Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 511
Filter
Add more filters

Publication year range
1.
J Biol Chem ; 289(10): 6991-7002, 2014 Mar 07.
Article in English | MEDLINE | ID: mdl-24451378

ABSTRACT

Atrial natriuretic peptide (ANP) binds guanylyl cyclase-A/natriuretic peptide receptor-A (GC-A/NPRA) and produces the intracellular second messenger, cGMP, which regulates cardiovascular homeostasis. We sought to determine the function of histone deacetylases (HDACs) in regulating Npr1 (coding for GC-A/NPRA) gene transcription, using primary mouse mesangial cells treated with class-specific HDAC inhibitors (HDACi). Trichostatin A, a pan inhibitor, and mocetinostat (MGCD0103), a class I HDAC inhibitor, significantly enhanced Npr1 promoter activity (by 8- and 10-fold, respectively), mRNA levels (4- and 5.3-fold, respectively), and NPRA protein (2.7- and 3.5-fold, respectively). However, MC1568 (class II HDAC inhibitor) had no discernible effect. Overexpression of HDAC1 and HDAC2 significantly attenuated Npr1 promoter activity, whereas HDAC3 and HDAC8 had no effect. HDACi-treated cultured cells in vitro and intact animals in vivo showed significantly reduced binding of HDAC1 and -2 and increased accumulation of acetylated H3-K9/14 and H4-K12 at the Npr1 promoter. Deletional analyses of the Npr1 promoter along with ectopic overexpression and inhibition of Sp1 confirmed that HDACi-induced Npr1 gene transcription is accomplished by Sp1 activation. Furthermore, HDACi attenuated the interaction of Sp1 with HDAC1/2 and promoted Sp1 association with p300 and p300/cAMP-binding protein-associated factor; it also promoted the recruitment of p300 and p300/cAMP-binding protein-associated factor to the Npr1 promoter. Our results demonstrate that trichostatin A and MGCD0103 enhanced Npr1 gene expression through inhibition of HDAC1/2 and increased both acetylation of histones (H3-K9/14, H4-K12) and Sp1 by p300, and their recruitment to Npr1 promoter. Our findings define a novel epigenetic regulatory mechanism that governs Npr1 gene transcription.


Subject(s)
Epigenesis, Genetic , Histone Deacetylases/metabolism , Receptors, Atrial Natriuretic Factor/genetics , Transcription, Genetic/drug effects , Animals , Benzamides/pharmacology , Cell Line , Glomerular Mesangium/drug effects , Glomerular Mesangium/enzymology , Histone Acetyltransferases/metabolism , Histone Deacetylase Inhibitors/pharmacology , Histones/metabolism , Hydroxamic Acids/pharmacology , Mice , Promoter Regions, Genetic , Pyrimidines/pharmacology , Sp1 Transcription Factor/metabolism , p300-CBP Transcription Factors/metabolism
2.
Kidney Int ; 82(3): 292-303, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22475818

ABSTRACT

Angiotensin-converting enzyme 2 (ACE2) degrades angiotensin II to angiotensin-(1-7) and is expressed in podocytes. Here we overexpressed ACE2 in podocytes in experimental diabetic nephropathy using transgenic methods where a nephrin promoter drove the expression of human ACE2. Glomeruli from these mice had significantly increased mRNA, protein, and activity of ACE2 compared to wild-type mice. Male mice were treated with streptozotocin to induce diabetes. After 16 weeks, there was no significant difference in plasma glucose levels between wild-type and transgenic diabetic mice. Urinary albumin was significantly increased in wild-type diabetic mice at 4 weeks, whereas albuminuria in transgenic diabetic mice did not differ from wild-type nondiabetic mice. However, this effect was transient and by 16 weeks both transgenic and nontransgenic diabetic mice had similar rates of proteinuria. Compared to wild-type diabetic mice, transgenic diabetic mice had an attenuated increase in mesangial area, decreased glomerular area, and a blunted decrease in nephrin expression. Podocyte numbers decreased in wild-type diabetic mice at 16 weeks, but were unaffected in transgenic diabetic mice. At 8 weeks, kidney cortical expression of transforming growth factor-ß1 was significantly inhibited in transgenic diabetic mice as compared to wild-type diabetic mice. Thus, the podocyte-specific overexpression of human ACE2 transiently attenuates the development of diabetic nephropathy.


Subject(s)
Diabetic Nephropathies/enzymology , Diabetic Nephropathies/genetics , Peptidyl-Dipeptidase A/genetics , Peptidyl-Dipeptidase A/metabolism , Podocytes/enzymology , Angiotensin-Converting Enzyme 2 , Animals , Blood Pressure , Diabetic Nephropathies/pathology , Diabetic Nephropathies/physiopathology , Glomerular Filtration Rate , Glomerular Mesangium/enzymology , Glomerular Mesangium/pathology , Humans , Male , Mice , Mice, Transgenic , Podocytes/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transforming Growth Factor beta1/metabolism , Up-Regulation
3.
J Exp Med ; 186(2): 189-98, 1997 Jul 21.
Article in English | MEDLINE | ID: mdl-9221748

ABSTRACT

Endogenously released or exogenously administered glucocorticosteroids are relevant hormones for controlling inflammation. Only 11beta-hydroxy glucocorticosteroids, but not 11-keto glucocorticosteroids, activate glucocorticoid receptors. Since we found that glomerular mesangial cells (GMC) express 11beta-hydroxysteroid dehydrogenase 1 (11beta-OHSD1), which interconverts 11-keto glucocorticosteroids into 11beta-hydroxy glucocorticosteroids (cortisone/cortisol shuttle), we explored whether 11beta-OHSD1 determines the antiinflammatory effect of glucocorticosteroids. GMC exposed to interleukin (IL)-1beta or tumor necrosis factor alpha (TNF-alpha) release group II phospholipase A2 (PLA2), a key enzyme producing inflammatory mediators. 11beta-hydroxy glucocorticosteroids inhibited cytokine-induced transcription and release of PLA2 through a glucocorticoid receptor-dependent mechanism. This inhibition was enhanced by inhibiting 11beta-OHSD1. Interestingly, 11-keto glucocorticosteroids decreased cytokine-induced PLA2 release as well, a finding abrogated by inhibiting 11beta-OHSD1. Stimulating GMC with IL-1beta or TNF-alpha increased expression and reductase activity of 11beta-OHSD1. Similarly, this IL-1beta- and TNF-alpha-induced formation of active 11beta-hydroxy glucocorticosteroids from inert 11-keto glucocorticosteroids by the 11beta-OHSD1 was shown in the Kiki cell line that expresses the stably transfected bacterial beta-galactosidase gene under the control of a glucocorticosteroids response element. Thus, we conclude that 11beta-OHSD1 controls access of 11beta-hydroxy glucocorticosteroids and 11-keto glucocorticosteroids to glucocorticoid receptors and thus determines the anti-inflammatory effect of glucocorticosteroids. IL-1beta and TNF-alpha upregulate specifically the reductase activity of 11beta-OHSD1 and counterbalance by that mechanism their own proinflammatory effect.


Subject(s)
Cortisone/metabolism , Glomerular Mesangium/enzymology , Hydrocortisone/metabolism , Hydroxysteroid Dehydrogenases/metabolism , Interleukin-1/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , 11-beta-Hydroxysteroid Dehydrogenases , Animals , Phospholipases A/metabolism , Phospholipases A2 , Rats , Rats, Sprague-Dawley
4.
J Exp Med ; 172(6): 1843-52, 1990 Dec 01.
Article in English | MEDLINE | ID: mdl-1979590

ABSTRACT

Endothelium-derived nitric oxide (NO) causes vasodilatation by activating soluble guanylate cyclase, and glomerular mesangial cells respond to NO with elevations of intracellular guanosine 3',5'-cyclic monophosphate (cGMP). We explored whether mesangial cells can be stimulated to produce NO and whether NO modulates mesangial cell function in an autocrine or paracrine fashion. Tumor necrosis factor alpha (TNF-alpha) raised mesangial cell cGMP levels in a time- and concentration-dependent manner (threshold dose 1 ng/ml, IC50 13.8 ng/ml, maximal response 100 ng/ml). TNF-alpha-induced increases in mesangial cGMP content were evident at 8 h and maximal at 18-24 h. The TNF-alpha-induced stimulation of mesangial cell cGMP production was abrogated by actinomycin D or cycloheximide suggesting dependence on new RNA or protein synthesis. Hemoglobin and methylene blue, both known to inhibit NO action, dramatically reduced TNF-alpha-induced mesangial cell cGMP production. Superoxide dismutase, known to potentiate NO action, augmented the TNF-alpha-induced effect. Ng-monomethyl-L-arginine (L-NMMA) decreased cGMP levels in TNF-alpha-treated, but not vehicle-treated mesangial cells in a concentration-dependent manner (IC50 53 microM). L-arginine had no effect on cGMP levels in control or TNF-alpha-treated mesangial cells but reversed L-NMMA-induced inhibition. Interleukin 1 beta and lipopolysaccharide (LPS), but not interferon gamma, also increased mesangial cell cGMP content. Transforming growth factor beta 1 blunted the mesangial cell response to TNF-alpha. TNF-alpha-induced L-arginine-dependent increases in cGMP were also evident in bovine renal artery vascular smooth muscle cells, COS-1 cells, and 1502 human fibroblasts. These findings suggest that TNF-alpha induces expression in mesangial cell of an enzyme(s) involved in the formation of L-arginine-derived NO. Moreover, the data indicate that NO acts in an autocrine and paracrine fashion to activate mesangial cell soluble guanylate cyclase. Cytokine-induced formation of NO in mesangial and vascular smooth muscle cells may be implicated in the pathogenesis of septic shock.


Subject(s)
Arginine/metabolism , Glomerular Mesangium/metabolism , Guanylate Cyclase/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Animals , Arginine/analogs & derivatives , Arginine/pharmacology , Cattle , Cells, Cultured , Cyclic GMP/metabolism , Cycloheximide/pharmacology , Dactinomycin/pharmacology , Enzyme Activation , Glomerular Mesangium/drug effects , Glomerular Mesangium/enzymology , Humans , Interferon-gamma/pharmacology , Interleukin-1/pharmacology , Kinetics , Nitrates/metabolism , Nitrites/metabolism , Recombinant Proteins/pharmacology , omega-N-Methylarginine
5.
Nephrology (Carlton) ; 15(5): 584-6, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20649880

ABSTRACT

Immunoglobulin A nephropathy (IgAN), characterized by predominant or exclusive deposition of IgA1 in glomerular mesangium, is the most common primary glomerulonephritis worldwide. At present, the treatment is always limited due to the incomplete understanding of the pathogenesis of IgAN. Mesangial deposited IgA1 is the common final pathway leading to glomerulonephritis and renal injury. IgA1 protease, a proteolytic enzyme with strict substrate specificity for human IgA1, may be an effective therapeutic candidate for IgAN by removing the mesangial deposited IgA1.


Subject(s)
Glomerular Mesangium/drug effects , Glomerulonephritis, IGA/drug therapy , Immunoglobulin A/metabolism , Protease Inhibitors/therapeutic use , Serine Endopeptidases/metabolism , Glomerular Mesangium/enzymology , Glomerular Mesangium/immunology , Glomerulonephritis, IGA/enzymology , Glomerulonephritis, IGA/immunology , Humans
6.
Ren Fail ; 32(2): 254-8, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20199188

ABSTRACT

Mesangial cell (MC) proliferation is a hallmark of many progressive renal diseases. Heme oxygenase-1 (HO-1) has been shown to have an anti-proliferative effect on vascular smooth muscle cells. In the present study, we evaluated the role of HO-1 on MC proliferation and the involved molecular mechanism. Both epidermal growth factor (EGF) and hepatocyte growth factor (HGF) not only enhanced mesangial cell HO-1 expression but also stimulated proliferation of MCs. Interestingly, inhibition of HO-1 induction (by zinc protoporphyrin, ZnP) was associated with an accelerated mitogenic response to EGF and HGF in MCs. Induction of HO-1 was associated with enhanced mesangial cell p21 expression. On the other hand, hemoglobin and ZnP inhibited mesangial cell p21 expression. It appears that the effect of HO-1 on MC growth may be mediated through upregulation of p21 expression.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/metabolism , Glomerular Mesangium/cytology , Glomerular Mesangium/enzymology , Heme Oxygenase-1/pharmacology , Analysis of Variance , Animals , Apoptosis/drug effects , Blotting, Western , Cell Proliferation/drug effects , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21/drug effects , Glomerular Mesangium/drug effects , Hepatocyte Growth Factor/pharmacology , Immunohistochemistry , Mice , Up-Regulation
7.
Clin Invest Med ; 32(1): E20-7, 2009 Feb 01.
Article in English | MEDLINE | ID: mdl-19178875

ABSTRACT

PURPOSE: To investigate the influence of IgA1 isolated from IgA nephropathy (IgAN) patients on integrin-linked kinase (ILK) synthesis and adhesive capacity of podocytes through indirect pathways. METHODS: IgA1 was isolated from healthy control or IgAN patients' sera using jacalin affinity chromatography and S-200 chromatography. Podocytes were treated with medium from mesangial cells incubated with aggregated IgA1 (aIgA1, 100 microg/ml), in the presence or absence of valsartan (10(-5)M) or neutralizing antibodies of tumor necrosis factor-alpha (TNF-alpha, 50 ng/ml). Adhesive capacity of podocytes was assessed by cell counting manually and hexosaminidase assay. Real-time PCR and western blotting were used to detect the expression of ILK. RESULTS: Medium from mesangial cells incubated with aIgA1 from IgAN patients reduced podocyte adhesion to collagen compared with medium from mesangial cells incubated with control medium(RPMI-1640 with 0.5% FBS) (35.0+/-4.8% vs. 60.0+/-2.0%; P < 0.05). While medium from mesangial cells incubated with aIgA1 from IgAN patients upregulated ILK expression in podocytes at mRNA and protein levels compared with medium from mesangial cells without aIgA1 incubated (1.6-fold and 1.38-fold higher than control, respectively, P < 0.05). Defects in podocyte adhesion and up-regulation of ILK synthesis induced by medium from mesangial cells incubated with aIgA1 from IgAN patients can be partially reversed by the pre-treatment for 1 hour with valsartan(P < 0.05), while pre-treatment with neutralizing antibodies of TNF-alpha produced no protective effect on podocytes (P > 0.05). CONCLUSION: Serum IgA1 from IgAN patients may inhibit adhesive capacity and up-regulate ILK synthesis in podocytes through indirect pathways.


Subject(s)
Glomerulonephritis, IGA/blood , Immunoglobulin A/blood , Podocytes/cytology , Protein Serine-Threonine Kinases/metabolism , Up-Regulation , Angiotensin II/metabolism , Animals , Base Sequence , Blotting, Western , Cell Adhesion , Culture Media , DNA Primers , Enzyme-Linked Immunosorbent Assay , Glomerular Mesangium/enzymology , Glomerular Mesangium/metabolism , Humans , Mice , Protein Serine-Threonine Kinases/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/metabolism
8.
J Am Soc Nephrol ; 19(2): 298-309, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18184857

ABSTRACT

Several recent reports support the hypothesis that aldosterone contributes to the progression of renal injury. Mineralocorticoids increase the expression of serum- and glucocorticoid-inducible protein kinase 1 (SGK1), which is upregulated in several fibrotic diseases. It was hypothesized that SGK1 may mediate the effects of aldosterone on glomerular fibrosis and inflammation. In primary cultures of rat mesangial cells, aldosterone stimulated the expression, phosphorylation, and kinase activity of SGK1, as well as SGK1-dependent NF-kappaB activity. Furthermore, aldosterone augmented the promoter activity and protein expression of intercellular adhesion molecule-1 (ICAM-1), which modulates the inflammatory response, and the profibrotic cytokine connective tissue growth factor (CTGF) in an SGK1- and NF-kappaB-dependent manner. Similar to the in vitro results, uninephrectomized rats that were treated with aldosterone demonstrated increased glomerular expression of SGK1, ICAM-1, and CTGF proteins than untreated rats; these changes were accompanied by hypertension, glomerulosclerosis, and inflammation. In conclusion, these findings suggest that aldosterone stimulates ICAM-1 and CTGF transcription via the activation of SGK1 and NF-kappaB, effects that may contribute to the progression of aldosterone-induced mesangial fibrosis and inflammation.


Subject(s)
Aldosterone/metabolism , Immediate-Early Proteins/metabolism , Mesangial Cells/enzymology , Mesangial Cells/pathology , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/physiology , Aldosterone/pharmacology , Animals , Antibodies/pharmacology , Antibody Specificity , Cells, Cultured , Connective Tissue Growth Factor , Fibrosis , Glomerular Mesangium/enzymology , Glomerular Mesangium/pathology , I-kappa B Kinase/metabolism , Immediate-Early Proteins/genetics , Immediate-Early Proteins/immunology , Intercellular Adhesion Molecule-1/genetics , Intercellular Signaling Peptides and Proteins/genetics , MAP Kinase Kinase 1/metabolism , Male , Mesangial Cells/drug effects , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , RNA, Small Interfering , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Transcription, Genetic/drug effects , Transcription, Genetic/physiology
9.
Food Chem Toxicol ; 122: 87-94, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30296514

ABSTRACT

Paeonol is a natural flavonoid isolated from Moutan Cortex, which has been found to exhibit antioxidant, anti-apoptotic, anti-aging and anti-inflammatory bioactivities. Herein, we investigated the nephroprotective efficacy of paeonol against Pb-induced toxicity and elucidated the potential mechanisms. The results revealed that paeonol significantly ameliorated renal dysfunction and histology changes of Pb-treated mice. Paeonol inhibited oxidative stress and increased activities of antioxidant enzyme in the kidneys of Pb-treated mice. Paeonol decreased the nuclear factor-κB activation and over-production of inflammatory cytokines tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6). Paeonol suppressed endoplasmic reticulum (ER) stress in kidneys of in the Pb treatment group and primary kidney mesangial cells. Moreover, paeonol increased the denosine 5'-monophosphate-activated protein kinase (AMPK) phosphorylation and decreased the activations of glycogen synthase kinase-3 (GSK-3), protein kinase RNA-like ER kinase (PERK), inositol-requiring protein-1 (IRE1), c-jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase (MAPK). These results were further confirmed in primary kidney mesangial cells. Taken together, these findings indicate that paeonol could protect kidney form Pb-induced injury by inhibiting oxidative stress, ER stress and inflammation via the AMPK and GSK-3 pathway. Paeonol might be a potential therapeutic agent to inhibit ER stress-associated inflammation in lead-stimulated kidneys.


Subject(s)
Acetophenones/pharmacology , Adenylate Kinase/metabolism , Endoplasmic Reticulum Stress/drug effects , Glomerular Mesangium/drug effects , Glomerular Mesangium/enzymology , Lead/toxicity , Animals , Cells, Cultured , Drugs, Chinese Herbal/chemistry , Enzyme Activation , Glomerular Mesangium/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Inflammation/prevention & control , Inflammation Mediators/metabolism , Interleukin-6/biosynthesis , MAP Kinase Kinase 4/metabolism , Male , Membrane Proteins/metabolism , Mice, Inbred ICR , NF-kappa B/metabolism , Oxidative Stress/drug effects , Paeonia/chemistry , Protein Serine-Threonine Kinases/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , eIF-2 Kinase/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
10.
Diabetes ; 55(7): 2115-25, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16804083

ABSTRACT

Diabetic nephropathy is characterized early in its course by glomerular hypertrophy and, importantly, mesangial hypertrophy, which correlate with eventual glomerulosclerosis. The mechanism of hypertrophy, however, is not known. Gene disruption of the tumor suppressor PTEN, a negative regulator of the phosphatidylinositol 3-kinase/Akt pathway, in fruit flies and mice demonstrated its role in size control in a cell-specific manner. Here, we investigated the mechanism of mesangial hypertrophy in response to high extracellular glucose. We link early renal hypertrophy with significant reduction in PTEN expression in the streptozotocin-induced diabetic kidney cortex and glomeruli, concomitant with activation of Akt. Similarly, exposure of mesangial cells to high concentrations of glucose also decreased PTEN expression and its phosphatase activity, resulting in increased Akt activity. Expression of PTEN inhibited high-glucose-induced mesangial cell hypertrophy, and expression of dominant-negative PTEN was sufficient to induce hypertrophy. In diabetic nephropathy, the hypertrophic effect of hyperglycemia is thought to be mediated by transforming growth factor-beta (TGF-beta). TGF-beta significantly reduced PTEN expression in mesangial cells, with a reduction in its phosphatase activity and an increase in Akt activation. PTEN and dominant-negative Akt attenuated TGF-beta-induced hypertrophy of mesangial cells. Finally, we show that inhibition of TGF-beta signal transduction blocks the effect of high glucose on PTEN downregulation. These data identify a novel mechanism placing PTEN as a key regulator of diabetic mesangial hypertrophy involving TGF-beta signaling.


Subject(s)
Blood Glucose/metabolism , Gene Expression Regulation, Enzymologic , Glomerular Mesangium/pathology , Kidney Cortex/pathology , PTEN Phosphohydrolase/genetics , Animals , Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Experimental/genetics , Glomerular Mesangium/enzymology , Hyperglycemia , Hypertrophy , Kidney Cortex/enzymology , Rats , Signal Transduction , Transforming Growth Factor beta/physiology
11.
J Clin Invest ; 100(6): 1394-9, 1997 Sep 15.
Article in English | MEDLINE | ID: mdl-9294104

ABSTRACT

Automatic control over exogenous gene expression in response to the activity of disease is a crucial hurdle for gene transfer-based therapies. Towards achieving this goal, we created a "cytosensor" that perceives local inflammatory states and subsequently regulates foreign gene expression. alpha-Smooth muscle actin is known to be expressed in glomerular mesangial cells exclusively in pathologic situations. CArG box element, the crucial regulatory sequence of the alpha-smooth muscle actin promoter, was used as a sensor for glomerular inflammation. Rat mesangial cells were stably transfected with an expression plasmid that introduces a beta-galactosidase gene under the control of CArG box elements. In vitro, the established cells expressed beta-galactosidase exclusively after stimulation with serum. To examine whether the cells are able to automatically control transgene activity in vivo, serum-stimulated or unstimulated cells were transferred into normal rat glomeruli or glomeruli subjected to anti-Thy 1 glomerulonephritis. When stimulated cells were transferred into the normal glomeruli, beta-galactosidase expression was switched off in vivo within 3 d. In contrast, when unstimulated cells were transferred into the nephritic glomeruli, transgene expression was substantially induced. These data indicate the feasibility of using the CArG box element as a molecular sensor for glomerular injury. In the context of advanced forms of gene therapy, this approach provides a novel concept for automatic regulation of local transgene expression where the transgene is required to be activated during inflammation and deactivated when the inflammation has subsided.


Subject(s)
Biosensing Techniques , Gene Expression Regulation , Glomerular Mesangium/enzymology , Transgenes/physiology , Actins/genetics , Animals , Cells, Cultured , Culture Media, Conditioned/pharmacology , Dose-Response Relationship, Drug , Glomerulonephritis/chemically induced , Glomerulonephritis/metabolism , Rats , Regulatory Sequences, Nucleic Acid/genetics , Time Factors , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
12.
J Clin Invest ; 92(5): 2516-23, 1993 Nov.
Article in English | MEDLINE | ID: mdl-8227364

ABSTRACT

Potent pro-inflammatory cytokines, such as interleukin 1 (IL-1) or tumor necrosis factor (TNF) alpha have been found to increase group II phospholipase A2 (PLA2) synthesis and secretion by mesangial cells. In all cases 85-90% of the enzyme is secreted from the cells and a parallel increase in prostaglandin (PG)E2 synthesis is observed. We report here that co-incubation with a monoclonal antibody that specifically binds and neutralizes rat group II PLA2 attenuates IL-1 beta and TNF alpha-stimulated PGE2 production by 45% and 52%, respectively. CGP43182, a specific inhibitor of group II PLA2, potently blocks mesangial cell group II PLA2 in vitro with a half-maximal inhibitory concentration (IC50) of 1.5 microM, while only slightly affecting mesangial cell high molecular weight PLA2. CGP 43182 markedly attenuates IL-1 beta- and TNF alpha-stimulated PGE2 synthesis in intact mesangial cells with IC50's of 1.3 and 1.0 microM, respectively. PLA2 secreted from cytokine-stimulated mesangial cells was purified to homogeneity. Addition of the purified enzyme to unstimulated mesangial cells causes a marked release of arachidonic acid and a subsequent increased synthesis of PGE2. Moreover, addition of purified PLA2 to a cloned rat glomerular epithelial cell line and cultured bovine glomerular endothelial cells augmented both arachidonic acid release and PGE2 synthesis, with the endothelial cells being especially sensitive. Thus, cytokine-triggered synthesis and secretion of group II PLA2 by mesangial cells contributes, at least in part, to the observed synthesis of PGE2 that occurs in parallel to the enzyme secretion. Furthermore, extracellular PLA2 secreted by mesangial cells is able to stimulate arachidonic acid release and PGE2 synthesis by the adjacent endothelial and epithelial cells. These data suggest that expression and secretion of group II PLA2 triggered by pro-inflammatory cytokines may crucially participate in the pathogenesis of inflammatory processes within the glomerulus.


Subject(s)
Cytokines/pharmacology , Dinoprostone/biosynthesis , Glomerular Mesangium/drug effects , Phospholipases A/metabolism , Animals , Calcimycin/pharmacology , Cells, Cultured , Chlorobenzenes/pharmacology , Dose-Response Relationship, Drug , Female , Glomerular Mesangium/enzymology , Interleukin-1/pharmacology , Isoenzymes/metabolism , Male , Neutralization Tests , Phospholipases A/classification , Phospholipases A/immunology , Phospholipases A2 , Rats , Rats, Sprague-Dawley , Rats, Wistar , Tumor Necrosis Factor-alpha/pharmacology
13.
J Clin Invest ; 96(1): 401-10, 1995 Jul.
Article in English | MEDLINE | ID: mdl-7615811

ABSTRACT

We studied interactions between the mitogen-activated protein kinase (MAPK) signalling pathway and cAMP-protein kinase (PKA) signaling pathway in regulation of mitogenesis of mesangial cells (MC) determined by [3H]thymidine incorporation, with or without added EGF. Forskolin or dibutyryl cAMP strongly (by 60-70%) inhibited [3H]thymidine incorporation into MC. Cilostamide, lixazinone or cilostazol selective inhibitors of cAMP-phosphodiesterase (PDE) isozyme PDE-III, inhibited mitogenesis to similar extent as forskolin and DBcAMP and activated in situ PKA, but without detectable increase in cAMP levels. Cilostamide and cilostazol were more than three times more effective at inhibiting mesangial mitogenesis than rolipram and denbufylline, inhibitors of isozyme PDE-IV, even though PDE-IV was two times more abundant in MC than was PDE-III. On the other hand, when incubated with forskolin, rolipram-enhanced cAMP accumulation was far greater (10-100x) than with cilostamide. EGF increased MAPK activity (+300%); PDE isozyme inhibitors which suppressed mitogenesis also inhibited MAPK. PDE isozyme inhibitors also suppressed PDGF-stimulated MC proliferation. We conclude that cAMP inhibits the mitogen-dependent MAPK-signaling pathway probably by decreasing the activity of Raf-1 due to PKA-catalyzed phosphorylation. Further, we surmise that minor increase in the cAMP pool metabolized by PDE-III is intimately related to regulation of mesangial proliferation. Thus, PDE isozyme inhibitors have the potential to suppress MC proliferation by a focused effect upon signaling pathways.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/physiology , Glomerular Mesangium/drug effects , Isoenzymes/antagonists & inhibitors , Animals , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cells, Cultured , Epidermal Growth Factor/pharmacology , Glomerular Mesangium/cytology , Glomerular Mesangium/enzymology , Male , Platelet-Derived Growth Factor/pharmacology , Pyrrolidinones/pharmacology , Quinolones/pharmacology , Rats , Rats, Sprague-Dawley , Rolipram , Thymidine/metabolism
14.
J Clin Invest ; 92(5): 2524-32, 1993 Nov.
Article in English | MEDLINE | ID: mdl-8227365

ABSTRACT

IL-1 stimulates mesangial cells to synthesize specific proteins, including a non-pancreatic (Type II) phospholipase A2 (PLA2). We have studied the regulation of PLA2 by proinflammatory mediators, implicated in the pathogenesis of glomerulonephritis, and have assessed whether the activation of second messenger systems modulates or mimics PLA2 gene expression by cytokines. IL-1 alpha and beta, TNF alpha, and LPS, but not serum, IL-2, or PDGF, potently induce PLA2 mRNA, and enzyme expression. IL-1-stimulated mesangial cells express a 1.0 kB PLA2 mRNA transcript that is induced in a dose- and time-dependent manner. IL-1-stimulated increases in steady-state PLA2 mRNA abundance result from a moderate increase in PLA2 transcription rate that is amplified by the prolonged persistence of the transcript. Forskolin and dibutyryl cAMP potentiate IL-1-induced PLA2 mRNA and enzyme expression, but have no effect in the absence of cytokine. 12-tetradecanoyl phorbol 13-acetate, sn-1, 2-dioctanoyl glycerol or 1-oleoyl-2-acetyl-sn-glycerol fail to induce PLA2 expression or to alter the effect of IL-1 when coincubated with the cytokine. In contrast, serum deprivation for 24 h specifically enhances IL-1-stimulated PLA2. Genistein potentiates PLA2 mRNA expression in cells exposed to both IL-1 and serum. The inhibitory effect of serum on IL-1-induced PLA2 mRNA abundance is reproduced by PDGF but not dexamethasone. These data demonstrate that the signaling pathways directly engaged by IL-1 to induce PLA2 expression in mesangial cells interact with several second messenger systems in a cell-specific manner. We speculate that IL-1 induces specialized changes in mesangial cell structure and function through direct activation of a transcription factor(s), that result in induction of a specific gene set.


Subject(s)
Gene Expression Regulation, Enzymologic , Glomerular Mesangium/enzymology , Phospholipases A/biosynthesis , RNA, Messenger/biosynthesis , Animals , Bucladesine/pharmacology , Cell Nucleus/metabolism , Colforsin/pharmacology , Diglycerides/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Glomerular Mesangium/drug effects , Interleukin-1/pharmacology , Interleukin-2/pharmacology , Lipopolysaccharides/pharmacology , Phorbol Esters/pharmacology , Phosphatidic Acids/pharmacology , Phospholipases A/classification , Phospholipases A/genetics , Phospholipases A/metabolism , Phospholipases A2 , Platelet-Derived Growth Factor/pharmacology , Rats , Signal Transduction , Tissue Distribution , Transcription, Genetic
15.
J Clin Invest ; 92(6): 2889-96, 1993 Dec.
Article in English | MEDLINE | ID: mdl-8254044

ABSTRACT

Changes in glomerular eicosanoid production have been implicated in the development of diabetes-induced glomerular hyperfiltration and glomerular mesangial cells (GMC) are major eicosanoid-producing cells within the glomerulus. However, the mechanism for the effect of diabetes mellitus on glomerular mesangial eicosanoid production is unknown. The present study therefore examined whether elevated glucose concentrations activate protein kinase C (PKC) in GMC and whether this PKC activation mediates an effect of elevated glucose concentrations to increase the release of arachidonic acid and eicosanoid production by GMC. The percentage of [3H]arachidonic acid release per 30 min by preloaded GMC monolayers was significantly increased after 3-h exposure to high glucose (20 mM) medium (177% vs control medium) and this increase was sustained after 24-h exposure to high glucose concentrations. 3-h and 24-h exposure to high glucose medium also increased PGE2, 6-keto-PGF1 alpha, and thromboxane (TXB2) production by GMC. High glucose medium (20 mM) increased PKC activity in GMC at 3 and 24 h (168% vs control). In contrast, osmotic control media containing either L-glucose or mannitol did not increase arachidonic acid release, eicosanoid production, or PKC activity in GMC. Inhibiting glucose-induced PKC activation with either H-7 (50 microM) or staurosporine (1 microM) prevented glucose-induced increases in arachidonic acid release and eicosanoid production by GMC. These data demonstrate that elevated extracellular glucose concentrations directly increase the release of endogenous arachidonic acid and eicosanoids by GMC via mechanisms dependent on glucose-induced PKC activation.


Subject(s)
Arachidonic Acid/metabolism , Eicosanoids/metabolism , Glomerular Mesangium/metabolism , Glucose/pharmacology , Prostaglandins/biosynthesis , Protein Kinase C/metabolism , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine , Alkaloids/pharmacology , Amino Acid Sequence , Animals , Cells, Cultured , Culture Media , Glomerular Mesangium/drug effects , Glomerular Mesangium/enzymology , Isoquinolines/pharmacology , Kinetics , Mannitol/pharmacology , Molecular Sequence Data , Oligopeptides/metabolism , Phospholipases A/pharmacology , Piperazines/pharmacology , Protein Kinase C/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Staurosporine , Substrate Specificity , Thromboxane B2/metabolism , Tritium
16.
J Clin Invest ; 98(2): 365-71, 1996 Jul 15.
Article in English | MEDLINE | ID: mdl-8755646

ABSTRACT

Maintenance of renal function in liver cirrhosis requires increased synthesis of arachidonic acid derived prostaglandin metabolites. Arachidonate metabolites have been reported to be involved in modulation of liver damage. The purpose of the present study was to establish whether the first enzyme of the prostaglandin cascade synthesis, the phospholipase A2(PLA2) is altered in liver cirrhosis induced by bile duct excision. The mRNA of PLA2(group I and II) and annexin-I a presumptive inhibitor of PLA2 enzyme was measured by PCR using glyceraldehyde 3-phosphate dehydrogenase (GAPDH) as an internal standard. The mean mRNA ratio of group II PLA2/GAPDH was increased in liver tissue by 126% (P < 0.001) and in kidney tissue by 263% (P < 0.006) following induction of liver cirrhosis. The increase in group II PLA2 mRNA in cirrhotic animals was reflected by an increase in PLA2 protein and enzyme activity in both liver and kidney tissues. Since the mRNA of group I PLA2 was not detectable and Group IV PLA2 activity measured in liver and kidney tissue samples was very low and not changed following induction of cirrhosis, it is likely that the major PLA2 activity measured in liver and kidney corresponds to group II PLA2 enzyme. The mean mRNA ratio of annexin-I/GAPDH was increased in liver tissue by 115% (P < 0.05) but unchanged in kidney tissue following induction of cirrhosis. The protein content of annexin-I and -V were not affected by bile duct excision in liver and kidney tissue indicating that upregulation of group II PLA2 activity was not due to downregulation of annexin-I or -V. Group II PLA2 activity of glomerular mesangial cells stimulated by interleukin-1 beta was enhanced by bile juice and various bile salts. In conclusion, activity of group II PLA2 is upregulated partly due to enhanced transcription and translation in cirrhosis and is furthermore augmented by elevated levels of bile salts.


Subject(s)
Glomerular Mesangium/enzymology , Kidney/enzymology , Liver Cirrhosis, Experimental/enzymology , Liver/enzymology , Phospholipases A/biosynthesis , Animals , Annexin A1/biosynthesis , Annexin A1/isolation & purification , Annexin A5/biosynthesis , Annexin A5/isolation & purification , Antibodies, Monoclonal , Bile/metabolism , Bile Acids and Salts/pharmacology , Blotting, Western , Cells, Cultured , Enzyme Induction , Glomerular Mesangium/drug effects , Glyceraldehyde-3-Phosphate Dehydrogenases/biosynthesis , Lung/enzymology , Male , Organ Specificity , Phospholipases A/analysis , Phospholipases A2 , Polymerase Chain Reaction , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Transcription, Genetic
17.
Mol Cell Biol ; 16(10): 5915-23, 1996 Oct.
Article in English | MEDLINE | ID: mdl-8816505

ABSTRACT

Endothelin-1 (ET-1) triggers poorly understood nuclear signaling cascades that control gene expression, cell growth, and differentiation. To better understand how ET-1 regulates gene expression, we asked whether voltage-insensitive Ca2+ channels and Ca2+/calmodulin-dependent protein kinases (CaMKs) propagate signals from ET-1 receptors to the c-fos promoter in mesangial cells. Ca2+ influx through voltage-insensitive Ca2+ channels, one of the earliest postreceptor events in ET-1 signaling, mediated induction of c-fos mRNA and activation of the c-fos promoter by ET-1. A CaMK inhibitor (KN-93) blocked activation of the c-fos promoter by ET-1. Ectopic expression of CaMKII potentiated stimulation by ET-1, providing further evidence that CaMKs contribute to c-fos promoter activation by ET-1. The c-fos serum response element was necessary but not sufficient for CaMKII to activate the c-fos promoter. Activation of the c-fos promoter by ET-1 and CaMKII also required the FAP cis element, an AP-1-like sequence adjacent to the serum response element. Thus, voltage-insensitive Ca2+ channels and CaMKs apparently propagate ET-1 signals to the c-fos promoter that require multiple, interdependent cis elements. Moreover, these experiments suggest an important role for voltage-insensitive Ca2+ channels in nuclear signal transduction in nonexcitable cells.


Subject(s)
Calcium Channels/physiology , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Endothelin-1/pharmacology , Genes, fos , Glomerular Mesangium/physiology , Promoter Regions, Genetic , Receptors, Endothelin/physiology , Signal Transduction , Animals , Base Sequence , Benzylamines/pharmacology , Calcium/metabolism , Calcium Channels/drug effects , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Cell Nucleus/physiology , Cells, Cultured , DNA Primers , Enzyme Inhibitors/pharmacology , Genes, Reporter , Glomerular Mesangium/enzymology , Kinetics , Luciferases/biosynthesis , Male , Mutagenesis, Site-Directed , Point Mutation , Polymerase Chain Reaction , Proto-Oncogene Proteins c-fos/biosynthesis , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Receptor, Endothelin A , Receptors, Endothelin/biosynthesis , Recombinant Proteins/metabolism , Sulfonamides/pharmacology , Transcription, Genetic , Transfection
18.
Int J Biochem Cell Biol ; 38(1): 12-6, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16140560

ABSTRACT

Lipoprotein lipase expressed by the vasculature plays a key role in atherogenesis by enhancing the binding and uptake of lipoproteins and, thereby, leading to the formation of lipid-loaded foam cells. Hyperlipidemia also accelerates the progression of glomerular diseases and addition of exogenous lipoprotein lipase to mesangial cells has been shown to lead to an enhanced binding of lipoproteins to these cells. Despite such potential importance, the expression of endogenous lipoprotein lipase by cells of the glomeruli has, as yet, not been investigated. We show here for the first time that mesangial cells, but not epithelial cells, express lipoprotein lipase. The minimal lipoprotein lipase gene promoter was active in mesangial cells and inhibited by interferon-gamma, which is known to suppress its expression.


Subject(s)
Gene Expression Regulation, Enzymologic/physiology , Glomerular Mesangium/enzymology , Lipoprotein Lipase/biosynthesis , Promoter Regions, Genetic/physiology , Animals , Antineoplastic Agents/pharmacology , Atherosclerosis/enzymology , Atherosclerosis/pathology , Cell Line , Foam Cells/enzymology , Foam Cells/pathology , Gene Expression Regulation, Enzymologic/drug effects , Glomerular Mesangium/cytology , Glomerulonephritis/enzymology , Glomerulonephritis/pathology , Hyperlipidemias/enzymology , Hyperlipidemias/pathology , Interferon-gamma/pharmacology , Mice
19.
Biochem Pharmacol ; 71(12): 1711-9, 2006 Jun 14.
Article in English | MEDLINE | ID: mdl-16643859

ABSTRACT

We have recently shown that the pancreatic hormone glucagon-induced phosphorylation of mitogen-activated protein (MAP) kinase ERK 1/2 as well as growth and proliferation of rat glomerular mesangial cells (MCs) via activation of cAMP-dependent protein kinase A (PKA)- and phospholipase C (PLC)/Ca2+-mediated signaling pathways. Since circulating glucagon and tissue angiotensin II (Ang II) levels are inappropriately elevated in type 2 diabetes, we tested the hypothesis that glucagon induces phosphorylation of ERK 1/2 in MCs by interacting with Ang II receptor signaling. Stimulation of MCs by glucagon (10 nM) induced a marked increase in intracellular [Ca2+]i that was abolished by [Des-His1, Glu9]-glucagon (1 microM), a selective glucagon receptor antagonist. Both glucagon and Ang II-induced ERK 1/2 phosphorylation (glucagon: 214+/-14%; Ang II: 174+/-16%; p<0.001 versus control), and these responses were inhibited by the AT1 receptor blocker losartan (glucagon + losartan: 77+/-14%; Ang II + losartan: 84+/-18%; p<0.01 versus glucagon or Ang II) and the AT2 receptor blocker PD 123319 (glucagon + PD: 78+/-7%; Ang II + PD: 87+/-7%; p<0.01 versus glucagon or Ang II). Inhibition of cAMP-dependent PKA with H89 (1 microM) or PLC with U73122 (1 microM) also markedly attenuated the phosphorylation of ERK 1/2 induced by glucagon (glucagon + U73122: 109+/-15%; glucagon + H89: 113+/-16%; p<0.01 versus glucagon) or Ang II (Ang II + U73122: 111+/-13%; Ang II + H89: 86+/-10%; p<0.01 versus Ang II). Wortmannin (1 microM), a selective PI 3-kinase inhibitor, also blocked glucagon- or Ang II-induced ERK 1/2 phosphorylation. These results suggest that AT1 receptor-activated cAMP-dependent PKA, PLC and PI 3-kinase signaling is involved in glucagon-induced MAP kinase ERK 1/2 phosphorylation in MCs. The inhibitory effect of PD 123319 on glucagon-induced ERK 1/2 phosphorylation further suggests that AT2 receptors also play a similar role in this response.


Subject(s)
Angiotensins/metabolism , Glomerular Mesangium/enzymology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Receptor Cross-Talk , Receptors, Angiotensin/metabolism , Receptors, Glucagon/metabolism , Animals , Calcium/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Glomerular Mesangium/cytology , Glomerular Mesangium/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Rats , Type C Phospholipases/metabolism
20.
Hum Pathol ; 37(6): 637-47, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16733202

ABSTRACT

The mechanism of renal cell apoptosis involves transcriptional activation of the inducible nitric oxide synthase (iNOS) gene by nuclear factor (NF)-kappaB. The role of apoptosis in mediating tubulointerstitial injury in human lupus nephritis (LN) remains unclear. We examined the relationship between alterations in NF-kappaB activation and iNOS expression levels and the degree of apoptosis in both glomerular and tubulointerstitial compartments of subjects with LN. Studies were done in renal biopsies from 49 patients with LN and 10 normal kidney tissues. Apoptotic and proliferating cells were identified by terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling and staining with anti-proliferating cell nuclear antigen antibody, respectively. Nuclear factor-kappaB and iNOS expression was examined by Southwestern histochemistry and immunohistochemistry, respectively. Glomerular cell apoptosis and proliferation increased concomitantly in LN. Glomerular apoptosis correlated with the activity index, the degree of proliferation, and the level of glomerular overexpression of iNOS and activated NF-kappaB in LN. Tubular cell apoptosis correlated with the activity and chronicity indices, the degree of tubular atrophy, and decline in renal function at the time of biopsy. Tubular expression of iNOS and activated NF-kappaB correlated with tubular cell proliferation in LN. Nuclear factor-kappaB activation accompanied overexpression of iNOS in both glomerular and tubulointerstitium compartments in LN. Apoptosis of renal cells associated with NF-kappaB activation and iNOS overexpression may play an important role in mediating chronic renal injury, especially tubulointerstitial lesions that may manifest clinically as progressive renal insufficiency.


Subject(s)
Apoptosis , Cell Proliferation , Lupus Nephritis/enzymology , Lupus Nephritis/physiopathology , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/metabolism , Adolescent , Adult , Aged , Case-Control Studies , Female , Glomerular Mesangium/enzymology , Glomerular Mesangium/metabolism , Humans , Immunohistochemistry , Kidney Tubules/enzymology , Kidney Tubules/metabolism , Male , Middle Aged , Nitric Oxide Synthase Type II/genetics , Proliferating Cell Nuclear Antigen/metabolism , Retrospective Studies , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL