63 results on '"Galper JB"'
Search Results
2. Differential effects of statins (pravastatin or simvastatin) on ventricular ectopic complexes: Galpha(i2), a possible molecular marker for ventricular irritability.
- Author
-
Welzig CM, Park HJ, Naggar J, Confalone D, Rhofiry J, Shea J, Karas RH, Estes NA 3rd, Galper JB, Welzig, C Michael, Park, Ho-Jin, Naggar, Jack, Confalone, Deborah, Rhofiry, Joanne, Shea, Julie, Karas, Richard H, Estes, N A Mark 3rd, and Galper, Jonas B
- Abstract
Retrospective studies suggest that statins might exert an antiarrhythmic effect on the heart. The mechanism of this effect is unclear. Parasympathetic stimulation of the heart has been shown to protect against ventricular arrhythmias. The goal of this study was to determine the effect of statins on ventricular arrhythmias and its correlation with changes in parasympathetic responsiveness and Galpha(i2) expression. Patients were randomized to pravastatin and simvastatin in a double-blind crossover design. Ventricular arrhythmias were determined by analysis of 24-hour Holter recordings. Spectral RR interval analysis of Holter studies determined peak high-frequency power fraction, which reflects parasympathetic modulation of heart rate. Expression of Galpha(i2), a molecular component of the parasympathetic response pathway, was determined by Western blots of patients' lymphocytes. Pravastatin treatment decreased the incidence of ventricular premature complexes by 22.5 + or - 3.4% (n = 20, p <0.05), couplets, and runs of 3 to 6 beats of nonsustained ventricular tachycardia from 9.8 + or - 2.67 to 3.9 + or - 1.25 events/patient/24 hours (n = 12, p <0.05). Pravastatin increased peak high-frequency fraction by 29.8 + or - 4.3% (n = 33, p <0.001), while Galpha(i2) expression increased by 51.3 + or - 22.5% (n = 21, p <0.05). Effects of simvastatin on ventricular premature complexes and nonsustained ventricular tachycardia were not significant. Relative changes in couplets and nonsustained ventricular tachycardia in pravastatin-treated patients correlated negatively with changes in Galpha(i2) and high-frequency fraction (rho = -0.588 and rho = -0.763, respectively, n = 12, p <0.05). In conclusion, these data suggest that pravastatin might decrease cardiac irritability via an increase in parasympathetic responsiveness and that changes in Galpha(i2) expression might serve as a molecular marker for this effect, which might play a role in the molecular mechanism of the antiarrhythmic effect of statins. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
3. Toll-Like Receptor 2 Attenuates the Formation and Progression of Angiotensin II-Induced Abdominal Aortic Aneurysm in ApoE-/- Mice.
- Author
-
Zhang Y, Bagley J, Park HJ, Cao X, Maganto-Garcia E, Lichtman A, Beasley D, and Galper JB
- Abstract
Introduction: We demonstrated Toll-like receptor (TLR) 4 in the pathogenesis of angiotensin II (AngII)-mediated abdominal aortic aneurysm (AAA) formation. Here, we study TLR2 in the AAA formation., Methods: Male ApoE-/- and ApoE-/-TLR2-/- mice were treated with AngII. Mice were injected with the TLR2 agonist Pam3CSK4. The incidence and severity of AAA were determined. MCP-1, MCP-5, RANTES, CXCL10, CCR5, and CXCR3 were analyzed. M1 and M2 macrophages in the aorta were detected by flow cytometry., Results: These studies demonstrated an increase in AAA formation in TLR2-/- mice and a decrease by Pam3CSK4. Pam3CSK4 decreased the ratio of M1/M2 and the levels of RANTES, CXCL10, CCR5, and CXCR3. Furthermore, Pam3CSK4 treatment 1 week following AngII retarded the progression of AAA., Conclusion: These data demonstrated a protective effect of TLR2 signaling on AAA in association with a decrease in the ratio of M1 to M2 macrophages and the expression of chemokines and their receptors. Furthermore, the treatment of Pam3CSK4 after AngII demonstrated a marked retardation of lesion progression. Given the fact that most AAA patients are detected late in the disease process, these findings suggest that TLR2 stimulation may play a therapeutic role in retarding disease progression., (© 2024 The Author(s). Published by S. Karger AG, Basel.)
- Published
- 2024
- Full Text
- View/download PDF
4. Glycolysis regulated transglutaminase 2 activation in cardiopulmonary fibrogenic remodeling.
- Author
-
Bhedi CD, Nasirova S, Toksoz D, Warburton RR, Morine KJ, Kapur NK, Galper JB, Preston IR, Hill NS, Fanburg BL, and Penumatsa KC
- Subjects
- Animals, Carrier Proteins metabolism, Cell Proliferation, Fibroblasts metabolism, Glucose metabolism, Humans, Hyperglycemia metabolism, Membrane Proteins metabolism, Mice, Mice, Inbred C57BL, Protein Glutamine gamma Glutamyltransferase 2, Pulmonary Artery metabolism, Pyruvate Kinase metabolism, Signal Transduction, Thyroid Hormones metabolism, Up-Regulation, Thyroid Hormone-Binding Proteins, GTP-Binding Proteins genetics, GTP-Binding Proteins metabolism, Gene Expression Regulation, Enzymologic, Glycolysis, Hypertension, Pulmonary metabolism, Transglutaminases genetics, Transglutaminases metabolism
- Abstract
The pathophysiology of pulmonary hypertension (PH) and heart failure (HF) includes fibrogenic remodeling associated with the loss of pulmonary arterial (PA) and cardiac compliance. We and others have previously identified transglutaminase 2 (TG2) as a participant in adverse fibrogenic remodeling. However, little is known about the biologic mechanisms that regulate TG2 function. We examined physiological mouse models of experimental PH, HF, and type 1 diabetes that are associated with altered glucose metabolism/glycolysis and report here that TG2 expression and activity are elevated in pulmonary and cardiac tissues under all these conditions. We additionally used PA adventitial fibroblasts to test the hypothesis that TG2 is an intermediary between enhanced tissue glycolysis and fibrogenesis. Our in vitro results show that glycolytic enzymes and TG2 are upregulated in fibroblasts exposed to high glucose, which stimulates cellular glycolysis as measured by Seahorse analysis. We examined the relationship of TG2 to a terminal glycolytic enzyme, pyruvate kinase M2 (PKM2), and found that PKM2 regulates glucose-induced TG2 expression and activity as well as fibrogenesis. Our studies further show that TG2 inhibition blocks glucose-induced fibrogenesis and cell proliferation. Our findings support a novel role for glycolysis-mediated TG2 induction and tissue fibrosis associated with experimental PH, HF, and hyperglycemia., (© 2019 Federation of American Societies for Experimental Biology.)
- Published
- 2020
- Full Text
- View/download PDF
5. Targeted disruption of glycogen synthase kinase-3β in cardiomyocytes attenuates cardiac parasympathetic dysfunction in type 1 diabetic Akita mice.
- Author
-
Zhang Y, Welzig CM, Haburcak M, Wang B, Aronovitz M, Blanton RM, Park HJ, Force T, Noujaim S, and Galper JB
- Subjects
- Animals, Diabetes Mellitus, Type 1 enzymology, Diabetes Mellitus, Type 1 genetics, G Protein-Coupled Inwardly-Rectifying Potassium Channels metabolism, Glycogen Synthase Kinase 3 beta genetics, Glycogen Synthase Kinase 3 beta metabolism, Heart Atria innervation, Heart Atria physiopathology, Heart Rate physiology, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Myocytes, Cardiac physiology, Potassium Channels, Inwardly Rectifying metabolism, Diabetes Mellitus, Type 1 physiopathology, Glycogen Synthase Kinase 3 beta deficiency, Myocytes, Cardiac enzymology, Parasympathetic Nervous System physiopathology
- Abstract
Type 1 diabetic Akita mice develop severe cardiac parasympathetic dysfunction that we have previously demonstrated is due at least in part to an abnormality in the response of the end organ to parasympathetic stimulation. Specifically, we had shown that hypoinsulinemia in the diabetic heart results in attenuation of the G-protein coupled inward rectifying K channel (GIRK) which mediates the negative chronotropic response to parasympathetic stimulation due at least in part to decreased expression of the GIRK1 and GIRK4 subunits of the channel. We further demonstrated that the expression of GIRK1 and GIRK4 is under the control of the Sterol Regulatory element Binding Protein (SREBP-1), which is also decreased in response to hypoinsulinemia. Finally, given that hyperactivity of Glycogen Synthase Kinase (GSK)3β, had been demonstrated in the diabetic heart, we demonstrated that treatment of Akita mice with Li+, an inhibitor of GSK3β, increased parasympathetic responsiveness and SREBP-1 levels consistent with the conclusion that GSK3β might regulate IKACh via an effect on SREBP-1. However, inhibitor studies were complicated by lack of specificity for GSK3β. Here we generated an Akita mouse with cardiac specific inducible knockout of GSK3β. Using this mouse, we demonstrate that attenuation of GSK3β expression is associated with an increase in parasympathetic responsiveness measured as an increase in the heart rate response to atropine from 17.3 ± 3.5% (n = 8) prior to 41.2 ± 5.4% (n = 8, P = 0.017), an increase in the duration of carbamylcholine mediated bradycardia from 8.43 ± 1.60 min (n = 7) to 12.71 ± 2.26 min (n = 7, P = 0.028) and an increase in HRV as measured by an increase in the high frequency fraction from 40.78 ± 3.86% to 65.04 ± 5.64 (n = 10, P = 0.005). Furthermore, patch clamp measurements demonstrated a 3-fold increase in acetylcholine stimulated peak IKACh in atrial myocytes from GSK3β deficiency mice compared with control. Finally, western blot analysis of atrial extracts from knockout mice demonstrated increased levels of SREBP-1, GIRK1 and GIRK4 compared with control. Taken together with our prior observations, these data establish a role of increased GSK3β activity in the pathogenesis of parasympathetic dysfunction in type 1 diabetes via the regulation of IKACh and GIRK1/4 expression., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2019
- Full Text
- View/download PDF
6. QRS/T-wave and calcium alternans in a type I diabetic mouse model for spontaneous postmyocardial infarction ventricular tachycardia: A mechanism for the antiarrhythmic effect of statins.
- Author
-
Jin H, Welzig CM, Aronovitz M, Noubary F, Blanton R, Wang B, Rajab M, Albano A, Link MS, Noujaim SF, Park HJ, and Galper JB
- Subjects
- Animals, Calcium metabolism, Heart Ventricles diagnostic imaging, Mice, Mice, Inbred C57BL, Myocardial Infarction metabolism, Tachycardia, Ventricular etiology, Tachycardia, Ventricular physiopathology, Diabetes Mellitus, Experimental, Diabetes Mellitus, Type 1 complications, Electrocardiography, Heart Ventricles physiopathology, Hydroxymethylglutaryl-CoA Reductase Inhibitors therapeutic use, Myocardial Infarction complications, Tachycardia, Ventricular drug therapy
- Abstract
Background: The incidence of sudden arrhythmic death is markedly increased in diabetics., Objective: The purpose of this study was to develop a mouse model for postmyocardial infarction (post-MI) ventricular tachycardia (VT) in the diabetic heart and determine the mechanism of an antiarrhythmic effect of statins., Methods: ECG transmitters were implanted in wild-type (WT), placebo, and pravastatin-treated type I diabetic Akita mice. MIs were induced by coronary ligation, and Ca
2+ transients were studied by optical mapping, and Ca2+ transients and sparks in left ventricular myocytes (VM) by the Ionoptix system and confocal microscopy., Results: Burst pacing of Akita mouse hearts resulted in rate-related QRS/T-wave alternans, which was attenuated in pravastatin-treated mice. Post-MI Akita mice developed QRS/T-wave alternans and VT at 2820 ± 879 beats per mouse, which decreased to 343 ± 115 in pravastatin-treated mice (n = 13, P <.05). Optical mapping demonstrated pacing-induced VT originating in the peri-infarction zone and Ca2+ alternans, both attenuated in hearts of statin-treated mice. Akita VM displayed Ca2+ alternans, and triggered activity as well as increased Ca2+ transient decay time (Tau), Ca2+ sparks, and cytosolic Ca2+ and decreased SR Ca2+ stores all of which were in part reversed in cells from statin treated mice. Homogenates of Akita ventricles demonstrated decreased SERCA2a/PLB ratio and increased ratio of protein phosphatase (PP-1) to the PP-1 inhibitor PPI-1 which were reversed in homogenates of pravastatin-treated Akita mice., Conclusion: Pravastatin decreased the incidence of post-MI VT and Ca2+ alternans in Akita mouse hearts in part by revering abnormalities of Ca2+ handling via the PP-1/PPI-1 pathway., (Copyright © 2017 Heart Rhythm Society. Published by Elsevier Inc. All rights reserved.)- Published
- 2017
- Full Text
- View/download PDF
7. Angiotensin II-induced TLR4 mediated abdominal aortic aneurysm in apolipoprotein E knockout mice is dependent on STAT3.
- Author
-
Qin Z, Bagley J, Sukhova G, Baur WE, Park HJ, Beasley D, Libby P, Zhang Y, and Galper JB
- Subjects
- Angiotensin II toxicity, Animals, Aortic Aneurysm, Abdominal chemically induced, Aortic Aneurysm, Abdominal pathology, Apolipoproteins E genetics, Disease Models, Animal, Gene Expression Regulation, Humans, Macrophages metabolism, Macrophages pathology, Male, Mice, Mice, Knockout, STAT3 Transcription Factor biosynthesis, STAT3 Transcription Factor metabolism, Signal Transduction, Toll-Like Receptor 4 metabolism, Aortic Aneurysm, Abdominal genetics, STAT3 Transcription Factor genetics, Toll-Like Receptor 4 genetics
- Abstract
Abdominal Aortic Aneurysm (AAA) is a major cause of mortality and morbidity in men over 65 years of age. Male apolipoprotein E knockout (ApoE(-/-)) mice infused with angiotensin II (AngII) develop AAA. Although AngII stimulates both JAK/STAT and Toll-like receptor 4 (TLR4) signaling pathways, their involvement in AngII mediated AAA formation is unclear. Here we used the small molecule STAT3 inhibitor, S3I-201, the TLR4 inhibitor Eritoran and ApoE(-/-)TLR4(-/-) mice to evaluate the interaction between STAT3 and TLR4 signaling in AngII-induced AAA formation. ApoE(-/-) mice infused for 28 days with AngII developed AAAs and increased STAT3 activation and TLR4 expression. Moreover, AngII increased macrophage infiltration and the ratio of M1 (pro-inflammatory)/M2 (healing) macrophages in aneurysmal tissue as early as 7-10 days after AngII infusion. STAT3 inhibition with S3I-201 decreased the incidence and severity of AngII-induced AAA formation and decreased MMP activity and the ratio of M1/M2 macrophages. Furthermore, AngII-mediated AAA formation, MMP secretion, STAT3 phosphorylation and the ratio of M1/M2 macrophages were markedly decreased in ApoE(-/-)TLR4(-/-) mice, and in Eritoran-treated ApoE(-/-) mice. TLR4 and pSTAT3 levels were also increased in human aneurysmal tissue. These data support a role of pSTAT3 in TLR4 dependent AAA formation and possible therapeutic roles for TLR4 and/or STAT3 inhibition in AAA., (Copyright © 2015 Elsevier Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
8. Glycogen synthase kinase-3β inhibition ameliorates cardiac parasympathetic dysfunction in type 1 diabetic Akita mice.
- Author
-
Zhang Y, Welzig CM, Picard KL, Du C, Wang B, Pan JQ, Kyriakis JM, Aronovitz MJ, Claycomb WC, Blanton RM, Park HJ, and Galper JB
- Subjects
- Animals, Blotting, Western, Cells, Cultured, Diabetes Mellitus, Type 1 physiopathology, Diabetic Neuropathies physiopathology, Electrocardiography, Glycogen Synthase Kinase 3 beta, Heart Atria physiopathology, Mice, Mice, Mutant Strains, Parasympathetic Nervous System physiopathology, Patch-Clamp Techniques, Sterol Regulatory Element Binding Protein 1 metabolism, Diabetes Mellitus, Type 1 metabolism, Diabetic Neuropathies metabolism, G Protein-Coupled Inwardly-Rectifying Potassium Channels metabolism, Glycogen Synthase Kinase 3 metabolism, Heart Rate, Myocytes, Cardiac metabolism, Parasympathetic Nervous System metabolism, Sterol Regulatory Element Binding Protein 1 drug effects
- Abstract
Decreased heart rate variability (HRV) is a major risk factor for sudden death and cardiovascular disease. We previously demonstrated that parasympathetic dysfunction in the heart of the Akita type 1 diabetic mouse was due to a decrease in the level of the sterol response element-binding protein (SREBP-1). Here we demonstrate that hyperactivity of glycogen synthase kinase-3β (GSK3β) in the atrium of the Akita mouse results in decreased SREBP-1, attenuation of parasympathetic modulation of heart rate, measured as a decrease in the high-frequency (HF) fraction of HRV in the presence of propranolol, and a decrease in expression of the G-protein coupled inward rectifying K(+) (GIRK4) subunit of the acetylcholine (ACh)-activated inward-rectifying K(+) channel (IKACh), the ion channel that mediates the heart rate response to parasympathetic stimulation. Treatment of atrial myocytes with the GSK3β inhibitor Kenpaullone increased levels of SREBP-1 and expression of GIRK4 and IKACh, whereas a dominant-active GSK3β mutant decreased SREBP-1 and GIRK4 expression. In Akita mice treated with GSK3β inhibitors Li(+) and/or CHIR-99021, Li(+) increased IKACh, and Li(+) and CHIR-99021 both partially reversed the decrease in HF fraction while increasing GIRK4 and SREBP-1 expression. These data support the conclusion that increased GSK3β activity in the type 1 diabetic heart plays a critical role in parasympathetic dysfunction through an effect on SREBP-1, supporting GSK3β as a new therapeutic target for diabetic autonomic neuropathy., (© 2014 by the American Diabetes Association.)
- Published
- 2014
- Full Text
- View/download PDF
9. Increased inducibility of ventricular tachycardia and decreased heart rate variability in a mouse model for type 1 diabetes: effect of pravastatin.
- Author
-
Rajab M, Jin H, Welzig CM, Albano A, Aronovitz M, Zhang Y, Park HJ, Link MS, Noujaim SF, and Galper JB
- Subjects
- Animals, Disease Models, Animal, Heart Rate drug effects, Hydroxymethylglutaryl-CoA Reductase Inhibitors therapeutic use, Mice, Pravastatin therapeutic use, Tachycardia, Ventricular drug therapy, Diabetes Mellitus, Type 1 physiopathology, Heart Rate physiology, Hydroxymethylglutaryl-CoA Reductase Inhibitors pharmacology, Pravastatin pharmacology, Tachycardia, Ventricular physiopathology
- Abstract
Although a reduction in the high-frequency (HF) component of heart rate variability (HRV) is a major complication of diabetes and a risk factor for sudden death, its relationship to ventricular tachycardia (VT) is unknown. We developed a mouse model for the study of VT and its relationship to changes in HRV in the Akita type 1 diabetic mouse. Programmed ventricular stimulation of anesthetized mice demonstrated that Akita mice were more inducible for VT compared with wild-type mice: 78.6% versus 28.6% (P = 0.007). Optical mapping of perfused hearts demonstrated multifocal breakthroughs that occasionally gave rise to short-lived rotors consistent with focal initiation and maintenance of VT. Treatment of Akita mice with pravastatin, which had been previously shown clinically to decrease ventricular ectopy and to increase HRV, decreased the inducibility of VT: 36.8% compared with 75.0% with placebo treatment (P = 0.022). The HF fraction of HRV was reduced in Akita mice (48.6 ± 5.2% vs. 70.9 ± 4.8% in wild-type mice, P = 0.005) and was increased compared with placebo treatment in pravastatin-treated mice. Pretreatment of Akita mice with the muscarinic agonist carbamylcholine or the β-adrenergic receptor blocker propranolol decreased the inducibility of VT (P = 0.001). In conclusion, the increased inducibility of focally initiated VT and reduced HF fraction in Akita mice were partially reversed by both pravastatin treatment and pharmacologic reversal of parasympathetic dysfunction. In this new animal model for the study of the pathogenesis of VT in type 1 diabetes, pravastatin may play a role in the prevention of VT by attenuating parasympathetic dysfunction.
- Published
- 2013
- Full Text
- View/download PDF
10. Phenylephrine as a simulated intravascular epidural test dose in pediatrics: a pilot study.
- Author
-
Pancaro C, Nasr VG, Paulus JK, Bonney I, Flores AF, Galper JB, and Ahmed I
- Subjects
- Blood Pressure drug effects, Child, Child, Preschool, Dose-Response Relationship, Drug, Electrocardiography drug effects, Female, Heart Rate drug effects, Hemodynamics drug effects, Humans, Infant, Male, Monitoring, Intraoperative, Pilot Projects, Sample Size, Sevoflurane, Anesthesia, Epidural methods, Anesthesia, Inhalation methods, Anesthetics, Inhalation administration & dosage, Anesthetics, Local administration & dosage, Methyl Ethers administration & dosage, Phenylephrine, Vasoconstrictor Agents
- Abstract
Background: A test dose is used to detect intravascular injection during neuraxial block in pediatrics. Accidental intravascular epidural local anesthetic injection might be unrecognized in anesthetized children leading to potential life-threatening complications. In children, sevoflurane anesthesia blunts the hemodynamic response when intravascular cathecolamines are administered. No studies have explored the hemodynamics and the criteria for a positive test dose result following phenylephrine in sevoflurane anesthetized children., Methods: Healthy children undergoing minor procedures were randomly assigned to receive intravenous placebo, or 5 μg∙kg(-1) phenylephrine (n = 11/group) during sevoflurane anesthesia. Hemodynamic response was assessed using electrocardiography, pulse oxymetry and non-invasive blood pressure monitoring for 5 min following drug administration in anesthetized patients., Results: All patients receiving phenylephrine showed a decreased heart rate (HR) but not all of them met the positive criterion for test dose response. Overall, at 1 min, patients receiving phenylephrine showed a 25% decrease in HR from the baseline while an increase in blood pressure was noticed in 54% of patients receiving phenylephrine., Discussion: Phenylephrine might be a future indicator of positive intravascular test dose. Further investigation is needed to find out the phenylephrine dose that elicits a reliable hemodynamic response and whether phenylephrine needs to be dose age-adjusted in order to appreciate relevant hemodynamic changes in children receiving neuraxial blocks undergoing general anesthesia., (© 2013 Blackwell Publishing Ltd.)
- Published
- 2013
- Full Text
- View/download PDF
11. Role of toll-like receptor 4 in intimal foam cell accumulation in apolipoprotein E-deficient mice.
- Author
-
Higashimori M, Tatro JB, Moore KJ, Mendelsohn ME, Galper JB, and Beasley D
- Subjects
- Acetyl-CoA C-Acetyltransferase metabolism, Animals, Aorta, Thoracic metabolism, Aorta, Thoracic pathology, Aortic Diseases genetics, Aortic Diseases pathology, Apolipoproteins E genetics, Atherosclerosis genetics, Atherosclerosis pathology, Chemokine CCL2 genetics, Cholesterol blood, Disease Models, Animal, Female, Foam Cells pathology, Gene Expression Regulation, Inflammation Mediators metabolism, Interleukin-1alpha blood, Interleukin-1alpha genetics, Male, Mice, Mice, Knockout, Myocytes, Smooth Muscle metabolism, Myocytes, Smooth Muscle pathology, RNA, Messenger metabolism, Time Factors, Toll-Like Receptor 2 genetics, Toll-Like Receptor 2 metabolism, Toll-Like Receptor 4 deficiency, Toll-Like Receptor 4 genetics, Triglycerides blood, Vascular Cell Adhesion Molecule-1 genetics, Aortic Diseases metabolism, Apolipoproteins E deficiency, Atherosclerosis metabolism, Foam Cells metabolism, Signal Transduction, Toll-Like Receptor 4 metabolism
- Abstract
Objective: Atherosclerosis encompasses a conspicuously maladaptive inflammatory response that might involve innate immunity. Here, we compared the role of Toll-like receptor 4 (TLR4) with that of TLR2 in intimal foam cell accumulation and inflammation in apolipoprotein E (ApoE) knockout (KO) mice in vivo and determined potential mechanisms of upstream activation and downstream action., Methods and Results: We measured lipid accumulation and gene expression in the lesion-prone lesser curvature of the aortic arch. TLR4 deficiency reduced intimal lipid by ≈75% in ApoE KO mice, despite unaltered total serum cholesterol and triglyceride levels, whereas TLR2 deficiency reduced it by ≈45%. TLR4 deficiency prevented the increased interleukin-1α (IL-1α) and monocyte chemoattractant protein-1 mRNA levels seen within lesional tissue, and it also lowered serum IL-1α levels. Smooth muscle cells (SMC) were present within the intima of the lesser curvature of the aortic arch at this early lesion stage, and they enveloped and permeated nascent lesions, which consisted of focal clusters of foam cells. Cholesterol enrichment of SMC in vitro stimulated acyl-coenzyme A:cholesterol acyltransferase-1 mRNA expression, cytoplasmic cholesterol ester accumulation, and monocyte chemoattractant protein-1 mRNA and protein expression in a TLR4-dependent manner., Conclusions: TLR4 contributes to early-stage intimal foam cell accumulation at lesion-prone aortic sites in ApoE KO mice, as does TLR2 to a lesser extent. Intimal SMC surround and penetrate early lesions, where TLR4 signaling within them may influence lesion progression.
- Published
- 2011
- Full Text
- View/download PDF
12. Simvastatin inhibits angiotensin II-induced abdominal aortic aneurysm formation in apolipoprotein E-knockout mice: possible role of ERK.
- Author
-
Zhang Y, Naggar JC, Welzig CM, Beasley D, Moulton KS, Park HJ, and Galper JB
- Subjects
- Angiotensin II, Animals, Aortic Aneurysm, Abdominal chemically induced, Aortic Aneurysm, Abdominal metabolism, Aortic Aneurysm, Abdominal pathology, Apolipoproteins E metabolism, Apolipoproteins E pharmacology, Benzamides pharmacology, Blood Pressure drug effects, Blotting, Western, Disease Models, Animal, Immunohistochemistry, Matrix Metalloproteinase 2 metabolism, Matrix Metalloproteinase 9 metabolism, Mice, Mice, Inbred C57BL, Mice, Knockout, Random Allocation, Reference Values, Renin-Angiotensin System drug effects, Aortic Aneurysm, Abdominal prevention & control, Hydroxymethylglutaryl-CoA Reductase Inhibitors pharmacology, MAP Kinase Kinase Kinase 3 antagonists & inhibitors, Simvastatin pharmacology
- Abstract
Objective: Abdominal aortic aneurysm (AAA) is a life-threatening disease affecting almost 10% of the population over age 65. Generation of AAAs by infusion of angiotensin (Ang) II in apolipoprotein E-knockout (ApoE(-/-)) mice is an animal model which supports an imbalance of the renin-angiotensin system in the pathogenesis of AAA. The effect of statins on AngII-mediated AAA formation and the associated neovascularization is not known. Here we determined the effect of simvastatin and the ERK inhibitor, CI1040, on AngII-stimulated AAA formation., Methods and Results: ApoE(-/-) mice infused for 28 days with AngII using osmotic minipumps were treated with placebo, 10 mg/kg/d simvastatin, or 100 mg/kg/d CI1040. 95% of AngII-treated mice developed AAA with neovascularization of the lesion, increased ERK phosphorylation, MCP-1 secretion, and MMP activity. These effects were markedly reversed by simvastatin and in part by CI1040. Furthermore, simvastatin and the ERK inhibitor U0126 reversed AngII-stimulated angiogenesis and MMP secretion by human umbilical vein endothelial cells., Conclusions: These data support the conclusion that simvastatin interferes with AAA formation induced by AngII in ApoE(-/-) mice at least in part via ERK inhibition.
