Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
J Biol Chem ; 297(6): 101389, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34762910

RESUMO

SRY-box 2 (Sox2) is a transcription factor with critical roles in maintaining embryonic stem (ES) cell and adult stem cell functions and in tumorigenesis. However, how Sox2 exerts its transcriptional function remains unclear. Here, we used an in vitro protein-protein interaction assay to discover transcriptional regulators for ES cell core transcription factors (Oct4, Sox2, Klf4, and c-Myc) and identified members of the steroid receptor coactivators (SRCs) as Sox2-specific interacting proteins. The SRC family coactivators have broad roles in transcriptional regulation, but it is unknown whether they also serve as Sox2 coactivators. We demonstrated that these proteins facilitate Sox2 transcriptional activity and act synergistically with p300. Furthermore, we uncovered an acetylation-enhanced interaction between Sox2 and SRC-2/3, but not SRC-1, demonstrating it is Sox2 acetylation that promotes the interaction. We identified putative Sox2 acetylation sites required for acetylation-enhanced interaction between Sox2 and SRC-3 and demonstrated that acetylation on these sites contributes to Sox2 transcriptional activity and recruitment of SRC-3. We showed that activation domains 1 and 2 of SRC-3 both display a preferential binding to acetylated Sox2. Finally, functional analyses in mouse ES cells demonstrated that knockdown of SRC-2/3 but not SRC-1 in mouse ES cells significantly downregulates the transcriptional activities of various Sox2 target genes and impairs ES cell stemness. Taken together, we identify specific SRC family proteins as novel Sox2 coactivators and uncover the role of Sox2 acetylation in promoting coactivator recruitment and Sox2 transcriptional function.


Assuntos
Coativador 1 de Receptor Nuclear/metabolismo , Coativador 2 de Receptor Nuclear/metabolismo , Coativador 3 de Receptor Nuclear/metabolismo , Fatores de Transcrição SOXB1/metabolismo , Transcrição Gênica , Acetilação , Animais , Células HEK293 , Células HeLa , Humanos , Camundongos , Coativador 1 de Receptor Nuclear/genética , Coativador 2 de Receptor Nuclear/genética , Coativador 3 de Receptor Nuclear/genética , Fatores de Transcrição SOXB1/genética
2.
BMC Pulm Med ; 21(1): 60, 2021 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-33593309

RESUMO

BACKGROUND: To investigate the role and its potential mechanism of 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 4 (PFKFB4) in lung adenocarcinoma. METHODS: Co-immunoprecipitation was performed to analyze the interaction between PFKFB4 and SRC-2. Western blot was used to investigate the phosphorylation of steroid receptor coactivator-2 (SRC-2) on the condition that PFKFB4 was knockdown. Transcriptome sequencing was performed to find the downstream target of SRC-2. Cell Counting Kit-8 (CCK-8) assay, transwell assay and transwell-matrigel assay were used to examine the proliferation, migration and invasion abilities in A549 and NCI-H1975 cells with different treatment. RESULTS: In our study we found that PFKFB4 was overexpressed in lung adenocarcinoma associated with SRC family protein and had an interaction with SRC-2. PFKFB4 could phosphorylate SRC-2 at Ser487, which altered SRC-2 transcriptional activity. Functionally, PFKFB4 promoted lung adenocarcinoma cells proliferation, migration and invasion by phosphorylating SRC-2. Furthermore, we identified that CARM1 was transcriptionally regulated by SRC-2 and involved in PFKFB4-SRC-2 axis on lung adenocarcinoma progression. CONCLUSIONS: Our research reveal that PFKFB4 promotes lung adenocarcinoma cells proliferation, migration and invasion via enhancing phosphorylated SRC-2-mediated CARM1 expression.


Assuntos
Adenocarcinoma de Pulmão/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/genética , Coativador 2 de Receptor Nuclear/genética , Fosfofrutoquinase-2/genética , Células A549 , Adenocarcinoma de Pulmão/patologia , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Progressão da Doença , Perfilação da Expressão Gênica , Células HEK293 , Humanos , Neoplasias Pulmonares/patologia , Invasividade Neoplásica/genética , Fosforilação , Ativação Transcricional/genética
3.
Mol Med ; 26(1): 45, 2020 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-32410572

RESUMO

The large family of transcriptional coactivators originated with the cloning of the subfamily of Steroid Receptor Coactivators (SRC-1,2,3). These 3 coactivators serve as primary 'master genes' to direct the coordinate transcription of multiple genes required for physiological goals in cells, specifically, carbohydrate, lipid, or anabolic growth metabolisms. SRC-2 is of special interest in terms of lipid metabolism and energy accrual and is the topic of a collection of our research discoveries and publications described in this Perspective.


Assuntos
Suscetibilidade a Doenças , Metabolismo Energético , Homeostase , Coativador 2 de Receptor Nuclear/genética , Coativador 2 de Receptor Nuclear/metabolismo , Humanos
4.
Proc Natl Acad Sci U S A ; 112(44): E6068-77, 2015 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-26487680

RESUMO

Despite extensive efforts to understand the monogenic contributions to perturbed glucose homeostasis, the complexity of genetic events that fractionally contribute to the spectrum of this pathology remain poorly understood. Proper maintenance of glucose homeostasis is the central feature of a constellation of comorbidities that define the metabolic syndrome. The ability of the liver to balance carbohydrate uptake and release during the feeding-to-fasting transition is essential to the regulation of peripheral glucose availability. The liver coordinates the expression of gene programs that control glucose absorption, storage, and secretion. Herein, we demonstrate that Steroid Receptor Coactivator 2 (SRC-2) orchestrates a hierarchy of nutritionally responsive transcriptional complexes to precisely modulate plasma glucose availability. Using DNA pull-down technology coupled with mass spectrometry, we have identified SRC-2 as an indispensable integrator of transcriptional complexes that control the rate-limiting steps of hepatic glucose release and accretion. Collectively, these findings position SRC-2 as a major regulator of polygenic inputs to metabolic gene regulation and perhaps identify a previously unappreciated model that helps to explain the clinical spectrum of glucose dysregulation.


Assuntos
Glucose/metabolismo , Homeostase/fisiologia , Proteínas Adaptadoras da Sinalização Shc/fisiologia , Animais , Glucoquinase/genética , Glucoquinase/metabolismo , Camundongos , Camundongos Knockout , Transcrição Gênica
5.
Bioorg Med Chem ; 25(2): 568-574, 2017 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-27890450

RESUMO

Vitamin D receptor (VDR) is a member of the nuclear receptor superfamily and regulates the expression of target genes through ligand binding. To express the target gene, coactivator binding to the VDR/ligand complex is essential. Although there are many coactivators in living cells, precise interactions between coactivators and VDR have not been clarified. Here, we synthesized two coactivator peptides, DRIP205-2 and SRC2-3, evaluated their affinity for the ligand-binding domain (LBD) of VDR using 1α,25-dihydroxyvitamin D3, partial agonist 1, and antagonist 2 by surface plasmon resonance (SPR), and assessed their interaction modes with VDR-LBD using X-ray crystallographic analysis. This study showed that the SRC2-3 peptide is more sensitive to the ligands (agonist, partial agonist, and antagonist) and shows more intimate interactions with VDR-LBD than DRIP205-2 peptide.


Assuntos
Peptídeos/metabolismo , Peptídeos/farmacologia , Receptores de Calcitriol/metabolismo , Sítios de Ligação , Cristalografia por Raios X , Relação Dose-Resposta a Droga , Humanos , Modelos Moleculares , Estrutura Molecular , Peptídeos/síntese química , Peptídeos/química , Receptores de Calcitriol/agonistas , Receptores de Calcitriol/antagonistas & inibidores , Receptores de Calcitriol/química , Relação Estrutura-Atividade , Ressonância de Plasmônio de Superfície
6.
Biochem Biophys Res Commun ; 473(2): 415-20, 2016 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-26970301

RESUMO

Hepatic gluconeogenic programs are regulated by a variety of signaling cascades. Glucagon-cAMP signaling is the main initiator of the gluconeogenic programs, including glucose-6-phosphatase catalytic subunit (G6pc) gene expression. Pterosin B, an ingredient in Pteridium aquilinum, inhibits salt-inducible kinase 3 signaling that represses cAMP-response element-binding protein regulated transcription coactivator 2, an inducer of gluconeogenic programs. As the results, pterosin B promotes G6pc expression even in the absence of cAMP. In this work, however, we noticed that once cAMP signaling was initiated, pterosin B became a strong repressor of G6pc expression. The search for associated transcription factors for pterosin B actions revealed that retinoic acid receptor-related orphan receptor alpha-steroid receptor coactivator 2 (RORα-SRC2) complex on the G6pc promoter was the target. Meanwhile, pterosin B impaired the oxidation-reduction cycle of coenzyme Q in mitochondrial oxidative phosphorylation (OXPHOS); and antimycin A, an inhibitor of coenzyme Q: cytochrome c-oxidoreductase (termed mitochondrial complex III), also mimicked pterosin B actions on RORα-SRC2 signaling. Although other respiratory toxins (rotenone and oligomycin) also suppressed G6pc expression accompanied by lowered ATP levels following the activation of AMP-activated kinase, minimal or no effect of these other toxins on RORα-SRC2 activity was observed. These results suggested that individual components in OXPHOS differentially linked to different transcriptional machineries for hepatic gluconeogenic programs, and the RORα-SRC2 complex acted as a sensor for oxidation-reduction cycle of coenzyme Q and regulated G6Pc expression. This was a site disrupted by pterosin B in gluconeogenic programs.


Assuntos
Gluconeogênese/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Indanos/farmacologia , Coativador 2 de Receptor Nuclear/metabolismo , Membro 1 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Transdução de Sinais/efeitos dos fármacos , Ubiquinona/metabolismo , Animais , Linhagem Celular , Regulação da Expressão Gênica/efeitos dos fármacos , Glucose/metabolismo , Glucose-6-Fosfatase/genética , Hepatócitos/metabolismo , Indanos/química , Camundongos , Oxirredução/efeitos dos fármacos , Regiões Promotoras Genéticas/efeitos dos fármacos , Mapas de Interação de Proteínas/efeitos dos fármacos , Pteridium/química
7.
Biochim Biophys Acta ; 1833(12): 2775-2780, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23872431

RESUMO

Reactive oxygen species (ROS) produced by NADPH oxidases play critical roles in plant environmental responses. Arabidopsis thaliana NADPH oxidase AtRbohF-mediated ROS-production is involved in abiotic stress responses. Because overproduction of ROS is highly toxic to cells, the activity of AtRbohF needs to be tightly regulated in response to diverse stimuli. The ROS-producing activity of AtRbohF is activated by Ca(2+) and protein phosphorylation, but other regulatory factors for AtRbohF are mostly unknown. In this study, we screened for proteins that interact with the N-terminal cytosolic region of AtRbohF by a yeast two-hybrid screen, and isolated AtSRC2, an A. thaliana homolog of SRC2 (soybean gene regulated by cold-2). A co-immunoprecipitation assay revealed that AtSRC2 interacts with the N-terminal region of AtRbohF in plant cells. Intracellular localization of GFP-tagged AtSRC2 was partially overlapped with that of GFP-tagged AtRbohF at the cell periphery. Co-expression of AtSRC2 enhanced the Ca(2+)-dependent ROS-producing activity of AtRbohF in HEK293T cells, but did not affect its phosphorylation-dependent activation. Low-temperature treatment induced expression of the AtSRC2 gene in Arabidopsis roots in proportion to levels of ROS production that was partially dependent on AtRbohF. Our findings suggest that AtSRC2 is a novel activator of Ca(2+)-dependent AtRbohF-mediated ROS production and may play a role in cold responses.


Assuntos
Proteínas de Arabidopsis/metabolismo , Arabidopsis/enzimologia , Temperatura Baixa , NADPH Oxidases/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Arabidopsis/efeitos dos fármacos , Arabidopsis/genética , Arabidopsis/fisiologia , Proteínas de Arabidopsis/química , Proteínas de Arabidopsis/isolamento & purificação , Cálcio/farmacologia , Regulação da Expressão Gênica de Plantas/efeitos dos fármacos , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Humanos , NADPH Oxidases/química , Células Vegetais/efeitos dos fármacos , Células Vegetais/metabolismo , Ligação Proteica/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Proteínas Recombinantes de Fusão/metabolismo , Estresse Fisiológico/efeitos dos fármacos , Estresse Fisiológico/genética , Frações Subcelulares/efeitos dos fármacos , Frações Subcelulares/metabolismo , Técnicas do Sistema de Duplo-Híbrido
8.
Cell Rep ; 37(10): 110075, 2021 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-34879284

RESUMO

The neuroendocrine system coordinates metabolic and behavioral adaptations to fasting, including reducing energy expenditure, promoting counterregulation, and suppressing satiation and anxiety to engage refeeding. Here, we show that steroid receptor coactivator-2 (SRC-2) in pro-opiomelanocortin (POMC) neurons is a key regulator of all these responses to fasting. POMC-specific deletion of SRC-2 enhances the basal excitability of POMC neurons; mutant mice fail to efficiently suppress energy expenditure during food deprivation. SRC-2 deficiency blunts electric responses of POMC neurons to glucose fluctuations, causing impaired counterregulation. When food becomes available, these mutant mice show insufficient refeeding associated with enhanced satiation and discoordination of anxiety and food-seeking behavior. SRC-2 coactivates Forkhead box protein O1 (FoxO1) to suppress POMC gene expression. POMC-specific deletion of SRC-2 protects mice from weight gain induced by an obesogenic diet feeding and/or FoxO1 overexpression. Collectively, we identify SRC-2 as a key molecule that coordinates multifaceted adaptive responses to food shortage.


Assuntos
Metabolismo Energético , Jejum/metabolismo , Comportamento Alimentar , Hipotálamo/metabolismo , Neurônios/metabolismo , Coativador 2 de Receptor Nuclear/metabolismo , Obesidade/metabolismo , Hipernutrição/metabolismo , Pró-Opiomelanocortina/metabolismo , Animais , Ansiedade/metabolismo , Ansiedade/fisiopatologia , Ansiedade/psicologia , Modelos Animais de Doenças , Jejum/psicologia , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Células HEK293 , Humanos , Hipotálamo/fisiopatologia , Masculino , Camundongos Knockout , Coativador 2 de Receptor Nuclear/genética , Obesidade/genética , Obesidade/fisiopatologia , Obesidade/psicologia , Hipernutrição/genética , Hipernutrição/fisiopatologia , Hipernutrição/psicologia , Pró-Opiomelanocortina/genética , Resposta de Saciedade , Transdução de Sinais , Aumento de Peso
9.
Sex Med ; 9(3): 100368, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-34049263

RESUMO

INTRODUCTION: Brain sexual differentiation results from the effects of sex steroids on the developing brain. The presumptive route for brain masculinization is the direct induction of gene expression via activation of the estrogen receptors α and ß and the androgen receptor through their binding to ligands and to coactivators, regulating the transcription of multiple genes in a cascade effect. AIM: To analyze the implication of the estrogen receptor coactivators SRC-1, SRC-2, and SRC-3 in the genetic basis of gender incongruence. MAIN OUTCOME MEASURES: Analysis of 157 polymorphisms located at the estrogen receptor coactivators SRC-1, SRC-2, and SRC-3, in 94 transgender versus 94 cisgender individuals. METHOD: Using SNPStats software, the allele and genotype frequencies were analyzed by χ2, the strength of the association was measured by binary logistic regression, estimating the odds ratio for each genotype. Measurements of linkage disequilibrium and haplotype frequencies were also performed. RESULTS: We found significant differences at level P < .05 in 8 polymorphisms that correspond to 5.09% of the total. Three were located in SRC-1 and 5 in SRC-2. The odds ratio analysis showed significant differences at level P < .05 for multiple patterns of inheritance. The polymorphisms analyzed were in linkage disequilibrium. The SRC-1 haplotypes CGA and CGG (global haplotype association P < .009) and the SRC-2 haplotypes GGTAA and GGTAG (global haplotype association P < .005) were overrepresented in the transgender population. CONCLUSION: The coactivators SRC-1 and SRC-2 could be considered as candidates for increasing the list of potential genes for gender incongruence. Ramírez KDV, Fernández R, Delgado-Zayas E, et al. Implications of the Estrogen Receptor Coactivators SRC1 and SRC2 in the Biological Basis of Gender Incongruence. Sex Med 2021;9:100368.

10.
Mol Cell Endocrinol ; 419: 92-101, 2016 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-26455881

RESUMO

Fasting hormones activate the cAMP/PKA signaling pathway and stimulate expression of hepatic gluconeogenic enzymes including glucose-6-phosphatase (G6Pase). Previously it was shown that steroid receptor coactivator 2 (SRC-2) knock-out mice exhibit fasting hypoglycemia and that SRC-2 coactivates RAR-related orphan receptor alpha (RORα) at the proximal G6Pase promoter. We have investigated the upstream regulation and functional implications of this RORα/SRC-2 complex on G6Pase expression. In HepG2 cells, overexpression of the catalytic PKA subunit (PKA-Cα) reduced the SRC-2 protein level, recruitment to the G6Pase promoter, and its ability to coactivate RORα. Knock-down and transactivation experiments employing G6Pase promoter constructs demonstrated that RORα and SRC-2 are required for PGC-1α to stimulate G6Pase expression. These results suggest that PKA inhibits SRC-2 coactivation of RORα and in turn reduces PGC-1α dependent regulation of G6Pase. This indirect feedback mechanism may underlie the suppression of gluconeogenesis throughout long-term starvation.


Assuntos
Subunidades Catalíticas da Proteína Quinase Dependente de AMP Cíclico/metabolismo , Regulação para Baixo , Glucose-6-Fosfatase/genética , Coativador 2 de Receptor Nuclear/genética , Membro 1 do Grupo F da Subfamília 1 de Receptores Nucleares/genética , Subunidades Catalíticas da Proteína Quinase Dependente de AMP Cíclico/genética , Regulação da Expressão Gênica , Gluconeogênese , Células Hep G2 , Humanos , Coativador 2 de Receptor Nuclear/metabolismo , Membro 1 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Regiões Promotoras Genéticas , Transcrição Gênica
11.
J Biol Rhythms ; 31(5): 443-60, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27432117

RESUMO

Circadian rhythmicity is a fundamental process that synchronizes behavioral cues with metabolic homeostasis. Disruption of daily cycles due to jet lag or shift work results in severe physiological consequences including advanced aging, metabolic syndrome, and even cancer. Our understanding of the molecular clock, which is regulated by intricate positive feedforward and negative feedback loops, has expanded to include an important metabolic transcriptional coregulator, Steroid Receptor Coactivator-2 (SRC-2), that regulates both the central clock of the suprachiasmatic nucleus (SCN) and peripheral clocks including the liver. We hypothesized that an environmental uncoupling of the light-dark phases, termed chronic circadian disruption (CCD), would lead to pathology similar to the genetic circadian disruption observed with loss of SRC-2 We found that CCD and ablation of SRC-2 in mice led to a common comorbidity of metabolic syndrome also found in humans with circadian disruption, non-alcoholic fatty liver disease (NAFLD). The combination of SRC-2(-/-) and CCD results in a more robust phenotype that correlates with human non-alcoholic steatohepatitis (NASH) and hepatocellular carcinoma (HCC) gene signatures. Either CCD or SRC-2 ablation produces an advanced aging phenotype leading to increased mortality consistent with other circadian mutant mouse models. Collectively, our studies demonstrate that SRC-2 provides an essential link between the behavioral activities influenced by light cues and the metabolic homeostasis maintained by the liver.


Assuntos
Envelhecimento , Fígado/patologia , Coativador 2 de Receptor Nuclear/genética , Coativador 2 de Receptor Nuclear/fisiologia , Animais , Carcinoma Hepatocelular/genética , Relógios Circadianos , Ritmo Circadiano/fisiologia , Modelos Animais de Doenças , Humanos , Fígado/metabolismo , Neoplasias Hepáticas/genética , Camundongos , Hepatopatia Gordurosa não Alcoólica/etiologia , Hepatopatia Gordurosa não Alcoólica/fisiopatologia , Coativador 2 de Receptor Nuclear/deficiência , Proteínas Circadianas Period/genética , Fotoperíodo , Núcleo Supraquiasmático/fisiologia
12.
Leuk Lymphoma ; 1(1): 35-41, 1989.
Artigo em Inglês | MEDLINE | ID: mdl-27456405

RESUMO

Cellular oncogenes are frequently activated or deregulated in human malignant tumors. We have analyzed the expression of cellular oncogenes in human leukemia by Northern blot experiments in a case of acute myeloid leukemia (AML). In this case the cellular oncogenes N-myc, c-myb, c-fes and c-met were expressed at high levels. This is in contrast to normal peripheral blood mononuclear cells (PBMC) or chronic lymphocytic leukemia (CLL) cells, where these genes could not be detected. c-src2-specific RNA was not seen in either AML or PBMC cells but readily appeared in CLL cells. This appears to represent further evidence that in leukemic cells multiple cellular oncogenes might be activated.

13.
J Neuroendocrinol ; 25(11): 1209-18, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23795583

RESUMO

Steroid hormones act in specific regions of the brain to alter behaviour and physiology. Although it has been well established that the bioavailability of the steroid and the expression of its receptor is critical for understanding steroid action in the brain, the importance of nuclear receptor coactivators in the brain is becoming more apparent. The present review focuses on the function of the p160 family of coactivators, which includes steroid receptor coactivator-1 (SRC-1), SRC-2 and SRC-3, in steroid receptor action in the brain. The expression, regulation and function of these coactivators in steroid-dependent gene expression in both brain and behaviour are discussed.


Assuntos
Encéfalo/fisiologia , Regulação da Expressão Gênica/fisiologia , Hormônios/fisiologia , Coativadores de Receptor Nuclear/fisiologia , Receptores de Esteroides/fisiologia , Comportamento Sexual Animal/fisiologia , Animais , Encéfalo/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa