Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Cancer Lett ; 571: 216333, 2023 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-37543278

RESUMO

The mechanisms underlying the progression of prostate cancer (PCa) to neuroendocrine prostate cancer (NEPC), an aggressive PCa variant, are largely unclear. Two prominent NEPC phenotypes are elevated NE marker expression and heightened angiogenesis. Identifying the still elusive direct molecular links connecting angiogenesis and neuroendocrine differentiation (NED) is crucial for our understanding and targeting of NEPC. Here we found that histone deacetylase 2 (HDAC2), whose role in NEPC has not been reported, is one of the most upregulated epigenetic regulators in NEPC. HDAC2 promotes both NED and angiogenesis. G protein-coupled receptor kinase 3 (GRK3), also upregulated in NEPC, is a critical promoter for both phenotypes too. Of note, GRK3 phosphorylates HDAC2 at S394, which enhances HDAC2's epigenetic repression of potent anti-angiogenic factor Thrombospondin 1 (TSP1) and master NE-repressor RE1 Silencing Transcription Factor (REST). Intriguingly, REST suppresses angiogenesis while TSP1 suppresses NE marker expression in PCa cells, indicative of their novel functions and their synergy in cross-repressing the two phenotypes. Furthermore, the GRK3-HDAC2 pathway is activated by androgen deprivation therapy and hypoxia, both known to promote NED and angiogenesis in PCa. These results indicate that NED and angiogenesis converge on GRK3-enhanced HDAC2 suppression of REST and TSP1, which constitutes a key missing link between two prominent phenotypes of NEPC.


Assuntos
Quinase 3 de Receptor Acoplado a Proteína G , Histona Desacetilase 2 , Neoplasias da Próstata , Humanos , Masculino , Antagonistas de Androgênios , Diferenciação Celular , Linhagem Celular Tumoral , Histona Desacetilase 2/genética , Histona Desacetilase 2/metabolismo , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Transdução de Sinais/genética , Quinase 3 de Receptor Acoplado a Proteína G/genética , Quinase 3 de Receptor Acoplado a Proteína G/metabolismo
2.
Cells ; 12(7)2023 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-37048054

RESUMO

Paget's Disease of Bone (PDB) is a metabolic bone disease that is characterized by dysregulated osteoclast function leading to focal abnormalities of bone remodeling. It can lead to pain, fracture, and bone deformity. G protein-coupled receptor kinase 3 (GRK3) is an important negative regulator of G protein-coupled receptor (GPCR) signaling. GRK3 is known to regulate GPCR function in osteoblasts and preosteoblasts, but its regulatory function in osteoclasts is not well defined. Here, we report that Grk3 expression increases during osteoclast differentiation in both human and mouse primary cells and established cell lines. We also show that aged mice deficient in Grk3 develop bone lesions similar to those seen in human PDB and other Paget's Disease mouse models. We show that a deficiency in Grk3 expression enhances osteoclastogenesis in vitro and proliferation of hematopoietic osteoclast precursors in vivo but does not affect the osteoclast-mediated bone resorption function or cellular senescence pathway. Notably, we also observe decreased Grk3 expression in peripheral blood mononuclear cells of patients with PDB compared with age- and gender-matched healthy controls. Our data suggest that GRK3 has relevance to the regulation of osteoclast differentiation and that it may have relevance to the pathogenesis of PDB and other metabolic bone diseases associated with osteoclast activation.


Assuntos
Doenças Ósseas Metabólicas , Reabsorção Óssea , Quinase 3 de Receptor Acoplado a Proteína G , Osteíte Deformante , Animais , Humanos , Camundongos , Doenças Ósseas Metabólicas/patologia , Reabsorção Óssea/metabolismo , Leucócitos Mononucleares/metabolismo , Osteíte Deformante/genética , Osteíte Deformante/metabolismo , Osteoclastos/metabolismo , Osteogênese , Quinase 3 de Receptor Acoplado a Proteína G/genética
3.
Sci Rep ; 10(1): 17395, 2020 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-33060647

RESUMO

Most G protein-coupled receptors (GPCRs) recruit ß-arrestins and internalize upon agonist stimulation. For the µ-opioid receptor (µ-OR), this process has been linked to development of opioid tolerance. GPCR kinases (GRKs), particularly GRK2 and GRK3, have been shown to be important for µ-OR recruitment of ß-arrestin and internalization. However, the contribution of GRK2 and GRK3 to ß-arrestin recruitment and receptor internalization, remain to be determined in their complete absence. Using CRISPR/Cas9-mediated genome editing we established HEK293 cells with knockout of GRK2, GRK3 or both to dissect their individual contributions in ß-arrestin2 recruitment and µ-OR internalization upon stimulation with four different agonists. We showed that GRK2/3 removal reduced agonist-induced µ-OR internalization and ß-arrestin2 recruitment substantially and we found GRK2 to be more important for these processes than GRK3. Furthermore, we observed a sustained and GRK2/3 independent component of ß-arrestin2 recruitment to the plasma membrane upon µ-OR activation. Rescue expression experiments restored GRK2/3 functions. Inhibition of GRK2/3 using the small molecule inhibitor CMPD101 showed a high similarity between the genetic and pharmacological approaches, cross-validating the specificity of both. However, off-target effects were observed at high CMPD101 concentrations. These GRK2/3 KO cell lines should prove useful for a wide range of studies on GPCR function.


Assuntos
Sistemas CRISPR-Cas , Endocitose/fisiologia , Quinase 2 de Receptor Acoplado a Proteína G/fisiologia , Quinase 3 de Receptor Acoplado a Proteína G/fisiologia , Receptores Opioides mu/metabolismo , beta-Arrestina 2/metabolismo , Quinase 2 de Receptor Acoplado a Proteína G/genética , Quinase 3 de Receptor Acoplado a Proteína G/genética , Edição de Genes , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Fosforilação , Reprodutibilidade dos Testes
4.
Biochem Biophys Res Commun ; 520(2): 327-332, 2019 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-31604529

RESUMO

The Raf kinase inhibitor protein (RKIP) activates ß-adrenoceptors (ß-AR) and thereby induces a well-tolerated cardiac contractility and prevents heart failure in mice. Different to RKIP-mediated ß-AR activation, chronic activation of ß-AR by catecholamines was shown to be detrimental for the heart. RKIP is an endogenous inhibitor of G protein coupled receptor kinase 2 (GRK2); it binds GRK2 and thereby inhibits GRK2 mediated ß-AR phosphorylation and desensitization. Here, we evaluate RKIP-mediated effects on ß-AR to explore new strategies for ß-AR modulation. Co-immunoprecipitation assays and pull-down assays revealed subtype specificity of RKIP for the cardiac GRK isoforms GRK2 and GRK3 - not GRK5 - as well as several RKIP binding sites within their N-termini (GRK21-185 and GRK31-185). Overexpression of these N-termini prevented ß2-AR phosphorylation and internalization, subsequently increased receptor signaling in HEK293 cells and cardiomyocyte contractility. Co-immunoprecipitation assays of ß2-AR with these N-terminal GRK fragments revealed a direct interaction suggesting a steric interference of the fragments with the functional GRK-receptor interaction. Altogether, N-termini of GRK2 and GRK3 efficiently simulate RKIP effects on ß-AR signaling in HEK293 cells and in cardiomyocytes by their binding to ß2-AR and, thus, provide important insights for the development of new strategies to modulate ß2-AR signaling.


Assuntos
Quinase 2 de Receptor Acoplado a Proteína G/metabolismo , Quinase 3 de Receptor Acoplado a Proteína G/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Animais , Sítios de Ligação , Células Cultivadas , Quinase 2 de Receptor Acoplado a Proteína G/genética , Quinase 3 de Receptor Acoplado a Proteína G/genética , Quinase 5 de Receptor Acoplado a Proteína G/metabolismo , Células HEK293 , Humanos , Camundongos Endogâmicos , Miócitos Cardíacos , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Proteína de Ligação a Fosfatidiletanolamina/metabolismo , Fosforilação , Receptores Adrenérgicos beta 2/genética
5.
Int J Mol Sci ; 20(15)2019 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-31366084

RESUMO

Duration of receptor antagonism, measured as the recovery of agonist responsiveness, is gaining attention as a method to evaluate the 'effective' target-residence for antagonists. These functional assays might be a good alternative for kinetic binding assays in competition with radiolabeled or fluorescent ligands, as they are performed on intact cells and better reflect consequences of dynamic cellular processes on duration of receptor antagonism. Here, we used a bioluminescence resonance energy transfer (BRET)-based assay that monitors heterotrimeric G protein activation via scavenging of released Venus-Gß1γ2 by NanoLuc (Nluc)-tagged membrane-associated-C-terminal fragment of G protein-coupled receptor kinase 3 (masGRK3ct-Nluc) as a tool to probe duration of G protein-coupled receptor (GPCR) antagonism. The Gαi-coupled histamine H3 receptor (H3R) was used in this study as prolonged antagonism is associated with adverse events (e.g., insomnia) and consequently, short-residence time ligands might be preferred. Due to its fast and prolonged response, this assay can be used to determine the duration of functional antagonism by measuring the recovery of agonist responsiveness upon washout of pre-bound antagonist, and to assess antagonist re-equilibration time via Schild-plot analysis. Re-equilibration of pre-incubated antagonist with agonist and receptor could be followed in time to monitor the transition from insurmountable to surmountable antagonism. The BRET-based G protein activation assay can detect differences in the recovery of H3R responsiveness and re-equilibration of pre-bound antagonists between the tested H3R antagonists. Fast dissociation kinetics were observed for marketed drug pitolisant (Wakix®) in this assay, which suggests that short residence time might be beneficial for therapeutic targeting of the H3R.


Assuntos
Técnicas de Transferência de Energia por Ressonância de Bioluminescência/métodos , Antagonistas dos Receptores Histamínicos H3/farmacologia , Receptores Histamínicos H3/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Quinase 3 de Receptor Acoplado a Proteína G/genética , Quinase 3 de Receptor Acoplado a Proteína G/metabolismo , Células HEK293 , Humanos , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Ligação Proteica
6.
Methods Mol Biol ; 1820: 33-41, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29884935

RESUMO

Like in other sensory systems, adaptation is an essential process in the olfactory system, required for its proper functioning. However, the precise molecular mechanism underlying the adaptation process has not been fully understood, especially at the receptor level. Here, we describe methods to evaluate the role of GRK3, one of the members of the GRK family responsible for the desensitization of non-olfactory G-protein-coupled receptor (GPCR), in desensitization of olfactory receptor (OR) using a heterologous expression system. As a parameter to characterize the degree of desensitization, we measure (1) the maximal response to an agonist by either cAMP or Ca2+ imaging assay and (2) the kinetic time course for recovery to basal levels by Ca2+ imaging assay. Differences in the degree of desensitization in the presence or absence of GRK3 can be examined by comparing these parameters, leading to evaluation of GRK3.


Assuntos
Sinalização do Cálcio , AMP Cíclico/metabolismo , Quinase 3 de Receptor Acoplado a Proteína G/metabolismo , Receptores Odorantes/metabolismo , Olfato , Quinase 3 de Receptor Acoplado a Proteína G/genética , Células HEK293 , Humanos , Receptores Odorantes/genética
7.
Am J Physiol Regul Integr Comp Physiol ; 314(4): R574-R583, 2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29212811

RESUMO

Increased ß-adrenergic receptor (ß-AR)-mediated activation of adenylyl cyclase (AC) in rat liver during aging has been linked to age-related increases in hepatic glucose output and hepatosteatosis. In this study, we investigated the expression of ß-ARs, individual receptor subtypes, and G protein-coupled receptor (GPCR) regulatory proteins in livers from aging rats. Radioligand-binding studies demonstrated that ß-AR density increased by greater than threefold in hepatocyte membranes from senescent (24-mo-old) compared with young adult (7-mo-old) rats and that this phenomenon was blocked by food restriction, which is known to retard aging processes in rodents. Competition-binding studies revealed a mixed population of ß1- and ß2-AR subtypes in liver membranes over the adult life span, with a trend for greater ß2-AR density with age. Expression of both ß-AR subtype mRNAs in rat liver increased with age, whereas ß2- but not ß1-AR protein levels declined in livers of old animals. Immunoreactive ß2- but not ß1-ARs were preferentially distributed in pericentral hepatic regions. Levels of GRK2/3 and ß-arrestin 2 proteins, which are involved in downregulation of agonist-activated GPCRs, including ß-ARs, increased during aging. Insofar as sympathetic tone increases with age, our findings suggest that, despite enhanced agonist-mediated downregulation of hepatic ß-ARs preferentially affecting the ß2-AR subtype, increased generation of both receptor subtypes during aging augments the pool of plasma membrane-bound ß-ARs coupled to AC in hepatocytes. This study thus identifies one or both ß-AR subtypes as possible therapeutic targets involved in aberrant hepatic processes of glucose and lipid metabolism during aging.


Assuntos
Envelhecimento/metabolismo , Membrana Celular/metabolismo , Metabolismo Energético , Hepatócitos/metabolismo , Fígado/metabolismo , Receptores Adrenérgicos beta 1/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Fatores Etários , Envelhecimento/genética , Animais , Restrição Calórica , Metabolismo Energético/genética , Quinase 2 de Receptor Acoplado a Proteína G/genética , Quinase 2 de Receptor Acoplado a Proteína G/metabolismo , Quinase 3 de Receptor Acoplado a Proteína G/genética , Quinase 3 de Receptor Acoplado a Proteína G/metabolismo , Regulação da Expressão Gênica , Glucose/metabolismo , Ligantes , Metabolismo dos Lipídeos , Fígado/fisiopatologia , Masculino , Ratos Endogâmicos F344 , Receptores Adrenérgicos beta 1/genética , Receptores Adrenérgicos beta 2/genética , beta-Arrestina 2/genética , beta-Arrestina 2/metabolismo
8.
J Mol Neurosci ; 63(3-4): 364-376, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29081032

RESUMO

ß-Adrenergic signaling plays an important role in regulating diverse brain functions and alterations in this signaling have been observed in different neuropathological conditions. In this study, we investigated the effect of a 10-day treatment with high doses of morphine (10 mg/kg per day) on major components and functional state of the ß-adrenergic receptor (ß-AR) signaling system in the rat cerebral cortex. ß-ARs were characterized by radioligand binding assays and amounts of various G protein subunits, adenylyl cyclase (AC) isoforms, G protein-coupled receptor kinases (GRKs), and ß-arrestin were examined by Western blot analysis. AC activity was determined as a measure of functionality of the signaling system. We also assessed the partitioning of selected signaling proteins between the lipid raft and non-raft fractions prepared from cerebrocortical plasma membranes. Morphine treatment resulted in a significant upregulation of ß-ARs, GRK3, and some AC isoforms (AC-I, -II, and -III). There was no change in quantity of G proteins and some other signaling molecules (AC-IV, AC-V/VI, GRK2, GRK5, GRK6, and ß-arrestin) compared with controls. Interestingly, morphine exposure caused a partial redistribution of ß-ARs, Gsα, Goα, and GRK2 between lipid rafts and bulk plasma membranes. Spatial localization of other signaling molecules within the plasma membrane was not changed. Basal as well as fluoride- and forskolin-stimulated AC activities were not significantly different in membrane preparations from control and morphine-treated animals. However, AC activity stimulated by the beta-AR agonist isoprenaline was markedly increased. This is the first study to demonstrate lipid raft association of key components of the cortical ß-AR system and its sensitivity to morphine.


Assuntos
Córtex Cerebral/efeitos dos fármacos , Microdomínios da Membrana/metabolismo , Morfina/farmacologia , Entorpecentes/farmacologia , Receptores Adrenérgicos beta/metabolismo , Transdução de Sinais , Adenilil Ciclases/genética , Adenilil Ciclases/metabolismo , Animais , Córtex Cerebral/metabolismo , Quinase 3 de Receptor Acoplado a Proteína G/genética , Quinase 3 de Receptor Acoplado a Proteína G/metabolismo , Masculino , Ratos , Ratos Wistar , beta-Arrestinas/genética , beta-Arrestinas/metabolismo
9.
Dis Markers ; 2017: 1202710, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29445249

RESUMO

Deregulation of G protein-coupled receptor kinase 3 (GRK3), which belongs to a subfamily of kinases called GRKs, acts as a promoter mechanism in some cancer types. Our study found that GRK3 was significantly overexpressed in 162 pairs of colon cancer tissues than in the matched noncancerous mucosa (P < 0.01). Based on immunohistochemistry staining of TMAs, GRK3 was dramatically stained positive in primary colon cancer (130/180, 72.22%), whereas it was detected minimally or negative in paired normal mucosa specimens (50/180, 27.78%). Overexpression of GRK3 was closely correlated with AJCC stage (P = 0.001), depth of tumor invasion (P < 0.001), lymph node involvement (P = 0.004), distant metastasis (P = 0.016), and histologic differentiation (P = 0.004). Overexpression of GRK3 is an independent prognostic indicator that correlates with poor survival in colon cancer patients. Consistent with this, downregulation of GRK3 exhibited decreased cell growth index, reduction in colony formation ability, elevated cell apoptosis rate, and impaired colon tumorigenicity in a xenograft model. Hence, a specific overexpression of GRK3 was observed in colon cancer, GRK3 potentially contributing to progression by mediating cancer cell proliferation and functions as a poor prognostic indicator in colon cancer and potentially represent a novel therapeutic target for the disease.


Assuntos
Neoplasias do Colo/patologia , Quinase 3 de Receptor Acoplado a Proteína G/genética , Quinase 3 de Receptor Acoplado a Proteína G/metabolismo , Regulação para Cima , Animais , Apoptose , Estudos de Casos e Controles , Linhagem Celular Tumoral , Proliferação de Células , Neoplasias do Colo/genética , Neoplasias do Colo/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Células HT29 , Humanos , Masculino , Camundongos , Metástase Neoplásica , Estadiamento de Neoplasias , Transplante de Neoplasias , Prognóstico , Análise de Sobrevida
10.
J Neuroimmunol ; 297: 103-16, 2016 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-27397083

RESUMO

The study investigated the influence of peripubertal ovariectomy on the thymic noradrenaline (NA) concentration, and the thymocyte NA content and ß2- and α1-adrenoceptor (AR) expression in adult 2- and 11-month-old rats. In control rats, the thymic NA concentration increased with age. This increase reflected rise in the density of catecholamine (CA)-containing fluorescent nerve fibers and cells and their CA content. Additionally, the average ß2- and α1-AR thymocyte surface density changed in the opposite direction with age; the density of ß2-AR decreased, whereas that of α1-AR increased. Ovariectomy diminished the thymic NA concentration in 2-month-old rats. This reflected the decrease in the density of fluorescent nerve fibers, and CA content in fluorescent nerve fibers and non-lymphoid cells, since the thymocyte NA content was increased in ovariectomized (Ox) rats. Estrogen supplementation prevented the ovariectomy-induced changes. In Ox rats, the density of CA-synthesizing nerve fibers and non-lymphoid cells diminished with age. To the contrary, NA content in thymocytes increased with age, but it did not exceed that in 11-month-old controls. Additionally, ovariectomy diminished the average thymocyte surface density of ß2-ARs, but it increased that of α1-ARs in 2-month-old-rats (due to estrogen, and estrogen and progesterone deficiency, respectively). These changes, despite of the rise in circulating estrogen level post-ovariectomy, remained stable with age. This most likely reflected a decreased sensitivity to estrogen action, as a consequence of the hormone misprinting in peripubertal age. The analysis of thymocyte proliferation in culture suggested that age- and ovariectomy-induced alterations in thymocyte NA synthesis and AR expression altered NA autocrine/paracrine action on thymocytes. In conclusion, the study indicates that the ovarian hormone deficiency in peripubertal age affects ovarian steroid-dependent remodeling of thymic adrenergic regulatory network in adult rats.


Assuntos
Regulação da Expressão Gênica/fisiologia , Rede Nervosa/metabolismo , Norepinefrina/metabolismo , Ovariectomia , Timócitos/metabolismo , Timo/citologia , Fatores Etários , Animais , Proliferação de Células , Cromatografia Líquida de Alta Pressão , Estradiol/sangue , Feminino , Citometria de Fluxo , Quinase 3 de Receptor Acoplado a Proteína G/genética , Quinase 3 de Receptor Acoplado a Proteína G/metabolismo , Progesterona/sangue , RNA Mensageiro/metabolismo , Ratos , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo , Tirosina 3-Mono-Oxigenase/genética , Tirosina 3-Mono-Oxigenase/metabolismo
11.
Oncotarget ; 7(29): 45171-45185, 2016 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-27191986

RESUMO

Neuroendocrine prostate cancer (NEPC) is an aggressive subtype of prostate cancer that commonly arises through neuroendocrine differentiation (NED) of prostate adenocarcinoma (PAC) after therapy, such as radiation therapy and androgen deprivation treatment (ADT). No effective therapeutic is available for NEPC and its molecular mechanisms remain poorly understood. We have reported that G protein-coupled receptor kinase 3 (GRK3, also called ADRBK2) promotes prostate cancer progression. In this study, we demonstrate that the ADT-activated cAMP response element binding protein (CREB) directly targets and induces GRK3. We show GRK3 expression is higher in NEPC than in PAC cells and mouse models, and it positively correlates with the expression and activity of CREB in human prostate cancers. Notably, overexpression of GRK3 in PAC cells increased the expression of NE markers in a kinase activity dependent manner. Conversely, silencing GRK3 blocked CREB-induced NED in PAC cells, reversed NE phenotypes and inhibited proliferation of NEPC cells. Taken together, these results indicate that GRK3 is a new critical activator of NE phenotypes and mediator of CREB activation in promoting NED of prostate cancer cells.


Assuntos
Carcinoma Neuroendócrino/patologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/fisiologia , Quinase 3 de Receptor Acoplado a Proteína G/fisiologia , Neoplasias da Próstata/patologia , Adenocarcinoma/patologia , Antagonistas de Androgênios/uso terapêutico , Animais , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Quinase 3 de Receptor Acoplado a Proteína G/análise , Quinase 3 de Receptor Acoplado a Proteína G/genética , Humanos , Masculino , Camundongos
12.
PLoS One ; 11(4): e0152856, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27049755

RESUMO

Triple negative breast cancer (TNBC) is a heterogeneous disease that has a poor prognosis and limited treatment options. Chemokine receptor interactions are important modulators of breast cancer metastasis; however, it is now recognized that quantitative surface expression of one important chemokine receptor, CXCR4, may not directly correlate with metastasis and that its functional activity in breast cancer may better inform tumor pathogenicity. G protein coupled receptor kinase 3 (GRK3) is a negative regulator of CXCR4 activity, and we show that GRK expression correlates with tumorigenicity, molecular subtype, and metastatic potential in human tumor microarray analysis. Using established human breast cancer cell lines and an immunocompetent in vivo mouse model, we further demonstrate that alterations in GRK3 expression levels in tumor cells directly affect migration and invasion in vitro and the establishment of distant metastasis in vivo. The effects of GRK3 modulation appear to be specific to chemokine-mediated migration behaviors without influencing tumor cell proliferation or survival. These data demonstrate that GRK3 dysregulation may play an important part in TNBC metastasis.


Assuntos
Neoplasias da Mama/patologia , Quinase 3 de Receptor Acoplado a Proteína G/fisiologia , Animais , Feminino , Quinase 3 de Receptor Acoplado a Proteína G/genética , Inativação Gênica , Humanos , Camundongos , Invasividade Neoplásica , Metástase Neoplásica
13.
Cell Signal ; 27(9): 1799-806, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26056051

RESUMO

G protein-coupled receptor desensitization is typically mediated by receptor phosphorylation by G protein-coupled receptor kinase (GRK) and subsequent arrestin binding; morphine, however, was previously found to activate a c-Jun N-terminal kinase (JNK)-dependent, GRK/arrestin-independent pathway to produce mu opioid receptor (MOR) inactivation in spinally-mediated, acute anti-nociceptive responses [Melief et al.] [1]. In the current study, we determined that JNK2 was also required for centrally-mediated analgesic tolerance to morphine using the hotplate assay. We compared JNK activation by morphine and fentanyl in JNK1(-/-), JNK2(-/-), JNK3(-/-), and GRK3(-/-) mice and found that both compounds specifically activate JNK2 in vivo; however, fentanyl activation of JNK2 was GRK3-dependent, whereas morphine activation of JNK2 was GRK3-independent. In MOR-GFP expressing HEK293 cells, treatment with either arrestin siRNA, the Src family kinase inhibitor PP2, or the protein kinase C (PKC) inhibitor Gö6976 indicated that morphine activated JNK2 through an arrestin-independent Src- and PKC-dependent mechanism, whereas fentanyl activated JNK2 through a Src-GRK3/arrestin-2-dependent and PKC-independent mechanism. This study resolves distinct ligand-directed mechanisms of JNK activation by mu opioid agonists and understanding ligand-directed signaling at MOR may improve opioid therapeutics.


Assuntos
Arrestina/metabolismo , Fentanila/farmacologia , Proteína Quinase 9 Ativada por Mitógeno/metabolismo , Morfina/farmacologia , Receptores Opioides mu/metabolismo , Animais , Arrestina/genética , Carbazóis/farmacologia , Ativação Enzimática/efeitos dos fármacos , Quinase 3 de Receptor Acoplado a Proteína G/genética , Quinase 3 de Receptor Acoplado a Proteína G/metabolismo , Células HEK293 , Humanos , Camundongos , Camundongos Knockout , Proteína Quinase 9 Ativada por Mitógeno/genética , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/genética , Proteína Quinase C/metabolismo , Receptores Opioides mu/genética , Quinases da Família src/genética , Quinases da Família src/metabolismo
14.
Sci Rep ; 5: 10920, 2015 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-26043205

RESUMO

Degeneration of dopaminergic neurons causes Parkinson's disease. Dopamine replacement therapy with L-DOPA is the best available treatment. However, patients develop L-DOPA-induced dyskinesia (LID). In the hemiparkinsonian rat, chronic L-DOPA increases rotations and abnormal involuntary movements modeling LID, via supersensitive dopamine receptors. Dopamine receptors are controlled by G protein-coupled receptor kinases (GRKs). Here we demonstrate that LID is attenuated by overexpression of GRK3 in the striatum, whereas knockdown of GRK3 by microRNA exacerbated it. Kinase-dead GRK3 and its separated RGS homology domain (RH) suppressed sensitization to L-DOPA, whereas GRK3 with disabled RH did not. RH alleviated LID without compromising anti-akinetic effect of L-DOPA. RH binds striatal Gq. GRK3, kinase-dead GRK3, and RH inhibited accumulation of ∆FosB, a marker of LID. RH-dead mutant was ineffective, whereas GRK3 knockdown exacerbated ∆FosB accumulation. Our findings reveal a novel mechanism of GRK3 control of the dopamine receptor signaling and the role of Gq in LID.


Assuntos
Discinesias/metabolismo , Quinase 3 de Receptor Acoplado a Proteína G/metabolismo , Levodopa/metabolismo , Doença de Parkinson/metabolismo , Domínios e Motivos de Interação entre Proteínas , Proteínas RGS/metabolismo , Animais , Comportamento Animal , Corpo Estriado/metabolismo , Modelos Animais de Doenças , Discinesias/etiologia , Quinase 3 de Receptor Acoplado a Proteína G/química , Quinase 3 de Receptor Acoplado a Proteína G/genética , Expressão Gênica , Técnicas de Silenciamento de Genes , Doença de Parkinson/complicações , Doença de Parkinson/genética , Interferência de RNA , RNA Interferente Pequeno/genética , Ratos , Transdução de Sinais
15.
Biol Psychiatry ; 76(10): 767-74, 2014 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-24629717

RESUMO

BACKGROUND: The clinical benefits of opioid drugs are counteracted by the development of tolerance and addiction. We provide in vivo evidence for the involvement of G protein-coupled receptor kinases (GRKs) in opioid dependence in addition to their roles in agonist-selective mu-opioid receptor (MOR) phosphorylation. METHODS: In vivo MOR phosphorylation was examined by immunoprecipitation and nanoflow liquid chromatography-tandem mass spectrometry analysis. Using the hot-plate and conditioned place preference test, we investigated opioid-related antinociception and reward effects in mice lacking GRK3 or GRK5. RESULTS: Etonitazene and fentanyl stimulated the in vivo phosphorylation of multiple carboxyl-terminal phosphate acceptor sites, including threonine 370, serine 375, and threonine 379, which was predominantly mediated by GRK3. By contrast, morphine promoted a selective phosphorylation of serine 375 that was predominantly mediated by GRK5. In contrast to GRK3 knockout mice, GRK5 knockout mice exhibited reduced antinociceptive responses after morphine administration and developed morphine tolerance similar to wild-type mice but fewer signs of physical dependence. Also, morphine was ineffective in inducing conditioned place preference in GRK5 knockout mice, whereas cocaine conditioned place preference was retained. However, the reward properties of morphine were evident in knock-in mice expressing a phosphorylation-deficient S375A mutation of the MOR. CONCLUSIONS: These findings show for the first time that MOR phosphorylation is regulated by agonist-selective recruitment of distinct GRK isoforms that influence different opioid-related behaviors. Modulation of GRK5 function could serve as a new approach for preventing addiction to opioids, while maintaining the analgesic properties of opioid drugs at an effective level.


Assuntos
Analgésicos Opioides/farmacologia , Quinase 5 de Receptor Acoplado a Proteína G/metabolismo , Dependência de Morfina/enzimologia , Morfina/farmacologia , Receptores Opioides mu/metabolismo , Recompensa , Animais , Benzimidazóis/farmacologia , Condicionamento Psicológico/efeitos dos fármacos , Condicionamento Psicológico/fisiologia , Tolerância a Medicamentos , Fentanila/farmacologia , Quinase 3 de Receptor Acoplado a Proteína G/genética , Quinase 3 de Receptor Acoplado a Proteína G/metabolismo , Quinase 5 de Receptor Acoplado a Proteína G/genética , Técnicas de Inativação de Genes , Camundongos , Camundongos Knockout , Nociceptividade/efeitos dos fármacos , Fosforilação , Isoformas de Proteínas/efeitos dos fármacos , Receptores Opioides mu/agonistas , Receptores Opioides mu/genética
16.
J Leukoc Biol ; 94(6): 1243-51, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23935208

RESUMO

Chemokine receptor interactions coordinate leukocyte migration in inflammation. Chemokine receptors are GPCRs that when activated, are phosphorylated by GRKs to turn off G protein-mediated signaling yet recruit additional signaling machinery. Recently, GRK3 was identified as a negative regulator of CXCL12/CXCR4 signaling that is defective in human WHIM syndrome. Here, we report that GRK3-/- mice exhibit numerous features of human WHIM, such as impaired CXCL12-mediated desensitization, enhanced CXCR4 signaling to ERK activation, altered granulocyte migration, and a mild myelokathexis. Moreover, GRK3-/- protects mice from two acute models of inflammatory arthritis (K/BxN serum transfer and CAIA). In these granulocyte-dependent disease models, protection of GRK3-/- mice is mediated by retention of cells in the marrow, fewer circulating granulocytes in the peripheral blood, and reduced granulocytes in the joints during active inflammation. In contrast to WHIM, GRK3-/- mice have minimal hypogammaglobulinemia and a peripheral leukocytosis with increased lymphocytes and absent neutropenia. Thus, we conclude that the loss of GRK3-mediated regulation of CXCL12/CXCR4 signaling contributes to some, but not all, of the complete WHIM phenotype and that GRK3 inhibition may be beneficial in the treatment of inflammatory arthritis.


Assuntos
Quinase 3 de Receptor Acoplado a Proteína G/imunologia , Síndromes de Imunodeficiência/imunologia , Sistema de Sinalização das MAP Quinases/imunologia , Verrugas/imunologia , Animais , Linhagem Celular Transformada , Quimiocina CXCL12/genética , Quimiocina CXCL12/imunologia , Quimiocina CXCL12/metabolismo , Modelos Animais de Doenças , Quinase 3 de Receptor Acoplado a Proteína G/genética , Quinase 3 de Receptor Acoplado a Proteína G/metabolismo , Granulócitos/enzimologia , Granulócitos/imunologia , Granulócitos/patologia , Humanos , Síndromes de Imunodeficiência/enzimologia , Síndromes de Imunodeficiência/genética , Síndromes de Imunodeficiência/patologia , Inflamação/enzimologia , Inflamação/genética , Inflamação/imunologia , Inflamação/patologia , Sistema de Sinalização das MAP Quinases/genética , Camundongos , Camundongos Knockout , Doenças da Imunodeficiência Primária , Receptores CXCR4/genética , Receptores CXCR4/imunologia , Receptores CXCR4/metabolismo , Verrugas/enzimologia , Verrugas/genética , Verrugas/patologia
17.
PLoS One ; 8(5): e64975, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23734232

RESUMO

The chemokine receptor CXCR4 regulates cell migration during ontogenesis and disease states including cancer and inflammation. Upon stimulation by the endogenous ligand CXCL12, CXCR4 becomes phosphorylated at multiple sites in its C-terminal domain. Mutations in the CXCR4 gene affecting C-terminal phosphorylation sites are a hallmark of WHIM syndrome, a genetic disorder characterized by a gain-of-CXCR4-function. To better understand how multi-site phosphorylation of CXCR4 is organized and how perturbed phosphorylation might affect CXCR4 function, we developed novel phosphosite-specific CXCR4 antibodies and studied the differential regulation and interaction of three C-terminal phosphorylation sites in human embryonic kidney cells (HEK293). CXCL12 promoted a robust phosphorylation at S346/347 which preceded phosphorylation at S324/325 and S338/339. After CXCL12 washout, the phosphosites S338/339 and S324/325 were rapidly dephosphorylated whereas phosphorylation at S346/347 was long-lasting. CXCL12-induced phosphorylation at S346/347 was staurosporine-insensitive and mediated by GRK2/3. WHIM syndrome-associated CXCR4 truncation mutants lacking the S346/347 phosphosite and the recently identified E343K WHIM mutant displayed strongly impaired phosphorylation at S324/325 and S338/339 as well as reduced CXCL12-induced receptor internalization. Relevance of the S346-S348 site was confirmed by a S346-348A mutant showing strongly impaired CXCL12-promoted phosphorylation at S324/325 and S338/339, defective internalization, gain of calcium mobilization, and reduced desensitization. Thus, the triple serine motif S346-S348 contains a major initial CXCR4 phosphorylation site and is required for efficient subsequent multi-site phosphorylation and receptor regulation. Hierarchical organization of CXCR4 phosphorylation explains why small deletions at the extreme CXCR4 C terminus typically associated with WHIM syndrome severely alter CXCR4 function.


Assuntos
Quimiocina CXCL12/metabolismo , Quinase 2 de Receptor Acoplado a Proteína G/metabolismo , Quinase 3 de Receptor Acoplado a Proteína G/metabolismo , Receptores CXCR4/metabolismo , Sítios de Ligação/genética , Cálcio/metabolismo , Quinase 2 de Receptor Acoplado a Proteína G/genética , Quinase 3 de Receptor Acoplado a Proteína G/genética , Células HEK293 , Humanos , Immunoblotting , Síndromes de Imunodeficiência/genética , Síndromes de Imunodeficiência/metabolismo , Cinética , Mutação , Fosforilação , Doenças da Imunodeficiência Primária , Interferência de RNA , Receptores CXCR4/genética , Serina/genética , Serina/metabolismo , Verrugas/genética , Verrugas/metabolismo
18.
Mol Biol Cell ; 24(11): 1649-60, S1-3, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23576548

RESUMO

Most α-synuclein (α-syn) deposited in Lewy bodies, the pathological hallmark of Parkinson disease (PD), is phosphorylated at Ser-129. However, the physiological and pathological roles of this modification are unclear. Here we investigate the effects of Ser-129 phosphorylation on dopamine (DA) uptake in dopaminergic SH-SY5Y cells expressing α-syn. Subcellular fractionation of small interfering RNA (siRNA)-treated cells shows that G protein-coupled receptor kinase 3 (GRK3), GRK5, GRK6, and casein kinase 2 (CK2) contribute to Ser-129 phosphorylation of membrane-associated α-syn, whereas cytosolic α-syn is phosphorylated exclusively by CK2. Expression of wild-type α-syn increases DA uptake, and this effect is diminished by introducing the S129A mutation into α-syn. However, wild-type and S129A α-syn equally increase the cell surface expression of dopamine transporter (DAT) in SH-SY5Y cells and nonneuronal HEK293 cells. In addition, siRNA-mediated knockdown of GRK5 or GRK6 significantly attenuates DA uptake without altering DAT cell surface expression, whereas knockdown of CK2 has no effect on uptake. Taken together, our results demonstrate that membrane-associated α-syn enhances DA uptake capacity of DAT by GRKs-mediated Ser-129 phosphorylation, suggesting that α-syn modulates intracellular DA levels with no functional redundancy in Ser-129 phosphorylation between GRKs and CK2.


Assuntos
Dopamina/metabolismo , Quinase 5 de Receptor Acoplado a Proteína G/genética , Quinases de Receptores Acoplados a Proteína G/genética , Neurônios/metabolismo , Serina/metabolismo , alfa-Sinucleína/genética , Transporte Biológico , Caseína Quinase II/antagonistas & inibidores , Caseína Quinase II/genética , Caseína Quinase II/metabolismo , Linhagem Celular , Membrana Celular/genética , Membrana Celular/metabolismo , Quinase 3 de Receptor Acoplado a Proteína G/genética , Quinase 3 de Receptor Acoplado a Proteína G/metabolismo , Quinase 5 de Receptor Acoplado a Proteína G/antagonistas & inibidores , Quinase 5 de Receptor Acoplado a Proteína G/metabolismo , Quinases de Receptores Acoplados a Proteína G/antagonistas & inibidores , Quinases de Receptores Acoplados a Proteína G/metabolismo , Regulação da Expressão Gênica , Células HEK293 , Humanos , Neurônios/citologia , Fosforilação , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , alfa-Sinucleína/metabolismo
19.
Immunobiology ; 218(7): 984-94, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23312955

RESUMO

The interaction between mycobacteria and epithelium is unexplored, but may determine the outcome of the infection. We have analyzed the role of two G protein-coupled receptors, CXCR1 and CXCR2 that are important regulators of many pulmonary diseases. We found that mycobacteria significantly increased the expression of both CXCR1 and CXCR2 on alveolar epithelial cells and both receptors were found to be important for neutrophil diapedesis across primary endothelial cells towards infected mucosa. Mycobacteria, lipoarabinomannan or 19-kDa glycolipoprotein up-regulated the inhibitory G protein-coupled receptor kinase (GRK)2, while GRK3 was less affected. Mycobacteria-induced GRK2 up-regulation decreased chemokine transcription and secretion thereby affecting the neutrophil recruitment to infected mucosa. These events were completely abolished by blocking these receptors prior to infection as the blocking increased epithelial immune responses. We have identified novel interactions occurring in the initial phase of mycobacterial infections by which mycobacterial manipulate epithelial inflammatory responses.


Assuntos
Células Epiteliais/imunologia , Quinase 2 de Receptor Acoplado a Proteína G/imunologia , Mycobacterium bovis/imunologia , Receptores de Interleucina-8A/imunologia , Receptores de Interleucina-8B/imunologia , Mucosa Respiratória/imunologia , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Quimiocinas/genética , Quimiocinas/imunologia , Técnicas de Cocultura , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/microbiologia , Quinase 2 de Receptor Acoplado a Proteína G/genética , Quinase 3 de Receptor Acoplado a Proteína G/genética , Quinase 3 de Receptor Acoplado a Proteína G/imunologia , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno/imunologia , Células Endoteliais da Veia Umbilical Humana/imunologia , Células Endoteliais da Veia Umbilical Humana/microbiologia , Humanos , Lipopolissacarídeos/farmacologia , Mycobacterium bovis/crescimento & desenvolvimento , Neutrófilos/imunologia , Neutrófilos/microbiologia , Receptores de Interleucina-8A/genética , Receptores de Interleucina-8B/genética , Mucosa Respiratória/efeitos dos fármacos , Mucosa Respiratória/microbiologia
20.
J Neurosci ; 32(49): 17582-96, 2012 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-23223282

RESUMO

Activation of the dynorphin/κ-opioid receptor (KOR) system by repeated stress exposure or agonist treatment produces place aversion, social avoidance, and reinstatement of extinguished cocaine place preference behaviors by stimulation of p38α MAPK, which subsequently causes the translocation of the serotonin transporter (SERT, SLC6A4) to the synaptic terminals of serotonergic neurons. In the present study we extend those findings by showing that stress-induced potentiation of cocaine conditioned place preference occurred by a similar mechanism. In addition, SERT knock-out mice did not show KOR-mediated aversion, and selective reexpression of SERT by lentiviral injection into the dorsal raphe restored the prodepressive effects of KOR activation. Kinetic analysis of several neurotransporters demonstrated that repeated swim stress exposure selectively increased the V(max) but not K(m) of SERT without affecting dopamine transport or the high-capacity, low-affinity transporters. Although the serotonergic neurons in the dorsal raphe project throughout the forebrain, a significant stress-induced increase in cell-surface SERT expression was only evident in the ventral striatum, and not in the dorsal striatum, hippocampus, prefrontal cortex, amygdala, or dorsal raphe. Stereotaxic microinjections of the long-lasting KOR antagonist norbinaltorphimine demonstrated that local KOR activation in the nucleus accumbens, but not dorsal raphe, mediated this stress-induced increase in ventral striatal surface SERT expression. Together, these results support the hypothesis that stress-induced activation of the dynorphin/KOR system produces a transient increase in serotonin transport locally in the ventral striatum that may underlie some of the adverse consequences of stress exposure, including the potentiation of the rewarding effects of cocaine.


Assuntos
Aprendizagem da Esquiva/fisiologia , Cocaína/farmacologia , Corpo Estriado/metabolismo , Dinorfinas/fisiologia , Recompensa , Proteínas da Membrana Plasmática de Transporte de Serotonina/metabolismo , Estresse Psicológico/metabolismo , Estresse Psicológico/psicologia , Animais , Aprendizagem da Esquiva/efeitos dos fármacos , Encéfalo/metabolismo , Dopamina/metabolismo , Dinorfinas/metabolismo , Quinase 3 de Receptor Acoplado a Proteína G/genética , Quinase 3 de Receptor Acoplado a Proteína G/fisiologia , Masculino , Proteínas de Membrana Transportadoras/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Microinjeções/métodos , Naltrexona/administração & dosagem , Naltrexona/análogos & derivados , Naltrexona/farmacologia , Antagonistas de Entorpecentes/administração & dosagem , Antagonistas de Entorpecentes/farmacocinética , Nicotina/efeitos adversos , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/metabolismo , Núcleos da Rafe/efeitos dos fármacos , Núcleos da Rafe/metabolismo , Núcleos da Rafe/fisiologia , Receptores Opioides kappa/antagonistas & inibidores , Receptores Opioides kappa/fisiologia , Serotonina/metabolismo , Proteínas da Membrana Plasmática de Transporte de Serotonina/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Síndrome de Abstinência a Substâncias/metabolismo , Sinaptossomos/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...