Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.047
Filtrar
1.
Am J Physiol Cell Physiol ; 326(2): C400-C413, 2024 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-38105755

RESUMO

Kidney fibrosis is a prominent pathological feature of hypertensive kidney diseases (HKD). Recent studies have highlighted the role of ubiquitinating/deubiquitinating protein modification in kidney pathophysiology. Ovarian tumor domain-containing protein 6 A (OTUD6A) is a deubiquitinating enzyme involved in tumor progression. However, its role in kidney pathophysiology remains elusive. We aimed to investigate the role and underlying mechanism of OTUD6A during kidney fibrosis in HKD. The results revealed higher OTUD6A expression in kidney tissues of nephropathy patients and mice with chronic angiotensin II (Ang II) administration than that from the control ones. OTUD6A was mainly located in tubular epithelial cells. Moreover, OTUD6A deficiency significantly protected mice against Ang II-induced kidney dysfunction and fibrosis. Also, knocking OTUD6A down suppressed Ang II-induced fibrosis in cultured tubular epithelial cells, whereas overexpression of OTUD6A enhanced fibrogenic responses. Mechanistically, OTUD6A bounded to signal transducer and activator of transcription 3 (STAT3) and removed K63-linked-ubiquitin chains to promote STAT3 phosphorylation at tyrosine 705 position and nuclear translocation, which then induced profibrotic gene transcription in epithelial cells. These studies identified STAT3 as a direct substrate of OTUD6A and highlighted the pivotal role of OTUD6A in Ang II-induced kidney injury, indicating OTUD6A as a potential therapeutic target for HKD.NEW & NOTEWORTHY Ovarian tumor domain-containing protein 6 A (OTUD6A) knockout mice are protected against angiotensin II-induced kidney dysfunction and fibrosis. OTUD6A promotes pathological kidney remodeling and dysfunction by deubiquitinating signal transducer and activator of transcription 3 (STAT3). OTUD6A binds to and removes K63-linked-ubiquitin chains of STAT3 to promote its phosphorylation and activation, and subsequently enhances kidney fibrosis.


Assuntos
Hipertensão Renal , Nefrite , Neoplasias Ovarianas , Humanos , Camundongos , Animais , Feminino , Angiotensina II/farmacologia , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Rim/metabolismo , Hipertensão Renal/metabolismo , Hipertensão Renal/patologia , Células Epiteliais/metabolismo , Fibrose , Neoplasias Ovarianas/metabolismo , Ubiquitinas/metabolismo , Camundongos Endogâmicos C57BL
2.
Circ Res ; 131(9): 731-747, 2022 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-36169218

RESUMO

BACKGROUND: SH2B3 (SH2B adaptor protein 3) is an adaptor protein that negatively regulates cytokine signaling and cell proliferation. A common missense single nucleotide polymorphism in SH2B3 (rs3184504) results in substitution of tryptophan (Trp) for arginine (Arg) at amino acid 262 and is a top association signal for hypertension in human genome-wide association studies. Whether this variant is causal for hypertension, and if so, the mechanism by which it impacts pathogenesis is unknown. METHODS: We used CRISPR-Cas9 technology to create mice homozygous for the major (Arg/Arg) and minor (Trp/Trp) alleles of this SH2B3 polymorphism. Mice underwent angiotensin II (Ang II) infusion to evaluate differences in blood pressure (BP) elevation and end-organ damage including albuminuria and renal fibrosis. Cytokine production and Stat4 phosphorylation was also assessed in Arg/Arg and Trp/Trp T cells. RESULTS: Trp/Trp mice exhibit 10 mmHg higher systolic BP during chronic Ang II infusion compared to Arg/Arg controls. Renal injury and perivascular fibrosis are exacerbated in Trp/Trp mice compared to Arg/Arg controls following Ang II infusion. Renal and ex vivo stimulated splenic CD8+ T cells from Ang II-infused Trp/Trp mice produce significantly more interferon gamma (IFNg) compared to Arg/Arg controls. Interleukin-12 (IL-12)-induced IFNg production is greater in Trp/Trp compared to Arg/Arg CD8+ T cells. In addition, IL-12 enhances Stat4 phosphorylation to a greater degree in Trp/Trp compared to Arg/Arg CD8+ T cells, suggesting that Trp-encoding SH2B3 exhibits less negative regulation of IL-12 signaling to promote IFNg production. Finally, we demonstrated that a multi-SNP model genetically predicting increased SH2B3 expression in lymphocytes is inversely associated with hypertension and hypertensive chronic kidney disease in humans.. CONCLUSIONS: Taken together, these results suggest that the Trp encoding allele of rs3184504 is causal for BP elevation and renal dysfunction, in part through loss of SH2B3-mediated repression of T cell IL-12 signaling leading to enhanced IFNg production.


Assuntos
Hipertensão Renal , Hipertensão , Proteínas Adaptadoras de Transdução de Sinal , Angiotensina II/metabolismo , Angiotensina II/toxicidade , Animais , Arginina/efeitos adversos , Arginina/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Fibrose , Estudo de Associação Genômica Ampla , Humanos , Hipertensão/metabolismo , Hipertensão Renal/metabolismo , Interferon gama/metabolismo , Interleucina-12/efeitos adversos , Interleucina-12/metabolismo , Rim/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Polimorfismo de Nucleotídeo Único , Triptofano
3.
Cell Mol Life Sci ; 80(7): 184, 2023 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-37340199

RESUMO

Macrophage activation has been shown to play an essential role in renal fibrosis and dysfunction in hypertensive chronic kidney disease. Dectin-1 is a pattern recognition receptor that is also involved in chronic noninfectious diseases through immune activation. However, the role of Dectin-1 in Ang II-induced renal failure is still unknown. In this study, we found that Dectin-1 expression on CD68 + macrophages was significantly elevated in the kidney after Ang II infusion. We assessed the effect of Dectin-1 on hypertensive renal injury using Dectin-1-deficient mice infused by Angiotensin II (Ang II) at 1000 ng/kg/min for 4 weeks. Ang II-induced renal dysfunction, interstitial fibrosis, and immune activation were significantly attenuated in Dectin-1-deficient mice. A Dectin-1 neutralizing antibody and Syk inhibitor (R406) were used to examine the effect and mechanism of Dectin-1/Syk signaling axle on cytokine secretion and renal fibrosis in culturing cells. Blocking Dectin-1 or inhibiting Syk significantly reduced the expression and secretion of chemokines in RAW264.7 macrophages. The in vitro data showed that the increase in TGF-ß1 in macrophages enhanced the binding of P65 and its target promotor via the Ang II-induced Dectin-1/Syk pathway. Secreted TGF-ß1 caused renal fibrosis in kidney cells through Smad3 activation. Thus, macrophage Dectin-1 may be involved in the activation of neutrophil migration and TGF-ß1 secretion, thereby promoting kidney fibrosis and dysfunction.


Assuntos
Angiotensina II , Hipertensão Renal , Camundongos , Animais , Angiotensina II/farmacologia , Angiotensina II/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Neutrófilos/metabolismo , Rim/metabolismo , Hipertensão Renal/metabolismo , Hipertensão Renal/patologia , Macrófagos/metabolismo , Fibrose
4.
Acta Pharmacol Sin ; 44(6): 1206-1216, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36635422

RESUMO

Hypertensive nephropathy (HTN) ranks as the second-leading cause of end-stage renal disease (ESRD). Accumulating evidence suggests that persistent hypertension injures tubular cells, leading to tubulointerstitial fibrosis (TIF), which is involved in the pathogenesis of HTN. G protein-coupled receptors (GPCRs) are implicated in many important pathological and physiological processes and act as important drug targets. In this study, we explored the intrarenal mechanisms underlying hypertension-associated TIF, and particularly, the potential role of GPR97, a member of the adhesion GPCR subfamily, in TIF. A deoxycorticosterone acetate (DOCA)/salt-induced hypertensive mouse model was used. We revealed a significantly upregulated expression of GPR97 in the kidneys, especially in renal tubules, of the hypertensive mice and 10 patients with biopsy-proven hypertensive kidney injury. GPR97-/- mice showed markedly elevated blood pressure, which was comparable to that of wild-type mice following DOCA/salt treatment, but dramatically ameliorated renal injury and TIF. In NRK-52E cells, we demonstrated that knockdown of GPR97 suppressed the activation of TGF-ß signaling by disturbing small GTPase RhoA-mediated cytoskeletal reorganization, thus inhibiting clathrin-mediated endocytosis of TGF-ß receptors and subsequent Smad activation. Collectively, this study demonstrates that GPR97 contributes to hypertension-associated TIF at least in part by facilitating TGF-ß signaling, suggesting that GPR97 is a pivotal intrarenal factor for TIF progression under hypertensive conditions, and therapeutic strategies targeting GPR97 may improve the outcomes of patients with HTN.


Assuntos
Acetato de Desoxicorticosterona , Hipertensão Renal , Hipertensão , Camundongos , Animais , Acetato de Desoxicorticosterona/efeitos adversos , Rim/patologia , Hipertensão Renal/tratamento farmacológico , Hipertensão Renal/metabolismo , Hipertensão Renal/patologia , Hipertensão/tratamento farmacológico , Fator de Crescimento Transformador beta/metabolismo , Fibrose
5.
Int J Mol Sci ; 24(11)2023 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-37298378

RESUMO

Arterial hypertension (AH) is a global challenge that greatly impacts cardiovascular morbidity and mortality worldwide. AH is a major risk factor for the development and progression of kidney disease. Several antihypertensive treatment options are already available to counteract the progression of kidney disease. Despite the implementation of the clinical use of renin-angiotensin aldosterone system (RAAS) inhibitors, gliflozins, endothelin receptor antagonists, and their combination, the kidney damage associated with AH is far from being resolved. Fortunately, recent studies on the molecular mechanisms of AH-induced kidney damage have identified novel potential therapeutic targets. Several pathophysiologic pathways have been shown to play a key role in AH-induced kidney damage, including inappropriate tissue activation of the RAAS and immunity system, leading to oxidative stress and inflammation. Moreover, the intracellular effects of increased uric acid and cell phenotype transition showed their link with changes in kidney structure in the early phase of AH. Emerging therapies targeting novel disease mechanisms could provide powerful approaches for hypertensive nephropathy management in the future. In this review, we would like to focus on the interactions of pathways linking the molecular consequences of AH to kidney damage, suggesting how old and new therapies could aim to protect the kidney.


Assuntos
Hipertensão Renal , Hipertensão , Humanos , Rim/metabolismo , Sistema Renina-Angiotensina , Anti-Hipertensivos/farmacologia , Anti-Hipertensivos/uso terapêutico , Anti-Hipertensivos/metabolismo , Hipertensão Renal/metabolismo
6.
Mol Med ; 27(1): 147, 2021 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-34773993

RESUMO

BACKGROUND: Patients with salt-sensitive hypertension are often accompanied with severe renal damage and accelerate to end-stage renal disease, which currently lacks effective treatment. Fibroblast growth factor 21 (FGF21) has been shown to suppress nephropathy in both type 1 and type 2 diabetes mice. Here, we aimed to investigate the therapeutic effect of FGF21 in salt-sensitive hypertension-induced nephropathy. METHODS: Changes of FGF21 expression in deoxycorticosterone acetate (DOCA)-salt-induced hypertensive mice were detected. The influence of FGF21 knockout in mice on DOCA-salt-induced nephropathy were determined. Recombinant human FGF21 (rhFGF21) was intraperitoneally injected into DOCA-salt-induced nephropathy mice, and then the inflammatory factors, oxidative stress levels and kidney injury-related indicators were observed. In vitro, human renal tubular epithelial cells (HK-2) were challenged by palmitate acid (PA) with or without FGF21, and then changes in inflammation and oxidative stress indicators were tested. RESULTS: We observed significant elevation in circulating levels and renal expression of FGF21 in DOCA-salt-induced hypertensive mice. We found that deletion of FGF21 in mice aggravated DOCA-salt-induced nephropathy. Supplementation with rhFGF21 reversed DOCA-salt-induced kidney injury. Mechanically, rhFGF21 induced AMPK activation in DOCA-salt-treated mice and PA-stimulated HK-2 cells, which inhibited NF-κB-regulated inflammation and Nrf2-mediated oxidative stress and thus, is important for rhFGF21 protection against DOCA-salt-induced nephropathy. CONCLUSION: These findings indicated that rhFGF21 could be a promising pharmacological strategy for the treatment of salt-sensitive hypertension-induced nephropathy.


Assuntos
Fatores de Crescimento de Fibroblastos , Hipertensão Renal , Nefrite , Animais , Anti-Inflamatórios/uso terapêutico , Antioxidantes/uso terapêutico , Linhagem Celular , Acetato de Desoxicorticosterona , Fatores de Crescimento de Fibroblastos/sangue , Fatores de Crescimento de Fibroblastos/genética , Fatores de Crescimento de Fibroblastos/metabolismo , Fatores de Crescimento de Fibroblastos/uso terapêutico , Humanos , Hipertensão Renal/induzido quimicamente , Hipertensão Renal/tratamento farmacológico , Hipertensão Renal/metabolismo , Hipertensão Renal/patologia , Interleucina-6/metabolismo , Rim/metabolismo , Rim/patologia , Masculino , Camundongos Endogâmicos C57BL , Nefrite/induzido quimicamente , Nefrite/tratamento farmacológico , Nefrite/metabolismo , Nefrite/patologia , Estresse Oxidativo , Proteínas Recombinantes/uso terapêutico , Cloreto de Sódio na Dieta , Fator de Necrose Tumoral alfa/metabolismo
7.
J Pharmacol Exp Ther ; 377(1): 189-198, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33414130

RESUMO

Previous studies identified a region on chromosome 1 associated with NG-nitro-L-arginine methyl ester (L-NAME) hypertension-induced renal disease in fawn-hooded hypertensive (FHH) rats. This region contains a mutant γ-adducin (Add3) gene that impairs renal blood flow (RBF) autoregulation, but its contribution to renal injury is unknown. The present study evaluated the hypothesis that knockout (KO) of Add3 impairs the renal vasoconstrictor response to the blockade of nitric oxide synthase and enhances hypertension-induced renal injury after chronic administration of L-NAME plus a high-salt diet. The acute hemodynamic effect of L-NAME and its chronic effects on hypertension and renal injury were compared in FHH 1Brown Norway (FHH 1BN) congenic rats (WT) expressing wild-type Add3 gene versus FHH 1BN Add3 KO rats. RBF was well autoregulated in WT rats but impaired in Add3 KO rats. Acute administration of L-NAME (10 mg/kg) raised mean arterial pressure (MAP) similarly in both strains, but RBF and glomerular filtration rate (GFR) fell by 38% in WT versus 15% in Add3 KO rats. MAP increased similarly in both strains after chronic administration of L-NAME and a high-salt diet; however, proteinuria and renal injury were greater in Add3 KO rats than in WT rats. Surprisingly, RBF, GFR, and glomerular capillary pressure were 41%, 82%, and 13% higher in L-NAME-treated Add3 KO rats than in WT rats. Hypertensive Add3 KO rats exhibited greater loss of podocytes and glomerular nephrin expression and increased interstitial fibrosis than in WT rats. These findings indicate that loss of ADD3 promotes L-NAME-induced renal injury by altering renal hemodynamics and enhancing the transmission of pressure to glomeruli. SIGNIFICANCE STATEMENT: A mutation in the γ-adducin (Add3) gene in fawn-hooded hypertensive rats that impairs autoregulation of renal blood flow is in a region of rat chromosome 1 homologous to a locus on human chromosome 10 associated with diabetic nephropathy. The present results indicate that loss of ADD3 enhanced NG-nitro-L-arginine methyl ester-induced hypertensive renal injury by altering the transmission of pressure to the glomerulus.


Assuntos
Proteínas de Ligação a Calmodulina/metabolismo , Hipertensão Renal/metabolismo , Insuficiência Renal Crônica/metabolismo , Animais , Pressão Sanguínea , Proteínas de Ligação a Calmodulina/genética , Inibidores Enzimáticos/toxicidade , Deleção de Genes , Taxa de Filtração Glomerular , Homeostase , Hipertensão Renal/etiologia , Hipertensão Renal/fisiopatologia , Masculino , NG-Nitroarginina Metil Éster/toxicidade , Podócitos/efeitos dos fármacos , Podócitos/metabolismo , Ratos , Circulação Renal , Insuficiência Renal Crônica/etiologia , Insuficiência Renal Crônica/fisiopatologia , Vasoconstrição
8.
J Pharmacol Exp Ther ; 376(2): 240-249, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33277348

RESUMO

Lysophosphatidic acid (LPA) is a bioactive lipid mediator that has been implicated in the pathophysiology of kidney disease. However, few studies have attempted to measure changes in the levels of various LPA species in the kidney after the development of renal disease. The present study measured the renal LPA levels during the development of kidney disease in rat models of hypertension, diabetes, and obstructive nephropathy using liquid chromatography/mass spectrometry/mass spectrometry. LPA levels (sum of 16:0, 18:0, 18:1, 18:2, and 20:4 LPA) were higher in the renal cortex of hypertensive Dahl salt-sensitive (Dahl S) rats fed a high-salt diet than those in normotensive rats fed a low-salt diet (296.6 ± 22.9 vs. 196.3 ± 8.5 nmol/g protein). LPA levels were elevated in the outer medulla of the kidney of streptozotocin-induced type 1 diabetic Dahl S rats compared with control rats (624.6 ± 129.5 vs. 318.8 ± 17.1 nmol/g protein). LPA levels were also higher in the renal cortex of 18-month-old, type 2 diabetic nephropathy (T2DN) rats with more severe renal injury than in 6-month-old T2DN rats (184.9 ± 20.9 vs. 116.9 ± 6.0 nmol/g protein). LPA levels also paralleled the progression of renal fibrosis in the renal cortex of Sprague-Dawley rats after unilateral ureteral obstruction (UUO). Administration of an LPA receptor antagonist, Ki16425, reduced the degree of renal fibrosis in UUO rats. These results suggest that the production of renal LPA increases during the development of renal injury and contributes to renal fibrosis. SIGNIFICANCE STATEMENT: The present study reveals that the lysophosphatidic acid (LPA) levels increase in the kidney in rat models of hypertension, diabetes, and obstructive nephropathy, and administration of an LPA receptor antagonist attenuates renal fibrosis. Therapeutic approaches that target the formation or actions of renal LPA might be renoprotective and have therapeutic potential.


Assuntos
Nefropatias Diabéticas/metabolismo , Hipertensão Renal/metabolismo , Lisofosfolipídeos/metabolismo , Animais , Nefropatias Diabéticas/patologia , Fibrose , Hipertensão Renal/patologia , Isoxazóis/farmacologia , Rim/efeitos dos fármacos , Rim/metabolismo , Rim/patologia , Lisofosfolipídeos/antagonistas & inibidores , Masculino , Propionatos/farmacologia , Ratos , Ratos Endogâmicos Dahl , Ratos Sprague-Dawley
9.
Circ Res ; 124(10): 1448-1461, 2019 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-30894089

RESUMO

RATIONALE: Endothelial dysfunction is an important determinant risk factor for the development of hypertension and its complications. Thus, identification of potential therapeutic targets for preventing endothelial dysfunction has major clinical importance. Emerging evidence indicates that epigenetic modifications are closely associated with the regulation of endothelial function. Among them, HDAC (histone deacetylase)-mediated epigenetic processes in vascular homeostasis and cardiovascular disease have attracted much attention. SIRT6 (sirtuin 6) is one member of SIRTs (class III HDAC) that are highly conserved NAD+-dependent deacetylases. OBJECTIVE: This study was designed to elucidate the role of SIRT6 in the pathogenesis of hypertension, discover the new targets of SIRT6, and explore related mechanisms on the regulation of endothelial function. METHODS AND RESULTS: The levels of endothelial SIRT6 were significantly reduced in 2 independent hypertension models: desoxycorticosterone acetate/salt-induced and Ang II (angiotensin II)-induced hypertensive mice. Utilizing genetically engineered endothelial-specific SIRT6 knockout (Cre+/SIRT6fl/fl) mice, we found that endothelial-specific deletion of SIRT6 significantly enhanced blood pressure, exacerbated endothelial dysfunction and cardiorenal injury in experimental hypertension. Functionally, SIRT6 has pleiotropic protective actions in endothelial cells, which include promoting endothelium-dependent vasodilatation and vascular NO bioavailability, reducing cellular permeability, ameliorating endothelial senescence and apoptosis, and facilitating autophagy. Mechanistically, SIRT6 induced the expression of GATA5 (GATA-binding protein 5), a novel regulator of blood pressure, through inhibiting Nkx3.2 (NK3 homeobox 2) transcription by deacetylating histone H3K9 (histone H3 lysine 9), thereby regulating GATA5-mediated signaling pathways to prevent endothelial injury. Finally, we provide direct evidence for the therapeutic potential of SIRT6 in desoxycorticosterone acetate/salt-induced hypertensive mice by overexpression of SIRT6 in vivo. CONCLUSIONS: This study for the first time demonstrates that SIRT6 prevents hypertension and its complications by maintaining endothelial function. Pharmacological targeting of SIRT6 may be an innovative therapeutic strategy for treating patients with hypertension.


Assuntos
Endotélio Vascular/fisiologia , Hipertensão/prevenção & controle , Sirtuínas/fisiologia , Acetilação , Angiotensina II , Animais , Acetato de Desoxicorticosterona , Endotélio Vascular/lesões , Epigênese Genética , Fator de Transcrição GATA5/metabolismo , Histona Desacetilases , Histonas/metabolismo , Proteínas de Homeodomínio/metabolismo , Hipertensão/induzido quimicamente , Hipertensão Renal/metabolismo , Rim/lesões , Camundongos , Camundongos Knockout , Nefrite/metabolismo , Sirtuínas/sangue , Sirtuínas/deficiência , Sirtuínas/genética , Cloreto de Sódio , Fatores de Transcrição/metabolismo , Vasoconstritores , Vasodilatação
10.
Exp Cell Res ; 386(1): 111706, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31697927

RESUMO

Krüppel-like factor 15 (KLF15) is a well-known transcription factor associated with podocyte injury and fibrosis. Recently, hypertensive nephropathy was discovered to be closely related to podocyte injury and fibrosis. However, methods to stimulate hypertension in vitro are lacking. Here, we constructed an in vitro model mimicking hypertension using a rotational force device to identify the role of KLF15 in fibrosis due to mechanically induced hypertensive injury. First, we found that KLF15 expression was decreased in patients with hypertensive nephropathy. Then, an in vitro study of hypertension due to rotational force was conducted, and an increase in fibrosis markers and decrease in KLF15 levels were determined after application of 4 mmHg pressure in primary cultured human podocytes. KLF15 and tight junction protein levels increased with retinoic acid treatment. siRNA-mediated inhibition of KLF15 exacerbated pressure-induced fibrosis injury, and KLF15 expression after treatment with angiotensin II was similar to that observed after treatment with the blood pressure modeling device. Furthermore, the reduced KLF15 levels after mechanical pressure application were restored after the administration of an antihypertensive drug. KLF15 expression was also low in vivo. We confirmed the protective role of KLF15 in fibrosis using a mechanically induced in vitro model of hypertensive injury.


Assuntos
Hipertensão Renal/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Nefrite/metabolismo , Podócitos/metabolismo , Adulto , Angiotensina II/farmacologia , Animais , Células Cultivadas , Feminino , Fibrose , Humanos , Hipertensão Renal/genética , Hipertensão Renal/patologia , Fatores de Transcrição Kruppel-Like/genética , Masculino , Camundongos , Nefrite/genética , Nefrite/patologia , Podócitos/efeitos dos fármacos , Podócitos/patologia , Pressão , Cultura Primária de Células/instrumentação , Rotação , Proteínas de Junções Íntimas/genética , Proteínas de Junções Íntimas/metabolismo
11.
Can J Physiol Pharmacol ; 99(10): 1000-1006, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-33852804

RESUMO

The Hippo/YAP (yes-associated protein) pathway is an important signaling pathway to control organ development and tissue homeostasis. YAP is a downstream effector of the Hippo pathway and a critical mediator of mechanic stress. Hypertensive nephropathy is characterized with glomerular sclerosis stiffness and renal fibrosis. The present study investigated the role of YAP pathway in angiotensin (Ang) II hypertensive renal injury by using YAP activation inhibitor verteporfin. Ang II increased the protein expression of YAP in renal nucleus fraction, decreased phospho-YAP, and phospho-LATS1/2 (large tumor suppressors 1 and 2) expressions in renal cytoplasmic fraction, suggesting Ang II activation of renal YAP. Ang II significantly increased systolic blood pressure (SBP), proteinuria, glomerular sclerosis, and fibrosis; treatment with verteporfin attenuated Ang II-induced proteinuria and renal injury with a mild reduction in SBP. Moreover, Ang II increased the protein expressions of inflammatory factors including tumor necrosis factor α, interleukin 1ß, and monocyte chemoattractant protein-1, and profibrotic factors including transforming growth factor ß, phospho-Smad3 and fibronectin. Verteporfin reversed abovementioned Ang II-induced molecule expressions. Our results for the first time demonstrate that the activation of the YAP pathway promotes hypertensive renal inflammation and fibrosis, which may promote hypertensive renal injury. YAP may be a new target for prevention and treatment of hypertensive renal diseases.


Assuntos
Injúria Renal Aguda/tratamento farmacológico , Angiotensina II/toxicidade , Hipertensão Renal/tratamento farmacológico , Hipertensão/metabolismo , Nefrite/tratamento farmacológico , Verteporfina/farmacologia , Proteínas de Sinalização YAP/antagonistas & inibidores , Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/patologia , Animais , Pressão Sanguínea , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Fibrose , Hipertensão/induzido quimicamente , Hipertensão/patologia , Hipertensão Renal/etiologia , Hipertensão Renal/metabolismo , Hipertensão Renal/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Nefrite/etiologia , Nefrite/metabolismo , Nefrite/patologia , Fármacos Fotossensibilizantes/farmacologia , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo , Vasoconstritores/toxicidade
12.
Int J Mol Sci ; 22(8)2021 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-33921823

RESUMO

Hypertension induces renal fibrosis or tubular interstitial fibrosis, which eventually results in end-stage renal disease. Epithelial-to-mesenchymal transition (EMT) is one of the underlying mechanisms of renal fibrosis. Though previous studies showed that Ecklonia cava extracts (ECE) and dieckol (DK) had inhibitory action on angiotensin (Ang) I-converting enzyme, which converts Ang I to Ang II. It is known that Ang II is involved in renal fibrosis; however, it was not evaluated whether ECE or DK attenuated hypertensive nephropathy by decreasing EMT. In this study, the effect of ECE and DK on decreasing Ang II and its down signal pathway of angiotensin type 1 receptor (AT1R)/TGFß/SMAD, which is related with the EMT and restoring renal function in spontaneously hypertensive rats (SHRs), was investigated. Either ECE or DK significantly decreased the serum level of Ang II in the SHRs. Moreover, the renal expression of AT1R/TGFß/SMAD was decreased by the administration of either ECE or DK. The mesenchymal cell markers in the kidney of SHRs was significantly decreased by ECE or DK. The fibrotic tissue of the kidney of SHRs was also significantly decreased by ECE or DK. The ratio of urine albumin/creatinine of SHRs was significantly decreased by ECE or DK. Overall, the results of this study indicate that ECE and DK decreased the serum levels of Ang II and expression of AT1R/TGFß/SMAD, and then decreased the EMT and renal fibrosis in SHRs. Furthermore, the decrease in EMT and renal fibrosis could lead to the restoration of renal function. It seems that ECE or DK could be beneficial for decreasing hypertensive nephropathy by decreasing EMT and renal fibrosis.


Assuntos
Benzofuranos/uso terapêutico , Angiotensina II/metabolismo , Animais , Benzofuranos/farmacologia , Pressão Sanguínea/efeitos dos fármacos , Dioxinas/farmacologia , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Fibrose/tratamento farmacológico , Fibrose/metabolismo , Hipertensão Renal/tratamento farmacológico , Hipertensão Renal/metabolismo , Rim/efeitos dos fármacos , Rim/metabolismo , Masculino , Nefrite/tratamento farmacológico , Nefrite/metabolismo , Ratos , Transdução de Sinais/efeitos dos fármacos
13.
J Cell Mol Med ; 24(22): 13093-13103, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32971570

RESUMO

Angiotensin-converting enzyme-2 (ACE2) and Mas receptor are the major components of the ACE2/Ang 1-7/Mas axis and have been shown to play a protective role in hypertension and hypertensive nephropathy individually. However, the effects of dual deficiency of ACE2 and Mas (ACE2/Mas) on Ang II-induced hypertensive nephropathy remain unexplored, which was investigated in this study in a mouse model of hypertension induced in either ACE2 knockout (KO) or Mas KO mice and in double ACE2/Mas KO mice by subcutaneously chronic infusion of Ang II. Compared with wild-type (WT) animals, mice lacking either ACE2 or Mas significantly increased blood pressure over 7-28 days following a chronic Ang II infusion (P < .001), which was further exacerbated in double ACE2/Mas KO mice (P < .001). Furthermore, compared to a single ACE2 or Mas KO mice, mice lacking ACE2/Mas developed more severe renal injury including higher levels of serum creatinine and a further reduction in creatinine clearance, and progressive renal inflammation and fibrosis. Mechanistically, worsen hypertensive nephropathy in double ACE2/Mas KO mice was associated with markedly enhanced AT1-ERK1/2-Smad3 and NF-κB signalling, thereby promoting renal fibrosis and renal inflammation in the hypertensive kidney. In conclusion, ACE2 and Mas play an additive protective role in Ang II-induced hypertension and hypertensive nephropathy. Thus, restoring the ACE2/Ang1-7/Mas axis may represent a novel therapy for hypertension and hypertensive nephropathy.


Assuntos
Angiotensina II/metabolismo , Enzima de Conversão de Angiotensina 2/genética , Enzima de Conversão de Angiotensina 2/metabolismo , Hipertensão Renal/metabolismo , Nefrite/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animais , Pressão Sanguínea , Fibrose , Deleção de Genes , Hipertensão Renal/genética , Inflamação , Rim/metabolismo , Rim/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nefrite/genética , Proteinúria/genética , Proto-Oncogene Mas , Transdução de Sinais
14.
Lab Invest ; 100(3): 414-425, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31527829

RESUMO

Prostaglandin E2 receptor EP1 (PGE2/EP1) promotes diabetic renal injury, and EP1 receptor deletion improves hyperfiltration, albuminuria, and fibrosis. The role of EP1 receptors in hypertensive kidney disease (HKD) remains controversial. We examined the contribution of EP1 receptors to HKD. EP1 null (EP1-/-) mice were bred with hypertensive TTRhRen mice (Htn) to evaluate kidney function and injury at 24 weeks. EP1 deletion had no effect on elevation of systolic blood pressure in Htn mice (HtnEP1-/-) but resulted in pronounced albuminuria and reduced FITC-inulin clearance, compared with Htn or wild-type (WT) mice. Ultrastructural injury to podocytes and glomerular endothelium was prominent in HtnEP1-/- mice; including widened subendothelial space, subendothelial lucent zones and focal lifting of endothelium from basement membrane, with focal subendothelial cell debris. Cortex COX2 mRNA was increased by EP1 deletion. Glomerular EP3 mRNA was reduced by EP1 deletion, and EP4 by Htn and EP1 deletion. In WT mice, PGE2 increased chloride reabsorption via EP1 in isolated perfused thick ascending limb (TAL), but PGE2 or EP1 deletion did not affect vasopressin-mediated chloride reabsorption. In WT and Htn mouse inner medullary collecting duct (IMCD), PGE2 inhibited vasopressin-water transport, but not in EP1-/- or HtnEP1-/- mice. Overall, EP1 mediated TAL and IMCD transport in response to PGE2 is unaltered in Htn, and EP1 is protective in HKD.


Assuntos
Hipertensão Renal , Podócitos , Receptores de Prostaglandina E Subtipo EP1 , Animais , Modelos Animais de Doenças , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Deleção de Genes , Taxa de Filtração Glomerular/genética , Hipertensão Renal/metabolismo , Hipertensão Renal/patologia , Glomérulos Renais/metabolismo , Glomérulos Renais/patologia , Masculino , Camundongos , Camundongos Transgênicos , Podócitos/citologia , Podócitos/metabolismo , Podócitos/patologia , Receptores de Prostaglandina E Subtipo EP1/genética , Receptores de Prostaglandina E Subtipo EP1/metabolismo
15.
Mol Cell Biochem ; 466(1-2): 1-15, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31912277

RESUMO

Progressive alteration of the extracellular matrix (ECM) is the characteristic of hypertensive nephropathy (HN). Both mesangial and endothelial cells have the ability to synthesize and degrade ECM components, including collagens through the activation of matrix metalloproteinases (MMPs) in stress conditions, such as in hypertension. On the other hand, hydrogen sulfide (H2S) has been shown to mitigate hypertensive renal matrix remodeling. Surprisingly, whether H2S ameliorates receptor-mediated (urokinase plasminogen activator receptor-associated protein, uPARAP/Endo180) collagen dysregulation in Ang-II hypertension is not clear. The purpose of this study was to determine whether Ang-II alters the expression of Endo180, tissue plasminogen activator (tPA), MMPs, and their tissue inhibitors (TIMPs) leading to the dysregulation of cellular collagen homeostasis and whether H2S mitigates the collagen turnover. Mouse mesangial cells (MCs) and glomerular endothelial cells (MGECs) were treated without or with Ang-II and H2S donor GYY (GYY4137) for 48 h. Cell lysates were analyzed by Western blot and RT-PCR, and cells were analyzed by immunocytochemistry. The results indicated that, while Ang-II differentially expressed MMP-13 and TIMP-1 in MCs and in MGECs, it predominantly decreased tPA, Endo 180, and increased plasminogen activator inhibitor-1 (PAI-1), MMP-14, and collagen IIIA and IV in both the cell types. Interestingly, H2S donor GYY treatment normalized the above changes in both the cell types. We conclude that Ang-II treatment causes ECM remodeling in MCs and MGECs through PAI-1/tPA/Endo180 and MMP/TIMP-dependent collagen remodeling, and H2S treatment mitigates remodeling, in part, by modulating these pathways.


Assuntos
Células Endoteliais/metabolismo , Mesângio Glomerular/metabolismo , Hipertensão Renal/metabolismo , Metaloproteinase 13 da Matriz/metabolismo , Metaloproteinase 14 da Matriz/metabolismo , Receptores de Colágeno/metabolismo , Estresse Fisiológico , Animais , Células Endoteliais/patologia , Mesângio Glomerular/patologia , Hipertensão Renal/patologia , Camundongos
16.
BMC Nephrol ; 21(1): 476, 2020 11 11.
Artigo em Inglês | MEDLINE | ID: mdl-33176720

RESUMO

BACKGROUND: Hypertensive nephropathy (HTN) is a kind of renal injury caused by chronic hypertension, which seriously affect people's life. The purpose of this study was to identify the potential biomarkers of HTN and understand its possible mechanisms. METHODS: The dataset numbered GSE28260 related to hypertensive and normotensive was downloaded from NCBI Gene Expression Omnibus. Then, the differentially expressed RNAs (DERs) were screened using R limma package, and functional analyses of DE-mRNA were performed by DAVID. Afterwards, a ceRNA network was established and KEGG pathway was analyzed based on the Gene Set Enrichment Analysis (GSEA) database. Finally, a ceRNA regulatory network directly associated with HTN was proposed. RESULTS: A total of 947 DERs were identified, including 900 DE-mRNAs, 20 DE-lncRNAs and 27 DE-miRNAs. Based on these DE-mRNAs, they were involved in biological processes such as fatty acid beta-oxidation, IRE1-mediated unfolded protein response, and transmembrane transport, and many KEGG pathways like glycine, serine and threonine metabolism, carbon metabolism. Subsequently, lncRNAs KCTD21-AS1, LINC00470 and SNHG14 were found to be hub nodes in the ceRNA regulatory network. KEGG analysis showed that insulin signaling pathway, glycine, serine and threonine metabolism, pathways in cancer, lysosome, and apoptosis was associated with hypertensive. Finally, insulin signaling pathway was screened to directly associate with HTN and was regulated by mRNAs PPP1R3C, PPKAR2B and AKT3, miRNA has-miR-107, and lncRNAs SNHG14, TUG1, ZNF252P-AS1 and MIR503HG. CONCLUSIONS: Insulin signaling pathway was directly associated with HTN, and miRNA has-miR-107 and lncRNAs SNHG14, TUG1, ZNF252P-AS1 and MIR503HG were the biomarkers of HTN. These results would improve our understanding of the occurrence and development of HTN.


Assuntos
Biomarcadores/metabolismo , Redes Reguladoras de Genes , Hipertensão Renal/metabolismo , Insulina/metabolismo , MicroRNAs/metabolismo , Nefrite/metabolismo , Transdução de Sinais/fisiologia , Conjuntos de Dados como Assunto , Humanos , Hipertensão Renal/genética , Nefrite/genética
17.
Ren Fail ; 42(1): 1166-1172, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33241962

RESUMO

The continuous accumulation of extracellular matrix will eventually lead to glomerular sclerosis, interstitial fibrosis, tubular atrophy and vascular sclerosis, which are involved in the progression of chronic kidney disease (CKD). If these processes can be discovered early and effective interventions given in time, the progression of kidney disease may be delayed. Therefore, exploring new biomarkers and therapeutic targets that can identify CKD at an early stage is urgently needed. In recent years, studies have shown that urine periostin may be used as a marker of early renal tubular injury. And in an animal model experiment of hypertensive nephropathy, periostin is involved in the progression of kidney injury and reflects its progression. Here we review the current progress on the role of periostin in pathologic pathways of kidney system to explore whether periostin is a potential therapeutic target for the treatment of CKD.


Assuntos
Moléculas de Adesão Celular/metabolismo , Hipertensão Renal/metabolismo , Túbulos Renais/patologia , Nefrite/metabolismo , Insuficiência Renal Crônica/metabolismo , Animais , Biomarcadores/metabolismo , Moléculas de Adesão Celular/genética , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patologia , Progressão da Doença , Matriz Extracelular , Fibrose , Humanos , Hipertensão Renal/genética , Hipertensão Renal/patologia , Túbulos Renais/metabolismo , Nefrite/genética , Nefrite/patologia , Insuficiência Renal Crônica/genética , Insuficiência Renal Crônica/patologia , Urinálise
18.
J Intern Med ; 285(1): 2-18, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30039620

RESUMO

Cardiovascular disorders including hypertension and associated renal disease are major health problems affecting more than 1.5 billion people worldwide. Apart from nonmodifiable factors such as ageing, family history and gender, both sedentary lifestyle and unhealthy dietary habits are considered as major risk factors. The disorders are interrelated suggesting common pathological pathways. Mechanistically, oxidative stress and compromised function of the nitric oxide synthase (NOS) system leading to endothelial dysfunction and reduction in nitric oxide (NO) bioavailability have been widely implicated and associated with development and progression of disease. New strategies that correct this redox imbalance and increase NO bioactivity may have major clinical implications. The inorganic anions, nitrate and nitrite, are endogenously formed by oxidization of NOS-derived NO, but there are also high amounts of nitrate in our daily diet. In this regard, accumulated evidence over the past two decades demonstrates that these anions can be recycled back to NO and other bioactive nitrogen oxides, thus offering an attractive alternative strategy for therapeutic exploitation. In this review, we describe how dietary stimulation of the nitrate-nitrite-NO pathway affects cardiovascular and renal functions in health and disease via modulation of oxidative stress and NO bioavailability. Clinical studies addressing potential effects on the renal system are still limited, but blood pressure-lowering effects of nitrate supplementation have been demonstrated in healthy and hypertensive subjects as well as in patients with chronic kidney disease. However, larger clinical studies are warranted to reveal whether chronic nitrate treatment can slow-down the progression of cardiorenal disease and associated complications.


Assuntos
Hipertensão Renal/metabolismo , Nefrite/metabolismo , Nitratos/metabolismo , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico/metabolismo , Nitritos/metabolismo , Disponibilidade Biológica , Humanos , Estresse Oxidativo
19.
J Pharmacol Exp Ther ; 370(2): 206-217, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31118214

RESUMO

Dual-specificity protein phosphatase 5 (DUSP5) is a member of the tyrosine-threonine phosphatase family with the ability to dephosphorylate and inactivate extracellular signal-related kinase (ERK). The present study investigates whether knockout (KO) of Dusp5 improves renal hemodynamics and protects against hypertension-induced renal injury. The renal expression of DUSP5 was reduced, and the levels of phosphorylated (p) ERK1/2 and p-protein kinase C (PKC) α were elevated in the KO rats. KO of Dusp5 enhanced the myogenic tone of the renal afferent arteriole and interlobular artery in vitro with or without induction of deoxycorticosterone acetate-salt hypertension. Inhibition of ERK1/2 and PKC diminished the myogenic response to a greater extent in Dusp5 KO rats. Autoregulation of renal blood flow was significantly impaired in hypertensive wild-type (WT) rats but remained intact in Dusp5 KO animals. Proteinuria was markedly decreased in hypertensive KO versus WT rats. The degree of glomerular injury was reduced, and the expression of nephrin in the glomerulus was higher in hypertensive Dusp5 KO rats. Renal fibrosis and medullary protein cast formation were attenuated in hypertensive Dusp5 KO rats in association with decreased expression of monocyte chemoattractant protein 1, transforming growth factor-ß1, matrix metalloproteinase (MMP) 2, and MMP9. These results indicate that KO of Dusp5 protects against hypertension-induced renal injury, at least in part, by maintaining the myogenic tone of the renal vasculature and extending the range of renal blood flow autoregulation to higher pressures, which diminish glomerular injury, protein cast formation, macrophage infiltration, and epithelial-mesenchymal transformation in the kidney. SIGNIFICANCE STATEMENT: Dual-specificity protein phosphatase 5 (DUSP5) is a tyrosine-threonine phosphatase that inactivates extracellular signal-related kinase (ERK). We previously reported that knockout (KO) of Dusp5 enhanced the myogenic response and autoregulation in the cerebral circulation. The present study investigates whether KO of DUSP5 improves renal hemodynamics and protects against hypertension-induced renal injury. Downregulation of DUSP5 enhanced the myogenic tone of renal arteriole and artery and autoregulation of renal blood flow in association with reduced proteinuria, glomerular injury, and interstitial fibrosis after the induction of hypertension. Inhibition of ERK1/2 and protein kinase C diminished the myogenic response to a greater extent in Dusp5 KO rats. These results suggest that DUSP5 might be a viable drug target for the treatment of hypertension nephropathy.


Assuntos
Fosfatases de Especificidade Dupla/deficiência , Fosfatases de Especificidade Dupla/genética , Técnicas de Inativação de Genes , Hipertensão Renal/genética , Nefrite/genética , Animais , Quimiocina CCL2/metabolismo , Regulação para Baixo/genética , Transição Epitelial-Mesenquimal/genética , Fibrose , Regulação Enzimológica da Expressão Gênica/genética , Hemodinâmica/genética , Hipertensão Renal/metabolismo , Hipertensão Renal/patologia , Hipertensão Renal/fisiopatologia , Glomérulos Renais/irrigação sanguínea , Glomérulos Renais/metabolismo , Glomérulos Renais/patologia , Masculino , Metaloproteinases da Matriz/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Desenvolvimento Muscular/genética , Nefrite/metabolismo , Nefrite/patologia , Nefrite/fisiopatologia , Proteína Quinase C/metabolismo , Ratos , Fluxo Sanguíneo Regional/genética , Fator de Crescimento Transformador beta/metabolismo
20.
Clin Exp Pharmacol Physiol ; 46(3): 246-254, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30304543

RESUMO

Hypertensive nephropathy, clinically characterized by progressive renal fibrosis and inflammation, is a severe complication of hypertension. The objectives of this study were to investigate the roles of miR-101a in relieving angiotensin II (Ang II)-mediated hypertensive nephropathy and uncover the possible underlying mechanisms. A hypertensive mouse model was established via continuous 28-day AngII infusion. Systolic blood pressure (SBP), ratio of urine albumin to creatinine, blood urea nitrogen (BUN), serum creatinine (Scr) and glomerular filtration rate (GFR) were evaluated. Dual luciferase reporter assay was used to explore the target of miR-101a. mRNA levels of miR-101a, TGFßRI, fibrotic markers (Collagen I and α-SMA) and pro-inflammatory cytokines (IL-1ß and TNF-α) were determined by real-time PCR. Protein levels of TGFßRI, Collagen I, α-SMA, IL-1ß, TNF-α, t-p65, P-p65, t-Smad3, P-Smad3, t-IκBα and P-IκBα were detected by western blot. MiR-101a mimics significantly improved GFR and inhibited AngII-induced increase in the ratio of urine albumin to creatinine, BUN and Scr. MiR-101a mimics partially abolished AngII-induced increase in the mRNA and protein level of fibrotic markers by targeting TGFßRI and inhibiting TGFß/Smad3 pathway. Moreover, TGFßRI inhibitor galunisertib inhibited AngII-mediated renal injury in mice with hypertensive nephropathy. Additionally, miR-101a overexpression blocked AngII-induced up-regulation of pro-inflammatory markers via suppressing NF-κB pathway. MiR-101a exhibited protective effects against hypertensive nephropathy via inhibiting TGFß/Smad3 and NF-κB signalling pathways.


Assuntos
Angiotensina II/metabolismo , Hipertensão Renal/patologia , MicroRNAs/genética , NF-kappa B/metabolismo , Nefrite/patologia , Transdução de Sinais/genética , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Sequência de Bases , Linhagem Celular , Modelos Animais de Doenças , Fibrose , Hipertensão Renal/genética , Hipertensão Renal/metabolismo , Hipertensão Renal/fisiopatologia , Rim/patologia , Rim/fisiopatologia , Masculino , Camundongos , Nefrite/genética , Nefrite/metabolismo , Nefrite/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA