High GNB4 protein levels independently and significantly predicted poor disease-specific and metastasis-free in UTUC and UBUC, respectively. Ingenuity pathway analysis furthermore showed that multiple signaling pathways were enriched including 'Communication between Innate and Adaptive Immune Cells' and 'NFκB Signaling'. Our findings demonstrated that the upregulation of the GNB4 protein is an independent unfavorable prognosticator in UC. High GNB4 gene expression plays an important role in UC progression.


Assuntos
Carcinoma de Células de Transição , Subunidades beta da Proteína de Ligação ao GTP/metabolismo , Neoplasias da Bexiga Urinária , Carcinoma de Células de Transição/diagnóstico , Humanos , Imuno-Histoquímica , Subunidades Proteicas , Estudos Retrospectivos , Neoplasias da Bexiga Urinária/diagnóstico
13.
Plant Mol Biol ; 106(6): 505-520, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34176052

RESUMO

KEY MESSAGE: Gene expression analysis coupled with in-planta studies showed that specific Gßγ combination regulates plant growth and defence traits in the allotetraploid Brassica juncea. Plant heterotrimeric G-proteins regulate a wide range of responses despite their limited repertoire of core components. The roles and functional interactions between different G-protein subunits are quite perplexing, which get further complicated with polyploidy. Here, we show that the allotetraploid Brassica juncea comprises multiple homologs of G-protein genes, encoding six BjuGß and ten highly divergent BjuGγ subunit proteins, later being classified into type-A1, type-A2 and type-C Gγ proteins. The encoded BjuGß and BjuGγ proteins shared close evolutionary relationship and have retained distinct spatio-temporal expression patterns during plant developmental stages and in response to the necrotrophic pathogen, Sclerotinia sclerotiorum. RNAi based suppression of BjuGß and BjuGγ genes suggested functional overlap and selectivity of BjuGßs with three distinct BjuGγ type subunits, to regulate plant height (BjuGßγA2 and BjuGßγC), seed weight (BjuGßGγA1 and BjuGßGγC), silique size (BjuGßGγC) and pathogen response (BjuGßGγA1 and BjuGßGγC). Further, the triplicated BjuGß genes, formed due to Brassica specific whole-genome-triplication event, showed differential involvement during pathogen response, wherein overexpression of BjuGß2 displayed higher resistance to Sclerotinia infection. Taken together, our study demonstrates that multiple BjuGß and BjuGγ proteins have retained distinct spatio-temporal expression and functional selectivity to regulate specific plant growth and defence traits in the oilseed B. juncea.


Assuntos
Subunidades beta da Proteína de Ligação ao GTP/genética , Subunidades gama da Proteína de Ligação ao GTP/genética , Mostardeira/genética , Doenças das Plantas/genética , Proteínas de Plantas/genética , Poliploidia , Ascomicetos/fisiologia , Resistência à Doença/genética , Subunidades beta da Proteína de Ligação ao GTP/classificação , Subunidades beta da Proteína de Ligação ao GTP/metabolismo , Subunidades gama da Proteína de Ligação ao GTP/classificação , Subunidades gama da Proteína de Ligação ao GTP/metabolismo , Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica de Plantas , Interações Hospedeiro-Patógeno , Modelos Genéticos , Mostardeira/crescimento & desenvolvimento , Mostardeira/microbiologia , Filogenia , Doenças das Plantas/microbiologia , Proteínas de Plantas/classificação , Proteínas de Plantas/metabolismo , Plantas Geneticamente Modificadas , Ligação Proteica , Interferência de RNA
14.
J Biol Chem ; 296: 100805, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34022220

RESUMO

After activation of G protein-coupled receptors, G protein ßγ dimers may translocate from the plasma membrane to the Golgi apparatus (GA). We recently report that this translocation activates extracellular signal-regulated protein kinases 1 and 2 (ERK1/2) via PI3Kγ; however, how Gßγ-PI3Kγ activates the ERK1/2 pathway is unclear. Here, we demonstrate that chemokine receptor CXCR4 activates ADP-ribosylation factor 1 (ARF1), a small GTPase important for vesicle-mediated membrane trafficking. This activation is blocked by CRISPR-Cas9-mediated knockout of the GA-translocating Gγ9 subunit. Inducible targeting of different Gßγ dimers to the GA can directly activate ARF1. CXCR4 activation and constitutive Gßγ recruitment to the GA also enhance ARF1 translocation to the GA. We further demonstrate that pharmacological inhibition and CRISPR-Cas9-mediated knockout of PI3Kγ markedly inhibit CXCR4-mediated and Gßγ translocation-mediated ARF1 activation. We also show that depletion of ARF1 by siRNA and CRISPR-Cas9 and inhibition of GA-localized ARF1 activation abolish ERK1/2 activation by CXCR4 and Gßγ translocation to the GA and suppress prostate cancer PC3 cell migration and invasion. Collectively, our data reveal a novel function for Gßγ translocation to the GA to activate ARF1 and identify GA-localized ARF1 as an effector acting downstream of Gßγ-PI3Kγ to spatiotemporally regulate G protein-coupled receptor signaling to mitogen-activated protein kinases.


Assuntos
Fator 1 de Ribosilação do ADP/metabolismo , Subunidades beta da Proteína de Ligação ao GTP/metabolismo , Complexo de Golgi/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Fator 1 de Ribosilação do ADP/análise , Subunidades beta da Proteína de Ligação ao GTP/análise , Células HEK293 , Humanos , Proteínas Quinases Ativadas por Mitógeno/análise , Células PC-3 , Multimerização Proteica , Transporte Proteico , Receptores Acoplados a Proteínas G/análise , Transdução de Sinais
15.
Biochem Biophys Res Commun ; 549: 98-104, 2021 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-33667715

RESUMO

The ßγ subunit of heterotrimeric G proteins, a key molecule in the G protein-coupled receptors (GPCRs) signaling pathway, has been shown to be an important factor in the modulation of the microtubule cytoskeleton. Gßγ has been shown to bind to tubulin, stimulate microtubule assembly, and promote neurite outgrowth of PC12 cells. In this study, we demonstrate that in addition to microtubules, Gßγ also interacts with actin filaments, and this interaction increases during NGF-induced neuronal differentiation of PC12 cells. We further demonstrate that the Gßγ-actin interaction occurs independently of microtubules as nocodazole, a well-known microtubule depolymerizing agent did not inhibit Gßγ-actin complex formation in PC12 cells. A confocal microscopic analysis of NGF-treated PC12 cells revealed that Gßγ co-localizes with both actin and microtubule cytoskeleton along neurites, with specific co-localization of Gßγ with actin at the distal end of these neuronal processes. Furthermore, we show that Gßγ interacts with the actin cytoskeleton in primary hippocampal and cerebellar rat neurons. Our results indicate that Gßγ serves as an important modulator of the neuronal cytoskeleton by interacting with both microtubules and actin filaments, and is likely to participate in various aspects of neuronal differentiation including axon and growth cone formation.


Assuntos
Citoesqueleto de Actina/metabolismo , Diferenciação Celular , Subunidades beta da Proteína de Ligação ao GTP/metabolismo , Subunidades gama da Proteína de Ligação ao GTP/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Citoesqueleto de Actina/efeitos dos fármacos , Actinas/metabolismo , Animais , Axônios/efeitos dos fármacos , Axônios/metabolismo , Diferenciação Celular/efeitos dos fármacos , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/metabolismo , Hipocampo/citologia , Modelos Biológicos , Fator de Crescimento Neural/farmacologia , Neurônios/efeitos dos fármacos , Células PC12 , Polimerização/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley
16.
Bratisl Lek Listy ; 121(12): 901-906, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33300361

RESUMO

Proteome indicates the protein content of a genome. Proteome analysis is effective in a new system formulation and prediction, prevention, and treatment based on protein. One of the purposes of proteomics researches is to know and understand the cancer mechanism. In this study, we separated the proteins by the Two-Dimensional Electrophorese method and analyzed and compared protein spots by statistical and software data. The spots were separated and identified by the proteins' Isoelectric PH differences, molecular weights, and data bank. In continuation, the protein profiles were clustered by MALDI-TOF-TOF and the main element was identified and confirmed. We have used site PhosphoSitePlus® to review post-translational modifications. The findings indicated that the G protein Beta subunit rate increased in the astrocytoma, oligodendroglia, and glioblastoma cerebral malignant tumors. The ßγ complex formation may prevent and activates many paths of cellular growth. The ßγ complex activity control of the changes after the conversion parallel to GTPase activity of this α subunit may be a formulation mechanism for the G signal path (Tab. 5, Fig. 4, Diagram 2, Ref. 29). Keywords: glioma, G protein ß subunit, proteomics.


Assuntos
Neoplasias Encefálicas/metabolismo , Subunidades beta da Proteína de Ligação ao GTP/metabolismo , Glioma/metabolismo , Processamento de Proteína Pós-Traducional , Encéfalo/metabolismo , Eletroforese em Gel Bidimensional , Humanos , Proteoma/metabolismo , Proteômica
17.
Sci Signal ; 13(656)2020 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-33144519

RESUMO

The PI3Kγ isoform is activated by Gi-coupled GPCRs in myeloid cells, but the extent to which the two endogenous complexes of PI3Kγ, p101/p110γ and p84/p110γ, receive direct regulation through Gßγ or indirect regulation through RAS and the sufficiency of those inputs is controversial or unclear. We generated mice with point mutations that prevent Gßγ binding to p110γ (RK552DD) or to p101 (VVKR777AAAA) and investigated the effects of these mutations in primary neutrophils and in mouse models of neutrophilic inflammation. Loss of Gßγ binding to p110γ substantially reduced the activation of both p101/p110γ and p84/p110γ in neutrophils by various GPCR agonists. Loss of Gßγ binding to p101 caused more variable effects, depending on both the agonist and cellular response, with the biggest reductions seen in PIP3 production by primary neutrophils in response to LTB4 and MIP-2 and in the migration of neutrophils during thioglycolate-induced peritonitis or MIP2-induced ear pouch inflammation. We also observed that p101VVKR777AAAA neutrophils showed enhanced p84-dependent ROS responses to fMLP and C5a, suggesting that competition may exist between p101/p110γ and p84/p110γ for Gßγ subunits downstream of GPCR activation. GPCRs did not activate p110γ in neutrophils from mice lacking both the p101 and p84 regulatory subunits, indicating that RAS binding to p110γ is insufficient to support GPCR activation in this cell type. These findings define a direct role for Gßγ subunits in activating both of the endogenous PI3Kγ complexes and indicate that the regulatory PI3Kγ subunit biases activation toward different GPCRs.


Assuntos
Classe Ib de Fosfatidilinositol 3-Quinase/metabolismo , Subunidades beta da Proteína de Ligação ao GTP/metabolismo , Subunidades gama da Proteína de Ligação ao GTP/metabolismo , Complexos Multienzimáticos/metabolismo , Neutrófilos/enzimologia , Transdução de Sinais , Animais , Classe Ib de Fosfatidilinositol 3-Quinase/genética , Subunidades beta da Proteína de Ligação ao GTP/genética , Subunidades gama da Proteína de Ligação ao GTP/genética , Camundongos , Camundongos Knockout , Complexos Multienzimáticos/genética
18.
Nat Chem Biol ; 16(12): 1368-1375, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32929279

RESUMO

Smoothened (SMO), a class Frizzled G protein-coupled receptor (class F GPCR), transduces the Hedgehog signal across the cell membrane. Sterols can bind to its extracellular cysteine-rich domain (CRD) and to several sites in the seven transmembrane helices (7-TMs) of SMO. However, the mechanism by which sterols regulate SMO via multiple sites is unknown. Here we determined the structures of SMO-Gi complexes bound to the synthetic SMO agonist (SAG) and to 24(S),25-epoxycholesterol (24(S),25-EC). A novel sterol-binding site in the extracellular extension of TM6 was revealed to connect other sites in 7-TMs and CRD, forming an intramolecular sterol channel from the middle side of 7-TMs to CRD. Additional structures of two gain-of-function variants, SMOD384R and SMOG111C/I496C, showed that blocking the channel at its midpoints allows sterols to occupy the binding sites in 7-TMs, thereby activating SMO. These data indicate that sterol transport through the core of SMO is a major regulator of SMO-mediated signaling.


Assuntos
Colesterol/análogos & derivados , Subunidades alfa de Proteínas de Ligação ao GTP/metabolismo , Subunidades beta da Proteína de Ligação ao GTP/metabolismo , Subunidades gama da Proteína de Ligação ao GTP/metabolismo , Transdução de Sinais , Receptor Smoothened/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Sítios de Ligação , Colesterol/química , Colesterol/metabolismo , Cicloexilaminas/química , Cicloexilaminas/farmacologia , Subunidades alfa de Proteínas de Ligação ao GTP/química , Subunidades alfa de Proteínas de Ligação ao GTP/genética , Subunidades beta da Proteína de Ligação ao GTP/química , Subunidades beta da Proteína de Ligação ao GTP/genética , Subunidades gama da Proteína de Ligação ao GTP/química , Subunidades gama da Proteína de Ligação ao GTP/genética , Regulação da Expressão Gênica , Células HEK293 , Humanos , Simulação de Acoplamento Molecular , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Domínios e Motivos de Interação entre Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Anticorpos de Cadeia Única , Receptor Smoothened/agonistas , Receptor Smoothened/química , Receptor Smoothened/genética , Tiofenos/química , Tiofenos/farmacologia
19.
Life Sci ; 262: 118481, 2020 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-32971104

RESUMO

AIMS: G-protein coupled receptors (GPCRs) tightly regulate platelet function by interacting with various physiological agonists. An essential mediator of GPCR signaling is the G protein αßγ heterotrimers, in which the ßγ subunits are central players in downstream signaling. Herein, we investigated the role of Gßγ subunits in platelet function, hemostasis and thrombogenesis. METHODS: To achieve this goal, platelets from both mice and humans were employed in the context of a small molecule inhibitor of Gßγ, namely gallein. We used an aggregometer to examine aggregation and dense granules secretion. We also used flow cytometry for P-selectin and PAC1 to determine the impact of inhibiting Gßγ on α -granule secretion and αIIbß3 activation. Clot retraction and the platelet spreading assay were used to examine Gßγ role in outside-in platelet signaling, whereas Western blot was employed to examine its role in Akt activation. Finally, we used the bleeding time assay and the FeCl3-induced carotid-artery injury thrombosis model to determine Gßγ contribution to in vivo platelet function. RESULTS: We observed that gallein inhibits platelet aggregation and secretion in response to agonist stimulation, in both mouse and human platelets. Furthermore, gallein also exerted inhibitory effects on integrin αIIbß3 activation, clot retraction, platelet spreading and Akt activation/phosphorylation. Finally, gallein's inhibitory effects manifested in vivo, as documented by its ability to modulate physiological hemostasis and delay thrombus formation. CONCLUSION: Our findings demonstrate, for the first time, that Gßγ subunits directly regulate GPCR-dependent platelet function, in vitro and in vivo. Moreover, these data highlight Gßγ as a novel therapeutic target for managing thrombotic disorders.


Assuntos
Plaquetas/fisiologia , Subunidades beta da Proteína de Ligação ao GTP/metabolismo , Subunidades gama da Proteína de Ligação ao GTP/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Trombose/patologia , Animais , Retração do Coágulo/fisiologia , Modelos Animais de Doenças , Hemostasia/fisiologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Agregação Plaquetária/fisiologia , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/fisiologia , Xantenos/farmacologia
20.
PLoS Genet ; 16(8): e1008644, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32776941

RESUMO

Correct regulation of cell contractility is critical for the function of many biological systems. The reproductive system of the hermaphroditic nematode C. elegans contains a contractile tube of myoepithelial cells known as the spermatheca, which stores sperm and is the site of oocyte fertilization. Regulated contraction of the spermatheca pushes the embryo into the uterus. Cell contractility in the spermatheca is dependent on actin and myosin and is regulated, in part, by Ca2+ signaling through the phospholipase PLC-1, which mediates Ca2+ release from the endoplasmic reticulum. Here, we describe a novel role for GSA-1/Gαs, and protein kinase A, composed of the catalytic subunit KIN-1/PKA-C and the regulatory subunit KIN-2/PKA-R, in the regulation of Ca2+ release and contractility in the C. elegans spermatheca. Without GSA-1/Gαs or KIN-1/PKA-C, Ca2+ is not released, and oocytes become trapped in the spermatheca. Conversely, when PKA is activated through either a gain of function allele in GSA-1 (GSA-1(GF)) or by depletion of KIN-2/PKA-R, the transit times and total numbers, although not frequencies, of Ca2+ pulses are increased, and Ca2+ propagates across the spermatheca even in the absence of oocyte entry. In the spermathecal-uterine valve, loss of GSA-1/Gαs or KIN-1/PKA-C results in sustained, high levels of Ca2+ and a loss of coordination between the spermathecal bag and sp-ut valve. Additionally, we show that depleting phosphodiesterase PDE-6 levels alters contractility and Ca2+ dynamics in the spermatheca, and that the GPB-1 and GPB-2 Gß subunits play a central role in regulating spermathecal contractility and Ca2+ signaling. This work identifies a signaling network in which Ca2+ and cAMP pathways work together to coordinate spermathecal contractions for successful ovulations.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Sinalização do Cálcio , Subunidades Catalíticas da Proteína Quinase Dependente de AMP Cíclico/metabolismo , Contração Muscular , 3',5'-AMP Cíclico Fosfodiesterases/metabolismo , Animais , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/genética , Subunidades Catalíticas da Proteína Quinase Dependente de AMP Cíclico/genética , Células Epiteliais/metabolismo , Células Epiteliais/fisiologia , Subunidades beta da Proteína de Ligação ao GTP/metabolismo , Mutação com Ganho de Função , Células Musculares/metabolismo , Células Musculares/fisiologia , Oócitos/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA