Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
1.
Pharmacol Res ; 170: 105744, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34182131

RESUMO

Enhanced platelet activation has been reported in patients with essential hypertension and heart failure. The possible contribution of platelet-derived thromboxane (TX)A2 in their pathophysiology remains unclear. We investigated the systemic TXA2 biosynthesis in vivo and gene expression of its receptor TP in 22 essential hypertension patients and a mouse model of salt-sensitive hypertension. The contribution of platelet TXA2 biosynthesis on enhanced blood pressure (BP) and overload-induced cardiac fibrosis was explored in mice by treating with low-dose Aspirin, resulting in selective inhibition of platelet cyclooxygenase (COX)-1-dependent TXA2 generation. In essential hypertensive patients, systemic biosynthesis of TXA2 [assessed by measuring its urinary metabolites (TXM) reflecting predominant platelet source] was enhanced together with higher gene expression of circulating leukocyte TP and TGF-ß, vs. normotensive controls. Similarly, in hypertensive mice with prostacyclin (PGI2) receptor (IP) deletion (IPKO) fed with a high-salt diet, enhanced urinary TXM, and left ventricular TP overexpression were detected vs. normotensive wildtype (WT) mice. Increased cardiac collagen deposition and profibrotic gene expression (including TGF-ß) was found. Low-dose Aspirin administration caused a selective inhibition of platelet TXA2 biosynthesis and mitigated enhanced blood pressure, cardiac fibrosis, and left ventricular profibrotic gene expression in IPKO but not WT mice. Moreover, the number of myofibroblasts and extravasated platelets in the heart was reduced. In cocultures of human platelets and myofibroblasts, platelet TXA2 induced profibrotic gene expression, including TGF-ß1. In conclusion, our results support tailoring low-dose Aspirin treatment in hypertensive patients with unconstrained TXA2/TP pathway to reduce blood pressure and prevent early cardiac fibrosis.


Assuntos
Antifibróticos/farmacologia , Anti-Hipertensivos/farmacologia , Aspirina/farmacologia , Plaquetas/efeitos dos fármacos , Pressão Sanguínea/efeitos dos fármacos , Cardiomiopatias/prevenção & controle , Hipertensão Essencial/tratamento farmacológico , Miócitos Cardíacos/efeitos dos fármacos , Inibidores da Agregação Plaquetária/farmacologia , Tromboxano A2/sangue , Adulto , Animais , Biomarcadores/sangue , Plaquetas/metabolismo , Cardiomiopatias/sangue , Cardiomiopatias/etiologia , Cardiomiopatias/patologia , Estudos de Casos e Controles , Células Cultivadas , Modelos Animais de Doenças , Hipertensão Essencial/sangue , Hipertensão Essencial/complicações , Hipertensão Essencial/fisiopatologia , Feminino , Fibrose , Humanos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Miofibroblastos/efeitos dos fármacos , Miofibroblastos/metabolismo , Miofibroblastos/patologia , Receptores de Epoprostenol/genética , Receptores de Epoprostenol/metabolismo , Receptores de Tromboxanos/metabolismo
2.
J Clin Invest ; 131(7)2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33529171

RESUMO

Tregs restrain both the innate and adaptive immune systems to maintain homeostasis. Allergic airway inflammation, characterized by a Th2 response that results from a breakdown of tolerance to innocuous environmental antigens, is negatively regulated by Tregs. We previously reported that prostaglandin I2 (PGI2) promoted immune tolerance in models of allergic inflammation; however, the effect of PGI2 on Treg function was not investigated. Tregs from mice deficient in the PGI2 receptor IP (IP KO) had impaired suppressive capabilities during allergic airway inflammatory responses compared with mice in which PGI2 signaling was intact. IP KO Tregs had significantly enhanced expression of immunoglobulin-like transcript 3 (ILT3) compared with WT Tregs, which may contribute to the impairment of the IP KO Treg's ability to suppress Th2 responses. Using fate-mapping mice, we reported that PGI2 signaling prevents Treg reprogramming toward a pathogenic phenotype. PGI2 analogs promoted the differentiation of naive T cells to Tregs in both mice and humans via repression of ß-catenin signaling. Finally, a missense variant in IP in humans was strongly associated with chronic obstructive asthma. Together, these data support that PGI2 signaling licenses Treg suppressive function and that PGI2 is a therapeutic target for enhancing Treg function.


Assuntos
Asma/imunologia , Reprogramação Celular/imunologia , Epoprostenol/imunologia , Tolerância Imunológica , Transdução de Sinais/imunologia , Linfócitos T Reguladores/imunologia , Animais , Asma/genética , Asma/patologia , Reprogramação Celular/genética , Doença Crônica , Epoprostenol/genética , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Receptores de Epoprostenol/genética , Receptores de Epoprostenol/imunologia , Transdução de Sinais/genética , Linfócitos T Reguladores/patologia
3.
Cardiovasc Res ; 117(4): 1154-1165, 2021 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-32531060

RESUMO

AIMS: Fibromuscular dysplasia (FMD) and spontaneous coronary artery dissection (SCAD) are related, non-atherosclerotic arterial diseases mainly affecting middle-aged women. Little is known about their physiopathological mechanisms. We aimed to identify rare genetic causes to elucidate molecular mechanisms implicated in FMD and SCAD. METHODS AND RESULTS: We analysed 29 exomes that included familial and sporadic FMD. We identified one rare loss-of-function variant (LoF) (frequencygnomAD = 0.000075) shared by two FMD sisters in the prostaglandin I2 receptor gene (PTGIR), a key player in vascular remodelling. Follow-up was conducted by targeted or Sanger sequencing (1071 FMD and 363 SCAD patients) or lookups in exome (264 FMD) or genome sequences (480 SCAD), all independent and unrelated. It revealed four additional LoF allele carriers, in addition to several rare missense variants, among FMD patients, and two LoF allele carriers among SCAD patients, including one carrying a rare splicing mutation (c.768 + 1C>G). We used burden test to test for enrichment in patients compared to gnomAD controls, which detected a putative enrichment in FMD (PTRAPD = 8 × 10-4), but not a significant enrichment (PTRAPD = 0.12) in SCAD. The biological effects of variants on human prostaclycin receptor (hIP) signalling and protein expression were characterized using transient overexpression in human cells. We confirmed the LoFs (Q163X and P17RfsX6) and one missense (L67P), identified in one FMD and one SCAD patient, to severely impair hIP function in vitro. CONCLUSIONS: Our study shows that rare genetic mutations in PTGIR are enriched among FMD patients and found in SCAD patients, suggesting a role for prostacyclin signalling in non-atherosclerotic stenosis and dissection.


Assuntos
Anomalias dos Vasos Coronários/genética , Displasia Fibromuscular/genética , Mutação com Perda de Função , Mutação de Sentido Incorreto , Receptores de Epoprostenol/genética , Doenças Vasculares/congênito , Adulto , Idoso , Austrália , Anomalias dos Vasos Coronários/diagnóstico , Anomalias dos Vasos Coronários/metabolismo , Análise Mutacional de DNA , Bases de Dados Genéticas , Europa (Continente) , Feminino , Displasia Fibromuscular/diagnóstico , Displasia Fibromuscular/metabolismo , Predisposição Genética para Doença , Células HEK293 , Humanos , Masculino , Pessoa de Meia-Idade , Fenótipo , Valor Preditivo dos Testes , Receptores de Epoprostenol/metabolismo , Medição de Risco , Fatores de Risco , Estados Unidos , Doenças Vasculares/diagnóstico , Doenças Vasculares/genética , Doenças Vasculares/metabolismo
4.
Cardiovasc Res ; 116(12): 1972-1980, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31688905

RESUMO

AIMS: Cardiovascular side effects caused by non-steroidal anti-inflammatory drugs (NSAIDs), which all inhibit cyclooxygenase (COX)-2, have prevented development of new drugs that target prostaglandins to treat inflammation and cancer. Microsomal prostaglandin E synthase-1 (mPGES-1) inhibitors have efficacy in the NSAID arena but their cardiovascular safety is not known. Our previous work identified asymmetric dimethylarginine (ADMA), an inhibitor of endothelial nitric oxide synthase, as a potential biomarker of cardiovascular toxicity associated with blockade of COX-2. Here, we have used pharmacological tools and genetically modified mice to delineate mPGES-1 and COX-2 in the regulation of ADMA. METHODS AND RESULTS: Inhibition of COX-2 but not mPGES-1 deletion resulted in increased plasma ADMA levels. mPGES-1 deletion but not COX-2 inhibition resulted in increased plasma prostacyclin levels. These differences were explained by distinct compartmentalization of COX-2 and mPGES-1 in the kidney. Data from prostanoid synthase/receptor knockout mice showed that the COX-2/ADMA axis is controlled by prostacyclin receptors (IP and PPARß/δ) and the inhibitory PGE2 receptor EP4, but not other PGE2 receptors. CONCLUSION: These data demonstrate that inhibition of mPGES-1 spares the renal COX-2/ADMA pathway and define mechanistically how COX-2 regulates ADMA.


Assuntos
Aorta/enzimologia , Arginina/análogos & derivados , Ciclo-Oxigenase 2/metabolismo , Rim/enzimologia , Prostaglandina-E Sintases/metabolismo , Animais , Aorta/efeitos dos fármacos , Arginina/sangue , Inibidores de Ciclo-Oxigenase 2/farmacologia , Sistema Enzimático do Citocromo P-450/genética , Sistema Enzimático do Citocromo P-450/metabolismo , Feminino , Oxirredutases Intramoleculares/genética , Oxirredutases Intramoleculares/metabolismo , Rim/efeitos dos fármacos , Masculino , Camundongos Knockout , PPAR beta/genética , PPAR beta/metabolismo , Prostaglandina-E Sintases/antagonistas & inibidores , Prostaglandina-E Sintases/genética , Prostaglandinas I/sangue , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores de Epoprostenol/genética , Receptores de Epoprostenol/metabolismo , Receptores de Prostaglandina E/genética , Receptores de Prostaglandina E/metabolismo
5.
Biol Reprod ; 100(1): 162-174, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30481284

RESUMO

The prostacyclin (prostaglandin I2) signaling system is an essential regulator of vascular homeostasis. Since the corpus luteum is a highly vascularized gland, prostacyclin seems to be crucial for luteal development and function. Although progress has been made in understanding the luteotropic action of prostacyclin in mammals, its role in the porcine corpus luteum remains to be determined. Therefore, studies were conducted to (1) determine profiles of prostacyclin synthase expression and prostacyclin metabolite concentration, as well as prostacyclin G-protein-coupled receptor expression in porcine luteal tissue on days 2 to 16 of the estrous cycle and days 10 to 30 of pregnancy using real-time PCR, western blot, or enzyme immunoassay; and (2) examine the effect of prostacyclin on progesterone synthesis in vitro. To accomplish the second aim, luteal cells were treated with prostacyclin analogs, iloprost and carbaprostacyclin, in the presence or absence of prostacyclin receptor antagonists. The mRNA expression of cytochrome P450 family 11 subfamily A member 1 and hydroxy-delta-5-steroid dehydrogenase, 3 beta- and steroid delta-isomerase 1 was analyzed using real-time PCR, while progesterone concentration in culture medium was assessed by radioimmunoassay.Dynamic changes of prostacyclin synthase and prostacyclin receptor expression were observed in porcine luteal tissue during the estrous cycle and early pregnancy. Moreover, prostacyclin stimulated progesterone production and this effect was abolished by the addition of prostacyclin receptor antagonists. Our findings provide strong evidence that prostacyclin and its signaling system are present in corpus luteum of the pig and may directly promote luteotropic activity through upregulation of progesterone synthesis.


Assuntos
Corpo Lúteo/metabolismo , Epoprostenol/biossíntese , Células Lúteas/metabolismo , Receptores de Epoprostenol/genética , Animais , Células Cultivadas , Corpo Lúteo/citologia , Feminino , Expressão Gênica , Gravidez , Receptores de Epoprostenol/metabolismo , Suínos
6.
Am J Respir Cell Mol Biol ; 60(5): 578-591, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30537446

RESUMO

Idiopathic pulmonary fibrosis is a life-threatening progressive disease characterized by loss of alveolar epithelial cells, inflammation, and aberrant fibroblast activation. The two currently approved therapies do not halt or reverse tissue remodeling, and therefore novel disease-modifying mechanisms are needed. Our results describe YAP/TAZ inhibition through prostacyclin (IP) receptor activation as a novel mechanism that suppresses profibrotic (myo)fibroblast activity. We investigated the antifibrotic properties of the selective IP receptor agonist ACT-333679 using primary human lung fibroblasts. ACT-333679 prevented transforming growth factor ß1-induced fibroblast-to-myofibroblast transition, proliferation, extracellular matrix synthesis, and IL-6 and PAI-1 secretion, and exerted relaxant effects in cell contraction assays. ACT-333679 treatment also reverted an established myofibroblast phenotype. Unbiased analysis of ACT-333679-induced whole-genome expression changes in transforming growth factor ß1-treated fibroblasts identified significant attenuation of genes regulated by YAP/TAZ, two transcriptional cofactors that are essential for fibrosis. We then demonstrated that ACT-333679, via elevation of cAMP, caused YAP/TAZ nuclear exclusion and subsequent suppression of YAP/TAZ-dependent profibrotic gene transcription. In summary, we offer a rationale for further exploring the potential of IP receptor agonists for the treatment of idiopathic pulmonary fibrosis.


Assuntos
Acetatos/farmacologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Fibroblastos/efeitos dos fármacos , Miofibroblastos/efeitos dos fármacos , Pirazinas/farmacologia , Receptores de Epoprostenol/genética , Fatores de Transcrição/genética , Aciltransferases , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Estudos de Casos e Controles , Diferenciação Celular , Proliferação de Células , AMP Cíclico/metabolismo , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patologia , Regulação da Expressão Gênica , Humanos , Fibrose Pulmonar Idiopática/tratamento farmacológico , Fibrose Pulmonar Idiopática/genética , Fibrose Pulmonar Idiopática/metabolismo , Fibrose Pulmonar Idiopática/patologia , Interleucina-6/genética , Interleucina-6/metabolismo , Pulmão/metabolismo , Pulmão/patologia , Masculino , Miofibroblastos/metabolismo , Miofibroblastos/patologia , Inibidor 1 de Ativador de Plasminogênio/genética , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Receptores de Epoprostenol/agonistas , Receptores de Epoprostenol/metabolismo , Transdução de Sinais , Fatores de Transcrição/metabolismo , Transcrição Gênica , Fator de Crescimento Transformador beta1/farmacologia , Proteínas de Sinalização YAP
7.
Arterioscler Thromb Vasc Biol ; 38(5): 1115-1124, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29599139

RESUMO

OBJECTIVE: Deletion of mPGES-1 (microsomal prostaglandin E synthase-1)-an anti-inflammatory target alternative to COX (cyclooxygenase)-2-attenuates injury-induced neointima formation in mice. This is attributable to the augmented levels of PGI2 (prostacyclin)-a known restraint of the vascular response to injury, acting via IP (I prostanoid receptor). To examine the role of mPGES-1-derived PGE2 (prostaglandin E2) in vascular remodeling without the IP. APPROACH AND RESULTS: Mice deficient in both IP and mPGES-1 (DKO [double knockout] and littermate controls [IP KO (knockout)]) were subjected to angioplasty wire injury. Compared with the deletion of IP alone, coincident deletion of IP and mPGES-1 increased neointima formation, without affecting media area. Early pathological changes include impaired reendothelialization and increased leukocyte invasion in neointima. Endothelial cells (ECs), but not vascular smooth muscle cells, isolated from DKOs exhibited impaired cell proliferation. Activation of EP (E prostanoid receptor) 4 (and EP2, to a lesser extent), but not of EP1 or EP3, promoted EC proliferation. EP4 antagonism inhibited proliferation of mPGES-1-competent ECs, but not of mPGES-1-deficient ECs, which showed suppressed PGE2 production. EP4 activation inhibited leukocyte adhesion to ECs in vitro, promoted reendothelialization, and limited neointima formation post-injury in the mouse. Endothelium-restricted deletion of EP4 in mice suppressed reendothelialization, increased neointimal leukocytes, and exacerbated neointimal formation. CONCLUSIONS: Removal of the IP receptors unmasks a protective role of mPGES-1-derived PGE2 in limiting injury-induced vascular hyperplasia. EP4, in the endothelial compartment, is essential to promote reendothelialization and restrain neointimal formation after injury. Activating EP4 bears therapeutic potential to prevent restenosis after percutaneous coronary intervention.


Assuntos
Proliferação de Células , Dinoprostona/metabolismo , Células Endoteliais/enzimologia , Artéria Femoral/enzimologia , Prostaglandina-E Sintases/metabolismo , Receptores de Epoprostenol/metabolismo , Receptores de Prostaglandina E Subtipo EP4/metabolismo , Lesões do Sistema Vascular/enzimologia , Animais , Adesão Celular , Células Cultivadas , Modelos Animais de Doenças , Células Endoteliais/patologia , Feminino , Artéria Femoral/lesões , Artéria Femoral/patologia , Humanos , Leucócitos/metabolismo , Leucócitos/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso/enzimologia , Músculo Liso/patologia , Neointima , Prostaglandina-E Sintases/deficiência , Prostaglandina-E Sintases/genética , Reepitelização , Receptores de Epoprostenol/deficiência , Receptores de Epoprostenol/genética , Receptores de Prostaglandina E Subtipo EP4/deficiência , Receptores de Prostaglandina E Subtipo EP4/genética , Transdução de Sinais , Lesões do Sistema Vascular/genética , Lesões do Sistema Vascular/patologia
8.
Odontology ; 106(1): 2-10, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28631175

RESUMO

This study aimed to analyze the mRNA expression and protein localization of prostaglandin I2 (PGI2) synthase (PGIS), the PGI2 receptor (IP receptor) and transient receptor potential cation channel, subfamily V, member 1 (TRPV1) in force-stimulated rat molars, toward the elucidation of the PGI2-IP receptor-TRPV1 pathway that is in operation in the pulp and possibly associated with orthodontic pain and inflammation. Experimental force was applied to the maxillary first and second molars by inserting an elastic band between them for 6-72 h. PGIS, PTGIR (the IP receptor gene), and TRPV1 mRNA levels in the coronal pulp were analyzed with real-time PCR. PGIS, IP receptor, and TRPV1 proteins were immunostained. The force stimulation induced significant upregulation of PGIS at 6-24 h, and PTGIR and TRPV1 at 6 and 12 h in the pulp. PGIS was immunolocalized in odontoblasts and some fibroblasts in the force-stimulated pulp. The IP receptor and TRPV1 immunoreactivities were detected on odontoblasts and some nerve fibers. It was concluded that PGIS, PTGIR, and TRPV1 in rat molar pulp were significantly upregulated shortly after the force application, and that the IP receptor was co-expressed on TRPV1-expressing nerves and odontoblasts. These findings suggest that the PGI2-IP receptor-TRPV1 pathway is associated with the acute phase of force-induced pulp changes involving odontoblasts and nerves.


Assuntos
Sistema Enzimático do Citocromo P-450/genética , Polpa Dentária/metabolismo , Expressão Gênica , Oxirredutases Intramoleculares/genética , Receptores de Epoprostenol/genética , Canais de Cátion TRPV/genética , Técnicas de Movimentação Dentária , Animais , Técnicas Imunoenzimáticas , Masculino , Dente Molar , Odontoblastos/metabolismo , RNA Mensageiro/genética , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase em Tempo Real , Fatores de Tempo , Regulação para Cima
9.
FASEB J ; 32(5): 2354-2365, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29247122

RESUMO

Nonalcoholic steatohepatitis (NASH) is a hepatic manifestation of metabolic syndrome. Although the prostaglandin (PG)I2 receptor IP is expressed broadly in the liver, the role of PGI2-IP signaling in the development of NASH remains to be determined. Here, we investigated the role of the PGI2-IP system in the development of steatohepatitis using mice lacking the PGI2 receptor IP [IP-knockout (IP-KO) mice] and beraprost (BPS), a specific IP agonist. IP-KO and wild-type (WT) mice were fed a methionine- and choline-deficient diet (MCDD) for 2, 5, or 10 wk. BPS was administered orally to mice every day during the experimental periods. The effect of BPS on the expression of chemokine and inflammatory cytokines was examined also in cultured Kupffer cells. WT mice fed MCDD developed steatohepatitis at 10 wk. IP-KO mice developed steatohepatitis at 5 wk with augmented histologic derangements accompanied by increased hepatic monocyte chemoattractant protein-1 (MCP-1) and TNF-α concentrations. After 10 wk of MCDD, IP-KO mice had greater hepatic iron deposition with prominent oxidative stress, resulting in hepatocyte damage. In WT mice, BPS improved histologic and biochemical parameters of steatohepatitis, accompanied by reduced hepatic concentration of MCP-1 and TNF-α. Accordingly, BPS suppressed the LPS-stimulated Mcp-1 and Tnf-α mRNA expression in cultured Kupffer cells prepared from WT mice. PGI2-IP signaling plays a crucial role in the development and progression of steatohepatitis by modulating the inflammatory response, leading to augmented oxidative stress. We suggest that the PGI2-IP system is an attractive therapeutic target for treating patients with NASH.-Kumei, S., Yuhki, K.-I., Kojima, F., Kashiwagi, H., Imamichi, Y., Okumura, T., Narumiya, S., Ushikubi, F. Prostaglandin I2 suppresses the development of diet-induced nonalcoholic steatohepatitis in mice.


Assuntos
Epoprostenol/farmacologia , Alimentos Formulados/efeitos adversos , Hepatócitos/metabolismo , Células de Kupffer/metabolismo , Fígado/metabolismo , Hepatopatia Gordurosa não Alcoólica/prevenção & controle , Animais , Quimiocina CCL2/biossíntese , Quimiocina CCL2/genética , Epoprostenol/análogos & derivados , Regulação da Expressão Gênica/efeitos dos fármacos , Hepatócitos/patologia , Células de Kupffer/patologia , Fígado/patologia , Camundongos , Camundongos Knockout , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/metabolismo , Hepatopatia Gordurosa não Alcoólica/patologia , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/genética , Receptores de Epoprostenol/agonistas , Receptores de Epoprostenol/genética , Receptores de Epoprostenol/metabolismo , Fatores de Tempo , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/genética
10.
Biochim Biophys Acta Mol Cell Res ; 1864(10): 1819-1832, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28739266

RESUMO

The cellular trafficking of numerous G protein-coupled receptors (GPCRs) is known to be regulated by Rab proteins that involves a direct protein:protein interaction between the receptor and the GTPase. In the case of the human prostacyclin receptor (hIP), it undergoes agonist-induced internalization and subsequent Rab11a-dependent recyclization involving an interaction between a Rab11-binding domain (RBD) localized within its carboxyl-tail domain with Rab11a. However, the GPCR-interacting domain on Rab11a itself is unknown. Hence, we sought to identify the region within Rab11a that mediates its interaction with the RBD of the hIP. The α4 helix region of Rab11 was identified as a novel binding domain for the hIP, a site entirely distinct from the Switch I/Switch II -regions that act as specific binding domain for most other Rab and Ras-like GTPase interactants. Specifically, Glu138 within α4 helix of Rab11a appears to contact with key residues (e.g. Lys304) within the RBD of the hIP, where such contacts differ depending on the agonist-activated versus -inactive status of the hIP. Through mutational studies, supported by in silico homology modelling of the inactive and active hIP:Rab11a complexes, a mechanism is proposed to explain both the constitutive and agonist-induced binding of Rab11a to regulate intracellular trafficking of the hIP. Collectively, these studies are not only the first to identify α4 helix of Rab11a as a protein binding domain on the GTPase but also reveal novel mechanistic insights into the intracellular trafficking of the hIP, and potentially of other members of the GPCR superfamily, involving Rab11-dependent mechanisms.


Assuntos
Domínios e Motivos de Interação entre Proteínas/genética , Receptores de Epoprostenol/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo , Sequência de Aminoácidos/genética , Células HeLa , Humanos , Ligação Proteica , Conformação Proteica em alfa-Hélice/genética , Transporte Proteico/genética , Receptores de Epoprostenol/genética , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Proteínas rab de Ligação ao GTP/química , Proteínas rab de Ligação ao GTP/genética
11.
Andrology ; 5(5): 971-978, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28544631

RESUMO

Infertility affects 10-15% of couples, and approximately 50% of cases are linked to male factor infertility. The purpose of this study was to evaluate the DNA methylation patterns in spermatozoa from males who are suffering from a reduction in fecundity. Thirty samples were subjected to 450K arrays as a screening study to evaluate the variation in sperm DNA methylation levels between cases and controls groups, and then four CpG sites (cg05799088, cg07227024, cg16338278, and cg08408433) underwent to deep bisulfite sequencing to validate the observed methylation differences in 111 samples (56 proven fertile males as 'controls' and 55 males suffering from a reduction in fecundity as 'cases'). A significant difference in the mean methylation level was found between cases and controls in the CpGs of PRICKLE2 gene-related amplicon (CpG1, p ≤ 0.002, and CpG2, p ≤ 0.004) and CpG of ALS2CR12 gene-related amplicon (CpG1, p ≤ 0.015, and CpG2, p ≤ 0.009). Besides, a significant difference was found at seven from thirteen CpGs tested in the ALDH3B2 gene amplicon CpG2, CpG6, CpG9, CpG10, CpG11, CpG12, and CpG13 (p ≤ 0.005, p ≤ 0.004, p ≤ 0.012, p ≤ 0.028, p ≤ 0.012, p ≤ 0.009, and p ≤ 0.001, respectively). In addition, the results showed that nine CpGs out of the twenty-six within the PTGIR gene-related amplicon (CpG4, CpG6, CpG8, CpG9, CpG11, CpG15, CpG19, CpG23, and CpG26) had a significant difference in their mean methylation level (p ≤ 0.006, p ≤ 0.009, p ≤ 0.003, p ≤ 0.003, p ≤ 0.007, p ≤ 0.002, p ≤ 0.018, p ≤ 0.018, and p ≤ 0.040, respectively) in the case vs. CONTROL GROUP: In conclusion, an alteration in the methylation levels of sperm DNA from males with reduced fecundity was observed. In addition, an association between changes in the methylation level for these CpGs and different semen parameters has been found.


Assuntos
Metilação de DNA , Infertilidade Masculina/genética , Espermatozoides/metabolismo , Adulto , Ilhas de CpG , Humanos , Proteínas com Domínio LIM/genética , Masculino , Proteínas de Membrana/genética , Metilação , Pessoa de Meia-Idade , Proteínas/genética , Receptores de Epoprostenol/genética , Adulto Jovem
12.
Bioorg Med Chem Lett ; 27(11): 2567-2570, 2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28462839

RESUMO

We present a practical synthesis of both enantiomers of 1,2,3,4-tetrahydroisoquinoline derivative IPPAM-1 (1), which is a positive allosteric modulator (PAM) of prostacyclin receptor (IP) and a candidate for treatment of pulmonary arterial hypertension without the side effects caused by IP agonists. Assay of cAMP production by CHO-K1 cells stably expressing human IP clearly demonstrated that the IPPAM activity resides exclusively on the R-form of 1.


Assuntos
Anti-Hipertensivos/química , Receptores de Epoprostenol/metabolismo , Tetra-Hidroisoquinolinas/química , Regulação Alostérica , Animais , Anti-Hipertensivos/síntese química , Anti-Hipertensivos/uso terapêutico , Células CHO , Cricetinae , Cricetulus , AMP Cíclico/metabolismo , Humanos , Hipertensão Pulmonar/tratamento farmacológico , Receptores de Epoprostenol/agonistas , Receptores de Epoprostenol/genética , Estereoisomerismo , Tetra-Hidroisoquinolinas/síntese química , Tetra-Hidroisoquinolinas/uso terapêutico
13.
Eur J Haematol ; 98(6): 615-621, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28306170

RESUMO

AIMS: Platelets retain cytoplasmic messenger RNA and are capable of protein biosynthesis. Several diseases are known to impact the platelet transcriptome but the effect of non-valvular atrial fibrillation (NVAF) on platelet RNA transcript is essentially unknown. The aim of this study was to evaluate the impact of NVAF on platelet RNA transcript by measuring platelet genes expression in consecutive NVAF patients before and 3-4 months after pulmonary vein isolation (PVI) and compared to normal sinus rhythm controls (NSR). METHODS AND RESULTS: RNA from isolated platelets were reverse transcribed, assayed against 15 genes using real-time qPCR, and expressed as mean cycle threshold (ΔCt) using beta-2-microglobulin as endogenous control. Expression of all evaluated genes, except cathepsin A gene, was significantly lower (higher ΔCt) in 103 NVAF patients compared to 55 NSR controls. Insulin-like growth factor binding protein acid labile subunit gene (IGFALS) had expression more than 16 fold-lower (17.0±2.8 vs 12.5±3.8, P<.001), follow by genes encoding for prostacyclin receptor, and for von Willebrand factor which had fourfold lower expression compared to NSR controls. Gender, type of atrial fibrillation, heart failure, hypertension, prior stroke, diabetes mellitus, and atherosclerosis were associated with different gene expression. Following PVI, expression of four genes significantly increased, particularly IGFALS gene (increased 256-fold) and ADAMT gene increased 16-fold); expression of three genes significantly decreased, and expression of eight genes has not changed. CONCLUSIONS: Platelets are capable to respond to the circulatory environment of NVAF by altering transcript and changing prothrombotic status. This shows platelet potential for molecular "reprogramming" possibly induced by flow disturbances of NVAF.


Assuntos
Aterosclerose/sangue , Fibrilação Atrial/sangue , Plaquetas/metabolismo , Diabetes Mellitus/sangue , Insuficiência Cardíaca/sangue , Hipertensão/sangue , Proteína ADAMTS13/sangue , Proteína ADAMTS13/genética , Idoso , Aterosclerose/fisiopatologia , Aterosclerose/cirurgia , Fibrilação Atrial/fisiopatologia , Fibrilação Atrial/cirurgia , Plaquetas/patologia , Proteínas de Transporte/sangue , Proteínas de Transporte/genética , Estudos de Casos e Controles , Diabetes Mellitus/fisiopatologia , Diabetes Mellitus/cirurgia , Feminino , Expressão Gênica , Glicoproteínas/sangue , Glicoproteínas/genética , Insuficiência Cardíaca/fisiopatologia , Insuficiência Cardíaca/cirurgia , Humanos , Hipertensão/fisiopatologia , Hipertensão/cirurgia , Masculino , Pessoa de Meia-Idade , Veias Pulmonares/cirurgia , Receptores de Epoprostenol/sangue , Receptores de Epoprostenol/genética , Fatores Sexuais , Fator de von Willebrand/genética , Fator de von Willebrand/metabolismo
14.
J Ocul Pharmacol Ther ; 33(3): 186-192, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28072560

RESUMO

PURPOSE: To investigate the therapeutic potential of a prostacyclin (IP) receptor agonist for ocular inflammation and the effect on immune cells. METHODS: The anti-inflammatory activities of cicaprost were determined in primary human monocyte-derived macrophages and human monocyte-derived dendritic cells (MoDC), as well as a lipopolysaccharides (LPS)-induced rat uveitis model. Multiple cytokine release was measured by utilizing Luminex Technology. Prostacyclin (IP) Receptor expression was detected by reverse transcription-polymerase chain receptor. Leukocyte infiltration and protein exudation in the rat uveitis model were measured using a hemocytometer and protein concentration by a NanoDrop instrument. RESULTS: Cicapost, an IP receptor agonist, potently inhibits proinflammatory chemokines/cytokine production not only from LPS- or TNFα (tumor necrosis factor-alpha)-induced primary human monocyte-derived macrophages, but also from LPS-stimulated MoDC. While constitutively expressed in macrophages, the IP receptor was inducible by LPS stimulation in MoDCs. In a LPS-induced rat uveitis model, cicaprost efficaciously prevents ocular inflammatory cell and protein leakage, as well as inflammatory cytokine release. CONCLUSION: The IP receptor agonist cicaprost is a potent anti-inflammatory agent, implicating that the tightly controlled PGI2/IP signaling pathway is important in regulating inflammation. This response could be harnessed in ocular inflammatory disease where steroids are currently the standard of care.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Células Dendríticas/efeitos dos fármacos , Modelos Animais de Doenças , Epoprostenol/análogos & derivados , Macrófagos/efeitos dos fármacos , Uveíte/tratamento farmacológico , Animais , Anti-Inflamatórios não Esteroides/administração & dosagem , Citocinas/antagonistas & inibidores , Citocinas/biossíntese , Células Dendríticas/metabolismo , Epoprostenol/administração & dosagem , Epoprostenol/farmacologia , Humanos , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Lipopolissacarídeos/antagonistas & inibidores , Macrófagos/metabolismo , Masculino , Ratos Endogâmicos Lew , Receptores de Epoprostenol/agonistas , Receptores de Epoprostenol/genética , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Uveíte/induzido quimicamente
15.
J Immunol ; 197(5): 1577-86, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27456482

RESUMO

Allergic airway diseases are immune disorders associated with heightened type 2 immune responses and IL-5 and IL-13 production at the site of inflammation. We have previously reported that cyclooxygenase (COX) inhibition by indomethacin augmented allergic airway inflammation in a STAT6-independent manner. However, the key COX product(s) responsible for restraining indomethacin-mediated STAT6-independent allergic inflammation is unknown. In this study, using the mouse model of OVA-induced allergic airway inflammation, we identified that PGI2 receptor (IP) signaling was critical for indomethacin-induced, STAT6-independent proallergic effects. We demonstrated that IP deficiency increased inflammatory cell infiltration, eosinophilia, and IL-5 and IL-13 expression in the lung in a STAT6-independent manner. The augmented STAT6-independent allergic inflammation correlated with enhanced primary immune responses to allergic sensitization and elevated production of multiple inflammatory chemokines (CCL11, CCL17, CCL22, and CXCL12) in the lung after allergen challenge. We also showed that the PGI2 analogue cicaprost inhibited CD4 T cell proliferation and IL-5 and IL-13 expression in vitro, and IP deficiency diminished the stimulatory effect of indomethacin on STAT6-independent IL-5 and IL-13 responses in vivo. The inhibitory effects of PGI2 and the IP signaling pathway on CD4 T cell activation, inflammatory chemokine production, and allergic sensitization and airway inflammation suggest that PGI2 and its analogue iloprost, both Food and Drug Administration-approved drugs, may be useful in treating allergic diseases and asthma. In addition, inhibiting PGI2 signaling by drugs that either block PGI2 production or restrain IP signaling may augment STAT6-independent pathways of allergic inflammation.


Assuntos
Alérgenos/imunologia , Pulmão/imunologia , Ativação Linfocitária/efeitos dos fármacos , Receptores de Epoprostenol/metabolismo , Fator de Transcrição STAT6/metabolismo , Alérgenos/administração & dosagem , Animais , Anti-Hipertensivos/farmacologia , Asma/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/fisiologia , Proliferação de Células , Quimiocinas/biossíntese , Quimiocinas/imunologia , Epoprostenol/administração & dosagem , Epoprostenol/análogos & derivados , Epoprostenol/farmacologia , Hipersensibilidade , Indometacina , Inflamação , Interleucina-13/genética , Interleucina-13/imunologia , Interleucina-5/genética , Interleucina-5/imunologia , Pulmão/fisiopatologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Ovalbumina/imunologia , Receptores de Epoprostenol/deficiência , Receptores de Epoprostenol/genética , Fator de Transcrição STAT6/deficiência , Fator de Transcrição STAT6/genética , Fator de Transcrição STAT6/imunologia , Transdução de Sinais , Células Th2/imunologia
16.
Diabetes ; 65(5): 1398-409, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26868296

RESUMO

Discovery of common pathways that mediate both pancreatic ß-cell function and end-organ function offers the opportunity to develop therapies that modulate glucose homeostasis and separately slow the development of diabetes complications. Here, we investigated the in vitro and in vivo effects of pharmacological agonism of the prostaglandin I2 (IP) receptor in pancreatic ß-cells and in glomerular podocytes. The IP receptor agonist MRE-269 increased intracellular 3',5'-cyclic adenosine monophosphate (cAMP), augmented glucose-stimulated insulin secretion (GSIS), and increased viability in MIN6 ß-cells. Its prodrug form, selexipag, augmented GSIS and preserved islet ß-cell mass in diabetic mice. Determining that this preservation of ß-cell function is mediated through cAMP/protein kinase A (PKA)/nephrin-dependent pathways, we found that PKA inhibition, nephrin knockdown, or targeted mutation of phosphorylated nephrin tyrosine residues 1176 and 1193 abrogated the actions of MRE-269 in MIN6 cells. Because nephrin is important to glomerular permselectivity, we next set out to determine whether IP receptor agonism similarly affects nephrin phosphorylation in podocytes. Expression of the IP receptor in podocytes was confirmed in cultured cells by immunoblotting and quantitative real-time PCR and in mouse kidneys by immunogold electron microscopy, and its agonism 1) increased cAMP, 2) activated PKA, 3) phosphorylated nephrin, and 4) attenuated albumin transcytosis. Finally, treatment of diabetic endothelial nitric oxide synthase knockout mice with selexipag augmented renal nephrin phosphorylation and attenuated albuminuria development independently of glucose change. Collectively, these observations describe a pharmacological strategy that posttranslationally modifies nephrin and the effects of this strategy in the pancreas and in the kidney.


Assuntos
Nefropatias Diabéticas/prevenção & controle , Células Secretoras de Insulina/efeitos dos fármacos , Proteínas de Membrana/metabolismo , Podócitos/efeitos dos fármacos , Receptores de Epoprostenol/agonistas , Acetamidas/uso terapêutico , Acetatos/farmacologia , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patologia , Nefropatias Diabéticas/metabolismo , Nefropatias Diabéticas/patologia , Nefropatias Diabéticas/fisiopatologia , Humanos , Hipoglicemiantes/farmacologia , Hipoglicemiantes/uso terapêutico , Insulina/agonistas , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/genética , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação , Fosforilação/efeitos dos fármacos , Podócitos/metabolismo , Podócitos/patologia , Podócitos/ultraestrutura , Pró-Fármacos/uso terapêutico , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Pirazinas/farmacologia , Pirazinas/uso terapêutico , Interferência de RNA , Receptores de Epoprostenol/genética , Receptores de Epoprostenol/metabolismo , Insuficiência Renal/complicações , Insuficiência Renal/metabolismo , Insuficiência Renal/patologia , Insuficiência Renal/prevenção & controle
17.
Prostaglandins Other Lipid Mediat ; 121(Pt A): 70-82, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25936507

RESUMO

The prostanoid Prostacyclin plays diverse physiologic roles within the vasculature and other systems, and is widely implicated in several cardiovascular, pulmonary and renal diseases. Despite this, knowledge of the factors regulating expression of the I prostanoid receptor (the IP) remained largely unknown. This review details recent advances in understanding the key transcriptional regulators determining expression of the PTGIR gene in the human vasculature and the identification of novel interacting partners of the IP that impact on its function therein. Included in this are the trans-acting factors that regulate expression of the PTGIR under basal- and regulated-conditions, particularly those determining its up-regulation in response to cellular differentiation, estrogen and low serum-cholesterol. Moreover, the functional implications of the interactions between the IP with PDZK1, a multi PDZ-domain containing protein essential for reverse-cholesterol transport and endothelialization, and the IP with IKEPP, the intestinal and kidney enriched PDZ protein, for the role of the prostacyclin-IP axis within the vasculature are reviewed.


Assuntos
Regulação da Expressão Gênica , Receptores de Epoprostenol/genética , Receptores de Epoprostenol/metabolismo , Transdução de Sinais , Transcrição Gênica , Sequência de Aminoácidos , Animais , Humanos , Dados de Sequência Molecular , Receptores de Epoprostenol/química
18.
J Allergy Clin Immunol ; 134(3): 698-705.e5, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25042746

RESUMO

BACKGROUND: The prevalence of allergic diseases has doubled in developed countries in the past several decades. Cyclooxygenase (COX)-inhibiting drugs augmented allergic diseases in mice by increasing allergic sensitization and memory immune responses. However, whether COX inhibition can promote allergic airway diseases by inhibiting immune tolerance is not known. OBJECTIVE: To determine the role of the COX pathway and prostaglandin I2 (PGI2) signaling through the PGI2 receptor (IP) in aeroallergen-induced immune tolerance. METHODS: Wild-type (WT) BALB/c mice and IP knockout mice were aerosolized with ovalbumin (OVA) to induce immune tolerance prior to immune sensitization with an intraperitoneal injection of OVA/alum. The COX inhibitor indomethacin or vehicle was administered in drinking water to inhibit enzyme activity during the sensitization phase. Two weeks after sensitization, the mice were challenged with OVA aerosols. Mouse bronchoalveolar lavage fluid was harvested for cell counts and TH2 cytokine measurements. RESULTS: WT mice treated with indomethacin had greater numbers of total cells, eosinophils, and lymphocytes, and increased IL-5 and IL-13 protein expression in BAL fluid compared to vehicle-treated mice. Similarly, IP knockout mice had augmented inflammation and TH2 cytokine responses compared to WT mice. In contrast, the PGI2 analog cicaprost attenuated the anti-tolerance effect of COX inhibition. CONCLUSION: COX inhibition abrogated immune tolerance by suppressing PGI2 IP signaling, suggesting that PGI2 signaling promotes immune tolerance and that clinical use of COX-inhibiting drugs may increase the risk of developing allergic diseases.


Assuntos
Inibidores Enzimáticos/administração & dosagem , Epoprostenol/metabolismo , Hipersensibilidade/imunologia , Indometacina/administração & dosagem , Receptores de Epoprostenol/metabolismo , Poluição do Ar/efeitos adversos , Alérgenos/efeitos adversos , Alérgenos/imunologia , Animais , Humanos , Tolerância Imunológica , Indometacina/farmacologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ovalbumina/imunologia , Prostaglandina-Endoperóxido Sintases/metabolismo , Receptores de Epoprostenol/genética , Transdução de Sinais
19.
Artigo em Inglês | MEDLINE | ID: mdl-24035274

RESUMO

Prostaglandin I2 (PGI2) is an important immunoregulatory lipid mediator. In this study, we analysed the effects of the PGI2 analogue (Iloprost) on the differentiation of Th17 cells and Tregs from human naïve CD4(+) T cells. PGI2 receptors (IP) are expressed on human naïve CD4(+) T cells. Via IP binding, the PGI2 analogue decreased the proportion of Tregs and Foxp3 mRNA expression but increased the percentage of Th17 cells, RORC mRNA and IL-17A production. The regulatory effects of Iloprost correlated with elevated intracellular cAMP levels. The effects were mimicked by a cAMP agonist (db-cAMP) but attenuated by a protein kinase A inhibitor (H-89). STAT3 and STAT5 signalling play direct and crucial roles in the development of Th17 and Tregs, respectively. The PGI2 analogue enhanced the activation of STAT3 in response to IL-6, whereas it decreased STAT5 activation in response to IL-2. Moreover, db-cAMP imitated the above effects of Iloprost, which were weakened by H-89. These results demonstrate that the PGI2-IP interaction promoted the phosphorylation of STAT3 and reduced the phosphorylation of STAT5, likely via the upregulation of cAMP-PKA signalling, thus facilitated Th17 differentiation and suppressed Treg differentiation. Together with previous results, these data suggest that prostanoids play an important role in the pathogenesis of autoimmune diseases, such as rheumatoid arthritis.


Assuntos
Epoprostenol/metabolismo , RNA Mensageiro/metabolismo , Receptores de Epoprostenol/metabolismo , Transdução de Sinais , Linfócitos T Reguladores/metabolismo , Células Th17/metabolismo , Bucladesina/farmacologia , Diferenciação Celular , AMP Cíclico/antagonistas & inibidores , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica , Humanos , Iloprosta/farmacologia , Interleucina-17/genética , Interleucina-17/metabolismo , Interleucina-2/genética , Interleucina-2/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Isoquinolinas/farmacologia , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/genética , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Fosforilação/efeitos dos fármacos , Inibidores da Agregação Plaquetária/farmacologia , RNA Mensageiro/genética , Receptores de Epoprostenol/genética , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Fator de Transcrição STAT5/genética , Fator de Transcrição STAT5/metabolismo , Sulfonamidas/farmacologia , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/efeitos dos fármacos , Células Th17/citologia , Células Th17/efeitos dos fármacos
20.
Mol Cell Biol ; 33(2): 184-93, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23109431

RESUMO

G protein-coupled receptors (GPCRs) interact with heterotrimeric G proteins and initiate a wide variety of signaling pathways. The molecular nature of GPCR-G protein interactions in the clinically important thromboxane A2 (TxA(2)) receptor (TP) and prostacyclin (PGI(2)) receptor (IP) is poorly understood. The TP activates its cognate G protein (Gαq) in response to the binding of thromboxane, while the IP signals through Gαs in response to the binding of prostacyclin. Here, we utilized a combination of approaches consisting of chimeric receptors, molecular modeling, and site-directed mutagenesis to precisely study the specificity of G protein coupling. Multiple chimeric receptors were constructed by replacing the TP intracellular loops (ICLs) with the ICL regions of the IP. Our results demonstrate that both the sequences and lengths of ICL2 and ICL3 influenced G protein specificity. Importantly, we identified a precise ICL region on the prostanoid receptors TP and IP that can switch G protein specificities. The validities of the chimeric technique and the derived molecular model were confirmed by introducing clinically relevant naturally occurring mutations (R60L in the TP and R212C in the IP). Our findings provide new molecular insights into prostanoid receptor-G protein interactions, which are of general significance for understanding the structural basis of G protein activation by GPCRs in basic health and cardiovascular disease.


Assuntos
Proteínas de Ligação ao GTP/genética , Receptores de Epoprostenol/química , Receptores de Tromboxano A2 e Prostaglandina H2/química , Sequência de Aminoácidos , Sítios de Ligação , Cálcio/análise , Imunofluorescência , Células HEK293 , Humanos , Inositol 1,4,5-Trifosfato/análise , Ligantes , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Dinâmica não Linear , Ligação Proteica , Conformação Proteica , Receptores de Epoprostenol/genética , Receptores de Tromboxano A2 e Prostaglandina H2/genética , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA