Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 119
Filtrar
1.
Int J Mol Sci ; 24(16)2023 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-37629047

RESUMEN

Current drugs for treating heart failure (HF), for example, angiotensin II receptor blockers and ß-blockers, possess specific target molecules involved in the regulation of the cardiac circulatory system. However, most clinically approved drugs are effective in the treatment of HF with reduced ejection fraction (HFrEF). Novel drug classes, including angiotensin receptor blocker/neprilysin inhibitor (ARNI), sodium-glucose co-transporter-2 (SGLT2) inhibitor, hyperpolarization-activated cyclic nucleotide-gated (HCN) channel blocker, soluble guanylyl cyclase (sGC) stimulator/activator, and cardiac myosin activator, have recently been introduced for HF intervention based on their proposed novel mechanisms. SGLT2 inhibitors have been shown to be effective not only for HFrEF but also for HF with preserved ejection fraction (HFpEF). In the myocardium, excess cyclic adenosine monophosphate (cAMP) stimulation has detrimental effects on HFrEF, whereas cyclic guanosine monophosphate (cGMP) signaling inhibits cAMP-mediated responses. Thus, molecules participating in cGMP signaling are promising targets of novel drugs for HF. In this review, we summarize molecular pathways of cGMP signaling and clinical trials of emerging drug classes targeting cGMP signaling in the treatment of HF.


Asunto(s)
Insuficiencia Cardíaca , Inhibidores del Cotransportador de Sodio-Glucosa 2 , Humanos , Insuficiencia Cardíaca/tratamiento farmacológico , Volumen Sistólico , Corazón , Miocardio , Antagonistas de Receptores de Angiotensina , Bloqueadores de los Canales de Calcio , AMP Cíclico , GMP Cíclico , Vasodilatadores
2.
Int J Mol Sci ; 24(4)2023 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-36835211

RESUMEN

Nonalcoholic steatohepatitis (NASH) is a disease that progresses from nonalcoholic fatty liver (NAFL) and which is characterized by inflammation and fibrosis. The purinergic P2Y6 receptor (P2Y6R) is a pro-inflammatory Gq/G12 family protein-coupled receptor and reportedly contributes to intestinal inflammation and cardiovascular fibrosis, but its role in liver pathogenesis is unknown. Human genomics data analysis revealed that the liver P2Y6R mRNA expression level is increased during the progression from NAFL to NASH, which positively correlates with inductions of C-C motif chemokine 2 (CCL2) and collagen type I α1 chain (Col1a1) mRNAs. Therefore, we examined the impact of P2Y6R functional deficiency in mice crossed with a NASH model using a choline-deficient, L-amino acid-defined, high-fat diet (CDAHFD). Feeding CDAHFD for 6 weeks markedly increased P2Y6R expression level in mouse liver, which was positively correlated with CCL2 mRNA induction. Unexpectedly, the CDAHFD treatment for 6 weeks increased liver weights with severe steatosis in both wild-type (WT) and P2Y6R knockout (KO) mice, while the disease marker levels such as serum AST and liver CCL2 mRNA in CDAHFD-treated P2Y6R KO mice were rather aggravated compared with those of CDAHFD-treated WT mice. Thus, P2Y6R may not contribute to the progression of liver injury, despite increased expression in NASH liver.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico , Receptores Purinérgicos P2 , Animales , Humanos , Ratones , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Ratones Noqueados , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , ARN Mensajero/metabolismo , Receptores Purinérgicos P2/genética , Receptores Purinérgicos P2/metabolismo
3.
Yakugaku Zasshi ; 142(10): 1091-1101, 2022.
Artículo en Japonés | MEDLINE | ID: mdl-36184444

RESUMEN

It is well-established that G protein-coupled receptors (GPCRs) transduce signals into cells using G proteins as intermediary molecules. ß-Arrestins are molecules involved in regulating GPCRs; however, it has recently been reported that ß-arrestins can also mediate signaling through GPCRs. Signaling through G proteins or ß-arrestins can be activated selectively using specific agonists; of the latter, those that can selectively activate either G proteins or ß-arrestins are called biased agonists. The clinical use of biased agonists could potentially induce fewer side effects. However, partial agonists can also explain the mechanism of G protein-biased agonists; thus, appropriate assay systems must be considered. Endogenous agonists are known to bind to orthosteric and allosteric sites in the agonist binding site, and the allosteric site is associated with the activity of biased agonists. This current review presents a detailed discussion of biased agonists.


Asunto(s)
Arrestinas , Receptores Acoplados a Proteínas G , Arrestinas/metabolismo , Proteínas de Unión al GTP/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , beta-Arrestinas/metabolismo
4.
Cells ; 11(13)2022 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-35805125

RESUMEN

Retarded revascularization after progressive occlusion of large conductance arteries is a major cause of bad prognosis for peripheral artery disease (PAD). However, pharmacological treatment for PAD is still limited. We previously reported that suppression of transient receptor potential canonical (TRPC) 6 channel activity in vascular smooth muscle cells (VSMCs) facilitates VSMC differentiation without affecting proliferation and migration. In this study, we found that 1-benzilpiperadine derivative (1-BP), a selective inhibitor for TRPC3 and TRPC6 channel activities, induced VSMC differentiation. 1-BP-treated mice showed increased capillary arterialization and improvement of peripheral circulation and skeletal muscle mass after hind-limb ischemia (HLI) in mice. 1-BP had no additive effect on the facilitation of blood flow recovery after HLI in TRPC6-deficient mice, suggesting that suppression of TRPC6 underlies facilitation of the blood flow recovery by 1-BP. 1-BP also improved vascular nitric oxide bioavailability and blood flow recovery after HLI in hypercholesterolemic mice with endothelial dysfunction, suggesting the retrograde interaction from VSMCs to endothelium. These results suggest that 1-BP becomes a potential seed for PAD treatments that target vascular TRPC6 channels.


Asunto(s)
Isquemia , Miocitos del Músculo Liso , Canales Catiónicos TRPC/metabolismo , Canal Catiónico TRPC6/metabolismo , Animales , Arterias , Isquemia/tratamiento farmacológico , Ratones , Músculo Esquelético
5.
Biol Pharm Bull ; 45(6): 669-674, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35650094

RESUMEN

Agonists are defined as the ligands that activate intracellular signaling and evoke cellular responses. Synthetic and endogenous agonists should bind specific amino acids to activate G protein-coupled receptor (GPCR). Agonists that induce maximal responses are full agonists. Partial agonists cannot induce full responses unlike full agonists. In definition, antagonists inhibit agonist-stimulated responses by binding to orthosteric or allosteric sites. Antagonists modulate agonist-induced responses and are often related with inverse agonist activity. However, the relationship between antagonists and partial agonists is complex. An antagonist behaves as a partial agonist when the constitutive activity of the GPCR is high. In contrast, a partial agonist with very weak intrinsic activity may be classified as an antagonist. Thus, antagonisms of the compounds are influenced by constitutive activity of GPCRs, intrinsic activity and differences in the binding sites of GPCRs. Since "antagonism" has been revealed to have multiple aspects and more complex than previously thought, it may be difficult to classify each compound as simply "agonist" or "antagonist" as before. In this review, we discuss the recent findings and perspectives on the pharmacology of GPCR-binding antagonists, inverse agonists, and signaling.


Asunto(s)
Receptores Acoplados a Proteínas G , Transducción de Señal , Sitios de Unión , Ligandos , Receptores Acoplados a Proteínas G/metabolismo
6.
Sci Signal ; 15(716): eabj0644, 2022 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-35015570

RESUMEN

After ligand stimulation, many G protein­coupled receptors (GPCRs) undergo ß-arrestin­dependent desensitization, during which they are internalized and either degraded or recycled to the plasma membrane. Some GPCRs are not subject to this type of desensitization because they lack the residues required to interact with ß-arrestins. We identified a mechanism of redox-dependent alternative internalization (REDAI) that promotes the internalization and degradation of the purinergic P2Y6 receptor (P2Y6R). Synthetic and natural compounds containing electrophilic isothiocyanate groups covalently modified P2Y6R at Cys220, which promoted the ubiquitylation of Lys137 and receptor internalization and degradation in various mouse and human cultured cell lines. Endogenous electrophiles also promoted ligand-dependent P2Y6R internalization and degradation. P2Y6R is highly abundant in inflammatory cells and promotes the pathogenesis of colitis. Deficiency in P2Y6R protected mice against experimentally induced colitis, and mice expressing a form of P2Y6R in which Cys220 was mutated to nonmodifiable serine were more sensitive to the induction of colitis. Several other GPCRs, including A2BAR, contain cysteine and lysine residues at the appropriate positions to mediate REDAI, and isothiocyanate stimulated the internalization of A2BAR and of a form of P2Y2R with insertions of the appropriate residues. Thus, endogenous and exogenous electrophiles may limit colitis progression through cysteine modification of P2Y6R and may also mediate internalization of other GPCRs.


Asunto(s)
Colitis , Receptores Purinérgicos P2 , Animales , Colitis/genética , Humanos , Ratones , Oxidación-Reducción , Receptores Purinérgicos P2/metabolismo , beta-Arrestinas/metabolismo
7.
Cells ; 10(7)2021 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-34359886

RESUMEN

Cardiac fibrosis is the excess deposition of extracellular matrix (ECM), such as collagen. Myofibroblasts are major players in the production of collagen, and are differentiated primarily from resident fibroblasts. Collagen can compensate for the dead cells produced by injury. The appropriate production of collagen is beneficial for preserving the structural integrity of the heart, and protects the heart from cardiac rupture. However, excessive deposition of collagen causes cardiac dysfunction. Recent studies have demonstrated that myofibroblasts can change their phenotypes. In addition, myofibroblasts are found to have functions other than ECM production. Myofibroblasts have macrophage-like functions, in which they engulf dead cells and secrete anti-inflammatory cytokines. Research into fibroblasts has been delayed due to the lack of selective markers for the identification of fibroblasts. In recent years, it has become possible to genetically label fibroblasts and perform sequencing at single-cell levels. Based on new technologies, the origins of fibroblasts and myofibroblasts, time-dependent changes in fibroblast states after injury, and fibroblast heterogeneity have been demonstrated. In this paper, recent advances in fibroblast and myofibroblast research are reviewed.


Asunto(s)
Fibrosis Endomiocárdica/patología , Matriz Extracelular/patología , Fibroblastos/patología , Macrófagos/patología , Miofibroblastos/patología , Animales , Cardiotónicos/uso terapéutico , Diferenciación Celular , Linaje de la Célula , Colágeno/genética , Colágeno/metabolismo , Citocinas/genética , Citocinas/metabolismo , Receptor con Dominio Discoidina 2/genética , Receptor con Dominio Discoidina 2/metabolismo , Fibrosis Endomiocárdica/tratamiento farmacológico , Fibrosis Endomiocárdica/genética , Fibrosis Endomiocárdica/metabolismo , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Fibroblastos/clasificación , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Regulación de la Expresión Génica , Humanos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Miofibroblastos/efectos de los fármacos , Miofibroblastos/metabolismo , Transducción de Señal
8.
Nat Commun ; 12(1): 2783, 2021 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-33986294

RESUMEN

Dysfunction of the circadian clock has been implicated in the pathogenesis of cardiovascular disease. The CLOCK protein is a core molecular component of the circadian oscillator, so that mice with a mutated Clock gene (Clk/Clk) exhibit abnormal rhythms in numerous physiological processes. However, here we report that chronic kidney disease (CKD)-induced cardiac inflammation and fibrosis are attenuated in Clk/Clk mice even though they have high blood pressure and increased serum angiotensin II levels. A search for the underlying cause of the attenuation of heart disorder in Clk/Clk mice with 5/6 nephrectomy (5/6Nx) led to identification of the monocytic expression of G protein-coupled receptor 68 (GPR68) as a risk factor of CKD-induced inflammation and fibrosis of heart. 5/6Nx induces the expression of GPR68 in circulating monocytes via altered CLOCK activation by increasing serum levels of retinol and its binding protein (RBP4). The high-GPR68-expressing monocytes have increased potential for producing inflammatory cytokines, and their cardiac infiltration under CKD conditions exacerbates inflammation and fibrosis of heart. Serum retinol and RBP4 levels in CKD patients are also sufficient to induce the expression of GPR68 in human monocytes. Our present study reveals an uncovered role of monocytic clock genes in CKD-induced heart failure.


Asunto(s)
Proteínas CLOCK/genética , Relojes Circadianos/genética , Ritmo Circadiano/fisiología , Cardiopatías/patología , Monocitos/metabolismo , Insuficiencia Renal Crónica/patología , Factores de Transcripción ARNTL/metabolismo , Animales , Proteínas CLOCK/metabolismo , Células Cultivadas , Ritmo Circadiano/genética , Citocinas/biosíntesis , Fibrosis/patología , Hipertensión/genética , Hipertensión/patología , Inflamación/genética , Inflamación/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratones Transgénicos , Receptores Acoplados a Proteínas G/metabolismo
10.
Biochem Biophys Res Commun ; 529(2): 224-230, 2020 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-32703415

RESUMEN

Fibrosis is attributed to excess deposition of extracellular matrix (ECM) proteins including collagen and is associated with various organ dysfunction. This excessive ECM is produced by myofibroblasts, which are differentiated from various cells by a variety of stimuli, represented by TGF-ß. However, molecular mechanisms for the regulation of ECM production in myofibroblasts remain obscure. In this study, we demonstrate that the expression of drebrin, which binds to and increases the stability of actin filament in neurons, is increased in mouse hearts and lungs upon fibrosis. Drebrin is mainly expressed in myofibroblasts in the fibrotic hearts and lungs and promotes the expression of fibrosis-related genes, such as Acta2 and Col1a1. Taken together, our study identifies drebrin as a molecule that promotes the production of fibrosis-related genes in myofibroblasts.


Asunto(s)
Pulmón/patología , Miocardio/patología , Miofibroblastos/patología , Neuropéptidos/genética , Animales , Diferenciación Celular , Células Cultivadas , Fibrosis , Pulmón/metabolismo , Ratones , Ratones Endogámicos C57BL , Miocardio/metabolismo , Miofibroblastos/metabolismo , Células 3T3 NIH , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/patología , Regulación hacia Arriba
11.
Exp Mol Med ; 52(6): 896-910, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32576930

RESUMEN

As the key governors of diverse physiological processes, G protein-coupled receptors (GPCRs) have drawn attention as primary targets for several diseases, including diabetes and cardiovascular disease. Heterotrimeric G proteins converge signals from ~800 members of the GPCR family. Among the members of the G protein α family, the Gα12 family members comprising Gα12 and Gα13 have been referred to as gep oncogenes. Gα12/13 levels are altered in metabolic organs, including the liver and muscles, in metabolic diseases. The roles of Gα12/13 in metabolic diseases have been investigated. In this review, we highlight findings demonstrating Gα12/13 amplifying or dampening regulators of phenotype changes. We discuss the molecular basis of G protein biology in the context of posttranslational modifications to heterotrimeric G proteins and the cell signaling axis. We also highlight findings providing insights into the organ-specific, metabolic and pathological roles of G proteins in changes associated with specific cells, energy homeostasis, glucose metabolism, liver fibrosis and the immune and cardiovascular systems. This review summarizes the currently available knowledge on the importance of Gα12/13 in the physiology and pathogenesis of metabolic diseases, which is presented according to the basic understanding of their metabolic actions and underlying cellular and molecular bases.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Enfermedades Metabólicas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Subunidades alfa de la Proteína de Unión al GTP G12-G13/genética , Humanos , Enfermedades Metabólicas/genética , Receptores Acoplados a Proteínas G/genética
12.
FASEB J ; 34(6): 8749-8763, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32385915

RESUMEN

Leukotriene B4 receptor 1 (BLT1), a high-affinity G-protein-coupled receptor for leukotriene B4 (LTB4 ), is expressed on various inflammatory cells and plays critical roles in several inflammatory diseases. In myocardial infarction (MI), various inflammatory cells are known to be recruited to the infarcted area, but the function of BLT1 in MI is poorly understood. Here, we investigated the role of BLT1 in MI and the therapeutic effect of a BLT1 antagonist, ONO-4057, on MI. Mice with infarcted hearts showed increased BLT1 expression and LTB4 levels. BLT1-knockout mice with infarcted hearts exhibited attenuated leukocyte infiltration, proinflammatory cytokine production, and cell death, which led to reduced mortality and improved cardiac function after MI. Bone-marrow transplantation studies showed that BLT1 expressed on bone marrow-derived cells was responsible for the exacerbation of inflammation in infarcted hearts. Furthermore, ONO-4057 administration attenuated the inflammatory responses in hearts surgically treated for MI, which resulted in reduced mortality and improved cardiac function after MI. Our study demonstrated that BLT1 contributes to excessive inflammation after MI and could represent a new therapeutic target for MI.


Asunto(s)
Inflamación/metabolismo , Infarto del Miocardio/metabolismo , Receptores de Leucotrieno B4/metabolismo , Animales , Modelos Animales de Enfermedad , Leucotrieno B4/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal/fisiología
13.
J Biochem ; 168(1): 1-6, 2020 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-32302392

RESUMEN

Myocardial infarction is one of the major causes of death worldwide. Many heart cells die during myocardial infarction through various processes such as necrosis, apoptosis, necroptosis, autophagy-related cell death, pyroptosis and ferroptosis. These dead cells in infarcted hearts expose the so-called 'eat-me' signals, such as phosphatidylserine, on their surfaces, enhancing their removal by professional and non-professional phagocytes. Clearance of dead cells by phagocytes in the diseased hearts plays a crucial role in the pathology of myocardial infarction by inhibiting the inflammatory responses caused by the leakage of contents from dead cells. This review focuses on the rapidly growing understanding of the molecular mechanisms of dead cell phagocytosis, termed efferocytosis, during myocardial infarction, which contributes to the pathophysiology of myocardial infarction.


Asunto(s)
Apoptosis , Inflamación/fisiopatología , Macrófagos/fisiología , Infarto del Miocardio/fisiopatología , Fagocitos/fisiología , Fagocitosis , Humanos , Transducción de Señal
14.
Front Cardiovasc Med ; 7: 34, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32211422

RESUMEN

Transforming growth factor-ß (TGF-ß) is a common mediator of cancer progression and fibrosis. Fibrosis can be a significant pathology in multiple organs, including the heart. In this review, we explain how inhibitors of TGF-ß signaling can work as antifibrotic therapy. After cardiac injury, profibrotic mediators such as TGF-ß, angiotensin II, and endothelin-1 simultaneously activate cardiac fibroblasts, resulting in fibroblast proliferation and migration, deposition of extracellular matrix proteins, and myofibroblast differentiation, which ultimately lead to the development of cardiac fibrosis. The consequences of fibrosis include a wide range of cardiac disorders, including contractile dysfunction, distortion of the cardiac structure, cardiac remodeling, and heart failure. Among various molecular contributors, TGF-ß and its signaling pathways which play a major role in carcinogenesis are considered master fibrotic mediators. In fact, recently the inhibition of TGF-ß signaling pathways using small molecule inhibitors, antibodies, and gene deletion has shown that the progression of several cancer types was suppressed. Therefore, inhibitors of TGF-ß signaling are promising targets for the treatment of tissue fibrosis and cancers. In this review, we discuss the molecular mechanisms of TGF-ß in the pathogenesis of cardiac fibrosis and cancer. We will review recent in vitro and in vivo evidence regarding antifibrotic and anticancer actions of TGF-ß inhibitors. In addition, we also present available clinical data on therapy based on inhibiting TGF-ß signaling for the treatment of cancers and cardiac fibrosis.

16.
Front Pharmacol ; 9: 1336, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30538631

RESUMEN

Heart failure (HF) is a heart disease that is classified into two main types: HF with reduced ejection fraction (HFrEF) and HF with preserved ejection fraction (HFpEF). Both types of HF lead to significant risk of mortality and morbidity. Pharmacological treatment with ß-adrenergic receptor (ßAR) antagonists (also called ß-blockers) has been shown to reduce the overall hospitalization and mortality rates and improve the clinical outcomes in HF patients with HFrEF but not HFpEF. Although, the survival rate of patients suffering from HF continues to drop, the management of HF still faces several limitations and discrepancies highlighting the need to develop new treatment strategies. Overstimulation of the sympathetic nervous system is an adaptive neurohormonal response to acute myocardial injury and heart damage, whereas prolonged exposure to catecholamines causes defects in ßAR regulation, including a reduction in the amount of ßARs and an increase in ßAR desensitization due to the upregulation of G protein-coupled receptor kinases (GRKs) in the heart, contributing in turn to the progression of HF. Several studies show that myocardial GRK2 activity and expression are raised in the failing heart. Furthermore, ß-arrestins play a pivotal role in ßAR desensitization and, interestingly, can mediate their own signal transduction without any G protein-dependent pathway involved. In this review, we provide new insight into the role of GRKs and ß-arrestins on how they affect ßAR signaling regarding the molecular and cellular pathophysiology of HF. Additionally, we discuss the therapeutic potential of targeting GRKs and ß-arrestins for the treatment of HF.

17.
J Clin Invest ; 128(12): 5587-5602, 2018 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-30300140

RESUMEN

Nonalcoholic fatty liver disease (NAFLD) arises from mitochondrial dysfunction under sustained imbalance between energy intake and expenditure, but the underlying mechanisms controlling mitochondrial respiration have not been entirely understood. Heterotrimeric G proteins converge with activated GPCRs to modulate cell-signaling pathways to maintain metabolic homeostasis. Here, we investigated the regulatory role of G protein α12 (Gα12) on hepatic lipid metabolism and whole-body energy expenditure in mice. Fasting increased Gα12 levels in mouse liver. Gα12 ablation markedly augmented fasting-induced hepatic fat accumulation. cDNA microarray analysis from Gna12-KO liver revealed that the Gα12-signaling pathway regulated sirtuin 1 (SIRT1) and PPARα, which are responsible for mitochondrial respiration. Defective induction of SIRT1 upon fasting was observed in the liver of Gna12-KO mice, which was reversed by lentivirus-mediated Gα12 overexpression in hepatocytes. Mechanistically, Gα12 stabilized SIRT1 protein through transcriptional induction of ubiquitin-specific peptidase 22 (USP22) via HIF-1α increase. Gα12 levels were markedly diminished in liver biopsies from NAFLD patients. Consistently, Gna12-KO mice fed a high-fat diet displayed greater susceptibility to diet-induced liver steatosis and obesity due to decrease in energy expenditure. Our results demonstrate that Gα12 regulates SIRT1-dependent mitochondrial respiration through HIF-1α-dependent USP22 induction, identifying Gα12 as an upstream molecule that contributes to the regulation of mitochondrial energy expenditure.


Asunto(s)
Endopeptidasas/metabolismo , Hígado Graso/metabolismo , Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Mitocondrias Hepáticas/metabolismo , Obesidad/metabolismo , Consumo de Oxígeno , Transducción de Señal , Sirtuina 1/metabolismo , Animales , Grasas de la Dieta/efectos adversos , Grasas de la Dieta/farmacología , Endopeptidasas/genética , Metabolismo Energético/efectos de los fármacos , Metabolismo Energético/genética , Hígado Graso/inducido químicamente , Hígado Graso/genética , Hígado Graso/patología , Subunidades alfa de la Proteína de Unión al GTP G12-G13/genética , Hepatocitos/metabolismo , Hepatocitos/patología , Humanos , Ratones , Ratones Noqueados , Mitocondrias Hepáticas/genética , Mitocondrias Hepáticas/patología , Obesidad/inducido químicamente , Obesidad/genética , Obesidad/patología , Sirtuina 1/genética , Ubiquitina Tiolesterasa
18.
Development ; 145(14)2018 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-29986870

RESUMEN

Erk5 belongs to the mitogen-activated protein kinase (MAPK) family. Following its phosphorylation by Mek5, Erk5 modulates several signaling pathways in a number of cell types. In this study, we demonstrated that Erk5 inactivation in mesenchymal cells causes abnormalities in skeletal development by inducing Sox9, an important transcription factor of skeletogenesis. We further demonstrate that Erk5 directly phosphorylates and activates Smurf2 (a ubiquitin E3 ligase) at Thr249, which promotes the proteasomal degradation of Smad proteins and phosphorylates Smad1 at Ser206 in the linker region known to trigger its proteasomal degradation by Smurf1. Smads transcriptionally activated the expression of Sox9 in mesenchymal cells. Accordingly, removal of one Sox9 allele in mesenchymal cells from Erk5-deficient mice rescued some abnormalities of skeletogenesis. These findings highlight the importance of the Mek5-Erk5-Smurf-Smad-Sox9 axis in mammalian skeletogenesis.


Asunto(s)
Proteína Quinasa 7 Activada por Mitógenos/metabolismo , Osteogénesis , Factor de Transcripción SOX9/metabolismo , Transducción de Señal , Proteínas Smad/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Diferenciación Celular , Condrogénesis , Humanos , Mesodermo/citología , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Proteolisis , Cráneo/anomalías , Ubiquitina/metabolismo , Ubiquitinación
19.
Nat Neurosci ; 21(6): 820-833, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29713080

RESUMEN

Chronic morphine exposure upregulates adenylate cyclase signaling and reduces analgesic efficacy, a condition known as opioid tolerance. Nonopioid neurotransmitters can enhance morphine tolerance, but the mechanism for this is poorly understood. We show that morphine tolerance was delayed in mice lacking vasopressin 1b receptors (V1bRs) or after administration of V1bR antagonist into the rostral ventromedial medulla, where transcripts for V1bRs and µ-opioid receptors are co-localized. Vasopressin increased morphine-binding affinity in cells expressing both V1bR and µ-opioid receptors. Complex formation among V1bR, ß-arrestin-2, and µ-opioid receptor resulted in vasopressin-mediated upregulation of ERK phosphorylation and adenylate cyclase sensitization. A leucine-rich segment in the V1bR C-terminus was necessary for the association with ß-arrestin-2. Deletion of this leucine-rich segment increased morphine analgesia and reduced vasopressin-mediated adenylate cyclase sensitization. These findings indicate that inhibition of µ-opioid-receptor-associated V1bR provides an approach for enhancing morphine analgesia without increasing analgesic tolerance.


Asunto(s)
Tolerancia a Medicamentos/genética , Morfina/farmacología , Narcóticos/farmacología , Receptores Opioides mu/metabolismo , Receptores de Vasopresinas/metabolismo , Arrestina beta 2/metabolismo , Adenilil Ciclasas/metabolismo , Animales , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/genética , Inyecciones , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Bulbo Raquídeo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Morfina/farmacocinética , Dependencia de Morfina/psicología , Narcóticos/farmacocinética , Dimensión del Dolor/efectos de los fármacos , Umbral del Dolor/efectos de los fármacos , Fosforilación , Receptores Opioides mu/genética , Receptores de Vasopresinas/genética , Arrestina beta 2/genética
20.
Sci Rep ; 7(1): 7812, 2017 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-28798316

RESUMEN

Myocardial infarction (MI) is an ischaemic heart condition caused by the occlusion of coronary arteries. Following MI, lactic acid from anaerobic glycolysis increases and infiltrating immune cells produce severe inflammation, which leads to acidosis in the ischaemic heart. However, the physiological implication of this pH reduction remains largely unknown. T-cell death-associated gene 8 (TDAG8) is a proton-sensing G protein-coupled receptor found on cardiac macrophages that recognise increases in extracellular protons. We demonstrated that TDAG8 negatively regulates the transcription of the chemokine Ccl20. The infarcted hearts of TDAG8 KO mice showed an increase in CCL20 expression and the number of infiltrating IL-17A-producing γδT cells that express CCR6, a receptor for CCL20. Accordingly, excessive IL-17A production, which is linked to the functional deterioration after MI, was observed in MI-operated TDAG8 KO mice. The survival rate and cardiac function significantly decreased in TDAG8 KO mice compared with those in wild-type mice after MI. Thus, our results suggest that TDAG8 is a key regulator of MI and a potential therapeutic target.


Asunto(s)
Quimiocina CCL20/genética , Infarto del Miocardio/genética , Animales , Quimiocina CCL20/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Interleucina-17/metabolismo , Linfocitos Intraepiteliales/inmunología , Ratones , Ratones Noqueados , Infarto del Miocardio/inmunología , Infarto del Miocardio/metabolismo , Receptores CCR6/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Análisis de Supervivencia , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...