- Published
- 2009
- Full Text
- View/download PDF
13. Role of SREBP-1 in the development of parasympathetic dysfunction in the hearts of type 1 diabetic Akita mice.
- Author
-
Park HJ, Zhang Y, Du C, Welzig CM, Madias C, Aronovitz MJ, Georgescu SP, Naggar I, Wang B, Kim YB, Blaustein RO, Karas RH, Liao R, Mathews CE, and Galper JB
- Subjects
- Animals, Carbachol pharmacology, Cells, Cultured, Chick Embryo, Cholinergic Agents pharmacology, Diabetes Mellitus, Type 1 pathology, Diabetic Neuropathies pathology, Disease Models, Animal, G Protein-Coupled Inwardly-Rectifying Potassium Channels genetics, G Protein-Coupled Inwardly-Rectifying Potassium Channels metabolism, Heart Atria metabolism, Heart Atria pathology, Heart Ventricles metabolism, Heart Ventricles pathology, Insulin metabolism, Insulin pharmacology, Male, Mice, Mice, Mutant Strains, Myocardium metabolism, Myocardium pathology, Myocytes, Cardiac drug effects, Myocytes, Cardiac metabolism, Myocytes, Cardiac pathology, Parasympathetic Nervous System drug effects, Parasympathetic Nervous System metabolism, Patch-Clamp Techniques, Proinsulin metabolism, Sterol Regulatory Element Binding Protein 1 genetics, Diabetes Mellitus, Type 1 metabolism, Diabetic Neuropathies metabolism, Heart innervation, Parasympathetic Nervous System physiopathology, Sterol Regulatory Element Binding Protein 1 metabolism
- Abstract
Rationale: Diabetic autonomic neuropathy (DAN), a major complication of diabetes mellitus, is characterized, in part, by impaired cardiac parasympathetic responsiveness. Parasympathetic stimulation of the heart involves activation of an acetylcholine-gated K+ current, I(KAch), via a (GIRK1)2/(GIRK4)2 K+ channel. Sterol regulatory element binding protein-1 (SREBP-1) is a lipid-sensitive transcription factor., Objective: We describe a unique SREBP-1-dependent mechanism for insulin regulation of cardiac parasympathetic response in a mouse model for DAN., Methods and Results: Using implantable EKG transmitters, we demonstrated that compared with wild-type, Ins2(Akita) type I diabetic mice demonstrated a decrease in the negative chronotropic response to carbamylcholine characterized by a 2.4-fold decrease in the duration of bradycardia, a 52+/-8% decrease in atrial expression of GIRK1 (P<0.01), and a 31.3+/-2.1% decrease in SREBP-1 (P<0.05). Whole-cell patch-clamp studies of atrial myocytes from Akita mice exhibited a markedly decreased carbamylcholine stimulation of I(KAch) with a peak value of -181+/-31 pA/pF compared with -451+/-62 pA/pF (P<0.01) in cells from wild-type mice. Western blot analysis of extracts of Akita mice demonstrated that insulin treatment increased the expression of GIRK1, SREBP-1, and I(KAch) activity in atrial myocytes from these mice to levels in wild-type mice. Insulin treatment of cultured atrial myocytes stimulated GIRK1 expression 2.68+/-0.12-fold (P<0.01), which was reversed by overexpression of dominant negative SREBP-1. Finally, adenoviral expression of SREBP-1 in Akita atrial myocytes reversed the impaired I(KAch) to levels in cells from wild-type mice., Conclusions: These results support a unique molecular mechanism for insulin regulation of GIRK1 expression and parasympathetic response via SREBP-1, which might play a role in the pathogenesis of DAN in response to insulin deficiency in the diabetic heart.
- Published
- 2009
- Full Text
- View/download PDF
14. Parasympathetic response in chick myocytes and mouse heart is controlled by SREBP.
- Author
-
Park HJ, Georgescu SP, Du C, Madias C, Aronovitz MJ, Welzig CM, Wang B, Begley U, Zhang Y, Blaustein RO, Patten RD, Karas RH, Van Tol HH, Osborne TF, Shimano H, Liao R, Link MS, and Galper JB
- Subjects
- Acetylcholine genetics, Acetylcholine metabolism, Animals, Cells, Cultured, Chickens, G Protein-Coupled Inwardly-Rectifying Potassium Channels genetics, G Protein-Coupled Inwardly-Rectifying Potassium Channels metabolism, Heart Atria innervation, Heart Atria metabolism, Heart Atria pathology, Ion Transport genetics, Lipoproteins metabolism, Mice, Mice, Knockout, Myocardial Infarction genetics, Myocardial Infarction metabolism, Myocardial Infarction pathology, Myocardium pathology, Myocytes, Cardiac pathology, Parasympathetic Nervous System pathology, Potassium metabolism, Response Elements genetics, Sterol Regulatory Element Binding Protein 1 genetics, Tachycardia, Ventricular genetics, Tachycardia, Ventricular metabolism, Tachycardia, Ventricular pathology, Transcription, Genetic genetics, Ventricular Fibrillation genetics, Ventricular Fibrillation metabolism, Ventricular Fibrillation pathology, Lipid Metabolism genetics, Myocardium metabolism, Myocytes, Cardiac metabolism, Parasympathetic Nervous System metabolism, Sterol Regulatory Element Binding Protein 1 metabolism
- Abstract
Parasympathetic stimulation of the heart, which provides protection from arrhythmias and sudden death, involves activation of the G protein-coupled inward rectifying K+ channel GIRK1/4 and results in an acetylcholine-sensitive K+ current, I KACh. We describe a unique relationship between lipid homeostasis, the lipid-sensitive transcription factor SREBP-1, regulation of the cardiac parasympathetic response, and the development of ventricular arrhythmia. In embryonic chick atrial myocytes, lipid lowering by culture in lipoprotein-depleted serum increased SREBP-1 levels, GIRK1 expression, and I KACh activation. Regulation of the GIRK1 promoter by SREBP-1 and lipid lowering was dependent on interaction with 2 tandem sterol response elements and an upstream E-box motif. Expression of dominant negative SREBP-1 (DN-SREBP-1) reversed the effect of lipid lowering on I KACh and GIRK1. In SREBP-1 knockout mice, both the response of the heart to parasympathetic stimulation and the expression of GIRK1 were reduced compared with WT. I KACh, attenuated in atrial myocytes from SREBP-1 knockout mice, was stimulated by SREBP-1 expression. Following myocardial infarction, SREBP-1 knockout mice were twice as likely as WT mice to develop ventricular tachycardia in response to programmed ventricular stimulation. These results demonstrate a relationship between lipid metabolism and parasympathetic response that may play a role in arrhythmogenesis.
- Published
- 2008
- Full Text
- View/download PDF
15. Simvastatin potentiates tumor necrosis factor alpha-mediated apoptosis of human vascular endothelial cells via the inhibition of the geranylgeranylation of RhoA.
- Author
-
Tang D, Park HJ, Georgescu SP, Sebti SM, Hamilton AD, and Galper JB
- Subjects
- Apoptosis, Cell Membrane enzymology, Cells, Cultured, Diterpenes pharmacology, E-Selectin metabolism, Endothelial Cells enzymology, Humans, Mutation, Receptors, Tumor Necrosis Factor, Type I antagonists & inhibitors, Receptors, Tumor Necrosis Factor, Type I metabolism, Umbilical Veins cytology, Up-Regulation, rhoA GTP-Binding Protein analysis, rhoA GTP-Binding Protein genetics, Endothelial Cells drug effects, Hydroxymethylglutaryl-CoA Reductase Inhibitors pharmacology, Protein Prenylation drug effects, Simvastatin pharmacology, Tumor Necrosis Factor-alpha pharmacology, rhoA GTP-Binding Protein metabolism
- Abstract
HMG-CoA reductase inhibitors (statins) are widely used in the treatment and prevention of atherosclerosis. Here we demonstrate that the HMG-CoA reductase inhibitor simvastatin potentiates TNFalpha-mediated apoptosis and TNFalpha signaling in human umbilical vein endothelial cells (HUVECs). While 2.5 microM simvastatin or 40 ng/ml TNFalpha alone had only a small effect on apoptosis in HUVECs, co-incubation with simvastatin and TNFalpha markedly increased apoptosis in a time- and dose-dependent manner as measured by FACS analysis of propidium iodide-stained cells. Geranylgeraniol, which serves as a substrate for the geranylgeranylation of small GTP binding proteins such as RhoA, which is required for the function and membrane localization of Rho, reversed the effect of simvastatin on apoptosis. GGTI, an inhibitor of protein geranylgeranylation, mimicked the effect of simvastatin on apoptosis and interfered with the membrane localization of RhoA. Furthermore, simvastatin increased the expression of the TNFalpha type I receptor (TNFalphaRI) with a dose dependence and a dependence on geranylgeranylation similar to that demonstrated for the potentiation of TNFalpha-mediated apoptosis. Adenoviral expression of a dominant-negative RhoA mimicked the effect of simvastatin on the expression of TNFalphaRI, while adenoviral expression of a dominant-activating RhoA mutant reversed the effect of simvastatin on the expression of TNFalphaRI. Simvastatin also potentiated TNFalpha signaling as determined by increased TNFalpha-mediated E-selectin expression. These data support the conclusion that TNFalpha signaling is under the negative control of RhoA and that statins potentiate TNFalpha signaling at least in part via interference with RhoA inhibition of TNFalpha type I receptor expression.
- Published
- 2006
- Full Text
- View/download PDF
16. Transforming growth factor beta regulates the expression of the M2 muscarinic receptor in atrial myocytes via an effect on RhoA and p190RhoGAP.
- Author
-
Park HJ, Ward SM, Desgrosellier JS, Georgescu SP, Papageorge AG, Zhuang X, Barnett JV, and Galper JB
- Subjects
- Amides pharmacology, Animals, Cells, Cultured, Chick Embryo, Enzyme Inhibitors pharmacology, Gene Expression Regulation drug effects, Heart Atria embryology, Muscle Cells drug effects, Promoter Regions, Genetic drug effects, Pyridines pharmacology, Signal Transduction, Carrier Proteins metabolism, Muscle Cells physiology, Receptor, Muscarinic M2 genetics, Transforming Growth Factor beta pharmacology, rhoA GTP-Binding Protein metabolism
- Abstract
Transforming growth factor beta (TGFbeta) signaling is involved in the development and regulation of multiple organ systems and cellular signaling pathways. We recently demonstrated that TGFbeta regulates the response of atrial myocytes to parasympathetic stimulation. Here, TGFbeta(1) is shown to inhibit expression of the M(2) muscarinic receptor (M(2)), which plays a critical role in the parasympathetic response of the heart. This effect is mimicked by overexpression of a dominant negative mutant of RhoA and by the RhoA kinase inhibitor Y27632, whereas adenoviral expression of a dominant activating-RhoA reverses TGFbeta inhibition of M(2) expression. TGFbeta(1) also mediates a decrease in GTP-bound RhoA and a reciprocal increase in the expression of the RhoA GTPase-activating protein, p190RhoGAP, whereas total RhoA is unchanged. Inhibition of M(2) promoter activity by TGFbeta(1) is mimicked by overexpression of p190RhoGAP, whereas a dominant negative mutant of p190RhoGAP reverses this effect of TGFbeta(1). In contrast to atrial myocytes, in mink lung epithelial cells, in which TGFbeta signaling through activation of RhoA has been previously identified, TGFbeta(1) stimulated an increase in GTP-bound RhoA in association with a reciprocal decrease in the expression of p190RhoGAP. Both effects demonstrated a similar dose dependence on TGFbeta(1). Thus TGFbeta regulation of M(2) muscarinic receptor expression is dependent on RhoA, and TGFbeta regulation of p190RhoGAP expression may be a cell type-specific mechanism for TGFbeta signaling through RhoA.
- Published
- 2006
- Full Text
- View/download PDF
17. Human umbilical vein endothelial cells and human dermal microvascular endothelial cells offer new insights into the relationship between lipid metabolism and angiogenesis.
- Author
-
Park HJ, Zhang Y, Georgescu SP, Johnson KL, Kong D, and Galper JB
- Subjects
- Dermis drug effects, Endothelial Cells drug effects, Humans, Hydroxymethylglutaryl-CoA Reductase Inhibitors pharmacology, Umbilical Veins drug effects, Dermis blood supply, Dermis cytology, Endothelial Cells cytology, Lipid Metabolism drug effects, Neovascularization, Physiologic drug effects, Umbilical Veins cytology
- Abstract
Human umbilical vein endothelial cells (HUVECs) have played a major role as a model system for the study of the regulation of endothelial cell function and the role of the endothelium in the response of the blood vessel wall to stretch, shear forces, and the development of atherosclerotic plaques and angiogenesis. Here, we use HUVECs and human microvascular endothelial cells to study the role of the HMG-CoA reductase inhibitor, simvastatin, and the small GTP-binding protein Rho in the regulation of angiogenesis. Simvastatin inhibited angiogenesis in response to FGF-2 in the corneal pocket assay of the mouse and in vascular endothelial growth factor (VEGF)-stimulated angiogenesis in the chick chorioallontoic membrane. Furthermore, simvastatin inhibited VEGF-stimulated tube formation by human dermal microvascular endothelial cells and the formation of honeycomb-like structures by HUVECs. The effect was dose-dependent and was not secondary to apoptosis. Geranylgeranyl-pyrophosphate (GGPP), a product of the cholesterol metabolic pathway that serves as a substrate for the posttranslational lipidation of RhoA, was required for membrane localization, but not farnesylpyrophosphate (FPP), the substrate for the lipidation of Ras. Furthermore, GGTI, a specific inhibitor of GGPP, mimicked the effect of simvastatin of tube formation and the formation of honeycombs whereas FTI, a specific inhibitor of the farnesylation of Ras, had no effect. Adenoviral expression of a DN-RhoA mutant mimicked the effect of simvastatin on tube formation and the formation of honeycombs, whereas a dominant activating mutant of RhoA reversed the effect of simvastatin on tube formation. Finally, simvastatin interfered with the membrane localization of RhoA with a dose-dependence similar to that for the inhibition of tube formation. Simvastatin also inhibited the VEGFstimulated phosphorylation of the VEGF receptor KDR, and the tyrosine kinase FAK, which plays a role in cell migration. These data demonstrate that simvastatin interfered with angiogenesis via the inhibition of RhoA. Data supporting a role for angiogenesis in the development and growth of atherosclerotic plaques suggest that this antiangiogenic effect of Statins might prevent the progression of atherosclerosis via the inhibition of plaque angiogenesis.
- Published
- 2006
- Full Text
- View/download PDF
18. Lipid lowering by pravastatin increases parasympathetic modulation of heart rate: Galpha(i2), a possible molecular marker for parasympathetic responsiveness.
- Author
-
Welzig CM, Shin DG, Park HJ, Kim YJ, Saul JP, and Galper JB
- Subjects
- Adult, Aged, Biomarkers analysis, Blotting, Western, Cross-Over Studies, Electrocardiography, Ambulatory drug effects, Female, GTP-Binding Protein alpha Subunit, Gi2, GTP-Binding Protein alpha Subunits, Gi-Go analysis, Humans, Hyperlipidemias blood, Hyperlipidemias drug therapy, Lipids blood, Lymphocytes drug effects, Lymphocytes metabolism, Male, Middle Aged, Parasympathetic Nervous System physiopathology, Pravastatin therapeutic use, Predictive Value of Tests, Proto-Oncogene Proteins analysis, Simvastatin therapeutic use, Single-Blind Method, GTP-Binding Protein alpha Subunits, Gi-Go metabolism, Heart Rate drug effects, Hypolipidemic Agents therapeutic use, Parasympathetic Nervous System drug effects, Pravastatin pharmacology, Proto-Oncogene Proteins metabolism
- Abstract
Background: We have previously demonstrated in an in vitro model for lipid lowering that lipoprotein depletion resulted in a marked increase in the negative chronotropic response to the acetylcholine analogue carbamylcholine. In this study we used heart rate variability analysis to determine the effect of lipid lowering by statins on the response of the heart to parasympathetic stimulation. In parallel, we examined whether changes in parasympathetic responsiveness correlated with changes in the expression of Galpha(i2), a molecular component of the parasympathetic signaling pathway in the heart., Methods and Results: Patients were randomized in a crossover study of pravastatin and simvastatin. R-R interval analysis of Holter monitor studies demonstrated that in patients treated initially with pravastatin, the peak high-frequency power fraction during sleep, which reflects parasympathetic modulation of heart rate, increased by 24.0+/-5.02% (SEM, n=13, P<0.001) compared with the untreated control value. Simvastatin had no significant effect. Western blot analysis of lymphocytes from patients treated with pravastatin demonstrated a 90.1+/-27.3% (n=10, P=0.009) increase in Galpha(i2) expression, whereas simvastatin had no effect. Relative changes in Galpha(i2) correlated significantly with the changes in the fraction of high-frequency power (rho=0.574, P=0.016)., Conclusions: Taken together with our in vitro data, these data are the first to suggest that cholesterol lowering by pravastatin might increase the response of the heart to parasympathetic stimulation and that changes in Galpha(i2) expression might serve as a molecular marker for this effect.
- Published
- 2003
- Full Text
- View/download PDF
19. Transforming growth factor beta (TGFbeta ) signaling via differential activation of activin receptor-like kinases 2 and 5 during cardiac development. Role in regulating parasympathetic responsiveness.
- Author
-
Ward SM, Desgrosellier JS, Zhuang X, Barnett JV, and Galper JB
- Subjects
- Alkaline Phosphatase metabolism, Animals, Blotting, Western, Chick Embryo, Enzyme Activation, Genes, Reporter, Luciferases metabolism, Promoter Regions, Genetic, Protein Serine-Threonine Kinases, RNA, Messenger metabolism, Receptor, Transforming Growth Factor-beta Type I, Ribonucleases metabolism, Time Factors, Activin Receptors, Type I metabolism, Gene Expression Regulation, Developmental, Heart embryology, Proteins, Receptors, Transforming Growth Factor beta metabolism, Signal Transduction, Transforming Growth Factor beta metabolism
- Abstract
Little is known regarding factors that induce parasympathetic responsiveness during cardiac development. We demonstrated previously that in atrial cells cultured from chicks 14 days in ovo, transforming growth factor beta (TGFbeta) decreased parasympathetic inhibition of beat rate by the muscarinic agonist, carbamylcholine, by 5-fold and decreased expression of Galpha(i2). Here in atrial cells 5 days in ovo, TGFbeta increased carbamylcholine inhibition of beat rate 2.5-fold and increased expression of Galpha(i2). TGFbeta also stimulated Galpha(i2) mRNA expression and promoter activity at day 5 while inhibiting them at day 14 in ovo. Over the same time course expression of type I TGFbeta receptors, chick activin receptor-like kinase 2 and 5 increased with a 2.3-fold higher increase in activin receptor-like kinase 2. Constitutively active activin receptor-like kinase 2 inhibited Galpha(i2) promoter activity, whereas constitutively active activin receptor-like kinase 5 stimulated Galpha(i2) promoter activity independent of embryonic age. In 5-day atrial cells, TGFbeta stimulated the p3TP-lux reporter, which is downstream of activin receptor-like kinase 5 and had no effect on the activity of the pVent reporter, which is downstream of activin receptor-like kinase 2. In 14-day cells, TGFbeta stimulated both pVent and p3TP-lux. Thus TGFbeta exerts opposing effects on parasympathetic response and Galpha(i2) expression by activating different type I TGFbeta receptors at distinct stages during cardiac development.
- Published
- 2002
- Full Text
- View/download PDF
20. TGFbeta regulates the expression of G alpha(i2) via an effect on the localization of ras.
- Author
-
Ward SM, Gadbut AP, Tang D, Papageorge AG, Wu L, Li G, Barnett JV, and Galper JB
- Subjects
- Animals, Carbachol pharmacology, Cardiotonic Agents pharmacology, Cells, Cultured, Chick Embryo, Enzyme Inhibitors pharmacology, Farnesol pharmacology, GTP-Binding Protein alpha Subunit, Gi2, Heart Atria cytology, Heart Atria embryology, Heart Rate drug effects, Methionine pharmacology, Myocytes, Cardiac drug effects, Parasympathetic Nervous System drug effects, Promoter Regions, Genetic, ras Proteins drug effects, GTP-Binding Protein alpha Subunits, Gi-Go metabolism, Gene Expression Regulation, Developmental drug effects, Heart Atria metabolism, Methionine analogs & derivatives, Proto-Oncogene Proteins metabolism, Transforming Growth Factor beta pharmacology, ras Proteins metabolism
- Abstract
The negative chronotropic response of the heart to parasympathetic stimulation is mediated via the interaction of M(2) muscarinic receptors, Galpha(i2) and the G-protein coupled inward rectifying K(+) channel, GIRK1. Here TGFbeta(1) is shown to decrease the expression of Galpha(i2) in cultured chick atrial cells in parallel with attenuation of the negative chronotropic response to parasympathetic stimulation. The response to the acetylcholine analogue, carbamylcholine, decreased from a 95+/-2% (+/-SEM, n=8) inhibition of beat rate in control cells to 18+/-2% (+/-SEM,n =8) in TGFbeta(1) treated cells. Data support the conclusion that TGFbeta regulation of Galpha(i2) expression was mediated via an effect on Ras. TGFbeta(1) inhibited Galpha(i2) promoter activity by 56+/-6% (+/-SEM, n=4) compared to control. A dominant activating Ras mutant reversed the effect of TGFbeta on Galpha(i2) expression and stimulated Galpha(i2) promoter activity 1.7 fold above control. A dominant negative Ras mutant mimicked the effect of TGFbeta(1) on Galpha(i2) promoter activity. TGFbeta had no effect on the ratio of GDP/GTP bound Ras, but markedly decreased the level of membrane associated Ras and increased the level of cytoplasmic Ras compared to control. Furthermore, farnesol, a precursor to farnesylpyrophosphate, the substrate for the farnesylation of Ras, not only reversed TGFbeta(1) inhibition of Ras localization to the membrane, but also reversed TGFbeta(1) inhibition of Galpha(i2)promoter activity. FTI-277, a specific inhibitor of the farnesylation of Ras, mimicked the effect of TGFbeta(1) on Ras localization and Galpha(i2) promoter activity. These data suggest a novel relationship between TGFbeta signaling, regulation of Ras function and the autonomic response of the heart.
- Published
- 2002
- Full Text
- View/download PDF
21. 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors interfere with angiogenesis by inhibiting the geranylgeranylation of RhoA.
- Author
-
Park HJ, Kong D, Iruela-Arispe L, Begley U, Tang D, and Galper JB
- Subjects
- Animals, Cell Membrane chemistry, Cells, Cultured, Collagen pharmacology, Cornea blood supply, Cornea drug effects, Endothelial Growth Factors antagonists & inhibitors, Endothelium, Vascular cytology, Endothelium, Vascular drug effects, Endothelium, Vascular growth & development, Fibroblast Growth Factor 2 antagonists & inhibitors, Humans, Lymphokines antagonists & inhibitors, Mice, Mutation, Protein Prenylation drug effects, Signal Transduction drug effects, Vascular Endothelial Growth Factor A, Vascular Endothelial Growth Factors, rhoA GTP-Binding Protein genetics, rhoA GTP-Binding Protein metabolism, Angiogenesis Inhibitors pharmacology, Hydroxymethylglutaryl-CoA Reductase Inhibitors pharmacology, Neovascularization, Physiologic drug effects, Simvastatin pharmacology, rhoA GTP-Binding Protein antagonists & inhibitors
- Abstract
Angiogenesis is implicated in the pathogenesis of cancer, rheumatoid arthritis, and atherosclerosis and in the treatment of coronary artery and peripheral vascular disease. Here, cholesterol-lowering agents, 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors, are shown to interfere with angiogenesis. In vivo, the HMG-CoA reductase inhibitor simvastatin dose-dependently inhibited capillary growth in both vascular endothelial growth factor-stimulated chick chorioallantoic membranes and basic fibroblast growth factor-stimulated mouse corneas. In vitro, the development of tubelike structures by human microvascular endothelial cells cultured on 3D collagen gels was inhibited at simvastatin concentrations similar to those found in the serum of patients on therapeutic doses of this agent. HMG-CoA reductase inhibitors interfered with angiogenesis via inhibition of the geranylgeranylation and membrane localization of RhoA. Simvastatin inhibited membrane localization of RhoA with a concentration dependence similar to that for the inhibition of tube formation, whereas geranylgeranyl pyrophosphate, the substrate for the geranylgeranylation of Rho, reversed the effect of simvastatin on tube formation and on the membrane localization of RhoA. Furthermore, tube formation was inhibited by GGTI, a specific inhibitor of the geranylgeranylation of Rho; by C3 exotoxin, which inactivates Rho; and by the adenoviral expression of a dominant-negative RhoA mutant. The expression of a dominant-activating RhoA mutant reversed the effect of simvastatin on tube formation. Finally, HMG-CoA reductase inhibitors inhibited signaling by vascular endothelial growth factor, Akt, and focal adhesion kinase, three RhoA-dependent pathways known to be involved in angiogenesis. This study demonstrates a new relationship between lipid metabolism and angiogenesis and an antiangiogenic effect of HMG-CoA reductase inhibitors with possible important therapeutic implications.
- Published
- 2002
- Full Text
- View/download PDF
22. Role of sterol regulatory element binding proteins in the regulation of Galpha(i2) expression in cultured atrial cells.
- Author
-
Park HJ, Begley U, Kong D, Yu H, Yin L, Hillgartner FB, Osborne TF, and Galper JB
- Subjects
- Animals, Binding Sites genetics, CCAAT-Enhancer-Binding Proteins genetics, Chick Embryo, Culture Media pharmacology, DNA-Binding Proteins genetics, GTP-Binding Protein alpha Subunit, Gi2, GTP-Binding Protein alpha Subunits, Gi-Go genetics, Gene Expression Regulation drug effects, Heart Atria cytology, Heart Atria drug effects, Lipoproteins pharmacology, Luciferases genetics, Luciferases metabolism, Mutation, Proto-Oncogene Proteins genetics, RNA, Messenger genetics, RNA, Messenger metabolism, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins metabolism, Sterol Regulatory Element Binding Protein 1, Transfection, CCAAT-Enhancer-Binding Proteins physiology, DNA-Binding Proteins physiology, GTP-Binding Protein alpha Subunits, Gi-Go metabolism, Heart Atria metabolism, Proto-Oncogene Proteins metabolism, Transcription Factors
- Abstract
We have previously demonstrated that growth of embryonic chick atrial cells in medium supplemented with lipoprotein-depleted serum (LPDS) resulted in a coordinate increase in the expression of genes involved in the parasympathetic response of the heart (the M2 muscarinic receptor; the alpha-subunit of the heterotrimeric G protein, Galpha(i2); and the inward rectifying K+ channel protein, GIRK1) and a marked increase in the negative chronotropic response of atrial cells to muscarinic stimulation. In the present study, we demonstrate that regulation of Galpha(i2) promoter activity by LPDS is mediated by the binding of a sterol regulatory element binding protein (SREBP) to a sterol regulatory element (SRE) in the Galpha(i2) promoter. Deletion and point mutation of this putative SRE interfered with the regulation of the Galpha(i2) promoter by SREBP and LPDS. Furthermore gel shift assays demonstrated that point mutations in the putative Galpha(i2) SRE markedly inhibited the binding of purified SREBP to oligonucleotides containing the Galpha(i2) SRE sequence. The expression of a dominant-negative SREBP mutant interfered with LPDS stimulation of Galpha(i2) promoter activity. Finally, we demonstrate that SREBP-1 is markedly more potent than SREBP-2 for the stimulation of Galpha(i2) promoter activity, suggesting that SREBP1 may play a role in the regulation of Galpha(i2) expression. These are the first data to demonstrate SREBP regulation of a protein not involved in lipid homeostasis and suggest a new relationship between lipid metabolism and the parasympathetic response of the heart.
- Published
- 2002
- Full Text
- View/download PDF
23. 3-Hydroxy-3-methylglutaryl CoA reductase inhibitors up-regulate transforming growth factor-beta signaling in cultured heart cells via inhibition of geranylgeranylation of RhoA GTPase.
- Author
-
Park HJ and Galper JB
- Subjects
- Animals, Cells, Cultured, Chick Embryo, Cholesterol metabolism, Plasminogen Activator Inhibitor 1 genetics, Plasminogen Activator Inhibitor 1 physiology, Promoter Regions, Genetic, Protein Serine-Threonine Kinases, Receptor, Transforming Growth Factor-beta Type II, Receptors, LDL analysis, Receptors, Transforming Growth Factor beta biosynthesis, Transforming Growth Factor beta genetics, Up-Regulation, rhoA GTP-Binding Protein, GTP-Binding Proteins metabolism, Hydroxymethylglutaryl-CoA Reductase Inhibitors pharmacology, Myocardium metabolism, Protein Prenylation drug effects, Transforming Growth Factor beta biosynthesis
- Abstract
Transforming growth factor-beta (TGFbeta) signaling has been shown to play a role in cardiac development as well as in the pathogenesis of cardiovascular disease. Prior studies have suggested a relationship between cholesterol metabolism and TGFbeta signaling. Here we demonstrate that induction of the cholesterol metabolic pathway by growth of embryonic chicken atrial cells in medium supplemented with lipoprotein-depleted serum coordinately decreased the expression of the TGFbeta type II receptor (TGFbetaRII), TGFbeta(1), and TGFbeta signaling as measured by plasminogen activator inhibitor-1 (PAI-1) promoter activity. Inhibition of the cholesterol metabolic pathway by the hydrophobic 3-hydroxy-3-methylglutaryl CoA (HMGCoA) reductase inhibitors, simvastatin and atorvastatin, reversed the effect of lipoprotein-depleted serum and up-regulated TGFbetaRII expression, whereas the hydrophilic HMGCoA reductase inhibitor, pravastatin, had no effect. Simvastatin stimulated the expression of TGFbetaRII, TGFbeta(1), and PAI-1 at the level of transcription. Experiments using specific inhibitors of different branches of the cholesterol metabolic pathway demonstrated that simvastatin exerted its effect on TGFbeta signaling by inhibition of the geranylgeranylation pathway. C3 exotoxin, which specifically inactivates geranylgeranylated Rho GTPases, mimicked the effect of simvastatin on PAI-1 promoter activity. Cotransfection of cells with a PAI-1 promoter-reporter and a dominant-negative RhoA mutant increased PAI-1 promoter activity, whereas cotransfection with a dominant-active RhoA mutant decreased PAI-1 promoter activity. These data support the conclusion that TGFbeta signaling is regulated by RhoA GTPase and demonstrate a relationship between cholesterol metabolism and TGFbeta signaling. Our data suggest that in patients treated with HMGCoA reductase inhibitors, these agents may exert effects independent of cholesterol lowering on TGFbeta signaling in the heart.
- Published
- 1999
- Full Text
- View/download PDF
24. Increased incidence of myositis in patients treated with high-dose simvastatin.
- Author
-
Galper JB
- Subjects
- Animals, Cells, Cultured, Cholesterol, LDL blood, Cholesterol, LDL drug effects, Creatine Kinase metabolism, GTP-Binding Proteins drug effects, GTP-Binding Proteins metabolism, Heart drug effects, Humans, Hydroxymethylglutaryl-CoA Reductase Inhibitors administration & dosage, Hyperlipidemias drug therapy, Hyperlipidemias metabolism, Incidence, Myocardium metabolism, Myocardium pathology, Myositis epidemiology, Myositis metabolism, Rats, Simvastatin administration & dosage, Hydroxymethylglutaryl-CoA Reductase Inhibitors adverse effects, Myositis chemically induced, Simvastatin adverse effects
- Published
- 1998
25. Induction of the cholesterol metabolic pathway regulates the farnesylation of RAS in embryonic chick heart cells: a new role for ras in regulating the expression of muscarinic receptors and G proteins.
- Author
-
Gadbut AP, Wu L, Tang D, Papageorge A, Watson JA, and Galper JB
- Subjects
- Animals, Cells, Cultured, Chick Embryo, G Protein-Coupled Inwardly-Rectifying Potassium Channels, Gene Expression Regulation, Heart Atria, Mevalonic Acid metabolism, Models, Biological, Protein Prenylation, RNA, Messenger biosynthesis, Recombinant Proteins biosynthesis, Transfection, Cholesterol metabolism, GTP-Binding Proteins biosynthesis, Myocardium metabolism, Potassium Channels biosynthesis, Potassium Channels, Inwardly Rectifying, Receptors, Muscarinic biosynthesis, Transcription, Genetic, ras Proteins metabolism
- Abstract
We propose a novel mechanism for the regulation of the processing of Ras and demonstrate a new function for Ras in regulating the expression of cardiac autonomic receptors and their associated G proteins. We have demonstrated previously that induction of endogenous cholesterol synthesis in cultured cardiac myocytes resulted in a coordinated increase in expression of muscarinic receptors, the G protein alpha-subunit, G-alphai2, and the inward rectifying K+ channel, GIRK1. These changes in gene expression were associated with a marked increase in the response of heart cells to parasympathetic stimulation. In this study, we demonstrate that the induction of the cholesterol metabolic pathway regulates Ras processing and that Ras regulates expression of G-alphai2. We show that in primary cultured myocytes most of the RAS is localized to the cytoplasm in an unfarnesylated form. Induction of the cholesterol metabolic pathway results in increased farnesylation and membrane association of RAS. Studies of Ras mutants expressed in cultured heart cells demonstrate that activation of Ras by induction of the cholesterol metabolic pathway results in increased expression of G-alphai2 mRNA. Hence farnesylation of Ras is a regulatable process that plays a novel role in the control of second messenger pathways.
- Published
- 1997
- Full Text
- View/download PDF
26. Physiological and biochemical evidence for coordinate increases in muscarinic receptors and Gi during pacing-induced heart failure.
- Author
-
Vatner DE, Sato N, Galper JB, and Vatner SF
- Subjects
- Acetylcholine pharmacology, Adenylyl Cyclase Inhibitors, Animals, Carbachol pharmacology, Cardiac Output, Low etiology, Cardiac Output, Low physiopathology, Cardiac Pacing, Artificial, Dogs, Hemodynamics drug effects, Muscarinic Agonists metabolism, Muscarinic Agonists pharmacology, Cardiac Output, Low metabolism, GTP-Binding Proteins metabolism, Myocardium metabolism, Receptors, Muscarinic metabolism
- Abstract
Background: It is not clear whether the increase in the myocardial guanylyl nucleotide inhibitory protein (Gi), frequently observed in heart failure, is associated with any functional effects., Methods and Results: Eight sham-operated dogs and 10 dogs were studied with pacing-induced heart failure (240 bpm for 4 to 7 weeks), characterized by reduced (P<.05) left ventricular dP/dt (from 2926+/-99 to 1303+/-126 mm Hg/s). The muscarinic agonist acetylcholine (10 micrograms/kg IV) in the presence of ganglionic blockade reduced left ventricular dP/dt more (P<.05) in heart failure (-23+/-2%) than before heart failure (-8+/-2%), despite lesser reductions in arterial pressure. Gi alpha2 was increased by 55% in heart failure. Dose-response curves for carbachol (10-8 to 10-3 mol/L) inhibition of isoproterenol-stimulated adenylyl cyclase demonstrated significantly greater (P<.05) inhibition in heart failure compared with sham-operated dogs. These changes were associated with a coordinate increase in muscarinic receptor density, determined by antagonist binding with 3H-quinuclidinyl benzilate, in heart failure (153+/-6.2 fmol/mg protein) compared with sham-operated dogs (124+/-7.4 fmol/mg protein). Agonist binding with carbachol also revealed an increase in total muscarinic receptors in heart failure without a change in fraction of high- and low-affinity receptors., Conclusions: These data, in the aggregate, provide physiological and biochemical evidence to support the concept that the coordinate increases in muscarinic receptor number and Gi levels in heart failure are coupled to increased inhibition of adenylyl cyclase activity and an increased inhibition of myocardial contractility.
- Published
- 1996
- Full Text
- View/download PDF
27. Specificity of coupling of muscarinic receptor isoforms to a novel chick inward-rectifying acetylcholine-sensitive K+ channel.
- Author
-
Gadbut AP, Riccardi D, Wu L, Hebert SC, and Galper JB
- Subjects
- Acetylcholine pharmacology, Amino Acid Sequence, Animals, Brain metabolism, Chick Embryo, Chickens, Cloning, Molecular, Female, G Protein-Coupled Inwardly-Rectifying Potassium Channels, In Vitro Techniques, Molecular Sequence Data, Potassium Channels drug effects, Potassium Channels genetics, RNA, Messenger genetics, RNA, Messenger metabolism, Rats, Receptors, Muscarinic classification, Sequence Homology, Amino Acid, Tissue Distribution, Xenopus laevis, Potassium Channels metabolism, Potassium Channels, Inwardly Rectifying, Receptors, Muscarinic metabolism
- Abstract
The G-protein-gated inward-rectifying K+ channel GIRK1 has been demonstrated in heart and brain. These tissues also both express the M2, M3, and M4, muscarinic acetylcholine receptors (mAChR) (Gadbut, A.P., and Galper, J.B. (1994),J. Biol. Chem. 269,25823-25829). Only the M2 mAChR has been demonstrated to couple to GIRK1 (Kubo, Y., Reuveny, E., Slesinger, P. A., Jan, Y. N., and Jan, L. Y. (1993) Nature 264, 802-806). In this study we determined the specificity of coupling of the M3 and M4 mAChR to a new GIRK1 cloned from a chick brain cDNA library. This clone codes for a 492-amino acid protein that is 93% identical to rat GIRK1 and is expressed in brain, atrium, and ventricle, but not skeletal muscle. In Xenopus laetis oocytes co-expression of GIRK1 with either the chick M2 or M4 mAChR gave carbamylcholine (10 microm)-stimulated K+ currents of 308 +/-26 nA and 298 +/-29 nA, respectively, which were both Ba2+- and pertussis toxin-sensitive. Activation of the M3 receptor produced 2382 +/-478 nA of current which was insensitive to Ba2+ and pertussis toxin, but was 85% inhabitable by the Cl channel blocker 5-nitro-2-(3-phenylpropylamino)benzoic acid (10-20 microm) consistent with coupling to an endogenous Ca2+-activated Cl- channel via a phosphatidylinositol-dependent mechanism. Co-expression of the cardiac inward rectifier CIR with chick M2 or M4 mAChR and GIRK1 increased currents more than 10-fold, but had no effect on specificity of coupling. These data demonstrate a new function for the M4 mAChR and a high degree of specificity for coupling of each receptor subtype to GIRK1.
- Published
- 1996
- Full Text
- View/download PDF
28. Differential sensitivity of C2-C12 striated muscle cells to lovastatin and pravastatin.
- Author
-
Gadbut AP, Caruso AP, and Galper JB
- Subjects
- Animals, Anticholesteremic Agents chemistry, Cell Differentiation drug effects, Cell Line, Cell Survival drug effects, Cholesterol metabolism, Hydroxymethylglutaryl-CoA Reductase Inhibitors, Lovastatin chemistry, Mice, Molecular Structure, Morphogenesis drug effects, Muscle, Skeletal metabolism, Pravastatin chemistry, Stem Cells drug effects, Stem Cells metabolism, Stem Cells ultrastructure, Structure-Activity Relationship, Anticholesteremic Agents toxicity, Lovastatin toxicity, Muscle, Skeletal drug effects, Myositis chemically induced, Pravastatin toxicity
- Abstract
One of the major side-effects of the use of HMG CoA reductase inhibitors for the treatment of hypercholesterolemia is the development of myositis and, in some patients undergoing concomitant immunosuppressive treatment, the development of rhabdomyolysis. Experiments outlined in these studies demonstrate that inhibitors of HMG-CoA reductase activity which differ primary in the substitution of a methyl group for a hydroxyl group have differential effects on both cholesterol levels and cell viability in a striated muscle cell model, the mouse C2-C12 myoblast. Thus, concentrations as high as 200 microM of pravastatin had little effect on total cholesterol level while 25 microM of lovastatin decreased cellular cholesterol by over 90%. Simvastatin and lovastatin decreased viability of C2-C12 myoblasts by nearly 50% at concentrations as low as 1 and 5 microM, respectively, and decreased viability by almost 90% at 10 and 15 microM respectively. However, 300 microM of pravastatin decreased cell viability by less than 50%. The order of potency for the effects on cell viability wassimvastatin>lovastatin>>>pravastatin. The possible relationship between effects on cell viability and the development of myositis is discussed.
- Published
- 1995
- Full Text
- View/download PDF
29. Decreased adenylate cyclase activity and expression of Gs alpha in human myocardium after orthotopic cardiac transplantation.
- Author
-
Loh E, Barnett JV, Feldman AM, Couper GS, Vatner DE, Colucci WS, and Galper JB
- Subjects
- Adolescent, Adult, Biopsy, Colforsin pharmacology, Female, Humans, Magnesium pharmacology, Male, Middle Aged, Myocardium pathology, RNA, Messenger analysis, Adenylyl Cyclases analysis, GTP-Binding Proteins biosynthesis, Heart Transplantation, Myocardium metabolism
- Abstract
We studied several aspects of guanine nucleotide-stimulated adenylate cyclase function in patients after orthotopic cardiac transplantation. In 28 patients, adenylate cyclase activity was measured in endomyocardial biopsy samples obtained just before and at monthly intervals after cardiac transplantation. In biopsies obtained > or = 6 months after transplantation, basal adenylate cyclase activity was decreased by 67% (n = 12; P < .05), GTP gamma S-stimulated adenylate cyclase activity was decreased by 78% (n = 12; P < .05), Mn+2+forskolin-stimulated adenylate cyclase activity was decreased by 80% (n = 8; P < .05), and Mn+2-stimulated adenylate cyclase activity (a measure of activity of the catalytic subunit of adenylate cyclase) was decreased by 83% (n = 8, P < .05). Western blot analysis demonstrated that 6 months after cardiac transplantation, the level of Gs alpha protein was decreased by 61 +/- 12% (n = 8; P < .001). There was no change in the level of Gi alpha as assessed by pertussis toxin-catalyzed ADP-ribosylation (n = 4; P = NS). With the use of the quantitative polymerase chain reaction, a 50 +/- 10% (n = 6; P < .001) reduction in the steady-state level of Gs alpha mRNA was observed. There was no change in the level of mRNA for Gi-3 alpha. Thus, after orthotopic cardiac transplantation in humans, guanine nucleotide-stimulated adenylate cyclase activity is decreased in parallel with decreased levels of Gs alpha protein and mRNA.
- Published
- 1995
- Full Text
- View/download PDF
30. Cloning of cDNAs coding for the G alpha i1 and G alpha i2 G-proteins from chick brain.
- Author
-
Kilbourne EJ and Galper JB
- Subjects
- Amino Acid Sequence, Animals, Antisense Elements (Genetics), Base Sequence, Chick Embryo, Cloning, Molecular methods, DNA, Complementary biosynthesis, GTP-Binding Proteins biosynthesis, Gene Expression, Gene Library, Molecular Sequence Data, Organ Specificity, RNA Probes, RNA, Messenger analysis, RNA, Messenger biosynthesis, Brain metabolism, Chickens genetics, GTP-Binding Proteins genetics
- Abstract
We have cloned and characterized several cDNAs coding for G-protein inhibitory alpha subunits (G alpha i) from a chick brain cDNA library. Based on homology to G alpha subunits from other eukaryotes, these clones were designated chick G alpha i1 and G alpha i2. On the deduced amino-acid level, G alpha i1 and G alpha i2 were found to be 98 and 95% identical to rat G alpha i1 and G alpha i2, respectively. Using RNase protection analysis, the G alpha i1 and G alpha i2 mRNAs were found to be expressed in chick atria, ventricle, lung, liver, brain and kidney.
- Published
- 1994
- Full Text
- View/download PDF
31. Low density lipoproteins induce parasympathetic responsiveness in embryonic chick ventricular myocytes in parallel with a coordinate increase in expression of genes coding for the M2 muscarinic receptor, G alpha i2, and the acetylcholine-sensitive K+ channel.
- Author
-
Gadbut AP, Toupin DK, Kilbourne EJ, and Galper JB
- Subjects
- Animals, Base Sequence, Blotting, Western, Chick Embryo, GTP-Binding Proteins isolation & purification, Heart drug effects, Heart Rate drug effects, Heart Ventricles, Kinetics, Lovastatin pharmacology, Macromolecular Substances, Molecular Sequence Data, Potassium Channels drug effects, Quinuclidinyl Benzilate metabolism, RNA, Messenger analysis, RNA, Messenger biosynthesis, Receptors, Muscarinic metabolism, Acetylcholine pharmacology, Carbachol pharmacology, GTP-Binding Proteins biosynthesis, Gene Expression drug effects, Heart physiology, Lipoproteins, LDL pharmacology, Myocardium metabolism, Potassium Channels biosynthesis, Receptors, Muscarinic biosynthesis
- Abstract
Growth of chick atrial cells in medium supplemented with lipoprotein-depleted serum has been shown to result in an increase in total cell cholesterol, and an increase in the negative chronotropic response to muscarinic stimulation in parallel with an increase in levels of muscarinic receptors and the G-protein alpha-subunits alpha i and alpha o (Haigh, L. S., Leatherman, G. F., O'Hara, D. S., Smith, T. W., and Galper, J. B. (1988) J. Biol. Chem. 263, 15608-15618). In this study we determined whether growth of chick ventricular cells in medium supplemented with lipoprotein depleted serum could alter levels of muscarinic receptors and G-protein alpha-subunits and induce a negative chronotropic response to muscarinic stimulation. We further determined whether levels of mRNA coding for muscarinic receptors, G-proteins, and the acetylcholine-sensitive K+ channel were coordinately regulated. Growth of embryonic chick ventricular cells from hearts 14 days in ovo in medium supplemented with lipoprotein depleted serum resulted in a 21 +/- 5% (n = 3, +/- S.E.) increase in muscarinic receptor number as demonstrated by [3H]quinuclidinyl benzilate binding and a 4.7 +/- 1.0 (+/- S.E., n = 4)-fold increase in G alpha i2 as demonstrated by Western blot analysis. These changes in receptor and G-protein were associated with a coordinate increase in levels of mRNA coding for the M2 muscarinic receptor, G alpha i2 and the acetylcholine sensitive K+ channel as determined by RNase protection. These increases were reversed by addition of 30 microM mevinolin, an inhibitor of HMG-CoA reductase activity. Carbamylcholine (0.1 mM) had no effect on beat rate in ventricular cells grown in medium supplemented with fetal calf serum. Cells grown in medium supplemented with lipoprotein depleted serum demonstrated a 40 +/- 8% (+/- S.E., n = 10, p < 0.0001) decrease in beat rate in response to 0.1 mM carbamylcholine which was reversed by the addition of 30 microM mevinolin. These data suggest that, during growth in medium supplemented with lipoprotein depleted serum, a component of the cholesterol biosynthetic pathway plays a role in the coordinate induction of mRNAs coding for receptors, G-proteins, and an effector (ion channel) that results in the induction of a parasympathetic response in the ventricular cell characteristic of the atrial phenotype.
