Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 219
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 120(30): e2216329120, 2023 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-37478163

RESUMEN

To accomplish concerted physiological reactions, nature has diversified functions of a single hormone at at least two primary levels: 1) Different receptors recognize the same hormone, and 2) different cellular effectors couple to the same hormone-receptor pair [R.P. Xiao, Sci STKE 2001, re15 (2001); L. Hein, J. D. Altman, B.K. Kobilka, Nature 402, 181-184 (1999); Y. Daaka, L. M. Luttrell, R. J. Lefkowitz, Nature 390, 88-91 (1997)]. Not only these questions lie in the heart of hormone actions and receptor signaling but also dissecting mechanisms underlying these questions could offer therapeutic routes for refractory diseases, such as kidney injury (KI) or X-linked nephrogenic diabetes insipidus (NDI). Here, we identified that Gs-biased signaling, but not Gi activation downstream of EP4, showed beneficial effects for both KI and NDI treatments. Notably, by solving Cryo-electron microscope (cryo-EM) structures of EP3-Gi, EP4-Gs, and EP4-Gi in complex with endogenous prostaglandin E2 (PGE2)or two synthetic agonists and comparing with PGE2-EP2-Gs structures, we found that unique primary sequences of prostaglandin E2 receptor (EP) receptors and distinct conformational states of the EP4 ligand pocket govern the Gs/Gi transducer coupling selectivity through different structural propagation paths, especially via TM6 and TM7, to generate selective cytoplasmic structural features. In particular, the orientation of the PGE2 ω-chain and two distinct pockets encompassing agonist L902688 of EP4 were differentiated by their Gs/Gi coupling ability. Further, we identified common and distinct features of cytoplasmic side of EP receptors for Gs/Gi coupling and provide a structural basis for selective and biased agonist design of EP4 with therapeutic potential.


Asunto(s)
Dinoprostona , Transducción de Señal , Dinoprostona/metabolismo , Transducción de Señal/fisiología , Receptores de Prostaglandina/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Hormonas , Subtipo EP4 de Receptores de Prostaglandina E/metabolismo , Subtipo EP2 de Receptores de Prostaglandina E/metabolismo , Subtipo EP3 de Receptores de Prostaglandina E/metabolismo
2.
J Cell Mol Med ; 28(10): e18409, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38769917

RESUMEN

Farnesoid X receptor (FXR), a ligand-activated transcription factor, plays an important role in maintaining water homeostasis by up-regulating aquaporin 2 (AQP2) expression in renal medullary collecting ducts; however, its role in the survival of renal medullary interstitial cells (RMICs) under hypertonic conditions remains unclear. We cultured primary mouse RMICs and found that the FXR was expressed constitutively in RMICs, and that its expression was significantly up-regulated at both mRNA and protein levels by hypertonic stress. Using luciferase and ChIP assays, we found a potential binding site of nuclear factor kappa-B (NF-κB) located in the FXR gene promoter which can be bound and activated by NF-κB. Moreover, hypertonic stress-induced cell death in RMICs was significantly attenuated by FXR activation but worsened by FXR inhibition. Furthermore, FXR increased the expression and nuclear translocation of hypertonicity-induced tonicity-responsive enhance-binding protein (TonEBP), the expressions of its downstream target gene sodium myo-inositol transporter (SMIT), and heat shock protein 70 (HSP70). The present study demonstrates that the NF-κB/FXR/TonEBP pathway protects RMICs against hypertonic stress.


Asunto(s)
Médula Renal , FN-kappa B , Transducción de Señal , Animales , FN-kappa B/metabolismo , Ratones , Médula Renal/metabolismo , Médula Renal/citología , Presión Osmótica , Acuaporina 2/metabolismo , Acuaporina 2/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Masculino , Ratones Endogámicos C57BL , Proteínas HSP70 de Choque Térmico/metabolismo , Proteínas HSP70 de Choque Térmico/genética , Regiones Promotoras Genéticas , Células Cultivadas , Regulación de la Expresión Génica , Simportadores/metabolismo , Simportadores/genética , Receptores Citoplasmáticos y Nucleares
3.
Am J Physiol Renal Physiol ; 327(1): F146-F157, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38779753

RESUMEN

17ß-Hydroxysteroid dehydrogenase-13 (HSD17B13), a newly identified lipid droplet-associated protein, plays an important role in the development of nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH). Emerging evidence demonstrates that NASH is an independent risk factor for chronic kidney disease, which is frequently accompanied by renal lipid accumulation. In addition, the HSD17B13 rs72613567 variant is associated with lower levels of albuminuria in patients with biopsy-proven NAFLD. At present, the role of HSD17B13 in lipid accumulation in the kidney is unclear. This study utilized bioinformatic and immunostaining approaches to examine the expression and localization of HSD17B13 along the mouse urinary tract. We found that HSD17B13 is constitutively expressed in the kidney, ureter, and urinary bladder. Our findings reveal for the first time, to our knowledge, the precise localization of HSD17B13 in the mouse urinary system, providing a basis for further studying the pathogenesis of HSD17B13 in various renal and urological diseases.NEW & NOTEWORTHY HSD17B13, a lipid droplet-associated protein, is crucial in nonalcoholic fatty liver disease (NAFLD) development. NAFLD also independently raises chronic kidney disease (CKD) risk, often with renal lipid buildup. However, HSD17B13's role in CKD-related lipid accumulation is unclear. This study makes the first effort to examine HSD17B13 expression and localization along the urinary system, providing a basis for exploring its physiological and pathophysiological roles in the kidney and urinary tract.


Asunto(s)
17-Hidroxiesteroide Deshidrogenasas , Ratones Endogámicos C57BL , Animales , 17-Hidroxiesteroide Deshidrogenasas/genética , 17-Hidroxiesteroide Deshidrogenasas/metabolismo , Masculino , Ratones , Sistema Urinario/metabolismo , Sistema Urinario/patología , Riñón/metabolismo , Riñón/patología
4.
J Pineal Res ; 76(4): e12964, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38803014

RESUMEN

Circadian disruption such as shift work, jet lag, has gradually become a global health issue and is closely associated with various metabolic disorders. The influence and mechanism of circadian disruption on renal injury in chronic kidney disease (CKD) remains inadequately understood. Here, we evaluated the impact of environmental light disruption on the progression of chronic renal injury in CKD mice. By using two abnormal light exposure models to induce circadian disruption, we found that circadian disruption induced by weekly light/dark cycle reversal (LDDL) significantly exacerbated renal dysfunction, accelerated renal injury, and promoted renal fibrosis in mice with 5/6 nephrectomy and unilateral ureteral obstruction (UUO). Mechanistically, RNA-seq analysis revealed significant immune and metabolic disorder in the LDDL-conditioned CKD kidneys. Consistently, renal content of ATP was decreased and ROS production was increased in the kidney tissues of the LDDL-challenged CKD mice. Untargeted metabolomics revealed a significant buildup of lipids in the kidney affected by LDDL. Notably, the level of ß-NMN, a crucial intermediate in the NAD+ pathway, was found to be particularly reduced. Moreover, we demonstrated that both ß-NMN and melatonin administration could significantly rescue the light-disruption associated kidney dysfunction. In conclusion, environmental circadian disruption may exacerbate chronic kidney injury by facilitating inflammatory responses and disturbing metabolic homeostasis. ß-NMN and melatonin treatments may hold potential as promising approaches for preventing and treating light-disruption associated CKD.


Asunto(s)
Ritmo Circadiano , Insuficiencia Renal Crónica , Animales , Insuficiencia Renal Crónica/metabolismo , Insuficiencia Renal Crónica/patología , Insuficiencia Renal Crónica/etiología , Ratones , Masculino , Ritmo Circadiano/fisiología , Melatonina/metabolismo , Progresión de la Enfermedad , Ratones Endogámicos C57BL , Fotoperiodo , Riñón/metabolismo , Riñón/patología
5.
J Immunol ; 208(8): 1912-1923, 2022 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-35379745

RESUMEN

The mechanism regulating the life span of short-lived plasma cells (SLPCs) remains poorly understood. Here we demonstrated that the EP4-mediated activation of AKT by PGE2 was required for the proper control of inositol-requiring transmembrane kinase endoribonuclease-1α (IRE1α) hyperactivation and hence the endoplasmic reticulum (ER) homeostasis in IgM-producing SLPCs. Disruption of the PGE2-EP4-AKT signaling pathway resulted in IRE1α-induced activation of JNK, leading to accelerated death of SLPCs. Consequently, Ptger4-deficient mice (C57BL/6) exhibited a markedly impaired IgM response to T-independent Ags and increased susceptibility to Streptococcus pneumoniae infection. This study reveals a highly selective impact of the PGE2-EP4 signal on the humoral immunity and provides a link between ER stress response and the life span of SLPCs.


Asunto(s)
Supervivencia Celular , Dinoprostona , Estrés del Retículo Endoplásmico , Endorribonucleasas , Células Plasmáticas , Proteínas Serina-Treonina Quinasas , Animales , Supervivencia Celular/inmunología , Dinoprostona/inmunología , Estrés del Retículo Endoplásmico/inmunología , Endorribonucleasas/inmunología , Inmunoglobulina M/inmunología , Ratones , Ratones Endogámicos C57BL , Células Plasmáticas/inmunología , Prostaglandinas/inmunología , Prostaglandinas E/inmunología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/inmunología , Proteínas Proto-Oncogénicas c-akt/inmunología
6.
Sheng Li Xue Bao ; 76(2): 329-340, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38658381

RESUMEN

Chronic liver disease (CLD) is a major global health burden in terms of growing morbidity and mortality. Although many conditions can cause CLD, leading to cirrhosis and hepatocellular carcinoma (HCC), viral hepatitis, drug-induced liver injury (DILI), alcoholic liver disease (ALD) and non-alcoholic fatty liver disease (NAFLD) are the most common culprits. Prostaglandin E2 (PGE2), produced in the liver, is an important lipid mediator derived from the ω-6 polyunsaturated fatty acid, arachidonic acid, and plays a critical role in hepatic homeostasis. The physiological effects of PGE2 are mediated through four classes of E-type prostaglandin (EP) receptors, namely EP1, EP2, EP3 and EP4. In recent years, an increasing number of studies has been done to clarify the effects of PGE2 and EP receptors in regulating liver function and the pathogenesis of CLD to create a new potential clinical impact. In this review, we overview the biosynthesis and regulation of PGE2 and discuss the role of its synthesizing enzymes and receptors in the maintenance of normal liver function and the development and progress of CLD. We also discuss the potential of the PGE2-EP receptors system in treating CLD with various etiologies.


Asunto(s)
Dinoprostona , Hepatopatías , Receptores de Prostaglandina E , Humanos , Dinoprostona/metabolismo , Receptores de Prostaglandina E/metabolismo , Receptores de Prostaglandina E/fisiología , Hepatopatías/metabolismo , Enfermedad Crónica , Animales , Hígado/metabolismo , Hepatopatías Alcohólicas/metabolismo , Enfermedad del Hígado Graso no Alcohólico/metabolismo
7.
J Mol Cell Cardiol ; 181: 15-30, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37244057

RESUMEN

AIMS: Pulmonary arterial hypertension (PAH) is a progressive and lethal disease characterized by continuous proliferation of pulmonary arterial smooth muscle cell (PASMCs) and increased pulmonary vascular remodeling. Maresin-1 (MaR1) is a member of pro-resolving lipid mediators and exhibits protective effects on various inflammation-related diseases. Here we aimed to study the role of MaR1 in the development and progression of PAH and to explore the underlying mechanisms. METHODS AND RESULTS: We evaluated the effect of MaR1 treatment on PAH in both monocrotaline (MCT)-induced rat and hypoxia+SU5416 (HySu)-induced mouse models of pulmonary hypertension (PH). Plasma samples were collected from patients with PAH and rodent PH models to examine MaR1 production. Specific shRNA adenovirus or inhibitors were used to block the function of MaR1 receptors. The data showed that MaR1 significantly prevented the development and blunted the progression of PH in rodents. Blockade of the function of MaR1 receptor ALXR, but not LGR6 or RORα, with BOC-2, abolished the protective effect of MaR1 against PAH development and reduced its therapeutic potential. Mechanistically, we demonstrated that the MaR1/ALXR axis suppressed hypoxia-induced PASMCs proliferation and alleviated pulmonary vascular remodeling by inhibiting mitochondrial accumulation of heat shock protein 90α (HSP90α) and restoring mitophagy. CONCLUSION: MaR1 protects against PAH by improving mitochondrial homeostasis through ALXR/HSP90α axis and represents a promising target for PAH prevention and treatment.


Asunto(s)
Hipertensión Pulmonar , Hipertensión Arterial Pulmonar , Ratones , Ratas , Animales , Hipertensión Arterial Pulmonar/metabolismo , Proteínas de Choque Térmico/efectos adversos , Proteínas de Choque Térmico/metabolismo , Remodelación Vascular , Proliferación Celular , Células Cultivadas , Hipertensión Pulmonar Primaria Familiar , Hipertensión Pulmonar/metabolismo , Arteria Pulmonar , Hipoxia/metabolismo , Miocitos del Músculo Liso/metabolismo , Monocrotalina , Modelos Animales de Enfermedad
8.
Acta Pharmacol Sin ; 44(10): 2075-2090, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37344564

RESUMEN

Renal fibrosis is a common pathological feature of chronic kidney disease (CKD) with various etiologies, which seriously affects the structure and function of the kidney. Pregnane X receptor (PXR) is a member of the nuclear receptor superfamily and plays a critical role in regulating the genes related to xenobiotic and endobiotic metabolism in mammals. Previous studies show that PXR is expressed in the kidney and has protective effect against acute kidney injury (AKI). In this study, we investigated the role of PXR in CKD. Adenine diet-induced CKD (AD) model was established in wild-type and PXR humanized (hPXR) mice, respectively, which were treated with pregnenolone-16α-carbonitrile (PCN, 50 mg/kg, twice a week for 4 weeks) or rifampicin (RIF, 10 mg·kg-1·d-1, for 4 weeks). We showed that both PCN and RIF, which activated mouse and human PXR, respectively, improved renal function and attenuated renal fibrosis in the two types of AD mice. In addition, PCN treatment also alleviated renal fibrosis in unilateral ureter obstruction (UUO) mice. On the contrary, PXR gene deficiency exacerbated renal dysfunction and fibrosis in both adenine- and UUO-induced CKD mice. We found that PCN treatment suppressed the expression of the profibrotic Wnt7a and ß-catenin in AD mice and in cultured mouse renal tubular epithelial cells treated with TGFß1 in vitro. We demonstrated that PXR was colocalized and interacted with p53 in the nuclei of tubular epithelial cells. Overexpression of p53 increased the expression of Wnt7a, ß-catenin and its downstream gene fibronectin. We further revealed that p53 bound to the promoter of Wnt7a gene to increase its transcription and ß-catenin activation, leading to increased expression of the downstream profibrotic genes, which was inhibited by PXR. Taken together, PXR activation alleviates renal fibrosis in mice via interacting with p53 and inhibiting the Wnt7a/ß-catenin signaling pathway.


Asunto(s)
Receptor X de Pregnano , Insuficiencia Renal Crónica , Vía de Señalización Wnt , Animales , Humanos , Ratones , beta Catenina/metabolismo , Fibrosis , Mamíferos/metabolismo , Receptor X de Pregnano/metabolismo , Receptores de Esteroides/genética , Receptores de Esteroides/metabolismo , Insuficiencia Renal Crónica/inducido químicamente , Insuficiencia Renal Crónica/tratamiento farmacológico , Transducción de Señal , Proteína p53 Supresora de Tumor/metabolismo , Rifampin/farmacología
9.
FASEB J ; 35(5): e21530, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33813752

RESUMEN

Circadian clock is involved in regulating most renal physiological functions, including water and electrolyte balance and blood pressure homeostasis, however, the role of circadian clock in renal pathophysiology remains largely unknown. Here we aimed to investigate the role of Bmal1, a core clock component, in the development of renal fibrosis, the hallmark of pathological features in many renal diseases. The inducible Bmal1 knockout mice (iKO) whose gene deletion occurred in adulthood were used in the study. Analysis of the urinary water, sodium and potassium excretion showed that the iKO mice exhibit abolished diurnal variations. In the model of renal fibrosis induced by unilateral ureteral obstruction, the iKO mice displayed significantly decreased tubulointerstitial fibrosis reflected by attenuated collagen deposition and mitigated expression of fibrotic markers α-SMA and fibronectin. The hedgehog pathway transcriptional effectors Gli1 and Gli2, which have been reported to be involved in the pathogenesis of renal fibrosis, were significantly decreased in the iKO mice. Mechanistically, ChIP assay and luciferase reporter assay revealed that BMAL1 bound to the promoter of and activate the transcription of Gli2, but not Gli1, suggesting that the involvement of Bmal1 in renal fibrosis was possibly mediated via Gli2-dependent mechanisms. Furthermore, treatment with TGF-ß increased Bmal1 in cultured murine proximal tubular cells. Knockdown of Bmal1 abolished, while overexpression of Bmal1 increased, Gli2 and the expression of fibrosis-related genes. Collectively, these results revealed a prominent role of the core clock gene Bmal1 in tubulointerstitial fibrosis. Moreover, we identified Gli2 as a novel target of Bmal1, which may mediate the adverse effect of Bmal1 in obstructive nephropathy.


Asunto(s)
Factores de Transcripción ARNTL/fisiología , Fibrosis/prevención & control , Regulación de la Expresión Génica , Enfermedades Renales/prevención & control , Proteínas Circadianas Period/fisiología , Proteína Gli2 con Dedos de Zinc/antagonistas & inhibidores , Animales , Animales Recién Nacidos , Fibrosis/etiología , Fibrosis/metabolismo , Fibrosis/patología , Enfermedades Renales/etiología , Enfermedades Renales/metabolismo , Enfermedades Renales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Gli2 con Dedos de Zinc/genética , Proteína Gli2 con Dedos de Zinc/metabolismo
10.
Proc Natl Acad Sci U S A ; 116(17): 8457-8462, 2019 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-30948641

RESUMEN

Prostaglandin E2 (PGE2) plays an important role in vascular homeostasis. Its receptor, E-prostanoid receptor 4 (EP4) is essential for physiological remodeling of the ductus arteriosus (DA). However, the role of EP4 in pathological vascular remodeling remains largely unknown. We found that chronic angiotensin II (AngII) infusion of mice with vascular smooth muscle cell (VSMC)-specific EP4 gene knockout (VSMC-EP4-/-) frequently developed aortic dissection (AD) with severe elastic fiber degradation and VSMC dedifferentiation. AngII-infused VSMC-EP4-/- mice also displayed more profound vascular inflammation with increased monocyte chemoattractant protein-1 (MCP-1) expression, macrophage infiltration, matrix metalloproteinase-2 and -9 (MMP2/9) levels, NADPH oxidase 1 (NOX1) activity, and reactive oxygen species production. In addition, VSMC-EP4-/- mice exhibited higher blood pressure under basal and AngII-infused conditions. Ex vivo and in vitro studies further revealed that VSMC-specific EP4 gene deficiency significantly increased AngII-elicited vasoconstriction of the mesenteric artery, likely by stimulating intracellular calcium release in VSMCs. Furthermore, EP4 gene ablation and EP4 blockade in cultured VSMCs were associated with a significant increase in MCP-1 and NOX1 expression and a marked reduction in α-SM actin (α-SMA), SM22α, and SM differentiation marker genes myosin heavy chain (SMMHC) levels and serum response factor (SRF) transcriptional activity. To summarize, the present study demonstrates that VSMC EP4 is critical for vascular homeostasis, and its dysfunction exacerbates AngII-induced pathological vascular remodeling. EP4 may therefore represent a potential therapeutic target for the treatment of AD.


Asunto(s)
Angiotensina II/metabolismo , Disección Aórtica/metabolismo , Presión Sanguínea/fisiología , Inflamación/metabolismo , Subtipo EP4 de Receptores de Prostaglandina E , Animales , Aorta/química , Aorta/metabolismo , Aneurisma de la Aorta/metabolismo , Dinoprostona/metabolismo , Femenino , Hipertensión/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/metabolismo , Subtipo EP4 de Receptores de Prostaglandina E/genética , Subtipo EP4 de Receptores de Prostaglandina E/metabolismo , Remodelación Vascular/genética
11.
Am J Physiol Renal Physiol ; 321(5): F617-F628, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34569253

RESUMEN

The ligand-activated nuclear receptor, farnesoid X receptor (FXR), plays a pivotal role in regulating renal function. Activation of FXR by its specific agonists exerts renoprotective action in animals with acute kidney injury (AKI). In the present study, we aimed to identify naturally occurring agonists of FXR with potential as therapeutic agents in renal ischemia-reperfusion injury. In vitro and in vivo FXR activation was determined by a dual-luciferase assay, docking analysis, site-directed mutagenesis, and whole kidney transcriptome analysis. Wild-type (WT) and FXR knockout (FXR-/-) mice were used to determine the effect of potential FXR agonist on renal ischemia-reperfusion injury (IRI). We found that alisol B 23-acetate (ABA), a major active triterpenoid extracted from Alismatis rhizoma, a well-known traditional Chinese medicine, can activate renal FXR and induce FXR downstream gene expression in mouse kidney. ABA treatment significantly attenuated renal ischemia-reperfusion-induced AKI in WT mice but not in FXR-/- mice. Our results demonstrate that ABA can activate renal FXR to exert renoprotection against ischemia-reperfusion injury-induced AKI. Therefore, ABA may represent a potential therapeutic agent in the treatment of ischemic AKI.NEW & NOTEWORTHY In the present study, we found that alisol B 23-acetate (ABA), an identified natural farnesoid X receptor (FXR) agonist from the well-known traditional Chinese medicine Alismatis rhizoma, protects against ischemic acute kidney injury (AKI) in an FXR-dependent manner, as reflected by improved renal function, reduced renal tubular apoptosis, ameliorated oxidative stress, and suppressed inflammatory factor expression. Therefore, ABA may have great potential as a novel therapeutic agent in the treatment of AKI in the future.


Asunto(s)
Lesión Renal Aguda/prevención & control , Colestenonas/farmacología , Medicamentos Herbarios Chinos/farmacología , Riñón/efectos de los fármacos , Receptores Citoplasmáticos y Nucleares/agonistas , Daño por Reperfusión/prevención & control , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Animales , Apoptosis/efectos de los fármacos , Modelos Animales de Enfermedad , Células HEK293 , Células Hep G2 , Humanos , Mediadores de Inflamación/metabolismo , Riñón/metabolismo , Riñón/patología , Ligandos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Estrés Oxidativo/efectos de los fármacos , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Transducción de Señal
12.
Exp Cell Res ; 390(1): 111949, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32145254

RESUMEN

Farnesoid X receptor (FXR) is a metabolic nuclear receptor, which protects liver from many endogenous and exogenous injuries. Metallothioneins (MTs) belong to a low-molecular-weight protein family involved in metal homeostasis and the regulation of hepatic oxidative stress. In the present study, we aimed to investigate the effect of FXR on hepatic MT1 expression and the underlying mechanism. C57BL/6 mice or primary cultured mouse hepatocytes were treated with the synthetic FXR ligand GW4064 or natural ligand CDCA. RNA-Sequencing (RNA-seq) analysis was performed to identify gene expression profile in the livers of mice treated with GW4064. Real-time PCR and Western blot were applied to determine the expression of MT1 and other FXR target genes in the livers of mice and primary hepatocytes treated with GW4064 and CDCA. Cellular and subcellular locations of MT1 in the livers of mice treated with GW4064 were examined using immunohistochemistry assay. FXR small interfering RNAs (siRNA) was transfected to silence FXR. Luciferase reporter and chromatin immunoprecipitation (ChIP) assays were utilized to confirm the regulation of MT1 gene promoter activity by FXR. RNA-seq analysis revealed that GW4064 treatment significantly induced MT1 expression in mouse liver. Consistently, MT1 expression in the hepatocytes of mouse livers and cultured hepatocytes was upregulated by GW4064 as well as CDCA. In addition, adenovirus-mediated overexpression of FXR markedly increased, while siRNA-mediated FXR silencing significantly suppressed MT1 expression in cultured hepatocytes. Luciferase reporter and ChIP assays further confirmed that the MT1 gene was under the direct control of FXR. Collectively, our findings demonstrate that MT1 is a novel target gene of FXR and may contribute to antioxidative capacity of FXR in liver diseases.


Asunto(s)
Hígado/metabolismo , Metalotioneína/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Células Cultivadas , Hepatocitos/metabolismo , Humanos , Masculino , Metalotioneína/metabolismo , Ratones , Ratones Endogámicos C57BL , Receptores Citoplasmáticos y Nucleares/genética
13.
Proc Natl Acad Sci U S A ; 115(21): 5600-5605, 2018 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-29739889

RESUMEN

Hypertonicity in renal medulla is critical for the kidney to produce concentrated urine. Renal medullary cells have to survive high medullary osmolarity during antidiuresis. Previous study reported that farnesoid X receptor (FXR), a nuclear receptor transcription factor activated by endogenous bile acids, increases urine concentrating ability by up-regulating aquaporin 2 expression in medullary collecting duct cells (MCDs). However, whether FXR is also involved in the maintenance of cell survival of MCDs under dehydration condition and hypertonic stress remains largely unknown. In the present study, we demonstrate that 24-hours water restriction selectively up-regulated renal medullary expression of FXR with little MCD apoptosis in wild-type mice. In contrast, water deprivation caused a massive apoptosis of MCDs in both global FXR gene-deficient mice and collecting duct-specific FXR knockout mice. In vitro studies showed that hypertonicity significantly increased FXR and tonicity response enhancer binding protein (TonEBP) expression in mIMCD3 cell line and primary cultured MCDs. Activation and overexpression of FXR markedly increased cell viability and decreased cell apoptosis under hyperosmotic conditions. In addition, FXR can increase gene expression and nuclear translocation of TonEBP. We conclude that FXR protects MCDs from hypertonicity-induced cell injury very likely via increasing TonEBP expression and nuclear translocation. This study provides insights into the molecular mechanism by which FXR enhances urine concentration via maintaining cell viability of MCDs under hyperosmotic condition.


Asunto(s)
Capacidad de Concentración Renal/fisiología , Médula Renal/citología , Túbulos Renales Colectores/citología , Presión Osmótica/fisiología , Receptores Citoplasmáticos y Nucleares/fisiología , Estrés Fisiológico , Factores de Transcripción/metabolismo , Animales , Regulación de la Expresión Génica , Médula Renal/metabolismo , Túbulos Renales Colectores/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factores de Transcripción/genética
14.
Sheng Li Xue Bao ; 73(4): 657-664, 2021 Aug 25.
Artículo en Zh | MEDLINE | ID: mdl-34405221

RESUMEN

Arachidonic acid (AA) is an ω-6 polyunsaturated fatty acid, which mainly exists in the cell membrane in the form of phospholipid. Three major enzymatic pathways including the cyclooxygenase (COX), lipoxygenase (LOX) and cytochrome P450 monooxygenase (CYP450) pathways are involved in AA metabolism leading to the generation of a variety of lipid mediators such as prostaglandins, leukotrienes, hydroxyeicosatetraenoic acids (HETEs) and epoxyeicoastrienoic acids (EETs). These bioactive AA metabolites play an important role in the regulation of many physiological processes including the maintenance of liver glucose and lipid homeostasis. As the central metabolic organ, the liver is essential in metabolism of carbohydrates, lipids and proteins, and its dysfunction is associated with the pathogenesis of many metabolic diseases such as type 2 diabetes mellitus, dyslipidemia and nonalcoholic fatty liver disease (NAFLD). This article aims to provide an overview of the enzymatic pathways of AA and discuss the role of AA-derived lipid mediators in the regulation of hepatic glucose and lipid metabolism and their associations with the pathogenesis of major metabolic disorders.


Asunto(s)
Diabetes Mellitus Tipo 2 , Glucosa , Ácido Araquidónico/metabolismo , Glucosa/metabolismo , Homeostasis , Humanos , Metabolismo de los Lípidos , Hígado
15.
Sheng Li Xue Bao ; 73(4): 597-605, 2021 Aug 25.
Artículo en Zh | MEDLINE | ID: mdl-34405216

RESUMEN

Prostaglandin E2 (PGE2) plays an important role in cardiovascular system. PGE2 regulates blood pressure through its 4 G protein coupled receptors, i.e., EP1, EP2, EP3, and EP4. The aim of this study was to investigate the role of EP4 receptors in vascular smooth muscle cells (VSMC) in blood pressure regulation. VSMC-specific human EP4 transgenic (VSMC-hEP4 Tg) mice were generated and genotyped. The systolic blood pressure (SBP) of the VSMC-hEP4 Tg mice and the wild-type (WT) littermates was measured under normal, low-salt (LSD) and high-salt diet (HSD) conditions using a tail-cuff method. Both WT and VSMC-hEP4 Tg mice were administered with a chronic infusion of angiotensin II (Ang II) with an osmotic pump and SBP levels were monitored every week. The mean arterial blood pressure (MAP) of WT and VSMC-hEP4 Tg mice upon Ang II intravenous infusion was measured via carotid arterial catheterization. Ang II-induced vasoconstriction of the mesenteric arterial rings from WT and VSMC-hEP4 Tg mice was measured using the multi myograph system. The effect of PGE1-OH (a selective EP4 agonist) on Ang II-induced phosphorylation of myosin phosphatase target subunit 1 (MYPT1) was detected by Western blot. The effect of two additional EP4 specific agonists (CAY10580 and CAY10598, 0.5 mg/kg) on blood pressure of WT mice was measured by carotid arterial catheterization. The results showed that the VSMC-hEP4 Tg mice were successfully generated and their basal SBP levels were lower than those of WT mice. Although blood pressure levels were significantly altered in WT mice under LSD and HSD, little change was observed in the VSMC-hEP4 Tg mice. After a chronic infusion and an acute intravenous injection of Ang II, SBP levels of VSMC-hEP4 Tg mice were significantly lower than those of WT mice. In addition, both CAY10580 and CAY10598 significantly reduced MAP levels of WT mice. Ex vivo study showed that treatment of isolated mesenteric arteries with PGE1-OH inhibited Ang II-induced phosphorylation of MYPT1. Collectively, these results demonstrate that specific overexpression of human EP4 gene in VSMCs significantly reduces basal blood pressure levels and attenuates Ang II-induced hypertension, possibly via inhibiting Ang II/AT1 signaling pathway. Our findings suggest that EP4 may represent an attractive target for the treatment of hypertension.


Asunto(s)
Angiotensina II , Hipertensión , Animales , Humanos , Hipertensión/inducido químicamente , Ratones , Ratones Endogámicos C57BL , Músculo Liso Vascular , Miocitos del Músculo Liso
16.
Sheng Li Xue Bao ; 73(5): 795-804, 2021 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-34708236

RESUMEN

Farnesoid X receptor (FXR) has been identified as an inhibitor of platelet function and an inducer of fibrinogen protein complex. However, the regulatory mechanism of FXR in hemostatic system remains incompletely understood. In this study, we aimed to investigate the functions of FXR in regulating antithrombin III (AT III). C57BL/6 mice and FXR knockout (FXR KO) mice were treated with or without GW4064 (30 mg/kg per day). FXR activation significantly prolonged prothrombin time (PT) and activated partial thromboplastin time (APTT), lowered activity of activated factor X (FXa) and concentrations of thrombin-antithrombin complex (TAT) and activated factor II (FIIa), and increased level of AT III, whereas all of these effects were markedly reversed in FXR KO mice. In vivo, hepatic AT III mRNA and protein expression levels were up-regulated in wild-type mice after FXR activation, but down-regulated in FXR KO mice. In vitro study showed that FXR activation induced, while FXR knockdown inhibited, AT III expression in mouse primary hepatocytes. The luciferase assay and ChIP assay revealed that FXR can bind to the promoter region of AT III gene where FXR activation increased AT III transcription. These results suggest FXR activation inhibits coagulation process via inducing hepatic AT III expression in mice. The present study reveals a new role of FXR in hemostatic homeostasis and indicates that FXR might act as a potential therapeutic target for diseases related to hypercoagulation.


Asunto(s)
Antitrombina III , Hepatocitos , Receptores Citoplasmáticos y Nucleares , Animales , Coagulación Sanguínea , Hígado , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Citoplasmáticos y Nucleares/genética
17.
Pflugers Arch ; 472(11): 1631-1641, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32914211

RESUMEN

Crystallin zeta (CRYZ) is a phylogenetically restricted water-soluble protein and provides cytoprotection against oxidative stress via multiple mechanisms. Increasing evidence suggests that CRYZ is high abundantly expressed in the kidney where it acts as a transacting factor in increasing glutaminolysis and the Na+/K+/2Cl- cotransporter (BSC1/NKCC2) expression to help maintain acid-base balance and medullary hyperosmotic gradient. However, the mechanism by which CRYZ is regulated in the kidney remains largely uncharacterized. Here, we show that CRYZ is a direct target of farnesoid X receptor (FXR), a nuclear receptor important for renal physiology. We found that CRYZ was ubiquitously expressed in mouse kidney and constitutively expressed in the cytoplasm of medullary collecting duct cells (MCDs). In primary cultured mouse MCDs, CRYZ expression was significantly upregulated by the activation and overexpression of FXR. FXR-induced CRYZ expression was almost completely abolished in the MCD cells with siRNA-mediated FXR knockdown. Consistently, treatment with FXR agonists failed to induce CRYZ expression in the MCDs isolated from mice with global and collecting duct-specific FXR deficiency. We identified a putative FXR response element (FXRE) on the CRYZ gene promoter. The luciferase reporter and ChIP assays revealed that FXR can bind directly to the FXRE site, which was further markedly enhanced by FXR activation. Furthermore, we found CRYZ overexpression in MCDs significantly attenuated hypertonicity-induced cell death possibly via increasing Bcl-2 expression. Collectively, our findings demonstrate that CRYZ is constitutively expressed in renal medullary collecting duct cells, where it is transcriptionally controlled by FXR. Given a critical role of FXR in MCDs, CRYZ may be responsible for protective effect of FXR on the survival of MCDs under hypertonic condition during dehydration.


Asunto(s)
Túbulos Renales Colectores/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , zeta-Cristalinas/genética , Animales , Células Cultivadas , Citoplasma/metabolismo , Túbulos Renales Colectores/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Presión Osmótica , Receptores Citoplasmáticos y Nucleares/genética , Elementos de Respuesta , zeta-Cristalinas/metabolismo
18.
Am J Physiol Renal Physiol ; 318(1): F193-F203, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31682170

RESUMEN

The kidney is the central organ involved in maintaining water and sodium balance. In human kidneys, nine aquaporins (AQPs), including AQP1-8 and AQP11, have been found and are differentially expressed along the renal tubules and collecting ducts with distinct and critical roles in the regulation of body water homeostasis and urine concentration. Dysfunction and dysregulation of these AQPs result in various water balance disorders. This review summarizes current understanding of physiological and pathophysiological roles of AQPs in the kidney, with a focus on recent progress on AQP2 regulation by the nuclear receptor transcriptional factors. This review also provides an overview of AQPs as clinical biomarkers and therapeutic targets for renal diseases.


Asunto(s)
Acuaporinas/metabolismo , Enfermedades Renales/metabolismo , Riñón/metabolismo , Equilibrio Hidroelectrolítico/fisiología , Animales , Homeostasis/fisiología , Humanos
19.
J Pharmacol Exp Ther ; 375(1): 40-48, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32759273

RESUMEN

Deletion of microsomal prostaglandin E2 synthase-1 (mPGES-1) inhibits inflammation and protects against atherosclerotic vascular diseases but displayed variable influence on pathologic cardiac remodeling. Overactivation of ß-adrenergic receptors (ß-ARs) causes heart dysfunction and cardiac remodeling, whereas the role of mPGES-1 in ß-AR-induced cardiac remodeling is unknown. Here we addressed this question using mPGES-1 knockout mice, subjecting them to isoproterenol, a synthetic nonselective agonist for ß-ARs, at 5 or 15 mg/kg per day to induce different degrees of cardiac remodeling in vivo. Cardiac structure and function were assessed by echocardiography 24 hours after the last of seven consecutive daily injections of isoproterenol, and cardiac fibrosis was examined by Masson trichrome stain in morphology and by real-time polymerase chain reaction for the expression of fibrosis-related genes. The results showed that deletion of mPGES-1 had no significant effect on isoproterenol-induced cardiac dysfunction or hypertrophy. However, the cardiac fibrosis was dramatically attenuated in the mPGES-1 knockout mice after either low-dose or high-dose isoproterenol exposure. Furthermore, in vitro study revealed that overexpression of mPGES-1 in cultured cardiac fibroblasts increased isoproterenol-induced fibrosis, whereas knocking down mPGES-1 in cardiac myocytes decreased the fibrogenesis of fibroblasts. In conclusion, mPGES-1 deletion protects against isoproterenol-induced cardiac fibrosis in mice, and targeting mPGES-1 may represent a novel strategy to attenuate pathologic cardiac fibrosis, induced by ß-AR agonists. SIGNIFICANCE STATEMENT: Inhibitors of microsomal prostaglandin E2 synthase-1 (mPGES-1) are being developed as alternative analgesics that are less likely to elicit cardiovascular hazards than cyclooxygenase-2 selective nonsteroidal anti-inflammatory drugs. We have demonstrated that deletion of mPGES-1 protects inflammatory vascular diseases and promotes post-myocardial infarction survival. The role of mPGES-1 in ß-adrenergic receptor-induced cardiomyopathy is unknown. Here we illustrated that deletion of mPGES-1 alleviated isoproterenol-induced cardiac fibrosis without deteriorating cardiac dysfunction. These results illustrated that targeting mPGES-1 may represent an efficacious approach to the treatment of inflammatory cardiovascular diseases.


Asunto(s)
Cardiomiopatías/genética , Microsomas/metabolismo , Miocardio/patología , Prostaglandina-E Sintasas/genética , Receptores Adrenérgicos beta/metabolismo , Remodelación Ventricular/genética , Agonistas Adrenérgicos beta/farmacología , Animales , Cardiomiopatías/metabolismo , Cardiomiopatías/patología , Células Cultivadas , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibrosis , Isoproterenol/farmacología , Masculino , Ratones Noqueados , Microsomas/efectos de los fármacos , Miocardio/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Ratas , Ratas Sprague-Dawley , Remodelación Ventricular/efectos de los fármacos
20.
Proc Natl Acad Sci U S A ; 114(12): 3181-3185, 2017 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-28270609

RESUMEN

Nonalcoholic fatty liver disease (NAFLD) is characterized by simple hepatic steatosis (SS), nonalcoholic steatohepatitis (NASH), hepatic fibrosis, and cirrhosis. Dysregulated fatty acid metabolism in the liver plays a critical role in the pathogenesis of NAFLD. Cytochrome P450 omega-hydroxylase 4A14 (CYP4A14) is a homolog of human CYP4A hydroxylase that catalyzes omega-hydroxylation of medium-chain fatty acids and arachidonic acid in mice. The goal of this study was to determine the role of CYP4A14 in the development and the progression of NAFLD. Here, we showed that hepatic CYP4A expression was up-regulated in the livers of patients and three murine models of NAFLD. Adenovirus-mediated overexpression of CYP4A14 in the livers of C57BL/6 mice resulted in a fatty liver phenotype with a significant increase in hepatic fatty acid translocase (FAT/CD36) expression. In contrast, CYP4A14 gene-deficient mice fed a high-fat diet or a methionine and choline-deficient (MCD) diet exhibited attenuated liver lipid accumulation and reduced hepatic FAT/CD36 expression. In addition, hepatic inflammation and fibrosis was markedly ameliorated in MCD diet-fed CYP4A14-deficient mice. Collectively, CYP4A14 plays an important role in the pathogenesis of both SS and NASH and may represent a potential therapeutic target for the treatment of NAFLD.


Asunto(s)
Familia 4 del Citocromo P450/genética , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Cirrosis Hepática/genética , Cirrosis Hepática/patología , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/patología , Animales , Biomarcadores , Biopsia , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Ácidos Grasos/metabolismo , Expresión Génica , Hepatocitos/metabolismo , Metabolismo de los Lípidos , Cirrosis Hepática/etiología , Masculino , Ratones , Ratones Noqueados , Enfermedad del Hígado Graso no Alcohólico/etiología , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA