Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Am J Hypertens ; 25(6): 690-6, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22378033

ABSTRACT

BACKGROUND: Cardiotonic steroids (CTS) are implicated in pathophysiology of uremic cardiomyopathy. In the present study, we tested whether a monoclonal antibody (mAb) against the bufadienolide CTS, marinobufagenin (MBG), alleviates cardiac hypertrophy and fibrosis in partially nephrectomized (PNx) rats. METHODS: In PNx rats, we compared the effects of 3E9 anti-MBG mAb and of Digibind, an affinity-purified digoxin antibody, on blood pressure and cardiac hypertrophy and fibrosis following 4 weeks after the surgery. RESULTS: In PNx rats, a fourfold elevation in plasma MBG levels was associated with hypertension, increased cardiac levels of carbonylated protein, cardiac hypertrophy, a reduction in cardiac expression of a nuclear transcription factor which is a negative regulator of collagen synthesis, Friend leukemia integration-1 (Fli-1), and an increase in the levels of collagen-1. A single intraperitoneal administration of 3E9 mAb to PNx rats reduced blood pressure by 59 mm Hg for 7 days and produced a significant reduction in cardiac weight and cardiac levels of oxidative stress, an increase in the expression of Fli-1, and a reduction in cardiac fibrosis. The effects of Digibind were similar to those of 3E9 mAb, but were less pronounced. CONCLUSIONS: In experimental chronic renal failure, elevated levels of MBG contribute to hypertension and induce cardiac fibrosis via suppression of Fli-1, representing a potential target for therapy.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Bufanolides/immunology , Cardiomegaly/etiology , Cardiomegaly/prevention & control , Kidney Failure, Chronic/complications , Myocardium/pathology , Animals , Antibodies, Monoclonal/pharmacology , Blood Pressure/drug effects , Blood Pressure/physiology , Bufanolides/metabolism , Cardiomegaly/metabolism , Comorbidity , Disease Models, Animal , Fibrosis , Immunoglobulin Fab Fragments/pharmacology , Immunoglobulin Fab Fragments/therapeutic use , Kidney Failure, Chronic/metabolism , Kidney Failure, Chronic/surgery , Male , Myocardium/metabolism , Nephrectomy , Proto-Oncogene Protein c-fli-1/metabolism , Rats , Rats, Sprague-Dawley
2.
Hypertension ; 54(6): 1313-20, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19884563

ABSTRACT

Spironolactone has been noted to attenuate cardiac fibrosis. We have observed that the cardiotonic steroid marinobufagenin plays an important role in the diastolic dysfunction and cardiac fibrosis seen with experimental renal failure. We performed the following studies to determine whether and how spironolactone might ameliorate these changes. First, we studied rats subjected to partial nephrectomy or administration of exogenous marinobufagenin. We found that spironolactone (20 mg/kg per day) attenuated the diastolic dysfunction as assessed by ventricular pressure-volume loops and essentially eliminated cardiac fibrosis as assessed by trichrome staining and Western blot. Next, we examined the effects of spironolactone and its major metabolite, canrenone (both 100 nM), on marinobufagenin stimulation of rat cardiac fibroblasts. Both spironolactone and canrenone prevented the stimulation of collagen production by 1 nM marinobufagenin but not 100 nM marinobufagenin, as assessed by proline incorporation and procollagen 1 expression, as well as signaling through the sodium-potassium-ATPase, as evidenced by protein kinase C isoform delta translocation and extracellular signal regulated kinase 1/2 activation. Both spironolactone and canrenone also altered ouabain binding to cultured porcine cells in a manner consistent with competitive inhibition. Our data suggest that some of the antifibrotic effects of spironolactone may be attributed to antagonism of marinobufagenin signaling through the sodium-potassium-ATPase.


Subject(s)
Bufanolides/antagonists & inhibitors , Cardiomyopathies/drug therapy , Mineralocorticoid Receptor Antagonists/pharmacology , Spironolactone/pharmacology , Uremia/complications , Animals , Bufanolides/metabolism , Bufanolides/pharmacology , Canrenone/pharmacology , Cardiomyopathies/etiology , Cardiomyopathies/pathology , Cardiotonic Agents/antagonists & inhibitors , Cardiotonic Agents/metabolism , Cells, Cultured , Disease Models, Animal , Drug Interactions , Endomyocardial Fibrosis/drug therapy , Endomyocardial Fibrosis/etiology , Endomyocardial Fibrosis/pathology , Fibroblasts/cytology , Fibroblasts/drug effects , Myocardium/cytology , Nephrectomy , Ouabain/antagonists & inhibitors , Ouabain/metabolism , Procollagen/metabolism , Proline/pharmacokinetics , Rats , Renal Insufficiency/complications , Tritium
3.
Am J Physiol Renal Physiol ; 296(5): F1219-26, 2009 May.
Article in English | MEDLINE | ID: mdl-19261738

ABSTRACT

The cardiotonic steroid marinobufagenin (MBG) has been implicated in the pathogenesis of experimental uremic cardiomyopathy, which is characterized by progressive cardiac fibrosis. We examined whether the transcription factor Friend leukemia integration-1 (Fli-1) might be involved in this process. Fli-1-knockdown mice demonstrated greater cardiac collagen-1 expression and fibrosis compared with wild-type mice; both developed increased cardiac collagen expression and fibrosis after 5/6 nephrectomy. There was a strong inverse relationship between the expressions of Fli-1 and procollagen in primary culture of rat cardiac and human dermal fibroblasts as well as a cell line derived from renal fibroblasts and MBG-induced decreases in nuclear Fli-1 as well as increases in procollagen-1 expression in these cells. Transfection of a Fli-1 expression vector prevented increased procollagen-1 expression from MBG. MBG exposure induced a rapid translocation of the delta-isoform of protein kinase C (PKCdelta) to the nucleus. This translocation was prevented by pharmacological inhibition of phospholipase C, and MBG-induced increases in procollagen-1 expression were prevented with a PKCdelta- but not a PKCalpha-specific inhibitor. Finally, immunoprecipitation studies strongly suggest that MBG induced phosphorylation of Fli-1. We feel these data support a causal relationship with MBG-induced translocation of PKCdelta, which results in phosphorylation of as well as decreases in nuclear Fli-1 expression, which, in turn, leads to increases in collagen production. Should these findings be confirmed, we speculate that this pathway may represent a therapeutic target for uremic cardiomyopathy as well as other conditions associated with excessive fibrosis.


Subject(s)
Bufanolides/pharmacology , Cardiomyopathies/drug therapy , Enzyme Inhibitors/pharmacology , Procollagen/genetics , Protein Kinase C-delta/metabolism , Proto-Oncogene Protein c-fli-1/metabolism , Uremia/complications , Animals , Bufo marinus , Cardiomyopathies/etiology , Cardiomyopathies/pathology , Cell Nucleus/metabolism , Cells, Cultured , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/physiology , Fibrosis , Gene Expression/drug effects , Gene Expression/physiology , Mice , Mice, Inbred Strains , Mice, Mutant Strains , Myocardium/cytology , Nephrectomy , Protein Kinase C-delta/genetics , Proto-Oncogene Protein c-fli-1/genetics
4.
Am J Physiol Renal Physiol ; 296(4): F922-34, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19176701

ABSTRACT

We recently demonstrated that the cardiotonic steroid marinobufagenin (MBG) induced fibrosis in rat hearts through direct stimulation of collagen I secretion by cardiac fibroblasts. This stimulation was also responsible for the cardiac fibrosis seen in experimental renal failure. In this study, the effect of MBG on the development of renal fibrosis in rats was investigated. Four weeks of MBG infusion triggered mild periglomerular and peritubular fibrosis in the cortex and the appearance of fibrotic scars in the corticomedullary junction of the kidney. MBG also significantly increased the protein levels and nuclear localization of the transcription factor Snail in the tubular epithelia. It is known that activation of Snail is associated with epithelial-to-mesenchymal transition (EMT) during renal fibrosis. To examine whether MBG alone can trigger EMT, we used the porcine proximal tubular cell line LLC-PK1. MBG (100 nM) caused LLC-PK1 cells grown to confluence to acquire a fibroblast-like shape and have an invasive motility. The expressions of the mesenchymal proteins collagen I, fibronectin, and vimentin were increased twofold. However, the total level of E-cadherin remained unchanged. These alterations in LLC-PK1 cells in the presence of MBG were accompanied by elevated expression and nuclear translocation of Snail. During the time course of EMT, MBG did not have measurable inhibitory effects on the ion pumping activity of its natural ligand, Na(+)-K(+)-ATPase. Our data suggest that the MBG may be an important factor in inducing EMT and, through this mechanism, elevated levels of MBG in chronic renal failure may play a role in the progressive fibrosis.


Subject(s)
Bufanolides/toxicity , Cardiotonic Agents/toxicity , Cell Transdifferentiation/drug effects , Epithelial Cells/drug effects , Kidney Diseases/chemically induced , Kidney/drug effects , Mesoderm/drug effects , Animals , Bufanolides/administration & dosage , Cardiotonic Agents/administration & dosage , Cell Movement/drug effects , Cell Shape/drug effects , Collagen Type I/metabolism , Enzyme Inhibitors/toxicity , Epithelial Cells/metabolism , Epithelial Cells/pathology , Fibronectins/metabolism , Fibrosis , Infusion Pumps, Implantable , Kidney/metabolism , Kidney/pathology , Kidney Diseases/metabolism , Kidney Diseases/pathology , LLC-PK1 Cells , Male , Mesoderm/metabolism , Mesoderm/pathology , Ouabain/pharmacology , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Snail Family Transcription Factors , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Sodium-Potassium-Exchanging ATPase/metabolism , Swine , Time Factors , Transcription Factors/metabolism , Vimentin/metabolism
5.
J Appl Physiol (1985) ; 105(1): 30-6, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18483172

ABSTRACT

We previously reported that cardiotonic steroids stimulate collagen synthesis by cardiac fibroblasts in a process that involves signaling through the Na-K-ATPase pathway (Elkareh et al. Hypertension 49: 215-224, 2007). In this study, we examined the effect of cardiotonic steroids on dermal fibroblasts collagen synthesis and on wound healing. Increased collagen expression by human dermal fibroblasts was noted in response to the cardiotonic steroid marinobufagenin in a dose- and time-dependent fashion. An eightfold increase in collagen synthesis was noted when cells were exposed to 10 nM marinobufagenin for 24 h (P < 0.01). Similar increases in proline incorporation were seen following treatment with digoxin, ouabain, and marinobufagenin (10 nM x 24 h, all results P < 0.01 vs. control). The coadministration of the Src inhibitor PP2 or N-acetylcysteine completely prevented collagen stimulation by marinobufagenin. Next, we examined the effect of digoxin, ouabain, and marinobufagenin on the rate of wound closure in an in vitro model where human dermal fibroblasts cultures were wounded with a pipette tip and monitored by digital microscopy. Finally, we administered digoxin in an in vivo wound healing model. Olive oil was chosen as the digoxin carrier because of a favorable partition coefficient observed for labeled digoxin with saline. This application significantly accelerated in vivo wound healing in rats wounded with an 8-mm biopsy cut. Increased collagen accumulation was noted 9 days after wounding (both P < 0.01). The data suggest that cardiotonic steroids induce increases in collagen synthesis by dermal fibroblasts, as could potentially be exploited to accelerate wound healing.


Subject(s)
Cardiac Glycosides/pharmacology , Cardiotonic Agents/pharmacology , Collagen/biosynthesis , Skin/metabolism , Wound Healing/drug effects , Animals , Bufanolides/pharmacology , Digoxin/pharmacology , Dose-Response Relationship, Drug , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression/drug effects , Humans , Image Processing, Computer-Assisted , Male , Oligonucleotide Array Sequence Analysis , Ouabain/pharmacology , Proline/metabolism , Rats , Rats, Sprague-Dawley , Skin/cytology , Skin/drug effects , src-Family Kinases/antagonists & inhibitors
6.
Am J Physiol Renal Physiol ; 294(4): F859-66, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18094034

ABSTRACT

The ion transporter Na-K-ATPase functions as a cell signal transducer that mediates ouabain-induced activation of protein kinases, such as ERK. While Na-K-ATPase composed of the alpha(1)-polypeptide is involved in cell signaling, the role of other alpha-isoforms (alpha(2), alpha(3), and alpha(4)) in transmitting ouabain effects is unknown. We have explored this using baculovirus-directed expression of Na-K-ATPase polypeptides in insect cells and ERK phosphorylation as an indicator of ouabain-induced signaling. Ouabain addition to Sf-9 cells coexpressing Na-K-ATPase alpha(1)- and beta(1)-isoforms stimulated ERK phosphorylation. In contrast, expression of the alpha(1)- and beta(1)-polypeptides alone resulted in no effect, indicating that the alphabeta-complex is necessary for Na-K-ATPase signaling. Moreover, the ouabain effect was sensitive to genistein, suggesting that Na-K-ATPase-mediated tyrosine kinase activation is a critical event in the intracellular cascade leading to ERK phosphorylation. In addition, the Na-K-ATPases alpha(3)beta(1)- and alpha(4)beta(1)-isozymes, but not alpha(2)beta(1), responded to ouabain treatment. In agreement with the differences in ouabain affinity of the alpha-polypeptides, alpha(1)beta(1) required 100- to 1,000-fold more ouabain to signal than did alpha(4)beta(1) and alpha(3)beta(1), respectively. These results confirm the role of the Na-K-ATPase in ouabain signal transduction, show that there are important isoform-specific differences in Na-K-ATPase signaling, and demonstrate the suitability of the baculovirus expression system for studying Na-K-ATPase-mediated ouabain effects.


Subject(s)
Ouabain/pharmacology , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Baculoviridae/drug effects , Baculoviridae/physiology , Cell Line , Extracellular Signal-Regulated MAP Kinases/drug effects , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Isoenzymes/genetics , Isoenzymes/metabolism , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Sodium-Potassium-Exchanging ATPase/genetics , Spodoptera/enzymology
7.
Am J Physiol Renal Physiol ; 294(2): F450-4, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18032546

ABSTRACT

Because of the plethora of genetic manipulations available in the mouse, we performed a partial nephrectomy in the mouse and examined whether the phenotypical features of uremic cardiomyopathy described in humans and rats were also present in the murine model. A 5/6 nephrectomy was performed using a combination of electrocautory to decrease renal mass on the left kidney and right surgical nephrectomy. This procedure produced substantial and persistent hypertension as well as increases in circulating concentrations of marinobufagenin. Invasive physiological measurements of cardiac function demonstrated that the 5/6 nephrectomy resulted in impairment of both active and passive left ventricular relaxation at 4 wk whereas tissue Doppler imaging detected changes in diastolic function after 6 wk. Morphologically, hearts demonstrated enlargement and progressive fibrosis, and biochemical measurements demonstrated downregulation of the sarcoplasmic reticulum calcium ATPase as well as increases in collagen-1, fibronectin, and vimentin expression. Our results suggest that partial nephrectomy in the mouse establishes a model of uremic cardiomyopathy which shares phenotypical features with the rat model as well as patients with chronic renal failure.


Subject(s)
Cardiomyopathies/physiopathology , Disease Models, Animal , Nephrectomy , Renal Insufficiency/complications , Animals , Antihypertensive Agents/pharmacology , Blood Pressure/drug effects , Blood Pressure/physiology , Bufanolides/blood , Cardiomegaly/metabolism , Cardiomegaly/pathology , Cardiomyopathies/etiology , Cardiomyopathies/metabolism , Echocardiography, Doppler , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibrosis , Heart/drug effects , Heart/physiopathology , Male , Mice , Mice, Inbred Strains , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Stroke Volume/physiology , Ventricular Function, Left/physiology , src-Family Kinases/metabolism
8.
Hypertension ; 49(1): 215-24, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17145984

ABSTRACT

We have observed recently that experimental renal failure in the rat is accompanied by increases in circulating concentrations of the cardiotonic steroid, marinobufagenin (MBG), and substantial cardiac fibrosis. We performed the following studies to examine whether MBG might directly stimulate cardiac fibroblast collagen production. In vivo studies were performed using the 5/6th nephrectomy model of experimental renal failure (PNx), MBG infusion (MBG), PNx after immunization against MBG, and concomitant PNx and adrenalectomy. Physiological measurements with a Millar catheter and immunohistochemistry were performed. In vitro studies were then pursued with cultured isolated cardiac fibroblasts. We observed that PNx and MBG increased MBG levels, blood pressure, heart size, impaired diastolic function, and caused cardiac fibrosis. PNx after immunization against MBG and concomitant PNx and adrenalectomy had similar blood pressure as PNx but less cardiac hypertrophy, diastolic dysfunction, and cardiac fibrosis. MBG induced increases in procollagen-1 expression by cultured cardiac fibroblasts at 1 nM concentration. These increases in procollagen expression were accompanied by increases in collagen translation and increases in procollagen-1 mRNA without any demonstrable increase in procollagen-1 protein stability. The stimulation of fibroblasts with MBG could be prevented by administration of inhibitors of tyrosine phosphorylation, Src activation, epidermal growth factor receptor transactivation, and N-acetyl cysteine. Based on these findings, we propose that MBG directly induces increases in collagen expression by fibroblasts, and we suggest that this may be important in the cardiac fibrosis seen with experimental renal failure.


Subject(s)
Bufanolides/pharmacology , Cardiomyopathies/chemically induced , Cardiomyopathies/pathology , Collagen/biosynthesis , Fibroblasts/metabolism , Myocardium/metabolism , Uremia/complications , Animals , Blood Pressure/drug effects , Cells, Cultured , Fibroblasts/drug effects , Fibrosis , Heart/physiopathology , Male , Myocardium/cytology , Rats , Rats, Sprague-Dawley , Renal Insufficiency/physiopathology , Signal Transduction , Sodium-Potassium-Exchanging ATPase/metabolism , Transforming Growth Factor beta/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...