Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Can J Physiol Pharmacol ; 93(11): 923-34, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26291649

ABSTRACT

Using the UM-X7.1 hereditary cardiomyopathic and muscular dystrophy hamsters (HCMH), we tested the effects of lifelong preventive or curative treatments during the heart failure phase with the NHE-1 inhibitor EMD 87580 (EMD) or with the angiotensin-converting enzyme inhibitor cilazapril on the intracellular Na(+) and Ca(2+) overloads, elevated level of NHE-1, necrosis, hypertrophy, heart failure, and early death. Our results showed that 310-day pretreatment of 30-day-old HCMHs with EMD significantly prevented cardiac necrosis, cardiomyocyte hypertrophy, and reduced the heart to body mass ratio. This treatment significantly prevented Na(+) and Ca(2+) overloads and the increase in NHE-1 protein level observed in HCMHs. Importantly, this lifelong preventive treatment significantly decreased the levels of creatine kinase and prevented early death of HCMHs. Curative treatment of hypertrophic 275-day-old HCMHs for 85 days with EMD significantly prevented hypertrophy and early death of HCMHs. However, treatments with cilazapril did not have any significant effects on the cardiac parameters studied or on early death of HCMHs. Our results suggest that the increase in the NHE-1 level and the consequent Na(+) and Ca(2+) overloads are implicated in the pathological process leading to heart failure and early death in HCMHs, and treatment with the NHE-1 inhibitor is promising for preventing early death in hereditary cardiomyopathy.


Subject(s)
Cardiomyopathies/drug therapy , Guanidines/pharmacology , Guanidines/therapeutic use , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Sulfones/pharmacology , Sulfones/therapeutic use , Animals , Cardiomyopathies/metabolism , Cardiomyopathies/mortality , Cricetinae , Death , Female , Male , Sodium-Hydrogen Exchangers/metabolism
2.
Langenbecks Arch Surg ; 399(5): 629-38, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24770930

ABSTRACT

OBJECTIVES: The purpose of this "first-in-man" study was to investigate the safety of a novel vascular polyester prosthesis coated with a resorbable polymer and free of any animal-based coating agents such as gelatin or collagen. METHODS: In a nonrandomized first-in-man multicenter safety study, the frequency of perigraft seroma (PGS) as the primary endpoint was studied in consecutive patients undergoing aortic reconstructions. The follow-up control to study the primary endpoint was intended at 3 months under routine clinical conditions. Pre- and postoperative white blood cell counts (WBC), C-reactive protein (CRP), and liver enzyme levels to characterize the systemic inflammation response and possible metabolic consequences were determined at different postoperative time points (secondary endpoints). Additionally, the primary unassisted patency rate, perioperative complications and serious adverse events, as well as intraoperative handling properties of the graft based on a semiquantitative scale were assessed. Magnetic resonance angiography (MRA) follow-up investigations were scheduled postoperatively at 3 months to determine graft tissue integration and the presence of PGS. RESULTS: A total of 24 patients with comorbidities such as coronary artery disease (8.3 %, 2/24), chronic occlusive pulmonary disease (COPD, 8.3 %, 2/24), Fontaine III/IV (20.8 %, 5/24), and diabetes (20.8 %, 5/24) were enrolled from June 2011 to September 2012. Due to two early nongraft-related deaths, there were 22 patients that had a follow-up. In these 22 patients, the freedom from PGS was 90.9 % (20/22) suggesting that the graft/tissue integration was comparable to other vascular grafts described in the relevant literature. WBC counts were not significantly different (pre (8.67 ± 2.98 1/nl) vs. follow-up (7.97 ± 2.24 1/nlI, p = 0.203). Likewise, preoperative CRP serum levels (6.47 ± 11.59 mg/l) were not different from those at follow-up (7.87 ± 12.81 mg/l, p = 0.769). There were two patients with a documented coagulation disorder and two premature deaths (cardiac failure, cerebral bleeding). The primary unassisted patency at follow-up was 77.3 % (17/22) in all patients who reached the follow-up (85.0 % or 17/20 if two cases with documented coagulation disorders are excluded). The reasons for occlusions were technical/surgical difficulties (2/5) and documented coagulation disorders (2/5). In one occlusion, the cause was unknown. There were no graft infections. Intraoperative graft handling properties were evaluated less favorable as compared to the routinely used gelatin- or collagen-coated polyester grafts in each investigator's clinical practice. CONCLUSIONS: Our results suggest that Uni-Graft® Synthetic is a promising prosthetic vascular graft to reduce PGS. Our findings should be interpreted with caution noting the limitation of the lack of a control group.


Subject(s)
Absorbable Implants , Blood Vessel Prosthesis Implantation/methods , Blood Vessel Prosthesis , Polymers/pharmacology , Prosthesis Design/methods , Aged , Angiography/methods , Aortic Aneurysm, Abdominal/diagnosis , Aortic Aneurysm, Abdominal/surgery , Aortic Aneurysm, Thoracic/diagnosis , Aortic Aneurysm, Thoracic/surgery , Blood Vessel Prosthesis Implantation/instrumentation , Coated Materials, Biocompatible , Female , Germany , Humans , Magnetic Resonance Angiography/methods , Male , Middle Aged , Patient Safety , Postoperative Complications/epidemiology , Postoperative Complications/physiopathology , Prosthesis Failure , Plastic Surgery Procedures/methods , Risk Assessment , Treatment Outcome , Ultrasonography, Doppler, Pulsed/methods , Vascular Patency/physiology , Vascular Surgical Procedures/adverse effects , Vascular Surgical Procedures/methods
3.
Cell Physiol Biochem ; 32(4): 838-48, 2013.
Article in English | MEDLINE | ID: mdl-24081014

ABSTRACT

BACKGROUND: The serum and glucocorticoid inducible kinase SGK1, which was originally cloned from mammary tumor cells, is highly expressed in some but not all tumors. SGK1 confers survival to several tumor cells. Along those lines, the number of colonic tumors following chemical carcinogenesis was decreased in SGK1 knockout mice. Recently, a highly selective SGK inhibitor (EMD638683) has been developed. The present study explored whether EMD638683 affects survival of colon carcinoma cells in vitro and impacts on development of colonic tumors in vivo. METHODS: Colon carcinoma (Caco-2) cells were exposed to EMD638683 with or without exposure to radiation (3 Gray) and cell volume was estimated from forward scatter, phosphatidylserine exposure from annexin V binding, mitochondrial potential from JC-9 fluorescence, caspase 3 activity from CaspGlow Fluorescein staining, DNA degradation from propidium iodide staining as well as late apoptosis from annexin-V FITC and propidium iodide double staining. In vivo tumor growth was determined in wild type mice subjected to chemical carcinogenesis (intraperitoneal injection of 20 mg/kg 1,2-dimethylhydrazine followed by three cycles of 30 g/L synthetic dextran sulfate sodium in drinking water for 7 days). RESULTS: EMD638683 treatment significantly augmented the radiation-induced decrease of forward scatter, increase of phosphatidylserine exposure, decrease of mitochondrial potential, increase of caspase 3 activity, increase of DNA fragmentation and increase of late apoptosis. The in vivo development of tumors following chemical carcinogenesis was significantly blunted by treatment with EMD638683. CONCLUSIONS: EMD638683 promotes radiation-induced suicidal death of colon tumor cells in vitro and decreases the number of colonic tumors following chemical carcinogenesis in vivo.


Subject(s)
Benzamides/pharmacology , Benzamides/therapeutic use , Colonic Neoplasms/drug therapy , Hydrazines/pharmacology , Hydrazines/therapeutic use , Immediate-Early Proteins/antagonists & inhibitors , Membrane Potential, Mitochondrial/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Apoptosis/drug effects , Caco-2 Cells , Caspase 3/metabolism , Flow Cytometry , Humans , Mice
4.
Exp Biol Med (Maywood) ; 237(2): 201-10, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22312057

ABSTRACT

Cardiovascular disease is characterized by impaired exercise capacity and endothelial dysfunction, i.e. reduced bioavailability of nitric oxide (NO). Phosphodiesterase-5 (PDE5) inhibition is a promising vasodilator therapy, but its effects on pulmonary and systemic hemodynamic responses to exercise in the absence, and particularly in the presence, of endothelial dysfunction have not been studied. We investigated the effects of PDE5 inhibitor EMD360527 in chronically instrumented swine at rest and during exercise with and without NO synthase inhibition (N(ω)-nitro-l-arginine; NLA). PDE5 inhibition caused a 19 ± 3% decrease in systemic vascular resistance (SVR) and a 24 ± 4% decrease in pulmonary vascular resistance (PVR) at rest. At maximal exercise, PDE5 inhibition caused a 13 ± 1% decrease in SVR and a 29 ± 3% decrease in PVR. NLA enhanced PDE5-inhibition-induced pulmonary (decrease in PVR 32 ± 12% at rest and 41 ± 3% during exercise) and systemic (decrease in SVR 24 ± 5% at rest and 18 ± 3% during exercise) vasodilation. Similarly, NLA increased the pulmonary and systemic vasodilation to nitroprusside and 8-bromo-cyclic guanosine monophosphate (cGMP), indicating that inhibition of NO synthase increases responsiveness to stimulation of the NO/cGMP pathway. Thus, PDE5 inhibition causes pulmonary and systemic vasodilation that is, respectively, maintained and slightly blunted during exercise. The degree of dilation in both the pulmonary and systemic beds were paradoxically enhanced in the presence of reduced bioavailability of NO, suggesting that this vasodilator therapy is most effective in patients with cardiovascular disease.


Subject(s)
Endothelial Cells/cytology , Phosphodiesterase 5 Inhibitors/pharmacology , Physical Conditioning, Animal , Animals , Cyclic GMP/analogs & derivatives , Cyclic GMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , Humans , Hypertension, Pulmonary/drug therapy , Nitric Oxide/chemistry , Nitric Oxide Synthase/metabolism , Nitroprusside/pharmacology , Pulmonary Circulation , Swine , Vascular Resistance
5.
Cell Physiol Biochem ; 28(1): 137-46, 2011.
Article in English | MEDLINE | ID: mdl-21865856

ABSTRACT

UNLABELLED: The serum- and glucocorticoid-inducible kinase 1 (SGK1) is transcriptionally upregulated by mineralocorticoids and activated by insulin. The kinase enhances renal tubular Na(+)-reabsorption and accounts for blood pressure increase following high salt diet in mice made hyperinsulinemic by dietary fructose or fat. The present study describes the in vitro and in vivo efficacy of a novel SGK1 inhibitor (EMD638683). EMD638683 was tested in vitro by determination of SGK1-dependent phosphorylation of NDRG1 (N-Myc downstream-regulated gene 1) in human cervical carcinoma HeLa-cells. In vivo EMD638683 (4460 ppm in chow, i.e. approx. 600 mg/kg/day) was administered to mice drinking tap water or isotonic saline containing 10% fructose. Blood pressure was determined by the tail cuff method, and urinary electrolyte (flame photometry) concentrations determined in metabolic cages. In vitro testing disclosed EMD638683 as a SGK1 inhibitor with an IC50 of 3 µM. Within 24 hours in vivo EMD638683 treatment significantly decreased blood pressure in fructose/saline-treated mice but not in control animals or in SGK1 knockout mice. EMD638683 failed to alter the blood pressure in SGK1 knockout mice. Following chronic (4 weeks) fructose/high salt treatment, additional EMD638683 treatment again decreased blood pressure. EMD638683 thus abrogates the salt sensitivity of blood pressure in hyperinsulinism without appreciably affecting blood pressure in the absence of hyperinsulinism. EMD638683 tended to increase fluid intake and urinary excretion of Na(+), significantly increased urinary flow rate and significantly decreased body weight. CONCLUSION: EMD638683 could serve as a template for drugs counteracting hypertension in individuals with type II diabetes and metabolic syndrome.


Subject(s)
Antihypertensive Agents/pharmacology , Benzamides/pharmacology , Hydrazines/pharmacology , Immediate-Early Proteins/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Antihypertensive Agents/chemistry , Antihypertensive Agents/therapeutic use , Benzamides/chemistry , Benzamides/therapeutic use , Blood Pressure/drug effects , Blood Pressure/physiology , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/metabolism , Fructose/pharmacology , HeLa Cells , Humans , Hydrazines/chemistry , Hydrazines/therapeutic use , Hypertension/drug therapy , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Knockout , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/therapeutic use , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Sodium/urine , Sodium Chloride/pharmacology
6.
Cell Physiol Biochem ; 24(3-4): 193-200, 2009.
Article in English | MEDLINE | ID: mdl-19710534

ABSTRACT

PPARgamma agonists, such as pioglitazone, are widely used in the treatment of diabetes and several further disorders. They enhance transcription of the serum and glucocorticoid inducible kinase SGK1, which could in turn enhance gastric acid secretion by stimulating KCNQ1 K+ channels. The present study explored whether pioglitazone upregulates SGK1 protein expression in gastric glands and thus modifies gastric acid secretion. Food containing the PPARgamma agonist pioglitazone (approximately 25 mg/kg bw/day) was administered to gene-targeted mice lacking SGK1 (sgk1-/-, n=11) and their wild-type littermates (sgk1+/+, n=11). Western blotting was employed to analyze SGK1 expression, BCECF-fluorescence to determine acid secretion in isolated gastric glands and immunohistochemistry to elucidate KCNQ1 and H+/K+-ATPase protein abundance in the parietal cell membrane. Pioglitazone significantly increased SGK1 expression. Cytosolic pH and cellular buffer capacity were not significantly different between sgk1+/+ and sgk1-/- mice and not significantly modified in either genotype by pioglitazone. Without pioglitazone treatment, Na+-independent pH-recovery following an ammonium pulse (DeltapH/min) reflecting H+/K+-ATPase activity was again similar in sgk1+/+ and sgk1-/- mice. Pioglitazone significantly increased DeltapH/min (approximately 3 fold) in sgk1+/+ but not in sgk1-/- mice. H+/K+-ATPase inhibitor omeprazole (100 microM) abolished DeltapH/min in both genotypes irrespective of pioglitazone treatment. Increase in local K+ concentrations to 35 mM (replacing Na+/NMDG) significantly increased DeltapH/min and abrogated the differences between genotypes. KCNQ1 and H+/K+-ATPase protein abundance in the parietal cell membrane was enhanced by pioglitazone treatment in sgk1+/+ but not in sgk1-/- mice. In conclusion, pioglitazone increases gastric acid secretion, an effect at least partially due to stimulation of SGK1 expression and SGK1-dependent upregulation of KCNQ1.


Subject(s)
Gastric Acid/metabolism , Gastric Mucosa/metabolism , Hypoglycemic Agents/pharmacology , Immediate-Early Proteins/deficiency , Protein Serine-Threonine Kinases/deficiency , Thiazolidinediones/pharmacology , Animals , Enzyme Inhibitors/pharmacology , Fluorescent Antibody Technique , Gastric Mucosa/drug effects , Gene Expression/drug effects , Hydrogen-Ion Concentration/drug effects , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Immunohistochemistry , KCNQ1 Potassium Channel/metabolism , Mice , Mice, Knockout , Omeprazole/pharmacology , PPAR gamma/agonists , Parietal Cells, Gastric/metabolism , Pioglitazone , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Sodium-Potassium-Exchanging ATPase/genetics , Sodium-Potassium-Exchanging ATPase/metabolism , Up-Regulation/drug effects
7.
Pflugers Arch ; 456(2): 425-36, 2008 May.
Article in English | MEDLINE | ID: mdl-18172605

ABSTRACT

PPARgamma-agonists enhance insulin sensitivity and improve glucose utilization in diabetic patients. Adverse effects of PPARgamma-agonists include volume retention and edema formation. Recent observations pointed to the ability of PPARgamma agonists to enhance transcription of the serum and glucocorticoid-inducible kinase SGK1, a kinase that is genomically upregulated by mineralocorticoids and stimulates various renal channels and transporters including the renal epithelial Na+ channel ENaC. SGK1 has been proposed to mediate the volume retention after treatment with PPARgamma agonists. To test this hypothesis, food containing the PPARgamma agonist pioglitazone (0.02%, i.e., approximately 25 mg/kg bw/day) was administered to gene-targeted mice lacking SGK1 (sgk1-/-, n=12) and their wild-type littermates (sgk1+/+), n=12). According to in situ hybridization, quantitative reverse transcriptase-polymerase chain reaction (RT-PCR) and immunofluorescence, treatment with pioglitazone significantly increased renal SGK1 mRNA and protein expression in sgk1+/+ mice. The treatment increased body weight significantly in both, sgk1+/+ mice (+2.2+/-0.3 g) and sgk-/- mice (+1.3+/-0.2 g), and decreased hematocrit significantly in sgk1+/+ mice (-6.5+/-1.0%) and sgk1-/- mice (-3.1+/-0.6%). Both effects were significantly (p<0.05) more pronounced in sgk1+/+ mice. According to Evans Blue distribution, pioglitazone increased plasma volume only in sgk1+/+ mice (from 50.9+/-3.9 to 63.7+/-2.5 microl/g bw) but not in sgk-/- mice (from 46.8+/-3.8 to 48.3+/-5.2 microl/g bw). Pioglitazone decreased aldosterone plasma levels and blood pressure and increased leptin plasma levels in both genotypes. We conclude that SGK1 contributes to but does not fully account for the volume retention during treatment with the PPARgamma agonist pioglitazone.


Subject(s)
Hypoglycemic Agents/pharmacology , Immediate-Early Proteins/metabolism , PPAR gamma/agonists , Plasma Volume/drug effects , Plasma Volume/physiology , Protein Serine-Threonine Kinases/metabolism , Thiazolidinediones/pharmacology , Animals , Epithelial Sodium Channels/metabolism , Immediate-Early Proteins/genetics , Kidney/enzymology , Kidney/physiology , Mice , Mice, Knockout , Mineralocorticoids/metabolism , Pioglitazone , Protein Serine-Threonine Kinases/genetics , RNA, Messenger/metabolism , Up-Regulation/drug effects
8.
J Biol Chem ; 282(39): 28501-28513, 2007 Sep 28.
Article in English | MEDLINE | ID: mdl-17686765

ABSTRACT

The human gene RSC1A1 codes for a 67-kDa protein named RS1 that mediates transcriptional and post-transcriptional regulation of Na(+)-D-glucose cotransporter SGLT1. The post-transcriptional regulation occurs at the trans-Golgi network (TGN). We identified two tripeptides in human RS1 (Gln-Cys-Pro (QCP) and Gln-Ser-Pro (QSP)) that induce posttranscriptional down-regulation of SGLT1 at the TGN leading to 40-50% reduction of SGLT1 in plasma membrane. For effective intracellular concentrations IC(50) values of 2.0 nM (QCP) and 0.16 nm (QSP) were estimated. Down-regulation of SGLT1 by tripeptides was attenuated by intracellular monosaccharides including non-metabolized methyl-alpha-D-glucopyranoside and 2-deoxyglucose. In small intestine post-transcriptional regulation of SGLT1 may contribute to glucose-dependent regulation of liver metabolism and intestinal mobility. QCP and QSP are transported by the H(+)-peptide cotransporter PepT1 that is colocated with SGLT1 in small intestinal enterocytes. Using coexpression of SGLT1 and PepT1 in Xenopus oocytes or polarized Caco-2 cells that contain both transporters we demonstrated that the tripeptides were effective when applied to the extracellular compartment. After a 1-h perfusion of intact rat small intestine with QSP, glucose absorption was reduced by 30%. The data indicate that orally applied tripeptides can be used to down-regulate small intestinal glucose absorption, e.g. in diabetes mellitus.


Subject(s)
Glucose/metabolism , Intestinal Absorption/drug effects , Monosaccharide Transport Proteins/metabolism , Oligopeptides/pharmacology , Protein Processing, Post-Translational/drug effects , Sodium-Glucose Transporter 1/metabolism , Animals , Antimetabolites/pharmacology , Biological Transport, Active/drug effects , Biological Transport, Active/physiology , Caco-2 Cells , Deoxyglucose/pharmacology , Diabetes Mellitus/drug therapy , Diabetes Mellitus/genetics , Diabetes Mellitus/metabolism , Female , Gastrointestinal Motility/drug effects , Gastrointestinal Motility/physiology , Gene Expression , Humans , Intestinal Absorption/physiology , Liver/metabolism , Male , Methylglucosides/pharmacology , Monosaccharide Transport Proteins/chemistry , Monosaccharide Transport Proteins/genetics , Oligopeptides/chemistry , Oligopeptides/therapeutic use , Oocytes/cytology , Peptide Transporter 1 , Protein Processing, Post-Translational/physiology , Rats , Rats, Wistar , Sodium-Glucose Transporter 1/genetics , Symporters/genetics , Symporters/metabolism , Xenopus laevis , trans-Golgi Network/metabolism
9.
Cardiovasc Drugs Ther ; 19(5): 315-22, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16382293

ABSTRACT

BACKGROUND: Ischemic preconditioning (IPC) elicits two distinct windows of cardioprotection, an early phase that lasts for 1-2 h and a delayed phase that lasts for 24-72 h. However, there is conflicting data as to how long the heart is resistant to IPC-induced cardioprotection after the initial protection wanes, leading to the demonstration of IPC-resistance. This resistance to IPC appears to be dependent on the timing of the next IPC stimulus, the species of animals used and the model studied. Furthermore, the mechanisms responsible IPC-resistance are unknown. It is also important to demonstrate therapeutic interventions that will produce cardioprotection during this period of IPC-resistance. METHODS AND RESULTS: To examine potential mechanisms responsible for acute IPC-induced resistance, the NHE-1 inhibitor EMD 85131 (2-methyl-5-methylsulfonyl-1-(1-pyrrollyl)-benzoylguanidine), which exerts its effects via mechanisms distinct from IPC, and the K(ATP) channel opener bimakalim, which bypasses the signaling mechanisms of IPC to directly open K(ATP) channels, were examined in a canine model of IPC-resistance. One 10 min. IPC stimulus followed by 10 min. of reperfusion produced a significant reduction in IS/AAR compared to Control (7.1 +/- 2.6% versus 26.0 +/- 6.2%; P < 0.05). However, IPC did not significantly protect the myocardium if a 2 h reperfusion period occurred between the initial IPC stimulus and the subsequent prolonged (60 min) ischemic challenge (IS/AAR: 22.5 +/- 4.8%: P > 0.05). Furthermore, hearts treated with IPC followed by 2 h of reperfusion were resistant to an additional IPC stimulus administered just prior to the subsequent 60 min. occlusion period (IS/AAR: 22.9 +/- 3.2%: P > 0.05). In contrast, administration of the NHE-1 inhibitor EMD 85131 (IS/AAR: 7.4 +/- 2.5%: P < 0.05) or the K(ATP) channel opener bimakalim (IS/AAR: 11.8 +/- 2.4%: P < 0.05) both afforded significant cardioprotection when administered at 2 h of reperfusion in previously preconditioned canine hearts resistant to IPC. CONCLUSIONS: IPC resistance occurs in this canine model of ischemia-reperfusion injury. However, in spite of IPC resistance, hearts can still be pharmacologically protected by direct application of the K(ATP) channel opener bimakalim or the NHE inhibitor EMD 85131.


Subject(s)
Cardiotonic Agents/therapeutic use , Myocardial Infarction/prevention & control , Reperfusion Injury/prevention & control , Animals , Benzamides/pharmacology , Benzamides/therapeutic use , Benzopyrans/pharmacology , Benzopyrans/therapeutic use , Cardiotonic Agents/pharmacology , Dihydropyridines/pharmacology , Dihydropyridines/therapeutic use , Disease Models, Animal , Dogs , Heart/drug effects , Ischemic Preconditioning, Myocardial , Myocardial Infarction/pathology , Potassium Channels/agonists , Pyrroles/pharmacology , Pyrroles/therapeutic use , Reperfusion Injury/pathology , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Time Factors
10.
J Mol Cell Cardiol ; 38(4): 571-82, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15808834

ABSTRACT

The purpose of this study was to verify whether myocardial intracellular Na(+) overload may take place in the hereditary cardiomyopathic hamster (CMH), as a result of an increased activity of the Na(+)-H(+) exchanger isoform-1 (NHE-1). Our results showed that simultaneous intracellular Na(+) and Ca(2+) overloads as well as an increase of NHE-1 protein level took place during the development of necrosis and hypertrophy in the CMH. Treatment of 30-day-old CMHs during the development of necrosis and in the absence of hypertrophy with the specific NHE-1 inhibitor EMD87580 (EMD) for 50 days significantly prevented the increase of NHE-1 protein level and intracellular Na(+) and Ca(2+) overloads as well as necrosis. Treatment of CMHs during the development of hypertrophy with EMD for 198 days prevented the development of both necrosis and hypertrophy. In conclusion, our results suggest that NHE-1 overexpression as well as Na(+) and Ca(2+) overloads do take place during the development of necrosis and hypertrophy in hereditary CMHs. Moreover, our results suggest that the blockade of NHE-1 by EMD87580 prevents these diseases by preventing the increase of Na(+) influx through the NHE-1.


Subject(s)
Calcium/metabolism , Cardiomyopathy, Hypertrophic/prevention & control , Guanidines/therapeutic use , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Sodium/metabolism , Sulfones/therapeutic use , Animals , Cardiomyopathy, Hypertrophic/metabolism , Cricetinae , Female , Guanidines/pharmacology , Male , Myocardium/pathology , Necrosis/prevention & control , Sodium-Hydrogen Exchangers/metabolism , Sulfones/pharmacology
11.
Life Sci ; 76(14): 1553-73, 2005 Feb 18.
Article in English | MEDLINE | ID: mdl-15680166

ABSTRACT

Energy intake and expenditure is a highly conserved and well-controlled system with a bias toward energy intake. In times of abundant food supply, individuals tend to overeat and in consequence to increase body weight, sometimes to the point of clinical obesity. Obesity is a disease that is not only characterized by enormous body weight but also by rising morbidity for diabetes type II and cardiovascular complications. To better understand the critical factors contributing to obesity we performed the present study in which the effects of energy expenditure and energy intake were examined with respect to body weight, localization of fat and insulin resistance in normal Wistar rats. It was found that a diet rich in fat and carbohydrates similar to "fast food" (cafeteria diet) has pronounced implication in the development of obesity, leading to significant body weight gain, fat deposition and also insulin resistance. Furthermore, an irregularly presented cafeteria diet (yoyo diet) has similar effects on body weight and fat deposition. However, these rats were not resistant to insulin, but showed an increased insulin secretion in response to glucose. When rats were fed with a specified high fat/carbohydrate diet (10% fat, 56.7% carbohydrate) ad lib or at the beginning of their activity phase they were able to detect the energy content of the food and compensate this by a lower intake. They, however, failed to compensate when food was given in the resting phase and gained more body weight as controls. Exercise, even of short duration, was able to keep rats on lower body weight and reduced fat deposition. Thus, inappropriate food intake with different levels of energy content is able to induce obesity in normal rats with additional metabolic changes that can be also observed in humans.


Subject(s)
Body Weight/physiology , Eating/physiology , Food , Insulin Resistance/physiology , Physical Conditioning, Animal/physiology , Physical Exertion/physiology , Adipose Tissue/pathology , Animals , Blood Glucose/metabolism , Caloric Restriction , Diet , Dietary Carbohydrates/pharmacology , Dietary Fats/pharmacology , Drinking , Glucose Tolerance Test , Housing, Animal , Insulin/blood , Lighting , Male , Obesity/pathology , Rats , Rats, Wistar , Social Isolation
12.
Biochem Biophys Res Commun ; 329(1): 275-80, 2005 Apr 01.
Article in English | MEDLINE | ID: mdl-15721303

ABSTRACT

Preferential expression of oncofetal extra domain-B fibronectin (EDB(+) FN), a proposed angiogenic marker, has been shown in proliferative diabetic retinopathy. High levels of glucose also increase EDB(+) FN expression in endothelial cells (ECs) via transforming growth factor-beta1 (TGF-beta1) and endothelin-1 (ET-1). The present study was aimed at elucidating the role of serum- and glucocorticoid-regulated kinase (SGK-1) in glucose-induced EDB(+) FN expression. Using human macro- and microvascular ECs, we show that high levels of glucose, TGF-beta1, and ET-1 increase the EDB(+) FN expression via SGK-1 alteration at the mRNA, protein, and activity levels. Inhibition of TGF-beta1 and ET-1 prevented glucose-induced SGK-1 activation and the EDB(+) FN expression. Furthermore, using siRNA-mediated SGK-1 gene silencing, we show that glucose-induced EDB(+) FN expression can be completely prevented. These findings provide first evidence of glucose-induced SGK-1 activation in altered EDB(+) FN expression and provide novel avenues for therapeutic modalities.


Subject(s)
Endothelium, Vascular/cytology , Fibronectins/biosynthesis , Fibronectins/metabolism , Glucose/metabolism , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Blotting, Western , Cells, Cultured , Diabetic Retinopathy , Endothelin-1/metabolism , Enzyme Activation , Fibronectins/chemistry , Gene Silencing , Humans , Immediate-Early Proteins , Immunoblotting , Microcirculation , Microscopy, Fluorescence , Protein Structure, Tertiary , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1 , Umbilical Veins/cytology
13.
BMC Pharmacol ; 4: 10, 2004 Jun 25.
Article in English | MEDLINE | ID: mdl-15219232

ABSTRACT

BACKGROUND: Type 4 phosphodiesterase (PDE4) inhibitors have been shown to stimulate bone formation in vivo and to stimulate osteoblastic differentiation in vitro. As one possible mechanism for the stimulation of bone formation is the recruitment of osteoprogenitor cells from the bone marrow, we have investigated the effect of the PDE4 inhibitors EMD273316, EMD95833, EMD249615 and EMD 219906 on fibroblastic colony formation by whole bone marrow cells and on the ability of these colonies to adopt an osteoblastic phenotype. RESULTS: All four agents stimulated colony formation in a concentration dependent manner, however, in the case of EMD273316 & EMD95833, the effect was evident at lower concentrations and the addition of prostaglandin E2 (PGE2) was not necessary for maximal stimulation. It was subsequently found that co-incubation with indomethacin reduced the stimulatory effects of EMD273316 & EMD95833 but had no effect on the actions of EMD249615 and EMD 219906 and that EMD273316 & EMD95833 stimulated the synthesis of endogenous PGE2 by whole bone marrow cells whereas EMD249615 and EMD 219906 had no significant effect. CONCLUSIONS: These data suggest that EMD249615, EMD 219906, EMD273316 & EMD95833 can promote the recruitment of bone marrow osteoprogenitor cells leading to a stimulation of bone formation via their direct inhibitory effects on PDE4. The actions of EMD273316 & EMD95833 however, are augmented by their ability to stimulate endogenous prostanoids synthesis which acts synergistically with their direct effects on PDE4.


Subject(s)
Bone Marrow Cells/drug effects , Fibroblasts/drug effects , Phosphodiesterase Inhibitors/pharmacology , Animals , Bone Marrow Cells/metabolism , Calcium/metabolism , Dinoprostone/metabolism , Fibroblasts/physiology , Male , Osteogenesis/drug effects , Prostaglandins/metabolism , Rats , Rats, Wistar
14.
J Cardiovasc Electrophysiol ; 15(4): 440-6, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15089994

ABSTRACT

INTRODUCTION: It has been suggested that blockade of the Na+/H+ exchanger (NHE1) can prevent atrial fibrillation (AF)-induced electrical remodeling and the development of AF. METHODS AND RESULTS: AF was maintained by burst pacing in 10 chronically instrumented conscious goats. Intravenous and oral dosages of two NHE1 blockers (EMD87580 and EMD125021) resulted in plasma levels several magnitudes higher than required for effective NHE1 blockade. Shortening of atrial refractoriness immediately after 5 minutes of AF was not prevented by NHE1 blockade. In remodeled atria, increasing dosages of EMD87580 and EMD125021 did not reverse shortening of the atrial refractory period or reduce the duration of AF episodes. The cycle length during persistent AF also was not affected. Oral pretreatment with EMD87580 (8 mg/kg bid) starting 3 days before AF could not prevent electrical remodeling. After 24 and 48 hours of remodeling, the duration of AF paroxysms was 47 +/- 32 seconds and 135 +/- 63 seconds compared to 56 +/- 17 seconds and 136 +/- 52 seconds in placebo-treated animals (P > 0.8), respectively. CONCLUSION: In the goat model of AF, the Na+/H+ exchanger inhibitors EMD87580 and EMD125021 did not prevent or revert AF-induced electrical remodeling. This indicates that activation of the Na+/H+ exchanger is not involved in the intracellular pathways of electrical remodeling. This does not support the suggestion that blockers of the Na+/H+ exchanger may be beneficial for prevention and treatment of AF.


Subject(s)
Atrial Fibrillation/physiopathology , Guanidines/administration & dosage , Heart Atria/drug effects , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Sulfones/administration & dosage , Ventricular Remodeling/drug effects , Animals , Atrial Fibrillation/prevention & control , Electrophysiologic Techniques, Cardiac , Female , Goats , Guanidines/pharmacokinetics , Heart Atria/physiopathology , Sulfones/pharmacokinetics , Ventricular Remodeling/physiology
15.
Am J Physiol Heart Circ Physiol ; 286(1): H381-7, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14684366

ABSTRACT

Sodium/hydrogen exchange (NHE) inhibitors show promise as potential therapeutic agents for the treatment of heart failure, but it is not known whether they can reverse the maladaptive remodeling that results in heart failure. We sought to determine the effect of the NHE-1-specific inhibitor EMD-87580 (EMD) on heart failure produced by myocardial infarction in the rat and to assess whether up to 4 wk of treatment delay results in beneficial effects. Male Sprague-Dawley rats were subjected to coronary artery ligation (or a sham procedure) and followed for up to 3 mo, at which time hypertrophy and hemodynamics were determined. EMD was provided in the diet, and treatment commenced immediately or 2-4 wk after ligation. EMD significantly reduced hemodynamic abnormalities, including the elevation in left ventricular end-diastolic pressure, and diminished the loss of systolic function with all treatment protocols. Left ventricular dilatation and hypertrophy, as assessed by heart weight, cell size, and atrial natriuretic peptide (ANP) expression, were similarly reversed to sham or near-sham levels. In addition, the increased plasma ANP and pro-ANP values were reversed to levels not significantly different from sham. Surprisingly, virtually all beneficial effects were identical with all treatment protocols. These effects were observed in the absence of infarct size reduction or blood pressure-lowering effects. Our results suggest that NHE-1 inhibition attenuates and reverses postinfarction remodeling and heart failure with a treatment delay of up to 4 wk after infarction. The effect is independent of infarct size or afterload reduction, indicating a direct effect on the myocardium.


Subject(s)
Cardiac Output, Low/prevention & control , Cardiac Output, Low/physiopathology , Cardiomegaly/prevention & control , Cardiomegaly/physiopathology , Myocardial Infarction/complications , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Animals , Body Weight/drug effects , Cardiac Output, Low/etiology , Cardiomegaly/etiology , Cardiomegaly/pathology , Cell Line , Coronary Disease/mortality , Guanidines/blood , Guanidines/pharmacology , Hemodynamics , Male , Mice , Myocardial Infarction/pathology , Myocardium/pathology , Myocytes, Cardiac , Osmolar Concentration , Rats , Rats, Sprague-Dawley , Sulfones/blood , Sulfones/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...