Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 93
Filter
1.
Aging (Albany NY) ; 15(22): 12738-12748, 2023 11 17.
Article in English | MEDLINE | ID: mdl-38032896

ABSTRACT

Recently we have shown that adipokine visfatin-induced NLRP3 inflammasome activation contributes to podocyte injury. However, the molecular mechanisms of how visfatin-induces the Nlrp3 inflammasome activation and podocyte damage is still unknown. The present study tested whether membrane raft (MR) redox signalling pathway plays a central role in visfatin-induced NLRP3 inflammasomes formation and activation in podocytes. Upon visfatin stimulation an aggregation of NADPH oxidase subunits, gp91phox and p47phox was observed in the membrane raft (MR) clusters, forming a MR redox signalling platform in podocytes. The formation of this signalling platform was blocked by prior treatment with MR disruptor MCD or NADPH oxidase inhibitor DPI. In addition, visfatin stimulation significantly increased the colocalization of Nlrp3 with Asc or Nlrp3 with caspase-1, IL-ß production, cell permeability in podocytes compared to control cells. Pretreatment with MCD, DPI, WEHD significantly abolished the visfatin-induced colocalization of NLRP3 with Asc or NLRP3 with caspase-1, IL-1ß production and cell permeability in podocytes. Furthermore, Immunofluorescence analysis demonstrated that visfatin treatment significantly decreased the podocin and nephrin expression (podocyte damage) and prior treatments with DPI, WEHD, MCD attenuated this visfatin-induced podocin and nephrin reduction. In conclusion, our results suggest that visfatin stimulates membrane raft clustering in the membrane of podocytes to form redox signaling platforms by aggregation and activation of NADPH oxidase subunits enhancing O2·- production and leading to NLRP3 inflammasome activation in podocytes and ultimate podocyte injury.


Subject(s)
Inflammasomes , Podocytes , Inflammasomes/metabolism , Podocytes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nicotinamide Phosphoribosyltransferase/metabolism , NADPH Oxidases/metabolism , Caspase 1/metabolism , Oxidation-Reduction
2.
Aging (Albany NY) ; 11(24): 12810-12821, 2019 12 13.
Article in English | MEDLINE | ID: mdl-31835256

ABSTRACT

BACKGROUND/AIMS: Recent studies have shown that nicotine induces podocyte damage. However, it remains unknown how nicotine induces podocyte injury. The present study tested whether nicotine induces NLRP3 inflammasomes activation and thereby contributes to podocyte injury. RESULTS: Nicotine treatment significantly increased the colocalization of NLRP3 with Asc, caspase-1 activity, IL-ß production, cell permeability in podocytes compared to control cells. Pretreatment with caspase-1 inhibitor, WEHD significantly abolished the nicotine-induced colocalization of NLRP3 with Asc, caspase-1 activity, IL-1ß production and cell permeability in podocytes. Immunofluorescence analysis showed that nicotine treatment significantly decreased the podocin and nephrin expression compared to control cells. However, prior treatment with WEHD attenuated the nicotine-induced podocin and nephrin reduction. In addition, we found that nicotine treatment significantly increased the O2.- production compared to control cells. However, prior treatment with WEHD did not alter the nicotine-induced O2.- production. Furthermore, prior treatment with ROS scavenger, NAC significantly attenuated the nicotine-induced caspase-1 activity, IL-1ß production, podocin and nephrin reduction in podocytes. CONCLUSIONS: Nicotine-induced the NLRP3 inflammasome activation in podocytes and thereby results in podocyte injury. METHODS: Inflammasome formation and immunofluorescence expressions were quantified by confocal microscopy. Caspase-1 activity, IL-1ß production and O2.- production were measured by ELISA and ESR.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , Inflammasomes/metabolism , Nicotine/pharmacology , Podocytes/drug effects , Adaptor Proteins, Signal Transducing/genetics , Animals , Apoptosis Regulatory Proteins/genetics , Caspase 1/genetics , Caspase 1/metabolism , Cell Line , Gene Expression Regulation/drug effects , Inflammasomes/drug effects , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Mice , Nicotinic Agonists/pharmacology , Permeability , Podocytes/metabolism
3.
J Cell Mol Med ; 23(12): 8139-8150, 2019 12.
Article in English | MEDLINE | ID: mdl-31565858

ABSTRACT

Sodium butyrate (NaBu) is reported to play important roles in a number of chronic diseases. The present work is aimed to investigate the effect of NaBu on angiotensin II (Ang II)-induced cardiac hypertrophy and the underlying mechanism in in vivo and in vitro models. Sprague Dawley rats were infused with vehicle or Ang II (200 ng/kg/min) and orally administrated with or without NaBu (1 g/kg/d) for two weeks. Cardiac hypertrophy parameters and COX2/PGE2 pathway were analysed by real-time PCR, ELISA, immunostaining and Western blot. The cardiomyocytes H9C2 cells were used as in vitro model to investigate the role of NaBu (2 mmol/L) in inhibition of Ang II-induced cardiac hypertrophy. NaBu significantly attenuated Ang II-induced increase in the mean arterial pressure. Ang II treatment remarkably increased cardiac hypertrophy as indicated by increased ratio of heart weight/body weight and enlarged cardiomyocyte size, extensive fibrosis and inflammation, as well as enhanced expression of hypertrophic markers, whereas hearts from NaBu-treated rats exhibited a significant reduction in these hypertrophic responses. Mechanistically, NaBu inhibited the expression of COX2/PGE2 along with production of ANP and phosphorylated ERK (pERK) stimulated by Ang II in in vivo and in vitro, which was accompanied by the suppression of HDAC5 and HDAC6 activities. Additionally, knocking down the expression of HDAC5 and HDAC6 via gene-editing strategy dramatically blocked Ang II-induced hypertrophic responses through COX2/PGE2 pathway. These results provide solid evidence that NaBu attenuates Ang II-induced cardiac hypertrophy by inhibiting the activation of COX2/PGE2 pathway in a HDAC5/HDAC6-dependent manner.


Subject(s)
Butyric Acid/pharmacology , Cardiomegaly/prevention & control , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Histone Deacetylase 6/metabolism , Histone Deacetylases/metabolism , Angiotensin II , Animals , Cardiomegaly/chemically induced , Cardiomegaly/metabolism , Cell Line , Cyclooxygenase 2/genetics , Gene Expression/drug effects , Gene Knockdown Techniques , Histone Deacetylase 6/genetics , Histone Deacetylases/genetics , Hypertension/chemically induced , Hypertension/genetics , Hypertension/physiopathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/genetics
4.
Cell Physiol Biochem ; 53(2): 355-365, 2019.
Article in English | MEDLINE | ID: mdl-31385664

ABSTRACT

BACKGROUND/AIMS: NLRP3 inflammasome activation has been reported to be an early mechanism responsible for glomerular inflammation and injury in obese mice. However, the precise mechanism of obesity-induced NLRP3 inflammasome activation remains unknown. The present study explored whether adipokine visfatin mediates obesity-induced NLRP3 inflammasome activation and consequent podocyte injury. METHODS: Inflammasome formation and immunofluorescence expressions were quantified by confocal microscopy. Caspase-activity, IL-1ß production and VEGF concentrations were measured by ELISA. RESULTS: Confocal microscopic analysis showed that visfatin treatment increased the colocalization of Nlrp3 with Asc or Nlrp3 with caspase-1 in podocytes indicating the formation of NLRP3 inflammasomes. This visfatin-induced NLRP3 inflammasome formation was abolished by pretreatment of podocytes with Asc siRNA. Correspondingly, visfatin treatment significantly increased the caspase-1 activity and IL-1ß production in podocytes, which was significantly attenuated by Asc siRNA transfection. Further RT-PCR and confocal microscopic analysis demonstrated that visfatin treatment significantly decreased the podocin expression (podocyte damage). Podocytes pretreatment with Asc siRNA or caspase-1 inhibitor, WEHD attenuated this visfatin-induced podocin reduction. Furthermore, Asc siRNA transfection was found to preserve podocyte morphology by maintaining the distinct arrangement of F-actin fibers normally lost in response to visfatin. It also prevented podocyte dysfunction by restoring visfatin-induced suppression of VEGF production and secretion. CONCLUSION: Visfatin induces NLRP3 inflammasome activation in podocytes and thereby resulting in podocyte injury.


Subject(s)
Adipokines/immunology , Inflammasomes/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Nicotinamide Phosphoribosyltransferase/immunology , Podocytes/immunology , Animals , Cell Line , Inflammation/immunology , Inflammation/pathology , Interleukin-1beta/immunology , Mice , Obesity/immunology , Obesity/pathology , Podocytes/cytology , Podocytes/pathology , Vascular Endothelial Growth Factor A/immunology
5.
Int J Mol Sci ; 20(14)2019 Jul 22.
Article in English | MEDLINE | ID: mdl-31336567

ABSTRACT

The intestinal microbe-derived metabolite trimethylamine N-oxide (TMAO) is implicated in the pathogenesis of cardiovascular diseases (CVDs). The molecular mechanisms of how TMAO induces atherosclerosis and CVDs' progression are still unclear. In this regard, high-mobility group box protein 1 (HMGB1), an inflammatory mediator, has been reported to disrupt cell-cell junctions, resulting in vascular endothelial hyper permeability leading to endothelial dysfunction. The present study tested whether TMAO associated endothelial dysfunction results via HMGB1 activation. Biochemical and RT-PCR analysis showed that TMAO increased the HMGB1 expression in a dose-dependent manner in endothelial cells. However, prior treatment with glycyrrhizin, an HMGB1 binder, abolished the TMAO-induced HMGB1 production in endothelial cells. Furthermore, Western blot and immunofluorescent analysis showed significant decrease in the expression of cell-cell junction proteins ZO-2, Occludin, and VE-cadherin in TMAO treated endothelial cells compared with control cells. However, prior treatment with glycyrrhizin attenuated the TMAO-induced cell-cell junction proteins' disruption. TMAO increased toll-like receptor 4 (TLR4) expression in endothelial cells. Inhibition of TLR4 expression by TLR4 siRNA protected the endothelial cells from TMAO associated tight junction protein disruption via HMGB1. In conclusion, our results demonstrate that HMGB1 is one of the important mediators of TMAO-induced endothelial dysfunction.


Subject(s)
Endothelium/drug effects , Endothelium/metabolism , HMGB1 Protein/metabolism , Methylamines/pharmacology , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Extracellular Space/metabolism , Humans , Intercellular Junctions/drug effects , Intercellular Junctions/metabolism , Tight Junctions/drug effects , Tight Junctions/metabolism , Toll-Like Receptor 4/metabolism
6.
Basic Clin Pharmacol Toxicol ; 124(4): 370-384, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30311396

ABSTRACT

Amitriptyline is a tricyclic antidepressant and an inhibitor of lysosomal acid sphingomyelinase (ASM). Amitriptyline is well known for its cardiovascular side effects and toxicity in psychiatric patients. However, the mechanisms underlying the cardiovascular side effects of amitriptyline remain largely undefined. This study aimed to determine the effects of amitriptyline on angiogenic capability of vascular endothelial cells in physiological settings and identify its mechanism of action. The ex vivo aortic ring angiogenesis and in vitro-cultured endothelial cell tube formation assay were used to assess the effects of amitriptyline on endothelial angiogenic capability. It was demonstrated that amitriptyline impaired the angiogenesis of aortic rings, which was similar to that found in aortic rings with haploinsufficiency of the ASM gene. In cultured mouse microvascular endothelial cells (MVECs), amitriptyline impaired the proliferation and tube formation under basal condition, which were accompanied by attenuated angiogenic signalling pathways such as endothelial nitric oxide synthase, Akt and Erk1/2 pathways. Mechanistically, amitriptyline inhibited autophagic flux without affecting autophagosome biogenesis at basal condition. ASM gene silencing or autophagy inhibition mimics the inhibitory effects of amitriptyline on endothelial cell proliferation and tube formation. Collectively, our data suggest that amitriptyline inhibits endothelial cell proliferation and angiogenesis via blockade of ASM-autophagic flux axis. It is implicated that the cardiovascular side effects of amitriptyline may be associated with its inhibitory action on physiological angiogenesis.


Subject(s)
Amitriptyline/toxicity , Antidepressive Agents, Tricyclic/toxicity , Autophagy/drug effects , Neovascularization, Physiologic/drug effects , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/pathology , Cell Proliferation/drug effects , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/pathology , Gene Knockdown Techniques , Male , Mice , Mice, Inbred C57BL , Signal Transduction/drug effects
7.
Cell Death Dis ; 9(12): 1145, 2018 11 19.
Article in English | MEDLINE | ID: mdl-30451833

ABSTRACT

Accumulating evidence indicates a critical role of autophagy in regulating vascular smooth muscle cell (SMC) homeostasis in atherogenesis. However, little is known about the modulatory role of autophagy in PDGF-BB-induced SMC transition towards the synthetic phenotype and extracellular matrix remodeling. We recently demonstrated that acid sphingomyelinase (ASM, encoded by Smpd1 gene) controls autophagy maturation in coronary arterial SMCs. Here, we demonstrate that PDGF-BB stimulation causes a myofibroblast-like non-canonical synthetic phenotype transition in Smpd1-/- SMCs. These non-canonical phenotypic changes induced by PDGF-BB in Smpd1-/- SMCs were characterized by increased expression of fibroblast-specific protein (FSP-1), massive deposition of collagen type I, decreased cell size, elevated inflammatory status with enhanced cytokine release and adhesion molecule expression. Mechanistically, PDGF-BB induces prolonged Akt activation that causes decreased autophagosome biogenesis and thereby exaggerates p62/SQSTM1 accumulation in Smpd1-/- SMCs. More importantly, Akt inhibition or p62/SQSTM1 gene silencing attenuates PDGF-BB-induced phenotypic changes in Smpd1-/- SMCs. This first demonstration of a p62/SQSTM1-dependent myofibroblast-like phenotypic transition in Smpd1-/- SMCs suggests that ASM-mediated autophagy pathway contributes to maintaining the arterial smooth muscle homeostasis in situation of vascular remodeling during atherosclerosis.


Subject(s)
Atherosclerosis/genetics , Autophagy/genetics , Sequestosome-1 Protein/genetics , Sphingomyelin Phosphodiesterase/genetics , Animals , Atherosclerosis/metabolism , Atherosclerosis/pathology , Becaplermin/genetics , Calcium-Binding Proteins/genetics , Coronary Vessels/metabolism , Coronary Vessels/pathology , Disease Models, Animal , Gene Expression Regulation/genetics , Humans , Mice , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Myofibroblasts/metabolism , Myofibroblasts/pathology , Phenotype , Phosphoproteins/genetics , Proto-Oncogene Proteins c-sis/genetics
8.
Cell Physiol Biochem ; 50(4): 1585-1600, 2018.
Article in English | MEDLINE | ID: mdl-30359991

ABSTRACT

BACKGROUND/AIMS: Angiotensin II (Ang II) is an octapeptide hormone that plays a significant role in mediating hypertension. Although hypertension is considered a chronic inflammatory disease, the molecular basis of the sterile inflammatory response involved in hypertension remains unclear. METHODS: We investigated the role of macrophage NLRP3 inflammasomes in engulfing and digesting microbes, a key macrophage function, and in early onset of hypertension-associated macrophage injury using biochemical analyses, gene silencing, molecular biotechnology, immunofluorescence, and microbiology. RESULTS: Ang II stimulation decreased nitric oxide (NO) release and macrophage digestion in cultured THP-1 cells and markedly increased NLRP3 inflammasome formation and activation. NO release and macrophage digestion were restored by NLRP3 inflammasome inhibition with isoliquiritigenin and gene silencing. This Ang II-induced upregulation of NLRP3 inflammasomes in macrophages was attributed to lysosomal damage and release of cathepsin B. Mechanistically, losartan, a nonpeptide Ang II receptor antagonist, decreased Ang II-induced NLRP3 inflammasome activation, lysosomal membrane permeability, lysosomal cathepsin B release, and macrophage digestion dysfunction. Similarly, Ang II-induced macrophage microbe digestion and NO production, which were blocked by ATI gene silencing. In addition, in vivo experiments showed that the bacteria scavenging function was clearly decreased in macrophages from Ang II-induced hypertensive mice. CONCLUSION: Angiotensin II enhances lysosomal membrane permeabilization and the consequent release of lysosomal cathepsin B, resulting in activation of the macrophage NLRP3 inflammasome. This may contribute to NO mediation of dysfunction in digesting microbes.


Subject(s)
Angiotensin II/pharmacology , Cathepsin B/metabolism , Inflammasomes/metabolism , Macrophages/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Animals , Cathepsin B/antagonists & inhibitors , Cell Line , Cell Membrane Permeability/drug effects , Chalcones/pharmacology , Escherichia coli/physiology , Hypertension/metabolism , Hypertension/pathology , Losartan/pharmacology , Lysosomes/metabolism , Macrophages/cytology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Nitric Oxide/metabolism , Phagocytosis/drug effects , RNA Interference , RNA, Small Interfering/metabolism
9.
Stroke Vasc Neurol ; 3(3): 153-159, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30294471

ABSTRACT

Background and purpose: Recent reports from our laboratory demonstrated the post-ischaemic expression profile of various matrix metalloproteinases (MMPs) in rats and the detrimental role of MMP-12 in post-stroke brain damage. We hypothesise that the post-stroke dysregulation of MMPs is similar across species and that genetic deletion of MMP-12 would not affect the post-stroke expression of other MMPs. We tested our hypothesis by determining the pre-ischaemic and post-ischaemic expression profile of MMPs in wild-type and MMP-12 knockout mice. Methods: Focal cerebral ischaemia was induced in wild-type and MMP-12 knockout mice by middle cerebral artery occlusion procedure by insertion of a monofilament suture. One hour after ischaemia, reperfusion was initiated by removing the monofilament. One day after reperfusion, ischaemic brain tissues from various groups of mice were collected, and total RNA was isolated and subjected to cDNA synthesis followed by PCR analysis. Results: Although the post-stroke expression profile of MMPs in the ischaemic brain of mice is different from rats, there is a clear species similarity in the expression of MMP-12, which was found to be predominantly upregulated in both species. Further, the post-stroke induction or inhibition of various MMPs in MMP-12 knockout mice is different from their respective expression profile in wild-type mice. Moreover, the brain mRNA expression profile of various MMPs in MMP-12 knockout mice under normal conditions is also different to their expression in wild-type mice. Conclusions: In the ischaemic brain, MMP-12 upregulates several fold higher than any other MMP. Mice derived with the genetic deletion of MMP-12 are constitutive and have altered MMP expression profile both under normal and ischaemic conditions.


Subject(s)
Gene Deletion , Infarction, Middle Cerebral Artery/enzymology , Matrix Metalloproteinase 12/deficiency , RNA, Messenger/metabolism , Transcriptome , Animals , Disease Models, Animal , Gene Expression Regulation, Enzymologic , Infarction, Middle Cerebral Artery/genetics , Male , Matrix Metalloproteinase 12/genetics , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/genetics , Rats , Species Specificity , Time Factors
10.
Sci Rep ; 7(1): 15878, 2017 Nov 20.
Article in English | MEDLINE | ID: mdl-29158549

ABSTRACT

Proteinuria is closely associated with the progression of chronic kidney diseases (CKD) by producing renal tubulointerstitial fibrosis. Over-activation of hypoxia inducible factor (HIF)-1α has been implicated in the progression of CKD. The present study tested the hypothesis that HIF-1α mediates albumin-induced profibrotic effect in cultured renal proximal tubular cells. Incubation of the cells with albumin (40 µg/ml) for 72 hrs significantly increased the protein levels of HIF-1α, tissue inhibitor of metalloproteinase (TIMP)-1 and collagen-I, which were blocked by HIF-1α shRNA. Albumin also stimulated an epithelial-mesenchymal transition (EMT) as indicated by the decrease in epithelial marker E-cadherin, and the increase in mesenchymal markers α-smooth muscle actin and fibroblast-specific protein 1. HIF-1α shRNA blocked albumin-induced changes in these EMT markers as well. Furthermore, albumin reduced the level of hydroxylated HIF-1α, indicating an inhibition of the activity of prolyl-hydroxylases, enzymes promoting the degradation of HIF-1α. An anti-oxidant ascorbate reversed albumin-induced inhibition of prolyl-hydroxylase activity. Overexpression of prolyl-hydroxylase 2 (PHD2) transgene, a predominant isoform of PHDs in renal tubules, to reduce HIF-1α level significantly attenuated albumin-induced increases in TIMP-1 and collagen-I levels. These results suggest that albumin-induced oxidative stress inhibits PHD activity to accumulate HIF-1α, which mediates albumin-induced profibrotic effects in renal tubular cells.


Subject(s)
Fibrosis/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Procollagen-Proline Dioxygenase/genetics , Renal Insufficiency, Chronic/genetics , Tissue Inhibitor of Metalloproteinase-1/genetics , Actins/genetics , Albumins/pharmacology , Animals , Cadherins/genetics , Epithelial-Mesenchymal Transition/genetics , Fibrosis/pathology , Gene Expression Regulation/genetics , Humans , Hypoxia-Inducible Factor-Proline Dioxygenases , Kidney/metabolism , Kidney/pathology , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Proteinuria/genetics , Rats , Renal Insufficiency, Chronic/pathology , S100 Calcium-Binding Protein A4/genetics , Signal Transduction/genetics
11.
Cell Physiol Biochem ; 44(1): 152-162, 2017.
Article in English | MEDLINE | ID: mdl-29130962

ABSTRACT

BACKGROUND/AIM: Plasma trimethylamine-N-oxide (TMAO), a product of intestinal microbial metabolism of dietary phosphatidylcholine has been recently associated with atherosclerosis and increased risk of cardiovascular diseases (CVD) in rodents and humans. However, the molecular mechanisms of how TMAO induces atherosclerosis and CVD progression are still unclear. The present study tested whether TMAO induces NLRP3 inflammasome formation and activation and thereby contributes to endothelial injury initiating atherogenesis. METHODS: Inflammasome formation and activation was determined by confocal microscopy, caspase-1 activity was measured by colorimetric assay, IL-1ß production was measured using ELISA, cell permeability was determined by microplate reader and ZO-1 expression was determined by western blot analysis and confocal microscopy. In in vivo experiments, TMAO was infused by osmotic pump implantation. RESULTS: TMAO treatment significantly increased the colocalization of NLRP3 with Asc or NLRP3 with caspase-1, caspase-1 activity, IL-1ß production, cell permeability in carotid artery endothelial cells (CAECs) compared to control cells. Pretreatment with caspase-1 inhibitor, WEHD or Nlrp3 siRNA abolished the TMAO-induced inflammasome formation, activation and cell permeability in these cells. In addition, we explored the mechanisms by which TMAO activates NLRP3 inflammasomes. TMAO-induced the activation of NLRP3 inflammasomes was associated with both redox regulation and lysosomal dysfunction. In animal experiments, direct infusion of TMAO in mice with partially ligated carotid artery were found to have increased NLRP3 inflammasome formation and IL-1ß production in the intima of wild type mice. CONCLUSION: The formation and activation of NLRP3 inflammasomes by TMAO may be an important initiating mechanism to turn on the endothelial inflammatory response leading to endothelial dysfunction.


Subject(s)
Inflammasomes/drug effects , Methylamines/toxicity , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Carotid Arteries/cytology , Carotid Arteries/pathology , Caspase 1/chemistry , Caspase 1/metabolism , Caspase Inhibitors/pharmacology , Cells, Cultured , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Inflammasomes/metabolism , Interleukin-1beta/metabolism , Male , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Permeability/drug effects , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction/drug effects
12.
Cell Physiol Biochem ; 42(3): 1264-1273, 2017.
Article in English | MEDLINE | ID: mdl-28693025

ABSTRACT

BACKGROUND: Our previous study has detected a stem cell deficiency in the renal medulla in Dahl salt-sensitive (S) rats. This study determined whether infusion of valproic acid (VA), an agent known to stimulate the stem cell function, attenuated salt-sensitive hypertension in Dahl S rats. METHODS: Uninephrectomized Dahl S rats were infused with vehicle or VA (50mg/kg/d) into the renal medulla and fed with a low (LS) or high salt diet (HS). Stem cell marker and number were analyzed by immunohistochemistry, Real-time RT-PCR and Western blot. Sodium excretion and blood pressure were measured. RESULTS: VA significantly increased the mRNA and protein levels of FGF2, a stem cell niche factor, and CD133, a stem cell marker. The number of CD133+ cells was significantly increased in the renal medulla in VA-treated rats. Meanwhile, high salt-induced increases in the mRNA level of proinflammatory factors interleukin-1ß and interleukin-6 were blocked in VA-treated rats. Functionally, sodium excretion in response to the blood pressure increase and acute sodium loading was significantly enhanced, sodium retention attenuated, high salt-induced increase of blood pressure reduced in VA-treated rats. CONCLUSION: Activation of stem cell function by VA inhibits the activation of proinflammatory factors and attenuates salt-sensitive hypertension in Dahl S rats.


Subject(s)
Antihypertensive Agents/pharmacology , Enzyme Inhibitors/pharmacology , Hypertension/drug therapy , Kidney Medulla/cytology , Kidney Medulla/drug effects , Stem Cells/drug effects , Valproic Acid/pharmacology , AC133 Antigen/analysis , AC133 Antigen/metabolism , Animals , Fibroblast Growth Factor 2/analysis , Fibroblast Growth Factor 2/metabolism , Hypertension/metabolism , Male , Rats, Inbred Dahl , Sodium Chloride, Dietary/metabolism , Stem Cells/cytology , Valproic Acid/administration & dosage
13.
Redox Biol ; 13: 336-344, 2017 10.
Article in English | MEDLINE | ID: mdl-28633109

ABSTRACT

The NLRP3 inflammasome has been reported to be activated by atherogenic factors, whereby endothelial injury and consequent atherosclerotic lesions are triggered in the arterial wall. However, the mechanisms activating and regulating NLRP3 inflammasomes remain poorly understood. The present study tested whether acid sphingomyelinase (ASM) and ceramide associated membrane raft (MR) signaling platforms contribute to the activation of NLRP3 inflammasomes and atherosclerotic lesions during hypercholesterolemia. We found that 7-ketocholesterol (7-Keto) or cholesterol crystal (ChC) markedly increased the formation and activation of NLRP3 inflammasomes in mouse carotid arterial endothelial cells (CAECs), as shown by increased colocalization of NLRP3 with ASC or caspase-1, enhanced caspase-1 activity and elevated IL-1ß levels, which were markedly attenuated by mouse Asm siRNA, ASM inhibitor- amitriptyline, and deletion of mouse Asm gene. In CAECs with NLRP3 inflammasome formation, membrane raft (MR) clustering with NADPH oxidase subunits was found remarkably increased as shown by CTXB (MR marker) and gp91phox aggregation indicating the formation of MR redox signaling platforms. This MR clustering was blocked by MR disruptor (MCD), ROS scavenger (Tempol) and TXNIP inhibitor (verapamil), accompanied by attenuation of 7-Keto or ChC-induced increase in caspase-1 activity. In animal experiments, Western diet fed mice with partially ligated left carotid artery (PLCA) were found to have significantly increased neointimal formation, which was associated with increased NLRP3 inflammasome formation and IL-1ß production in the intima of Asm+/+ mice but not in Asm-/- mice. These results suggest that Asm gene and ceramide associated MR clustering are essential to endothelial inflammasome activation and dysfunction in the carotid arteries, ultimately determining the extent of atherosclerotic lesions.


Subject(s)
Hypercholesterolemia/metabolism , Inflammasomes/metabolism , Neointima/metabolism , Sphingomyelin Phosphodiesterase/metabolism , Animals , Carotid Arteries/cytology , Carotid Arteries/metabolism , Caspase 1/metabolism , Cells, Cultured , Hypercholesterolemia/pathology , Inflammasomes/genetics , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Male , Membrane Microdomains/metabolism , Mice , NADP/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Sphingomyelin Phosphodiesterase/genetics
14.
J Hypertens ; 35(9): 1872-1880, 2017 09.
Article in English | MEDLINE | ID: mdl-28445205

ABSTRACT

BACKGROUND: We have previously shown that high salt intake suppresses the expression of prolyl hydroxylase domain-containing protein 2 (PHD2), an enzyme promoting the degradation of hypoxia-inducible factor (HIF)-1α, and increases HIF-1α along with its target genes in the renal medulla, which promotes sodium excretion and regulates salt sensitivity of blood pressure. However, it remains unknown how high salt inhibits the expression of PHD2. METHOD AND RESULTS: The current study first revealed that high-salt-induced PHD2 inhibition was due to the enhanced decay of mRNA. We then found that high salt significantly increased the expression of miR-429, which was subsequently proven to target the 3'-untranslated region of PHD2 and reduce PHD2 levels, in the renal medulla. To define the functional role of renal medullary miR-429 in the regulation of PHD2/HIF-1α-mediated renal adaptation to high salt intake and salt sensitivity of blood pressure, we locally inhibited miR-429 in the renal medulla by locked nucleic acid anti-miR-429 in uninephrectomized rats. Our results demonstrated that inhibition of miR-429 remarkably increased the levels of PHD2, which disrupted PHD2-associated adaptive activation of HIF-1α-mediated gene expression in response to high salt in the renal medulla and consequently inhibited urinary sodium excretion, enhanced sodium retention in response to chronic sodium overloading, and as a result, produced a salt-sensitive hypertension. CONCLUSION: It is concluded that miR-429 is an important upstream mediator in PHD2/HIF-1α-associated renal adaptation to high salt intake and that deficiency in miR-429-mediated PHD2 inhibition in response to high salt in the renal medulla may represent a pathogenic mechanism for salt-sensitive hypertension.


Subject(s)
Blood Pressure , Hypertension , Kidney Medulla , MicroRNAs , Sodium Chloride/pharmacology , Animals , Blood Pressure/drug effects , Blood Pressure/genetics , Hypertension/metabolism , Kidney Medulla/drug effects , Kidney Medulla/metabolism , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , MicroRNAs/metabolism , Rats , Rats, Sprague-Dawley
15.
Free Radic Biol Med ; 106: 236-244, 2017 05.
Article in English | MEDLINE | ID: mdl-28193546

ABSTRACT

NADPH oxidase (NOX)-derived reactive oxygen species (ROS) have been demonstrated to mediate the activation of NOD-like receptor protein 3 (NLRP3) inflammasomes in podocytes in response to elevated levels of homocysteine (Hcys). However, it remains unknown how NLRP3 inflammasome activation is triggered by NOX. The present study tested whether the guanine nucleotide exchange factor Vav2 mediates Rac1-mediated NOX activation in response to elevated Hcys leading to NLRP3 inflammasome activation in podocytes and consequent glomerular injury. In a mouse model of hyperhomocysteinemia (hHcys), we found that mice with hHcys (on the FF diet) or oncoVav2 (a constitutively active form of Vav2) transfection in the kidney exhibited increased colocalization of NLRP3 with apoptosis-associated speck-like protein (ASC) or caspase-1 and elevated IL-1ß levels in glomeruli, indicating the formation and activation of the NLRP3 inflammasome. This glomerular NLRP3 inflammasome activation was accompanied by podocyte dysfunction and glomerular injury, even sclerosis. Local transfection of Vav2 shRNA plasmids significantly attenuated hHcys-induced NLRP3 inflammasome activation, podocyte injury, and glomerular sclerosis. In cultured podocytes, Hcys treatment and oncoVav2 transfection were also found to increase NLRP3 inflammasome formation and activation, which were all inhibited by Vav2 shRNA. Furthermore, Vav2 shRNA prevented Hcys-induced podocyte damage as shown by restoring Hcys-impaired VEGF secretion and podocin production. This inhibitory action of Vav2 shRNA on Hcys-induced podocyte injury was associated with reduction of Rac1 activity and ROS production. These results suggest that elevated Hcys levels activate Vav2 and thereby increase NOX activity leading to ROS production, which triggers NLRP3 inflammasome activation, podocyte dysfunction and glomerular injury.


Subject(s)
Hyperhomocysteinemia/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neuropeptides/metabolism , Proto-Oncogene Proteins c-vav/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , CARD Signaling Adaptor Proteins/genetics , CARD Signaling Adaptor Proteins/metabolism , Disease Models, Animal , Homocysteine/biosynthesis , Humans , Hyperhomocysteinemia/metabolism , Hyperhomocysteinemia/pathology , Inflammasomes/genetics , Inflammasomes/metabolism , Kidney Glomerulus/injuries , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Neuropeptides/genetics , Podocytes/metabolism , Podocytes/pathology , Proto-Oncogene Proteins c-vav/genetics , RNA, Small Interfering/genetics , Reactive Oxygen Species/metabolism , rac1 GTP-Binding Protein/genetics
16.
Curr Drug Targets ; 18(9): 1019-1029, 2017.
Article in English | MEDLINE | ID: mdl-27538510

ABSTRACT

BACKGROUND: The intracellular multiprotein complex termed the inflammasome functions as a platform of pro-inflammatory cytokine production such as IL-1ß and IL-18. Under certain conditions, however, the inflammasome produces non-canonical effects such as induction of cell death, pyroptosis and cell metabolism alterations. OBJECTIVE: In mammalian cells, several types of inflammasomes were identified, but the most widely studied one is the inflammasome containing NOD-like receptor with pyrin domain 3 (NLRP3), which has recently been reported as a central pathogenic mechanism of chronic degenerative diseases. Many activators or risk factors exert their actions through the activation of the NLRP3 inflammasome to produce a variety of functional changes in different cells including inflammatory, metabolic or survival responses. Several molecular signaling pathways are shown to mediate the activation of the NLRP3 inflammasome, and they are related to the modifications in K+ efflux, increased lysosome leakage and activation of cathepsin B or enhanced reactive oxygen species (ROS) production. In the kidney, inflammation is believed to mediate or promote the progression of glomerular sclerotic pathologies resulting in end-stage renal disease (ESRD). NLRP3 inflammasome activation may turn on glomerular inflammation and other cell damages, contributing to the onset of glomerular injury and ESRD. This inflammasome activation not only occurs in immune cells, but also in residential cells such as endothelial cells and podocytes in the glomeruli. SUMMARY: This review briefly summarizes current evidence of NLRP3 inflammasome activation and related molecular mechanisms in renal glomeruli. The possible canonical and non-canonical effects of this inflammasome activation and its potential implication in the development of different glomerular diseases are highlighted.


Subject(s)
Glomerulonephritis/immunology , Inflammasomes/immunology , Chronic Disease , Glomerulonephritis/metabolism , Glomerulonephritis/therapy , Humans , Inflammasomes/metabolism , Kidney Glomerulus/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
17.
Front Biosci (Landmark Ed) ; 22(1): 96-116, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27814604

ABSTRACT

Sphingolipids are biologically active lipids ubiquitously produced in all vertebrate cells. Asides from structural components of cell membrane, sphingolipids also function as intracellular and extracellular mediators that regulate many important physiological cellular processes including cell survival, proliferation, apoptosis, differentiation, migration and immune processes. Recent studies have also indicated that disruption of sphingolipid metabolism is strongly associated with different diseases that exhibit diverse neurological and metabolic consequences. Here, we briefly summarize current evidence for understanding of sphingolipid pathways in obesity and associated complications. The regulation of sphingolipids and their enzymes may have a great impact in the development of novel therapeutic modalities for a variety of metabolic diseases.


Subject(s)
Obesity/metabolism , Sphingolipids/metabolism , Adipokines/biosynthesis , Adipose Tissue/metabolism , Animals , Atherosclerosis/etiology , Atherosclerosis/metabolism , Diabetes Mellitus/etiology , Diabetes Mellitus/metabolism , Energy Intake , Humans , Hypertension/etiology , Hypertension/metabolism , Inflammasomes/metabolism , Insulin Resistance , Obesity/complications , Obesity/etiology , Oxidative Stress , Renal Insufficiency, Chronic/etiology , Renal Insufficiency, Chronic/metabolism , Sphingolipids/antagonists & inhibitors
18.
Front Biosci (Landmark Ed) ; 22(4): 558-569, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27814632

ABSTRACT

Collagen deposition is a hallmark of atherosclerosis. Although compromised collagen I degradation has been implied in the pathogenesis of atherosclerosis, the molecular mechanisms are still unclear. Thus, we determined the role of CD38, an enzyme involved in cellular calcium modulation and autophagic flux, in the regulation of collagen I degradation in coronary arterial myocytes (CAMs).In primary cultured CAMs from CD38-/- mice, collagen I protein accumulation but not mRNA abundance was significantly increased compared with cells from CD38+/+ mice either under control or upon TGF-Beta stimulation. Pharmacological inhibition of the formation of autophagosomes with 3-methyladenine or of autophagolysosomes with a lysosomal functional blocker, bafilomycin A1, induced a similar increase in collagen protein levels, while inhibition of the proteasome by MG132 had no effects on collagen I accumulation. In addition, CD38-deficiency did not change the protein expression of matrix metalloprotein-9 (MMP-9) or tissue inhibitor of metalloproteinase-1 (TIMP-1) in CAMs. Confocal microscopy showed that collagen I deposition was mainly lied within lysosomes or autophagosomes in CD38-/- or TGF-Beta treated CAMs. Collagen I deposition increased when CAMs lack CD38 expression or if autophagy was blocked, which is associated with impaired autophagic degradation of collagen I. This CD38 regulation of autophagic flux may represent a novel mechanism for extracellular matrix (ECM) plasticity of coronary arteries upon atherogenic stimulation.


Subject(s)
ADP-ribosyl Cyclase 1/genetics , Collagen Type I/metabolism , Coronary Vessels/metabolism , Membrane Glycoproteins/genetics , Myocytes, Cardiac/metabolism , ADP-ribosyl Cyclase 1/deficiency , ADP-ribosyl Cyclase 1/metabolism , Animals , Autophagy , Cells, Cultured , Collagen Type I/genetics , Coronary Artery Disease/etiology , Coronary Artery Disease/metabolism , Coronary Artery Disease/pathology , Coronary Vessels/cytology , Diet, Western/adverse effects , Lysosomes/metabolism , Male , Matrix Metalloproteinase 9/metabolism , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/cytology , Phagosomes/metabolism , Proteasome Inhibitors/pharmacology , Proteolysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Tissue Inhibitor of Metalloproteinase-1/metabolism , Transforming Growth Factor beta/metabolism
19.
Oncotarget ; 7(26): 39316-39331, 2016 Jun 28.
Article in English | MEDLINE | ID: mdl-27322427

ABSTRACT

The major pathological changes during Schistosoma J. infection are characterized by granulomatous inflammation in the liver, a cellular immune response to schistosomal egg antigens. The molecular mechanisms initiating or promoting this schistosomal granulomatous inflammation remain poorly understood. In the present study, we first demonstrated that in mice infected with Schistosoma J. for 6 weeks exhibited increased levels of IL-1ß in liver, a major product of NLRP3 inflammasomes and collagen deposition around the eosinophilic granuloma with Schistosoma J. eggs, which was substantially attenuated by caspase-1 inhibitor, YVAD. This activation of the NLRP3 inflammasome occurred in hepatic stellate cells (HSCs), as shown by a marked increase in co-localization of IL-1ß with HSCs marker, desmin. Using isolated, cultured mouse HSCs, we further explored the mechanisms by which soluble egg antigen (SEA) from Schistosoma J. activates NLRP3 inflammasomes. SEA induced the formation and activation of NLRP3 inflammasomes, which was associated with both redox regulation and lysosomal dysfunction, but not with potassium channel activation. These results suggest that NLRP3 inflammasome activation in HSCs may serve as an early mechanism to turn on the inflammatory response and thereby instigate liver fibrosis during Schistosoma J infection.


Subject(s)
Hepatic Stellate Cells/parasitology , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Schistosomiasis/immunology , Animals , Antigens/chemistry , Carrier Proteins/immunology , Caspase 1/metabolism , Caspase Inhibitors/chemistry , Disease Models, Animal , Fibrosis/pathology , Hepatic Stellate Cells/metabolism , Inflammation , Interleukin-1beta/metabolism , Liver/microbiology , Liver Cirrhosis/pathology , Lysosomes/metabolism , Male , Mice , Mice, Inbred BALB C , Schistosoma
20.
Kidney Blood Press Res ; 41(2): 208-21, 2016.
Article in English | MEDLINE | ID: mdl-27010539

ABSTRACT

BACKGROUND/AIMS: Recent studies have indicated that local inflammatory mediators are importantly involved in the regulation of renal function. However, it remains unknown how such local inflammation is triggered intracellularly in the kidney. The present study was designed to characterize the inflammasome centered by Nlrp3 in the kidney and also test the effect of its activation in the renal medulla. METHODS AND RESULTS: By immunohistochemistry analysis, we found that inflammasome components, Nlrp3, Asc and caspase-1, were ubiquitously distributed in different kidney areas. The caspase-1 activity and IL-1ß production were particularly high in the renal outer medulla compared to other kidney regions. Further confocal microscopy and RT-PCR analysis showed that Nlrp3, Asc and caspase-1 were particularly enriched in the thick ascending limb of Henle's loop. In anesthetized mice, medullary infusion of Nlrp3 inflammasome activator, monosodium urate (MSU), induced significant decreases in sodium excretion and medullary blood flow without changes in mean arterial blood pressure and renal cortical blood flow. Caspase-1 inhibitor, Ac-YVAD-CMK and deletion of Nlrp3 or Asc gene abolished MSU-induced decreases in renal sodium excretion and MBF. CONCLUSION: Our results indicate that renal medullary Nlrp3 inflammasomes represent a new regulatory mechanism of renal MBF and sodium excretion which may not depend on classical inflammatory response.


Subject(s)
Kidney Medulla/blood supply , Kidney Medulla/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Animals , Blood Flow Velocity , Gene Deletion , Inflammasomes/genetics , Inflammasomes/metabolism , Kidney Medulla/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...