Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
1.
Nucleic Acids Res ; 51(19): 10194-10217, 2023 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-37638746

RESUMO

Incorporation of histone variant H3.3 comprises active territories of chromatin. Exploring the function of H3.3 in prostate cancer (PC), we found that knockout (KO) of H3.3 chaperone HIRA suppresses PC growth in vitro and in xenograft settings, deregulates androgen-induced gene expression and alters androgen receptor (AR) binding within enhancers of target genes. H3.3 affects transcription in multiple ways, including activation of p300 by phosphorylated H3.3 at Ser-31 (H3.3S31Ph), which results in H3K27 acetylation (H3K27Ac) at enhancers. In turn, H3K27Ac recruits bromodomain protein BRD4 for enhancer-promoter interaction and transcription activation. We observed that HIRA KO reduces H3.3 incorporation, diminishes H3.3S31Ph and H3K27Ac, modifies recruitment of BRD4. These results suggest that H3.3-enriched enhancer chromatin serves as a platform for H3K27Ac-mediated BRD4 recruitment, which interacts with and retains AR at enhancers, resulting in transcription reprogramming. In addition, HIRA KO deregulates glucocorticoid- (GR) driven transcription of genes co-regulated by AR and GR, suggesting a common H3.3/HIRA-dependent mechanism of nuclear receptors function. Expression of HIRA complex proteins is increased in PC compared with normal prostate tissue, especially in high-risk PC groups, and is associated with a negative prognosis. Collectively, our results demonstrate function of HIRA-dependent H3.3 pathway in regulation of nuclear receptors activity.


Assuntos
Histonas , Proteínas Nucleares , Humanos , Masculino , Androgênios/farmacologia , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Cromatina/genética , Chaperonas de Histonas/metabolismo , Histonas/genética , Histonas/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Elementos Facilitadores Genéticos
2.
Prostate ; 82(7): 816-825, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35226379

RESUMO

BACKGROUND: Prostate cancer (PC) is the most commonly diagnosed malignancy and the second leading cause of cancer-related deaths in males. The disease is initially treated with methods that inhibit androgen receptor (AR) signal transduction. Laboratory-based and clinical studies have identified alternative pathways that cause the failure of AR signal inhibition and consequent development of castration-resistant prostate cancer (CRPC). Glucocorticoid receptor (GR) signaling is activated in certain PC patients and promotes the emergence of CRPC, although by as yet incompletely understood mechanisms. We have previously demonstrated that ubiquitous ßarrestin1 (ßArr1) expression levels are linked to PC progression. Here, we consider the possibility that ßArr1 interacts with and activates GR in model CRPC cells. METHODS: Bioinformatic analysis of tumor xenograft and human PC datasets was used to correlate the expression of ßArr1 and GR. Western blot, immunohistochemistry and immunofluorescence microscopy, and subcellular fractionation were used to determine protein expression level and localization. Immunoprecipitation was applied to detect protein-protein interactions. RNA expression levels were determined using quantitative reverse transcription-polymerase chain reaction. Prostate sphere analysis was used to assess the rate of growth and invasion. The xenograft tumor implantation method was used to determine the tumor growth rate, local invasion, and metastasis. RESULTS: Elevated expression of ßArr1 positively correlated with increased GR expression and function in CRPC xenograft and in human PC patients. ßArr1 is expressed in the cell cytosol and nucleus, and it formed a complex with GR in the nucleus and not cytosol. Depletion of ßArr1 in AR-null CRPC cells inhibited GR function and CRPC growth and invasion in both in vitro and in vivo settings. CONCLUSIONS: ßArr1 binds GR that initiates mitogenic signaling cascades involved in the progression of PC to CRPC. The targeting of the ßArr1-GR axis may provide a new opportunity to better manage the CRPC disease.


Assuntos
Neoplasias de Próstata Resistentes à Castração , beta-Arrestina 1/metabolismo , Androgênios , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Mitógenos/uso terapêutico , Neoplasias de Próstata Resistentes à Castração/patologia , Receptores Androgênicos/metabolismo , Receptores de Glucocorticoides/metabolismo , Transdução de Sinais , beta-Arrestina 1/genética
3.
J Biol Chem ; 293(42): 16518-16527, 2018 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-30166343

RESUMO

Uropathogenic Escherichia coli (UPEC) is the causative bacterium in most urinary tract infections (UTIs). UPEC cells adhere to and invade bladder epithelial cells (BECs) and cause uropathogenicity. Invading UPEC cells may encounter one of several fates, including degradation in the lysosome, expulsion to the extracellular milieu for clearance, or survival as an intracellular bacterial community and quiescent intracellular reservoir that can cause later infections. Here we considered the possibility that UPEC cells secrete factors that activate specific host cell signaling networks to facilitate the UPEC invasion of BECs. Using GFP-based reporters and Western blot analysis, we found that the representative human cystitis isolate E. coli UTI89 and its derivative UTI89ΔFimH, which does not bind to BECs, equally activate phosphatidylinositol 4,5-bisphosphate 3-OH kinase (PI3K), Akt kinase, and mTOR complex (mTORC) 1 and 2 in BECs. We also found that conditioned medium taken from UTI89 and UTI89ΔFimH cultures similarly activates epidermal growth factor receptor (EGFR), PI3K, Akt, and mTORC and that inhibition of EGFR and mTORC2, but not mTORC1, abrogates UTI89 invasion in vitro and in animal models of UTI. Our results reveal a key molecular mechanism of UPEC invasion and the host cells it targets, insights that may have therapeutic utility for managing the ever-increasing number of persistent and chronic UTIs.


Assuntos
Células Epiteliais/microbiologia , Interações Hospedeiro-Patógeno , Bexiga Urinária/patologia , Escherichia coli Uropatogênica/patogenicidade , Animais , Meios de Cultivo Condicionados/química , Células Epiteliais/metabolismo , Receptores ErbB/metabolismo , Humanos , Proteínas Quinases/metabolismo , Transdução de Sinais , Infecções Urinárias/etiologia , Infecções Urinárias/microbiologia
4.
EMBO J ; 34(12): 1674-86, 2015 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-25964433

RESUMO

The biological underpinnings linking stress to Alzheimer's disease (AD) risk are poorly understood. We investigated how corticotrophin releasing factor (CRF), a critical stress response mediator, influences amyloid-ß (Aß) production. In cells, CRF treatment increases Aß production and triggers CRF receptor 1 (CRFR1) and γ-secretase internalization. Co-immunoprecipitation studies establish that γ-secretase associates with CRFR1; this is mediated by ß-arrestin binding motifs. Additionally, CRFR1 and γ-secretase co-localize in lipid raft fractions, with increased γ-secretase accumulation upon CRF treatment. CRF treatment also increases γ-secretase activity in vitro, revealing a second, receptor-independent mechanism of action. CRF is the first endogenous neuropeptide that can be shown to directly modulate γ-secretase activity. Unexpectedly, CRFR1 antagonists also increased Aß. These data collectively link CRF to increased Aß through γ-secretase and provide mechanistic insight into how stress may increase AD risk. They also suggest that direct targeting of CRF might be necessary to effectively modulate this pathway for therapeutic benefit in AD, as CRFR1 antagonists increase Aß and in some cases preferentially increase Aß42 via complex effects on γ-secretase.


Assuntos
Doença de Alzheimer/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Peptídeos beta-Amiloides/biossíntese , Hormônio Liberador da Corticotropina/metabolismo , Modelos Biológicos , Estresse Fisiológico/fisiologia , Doença de Alzheimer/etiologia , Análise de Variância , Animais , Western Blotting , AMP Cíclico/metabolismo , Ensaio de Imunoadsorção Enzimática , Células HEK293 , Humanos , Sistema Hipotálamo-Hipofisário/fisiologia , Imunoprecipitação , Microdomínios da Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência , Sistema Hipófise-Suprarrenal/fisiologia , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Hormônio Liberador da Corticotropina/metabolismo
5.
Semin Cell Dev Biol ; 50: 95-104, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26773211

RESUMO

GPCRs are ubiquitous in mammalian cells and present intricate mechanisms for cellular signaling and communication. Mechanistically, GPCR signaling was identified to occur vectorially through heterotrimeric G proteins that are negatively regulated by GRK and arrestin effectors. Emerging evidence highlights additional roles for GRK and Arrestin partners, and establishes the existence of interconnected feedback pathways that collectively define GPCR signaling. GPCRs influence cellular dynamics and can mediate pathologic development, such as cancer and cardiovascular remolding. Hence, a better understanding of their overall signal regulation is of great translational interest and research continues to exploit the pharmacologic potential for modulating their activity.


Assuntos
Arrestinas/metabolismo , Retroalimentação Fisiológica , Quinases de Receptores Acoplados a Proteína G/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais , Animais , Humanos , Modelos Biológicos
6.
Prostate ; 78(15): 1140-1156, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30009471

RESUMO

BACKGROUND: Mutations or truncation of the ligand-binding domain (LBD) of androgen receptor (AR) underlie treatment resistance for prostate cancer (PCa). Thus, targeting the AR N-terminal domain (NTD) could overcome such resistance. METHODS: Luciferase reporter assays after transient transfection of various DNA constructs were used to assess effects of E1A proteins on AR-mediated transcription. Immunofluorescence microscopy and subcellular fractionation were applied to assess intracellular protein localization. Immunoprecipitation and mammalian two-hybrid assays were used to detect protein-protein interactions. qRT-PCR was employed to determine RNA levels. Western blotting was used to detect protein expression in cells. Effects of adenoviruses on prostate cancer cell survival were evaluated with CellTiter-Glo assays. RESULTS: Adenovirus 12 E1A (E1A12) binds specifically to the AR. Interestingly, the full-length E1A12 (266 aa) preferentially binds to full-length AR, while the small E1A12 variant (235 aa) interacts more strongly with AR-V7. E1A12 promotes AR nuclear translocation, likely through mediating intramolecular AR NTD-LBD interactions. In the nucleus, AR and E1A12 co-expression in AR-null PCa cells results in E1A12 redistribution from nuclear foci containing CBX4 (also known as Pc2), suggesting a preferential AR-E1A12 interaction over other E1A12 interactors. E1A12 represses AR-mediated transcription in reporter gene assays and endogenous AR target genes such as ATAD2 and MYC in AR-expressing PCa cells. AR-expressing PCa cells are more sensitive to death induced by a recombinant adenovirus expressing E1A12 (Ad-E1A12) than AR-deficient PCa cells, which could be attributed to the increased viral replication promoted by androgen stimulation. Targeting the AR by E1A12 promotes apoptosis in PCa cells that express the full-length AR or C-terminally truncated AR variants. Importantly, inhibition of mTOR signaling that blocks the expression of anti-apoptotic proteins markedly augments Ad-E1A12-induced apoptosis of AR-expressing cells. Mechanistically, Ad-E1A12 infection triggers apoptotic response while activating the PI3K-AKT-mTOR signaling axis; thus, mTOR inhibition enhances apoptosis in AR-expressing PCa cells infected by Ad-E1A12. CONCLUSION: Ad12 E1A inhibits AR-mediated transcription and suppresses PCa cell survival, suggesting that targeting the AR by E1A12 might have therapeutic potential for treating advanced PCa with heightened AR signaling.


Assuntos
Proteínas E1A de Adenovirus/metabolismo , Neoplasias da Próstata/terapia , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Proteínas E1A de Adenovirus/genética , Adenovírus Humanos/genética , Adenovírus Humanos/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/fisiologia , Células HEK293 , Humanos , Masculino , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/virologia , Domínios Proteicos , Ativação Transcricional , Transfecção
7.
Biochem Biophys Res Commun ; 471(1): 41-6, 2016 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-26850854

RESUMO

Benign prostatic hyperplasia (BPH) is a common disease in older men that involves the enlargement of the prostate gland. This occurs in response to signal transduction initiated by α-adrenergic receptors (α-ARs). When bound to ligands, α-ARs stimulate the mitogenic extracellular signal-regulated kinases 1 and 2 (ERK) pathway, ultimately promoting stromal and epithelial cell hyperplasia in the prostate. Current knowledge of how α-ARs promote prostate cell growth remains incomplete, and despite decades of research, there is no cure for BPH. In this study, we aimed to exploit an in vitro model system of BPH in order to better understand the mechanisms of α-AR signaling in prostatic hyperplasia.


Assuntos
Arrestinas/metabolismo , Sistema de Sinalização das MAP Quinases , Hiperplasia Prostática/metabolismo , Receptores Adrenérgicos alfa/metabolismo , Transdução de Sinais , Idoso , Linhagem Celular , Humanos , Masculino , beta-Arrestinas
9.
J Biol Chem ; 289(19): 13638-50, 2014 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-24692549

RESUMO

ßArrestin proteins shuttle between the cytosol and nucleus and have been shown to regulate G protein-coupled receptor signaling, actin remodeling, and gene expression. Here, we tested the hypothesis that ßarrestin1 regulates actin remodeling and cell migration through the small GTPase Rac. Depletion of ßarrestin1 promotes Rac activation, leading to the formation of multipolar protrusions and increased cell circularity, and overexpression of a dominant negative form of Rac reverses these morphological changes. Small interfering RNA library screen identifies RasGRF2 as a target of ßarrestin1. RasGRF2 gene and protein expression levels are elevated following depletion of ßarrestin1, and the consequent activation of Rac results in dephosphorylation of cofilin that can promote actin polymerization and formation of multipolar protrusions, thereby retarding cell migration and invasion. Together, these results suggest that ßarrestin1 regulates rasgrf2 gene expression and Rac activation to affect membrane protrusion and cell migration and invasion.


Assuntos
Arrestinas/metabolismo , Estruturas da Membrana Celular/metabolismo , Regulação da Expressão Gênica/fisiologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fatores ras de Troca de Nucleotídeo Guanina/biossíntese , Animais , Arrestinas/genética , Estruturas da Membrana Celular/genética , Movimento Celular/fisiologia , Ativação Enzimática/fisiologia , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Camundongos , Fosforilação/fisiologia , Proteínas Proto-Oncogênicas c-akt/genética , beta-Arrestinas , Fatores ras de Troca de Nucleotídeo Guanina/genética
10.
J Biol Chem ; 287(23): 18925-36, 2012 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-22500016

RESUMO

ß(2)-Adrenergic receptors (ß(2)ARs) regulate cellular functions through G protein-transduced and ßArrestin-transduced signals. ß(2)ARs have been shown to regulate cancer cell migration, but the underlying mechanisms are not well understood. Here, we report that ß(2)AR regulates formation of focal adhesions, whose dynamic remodeling is critical for directed cell migration. ß(2)ARs induce activation of RhoA, which is dependent on ßArrestin2 but not G(s). ßArrestin2 forms a complex with p115RhoGEF, a guanine nucleotide exchange factor for RhoA that is well known to be activated by G(12/13)-coupled receptors. Our results show that ßArrestin2 forms a complex with p115RhoGEF in the cytosol in resting cells. Upon ß(2)AR activation, both ßArrestin2 and p115RhoGEF translocate to the plasma membrane, with concomitant activation of RhoA and formation of focal adhesions and stress fibers. Activation of RhoA and focal adhesion remodeling may explain, at least in part, the role of ß(2)ARs in cell migration. These results suggest that ßArrestin2 may serve as a convergence point for non-G(12/13) and non-G(q) protein-coupled receptors to activate RhoA.


Assuntos
Arrestinas/metabolismo , Adesões Focais/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Transdução de Sinais/fisiologia , Proteínas rho de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Fator 6 Ativador da Transcrição , Animais , Arrestinas/genética , Fatores de Transcrição de Zíper de Leucina Básica/genética , Fatores de Transcrição de Zíper de Leucina Básica/metabolismo , Movimento Celular/fisiologia , Ativação Enzimática/fisiologia , Adesões Focais/genética , Fatores de Troca do Nucleotídeo Guanina/genética , Células HEK293 , Humanos , Camundongos , Receptores Adrenérgicos beta 2/genética , Fatores de Troca de Nucleotídeo Guanina Rho , beta-Arrestinas , Proteínas rho de Ligação ao GTP/genética , Proteína rhoA de Ligação ao GTP/genética
11.
Biochim Biophys Acta ; 1820(6): 743-51, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21397660

RESUMO

G protein-coupled receptors (GPCRs) are the most numerous and diverse type of cell surface receptors, accounting for about 1% of the entire human genome and relaying signals from a variety of extracellular stimuli that range from lipid and peptide growth factors to ions and sensory inputs. Activated GPCRs regulate a multitude of target cell functions, including intermediary metabolism, growth and differentiation, and migration and invasion. The GPCRs contain a characteristic 7-transmembrane domain topology and their activation promotes complex formation with a variety of intracellular partner proteins, which form basis for initiation of distinct signaling networks as well as dictate fate of the receptor itself. Both termination of active GPCR signaling and removal from the plasma membrane are controlled by protein post-translational modifications of the receptor itself and its interacting partners. Phosphorylation, acylation and ubiquitination are the most studied post-translational modifications involved in GPCR signal transduction, subcellular trafficking and overall expression. Emerging evidence demonstrates that protein S-nitrosylation, the covalent attachment of a nitric oxide moiety to specified cysteine thiol groups, of GPCRs and/or their associated effectors also participates in the fine-tuning of receptor signaling and expression. This newly appreciated mode of GPCR system modification adds another set of controls to more precisely regulate the many cellular functions elicited by this large group of receptors. This article is part of a Special Issue entitled: Regulation of cellular processes by S-nitrosylation.


Assuntos
Processamento de Proteína Pós-Traducional , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais , Arrestinas/metabolismo , Cisteína/química , Cisteína/metabolismo , Dinaminas/metabolismo , Regulação da Expressão Gênica , Humanos , Óxido Nítrico/química , Óxido Nítrico/metabolismo , Nitrosação , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/genética , beta-Arrestinas
12.
Blood ; 118(19): 5355-64, 2011 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-21926356

RESUMO

Inflammation is increasingly recognized as a critical mediator of angiogenesis, and unregulated angiogenic response is involved in human diseases, including cancer. Proinflammatory prostaglandin E2 (PGE2) is secreted by many cell types and plays important roles in the process of angiogenesis via activation of cognate EP1-4 receptors. Here, we provide evidence that PGE2 promotes the in vitro tube formation of human microvascular endothelial cells, ex vivo vessel outgrowth of aortic rings, and actual in vivo angiogenesis. Use of EP subtype-selective agonists and antagonists suggested EP4 mediates the prostaglandin-induced tube formation, and this conclusion was substantiated with small interfering RNA to specifically knockdown the EP4 expression. EP4 couples to Gαs, leading to activation of protein kinase A (PKA). Inhibition of PKA activity or knockdown of PKA catalytic subunit γ with RNAi attenuates the PGE2-induced tube formation. Further, knocking down the expression of Rap1A, HSPB6, or endothelial NO synthase, which serve as PKA-activatable substrates, inhibits the tube formation, whereas knockdown of RhoA or glycogen synthase kinase 3ß that are inactivated after phosphorylation by PKA increases the tube formation. These results support the existence of EP4-to-PKA angiogenic signal and provide rationale for use of selective EP4 signal inhibitors as a probable strategy to control pathologic angiogenesis.


Assuntos
Subunidades Catalíticas da Proteína Quinase Dependente de AMP Cíclico/fisiologia , Dinoprostona/fisiologia , Neovascularização Fisiológica/fisiologia , Receptores de Prostaglandina E Subtipo EP4/fisiologia , Animais , Aorta Torácica/citologia , Aorta Torácica/efeitos dos fármacos , Aorta Torácica/fisiologia , Sequência de Bases , Embrião de Galinha , Subunidades Catalíticas da Proteína Quinase Dependente de AMP Cíclico/antagonistas & inibidores , Subunidades Catalíticas da Proteína Quinase Dependente de AMP Cíclico/genética , Dinoprostona/farmacologia , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/fisiologia , Técnicas de Silenciamento de Genes , Humanos , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Fisiológica/efeitos dos fármacos , RNA Interferente Pequeno/genética , Receptores de Prostaglandina E Subtipo EP4/antagonistas & inibidores , Receptores de Prostaglandina E Subtipo EP4/genética
13.
bioRxiv ; 2023 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-37214820

RESUMO

Incorporation of histone variant H3.3 comprises active territories of chromatin. Exploring the function of H3.3 in prostate cancer (PC), we found that knockout (KO) of H3.3 chaperone HIRA suppresses PC growth in vitro and in xenograft settings, deregulates androgen-induced gene expression and alters androgen receptor (AR) binding within enhancers of target genes. H3.3 affects transcription in multiple ways, including activation of p300 by phosphorylated H3.3 at Ser-31 (H3.3S31Ph), which results in H3K27 acetylation (H3K27Ac) at enhancers. In turn, H3K27Ac recruits bromodomain protein BRD4 for enhancer-promoter interaction and transcription activation. We observed that HIRA KO reduces H3.3 incorporation, diminishes H3.3S31Ph and H3K27Ac, modifies recruitment of BRD4. These results suggest that H3.3-enriched enhancer chromatin serves as a platform for H3K27Ac-mediated BRD4 recruitment, which interacts with and retains AR at enhancers, resulting in transcription reprogramming. AR KO reduced levels of H3.3 at enhancers, indicating feedback mechanism. In addition, HIRA KO deregulates glucocorticoid-driven transcription, suggesting a common H3.3/HIRA-dependent mechanism of nuclear receptors function. Expression of HIRA complex proteins is increased in PC compared with normal prostate tissue, especially in high-risk PC groups, and is associated with a negative prognosis. Collectively, our results demonstrate function of HIRA-dependent H3.3 pathway in regulation of nuclear receptors activity. Key points: *H3.3 at enhancers promotes acetylation of H3K27Ac and retention of AR/BRD4 complex for transcription regulation*Knockout of H3.3 chaperone HIRA suppresses PC cells growth and deregulates androgen-induced transcription*H3.3/HIRA pathway regulates both AR and GR, suggesting a common HIRA/H3.3 mechanism of nuclear receptors function.

14.
Nat Commun ; 14(1): 1927, 2023 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-37045819

RESUMO

Cancer cells exhibit elevated lipid synthesis. In breast and other cancer types, genes involved in lipid production are highly upregulated, but the mechanisms that control their expression remain poorly understood. Using integrated transcriptomic, lipidomic, and molecular studies, here we report that DAXX is a regulator of oncogenic lipogenesis. DAXX depletion attenuates, while its overexpression enhances, lipogenic gene expression, lipogenesis, and tumor growth. Mechanistically, DAXX interacts with SREBP1 and SREBP2 and activates SREBP-mediated transcription. DAXX associates with lipogenic gene promoters through SREBPs. Underscoring the critical roles for the DAXX-SREBP interaction for lipogenesis, SREBP2 knockdown attenuates tumor growth in cells with DAXX overexpression, and DAXX mutants unable to bind SREBP1/2 have weakened activity in promoting lipogenesis and tumor growth. Remarkably, a DAXX mutant deficient of SUMO-binding fails to activate SREBP1/2 and lipogenesis due to impaired SREBP binding and chromatin recruitment and is defective of stimulating tumorigenesis. Hence, DAXX's SUMO-binding activity is critical to oncogenic lipogenesis. Notably, a peptide corresponding to DAXX's C-terminal SUMO-interacting motif (SIM2) is cell-membrane permeable, disrupts the DAXX-SREBP1/2 interactions, and inhibits lipogenesis and tumor growth. These results establish DAXX as a regulator of lipogenesis and a potential therapeutic target for cancer therapy.


Assuntos
Lipogênese , Neoplasias , Carcinogênese/genética , Transformação Celular Neoplásica , Proteínas Correpressoras/genética , Proteínas Correpressoras/metabolismo , Lipídeos , Lipogênese/genética , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 1/genética , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Animais , Camundongos
15.
J Biol Chem ; 286(39): 33954-62, 2011 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-21832044

RESUMO

Prognosis for patients with early stage kidney cancer has improved, but the treatment options for patients with locally advanced disease and metastasis remain few. Understanding the molecular mechanisms that regulate invasion and metastasis is critical for developing successful therapies to treat these patients. Proinflammatory prostaglandin E(2) plays an important role in cancer initiation and progression via activation of cognate EP receptors that belong to the superfamily of G protein-coupled receptors. Here we report that prostaglandin E(2) promotes renal cancer cell invasion through a signal transduction pathway that encompasses EP4 and small GTPase Rap. Inactivation of Rap signaling with Rap1GAP, like inhibition of EP4 signaling with ligand antagonist or knockdown with shRNA, reduces the kidney cancer cell invasion. Human kidney cells evidence increased EP4 and decreased Rap1GAP expression levels in the malignant compared with benign samples. These results support the idea that targeted inhibition of EP4 signaling and restoration of Rap1GAP expression constitute a new strategy to control kidney cancer progression.


Assuntos
Carcinoma de Células Renais/metabolismo , Dinoprostona/metabolismo , Proteínas Ativadoras de GTPase/biossíntese , Neoplasias Renais/metabolismo , Proteínas de Neoplasias/biossíntese , Receptores de Prostaglandina E Subtipo EP4/biossíntese , Transdução de Sinais , Carcinoma de Células Renais/tratamento farmacológico , Carcinoma de Células Renais/genética , Carcinoma de Células Renais/patologia , Linhagem Celular Tumoral , Dinoprostona/genética , Proteínas Ativadoras de GTPase/genética , Regulação Neoplásica da Expressão Gênica , Células HEK293 , Humanos , Neoplasias Renais/tratamento farmacológico , Neoplasias Renais/genética , Neoplasias Renais/patologia , Invasividade Neoplásica , Proteínas de Neoplasias/genética , Receptores de Prostaglandina E Subtipo EP4/genética
16.
J Gen Virol ; 93(Pt 10): 2109-2117, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22791607

RESUMO

Dynamin2 is a large GTPase that regulates vesicle trafficking, and the GTPase activity of dynamin2 is required for the multistep process of adenovirus infection. Activity of dynamin2 may be regulated by post-translational phosphorylation and S-nitrosylation modifications. In this study, we demonstrate a role for dynamin2 S-nitrosylation in adenovirus infection of epithelial cells. We show that adenovirus serotype 5 (Ad5) infection augments production of nitric oxide (NO) in epithelial cells and causes the S-nitrosylation of dynamin2, mainly on cysteine 86 (C86) and 607 (C607) residues. Forced overexpression of dynamin2 bearing C86A and/or C607A mutations decreases Ad5 infection. Diminishing NO synthesis by RNAi-induced knockdown of endogenous endothelial NO synthase (eNOS) expression attenuates virus infection of target cells. Ad5 infection promotes the kinetically dynamic S-nitrosylation of dynamin2 and eNOS: there is a rapid decrease in eNOS S-nitrosylation and a concomitant increase in the dynamin2 S-nitrosylation. These results support the hypothesis that dynamin2 S-nitrosylation following eNOS activation facilitates adenovirus infection of host epithelial cells.


Assuntos
Infecções por Adenoviridae/enzimologia , Adenoviridae/patogenicidade , Dinamina II/metabolismo , Células Epiteliais/virologia , Adenoviridae/genética , Adenoviridae/metabolismo , Infecções por Adenoviridae/genética , Infecções por Adenoviridae/metabolismo , Infecções por Adenoviridae/virologia , Animais , Células COS , Linhagem Celular , Chlorocebus aethiops , Cisteína/genética , Cisteína/metabolismo , Dinamina II/genética , Células Epiteliais/enzimologia , Células Epiteliais/metabolismo , Técnicas de Silenciamento de Genes , Humanos , Mutação , Óxido Nítrico/genética , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo III/genética , Óxido Nítrico Sintase Tipo III/metabolismo , Fosforilação , Processamento de Proteína Pós-Traducional , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo
17.
J Urol ; 187(1): 322-9, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22099983

RESUMO

PURPOSE: The limited success of cancer therapeutics is largely attributable to the ability of cancer to become resistant to conventional cytotoxic chemotherapy. Thus, further identification of signaling molecules and pathways that influence tumorigenesis is needed to increase the overall therapeutic options. GRKs, originally recognized for their conserved role in GPCR signal control, have now emerged as regulators of additional biological molecules and functions. MATERIALS AND METHODS: We used Western blot analysis to determine GRK expression in prostate cancer and RNA interference to establish the role of GRK5 in prostate cancer growth and progression through the cell cycle. RESULTS: GRK5 was expressed highly in the aggressive prostate cancer PC3 cell line and its silencing by RNA interference attenuated in vitro cell proliferation. PC3 cells that stably expressed lentiviral small hairpin RNA and targeted GRK5 evidence reduced xenograft tumor growth in mice. This was reversed by rescuing expression with wild-type but not with kinase inactive K215R GRK5, implying the need of GRK5 kinase activity for tumor growth. To investigate possible cellular mechanism(s) for GRK5 in cell growth regulation we tested whether kinase activity would impact cell cycle progression. Like forced over expression of kinase-inactive K215R GRK5, GRK5 knockdown led to G2/M arrest in the cell cycle. Also, evidence revealed that the loss of GRK5 activity resulted in decreased cyclin D1 expression, Rb protein phosphorylation and E2F target gene expression involved in cell cycle control. CONCLUSIONS: Results provide direct evidence that GRK5 has an immediate role in the regulation of prostate tumor growth.


Assuntos
Quinase 5 de Receptor Acoplado a Proteína G/fisiologia , Neoplasias da Próstata/patologia , Animais , Linhagem Celular Tumoral , Proliferação de Células , Progressão da Doença , Humanos , Masculino , Camundongos
18.
Proc Natl Acad Sci U S A ; 106(23): 9379-84, 2009 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-19458261

RESUMO

Androgen receptor (AR) signaling regulates the development and homeostasis of male reproductive organs, including the prostate. Deregulation of AR and AR coregulators, expression, or activity is involved in the initiation of prostate cancer and contributes to the transition of the disease to hormone-refractory stage. The ubiquitous betaArrestin proteins are now recognized as bona fide adapters and signal transducers with target effectors found in both the cytosol and nucleus. Here, we provide evidence that betaArrestin2 forms a complex with AR and acts as an AR corepressor in androgen-dependent prostate cancer cells. Accordingly, the forced overexpression of betaArrestin2 diminishes, and knockdown of betaArrestin2 expression with RNAi increases the androgen-induced prostate-specific antigen (PSA) gene expression. betaArrestin2 serves as an adapter, bringing into close proximity the Mdm2 E3 ligase and AR, thereby promoting AR ubiquitylation and degradation. Human prostate tissues evidence an inverse relationship between the expression of betaArrestin2 and AR activity: glands that express high levels of betaArrestin2 exhibit low expression of PSA, and those glands that express low levels of betaArrestin2 evidence elevated PSA levels. We conclude that betaArrestin2 acts as a corepressor of AR by serving as a scaffold for Mdm2 leading to the AR ubiquitylation and degradation.


Assuntos
Arrestinas/metabolismo , Neoplasias da Próstata/metabolismo , Receptores Androgênicos/metabolismo , Transdução de Sinais , Humanos , Masculino , Antígeno Prostático Específico , Neoplasias da Próstata/patologia , Estabilidade Proteica , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Receptores Androgênicos/análise , Ubiquitinação , beta-Arrestinas
19.
Cell Death Dis ; 13(10): 868, 2022 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-36229449

RESUMO

Androgen ablation therapy is the standard of care for newly diagnosed prostate cancer (PC) patients. PC that relapsed after hormonal therapy, referred to as castration-resistant PC (CRPC), often presents with metastasis (mCRPC) and is the major cause of disease lethality. The few available therapies for mCRPC include the Taxanes Docetaxel (DTX) and Cabazitaxel (CBZ). Alas, clinical success of Taxanes in mCRPC is limited by high intrinsic and acquired resistance. Therefore, it remains essential to develop rationally designed treatments for managing therapy-resistant mCRPC disease. The major effect of Taxanes on microtubule hyper-polymerization is a prolonged mitotic block due to activation of the Spindle Assembly Checkpoint (SAC). Taxane-sensitive cells eventually inactivate SAC and exit mitosis by mitotic catastrophe, resulting in genome instability and blockade of proliferation. Resistant cells remain in mitotic block, and, upon drug decay, resume mitosis and proliferation, underlying one resistance mechanism. In our study we explored the possibility of forced mitotic exit to elevate Taxane efficacy. Inactivation of the SAC component, mitotic checkpoint kinase Mps1/TTK with a small molecule inhibitor (Msp1i), potentiated efficacy of Taxanes treatment in both 2D cell culture and 3D prostasphere settings. Mechanistically, Mps1 inhibition forced mitotic catastrophe in cells blocked in mitosis by Taxanes. Androgen receptor (AR), the main driver of PC, is often mutated or truncated in mCRPC. Remarkably, Mps1i significantly potentiated CBZ cytotoxicity regardless of AR status, in both AR-WT and in AR-truncated CRPC cells. Overall, our data demonstrate that forced mitotic exit by Mps1 inhibition potentiates Taxanes efficacy. Given that several Mps1i's are currently in different stages of clinical trials, our results point to Mps1 as a new therapeutic target to potentiate efficacy of Taxanes in mCRPC patients.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Neoplasias de Próstata Resistentes à Castração , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Tirosina Quinases/metabolismo , Receptores Androgênicos , Androgênios/farmacologia , Hidrocarbonetos Aromáticos com Pontes , Docetaxel/farmacologia , Docetaxel/uso terapêutico , Resistencia a Medicamentos Antineoplásicos/genética , Humanos , Masculino , Neoplasias de Próstata Resistentes à Castração/tratamento farmacológico , Neoplasias de Próstata Resistentes à Castração/genética , Neoplasias de Próstata Resistentes à Castração/patologia , Receptores Androgênicos/genética , Taxoides/farmacologia , Taxoides/uso terapêutico
20.
Oncogene ; 40(14): 2610-2620, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33692468

RESUMO

Progression of prostate cancer (PC) to terminal castration-resistant PC (CRPC) involves a diverse set of intermediates, and androgen receptor (AR) is the key mediator of PC initiation and progression to CRPC. Hence, identification of factors involved in the regulation of AR expression and function is a necessary first-step to improve disease outcome. In this study, we identified ubiquitous ßArrestin 1 (ßArr1) as a regulator of AR function in CRPC. Unbiased gene expression analysis of public datasets revealed increased levels of ARRB1 (the gene encoding ßArr1) in CRPC when compared to normal tissue. Further, ßArr1 expression correlated with enhanced AR transcriptional function in these datasets. The ßArr1 partitions to both nucleus and cytosol and mechanistic studies showed that nuclear, and not cytosolic, ßArr1 formed a complex with AR and AR-coregulator ßCatenin and that the heterotrimeric protein complex was recruited to androgen-response elements of AR-regulated genes. Functionally, we demonstrate that depletion of ßArr1 attenuates PC cell and tumor growth and metastasis, and rescued expression of nuclear, but not cytosolic, ßArr1 restores the PC colony growth and invasion of Matrigel in vitro and tumor growth and metastasis in mice. The targeting of ßArr1-regulated AR transcriptional function may be used in the development of new drugs to treat lethal CRPC.


Assuntos
Neoplasias de Próstata Resistentes à Castração/metabolismo , Receptores Androgênicos/metabolismo , beta-Arrestina 1/metabolismo , Animais , Progressão da Doença , Xenoenxertos , Humanos , Masculino , Camundongos , Camundongos Nus , Neoplasias de Próstata Resistentes à Castração/patologia , beta-Arrestina 1/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA