Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
1.
Arterioscler Thromb Vasc Biol ; 43(9): 1684-1699, 2023 09.
Article in English | MEDLINE | ID: mdl-37409531

ABSTRACT

BACKGROUND: Excess aldosterone is implicated in vascular calcification (VC), but the mechanism by which aldosterone-MR (mineralocorticoid receptor) complex promotes VC is unclear. Emerging evidence indicates that long-noncoding RNA H19 (H19) plays a critical role in VC. We examined whether aldosterone-induced osteogenic differentiation of vascular smooth muscle cells (VSMCs) through H19 epigenetic modification of Runx2 (runt-related transcription factor-2) in a MR-dependent manner. METHODS: We induced in vivo rat model of chronic kidney disease using a high adenine and phosphate diet to explore the relationship among aldosterone, MR, H19, and VC. We also cultured human aortic VSMCs to explore the roles of H19 in aldosterone-MR complex-induced osteogenic differentiation and calcification of VSMCs. RESULTS: H19 and Runx2 were significantly increased in aldosterone-induced VSMC osteogenic differentiation and VC, both in vitro and in vivo, which were significantly blocked by the MR antagonist spironolactone. Mechanistically, our findings reveal that the aldosterone-activated MR bound to H19 promoter and increased its transcriptional activity, as determined by chromatin immunoprecipitation, electrophoretic mobility shift assay, and luciferase reporter assay. Silencing H19 increased microRNA-106a-5p (miR-106a-5p) expression, which subsequently inhibited aldosterone-induced Runx2 expression at the posttranscriptional level. Importantly, we observed a direct interaction between H19 and miR-106a-5p, and downregulation of miR-106a-5p efficiently reversed the suppression of Runx2 induced by H19 silencing. CONCLUSIONS: Our study clarifies a novel mechanism by which upregulation of H19 contributes to aldosterone-MR complex-promoted Runx2-dependent VSMC osteogenic differentiation and VC through sponging miR-106a-5p. These findings highlight a potential therapeutic target for aldosterone-induced VC.


Subject(s)
MicroRNAs , RNA, Long Noncoding , Vascular Calcification , Humans , Rats , Animals , MicroRNAs/metabolism , Aldosterone/toxicity , RNA, Long Noncoding/metabolism , Osteogenesis , Vascular Calcification/chemically induced , Vascular Calcification/genetics , Vascular Calcification/metabolism , Myocytes, Smooth Muscle/metabolism , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism
2.
Circ Res ; 127(3): e80-e93, 2020 07 17.
Article in English | MEDLINE | ID: mdl-32329663

ABSTRACT

RATIONALE: Mitral valve prolapse (MVP) is one of the most common valvular disorders. However, the molecular and cellular mechanisms involved in fibromyxomatous changes in the mitral leaflet tissue have not been elucidated. Aldosterone (Aldo) promotes fibrosis in myocardium, and MR (mineralocorticoid receptor) antagonists (MRAs) improve cardiac function by decreasing cardiac fibrosis. OBJECTIVE: We investigated the role of the Aldo/MR in the fibromyxomatous modifications associated with MVP. METHODS AND RESULTS: Aldo enhanced valvular interstitial cell activation markers and induced endothelial-mesenchymal transition in valvular endothelial cells, resulting in increased proteoglycan secretion. MRA blocked all the above effects. Cytokine arrays showed CT-1 (cardiotrophin-1) to be a mediator of Aldo-induced valvular interstitial cell activation and proteoglycan secretion and CD (cluster of differentiation) 14 to be a mediator of Aldo-induced endothelial-mesenchymal transition and proteoglycan secretion in valvular endothelial cells. In an experimental mouse model of MVP generated by nordexfenfluramine administration, MRA treatment reduced mitral valve thickness and proteoglycan content. Endothelial-specific MR deletion prevented fibromyxomatous changes induced by nordexfenfluramine administration. Moreover, proteoglycan expression was slightly lower in the mitral valves of MVP patients treated with MRA. CONCLUSIONS: These findings demonstrate, for the first time, that the Aldo/MR pathway regulates the phenotypic, molecular, and histological changes of valvular interstitial cells and valvular endothelial cells associated with MVP development. MRA treatment appears to be a promising option to reduce fibromyxomatous alterations in MVP.


Subject(s)
Aldosterone/toxicity , Mitral Valve Prolapse/metabolism , Mitral Valve/drug effects , Receptors, Mineralocorticoid/agonists , Receptors, Mineralocorticoid/metabolism , Aged , Animals , Case-Control Studies , Cell Differentiation/drug effects , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Epithelial-Mesenchymal Transition/drug effects , Female , Fibrosis , Humans , Male , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Mineralocorticoid Receptor Antagonists/pharmacology , Mitral Valve/metabolism , Mitral Valve/pathology , Mitral Valve Prolapse/chemically induced , Mitral Valve Prolapse/pathology , Mitral Valve Prolapse/prevention & control , Paracrine Communication , Phenotype , Prospective Studies , Proteoglycans/metabolism , Receptors, Mineralocorticoid/deficiency , Receptors, Mineralocorticoid/genetics , Signal Transduction
3.
Vascul Pharmacol ; 122-123: 106598, 2019.
Article in English | MEDLINE | ID: mdl-31655164

ABSTRACT

An increase in aldosterone levels positively correlates with an increased risk of acute cardiovascular thrombotic events. The aim of the study was to determine the mechanism of action of prothrombotic aldosterone focusing on the rapid effects of the hormone on platelets, coagulation, and fibrinolysis. A wide panel of advanced ex vivo and in vitro techniques was used for the evaluation of coagulation and fibrinolysis in aldosterone-treated rats. Additionally, two experimental mice models of thrombosis, which allowed for the intravital observation of the first stage of thrombus formation in real time, were used. Acute administration of aldosterone in rats increased the density of fibrin net and platelet aggregates in clots as well as reduced fibrinolysis. These effects were observed within 10 min and were partially suppressed by eplerenone. Moreover, acute administration of aldosterone in mice enhanced platelet accumulation at the site of endothelial injury induced by laser and increased the area of irreversibly activated platelets in FeCl3-induced thrombus. These results demonstrate that aldosterone acutely affects platelets, coagulation, and fibrinolysis, leading to an enhanced thrombosis. The aldosterone effects were mediated partially via a mineralocorticoid receptor. The mechanism seems to involve non-genomic signaling since the effects were observed within a few minutes of aldosterone administration.


Subject(s)
Aldosterone/toxicity , Blood Coagulation/drug effects , Blood Platelets/drug effects , Fibrinolysis/drug effects , Thrombosis/chemically induced , Animals , Blood Platelets/metabolism , Cells, Cultured , Disease Models, Animal , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Male , Mice, Inbred C57BL , Mice, Transgenic , Platelet Aggregation/drug effects , Rats, Wistar , Thrombosis/blood , Time Factors
4.
Hypertens Res ; 42(10): 1518-1527, 2019 10.
Article in English | MEDLINE | ID: mdl-31168059

ABSTRACT

Tubulointerstitial damage is a crucial therapeutic target in preventing chronic kidney disease (CKD) progression. Inappropriately activated renin-angiotensin-aldosterone system (RAAS) in the tubulointerstitial area is strongly associated with tubulointerstitial damage progression. Therefore, this study aimed to determine whether there is a beneficial effect of voluntary running exercise training on aldosterone-induced renal injury. Human L-type fatty acid-binding protein (L-FABP) chromosomal transgenic (L-FABP+/-) male mice were used to evaluate the effect of exercise by using urinary L-FABP, a tubular marker. The mice were assigned to either the exercise group that performed voluntary running exercise training using a running wheel or the control group. Subsequently, two groups were injected with aldosterone (0.125 µg kg-1 min-1) and administered 1% NaCl water, and two groups were administered aldosterone only in solvent 4 weeks after initiating the exercise. Aldosterone was injected for another 4 weeks, and NaCl water was administered from 5 weeks after starting the exercise until 8 weeks. Although both aldosterone and NaCl water significantly decreased the running distance, tubulointerstitial damage involving interstitial infiltration of macrophages and fibrosis and the elevation of urinary human L-FABP induced by aldosterone injection was prevented by voluntary running exercise training. Urinary human L-FABP levels were significantly correlated with the degree of tubulointerstitial damage. In conclusion, voluntary running exercise training delayed tubulointerstitial damage progression in the aldosterone-induced renal injury model and therefore may be a promising nonpharmacological strategy in CKD.


Subject(s)
Aldosterone/toxicity , Exercise , Fatty Acid-Binding Proteins/physiology , Kidney/drug effects , Animals , Fatty Acid-Binding Proteins/urine , Humans , Kidney/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Running , Systole/drug effects
5.
J Cell Biochem ; 119(7): 5934-5943, 2018 07.
Article in English | MEDLINE | ID: mdl-29600538

ABSTRACT

Cardiovascular and renal inflammation induced by Aldosterone (Aldo) plays a pivotal role in the pathogenesis of hypertension and renal fibrosis. GSK-3ß contributes to inflammatory cardiovascular and renal diseases, but its role in Aldo-induced hypertension, and renal damage is not clear. In the present study, rats were treated with Aldo combined with SB-216763 (a GSK-3ß inhibitor) for 4 weeks. Hemodynamic, cardiac, and renal parameters were assayed at the indicated time. Here we found that rats treated with Aldo presented cardiac and renal hypertrophy and dysfunction. Cardiac and renal expression levels of molecular markers attesting inflammation and fibrosis were increased by Aldo infusion, whereas the treatment of SB-216763 reversed these alterations. SB-216763 suppressed cardiac and renal inflammatory cytokines levels (TNF-a, IL-1ß, and MCP-1). Meanwhile, SB-216763 increased the protein levels of LC3-II in the cardiorenal tissues as well as p62 degradation, indicating that SB-216763 induced autophagy activation in cardiac, and renal tissues. Importantly, inhibition of autophagy by 3-MA attenuated the role of SB-216763 in inhibiting perivascular fibrosis, and tubulointerstitial injury. These data suggest that SB-216763 protected against Aldo-induced cardiac and renal injury by activating autophagy, and might be a therapeutic option for salt-sensitive hypertension and renal fibrosis.


Subject(s)
Aldosterone/toxicity , Autophagy , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Heart Diseases/prevention & control , Indoles/pharmacology , Kidney Diseases/prevention & control , Maleimides/pharmacology , Animals , Cytokines/metabolism , Fibrosis/chemically induced , Fibrosis/metabolism , Fibrosis/prevention & control , Heart Diseases/chemically induced , Heart Diseases/metabolism , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/prevention & control , Kidney Diseases/chemically induced , Kidney Diseases/metabolism , Male , Rats , Rats, Wistar
6.
Am J Physiol Renal Physiol ; 314(5): F798-F808, 2018 05 01.
Article in English | MEDLINE | ID: mdl-28659272

ABSTRACT

Mitochondrial dysfunction is increasingly recognized as an important factor in glomerular diseases. Previous study has shown that mitochondrial fission contributed to mitochondrial dysfunction. However, the mechanism of mitochondrial fission on mitochondrial dysfunction in aldosterone-induced podocyte injury remains ambiguous. This study aimed to investigate the pathogenic effect of mitochondrial fission both in vivo and in vitro. In an animal model of aldosterone-induced nephropathy, inhibition of the mitochondrial fission protein dynamin-related protein 1 (Drp1) suppressed aldosterone-induced podocyte injury. In cultured podocytes, aldosterone dose dependently induced Drp1 expression. Knockdown of Drp1 inhibited aldosterone-induced mitochondrial fission, mitochondrial dysfunction, and podocyte apoptosis. Furthermore, aldosterone dose dependently induced p53 expression. Knockdown of p53 inhibited aldosterone-induced Drp1 expression, mitochondrial dysfunction, and podocyte apoptosis. These findings implicated that aldosterone induced mitochondrial dysfunction and podocyte injury mediated by p53/Drp1-dependent mitochondrial fission, which may provide opportunities for therapeutic intervention for podocyte injury.


Subject(s)
Aldosterone/toxicity , Apoptosis/drug effects , Dynamins/metabolism , Kidney Diseases/chemically induced , Mitochondria/drug effects , Mitochondrial Dynamics/drug effects , Podocytes/drug effects , Tumor Suppressor Protein p53/metabolism , Aldosterone/administration & dosage , Animals , Cell Line , Dose-Response Relationship, Drug , Dynamins/genetics , Infusions, Subcutaneous , Kidney Diseases/metabolism , Kidney Diseases/pathology , Kidney Diseases/prevention & control , Male , Mice, Inbred C57BL , Mitochondria/metabolism , Mitochondria/pathology , Podocytes/metabolism , Podocytes/pathology , Quinazolinones/pharmacology , Signal Transduction/drug effects , Tumor Suppressor Protein p53/genetics
7.
Cell Physiol Biochem ; 44(2): 741-750, 2017.
Article in English | MEDLINE | ID: mdl-29169180

ABSTRACT

BACKGROUND/AIMS: Growing evidence suggests mitochondrial dysfunction (MtD) and the Nlrp3 inflammasome play critical roles in chronic kidney disease (CKD) progression. We previously reported that Aldosterone (Aldo)-induced renal injury in vitro is directly caused by mitochondrial reactive oxygen species (mtROS)-mediated activation of the Nlrp3 inflammasome. Here we aimed to determine whether a mitochondria-targeted antioxidant (Mito-Tempo) could prevent Aldo-induced kidney damage in vivo. METHODS: C57BL/6J mice were treated with Aldo and/or Mito-Tempo (or ethanol as a control) for 4 weeks. Renal injury was evaluated by Periodic Acid-Schiff reagent or Masson's trichrome staining and electron microscopy. ROS were measured by DCFDA fluorescence and ELISA. MtD was determined by real-time PCR and electron microscopy. Activation of the Nlrp3 inflammasome and endoplasmic reticulum stress (ERS) was detected via western blot. RESULTS: Compared with control mice, Aldo-infused mice showed impaired renal function, increased mtROS production and MtD, Nlrp3 inflammasome activation, and elevated ERS. We showed administration of Mito-Tempo significantly improved renal function and MtD, and reduced Nlrp3 inflammasome activation and ERS in vivo. CONCLUSION: Mitochondria-targeted antioxidants may attenuate Aldo-infused renal injury by inhibiting MtD, the Nlrp3 inflammasome, and ERS in vivo. Therefore, targeting mtROS might be an effective strategy for preventing CKD.


Subject(s)
Antioxidants/pharmacology , Mitochondria/drug effects , Protective Agents/pharmacology , Aldosterone/toxicity , Animals , Antioxidants/therapeutic use , Cytokines/metabolism , Endoplasmic Reticulum Stress/drug effects , Inflammasomes/metabolism , Kidney/pathology , Kidney Diseases/etiology , Kidney Diseases/pathology , Kidney Diseases/prevention & control , Male , Mice , Mice, Inbred C57BL , Microscopy, Electron , Mitochondria/metabolism , Mitochondria/ultrastructure , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Protective Agents/therapeutic use , Reactive Oxygen Species/metabolism , Up-Regulation/drug effects
8.
PLoS One ; 12(9): e0184888, 2017.
Article in English | MEDLINE | ID: mdl-28926607

ABSTRACT

BACKGROUND: The novel nonsteroidal mineralocorticoid receptor (MR) antagonist finerenone holds promise to be safe and efficient in the treatment of patients with heart failure and/or chronic kidney disease. However, its effects on vascular function remain elusive. PURPOSE: The aim of this study was to determine the functional effect of selective MR antagonism by finerenone in vascular cells in vitro and the effect on vascular remodeling following acute vascular injury in vivo. METHODS AND RESULTS: In vitro, finerenone dose-dependently reduced aldosterone-induced smooth muscle cell (SMC) proliferation, as quantified by BrdU incorporation, and prevented aldosterone-induced endothelial cell (EC) apoptosis, as measured with a flow cytometry based caspase 3/7 activity assay. In vivo, oral application of finerenone resulted in an accelerated re-endothelialization 3 days following electric injury of the murine carotid artery. Furthermore, finerenone treatment inhibited intimal and medial cell proliferation following wire-induced injury of the murine femoral artery 10 days following injury and attenuated neointimal lesion formation 21 days following injury. CONCLUSION: Finerenone significantly reduces apoptosis of ECs and simultaneously attenuates SMC proliferation, resulting in accelerated endothelial healing and reduced neointima formation of the injured vessels. Thus, finerenone appears to provide favorable vascular effects through restoring vascular integrity and preventing adverse vascular remodeling.


Subject(s)
Carotid Artery Injuries/drug therapy , Mineralocorticoid Receptor Antagonists/therapeutic use , Naphthyridines/therapeutic use , Aldosterone/toxicity , Animals , Apoptosis/drug effects , Carotid Arteries/pathology , Carotid Artery Injuries/etiology , Carotid Artery Injuries/pathology , Cell Line , Cell Proliferation/drug effects , Disease Models, Animal , Human Umbilical Vein Endothelial Cells , Humans , Leukocytes/cytology , Leukocytes/immunology , Leukocytes/metabolism , Male , Mice , Mice, Inbred C57BL , Mineralocorticoid Receptor Antagonists/pharmacology , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Naphthyridines/pharmacology , Neointima/pathology , Neointima/prevention & control , Neovascularization, Physiologic/drug effects
9.
Cell Physiol Biochem ; 41(2): 835-848, 2017.
Article in English | MEDLINE | ID: mdl-28214884

ABSTRACT

BACKGROUND/AIMS: The present study investigated whether the transient receptor potential melastatin 4 (TRPM4) channel plays a role in high salt diet (HSD)-induced endothelial injuries. METHODS: Western blotting and immunofluorescence were used to examine TRPM4 expression in the mesenteric endothelium of Dahl salt-sensitive (SS) rats fed a HSD. The MTT, TUNEL, and transwell assays were used to evaluate the cell viability, cell apoptosis, and cell migration, respectively, of human umbilical vein endothelial cells (HUVECs). Enzyme-linked immunosorbent assays were used to determine the concentrations of intercellular adhesion molecule 1 (ICAM-1), vascular cell adhesion protein 1 (VCAM-1), and E-selectin. Carboxy-H2DCFDA, a membrane-permeable reactive oxygen species (ROS)-sensitive fluorescent probe, was used to detect intracellular ROS levels. RESULTS: TRPM4 was mainly expressed near the plasma membrane of mesenteric artery endothelial cells, and its expression level increased in SS hypertensive rats fed a HSD. Its protein expression was significantly upregulated upon treatment with exogenous hydrogen peroxide (H2O2) and aldosterone in cultured HUVECs. Cell viability decreased upon treatment with both agents in a concentration-dependent manner, which could be partially reversed by 9-phenanthrol, a specific TRPM4 inhibitor. Exogenous H2O2 induced apoptosis, enhanced cell migration, and increased the release of adhesion molecules, including ICAM-1, VCAM-1, and E-selectin, all of which were significantly attenuated upon treatment with 9-phenanthrol. Aldosterone and H2O2 induced the accumulation of intracellular ROS, which was significantly inhibited by 9-phenanthrol, suggesting that oxidative stress is one of the mechanisms underlying aldosterone-induced endothelial injury. CONCLUSIONS: Given the fact that oxidative stress and high levels of circulating aldosterone are present in hypertensive patients, we suggest that the upregulation of TRPM4 in the vascular endothelium may be involved in endothelial injuries caused by these stimuli.


Subject(s)
Diet , Endothelium, Vascular/metabolism , TRPM Cation Channels/metabolism , Aldosterone/toxicity , Animals , Apoptosis/drug effects , Cell Movement/drug effects , Cell Survival/drug effects , Cells, Cultured , E-Selectin/analysis , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Human Umbilical Vein Endothelial Cells , Humans , Hydrogen Peroxide/toxicity , Male , Mesenteric Arteries/cytology , Oxidative Stress/drug effects , Phenanthrenes/pharmacology , RNA, Small Interfering/metabolism , Rats , Rats, Inbred Dahl , Sodium Chloride/pharmacology , TRPM Cation Channels/antagonists & inhibitors , TRPM Cation Channels/genetics , Up-Regulation/drug effects
10.
Int J Cardiol ; 232: 233-242, 2017 Apr 01.
Article in English | MEDLINE | ID: mdl-28089144

ABSTRACT

OBJECTIVE: Elevated aldosterone is associated with increased risk of atherosclerosis complications, whereas treatment with mineralocorticoid receptor (MR) antagonists decreases the rate of cardiovascular events. Here we test the hypothesis that aldosterone promotes early atherosclerosis by modulating intercellular adhesion molecule-1 (ICAM-1) expression and investigate the molecular mechanisms by which aldosterone regulates ICAM-1 expression. METHODS AND RESULTS: Apolipoprotein-E (ApoE)-/- mice fed an atherogenic diet and treated with aldosterone for 4weeks showed increased vascular expression of ICAM-1, paralleled by enhanced atherosclerotic plaque size in the aortic root. Moreover, aldosterone treatment resulted in increased plaque lipid and inflammatory cell content, consistent with an unstable plaque phenotype. ApoE/ICAM-1 double knockout (ApoE-/-/ICAM-1-/-) littermates were protected from the aldosterone-induced increase in plaque size, lipid content and macrophage infiltration. Since aldosterone is known to regulate ICAM-1 transcription via MR in human endothelial cells, we explored MR regulation of the ICAM-1 promoter. Luciferase reporter assays performed in HUVECs using deletion constructs of the human ICAM-1 gene promoter showed that a region containing a predicted MR-responsive element (MRE) is required for MR-dependent transcriptional regulation of ICAM-1. CONCLUSIONS: Pro-atherogenic effects of aldosterone are mediated by increased ICAM-1 expression, through transcriptional regulation by endothelial MR. These data enhance our understanding of the molecular mechanism by which MR activation promotes atherosclerosis complications.


Subject(s)
Atherosclerosis/genetics , Gene Expression Regulation , Intercellular Adhesion Molecule-1/genetics , RNA/genetics , Aldosterone/toxicity , Animals , Atherosclerosis/metabolism , Atherosclerosis/pathology , Blotting, Western , Cells, Cultured , Disease Models, Animal , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Flow Cytometry , Genotype , Immunohistochemistry , Intercellular Adhesion Molecule-1/biosynthesis , Intercellular Adhesion Molecule-1/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Mineralocorticoid/metabolism
11.
Am J Physiol Renal Physiol ; 312(4): F556-F564, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28052869

ABSTRACT

Aldosterone (Aldo) has been shown as an important contributor of podocyte injury. However, the underlying molecular mechanisms are still elusive. Recently, the pathogenic role of NOD-like receptor family, pyrin domain containing 3 (NLRP3) inflammasome in mediating renal tubular damage was identified while its role in podocyte injury still needs evidence. Thus the present study was undertaken to investigate the role of NLRP3 inflammasome in Aldo-induced podocyte damage. In vitro, exposure of podocytes to Aldo enhanced NLRP3, caspase-1, and IL-18 expressions in dose- and time-dependent manners, indicating an activation of NLRP3 inflammasome, which was significantly blocked by the mineralocorticoid receptor antagonist eplerenone or the antioxidant N-acetylcysteine. Silencing NLRP3 by a siRNA approach strikingly attenuated Aldo-induced podocyte apoptosis and nephrin protein downregulation in line with the blockade of caspase-1 and IL-18. In vivo, since day 5 of Aldo infusion, NLRP3 inflammasome activation and podocyte injury evidenced by nephrin reduction occurred concurrently. More importantly, immunofluorescence analysis showed a significant induction of NLRP3 in podocytes of glomeruli following Aldo infusion. In the mice with NLRP3 gene deletion, Aldo-induced downregulation of nephrin and podocin, podocyte foot processes, and albuminuria was remarkably improved, indicating an amelioration of podocyte injury. Finally, we observed a striking induction of NLRP3 in glomeruli and renal tubules in line with an enhanced urinary IL-18 output in nephrotic syndrome patients with minimal change disease or focal segmental glomerular sclerosis. Together, these results demonstrated an important role of NLRP3 inflammasome in mediating the podocyte injury induced by Aldo.


Subject(s)
Aldosterone/toxicity , Apoptosis/drug effects , Inflammasomes/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nephrotic Syndrome/metabolism , Podocytes/drug effects , Albuminuria/chemically induced , Albuminuria/metabolism , Animals , Caspase 1/metabolism , Cell Line , Dose-Response Relationship, Drug , Genotype , Humans , Inflammasomes/immunology , Interleukin-18/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/deficiency , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Nephrotic Syndrome/immunology , Nephrotic Syndrome/pathology , Oxidative Stress , Phenotype , Podocytes/immunology , Podocytes/metabolism , Podocytes/pathology , RNA Interference , Receptors, Mineralocorticoid/agonists , Receptors, Mineralocorticoid/metabolism , Signal Transduction/drug effects , Time Factors , Transfection
12.
Am J Physiol Renal Physiol ; 312(4): F589-F598, 2017 04 01.
Article in English | MEDLINE | ID: mdl-27974319

ABSTRACT

MicroRNAs are essential for the maintenance of podocyte homeostasis. Emerging evidence has demonstrated a protective role of microRNA-30a (miR-30a), a member of the miR-30 family, in podocyte injury. However, the roles of other miR-30 family members in podocyte injury are unclear. The present study was undertaken to investigate the contribution of miR-30e to the pathogenesis of podocyte injury induced by aldosterone (Aldo), as well as the underlying mechanism. After Aldo treatment, miR-30e was reduced in a dose-and time-dependent manner. Notably, overexpression of miR-30e markedly attenuated Aldo-induced apoptosis in podocytes. In agreement with this finding, miR-30e silencing led to significant podocyte apoptosis. Mitochondrial dysfunction (MtD) has been shown to be an early event in Aldo-induced podocyte injury. Here we found that overexpression of miR-30e improved Aldo-induced MtD while miR-30e silencing resulted in MtD. Next, we found that miR-30e could directly target the BCL2/adenovirus E1B-interacting protein 3-like (BNIP3L) gene. Aldo markedly enhanced BNIP3L expression in podocytes, and silencing of BNIP3L largely abolished Aldo-induced MtD and cell apoptosis. On the contrary, overexpression of BNIP3L induced MtD and apoptosis in podocytes. Together, these findings demonstrate that miR-30e protects mitochondria and podocytes from Aldo challenge by targeting BNIP3L.


Subject(s)
Aldosterone/toxicity , Apoptosis/drug effects , Membrane Proteins/metabolism , MicroRNAs/metabolism , Mitochondria/drug effects , Mitochondrial Proteins/metabolism , Podocytes/drug effects , Animals , Cell Line , Dose-Response Relationship, Drug , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Proteins/genetics , Podocytes/metabolism , Podocytes/pathology , RNA Interference , Signal Transduction/drug effects , Time Factors , Transfection , Up-Regulation
13.
Mediators Inflamm ; 2016: 4387031, 2016.
Article in English | MEDLINE | ID: mdl-27721575

ABSTRACT

Aldosterone (Aldo) is critically involved in the development of renal injury via the production of reactive oxygen species and inflammation. Endoplasmic reticulum (ER) stress is also evoked in Aldo-induced renal injury. In the present study, we investigated the role of ER stress in inflammation-mediated renal injury in Aldo-infused mice. C57BL/6J mice were randomized to receive treatment for 4 weeks as follows: vehicle infusion, Aldo infusion, vehicle infusion plus tauroursodeoxycholic acid (TUDCA), and Aldo infusion plus TUDCA. The effect of TUDCA on the Aldo-infused inflammatory response and renal injury was investigated using periodic acid-Schiff staining, real-time PCR, Western blot, and ELISA. We demonstrate that Aldo leads to impaired renal function and inhibition of ER stress via TUDCA attenuates renal fibrosis. This was indicated by decreased collagen I, collagen IV, fibronectin, and TGF-ß expression, as well as the downregulation of the expression of Nlrp3 inflammasome markers, Nlrp3, ASC, IL-1ß, and IL-18. This paper presents an important role for ER stress on the renal inflammatory response to Aldo. Additionally, the inhibition of ER stress by TUDCA negatively regulates the levels of these inflammatory molecules in the context of Aldo.


Subject(s)
Endoplasmic Reticulum/drug effects , Kidney/drug effects , Kidney/injuries , Aldosterone/toxicity , Animals , Blotting, Western , Endoplasmic Reticulum/metabolism , Enzyme-Linked Immunosorbent Assay , Kidney Diseases/drug therapy , Kidney Diseases/metabolism , Mice , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Taurochenodeoxycholic Acid/therapeutic use
14.
J Pharmacol Sci ; 132(2): 145-153, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27773435

ABSTRACT

Emerging evidence has suggested that aldosterone has direct deleterious effects on the kidney independently of its hemodynamic effects. However, the detailed mechanisms of these direct effects remain to be elucidated. We have previously reported that camostat mesilate (CM), a synthetic serine protease inhibitor, attenuated kidney injuries in Dahl salt-sensitive rats, remnant kidney rats, and unilateral ureteral obstruction rats, suggesting that some serine proteases would be involved in the pathogenesis of kidney injuries. The current study was conducted to investigate the roles of serine proteases and the beneficial effects of CM in aldosterone-related kidney injuries. We observed a serine protease that was activated by aldosterone/salt in rat kidney lysate, and identified it as plasmin with liquid chromatography-tandem mass spectrometry. Plasmin increased pro-fibrotic and inflammatory gene expressions in rat renal fibroblast cells. CM inhibited the protease activity of plasmin and suppressed cell injury markers induced by plasmin in the fibroblast cells. Furthermore, CM ameliorated glomerulosclerosis and interstitial fibrosis in the kidney of aldosterone/salt-treated rats. Our findings indicate that plasmin has important roles in kidney injuries that are induced by aldosterone/salt, and that serine protease inhibitor could provide a new strategy for the treatment of aldosterone-associated kidney diseases in humans.


Subject(s)
Acute Kidney Injury/drug therapy , Acute Kidney Injury/metabolism , Aldosterone/toxicity , Antifibrinolytic Agents/therapeutic use , Fibrinolysin/metabolism , Serine Proteinase Inhibitors/therapeutic use , Acute Kidney Injury/chemically induced , Animals , Antifibrinolytic Agents/pharmacology , Fibrinolysin/antagonists & inhibitors , Male , Rats , Rats, Sprague-Dawley , Serine Proteinase Inhibitors/pharmacology
15.
Nat Commun ; 7: 10877, 2016 Mar 02.
Article in English | MEDLINE | ID: mdl-26932512

ABSTRACT

Hyper-aldosteronism is associated with myocardial dysfunction including induction of cardiac fibrosis and maladaptive hypertrophy. Mechanisms of these cardiotoxicities are not fully understood. Here we show that mineralocorticoid receptor (MR) activation by aldosterone leads to pathological myocardial signalling mediated by mitochondrial G protein-coupled receptor kinase 2 (GRK2) pro-death activity and GRK5 pro-hypertrophic action. Moreover, these MR-dependent GRK2 and GRK5 non-canonical activities appear to involve cross-talk with the angiotensin II type-1 receptor (AT1R). Most importantly, we show that ventricular dysfunction caused by chronic hyper-aldosteronism in vivo is completely prevented in cardiac Grk2 knockout mice (KO) and to a lesser extent in Grk5 KO mice. However, aldosterone-induced cardiac hypertrophy is totally prevented in Grk5 KO mice. We also show human data consistent with MR activation status in heart failure influencing GRK2 levels. Therefore, our study uncovers GRKs as targets for ameliorating pathological cardiac effects associated with high-aldosterone levels.


Subject(s)
Aldosterone/toxicity , G-Protein-Coupled Receptor Kinases/metabolism , Gene Expression Regulation, Enzymologic/physiology , Heart Diseases/chemically induced , Animals , Arrestins/genetics , Arrestins/metabolism , Cell Culture Techniques , Cell Movement , Heart Failure/pathology , Humans , Mice , Microscopy, Confocal , Muscle Cells/metabolism , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 1/metabolism , Signal Transduction , beta-Arrestins
16.
Hypertension ; 66(1): 158-66, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25987661

ABSTRACT

Activation of the mineralocorticoid receptor has been shown to be deleterious in cardiovascular diseases (CVDs). We have recently shown that lipocalin 2 (Lcn2), or neutrophil gelatinase-associated lipocalin (NGAL), is a primary target of aldosterone/mineralocorticoid receptor in the cardiovascular system. Lcn2 is a circulating protein, which binds matrix metalloproteinase 9 and modulates its stability. We hypothesized that Lcn2 could be a mediator of aldosterone/mineralocorticoid receptor profibrotic effects in the cardiovascular system. Correlations between aldosterone and profibrotic markers, such as procollagen type I N-terminal peptide, were investigated in healthy subjects and subjects with abdominal obesity. The implication of Lcn2 in the mineralocorticoid pathway was studied using Lcn2 knockout mice subjected to a nephrectomy/aldosterone/salt (NAS) challenge for 4 weeks. In human subjects, NGAL/matrix metalloproteinase 9 was positively correlated with plasma aldosterone and fibrosis biomarkers. In mice, loss of Lcn2 prevented the NAS-induced increase of plasma procollagen type I N-terminal peptide, as well as the increase of collagen fibers deposition and collagen I expression in the coronary vessels and the aorta. The lack of Lcn2 also blunted the NAS-induced increase in systolic blood pressure. Ex vivo, treatment of human fibroblasts with recombinant Lcn2 induced the expression of collagen I and the profibrotic galectin-3 and cardiotrophin-1 molecules. Our results showed that Lcn2 plays a key role in aldosterone/mineralocorticoid receptor-mediated vascular fibrosis. The clinical data indicate that this may translate in human patients. Lcn2 is, therefore, a new biotarget in cardiovascular fibrosis induced by mineralocorticoid activation.


Subject(s)
Acute-Phase Proteins/physiology , Aldosterone/toxicity , Lipocalins/physiology , Obesity, Abdominal/physiopathology , Oncogene Proteins/physiology , Proto-Oncogene Proteins/physiology , Acute-Phase Proteins/deficiency , Acute-Phase Proteins/genetics , Acute-Phase Proteins/pharmacology , Aldosterone/blood , Animals , Aorta/drug effects , Aorta/pathology , Cardiomyopathy, Hypertrophic/chemically induced , Cardiomyopathy, Hypertrophic/physiopathology , Cells, Cultured , Cytokines/biosynthesis , Cytokines/genetics , Female , Fibroblasts , Fibrosis , Galectin 3/biosynthesis , Galectin 3/blood , Galectin 3/genetics , Humans , Hypertension/physiopathology , Hypertrophy , Kidney/pathology , Lipocalin-2 , Lipocalins/blood , Lipocalins/genetics , Lipocalins/pharmacology , Male , Mice , Myocardium/cytology , Myocardium/metabolism , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/physiology , Nephrectomy/adverse effects , Obesity, Abdominal/blood , Oncogene Proteins/deficiency , Oncogene Proteins/genetics , Peptide Fragments/blood , Procollagen/blood , Proto-Oncogene Proteins/blood , Proto-Oncogene Proteins/pharmacology , Rats , Recombinant Proteins/pharmacology
18.
Mol Med Rep ; 12(2): 1631-8, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25872931

ABSTRACT

The present study aimed to investigate the effects of aldosterone on vascular endothelial cells and the viability of poly (ADP-ribose) polymerase 1 (PARP1) in cells, and to examine the molecular mechanisms underlying the effects of aldosterone on vascular endothelial cell injury. Cultured endothelial cells were treated either with different concentrations of aldosterone for the same duration or with the same concentrations of aldosterone for different durations, and the levels of apoptosis and activity of PARP1 in the cells were detected, respectively. Aldosterone receptor antagonists or PARP1 inhibitors were added to cells during treatment with aldosterone and the levels of apoptosis and activity of PARP1 were detected. As the concentration of aldosterone increased or the treatment time increased, the number of apoptotic cells and the activity of PARP1 increased. The aldosterone receptor antagonists and PARP1 inhibitors inhibited the increase of apoptosis and PARP1 activity caused by aldosterone treatment. Aldosterone activated the activity of PARP1 via the aldosterone receptor, inhibiting cell proliferation and inducing apoptosis. Treatment with PARP1 may be used as a target for vascular diseases caused by aldosterone at high concentrations.


Subject(s)
Aldosterone/toxicity , Apoptosis/drug effects , Poly(ADP-ribose) Polymerase Inhibitors/toxicity , Poly(ADP-ribose) Polymerases/metabolism , Benzimidazoles/pharmacology , Caspase 3/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/chemistry , Poly(ADP-ribose) Polymerases/genetics , Real-Time Polymerase Chain Reaction
19.
JACC Heart Fail ; 3(1): 59-67, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25458174

ABSTRACT

OBJECTIVES: This study investigated whether galectin (Gal)-3 inhibition could block aldosterone-induced cardiac and renal fibrosis and improve cardiorenal dysfunction. BACKGROUND: Aldosterone is involved in cardiac and renal fibrosis that is associated with the development of cardiorenal injury. However, the mechanisms of these interactions remain unclear. Gal-3, a ß-galactoside-binding lectin, is increased in heart failure and kidney injury. METHODS: Rats were treated with aldosterone-salt combined with spironolactone (a mineralocorticoid receptor antagonist) or modified citrus pectin (a Gal-3 inhibitor), for 3 weeks. Wild-type and Gal-3 knockout mice were treated with aldosterone for 3 weeks. Hemodynamic, cardiac, and renal parameters were analyzed. RESULTS: Hypertensive aldosterone-salt-treated rats presented cardiac and renal hypertrophy (at morphometric, cellular, and molecular levels) and dysfunction. Cardiac and renal expressions of Gal-3 as well as levels of molecular markers attesting fibrosis were also augmented by aldosterone-salt treatment. Spironolactone or modified citrus pectin treatment reversed all of these effects. In wild-type mice, aldosterone did not alter blood pressure levels but increased cardiac and renal Gal-3 expression, fibrosis, and renal epithelial-mesenchymal transition. Gal-3 knockout mice were resistant to aldosterone effects. CONCLUSIONS: In experimental hyperaldosteronism, the increase in Gal-3 expression was associated with cardiac and renal fibrosis and dysfunction but was prevented by pharmacological inhibition (modified citrus pectin) or genetic disruption of Gal-3. These data suggest a key role for Gal-3 in cardiorenal remodeling and dysfunction induced by aldosterone. Gal-3 could be used as a new biotarget for specific pharmacological interventions.


Subject(s)
Acute Kidney Injury/drug therapy , Galectin 3/antagonists & inhibitors , Heart Failure/drug therapy , Spironolactone/therapeutic use , Acute Kidney Injury/chemically induced , Acute Kidney Injury/metabolism , Aldosterone/toxicity , Animals , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Galectin 3/biosynthesis , Heart Failure/chemically induced , Heart Failure/metabolism , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mineralocorticoid Receptor Antagonists/therapeutic use , Rats , Rats, Wistar
20.
Cell Physiol Biochem ; 35(1): 116-25, 2015.
Article in English | MEDLINE | ID: mdl-25547416

ABSTRACT

BACKGROUND/AIM: Aldosterone (Aldo), a mediator of kidney fibrosis, is implicated in the pathogenesis of chronic kidney diseases (CKD). The aim of this study was to evaluate the regulatory role of rapamycin (Rap) in Aldo-induced tubulointerstitial inflammation and fibrosis. METHODS: Uninephrectomized, Sprague-Dawley rats were given 1% NaCl (salt) to drink and were randomized to receive treatment for 28 days as follows: vehicle infusion (control), 0.75 µg/h Aldo subcutaneous infusion, or Aldo infusion plus 1 mg/kg/day of Rap by intraperitoneal injection. The effect of Rap on Aldo-induced fibrosis and renal inflammation was investigated using Masson's technique, immunohistochemistry, and western blotting. The effects of Rap on the Aldo-induced epithelial-mesenchymal transition (EMT) process and on TNF-α mRNA expression and secretion in cultured HK-2 cells were investigated by immunofluorescent staining, western blot, qRT-PCR and ELISA. RESULTS: An in vivo study indicated that signaling by the mammalian target of Rap (mTOR) was activated in rats in the Aldo group compared to controls, as indicated by up-regulated expression of p-mTOR and p-S6K. In addition, the inflammatory response increased, as evidenced by increases in inflammatory markers (MCP-1, ICAM-1, F4/80), and the accumulation of extracellular matrix (ECM), as indicated by increased collagen I and fibronectin expression and pro-fibrogenic gene (PAI-1 and TGF-ß1) expression. These changes were attenuated by Rap treatment. An in vitro study showed that Rap significantly suppressed the Aldo-induced EMT process and TNF-α mRNA expression and secretion in cultured HK-2 cells. CONCLUSIONS: Rap can ameliorate tubulointerstitial inflammation and fibrosis by blocking mTOR signaling. Tubular cells may be a major cell type involved in this physiologic process.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Fibrosis/pathology , Nephritis, Interstitial/pathology , Sirolimus/pharmacology , Aldosterone/toxicity , Animals , Antibiotics, Antineoplastic/therapeutic use , Cell Line , Extracellular Matrix/metabolism , Fibrosis/chemically induced , Fibrosis/drug therapy , Humans , Male , Nephritis, Interstitial/chemically induced , Nephritis, Interstitial/drug therapy , Phosphorylation/drug effects , Plasminogen Activator Inhibitor 1/genetics , Rats , Rats, Sprague-Dawley , Ribosomal Protein S6 Kinases/metabolism , Signal Transduction/drug effects , Sirolimus/therapeutic use , TOR Serine-Threonine Kinases/metabolism , Transforming Growth Factor beta1/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL