Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 22(10)2021 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-34070125

RESUMO

The neuropeptide substance P (SP) mediates neurogenic inflammation and pain and contributes to atopic dermatitis in mice through the activation of mast cells (MCs) via Mas-related G protein-coupled receptor (GPCR)-B2 (MrgprB2, human ortholog MRGPRX2). In addition to G proteins, certain MRGPRX2 agonists activate an additional signaling pathway that involves the recruitment of ß-arrestins, which contributes to receptor internalization and desensitization (balanced agonists). We found that SP caused ß-arrestin recruitment, MRGPRX2 internalization, and desensitization. These responses were independent of G proteins, indicating that SP serves as a balanced agonist for MRGPRX2. A tyrosine residue in the highly conserved NPxxY motif contributes to the activation and internalization of many GPCRs. We have previously shown that Tyr279 of MRGPRX2 is essential for G protein-mediated signaling and degranulation. To assess its role in ß-arrestin-mediated MRGPRX2 regulation, we replaced Tyr279 in the NPxxY motif of MRGPRX2 with Ala (Y279A). Surprisingly, we found that, unlike the wild-type receptor, Y279A mutant of MRGPRX2 was resistant to SP-induced ß-arrestin recruitment and internalization. This study reveals the novel findings that activation of MRGPRX2 by SP is regulated by ß-arrestins and that a highly conserved tyrosine residue within MRGPRX2's NPxxY motif contributes to both G protein- and ß-arrestin-mediated responses.


Assuntos
Proteínas do Tecido Nervoso/agonistas , Receptores Acoplados a Proteínas G/agonistas , Receptores de Neuropeptídeos/agonistas , Substância P/metabolismo , beta-Arrestinas/metabolismo , Substituição de Aminoácidos , Animais , Degranulação Celular , Linhagem Celular , Feminino , Humanos , Masculino , Mastócitos/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutagênese Sítio-Dirigida , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/genética , Neuroimunomodulação/fisiologia , Ratos , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Neuropeptídeos/química , Receptores de Neuropeptídeos/genética , Tirosina/química , beta-Arrestina 2/deficiência , beta-Arrestina 2/genética , beta-Arrestina 2/metabolismo
2.
Eur J Pharmacol ; 899: 174007, 2021 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-33705801

RESUMO

G-protein-biased agonists with reduced ß-arrestin-2 activation are being investigated as safer alternatives to clinically-used opioids. ß-arrestin-2 has been implicated in the mechanism of opioid-induced antinociceptive tolerance. Opioid-induced analgesic tolerance is classically considered as centrally-mediated, but recent reports implicate nociceptive dorsal root ganglia neurons as critical mediators in this process. Here, we investigated the role of ß-arrestin-2 in the mechanism of opioid tolerance in dorsal root ganglia nociceptive neurons using ß-arrestin-2 knockout mice and the G-protein-biased µ-opioid receptor agonist, TRV130. Whole-cell current-clamp electrophysiology experiments revealed that 15-18-h overnight exposure to 10 µM morphine in vitro induced acute tolerance in ß-arrestin-2 wild-type but not knockout neurons. Furthermore, in wild-type neurons circumventing ß-arrestin-2 activation by overnight treatment with 200 nM TRV130 attenuated tolerance. Similarly, acute morphine tolerance in vivo in ß-arrestin-2 knockout mice was prevented in the warm-water tail-withdrawal assay. Treatment with 30 mg/kg TRV130 s.c. also inhibited acute antinociceptive tolerance in vivo in wild-type mice. Alternately, in ß-arrestin-2 knockout neurons tolerance induced by 7-day in vivo exposure to 50 mg morphine pellet was conserved. Likewise, ß-arrestin-2 deletion did not mitigate in vivo antinociceptive tolerance induced by 7-day exposure to 25 mg or 50 mg morphine pellet in both female or male mice, respectively. Consequently, these results indicated that ß-arrestin-2 mediates acute but not chronic opioid tolerance in dorsal root ganglia neurons and to antinociception in vivo. This suggests that opioid-induced antinociceptive tolerance may develop even in the absence of ß-arrestin-2 activation, and thus significantly affect the clinical utility of biased agonists.


Assuntos
Analgésicos Opioides/farmacologia , Tolerância a Medicamentos , Gânglios Espinais/efeitos dos fármacos , Morfina/farmacologia , Neurônios/efeitos dos fármacos , Dor Nociceptiva/prevenção & controle , Receptores Opioides mu/agonistas , Compostos de Espiro/farmacologia , Tiofenos/farmacologia , beta-Arrestina 2/metabolismo , Animais , Comportamento Animal/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Feminino , Gânglios Espinais/metabolismo , Gânglios Espinais/fisiopatologia , Masculino , Camundongos Knockout , Neurônios/metabolismo , Dor Nociceptiva/genética , Dor Nociceptiva/metabolismo , Dor Nociceptiva/fisiopatologia , Limiar da Dor/efeitos dos fármacos , Receptores Opioides mu/metabolismo , Fatores de Tempo , beta-Arrestina 2/deficiência , beta-Arrestina 2/genética
3.
J Neurophysiol ; 125(4): 1396-1407, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33656934

RESUMO

We have investigated the potential acute desensitizing role of the ß arrestin 2 (ß-arr2) pathway on the ventilatory depression produced by levels of fentanyl ranging from analgesic to life-threatening (0.1 to 60 mg/kg ip) in control and ß-arr2-deficient nonsedated mice. Fentanyl at doses of 0.1, 0.5, and 1 mg/kg ip-corresponding to the doses previously used to study the role of ß-arr2 pathway-decreased ventilation, but along the V̇e/V̇co2 relationship established in baseline conditions. This reduction in ventilation was therefore indistinguishable from the decrease in breathing during the periods of spontaneous immobility. Above 1.5 mg/kg, however, ventilation was depressed out of proportion of the changes in metabolic rate, suggesting a specific depression of the drive to breathe. The ventilatory responses were similar between the two groups. At high doses of fentanyl (60 mg/kg ip) 1 out of 20 control mice died by apnea versus 8 out of 20 ß-arr2-deficient mice (P = 0.008). In the surviving mice, ventilation was however identical in both groups. The ventilatory effects of fentanyl in ß-arr2-deficient mice, reported in the literature, are primarily mediated by the "indirect" effects of sedation/hypometabolism on breathing control. There was an excess mortality at very high doses of fentanyl in the ß-arr2-deficient mice, mechanisms of which are still open to question, as the capacity of maintaining a rhythmic, although profoundly depressed, breathing activity remains similar in all of the surviving control and ß-arr2-deficient mice.NEW & NOTEWORTHY When life-threatening doses of fentanyl are used in mice, the ß-arrestin 2 pathway appears to play a critical role in the recovery from opioid overdose. This observation calls into question the use of G protein-biased µ-opioid receptor agonists, as a strategy for safer opioid analgesic drugs.


Assuntos
Analgésicos Opioides/farmacologia , Apneia/induzido quimicamente , Fentanila/farmacologia , Overdose de Opiáceos/metabolismo , Respiração/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , beta-Arrestina 2/metabolismo , Analgésicos Opioides/administração & dosagem , Analgésicos Opioides/efeitos adversos , Animais , Comportamento Animal/efeitos dos fármacos , Modelos Animais de Doenças , Feminino , Fentanila/administração & dosagem , Fentanila/efeitos adversos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , beta-Arrestina 2/deficiência
4.
Acta Pharmacol Sin ; 42(5): 755-766, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-32855529

RESUMO

ß-arrestin2 (ß-arr2) is, a key protein that mediates desensitization and internalization of G protein-coupled receptors and participates in inflammatory and immune responses. Deficiency of ß-arr2 has been found to exacerbate collagen antibody-induced arthritis (CAIA) through unclear mechanisms. In this study we tried to elucidate the molecular mechanisms underlying ß-arr2 depletion-induced exacerbation of CAIA. CAIA was induced in ß-arr2-/- and wild-type (WT) mice by injection of collagen antibodies and LPS. The mice were sacrificed on d 13 after the injection, spleen, thymus and left ankle joints were collected for analysis. Arthritis index (AI) was evaluated every day or every 2 days. We showed that ß-arr2-/- mice with CAIA had a further increase in the percentage of plasma cells in spleen as compared with WT mice with CAIA, which was in accordance with elevated serum IgG1 and IgG2A expression and aggravating clinical performances, pathologic changes in joints and spleen, joint effusion, and joint blood flow. Both LPS stimulation of isolated B lymphocytes in vitro and TNP-LPS challenge in vivo led to significantly higher plasma cell formation and antibodies production in ß-arr2-/- mice as compared with WT mice. LPS treatment induced membrane distribution of toll-like receptor 4 (TLR4) on B lymphocytes, accordingly promoted the nuclear translocation of NF-κB and the transcription of Blimp1. Immunofluorescence analysis confirmed that more TLR4 colocalized with ß-arr2 in B lymphocytes in response to LPS stimulation. Depletion of ß-arr2 restrained TLR4 on B lymphocyte membrane after LPS treatment and further enhanced downstream NF-κB signaling leading to additional increment in plasma cell formation. In summary, ß-arr2 depletion exacerbates CAIA and further increases plasma cell differentiation and antibody production through inhibiting TLR4 endocytosis and aggravating NF-κB signaling.


Assuntos
Artrite Experimental/metabolismo , Artrite Reumatoide/metabolismo , Plasmócitos/metabolismo , beta-Arrestina 2/deficiência , Animais , Anticorpos Monoclonais/imunologia , Artrite Experimental/induzido quimicamente , Artrite Experimental/patologia , Artrite Reumatoide/induzido quimicamente , Artrite Reumatoide/patologia , Peso Corporal/fisiologia , Diferenciação Celular/fisiologia , Colágeno Tipo II/imunologia , Imunidade Humoral/fisiologia , Articulação do Joelho/metabolismo , Articulação do Joelho/patologia , Ativação Linfocitária/fisiologia , Masculino , Camundongos Endogâmicos C57BL , Transdução de Sinais/fisiologia , Receptor 4 Toll-Like/metabolismo
5.
Acta Pharmacol Sin ; 42(7): 1090-1100, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33116250

RESUMO

Hepatic fibrosis is a disease characterized by excessive deposition of extracellular matrix (ECM) in the liver. Activation of hepatic stellate cells (HSCs) is responsible for most of ECM production. Oxidative stress and reactive oxygen species (ROS) may be important factors leading to liver fibrosis. NADPH oxidase 4 (NOX4) is the main source of ROS in hepatic fibrosis, but the mechanism by which NOX4 regulates oxidative stress is not fully understood. ß-Arrestin2 is a multifunctional scaffold protein that regulates receptor endocytosis, signaling and trafficking. In this study, we investigated whether ß-arrestin2 regulated oxidative stress in hepatic fibrosis. Both ß-arrestin2 knockout (Arrb2 KO) mice and wild-type mice were intraperitoneally injected with carbon tetrachloride (CCl4) to induce hepatic fibrosis. Arrb2 KO mice showed significantly attenuated liver fibrosis, decreased ROS levels and NOX4 expression, and reduced collagen levels in their livers. In vitro, NOX4 knockdown significantly inhibited ROS production, and decreased expression of alpha-smooth muscle actin in angiotensin II-stimulated human HSC cell line LX-2. Through overexpression or depletion of ß-arrestin2 in LX-2 cells, we revealed that decreased ß-arrestin2 inhibited ROS levels and NOX4 expression, and reduced collagen production; it also inhibited activation of ERK and JNK signaling pathways. These results demonstrate that ß-arrestin2 deficiency protects against liver fibrosis by downregulating ROS production through NOX4. This effect appears to be mediated by ERK and JNK signaling pathways. Thus, targeted inhibition of ß-arrestin2 might reduce oxidative stress and inhibit the progression of liver fibrosis.


Assuntos
Cirrose Hepática/metabolismo , NADPH Oxidase 4/metabolismo , Estresse Oxidativo/fisiologia , beta-Arrestina 2/deficiência , Animais , Tetracloreto de Carbono , Colágeno/metabolismo , Regulação para Baixo/fisiologia , Técnicas de Inativação de Genes , Cirrose Hepática/induzido quimicamente , Cirrose Hepática/patologia , Metaloproteinase 13 da Matriz/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/fisiologia , Inibidor Tecidual de Metaloproteinase-1/metabolismo , beta-Arrestina 2/genética
6.
Cell Death Dis ; 11(5): 389, 2020 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-32439968

RESUMO

Hepatic fibrosis is a disease of the wound-healing response following chronic liver injury, and activated hepatic stellate cells (HSCs) play a crucial role in the progression of hepatic fibrosis. ß-arrestin2 functions as a multiprotein scaffold to coordinate complex signal transduction networks. Although ß-arrestin2 transduces diverse signals in cells, little is known about its involvement in the regulation of liver fibrosis. Our current study utilized a porcine serum-induced liver fibrosis model and found increased expression of ß-arrestin2 in hepatic tissues with the progression of hepatic fibrosis, which was positively correlated with collagen levels. Furthermore, changes in human fibrotic samples were also observed. We next used ß-arrestin2-/- mice to demonstrate that ß-arrestin2 deficiency ameliorates CCl4-induced liver fibrosis and decreases collagen deposition. The in vitro depletion and overexpression experiments showed that decreased ß-arrestin2 inhibited HSCs collagen production and elevated TßRIII expression, thus downregulating the TGF-ß1 pathway components Smad2, Smad3 and Akt. These findings suggest that ß-arrestin2 deficiency ameliorates liver fibrosis in mice, and ß-arrestin2 may be a potential treatment target in hepatic fibrosis.


Assuntos
Matriz Extracelular/metabolismo , Células Estreladas do Fígado/efeitos dos fármacos , Células Estreladas do Fígado/metabolismo , Cirrose Hepática/metabolismo , beta-Arrestina 2/deficiência , Animais , Células Cultivadas , Regulação para Baixo , Matriz Extracelular/efeitos dos fármacos , Fígado/metabolismo , Cirrose Hepática/patologia , Masculino , Substâncias Protetoras/metabolismo , Substâncias Protetoras/farmacologia , Ratos Wistar , Fator de Crescimento Transformador beta1/metabolismo , beta-Arrestina 2/metabolismo
7.
World J Gastroenterol ; 24(2): 216-225, 2018 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-29375207

RESUMO

AIM: To study the role and the possible mechanism of ß-arrestin 2 in lipopolysaccharide (LPS)-induced liver injury in vivo and in vitro. METHODS: Male ß-arrestin 2+/+ and ß-arrestin 2-/- C57BL/6J mice were used for in vivo experiments, and the mouse macrophage cell line RAW264.7 was used for in vitro experiments. The animal model was established via intraperitoneal injection of LPS or physiological sodium chloride solution. Blood samples and liver tissues were collected to analyze liver injury and levels of pro-inflammatory cytokines. Cultured cell extracts were collected to analyze the production of pro-inflammatory cytokines and expression of key molecules involved in the TLR4/NF-κB signaling pathway. RESULTS: Compared with wild-type mice, the ß-arrestin 2 knockout mice displayed more severe LPS-induced liver injury and significantly higher levels of pro-inflammatory cytokines, including interleukin (IL)-1ß, IL-6, tumor necrosis factor (TNF)-α, and IL-10. Compared with the control group, pro-inflammatory cytokines (including IL-1ß, IL-6, TNF-α, and IL-10) produced by RAW264.7 cells in the ß-arrestin 2 siRNA group were significantly increased at 6 h after treatment with LPS. Further, key molecules involved in the TLR4/NF-κB signaling pathway, including phospho-IκBα and phosho-p65, were upregulated. CONCLUSION: ß-arrestin 2 can protect liver tissue from LPS-induced injury via inhibition of TLR4/NF-κB signaling pathway-mediated inflammation.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas/prevenção & controle , Lipopolissacarídeos , Fígado/metabolismo , NF-kappa B/metabolismo , Transdução de Sinais , Receptor 4 Toll-Like/metabolismo , beta-Arrestina 2/metabolismo , Animais , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/patologia , Citocinas/metabolismo , Modelos Animais de Doenças , Mediadores da Inflamação/metabolismo , Fígado/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Inibidor de NF-kappaB alfa/metabolismo , Fosforilação , Células RAW 264.7 , Fator de Transcrição RelA/metabolismo , beta-Arrestina 2/deficiência , beta-Arrestina 2/genética
8.
JCI Insight ; 2(24)2017 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-29263312

RESUMO

Primary myelofibrosis is a myeloproliferative neoplasm associated with significant morbidity and mortality, for which effective therapies are lacking. ß-Arrestins are multifunctional adaptor proteins involved in developmental signaling pathways. One isoform, ß-arrestin2 (ßarr2), has been implicated in initiation and progression of chronic myeloid leukemia, another myeloproliferative neoplasm closely related to primary myelofibrosis. Accordingly, we investigated the relationship between ßarr2 and primary myelofibrosis. In a murine model of MPLW515L-mutant primary myelofibrosis, mice transplanted with donor ßarr2-knockout (ßarr2-/-) hematopoietic stem cells infected with MPL-mutant retrovirus did not develop myelofibrosis, whereas controls uniformly succumbed to disease. Although transplanted ßarr2-/- cells homed properly to marrow, they did not repopulate long-term due to increased apoptosis and decreased self-renewal of ßarr2-/- cells. In order to assess the effect of acute loss of ßarr2 in established primary myelofibrosis in vivo, we utilized a tamoxifen-induced Cre-conditional ßarr2-knockout mouse. Mice that received Cre (+) donor cells and developed myelofibrosis had significantly improved survival compared with controls. These data indicate that lack of antiapoptotic ßarr2 mediates marrow failure of murine hematopoietic stem cells overexpressing MPLW515L. They also indicate that ßarr2 is necessary for progression of primary myelofibrosis, suggesting that it may serve as a novel therapeutic target in this disease.


Assuntos
Mielofibrose Primária/metabolismo , beta-Arrestina 2/fisiologia , Animais , Apoptose/fisiologia , Medula Óssea/patologia , Proliferação de Células/fisiologia , Modelos Animais de Doenças , Progressão da Doença , Sobrevivência de Enxerto/fisiologia , Transplante de Células-Tronco Hematopoéticas/métodos , Células-Tronco Hematopoéticas/metabolismo , Masculino , Camundongos Knockout , Mielofibrose Primária/induzido quimicamente , Mielofibrose Primária/patologia , Tamoxifeno , beta-Arrestina 2/deficiência , beta-Arrestina 2/genética
9.
Cardiovasc Res ; 113(13): 1615-1626, 2017 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-29016703

RESUMO

AIMS: Ischemic heart disease is a leading cause of morbidity and mortality worldwide. Although timely restoration of coronary blood flow (reperfusion) is the most effective therapeutics of myocardial infarction, reperfusion causes further cardiac damage, i.e. ischemia-reperfusion (I/R) injury. ß-arrestins (Arrbs) have been traditionally defined as negative regulators of G protein-coupled receptor (GPCR) signalling, but recent studies have shown that they are essential for G protein-independent, GPCR-mediated biased signalling. Several ligands have been reported to be cardioprotective via Arrbs dependent pathway. However, it is unclear whether Arrbs exert receptor-independent physiological or pathological functions in the heart. Here, we sought to determine whether and how Arrbs play a role in regulating cardiomyocyte viability and myocardial remodelling following I/R injury. METHODS AND RESULTS: The expression of ß-arrestin 2 (Arrb2), but not ß-arrestin 1 (Arrb1), is upregulated in rat hearts subjected to I/R injury, or in cultured neonatal rat cardiomyocytes treated with hypoxia-reoxygenation (H/R) injury. Deficiency of Arrb2 in cultured neonatal rat cardiomyocytes alleviates H/R-induced cardiomyocyte death and Arrb2-/- mice are resistant to myocardial damage caused by I/R injury. In contrast, upregulation of Arrb2 triggers cardiomyocyte death and exaggerates I/R (or H/R)-induced detrimental effects. Mechanically, Arrb2 induces cardiomyocyte death by interacting with the p85 subunit of PI3K, and negatively regulating the formation of p85-PI3K/CaV3 survival complex, thus blocking activation of PI3K-Akt-GSK3ß cell survival signalling pathway. CONCLUSION: We define an upregulation of Arrb2 as a pathogenic factor in cardiac I/R injury, and also reveal a novel GPCR-independent mechanism of Arrb2-mediated cell death signalling in the heart.


Assuntos
Infarto do Miocárdio/metabolismo , Traumatismo por Reperfusão Miocárdica/metabolismo , Miócitos Cardíacos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , beta-Arrestina 2/metabolismo , Animais , Caveolina 3/metabolismo , Morte Celular , Sobrevivência Celular , Classe Ia de Fosfatidilinositol 3-Quinase/metabolismo , Modelos Animais de Doenças , Predisposição Genética para Doença , Glicogênio Sintase Quinase 3 beta/metabolismo , Preparação de Coração Isolado , Masculino , Camundongos Knockout , Infarto do Miocárdio/genética , Infarto do Miocárdio/patologia , Infarto do Miocárdio/prevenção & controle , Traumatismo por Reperfusão Miocárdica/genética , Traumatismo por Reperfusão Miocárdica/patologia , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Miócitos Cardíacos/patologia , Fenótipo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Transdução de Sinais , beta-Arrestina 1/genética , beta-Arrestina 1/metabolismo , beta-Arrestina 2/deficiência , beta-Arrestina 2/genética
10.
Circulation ; 135(1): 73-88, 2017 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-27815373

RESUMO

BACKGROUND: Type 2 diabetes mellitus (DM) and obesity independently increase the risk of heart failure by incompletely understood mechanisms. We propose that hyperinsulinemia might promote adverse consequences in the hearts of subjects with type-2 DM and obesity. METHODS: High-fat diet feeding was used to induce obesity and DM in wild-type mice or mice lacking ß2-adrenergic receptor (ß2AR) or ß-arrestin2. Wild-type mice fed with high-fat diet were treated with a ß-blocker carvedilol or a GRK2 (G-protein-coupled receptor kinase 2) inhibitor. We examined signaling and cardiac contractile function. RESULTS: High-fat diet feeding selectively increases the expression of phosphodiesterase 4D (PDE4D) in mouse hearts, in concert with reduced protein kinase A phosphorylation of phospholamban, which contributes to systolic and diastolic dysfunction. The expression of PDE4D is also elevated in human hearts with DM. The induction of PDE4D expression is mediated by an insulin receptor, insulin receptor substrate, and GRK2 and ß-arrestin2-dependent transactivation of a ß2AR-extracellular regulated protein kinase signaling cascade. Thus, pharmacological inhibition of ß2AR or GRK2, or genetic deletion of ß2AR or ß-arrestin2, all significantly attenuate insulin-induced phosphorylation of extracellular regulated protein kinase and PDE4D induction to prevent DM-related contractile dysfunction. CONCLUSIONS: These studies elucidate a novel mechanism by which hyperinsulinemia contributes to heart failure by increasing PDE4D expression and identify ß2AR or GRK2 as plausible therapeutic targets for preventing or treating heart failure in subjects with type 2 DM.


Assuntos
Diabetes Mellitus Tipo 2/complicações , Insuficiência Cardíaca/etiologia , Obesidade/complicações , Receptores Adrenérgicos beta 2/genética , Animais , Carbazóis/farmacologia , Carvedilol , Células Cultivadas , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/genética , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/metabolismo , Dieta Hiperlipídica , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Quinase 2 de Receptor Acoplado a Proteína G/antagonistas & inibidores , Quinase 2 de Receptor Acoplado a Proteína G/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Contração Miocárdica/efeitos dos fármacos , Miocárdio/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Propanolaminas/farmacologia , Receptores Adrenérgicos beta 2/deficiência , Transdução de Sinais , Vasodilatadores/farmacologia , beta-Arrestina 2/deficiência , beta-Arrestina 2/genética
11.
Proc Natl Acad Sci U S A ; 113(50): 14426-14431, 2016 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-27911784

RESUMO

The Frank-Starling law of the heart is a physiological phenomenon that describes an intrinsic property of heart muscle in which increased cardiac filling leads to enhanced cardiac contractility. Identified more than a century ago, the Frank-Starling relationship is currently known to involve length-dependent enhancement of cardiac myofilament Ca2+ sensitivity. However, the upstream molecular events that link cellular stretch to the length-dependent myofilament Ca2+ sensitivity are poorly understood. Because the angiotensin II type 1 receptor (AT1R) and the multifunctional transducer protein ß-arrestin have been shown to mediate mechanosensitive cellular signaling, we tested the hypothesis that these two proteins are involved in the Frank-Starling mechanism of the heart. Using invasive hemodynamics, we found that mice lacking ß-arrestin 1, ß-arrestin 2, or AT1R were unable to generate a Frank-Starling force in response to changes in cardiac volume. Although wild-type mice pretreated with the conventional AT1R blocker losartan were unable to enhance cardiac contractility with volume loading, treatment with a ß-arrestin-biased AT1R ligand to selectively activate ß-arrestin signaling preserved the Frank-Starling relationship. Importantly, in skinned muscle fiber preparations, we found markedly impaired length-dependent myofilament Ca2+ sensitivity in ß-arrestin 1, ß-arrestin 2, and AT1R knockout mice. Our data reveal ß-arrestin 1, ß-arrestin 2, and AT1R as key regulatory molecules in the Frank-Starling mechanism, which potentially can be targeted therapeutically with ß-arrestin-biased AT1R ligands.


Assuntos
Modelos Cardiovasculares , Contração Miocárdica/fisiologia , beta-Arrestina 1/fisiologia , beta-Arrestina 2/fisiologia , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Animais , Sinalização do Cálcio/fisiologia , Técnicas In Vitro , Losartan/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Contração Miocárdica/efeitos dos fármacos , Receptor Tipo 1 de Angiotensina/deficiência , Receptor Tipo 1 de Angiotensina/genética , Receptor Tipo 1 de Angiotensina/fisiologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , beta-Arrestina 1/deficiência , beta-Arrestina 1/genética , beta-Arrestina 2/deficiência , beta-Arrestina 2/genética
12.
J Pharmacol Exp Ther ; 359(3): 411-419, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27660244

RESUMO

Opioid receptors expressed by peripheral pain-sensing neurons are functionally inactive for antinociceptive signaling under most basal conditions; however, tissue damage or exposure to inflammatory mediators (e.g., bradykinin) converts these receptors from a nonresponsive state to a functionally competent state. Here we tested the hypothesis that the basal, nonresponsive state of the mu- and delta-opioid receptors (MOR and DOR, respectively) is the result of constitutive receptor activity that activates desensitization mechanisms, resulting in MOR and DOR receptor systems that are constitutively desensitized. Consistent with our previous findings, under basal conditions, neither the MOR agonist [d-Ala2,N-MePhe4,Gly-ol5]-enkephalin nor the DOR agonist [d-Pen2,5]-enkephalin, inhibited prostaglandin E2 (PGE2)-stimulated cAMP accumulation in peripheral sensory neurons in culture (ex vivo) or inhibited PGE2-stimulated thermal allodynia in the rat hind paw in vivo. Prolonged treatment with naloxone induced MOR and DOR responsiveness both in vivo and ex vivo to a similar magnitude as that produced by bradykinin. Also similar to bradykinin, the effect of naloxone persisted for 60 minutes after washout of the ligand. By contrast, prolonged treatment with 6ß-naltrexol, did not induce functional competence of MOR or DOR but blocked the effect of naloxone. Treatment with siRNA for ß-arrestin-2, but not ß-arrestin-1, also induced MOR and DOR functional competence in cultured peripheral sensory neurons. These data suggest that the lack of responsiveness of MOR and DOR to agonist for antinociceptive signaling in peripheral sensory neurons is due to constitutive desensitization that is likely mediated by ß-arrestin-2.


Assuntos
Receptores Opioides delta/metabolismo , Receptores Opioides mu/metabolismo , Células Receptoras Sensoriais/metabolismo , Animais , Bradicinina/farmacologia , Agonismo Inverso de Drogas , Regulação da Expressão Gênica/efeitos dos fármacos , Inativação Gênica , Masculino , Naloxona/farmacologia , Nociceptividade/efeitos dos fármacos , RNA Interferente Pequeno/genética , Ratos , Ratos Sprague-Dawley , Receptores Opioides delta/agonistas , Receptores Opioides mu/agonistas , Células Receptoras Sensoriais/efeitos dos fármacos , beta-Arrestina 2/deficiência , beta-Arrestina 2/genética
13.
J Pharmacol Exp Ther ; 359(1): 124-33, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27474750

RESUMO

Lysophosphatidic acid (LPA) is a lipid mediator that mediates cellular effects via G protein-coupled receptors (GPCRs). Epidermal growth factor (EGF) is a peptide that acts via a receptor tyrosine kinase. LPA and EGF both induce proliferation of prostate cancer cells and can transactivate each other's receptors. The LPA receptor LPA1 is particularly important for LPA response in human prostate cancer cells. Previous work in our laboratory has demonstrated that free fatty acid 4 (FFA4), a GPCR activated by ω-3 fatty acids, inhibits responses to both LPA and EGF in these cells. One potential mechanism for the inhibition involves negative interactions between FFA4 and LPA1, thereby suppressing responses to EGF that require LPA1 In the current study, we examined the role of LPA1 in mediating EGF and FFA4 agonist responses in two human prostate cancer cell lines, DU145 and PC-3. The results show that an LPA1-selective antagonist inhibits proliferation and migration to both LPA and EGF. Knockdown of LPA1 expression, using silencing RNA, blocks responses to LPA and significantly inhibits responses to EGF. The partial response to EGF that is observed after LPA1 knockdown is not inhibited by FFA4 agonists. Finally, the role of arrestin-3, a GPCR-binding protein that mediates many actions of activated GPCRs, was tested. Knockdown of arrestin-3 completely inhibits responses to both LPA and EGF in prostate cancer cells. Taken together, these results suggest that LPA1 plays a critical role in EGF responses and that FFA4 agonists inhibit proliferation by suppressing positive cross-talk between LPA1 and the EGF receptor.


Assuntos
Receptores ErbB/metabolismo , Neoplasias da Próstata/patologia , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Ácidos Lisofosfatídicos/metabolismo , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Fator de Crescimento Epidérmico/farmacologia , Técnicas de Silenciamento de Genes , Humanos , Lisofosfolipídeos/farmacologia , Masculino , Receptores Acoplados a Proteínas G/agonistas , Receptores de Ácidos Lisofosfatídicos/deficiência , Receptores de Ácidos Lisofosfatídicos/genética , beta-Arrestina 2/deficiência , beta-Arrestina 2/genética , beta-Arrestina 2/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA