Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
J Am Soc Nephrol ; 19(1): 125-34, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18178802

ABSTRACT

Hypokalemic nephropathy is associated with alterations in intrarenal vasoactive substances, leading to vasoconstriction, salt-sensitivity, and progression of interstitial fibrosis. In this study, we investigated whether hypokalemic nephropathy might also involve impaired renal angiogenesis. Sprague-Dawley rats that were fed low-potassium diets developed peritubular capillary loss that began in the inner stripe of the outer medulla (week 2) and progressed to the outer stripe of the outer medulla (week 4) and cortex (week 12). These changes were associated with increased macrophage infiltration, increased expression of both monocyte chemoattractant protein-1 and TNF-alpha, and a loss of vascular endothelial growth factor and endothelial nitric oxide synthase. Renal thiobarbituric acid-reactive substances, markers of oxidative stress, were increased late in disease. In conclusion, hypokalemic nephropathy is associated with impaired renal angiogenesis, evidenced by progressive capillary loss, reduced endothelial cell proliferation, and loss of VEGF expression.


Subject(s)
Hypokalemia/pathology , Hypokalemia/physiopathology , Kidney Diseases/physiopathology , Neovascularization, Pathologic/physiopathology , Animals , Body Weight , Creatinine/blood , Disease Models, Animal , Hypertrophy , Immunohistochemistry , Kidney/pathology , Kidney Diseases/pathology , Kidney Tubules/pathology , Neovascularization, Physiologic , Organ Size , Potassium/blood , Rats , Rats, Sprague-Dawley
2.
Regul Pept ; 141(1-3): 129-34, 2007 Jun 07.
Article in English | MEDLINE | ID: mdl-17307261

ABSTRACT

OBJECTIVE: We investigated the pathophysiological significance of molecular forms of adrenomedullin (AM) in patients after the Fontan procedure. METHODS: Plasma concentrations of mature AM (AM-m), an active form, glycine-extended AM (AM-Gly), an inactive form, and total AM (AM-T: AM-m+AM-Gly) were measured by specific immunoradiometric assay in the femoral vein, pulmonary artery and femoral artery of 29 consecutive patients after the Fontan procedure. The eleven patients who had history of Kawasaki disease and have normal coronary and hemodynamics served as control. RESULTS: Patients who underwent Fontan procedure had significantly higher venous concentrations of AM-T, AM-Gly, and AM-m than age-matched normal controls (AM-T, 12.0+/-3.3 vs. 9.6+/-2.0; AM-Gly, 10.4+/-3.0 vs. 8.5+/-1.6; AM-m, 1.6+/-0.7 vs. 1.0+/-0.6 pmol/l, each p<0.05). In patients with Fontan procedure, there were no differences in plasma AM-T, AM-Gly or AM-m levels between the femoral vein and pulmonary artery, however, there was a significant step-down in the AM-m levels, but not in plasma AM-T or AM-Gly levels, between the pulmonary artery and femoral artery (1.3+/-0.6 to 1.0+/-0.6, p<0.05). The venous concentrations of AM-m correlated negatively with systemic blood flow (cardiac output) (r=-0.46, p<0.05). CONCLUSIONS: Results suggest that in Fontan circulation plasma AM-m is increased in parallel with those of AM-T and AM-Gly and that AM-m is extracted in the lung. Extracted AM-m may be involved in the regulation of pulmonary arterial tonus, although further studies are necessary to elucidate the exact role of AM in Fontan circulation.


Subject(s)
Adrenomedullin/blood , Fontan Procedure/methods , Lung/blood supply , Lung/chemistry , Pulmonary Circulation/physiology , Adolescent , Adult , Case-Control Studies , Child , Child, Preschool , Female , Humans , Immunoradiometric Assay , Infant , Male
3.
Endocrinology ; 148(3): 1218-25, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17138658

ABSTRACT

Evidence has emerged that undernutrition in utero is a risk factor for cardiovascular disorders in adulthood, along with genetic and environmental factors. Recently, the local expression of angiotensinogen and related bioactive substances has been demonstrated to play a pivotal role in cardiac remodeling, i.e. fibrosis and hypertrophy. The aim of the present study was to clarify the possible involvement of the local cardiac angiotensin system in fetal undernutrition-induced cardiovascular disorders. We developed a mouse model of undernutrition in utero by maternal food restriction, in which offspring (UN offspring) showed an increase in systolic blood pressure (8 wk of age, P < 0.05; and 16 wk, P < 0.01), perivascular fibrosis of the coronary artery (16 wk, P < 0.05) and cardiac cardiomegaly (16 wk, P < 0.01), and cardiomyocyte enlargement, concomitant with a significant augmentation of angiotensinogen (P < 0.05) and endothelin-1 (P < 0.01) mRNA expression and a tendency to increase in immunostaining for both angiotensin II and endothelin-1 in the left ventricles (16 wk). These findings suggest that fetal undernutrition activated the local cardiac angiotensin system-associated bioactive substances, which contributed, at least partly, to the development of cardiac remodeling in later life, in concert with the effects of increase in blood pressure.


Subject(s)
Blood Pressure , Cardiovascular Diseases/etiology , Fetal Nutrition Disorders/physiopathology , Malnutrition/complications , Prenatal Exposure Delayed Effects/physiopathology , Renin-Angiotensin System , Ventricular Remodeling , Angiotensin II/blood , Angiotensin II/metabolism , Angiotensin II/physiology , Animals , Cardiovascular Diseases/embryology , Female , Fetal Nutrition Disorders/blood , Fetal Nutrition Disorders/etiology , Heart Ventricles/metabolism , Leptin/pharmacology , Malnutrition/blood , Mice , Mice, Inbred C57BL , Nitric Oxide/blood , Pregnancy , Prenatal Exposure Delayed Effects/blood , Sodium Glutamate/pharmacology , Ventricular Remodeling/drug effects
4.
Expert Rev Endocrinol Metab ; 2(5): 615-622, 2007 Sep.
Article in English | MEDLINE | ID: mdl-30736124

ABSTRACT

Diabetic nephropathy is a leading cause of end-stage renal disease. Several pathways, including the renin-angiotensin system, have been postulated as potential mechanisms of diabetic nephropathy. In addition, glomerulogenesis-related molecules are involved in the pathogenesis of diabetic nephropathy, especially at the early stage. They can be divided into three groups by function, that is, fibrosis-related, podocyte differentiation-related and angiogenesis-related molecules. Most of the molecules are expressed in the podocyte and upregulated, even during the normoalbuminuric stage. Expression of several podocyte structure-related molecules are also altered at the normoalbuminuric stage. They can contribute to the structural alteration of the podocyte in diabetic nephropathy. Thus, normalization of the expression of glomerulogenesis-related molecules could be a new target for preventing the initiation and progression of diabetic nephropathy.

5.
Diabetes ; 55(10): 2747-56, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17003339

ABSTRACT

Glomerular injury plays a pivotal role in the development of diabetic nephropathy. To elucidate molecular mechanisms underlying diabetic glomerulopathy, we compared glomerular gene expression profiles of db/db mice with those of db/m control mice at a normoalbuminuric stage characterized by hyperglycemia and at an early stage of diabetic nephropathy with elevated albuminuria, using cDNA microarray. In db/db mice at the normoalbuminuric stage, hypoxia-inducible factor-1alpha (HIF-1alpha), ephrin B2, glomerular epithelial protein 1, and Pod-1, which play key roles in glomerulogenesis, were already upregulated in parallel with an alteration of genes related to glucose metabolism, lipid metabolism, and oxidative stress. Podocyte structure-related genes, actinin 4alpha and dystroglycan 1 (DG1), were also significantly upregulated at an early stage. The alteration in the expression of these genes was confirmed by quantitative RT-PCR. Through pioglitazone treatment, gene expression of ephrin B2, Pod-1, actinin 4alpha, and DG1, as well as that of oxidative stress and lipid metabolism, was restored concomitant with attenuation of albuminuria. In addition, HIF-1alpha protein expression was partially attenuated by pioglitazone. These results suggest that not only metabolic alteration and oxidative stress, but also the alteration of gene expression related to glomerulogenesis and podocyte structure, may be involved in the pathogenesis of early diabetic glomerulopathy in type 2 diabetes.


Subject(s)
Diabetic Nephropathies/physiopathology , Gene Expression/drug effects , Kidney Glomerulus/growth & development , Podocytes/pathology , Thiazolidinediones/pharmacology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Diabetes Mellitus, Type 2/physiopathology , Disease Models, Animal , Ephrin-B2/genetics , Gene Expression Profiling , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Kidney Glomerulus/drug effects , Membrane Proteins/genetics , Mice , Mice, Obese , Oligonucleotide Array Sequence Analysis , Pioglitazone , Protein Tyrosine Phosphatases/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 3
7.
Nihon Rinsho ; 63 Suppl 3: 167-71, 2005 Mar.
Article in Japanese | MEDLINE | ID: mdl-15813063
8.
Regul Pept ; 127(1-3): 239-44, 2005 Apr 15.
Article in English | MEDLINE | ID: mdl-15680493

ABSTRACT

Specific adrenomedullin receptors have been identified as calcitonin receptor-like receptor (CRLR)/receptor activity-modifying proteins (RAMP2 and RAMP3) complexes. Although we have demonstrated that adrenomedullin is increased in volume overload-induced cardiac hypertrophy, it remains unknown whether the adrenomedullin receptor is altered or not. This study sought to investigate the significance of intracardiac adrenomedullin and its receptor system in volume overload-induced cardiac hypertrophy. Left ventricular adrenomedullin levels were higher in aortocaval shunt (ACS) rats than in controls (+58%). The left ventricular gene expressions of adrenomedullin, CRLR, RAMP2 and RAMP3 were increased (+27%, +76%, +108% and +131%, respectively) and the left ventricular collagen gene expressions were also increased (type I: +138%, type III: +87%). The left ventricular adrenomedullin level correlated with the gene expression of type III collagen (R=0.42). These results suggest that intracardiac adrenomedullin and its receptor system are upregulated and may participate in the regulation of cardiac remodeling in volume overload-induced cardiac hypertrophy.


Subject(s)
Gene Expression Regulation , Hypertrophy, Left Ventricular/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Myocardium/metabolism , Peptides/metabolism , Receptors, Calcitonin/metabolism , Up-Regulation , Adrenomedullin , Animals , Calcitonin Receptor-Like Protein , Cardiac Volume , Collagen Type III/genetics , Collagen Type III/metabolism , Hemodynamics , Hypertrophy, Left Ventricular/pathology , Hypertrophy, Left Ventricular/physiopathology , Intracellular Signaling Peptides and Proteins/genetics , Male , Membrane Proteins/genetics , Myocardium/pathology , Peptides/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Radioimmunoassay , Rats , Rats, Wistar , Receptor Activity-Modifying Proteins , Receptors, Calcitonin/genetics , Statistics as Topic , Ventricular Remodeling
9.
Regul Pept ; 128(1): 7-13, 2005 May 15.
Article in English | MEDLINE | ID: mdl-15721482

ABSTRACT

Adrenomedullin reduces systemic blood pressure and increases urinary sodium excretion partly through the release of nitric oxide. We hypothesized that chronic adrenomedullin infusion ameliorates salt-sensitive hypertension and increases the expression of renal nitric oxide synthase (NOS) in Dahl salt-sensitive (DS) rats, because the reduced renal NOS expression promotes salt sensitivity. DS rats and Dahl salt-resistant (DR) rats were fed a high sodium diet (8.0% NaCl) for 3 weeks. The high sodium diet resulted in an increase in blood pressure and a reduction of urinary sodium excretion in association with increased renal adrenomedullin concentrations and decreased expression of renal neuronal NOS (nNOS) and renal medullary endothelial NOS (eNOS) in DS rats compared with DR rats. Chronic adrenomedullin infusion partly inhibited the increase of blood pressure and proteinuria in association with a restoration of renal nNOS and medullary eNOS expression in DS rats under the high sodium diet. The immunohistochemical analysis revealed that the restored renal nNOS expression induced by chronic adrenomedullin infusion may reflect the restoration of nNOS expression in the macula densa and inner medullary collecting duct. These results suggest that adrenomedullin infusion has beneficial effects on this hypertension probably in part through restored renal NOS expression in DS rats.


Subject(s)
Kidney/drug effects , Nitric Oxide Synthase/metabolism , Peptides/pharmacology , Adrenomedullin , Animals , Blotting, Western , Immunohistochemistry , Kidney/enzymology , Kidney/physiopathology , Male , Peptides/administration & dosage , Rats
10.
Regul Pept ; 124(1-3): 65-72, 2005 Jan 15.
Article in English | MEDLINE | ID: mdl-15544842

ABSTRACT

Insulin-like growth factor (IGF)-1 appears to play an important role in cardiac hypertrophy or remodeling. However, the role of endogenous IGF-1 in the growth of cardiac myocytes and fibroblasts remains unclear. This study investigated the major site of the production of cardiac IGF-1 and the local effects of endogenous IGF-1 secreted from cardiac cells. A significant expression of IGF-1 mRNA was found in cultured neonatal and adult rat cardiac fibroblasts, but not in myocytes. In addition, an in vivo examination by in situ hybridization histochemical analyses demonstrated the IGF-1 transcripts in the interstitial fibrotic tissue of the ventricle. Time-dependent secretion of IGF-1 protein was also observed in cultured cardiac fibroblasts. An antibody against IGF-1 decreased collagen synthesis in cardiac fibroblasts under basal conditions. Fibroblast-conditioned medium, as well as exogenous IGF-1, increased protein synthesis in cardiac myocytes, and this increase was inhibited by antibodies against IGF-1 and IGF-1 receptor, IGF binding protein-3, and IGF-1 receptor antagonist. These observations suggest that IGF-1 is produced and released mainly from cardiac fibroblasts and that endogenous IGF-1 promotes collagen synthesis by cardiac fibroblasts and hypertrophy of myocytes as an autocrine and a paracrine factor. Cardiac IGF-1 may function as an endogenous modulator of cardiac hypertrophy or remodeling.


Subject(s)
Autocrine Communication , Insulin-Like Growth Factor I/metabolism , Myocardium/cytology , Myocardium/metabolism , Paracrine Communication , Animals , Animals, Newborn , Antibodies/immunology , Cell Size , Cells, Cultured , Dose-Response Relationship, Drug , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Regulation , Insulin-Like Growth Factor I/biosynthesis , Insulin-Like Growth Factor I/genetics , Muscle Cells/metabolism , Rats , Rats, Wistar
12.
Metabolism ; 53(6): 710-5, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15164316

ABSTRACT

The present study examined the direct effects of high glucose and insulin on protein synthesis in cardiac myocytes and DNA and collagen synthesis in cardiac fibroblasts. Cultured rat cardiac myocytes and fibroblasts were grown in media containing normal glucose, high glucose, or osmotic control, and incubated with or without insulin. In cardiac myocytes, high glucose had no effect, but insulin increased protein synthesis and atrial natriuretic peptide (ANP) secretion and gene expression. The extracellular signal-regulated protein kinase (ERK)/mitogen-activated protein kinase (MAPK) inhibitor and the protein kinase C (PKC) inhibitor blocked insulin-induced protein synthesis. In cardiac fibroblasts, high glucose and osmotic control media increased DNA synthesis. Collagen synthesis and fibronectin and transforming growth factor-beta1 (TGF-beta1) mRNA expression were stimulated by high glucose, but not by osmotic control. Insulin increased DNA and collagen synthesis in fibroblasts, and the insulin-induced increase in DNA synthesis was blocked by the phosphatidylinositol 3 kinase (PI3K) inhibitor. Our findings suggest that cardiomyocyte protein synthesis is mainly regulated by insulin rather than high glucose and both high glucose and insulin contribute to fibroblast DNA and collagen synthesis. High glucose accelerates fibroblast DNA synthesis and collagen synthesis, and fibronectin and TGF-beta1 mRNA expression, dependent or independent of osmotic stress. Insulin regulates myocyte protein synthesis and fibroblast DNA synthesis through different intracellular mechanisms.


Subject(s)
Atrial Natriuretic Factor/biosynthesis , Collagen/biosynthesis , DNA/biosynthesis , Fibroblasts/metabolism , Glucose/pharmacology , Insulin/pharmacology , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Animals , Animals, Newborn , Atrial Natriuretic Factor/immunology , Cells, Cultured , Enzyme Inhibitors/pharmacology , Fibroblasts/cytology , Fibroblasts/drug effects , Leucine/metabolism , Myocardium/cytology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , RNA, Messenger/biosynthesis , Rats , Up-Regulation/drug effects
13.
Endocrinology ; 145(5): 2131-40, 2004 May.
Article in English | MEDLINE | ID: mdl-14749356

ABSTRACT

C-type natriuretic peptide (CNP) is known to play a role in the local regulation of vascular tone. We recently found that CNP is also produced by cardiac ventricular cells. However, its local effect on myocyte hypertrophy remains to be elucidated. The present study investigated the effects of CNP on cultured cardiac myocyte hypertrophy and the interaction between CNP and endothelin-1 (ET-1) signaling pathways. CNP attenuated basal and ET-1-augumented protein synthesis, atrial natriuretic peptide secretion, hypertrophy-related gene expression, GATA-4 and MEF-2 DNA binding activities, Ca(2+)/calmodulin-dependent kinase II activity, and ERK phosphorylation. CNP also inhibited ET-1-induced increase in intracellular Ca(2+) concentration. These effects of CNP were mimicked by a cGMP analog, 8-bromo cGMP. However, the inhibitory effects of CNP on the hypertrophic response of myocytes were significantly diminished at high concentrations of ET-1. Although CNP increased intracellular cGMP levels in myocytes, ET-1 suppressed CNP-induced cellular cGMP accumulation. A protein kinase C activator and Ca(2+) ionophore mimicked this suppressive effect of ET-1. We further examined the effect of CNP on the paracrine action of ET-1 secreted from cardiac nonmyocytes. CNP and 8-bromo cGMP significantly inhibited ET-1 secretion from nonmyocytes. Although nonmyocyte-conditioned medium increased the protein synthesis in myocytes through endogenous ET-1 action, this increase was significantly attenuated by pretreatment of nonmyocytes with CNP and 8-bromo cGMP. These findings demonstrate that CNP inhibits ET-1-induced cardiac myocyte hypertrophy via a cGMP-dependent mechanism, and conversely, ET-1 inhibits CNP signaling by a protein kinase C- and Ca(2+)-dependent mechanism, suggesting mutual interference between CNP and ET-1 signaling pathways.


Subject(s)
Cardiomegaly , Cyclic GMP/analogs & derivatives , Endothelin-1/pharmacology , Myocardium/cytology , Natriuretic Peptide, C-Type/pharmacology , Signal Transduction , Animals , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cells, Cultured , Cyclic GMP/metabolism , Cyclic GMP/pharmacology , DNA/metabolism , DNA-Binding Proteins/metabolism , Drug Interactions , GATA4 Transcription Factor , MEF2 Transcription Factors , Mitogen-Activated Protein Kinases/metabolism , Myogenic Regulatory Factors , Phosphorylation , Rats , Rats, Wistar , Transcription Factors/metabolism
14.
Endocrinology ; 145(5): 2458-66, 2004 May.
Article in English | MEDLINE | ID: mdl-14736733

ABSTRACT

A cross-talk between cardiac myocytes and nonmyocytes via humoral factors plays an important role in the development of cardiac growth. However, it remains to be elucidated whether humoral factors produced from nonmyocytes have a protective effect on acute myocardial injury. The present in vitro study investigated the antiapoptotic effect of nonmyocytes on doxorubicin (DOX)-induced myocyte apoptosis and its molecular mechanism. Myocyte-nonmyocyte coculture and treatment with nonmyocyte-conditioned media significantly attenuated DOX-induced myocyte apoptosis. Treatment with nonmyocyte-conditioned media stimulated the phosphorylation of ERK, Akt, and cAMP response element-binding protein (CREB) in myocytes. Nonmyocyte-conditioned media also increased protein levels of Bcl-2 but not Bcl-xL and decreased caspase-3 activation induced by DOX. MAPK kinase-specific inhibitor PD98059, phosphatidylinositol-3 kinase-Akt inhibitor LY294002, and CREB antisense oligonucleotide significantly blocked the antiapoptotic effect of nonmyocyte-conditioned media. A considerable amount of endothelin (ET)-1 production was detected in nonmyocytes but not in myocytes. Exogenous ET-1 mimicked nonmyocyte-conditioned media-mediated ERK and CREB phosphorylation and Bcl-2 protein increase but not Akt phosphorylation. In addition, ET-A receptor antagonists BQ123 and BQ485 partially blocked nonmyocyte-conditioned media-mediated antiapoptotic effect, ERK and CREB phosphorylation, and Bcl-2 protein increase. Nonmyocyte-conditioned media and exogenous ET-1 unchanged protein levels of manganese superoxide dismutase and oxidative stress-related product levels augmented by DOX. The present findings demonstrate that cardiac nonmyocytes inhibit DOX-induced myocyte apoptosis, at least in part, via ET-1 secretion-mediated CREB activation independent of the decrease in oxidative stress.


Subject(s)
Apoptosis/drug effects , Doxorubicin/pharmacology , Endothelin-1/physiology , Myocardium/cytology , Myocytes, Cardiac/physiology , Protein Serine-Threonine Kinases , Caspase 3 , Caspases/metabolism , Cell Communication , Cells, Cultured , Coculture Techniques , Culture Media, Conditioned , Cyclic AMP Response Element-Binding Protein/metabolism , DNA Fragmentation , Heart Ventricles/cytology , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-bcl-2/analysis , bcl-X Protein
15.
Endocrinology ; 144(6): 2279-84, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12746286

ABSTRACT

C-type natriuretic peptide (CNP), the third member of the natriuretic peptide family, is known to be synthesized in the central nervous system and vascular endothelial cells, in contrast to atrial natriuretic peptide and brain natriuretic peptide. However, there have been no studies concerning CNP production in cultured cardiac cells. Here, we examined the production and the local effect of CNP in cultured ventricular cells. Under serum-free conditions, adult rat cardiac fibroblasts secreted immunoreactive CNP time dependently. TGF-beta1, basic fibroblast growth factor, and endothelin-1 significantly stimulated CNP secretion. Northern blot analysis detected significant expressions of CNP and its specific receptor (guanylyl cyclase-B) mRNA in cardiac fibroblasts. CNP stimulated intracellular cGMP production in fibroblasts more intensely than atrial and brain natriuretic peptides. CNP inhibited both DNA and collagen syntheses of cardiac fibroblasts, and these inhibitory effects by CNP were stronger than by atrial and brain natriuretic peptides. The inhibition by CNP of DNA and collagen syntheses was reproduced by a cGMP analog, 8-bromo cGMP. The present findings demonstrate that CNP is synthesized in and secreted from cardiac fibroblasts and suggest that CNP has a suppressive effect on fibroblast proliferation and extracellular matrix production, probably via the guanylyl cyclase-B-mediated cGMP-dependent process. CNP produced by cardiac fibroblasts may play a role as an autocrine regulator against excessive cardiac fibrosis.


Subject(s)
Fibroblasts/physiology , Myocardium/cytology , Natriuretic Peptide, C-Type/genetics , Natriuretic Peptide, C-Type/metabolism , Age Factors , Animals , Animals, Newborn , Atrial Natriuretic Factor/genetics , Atrial Natriuretic Factor/metabolism , Atrial Natriuretic Factor/pharmacology , Autocrine Communication/physiology , Cells, Cultured , Collagen/biosynthesis , Cyclic GMP/biosynthesis , DNA/biosynthesis , Fibroblasts/cytology , Fibroblasts/drug effects , Gene Expression/physiology , Heart Ventricles/cytology , Male , Natriuretic Peptide, Brain/genetics , Natriuretic Peptide, Brain/metabolism , Natriuretic Peptide, Brain/pharmacology , Natriuretic Peptide, C-Type/pharmacology , RNA, Messenger/analysis , Rats , Rats, Wistar
16.
Curr Opin Nephrol Hypertens ; 12(3): 233-41, 2003 May.
Article in English | MEDLINE | ID: mdl-12698060

ABSTRACT

PURPOSE OF REVIEW: Tubulointerstitial injury is characteristic of aging-associated renal injury and progressive renal disease. Salt-sensitive hypertension is also associated with tubulointerstitial inflammation, especially when accompanied by microvascular disease. Here we summarize recent studies on the pathogenesis and consequences of tubulointerstitial disease, emphasizing the role of ischemia and the microvasculature. RECENT FINDINGS: Tubulointerstitial injury occurs via several mechanisms of which one of the most important is chronic ischemia. Recent studies suggest that chronic vasoconstriction may contribute to the renal injury associated with angiotensin II, catecholamines, nitric oxide inhibition, hypokalemia, hyperuricemia, and cyclosporine nephropathy. Salt-sensitivity may result as a consequence of the tubulointerstitial inflammatory response to these conditions, and this appears to be perpetuated by the development of preglomerular vascular disease. With progression of tubulointerstitial disease there is also a loss of peritubular capillaries, and stimulating microvascular growth with angiogenic factors can stabilize renal function in these models. SUMMARY: Ischemia secondary to vasoconstriction or to structural changes of the renal vasculature may have important consequences both in terms of mediating salt-sensitive hypertension and renal progression. Angiogenic factors may have potential benefit in preventing or treating these conditions.


Subject(s)
Nephritis, Interstitial/etiology , Animals , Humans , Ischemia/complications , Ischemia/pathology , Ischemia/physiopathology , Kidney/blood supply , Microcirculation/pathology , Microcirculation/physiopathology , Nephritis, Interstitial/pathology , Nephritis, Interstitial/physiopathology , Vascular Diseases/complications , Vascular Diseases/pathology , Vascular Diseases/physiopathology , Vasoconstriction
17.
J Am Soc Nephrol ; 14(2): 397-406, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12538740

ABSTRACT

Hypokalemia causes renal tubulointerstitial injury with an elevation in renal endothelin-1 (ET-1). It was hypothesized that hypokalemic tubulointerstitial injury is ameliorated by the blockade of ET-A receptors (ETA), whereas ET-B receptor (ETB) antagonism may exacerbate the injury, because ETB is thought to mediate vasodilation. Rats were fed a K(+)-deficient diet alone (LC) or with an ETA-selective antagonist ABT-627 (LA) or an ETB-selective antagonist A-192621 (LB) for 8 wk. Control rats were on a normal K(+) diet alone or with the ETA-selective or ETB-selective antagonists. The severity of hypokalemia was not significantly different among LA, LB, and LC. LC developed tubulointerstitial injury with an elevation of renal preproET-1 mRNA level. There was an increase in tubular osteopontin expression, macrophage infiltration, collagen accumulation, and tubular cell hyperplasia. ETA blockade significantly ameliorated all parameters for renal injury in the cortex without suppressing local ET-1 and ETA expression. By contrast, ETB blockade significantly reduced local ET-1 and ETA expression and improved the injury to a similar extent in the cortex. In the medulla, ETA or ETB blockade only partially blocked renal injury. ETA blockade did not affect BP in normokalemic or hypokalemic rats. ETB blockade induced a BP elevation with a decrease in urinary Na(+) excretion in normokalemic but not in hypokalemic rats. These results indicate that ET-1 can mediate hypokalemic renal injury in two different ways: by directly stimulating ETA and by locally promoting endogenous ET-1 production via ETB. Thus, ETA as well as ETB blockade may be renoprotective in hypokalemic nephropathy.


Subject(s)
Endothelin Receptor Antagonists , Hypokalemia/complications , Kidney Diseases/etiology , Kidney Diseases/physiopathology , Pyrrolidines/pharmacology , Albuminuria/urine , Animals , Atrasentan , Endothelin-1/biosynthesis , Hypertension/chemically induced , Hypertrophy , Hypokalemia/metabolism , Kidney/metabolism , Kidney/pathology , Kidney Diseases/urine , Kidney Tubules/drug effects , Kidney Tubules/pathology , Male , Potassium/metabolism , Proteinuria/urine , Rats , Rats, Sprague-Dawley , Receptor, Endothelin A , Receptor, Endothelin B
18.
Endocrinology ; 143(9): 3515-21, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12193565

ABSTRACT

We previously reported that adrenomedullin produced by cardiac myocytes acts as a local modulator in some cardiac disorders. However, the role of adrenomedullin (AM) in cardiomyocyte apoptosis remains to be clarified. The present study investigated the effect of AM on doxorubicin-induced cardiac myocyte apoptosis. Doxorubicin increased the number of cells with pyknotic nuclei and lactate dehydrogenase release, and AM dose-dependently (10(-10)-10(-8)6 M) inhibited these increases produced by doxorubicin. Treatment with AM also suppressed doxorubicin-induced DNA fragmentation and caspase-3 activation. 8-Bromo-cAMP, a cAMP analog, mimicked these antiapoptotic effects of AM. An AM/calcitonin gene-related peptide (CGRP) receptor antagonist CGRP-(8-37) and a protein kinase A inhibitor H89 attenuated the antiapoptotic effect of AM. CGRP-(8-37) and H89 had no apoptotic effect alone, but accelerated doxorubicin-induced apoptosis. Under serum-free conditions, AM secretion into the culture medium and expression of AM mRNA were significantly increased after treatment with doxorubicin. Hydrogen peroxide scavenger catalase and antioxidant N-acetyl-L-cysteine inhibited the doxorubicin-mediated increase in AM secretion and its gene expression. These results indicate that AM inhibits doxorubicin-induced cardiac myocyte apoptosis through a cAMP-dependent mechanism and suggest that augmented production of AM by doxorubicin has an endogenous antiapoptotic effect. AM, as an autocrine factor, may play a protective role against cardiomyocyte injury by doxorubicin.


Subject(s)
Apoptosis/drug effects , Cardiotonic Agents/pharmacology , Cyclic AMP/pharmacology , Doxorubicin/pharmacology , Myocardium/cytology , Peptides/physiology , Sulfonamides , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Acetylcysteine/pharmacology , Adrenomedullin , Animals , Antineoplastic Agents/pharmacology , Antioxidants/pharmacology , Calcitonin Gene-Related Peptide/pharmacology , Calcitonin Gene-Related Peptide Receptor Antagonists , Catalase/pharmacology , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , DNA Fragmentation , Enzyme Inhibitors/pharmacology , Free Radical Scavengers/pharmacology , Humans , Isoquinolines/pharmacology , L-Lactate Dehydrogenase/metabolism , Peptide Fragments/pharmacology , Peptides/genetics , Peptides/metabolism , Peptides/pharmacology , Rats , Rats, Wistar , Recombinant Proteins/pharmacology
19.
Hypertens Res ; 25(1): 125-33, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11924718

ABSTRACT

Angiotensin II type 2 (AT2) receptor is developmentally regulated and exerts antiproliferative and proapoptotic actions. Genetic ablation of this receptor in mice affects regulation of blood pressure, but the involvement of the AT2 receptor in the pathogenesis of hypertension remains unknown. In the present study, we examined developmental changes of angiotensin receptor subtypes in the kidney of stroke-prone spontaneously hypertensive rats (SHRSP), and compared them with those in normotensive Wistar-Kyoto rats (WKY). We also investigated the regulation and functional role of the AT2 receptor in cultured mesangial cells. Receptor binding and Northern blot analyses revealed that AT2 receptor expression is significantly lower in the SHRSP kidney than in the WKY kidney during the perinatal period, while AT1 receptor expression is not different between them. In WKY mesangial cells, AT2 receptor stimulation exerted a potent antiproliferative effect; this effect was not observed in SHRSP cells lacking the AT2 receptor expression. The expression of interferon regulatory factor (IRF)-1 paralleled the growth-dependent induction of AT2 receptor in WKY mesangial cells, and transfection of IRF-1 antisense oligonucleotide significantly suppressed AT2 receptor expression, indicating IRF-1-dependent regulation of AT2 receptor expression in mesangial cells. However, this induction was inefficient in SHRSP cells. Thus, we found impaired AT2 receptor expression in the SHRSP kidney in vivo and in mesangial cells in vitro. The unbalanced expression of renal angiotensin receptor subtypes with exaggerated AT1 receptor signaling during early life in SHRSP may play a role in the programming for hypertension and related renal injury.


Subject(s)
Glomerular Mesangium/metabolism , Hypertension/metabolism , Kidney/metabolism , Rats, Inbred SHR/physiology , Receptors, Angiotensin/metabolism , Aging/metabolism , Animals , Blotting, Northern , Cell Division/physiology , Cells, Cultured , DNA-Binding Proteins/metabolism , Embryo, Mammalian/metabolism , Embryonic and Fetal Development , Genetic Predisposition to Disease , Glomerular Mesangium/embryology , Glomerular Mesangium/growth & development , Glomerular Mesangium/pathology , Hypertension/pathology , Interferon Regulatory Factor-1 , Kidney/embryology , Kidney/growth & development , Kidney/pathology , Phosphoproteins/metabolism , Promoter Regions, Genetic/genetics , Rats/embryology , Rats, Inbred SHR/genetics , Rats, Inbred WKY , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Receptors, Angiotensin/genetics , Stroke/genetics
20.
Kidney Int ; 61(3): 951-8, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11849449

ABSTRACT

BACKGROUND: Chronic potassium (K+) deficiency, one of the well-known causes of renal tubulointerstitial injury, is associated with an alteration in vasoactive mediators including persistent generation of renal cortical angiotensin (Ang) II despite the suppression of plasma Ang II, and suppression of urinary nitrite/nitrate excretion. We tested the hypothesis that K+-deficiency-induced renal tubulointerstitial injury could be mediated by Ang II or a reduction in nitric oxide. METHODS: Rats were fed a K+-deficient diet (0.01% K+) alone, or with either losartan or l-arginine (L-Arg) in drinking water. Control rats were fed with a normal K+ diet (0.36% K+). At the end of 10 weeks, kidneys were excised and renal injury was evaluated. RESULTS: Serum K+ was similarly depressed in all three groups receiving the K+-deficient diet. Rats on the K+-deficient diet alone developed renal hypertrophy and tubulointerstitial fibrosis with an increase in tubular osteopontin expression, macrophage infiltration and type III collagen deposition. Administration of losartan significantly reduced renal hypertrophy and prevented tubulointerstitial injury in the cortex, although some medullary injury occurred. In contrast, administration of L-Arg did not attenuate tubulointerstitial injury in the cortex, despite a complete recovery of urinary nitrate excretion. Mild but significant improvement of tubular osteopontin expression and macrophage infiltration were observed in the medulla of L-Arg-treated hypokalemic rats. CONCLUSIONS: These results indicate that hypokalemic renal injury is mediated, at least in part, by Ang II via the Ang II type 1 receptor, with a lesser contribution mediated by a reduction in nitric oxide. Losartan may be beneficial in preventing hypokalemic tubulointerstitial injury.


Subject(s)
Angiotensin Receptor Antagonists , Kidney/pathology , Potassium Deficiency/pathology , Animals , Arginine/pharmacology , Chronic Disease , Dinoprostone/urine , Kidney/drug effects , Losartan/pharmacology , Male , Nitrates/urine , Nitrites/urine , Potassium Deficiency/urine , Rats , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1
SELECTION OF CITATIONS
SEARCH DETAIL
...