- Published
- 1994
32. A novel M3 muscarinic acetylcholine receptor is expressed in chick atrium and ventricle.
- Author
-
Gadbut AP and Galper JB
- Subjects
- Amino Acid Sequence, Animals, Base Sequence, CHO Cells, Carbachol pharmacology, Cloning, Molecular, Cricetinae, Dose-Response Relationship, Drug, Gene Expression, Inositol Phosphates metabolism, Molecular Sequence Data, Muscarinic Antagonists, Pertussis Toxin, Quinuclidinyl Benzilate metabolism, RNA, Messenger analysis, Receptor, Muscarinic M3, Receptors, Muscarinic classification, Receptors, Muscarinic genetics, Restriction Mapping, Sequence Analysis, DNA, Sequence Homology, Amino Acid, Tissue Distribution, Transfection, Virulence Factors, Bordetella pharmacology, Acetylcholine metabolism, Chickens genetics, Heart Atria chemistry, Heart Ventricles chemistry, Receptors, Muscarinic biosynthesis
- Abstract
Prior studies have suggested that heart expresses only the M2 isoform of the muscarinic receptor (Peralta, E.G., Ashkenazi, A., Winslow, J.W., Smith, D.H., Ramachandran, J., and Capon, D.J. (1987) EMBO J. 6, 3923-3929). Tietje and Nathanson (Tietje, K.M., and Nathanson, N. M. (1991) J. Biol. Chem. 266, 17382-17387) have recently demonstrated that the chick heart may be unique since it expresses both the M2 and M4 isoforms of the muscarinic receptor. In this study, in order to determine whether other isoforms of the muscarinic receptor were present in the chick heart, a chick M3 muscarinic receptor receptor was cloned, characterized, and its expression in chick tissues determined. Using a human M3 muscarinic receptor cDNA as a probe, a 2.4-kilobase pair cDNA was isolated from a chick brain cDNA library which contained an open reading frame coding for a 639 amino acid protein. This protein demonstrated an 87 and 86% homology to the human and rat M3 muscarinic receptor, respectively. Chinese hamster ovary (CHO-GRA) cells were stably transfected with the chick M3 muscarinic receptor and one clone (CHO-CM3) expressed the M3 receptor, as measured by the binding of quinuclidinly benzilate at 116 +/- 14 (+/- S.E., n = 3) fmol/mg protein with a Kd of 76 +/- 17 pM. This receptor demonstrated a rank order of potency for muscarinic antagonist binding characteristic for the M3 receptor: with high affinity binding for hexahydrosiladifenidol, Kd: 16 +/- 2 nM (+/- S.E., n = 3); intermediate affinity for pirenzepine, Kd: 383 +/- 47 nM, and low affinity for methoctramine, Kd: 533 +/- 185 nM (+/- S.E., n = 3). Carbamylcholine stimulation of CHO-CM3 cells resulted in a 1.6-fold increase in cyclic AMP accumulation and a 3.5-fold increase in a pertussis toxin-insensitive inositol phosphate release. These data demonstrate that the chick M3 muscarinic receptor has the properties characteristic of M3 receptors from other species. RNase protection studies demonstrated the presence of M3 muscarinic receptor mRNA in the brain, atria, and ventricle of chicks 17 days in ovo. Hence, the chick heart appears to have the unique capacity to express mRNAs coding not only for the M2 and M4 muscarinic receptors but also for the M3 muscarinic receptor.
- Published
- 1994
33. Isolation and expression of a novel chick G-protein cDNA coding for a G alpha i3 protein with a G alpha 0 N-terminus.
- Author
-
Kilbourne EJ and Galper JB
- Subjects
- Amino Acid Sequence, Animals, Base Sequence, Brain, Chickens, Cloning, Molecular, DNA, Complementary, GTP-Binding Proteins chemistry, Gene Expression, Guanosine Triphosphate metabolism, Molecular Sequence Data, Peptide Mapping, RNA, Messenger genetics, Restriction Mapping, Sequence Alignment, Sequence Homology, Amino Acid, Tissue Distribution, GTP-Binding Proteins genetics
- Abstract
We have cloned cDNAs coding for G-protein alpha subunits from a chick brain cDNA library. Based on sequence similarity to G-protein alpha subunits from other eukaryotes, one clone was designated G alpha i3. A second clone, G alpha i3-o, was identical to the G alpha i3 clone over 932 bases on the 3' end. The 5' end of G alpha i3-o, however, contained an alternative sequence in which the first 45 amino acids coded for are 100% identical to the conserved N-terminus of G alpha o from species such as rat, mouse, human, bovine and hamster. Both clones were found to be expressed in all tissues studied. The unusual alpha o-alpha i3-like G-protein chimera, G alpha i3-o, was found to be expressed at significantly lower levels than G alpha i3. In vitro transcription and translation of the G alpha i3-o cDNA clone gave a protein of approx. 41 kDa which stably bound guanosine 5'-[gamma-thio]triphosphate. G alpha i3-o appears to be the first G-protein alpha subunit cloned which contains ends that are homologous to two different alpha subunit isoforms, G alpha o and G alpha i3.
- Published
- 1994
- Full Text
- View/download PDF
34. Cloning and developmental expression of the chick type II and type III TGF beta receptors.
- Author
-
Barnett JV, Moustakas A, Lin W, Wang XF, Lin HY, Galper JB, and Maas RL
- Subjects
- Amino Acid Sequence, Animals, Base Sequence, Chick Embryo growth & development, Cloning, Molecular, Conserved Sequence, Gene Expression Regulation, Molecular Sequence Data, Organ Specificity, Protein Serine-Threonine Kinases, RNA, Messenger biosynthesis, Receptor, Transforming Growth Factor-beta Type II, Receptors, Transforming Growth Factor beta chemistry, Receptors, Transforming Growth Factor beta genetics, Chick Embryo metabolism, Proteoglycans, Receptors, Transforming Growth Factor beta biosynthesis
- Abstract
To address the role of peptide growth factors in chick organogenesis, we have focused on TGF beta 2 and have cloned the chick Type II and Type III TGF beta receptors. The chick Type II receptor is a serine/threonine kinase with a ligand binding profile identical to the human receptor and a divergent N-terminus when compared to the mammalian receptors. The chick Type III receptor is a beta-glycan that demonstrates a binding profile identical to the rat receptor and contains a single transmembrane spanning domain and short cytoplasmic tail that are highly conserved when compared to the mammalian receptors. Both the Type II and Type III TGF beta receptors are coexpressed during chick embryogenesis in the developing heart, lung, and eye, and are developmentally upregulated in parallel in the heart and lung. Levels of both receptor proteins and mRNAs also increase in cardiocytes cultured from different developmental stages, in agreement with the increase in Type II and Type III receptor mRNA levels observed in the developing heart. Although exhibiting different temporal or spatial profiles from the receptors, TGF beta 2 is also expressed in the developing heart, lung, and eye. These findings are consistent with recent data indicating that co-expression of both the Type II and Type III TGF beta receptors is required for high affinity binding of TGF beta 2 by the Type II receptor and suggest that TGF beta 2 and the Type II and Type III TGF beta receptors participate in heart, lung, and eye development.
- Published
- 1994
- Full Text
- View/download PDF
35. Effect of low-density lipoproteins, mevinolin, and G proteins on Ca2+ response in cultured chick atrial cells.
- Author
-
Tan W, Barnett JV, Pietrobon D, Hehn G, Greiser C, Marsh JD, and Galper JB
- Subjects
- 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester pharmacology, Animals, Calcium Channels metabolism, Cattle blood, Cattle embryology, Cells, Cultured, Chick Embryo, Dihydropyridines metabolism, Extracellular Space metabolism, Fetal Blood, Heart Atria, Myocardium cytology, Pertussis Toxin, Virulence Factors, Bordetella pharmacology, Calcium metabolism, GTP-Binding Proteins physiology, Lipoproteins, LDL pharmacology, Lovastatin pharmacology, Myocardium metabolism
- Abstract
Growth of cells from atria of embryonic chick hearts 14 days in ovo in medium supplemented with lipoprotein-depleted serum (LPDS) results in an increase in total cell cholesterol, enhanced parasympathetic responsiveness (7), and decreased sympathetic responsiveness (1). These effects were reversed by the hydroxymethyl glutaryl CoA reductase inhibitor, mevinolin. In these studies, comparison of cell growth in medium supplemented with fetal calf serum (FCS) and LPDS demonstrated that, after growth with LPDS, the ability of Ca2+ and the Ca2+ channel agonist, BAY K 8644, to enhance the amplitude of contraction decreased by 25 and 50%, respectively. These effects of growth in LPDS were reversed by incubation with mevinolin. LPDS had no effect on either Ca2+ channel number as measured by (+)-[5-methyl-3H]PN200-110 binding or Ca2+ current density as measured by the whole cell patch method. Treatment of cells grown in LPDS with pertussis toxin, which inactivates alpha o and alpha i, returned the contractile response to 10(-7) M BAY K 8644 to control levels. Pertussis toxin had no effect on the contractile response or adenosine 3',5'-cyclic monophosphate levels in control cells grown in FCS alone. These data suggest that alterations in the relative levels of alpha o and alpha s in cells grown in LPDS may play a role in regulating the contractile response to Ca2+ channel agonists and to exogenous Ca2+.
- Published
- 1993
- Full Text
- View/download PDF
36. Co-culture of embryonic chick heart cells and ciliary ganglia induces parasympathetic responsiveness in embryonic chick heart cells.
- Author
-
Barnett JV, Taniuchi M, Yang MB, and Galper JB
- Subjects
- Animals, Carbachol pharmacology, Cells, Cultured, Chick Embryo, Culture Media, Conditioned pharmacology, GTP-Binding Proteins metabolism, Ganglia, Parasympathetic cytology, Guanosine Triphosphate metabolism, Heart drug effects, Heart innervation, Myocardium metabolism, Ganglia, Parasympathetic physiology, Heart physiology, Myocardium cytology
- Abstract
We have developed a system for the co-culture of embryonic chick heart cells obtained from embryos at 3.5 days in ovo with ciliary ganglia from chick embryos at 7 days in vivo. After 3 days of co-culture, removal of the ciliary ganglia resulted in complete degeneration of axons within 6-8 h, leaving the post-innervated heart cell culture devoid of neurons. Embryonic chick heart cells at 3.5 days in ovo are unresponsive to muscarinic stimulation. However, following 3 days of co-culture with ciliary ganglia, the heart cells developed a negative chronotropic response to muscarinic stimulation (paired t test, P < 0.02) which persisted for at least 24 h after removal of the ciliary ganglion. The development of muscarinic responsiveness was associated with an increase in the levels of specific alpha-subunits of the guanine nucleotide binding proteins (G-proteins), with a 3-fold increase in the level of alpha 39 (39 kDa subunit) and a 2.5-fold increase in the level of alpha 41. The level of the G-protein subunit alpha s remained unchanged. Culture of embryonic chick heart cells at 3.5 days in ovo with medium conditioned by the growth of embryonic chick heart cells and ciliary ganglia had an effect on the chronotropic response to muscarinic stimulation and on alpha 39 and alpha 41 levels identical to that of co-culture. These data suggest that a soluble factor released during the co-culture of embryonic chick heart cells and ciliary ganglia is capable of inducing muscarinic responsiveness. These studies suggest that innervation of the heart may induce parasympathetic responsiveness by increasing the availability of G-proteins which couple the muscarinic receptor to a physiological response.
- Published
- 1993
- Full Text
- View/download PDF
37. Direct contact between sympathetic neurons and rat cardiac myocytes in vitro increases expression of functional calcium channels.
- Author
-
Ogawa S, Barnett JV, Sen L, Galper JB, Smith TW, and Marsh JD
- Subjects
- Animals, Calcium metabolism, In Vitro Techniques, Myocardial Contraction, Myocardium metabolism, Rats, Receptors, Nicotinic analysis, Calcium Channels physiology, Cell Communication, Ganglia, Sympathetic physiology, Heart innervation, Myocardium cytology
- Abstract
To test the hypothesis that direct contact between sympathetic neurons and myocytes regulates expression and function of cardiac Ca channels, we prepared cultures of neonatal rat ventricular myocytes with and without sympathetic ganglia. Contractile properties of myocytes were assessed by an optical-video system. Contractility-pCa curves showed a 60% greater increase in contractility for innervated myocytes compared with control cells at 6.3 mM [Ca]0 (n = 8, P less than 0.05). Cells grown in medium conditioned by growth of ganglia and myocytes were indistinguishable physiologically from control cells. [Bay K 8644]-contractility curves revealed a 60 +/- 10% enhancement of the contractility response at 10(-6) M for innervated cells compared with control cells. The increased response to Bay K 8644 was not blocked by alpha- or beta-adrenergic antagonists. Moreover, increased efficacy of Bay K 8644 was maintained for at least 24 h after denervation produced by removal of ganglia from the culture. Dihydropyridine binding sites were assessed with the L channel-specific radioligand 3[H]PN200-110. PN200-110 binding sites were increased by innervation (51 +/- 5 to 108 +/- 20 fmol/mg protein, P less than 0.01), with no change in KD. Peak current-voltage curves were determined by whole-cell voltage clamp techniques for myocytes contacted by a neuron, control myocytes, and myocytes grown in conditioned medium. Current density of L-type Ca channels was significantly higher in innervated myocytes (10.5 +/- 0.4 pA/pF, n = 5) than in control myocytes (5.9 +/- 0.3 pA/pF, n = 8, P less than 0.01) or myocytes grown in conditioned medium (6.2 +/- 0.2 pA/pF, n = 10, P less than 0.01). Thus, physical contact between a sympathetic neuron and previously uninnervated neonatal rat ventricular myocytes increases expression of functional L-type calcium channels as judged by contractile responses to Ca0 and Bay K 8644, as well as by electrophysiological and radioligand binding properties.
- Published
- 1992
- Full Text
- View/download PDF
38. Effect of manganese (II) bis(glycinate)dichloride on Ca2+ channel function in cultured chick atrial cells.
- Author
-
Tan W, Barnett JV, Hehn GM, Quay SC, and Galper JB
- Subjects
- 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester pharmacology, Animals, Calcium metabolism, Calcium pharmacokinetics, Calcium Channel Blockers metabolism, Calcium Channels metabolism, Calcium Channels physiology, Calcium Radioisotopes, Cells, Cultured, Chick Embryo, Dihydropyridines metabolism, Glycine pharmacology, Heart Atria cytology, Heart Atria drug effects, Myocardial Contraction drug effects, Myocardium metabolism, Calcium Channels drug effects, Glycine analogs & derivatives, Heart drug effects, Manganese pharmacology, Organometallic Compounds pharmacology
- Abstract
Manganese (II) bis(glycinate)dichloride (Mn(glycinate)2) is a coordination complex of manganese with application as a contrast enhancement agent for magnetic resonance imaging in the heart. To determine the cardioactivity of the manganese ion in this chelation cage, the effects of Mn(glycinate)2 on Ca channel function in the cultured chick atrial cell was studied. Mn(glycinate)2 decreased amplitude of contraction in chick atrial cells from embryos 14 days in ovo with complete inhibition of beating at 1 mM and half-maximal effect at 0.1 mM. Under control conditions, Bay K 8644, a Ca channel activator increased amplitude of contraction by 86% with a half maximal effect at 3.2 x 10(-7) M. In the presence of 0.025 mM Mn(glycinate)2, a concentration which had no effect on the amplitude of contraction, the maximum response to Bay K 8644 was decreased to 31%. Mn(glycinate)2 had no effect on the EC50 for the response to Bay K 8644, 1.7 +/- 0.1 x 10(-9) M (S.E.M., n = 4) in control cells compared to 2.2 +/- 0.4 x 10(-9) M (S.E.M., n = 4) in cells incubated with Mn(glycinate)2. 45Ca2+ uptake over 5 min in cultured chick atrial cells decreased from 2.0 nmol/mg protein in control cells to 1.5 nmol/mg protein in the presence of 10(-5) M PN200-110, a Ca2+ channel blocker, a decrease of 28%. 45Ca2+ uptake decreased to 0.94 nmol/mg protein (53%) in the presence of 1 nmol Mn(glycinate)2. Effects of Mn(glycinate)2 and PN200 were not additive. These data demonstrate that Mn(glycinate)2 exerts its negative inotropic effect, at least partially, by interfering with the function of the L-type Ca channels at high concentrations.
- Published
- 1991
- Full Text
- View/download PDF
39. Muscarinic cholinergic stimulation of inositol phosphate production in cultured embryonic chick atrial cells. Evidence for a role of two guanine-nucleotide-binding proteins.
- Author
-
Barnett JV, Shamah SM, Lassegue B, Griendling KK, and Galper JB
- Subjects
- Adenosine Diphosphate Ribose metabolism, Animals, Atropine pharmacology, Cells, Cultured, Chick Embryo, Guanosine 5'-O-(3-Thiotriphosphate) pharmacology, Heart Atria drug effects, Heart Atria embryology, Heart Atria metabolism, Kinetics, NAD metabolism, Pertussis Toxin, Type C Phospholipases metabolism, Virulence Factors, Bordetella pharmacology, GTP-Binding Proteins physiology, Inositol Phosphates biosynthesis, Myocardium metabolism, Receptors, Muscarinic physiology
- Abstract
These studies demonstrate a novel mechanism for the coupling of the muscarinic receptor to phospholipase C activity in embryonic chick atrial cells. In monolayer cultures of atrial cells from hearts of embryonic chicks at 14 days in ovo, carbamylcholine stimulated the sequential appearance of InsP3, InsP2 and InsP1 with an EC50 (concn. causing 50% of maximal stimulation) of 30 microM. In the presence of 15 mM-Li, a 5 min exposure to carbamylcholine (0.1 mM) increased InsP3 levels to a maximum of 47 +/- 12% over basal, InsP2 to 108 +/- 13% over basal and InsP1 to 42 +/- 5% over basal. This effect was blocked by 5 microM-atropine. Incubation of these cells with pertussis toxin (15 h; 0.5 ng/ml) inhibited carbamylcholine-stimulated InsP3, InsP2 and InsP1 formation by 42 +/- 7%, 30 +/- 3% and 48 +/- 7% respectively. The IC50 (concn. causing 50% inhibition) for pertussis toxin inhibition of all three inositol phosphates was 0.01 ng/ml, with a half-time of 6 h at 0.5 ng/ml. This partial sensitivity to pertussis toxin was not due to incomplete ADP-ribosylation of the guanine-nucleotide-binding protein (G-protein), since autoradiography of polyacrylamide gels of cell homogenates incubated with [32P]NAD+ in the presence of pertussis toxin demonstrated that incubation of cells with 0.5 ng of pertussis toxin/ml for 15 h resulted in complete ADP-ribosylation of pertussis toxin substrates by endogenous NAD+. In cells permeabilized with saponin (10 micrograms/ml), 0.1 mM-GTP[S] (guanosine 5'-[gamma-thio]triphosphate) stimulated InsP1 by 102 +/- 15% (mean +/- S.E.M., n = 4), InsP2 by 421 +/- 67% and InsP3 by 124 +/- 33% above basal. Incubation of cells for 15 h with 0.5 ng of pertussis toxin/ml decreased GTP[S]-stimulated InsP1 production in saponin-treated cells by 30 +/- 10% (n = 3), InsP2 production by 45 +/- 7% (n = 4) and InsP3 production by 49 +/- 6% (n = 4). These data demonstrate that in embryonic chick atrial cells at least two independent G-proteins, a pertussis toxin-sensitive G-protein and a pertussis toxin-insensitive G-protein, play a role in coupling muscarinic agonist binding to phospholipase C activation and to inositol phosphate production.
- Published
- 1990
- Full Text
- View/download PDF
40. Development of muscarinic-cholinergic stimulation of inositol phosphate production in cultured embryonic chick atrial cells. Evidence for a switch in guanine-nucleotide-binding protein coupling.
- Author
-
Barnett JV, Shamah SM, Lassegue B, Griendling KK, and Galper JB
- Subjects
- Animals, Carbachol pharmacology, Cells, Cultured, Chick Embryo, Guanosine 5'-O-(3-Thiotriphosphate) pharmacology, Heart Atria drug effects, Heart Atria embryology, Heart Atria metabolism, Heart Ventricles drug effects, Heart Ventricles embryology, Heart Ventricles metabolism, Kinetics, Pertussis Toxin, Time Factors, Type C Phospholipases metabolism, Virulence Factors, Bordetella pharmacology, GTP-Binding Proteins physiology, Inositol Phosphates biosynthesis, Myocardium metabolism, Receptors, Muscarinic physiology
- Abstract
We have demonstrated that muscarinic stimulation of inositol phosphate production in cultured atrial cells from chicks at 14 days in ovo is partially sensitive to inhibition by pertussis toxin. In these cells, muscarinic agonist binding is coupled to phospholipase C activity via at least two guanine-nucleotide-binding proteins (G-proteins), one sensitive to pertussis toxin and the other (Gp) insensitive to pertussis toxin [Barnett, Shamah, Lassegue, Griendling & Galper (1990) Biochem. J. 271, 437-442]. In the current study we demonstrate that during embryonic development of the chick heart, muscarinic stimulation of inositol phosphate production decreases by 50% between days 5 and 14 in ovo in cells cultured from both atrium and ventricle. In atrial cells, however, pertussis toxin-sensitive muscarinic stimulation of inositol phosphate production increased from undetectable levels at day 5 in ovo to 40% of total stimulation at day 12 in ovo. Muscarinic stimulation of inositol phosphate production in the ventricle did not become sensitive to pertussis toxin at any age studied. In permeabilized atrial cells from embryonic chicks at 5 days in ovo, guanosine 5'-[gamma-thio]triphosphate (GTP[S]) stimulated InsP1 levels by 40 +/- 10% (mean +/- S.E.M., n = 3), InsP2 levels by 117 +/- 18% and InsP3 levels by 51 +/- 8%, suggesting that at day 5 in ovo all of the muscarinic-stimulated inositol phosphate production was coupled to phospholipase C via Gp. H.p.l.c. analysis demonstrated that, in spite of these changes in coupling of phospholipase C to different G-proteins, no changes could be demonstrated in the isomers of InsP3 produced in response to carbamylcholine at both days 5 and 14 in ovo. These data demonstrate that embryonic development of the chick atrium is associated with a switch in coupling of muscarinic receptors to phospholipase C from Gp to a pertussis toxin substrate. This developmental switch in coupling of G-proteins may be related to possible developmental switches in levels of muscarinic receptor isoforms or switches in the subtype of phospholipase C.
- Published
- 1990
- Full Text
- View/download PDF
41. The development of physiologic responsiveness to muscarinic stimulation in embryonic chick heart. Relationship to increased levels of pertussis toxin substrates.
- Author
-
Barnett JV, Shamah SM, and Galper JB
- Subjects
- Animals, Chick Embryo, Heart drug effects, Heart physiology, Immunoblotting, Isoproterenol pharmacology, Myocardium enzymology, Organ Culture Techniques, Poly(ADP-ribose) Polymerases metabolism, Receptors, Muscarinic drug effects, Substrate Specificity, Adenylate Cyclase Toxin, Adenylyl Cyclases metabolism, GTP-Binding Proteins metabolism, Heart embryology, Pertussis Toxin, Receptors, Muscarinic physiology, Virulence Factors, Bordetella metabolism
- Abstract
Studies of the development of parasympathetic responsiveness in embryonic chick hearts have demonstrated that between days 2.5 and 10 in ovo the ability of muscarinic agonists to inhibit adenylate cyclase activity increases 10-fold in parallel with a 2.7-fold increase in the level of alpha i and alpha o. Thus, muscarinic inhibition of adenylate cyclase increases in parallel with an increase in alpha o and alpha i. These data suggest that changes in levels of guanine nucleotide regulatory proteins control, at least in part, the appearance of a parasympathetic response in the heart during embryonic development of the chick.
- Published
- 1990
- Full Text
- View/download PDF
42. Non-co-ordinate development of beta-adrenergic receptors and adenylate cyclase in chick heart.
- Author
-
Alexander RW, Galper JB, Neer EJ, and Smith TW
- Subjects
- Animals, Chick Embryo, Dihydroalprenolol metabolism, Guanylyl Imidodiphosphate metabolism, Heart embryology, In Vitro Techniques, Isoproterenol metabolism, Kinetics, Time Factors, Adenylyl Cyclases metabolism, Myocardium metabolism, Receptors, Adrenergic metabolism, Receptors, Adrenergic, beta metabolism
- Abstract
We have studied the properties of beta-adrenergic receptors and of their interaction with adenylate cyclase in the chick myocardium during embryogenesis. Between 4.5 and 7.5 days in ovo the number of receptors determined by (-)-[3H]dihydroalprenolol ([3H]DHA) binding is constant at approx. 0.36 pmol of receptor/mg of protein. By day 9 the density decreases significantly to 0.22 pmol of receptor/mg of protein. At day 12.5--13.5 the number was 0.14--0.18 pmol of receptor/mg of protein. This number did not change further up to day 16. The same results were obtained with guanosine 5'-[beta, gamma-imido]triphosphate (p[NH]ppG) added to the assay mixtures. There was no significant change in receptor affinity for the antagonist [3H]DHA between days 5.5 and 13. Despite the decrease in numbers of beta-adrenergic receptors, there was no change in basal, p[NH]ppG-, isoprenaline- or isoprenaline-plus-p[NH]ppG-stimulated adenylate cyclase activity between days 3 and 12 of development. We conclude that beta-adrenergic receptors and adenylate cyclase are not co-ordinately regulated during early embryonic development of the chick heart. Some of the beta-adrenergic receptors present very early in the ontogeny of cardiac tissue appear not to be coupled to adenylate cyclase since their loss is not reflected in decreased activation of the enzyme.
- Published
- 1982
- Full Text
- View/download PDF
43. Developmental changes in the sensitivity of embryonic heart cells to tetrodotoxin and D600.
- Author
-
Galper JB and Catterall WA
- Subjects
- Age Factors, Animals, Biological Transport drug effects, Chick Embryo, Culture Techniques, Electric Conductivity, Sodium metabolism, Veratridine antagonists & inhibitors, Gallopamil pharmacology, Heart embryology, Myocardial Contraction drug effects, Tetrodotoxin pharmacology, Verapamil analogs & derivatives
- Published
- 1978
- Full Text
- View/download PDF
44. Agonist regulation of the muscarinic cholinergic receptor in embryonic chick heart.
- Author
-
Galper JB, Dziekan LC, and Smith TW
- Subjects
- Animals, Cell Membrane Permeability drug effects, Cells, Cultured, Chick Embryo, Embryo, Mammalian, Embryo, Nonmammalian, Myocardium cytology, Myocardium metabolism, N-Methylscopolamine, Potassium metabolism, Quinuclidinyl Benzilate pharmacology, Time Factors, Carbachol pharmacology, Myocardial Contraction drug effects, Receptors, Cholinergic drug effects, Receptors, Muscarinic drug effects, Scopolamine Derivatives pharmacology
- Published
- 1983
- Full Text
- View/download PDF
45. Effects of low density lipoproteins and mevinolin on cholesterol content and muscarinic cholinergic responsiveness in cultured chick atrial cells. Regulation of levels of muscarinic receptors and guanine nucleotide regulatory proteins.
- Author
-
Haigh LS, Leatherman GF, O'Hara DS, Smith TW, and Galper JB
- Subjects
- Algorithms, Animals, Carbachol pharmacology, Cells, Cultured, Chick Embryo, Hydroxymethylglutaryl CoA Reductases metabolism, Myocardial Contraction drug effects, Quinuclidinyl Benzilate metabolism, Cholesterol metabolism, GTP-Binding Proteins metabolism, Heart Atria cytology, Lipoproteins, LDL pharmacology, Lovastatin pharmacology, Receptors, Muscarinic metabolism
- Abstract
Cultures of myocytes from embryonic chick atria grown in medium supplemented with fetal calf serum from which lipoproteins had been removed demonstrated a nearly 10-fold increase in sensitivity of beating to the muscarinic cholinergic agonist carbamylcholine compared to cells grown with control serum. This effect was reversed by growth of cells in medium supplemented with lipoprotein-depleted serum (LPDS) reconstituted with the low density lipoprotein fraction from fetal calf serum. In cells grown in LPDS, total cell cholesterol was increased 32% over control levels and returned to control levels in cells grown with LPDS reconstituted with low density lipoprotein. Growth of cells in LPDS plus mevinolin, an inhibitor of endogenous cholesterol synthesis, also reversed the effects of LPDS on cholesterol content and sensitivity of beating to carbamylcholine. The ability of mevinolin (30 microM) to reverse the effect of LPDS on sensitivity of beating to carbamylcholine was inhibited by mevalonic acid, a metabolic precursor to cholesterol, with an IC50 of 7 x 10(-5) M. These data suggest that mevinolin reverses the effects of LPDS by altering cellular cholesterol levels. Enhanced responsiveness of embryonic chick heart cells to muscarinic stimulation was associated with a 2-fold increase in the number of muscarinic receptors with high affinity for agonist from 82 +/- 10 fmol/mg protein in media containing fetal calf serum to 175 +/- 12 fmol/mg protein in cells grown in the presence of LPDS. The distribution of receptors between high affinity (RH) and low affinity (RL) forms changed from 41% RH and 59% RL in cells grown in control serum to 66.5% RH and 33.5% RL in cells grown in LPDS. Quantitation of the effect of growth in LPDS on the levels of guanine nucleotide regulatory proteins No and Ni which couple the muscarinic receptor to a physiologic response, demonstrated that the relative levels of the 39-kDa alpha subunits of No and 41-kDa alpha subunits of Ni determined by ADP ribosylation with pertussis toxin and immunoblotting increased 2-fold compared to control cells grown with fetal calf serum. Growth of cells with medium supplemented with LPDS plus mevinolin reduced the levels of alpha 39 and alpha 41 to below the levels in control cells. Levels of the beta subunit of No and Ni were unaffected by growth with LPDS.(ABSTRACT TRUNCATED AT 400 WORDS)
- Published
- 1988
46. Effects of low density lipoproteins and mevinolin on sympathetic responsiveness in cultured chick atrial cells. Regulation of beta-adrenergic receptors and alpha s.
- Author
-
Barnett JV, Haigh LS, Marsh JD, and Galper JB
- Subjects
- 1-Methyl-3-isobutylxanthine pharmacology, Adenosine Diphosphate Ribose metabolism, Adenylyl Cyclases metabolism, Animals, Atrial Function, Cell Division drug effects, Cells, Cultured, Chick Embryo, Cholera Toxin pharmacology, Heart Atria drug effects, Myocardium cytology, Myocardium metabolism, Receptors, Adrenergic, beta drug effects, Heart physiology, Heart Rate drug effects, Isoproterenol pharmacology, Lipoproteins, LDL pharmacology, Lovastatin pharmacology, Myocardial Contraction drug effects, Receptors, Adrenergic, beta physiology
- Abstract
We have previously demonstrated that cultures of myocytes from embryonic chick atria grown in media supplemented with fetal calf serum from which lipoproteins have been removed demonstrate a nearly 10-fold increase in sensitivity of beating to the muscarinic cholinergic agonist carbamylcholine compared with cells grown with control medium. This increased response to carbamylcholine was associated with a 1.4-fold increase in total cell cholesterol, a 2-fold increase in the number of muscarinic receptors which bind agonist with high affinity, and a 2-fold increase in the levels of the alpha subunits of Go and Gi (Haigh, L. S., Leatherman, G. F., O'Hara, D. S., Smith, T. W., and Galper, J. B. (1988) J. Biol. Chem. 263, 15608-15618). In the studies reported here, we determined the responsiveness of cells grown in lipoprotein-depleted serum (LPDS) to beta-adrenergic stimulation. Isoproterenol stimulated a contractile response of 58% measured as an increase in amplitude of contraction with a half-maximal effect at 3 x 10(-7) M for cells grown in fetal calf serum, but had no significant effect on amplitude of contraction on cells grown in LPDS. In cells grown in media supplemented with fetal calf serum, isoproterenol (1 x 10(-3) M) stimulated adenylate cyclase activity 100% over basal with an EC50 of 7 x 10(-6) M compared with an increase of 32% in cells grown in media supplemented with LPDS. beta-Adrenergic receptor number as measured by the binding of 125I-pindolol decreased from 24 +/- 3 (+/- S.E., n = 6) fmol/mg protein in cells grown under control conditions to 12 +/- 2 (n = 6) fmol/mg protein in media supplemented with LPDS. The level of alpha s as measured both by ADP-ribosylation with cholera toxin in the presence of 32P-NAD and by immunoblotting with specific antibody to alpha s decreased by 3-fold in cells grown in media supplemented with LPDS compared with control. All of these effects of growth of cells in LPDS were reversed by incubating cells with LPDS plus 30 microM mevinolin, an inhibitor of endogenous cholesterol synthesis. These studies indicate that growth of cells in media supplemented with LPDS results in a coordinate decrease in the levels of beta-adrenergic receptors and alpha s. Taken together with our previous studies these data support the hypothesis that the receptors and guanine nucleotide-binding proteins which mediate sympathetic and parasympathetic responsiveness in the heart are reciprocally regulated.
- Published
- 1989
47. Inhibition of sodium channels by D600.
- Author
-
Galper JB and Catterall WA
- Subjects
- Binding, Competitive, Calcium metabolism, Cells, Cultured, Myocardium metabolism, Neoplasms, Experimental metabolism, Neuroblastoma metabolism, Veratridine pharmacology, Gallopamil pharmacology, Ion Channels drug effects, Sodium metabolism, Verapamil analogs & derivatives
- Published
- 1979
48. Reconstitution of muscarinic cholinergic inhibition of adenylate cyclase activity in homogenates of embryonic chick hearts by membranes of adult chick hearts.
- Author
-
Liang BT and Galper JB
- Subjects
- Adenylate Cyclase Toxin, Animals, Carbachol pharmacology, Chick Embryo, Chickens, Guanosine Triphosphate pharmacology, Isoproterenol pharmacology, Pertussis Toxin, Quinuclidinyl Benzilate metabolism, Virulence Factors, Bordetella pharmacology, Adenylyl Cyclase Inhibitors, Cell Extracts pharmacology, Cell Membrane analysis, Heart embryology, Myocardium enzymology, Receptors, Muscarinic metabolism, Tissue Extracts pharmacology
- Abstract
We have previously demonstrated that during embryonic development of the chick heart between days 2 1/2 and 10 days in ovo, muscarinic cholinergic inhibition of isoproterenol-stimulated adenylate cyclase activity increased 4-fold, and the sensitivity of isoproterenol-stimulated adenylate cyclase activity to inhibition by carbamylcholine increased 26-fold. Although the number of muscarinic receptors remained constant between days 2 1/2 and 10 in ovo, the levels of a 39- and 41-kDa pertussis toxin substrate increased in parallel with the ability of muscarinic agonist to inhibit adenylate cyclase activity (Liang. B.T., Hellmich, M. R., Neer, E. J., and Galper, J. B. (1986) J. Biol. Chem. 261, 9011-9021). These data are consistent with the hypothesis that between days 2 1/2 and 10 in ovo muscarinic receptors were uncoupled from inhibition of adenylate cyclase activity because of limiting levels of pertussis toxin substrates. In the current studies, in order to test this hypothesis homogenates of embryonic chick hearts 3 1/2 days in ovo were reconstituted with membranes from hearts of hatched chicks. In order to rule out reconstitution by factors from hatched chick hearts other than pertussis toxin substrates, muscarinic receptors from hatched chick hearts were inactivated by covalent binding of benzilycholine mustard and adenylate cyclase inactivated by N-ethylmaleimide prior to reconstitution. Reconstitution of benzilylcholine mustard/N-ethylmaleimide treated hatched chick heart membranes with homogenates of embryonic chick hearts 3 1/2 days in ovo resulted in a 2 1/2-fold increase in the ability of carbamylcholine to inhibit adenylate cyclase activity and reconstitution of hatched chick heart membranes with homogenates of hearts 2 1/2 days in ovo resulted in an approximately 10-fold increase in the sensitivity of isoproterenol-stimulated adenylate cyclase activity to inhibition by carbamylcholine. Membranes from hearts of hatched chicks which had been injected with pertussis toxin were incapable of reconstituting muscarinic inhibition of adenylate cyclase activity in homogenates of hearts 3 1/2 days in ovo. These data support the conclusion that early in embryonic development coupling of muscarinic receptors to inhibition of adenylate cyclase activity is limited by the availability of a pertussis toxin substrate.
- Published
- 1987
49. The biphasic response of muscarinic cholinergic receptors in cultured heart cells to agonists. Effects on receptor number and affinity in intact cells and homogenates.
- Author
-
Galper JB, Dziekan LC, O'Hara DS, and Smith TW
- Subjects
- Animals, Cell Membrane metabolism, Cells, Cultured, Chick Embryo, Kinetics, N-Methylscopolamine, Receptors, Muscarinic drug effects, Carbachol pharmacology, Guanosine Triphosphate analogs & derivatives, Guanylyl Imidodiphosphate pharmacology, Myocardium metabolism, Quinuclidines metabolism, Quinuclidinyl Benzilate metabolism, Receptors, Cholinergic metabolism, Receptors, Muscarinic metabolism, Scopolamine Derivatives metabolism
- Abstract
A biphasic time course of the agonist-mediated loss of muscarinic cholinergic receptors has been demonstrated in cultured chick embryo heart cells by radioligand binding studies using the muscarinic antagonist [3H]quinuclidinyl benzilate ([3H]QNB). This agonist-mediated receptor loss was associated with decreased affinity of the receptor for agonist as judged by competitive binding of the agonist carbamylcholine with [3H]QNB to cell homogenates (Galper, J. B., and Smith, T. W. (1980) J. Biol. Chem. 255, 9571-9579). In the current studies the concentration dependence of agonist-mediated receptor loss was also found to be biphasic. The apparent shift of affinity following brief (15 min) agonist exposure coincided with the agonist-mediated loss of a subclass of high affinity receptors with an IC50 for carbamylcholine inhibition of [3H]QNB binding of 3.9 x 10(-7) M. Those receptors remaining constituted a subclass of low affinity receptors with IC50 = 8.2 x 10(-5) M. The data further suggest that an apparent decrease in agonist affinity after guanine nucleotide exposure represents conversion of high affinity receptors to a similar low affinity state, IC50 = 8.6 x 10(-5) M. The rapid loss of [3H]QNB binding sites in the presence of agonist did not require interaction of agonist with intact cells, but also occurred if cells were homogenized and then subjected to a brief (15 min) exposure to agonist. The slow loss over 3 h of [3H]QNB binding sites could only be demonstrated in intact cells incubated with agonist prior to homogenization. To probe further the later phase of agonist-mediated receptor loss, we developed new assay methods for determining muscarinic antagonist binding to intact cells. In control cells, binding of the hydrophobic antagonist [3H]QNB was quite similar in extent to binding of the more hydrophilic antagonist [3H]methylscopolamine ([3H]MS), with Kd values of 0.11 and 0.47 nM, respectively. Kinetic analysis of the binding of these two ligands was performed to determine whether they might distinguish between two states of the receptor. Both ligands bound to the receptor by a two step mechanism consistent with the formation of a low affinity complex followed by conversion to a high affinity complex. However, the ratio of reverse to forward rate constants of the second step of [3H]MS binding was roughly 100-fold greater than that for the more hydrophobic ligand [3H]QNB. Comparison of the time course of agonist-induced receptor loss as measured by binding of [3H]MS or [3H]QNB was consistent with muscarinic agonist mediation of a stepwise alteration in receptor configuration from a form that bound both [3H]MS and [3H]QNB to a form that bound only [3H]QNB and thence to a form that bound neither [3H]MS nor [3H]QNB. The relationship of such a sequential mechanism to agonist-induced changes in the relationship of the receptor to the cell membrane and agonist-induced endocytosis of the receptor is discussed.
- Published
- 1982
50. Differential sensitivity of alpha o and alpha i to ADP-ribosylation by pertussis toxin in the intact cultured embryonic chick ventricular myocyte. Relationship to the role of G proteins in the coupling of muscarinic cholinergic receptors to inhibition of adenylate cyclase activity.
- Author
-
Liang BT and Galper JB
- Subjects
- Animals, Chick Embryo, Dose-Response Relationship, Drug, Heart embryology, Kinetics, Adenosine Diphosphate Ribose metabolism, Adenylate Cyclase Toxin, Adenylyl Cyclase Inhibitors, GTP-Binding Proteins metabolism, Myocardium metabolism, Pertussis Toxin, Receptors, Muscarinic metabolism, Virulence Factors, Bordetella pharmacology
- Abstract
The guanine nucleotide regulatory proteins, alpha i and alpha o, coexist in a variety of tissues, including heart, brain, and adipose tissues and are ADP-ribosylated by pertussis toxin (Gilman AG, G-proteins and dual control of adenylate cyclase. Cell 26: 577-579, 1984). Previous studies in which purified G proteins were reconstituted with cell membranes and/or phospholipid vesicles have suggested that an alpha i-like protein mediates GTP-dependent inhibition of adenylate cyclase activity. However, direct studies comparing the role of alpha i and alpha o in mediating the inhibition of adenylate cyclase activity in the intact cell have not appeared. In the present study, we demonstrated that, in the intact cell, alpha o was more sensitive to ADP-ribosylation in the presence of pertussis toxin than was alpha i. The T1/2 for pertussis toxin-mediated ADP-ribosylation of alpha i was 199 +/- 10 min (mean +/- SE, N = 10) compared to 157 +/- 7 min for alpha o. The IC50 for pertussis toxin-induced ADP-ribosylation of alpha i was 158 +/- 40 pg/ml (mean +/- SE, N = 11) compared to 35 +/- 8 pg/ml for alpha o. The differences in both T1/2 and IC50 for alpha i and alpha o were statistically significant (P less than 0.001). Studies were carried out to determine whether alpha o was involved in coupling the muscarinic cholinergic receptor to inhibition of adenylate cyclase activity in intact cells. The time course and dose dependence of the pertussis toxin-induced uncoupling of the muscarinic receptor from inhibition of adenylate cyclase closely paralleled the time course and dose dependence for the ADP-ribosylation of alpha i but differed significantly (P less than 0.001) from the time course and dose dependence for the ADP-ribosylation of alpha i but differed significantly (P less than 0.001) from the time course and dose dependence of the pertussis toxin mediated ADP-ribosylation of alpha o. The T1/2 and IC50 values for the pertussis toxin-induced decrease in the inhibition of adenylate cyclase activity were 210 +/- 6 min (mean +/- SE, N = 11) and 169 +/- 25 pg/ml (mean +/- SE, N = 12), respectively, which were not significantly different from the T1/2 and IC50 for pertussis toxin mediated ADP-ribosylation of alpha i. The data are consistent with the hypothesis that, in the intact cell, a pertussis toxin-sensitive alpha i-like protein, but not alpha o, couples muscarinic receptors to inhibition of adenylate cyclase activity.
- Published
- 1988
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.