Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 198
Filter
1.
J Cell Biol ; 135(6 Pt 1): 1609-17, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8978826

ABSTRACT

Angiotensin II (Ang II) stimulates expression of tyrosine hydroxylase and norepinephrine transporter genes in brain neurons; however, the signal-transduction mechanism is not clearly defined. This study was conducted to determine the involvement of the mitogen-activated protein (MAP) kinase signaling pathway in Ang II stimulation of these genes. MAP kinase was localized in the perinuclear region of the neuronal soma. Ang II caused activation of MAP kinase and its subsequent translocation from the cytoplasmic to nuclear compartment, both effects being mediated by AT1 receptor subtype. Ang II also stimulated SRE- and AP1-binding activities and fos gene expression and its translocation in a MAP kinase-dependent process. These observations are the first demonstration of a downstream signaling pathway involving MAP kinase in Ang II-mediated neuromodulation in noradrenergic neurons.


Subject(s)
Angiotensin II/physiology , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Neurons/physiology , Signal Transduction , Animals , Cell Nucleus/metabolism , Cells, Cultured , Enzyme Activation , Neurons/enzymology , Rats , Rats, Inbred WKY , Receptor, Angiotensin, Type 1 , Receptors, Angiotensin/metabolism
2.
J Cell Biol ; 142(1): 217-27, 1998 Jul 13.
Article in English | MEDLINE | ID: mdl-9660875

ABSTRACT

Angiotensin II (Ang II) exerts chronic stimulatory actions on tyrosine hydroxylase (TH), dopamine beta-hydroxylase (DbetaH), and the norepinephrine transporter (NET), in part, by influencing the transcription of their genes. These neuromodulatory actions of Ang II involve Ras-Raf-MAP kinase signal transduction pathways (Lu, D., H. Yang, and M.K. Raizada. 1997. J. Cell Biol. 135:1609-1617). In this study, we present evidence to demonstrate participation of another signaling pathway in these neuronal actions of Ang II. It involves activation of protein kinase C (PKC)beta subtype and phosphorylation and redistribution of myristoylated alanine-rich C kinase substrate (MARCKS) in neurites. Ang II caused a dramatic redistribution of MARCKS from neuronal varicosities to neurites. This was accompanied by a time-dependent stimulation of its phosphorylation, that was mediated by the angiotensin type 1 receptor subtype (AT1). Incubation of neurons with PKCbeta subtype specific antisense oligonucleotide (AON) significantly attenuated both redistribution and phosphorylation of MARCKS. Furthermore, depletion of MARCKS by MARCKS-AON treatment of neurons resulted in a significant decrease in Ang II-stimulated accumulation of TH and DbetaH immunoreactivities and [3H]NE uptake activity in synaptosomes. In contrast, mRNA levels of TH, DbetaH, and NET were not influenced by MARKS-AON treatment. MARCKS pep148-165, which contains PKC phosphorylation sites, inhibited Ang II stimulation of MARCKS phosphorylation and reduced the amount of TH, DbetaH, and [3H]NE uptake in neuronal synaptosomes. These observations demonstrate that phosphorylation of MARCKS by PKCbeta and its redistribution from varicosities to neurites is important in Ang II-induced synaptic accumulation of TH, DbetaH, and NE. They suggest that a coordinated stimulation of transcription of TH, DbetaH, and NET, mediated by Ras-Raf-MAP kinase followed by their transport mediated by PKCbeta-MARCKS pathway are key in persistent stimulation of Ang II's neuromodulatory actions.


Subject(s)
Angiotensin II/metabolism , Brain/metabolism , Intracellular Signaling Peptides and Proteins , Membrane Proteins , Neurons/metabolism , Protein Kinase C/metabolism , Proteins/metabolism , Amino Acid Sequence , Angiotensin II/pharmacology , Animals , Brain/cytology , Cells, Cultured , Dopamine beta-Hydroxylase/metabolism , Molecular Sequence Data , Myristoylated Alanine-Rich C Kinase Substrate , Neurons/drug effects , Norepinephrine/metabolism , Proteins/genetics , Rats , Rats, Inbred WKY , Signal Transduction , Tyrosine 3-Monooxygenase/metabolism
3.
Science ; 210(4471): 791-3, 1980 Nov 14.
Article in English | MEDLINE | ID: mdl-6254147

ABSTRACT

The organum vasculosum of the lamina terminalis has been implicated as the site of receptors mediating central responses of angiotensin II. Up to now, this had been based on indirect evidence, but direct visualization of angiotensin II at its site of action has now been achieved by the use of a biologically active fluorescent angiotensin II agonist. The ventricular surface of the organum vasculosum lamina terminalis showed intense fluorescence, which was virtually eliminated by an excess of unlabeled angiotensin II.


Subject(s)
Angiotensin II/metabolism , Cerebral Ventricles/metabolism , Receptors, Angiotensin/metabolism , Receptors, Cell Surface/metabolism , Angiotensin II/physiology , Animals , Drinking Behavior/physiology , Male , Microscopy, Fluorescence , Rats
4.
Circ Res ; 87(12): 1118-22, 2000 Dec 08.
Article in English | MEDLINE | ID: mdl-11110768

ABSTRACT

Systemic hypertension is a pathophysiological state that is manifested as high blood pressure and is a major risk factor for stroke, ischemic heart disease, peripheral vascular disease, and progressive renal damage. Pulmonary hypertension occurs in 3 distinct forms: primary pulmonary hypertension, pulmonary hypertension of the newborn, or secondary pulmonary hypertension attributable to a variety of lung and cardiovascular diseases. This review discusses the use of gene therapy in the control of systemic and pulmonary hypertension. Overexpression of vasodilator genes as well as antisense knockdown of vasoconstrictor genes has been successfully used in animal models of both forms of hypertension. Furthermore, the use of viral vectors to deliver these constructs has achieved long-term control of hypertension. The successful establishment of gene therapy techniques in the animal models of hypertension coupled with the anticipated advances in the genetic aspects of this disease would make it highly feasible to attempt gene delivery in the control of human hypertension.


Subject(s)
Genetic Therapy , Hypertension/therapy , Adrenergic Antagonists/therapeutic use , Animals , Forecasting , Humans , Hypertension/genetics , Hypertension/metabolism , Oligonucleotides, Antisense/therapeutic use , Receptors, Adrenergic, beta-1/drug effects , Receptors, Adrenergic, beta-1/genetics , Receptors, Adrenergic, beta-1/metabolism , Renin-Angiotensin System , Vasodilator Agents/metabolism
5.
Circ Res ; 86(11): 1167-72, 2000 Jun 09.
Article in English | MEDLINE | ID: mdl-10850969

ABSTRACT

Our previous studies have shown that neonatal delivery of angiotensin type 1 receptor antisense (AT(1)R-AS) in a retroviral vector prevents spontaneously hypertensive rats from developing hypertension for life but has no effect on blood pressure (BP) in normotensive animals. Based on these results, we hypothesized that AT(1)R-AS transduction in normotensive rats would protect them from developing experimental hypertension. The present study was designed to evaluate this hypothesis. A single intracardiac administration of AT(1)R-AS by a retroviral-mediated delivery system (LNSV-AT(1)R-AS) in 5-day-old normotensive Sprague-Dawley rats resulted in long-term expression of the AT(1)R-AS without an effect on basal BP. However, angiotensin II (Ang II)-induced BP, dipsogenic responses, and renovascular contractility were significantly attenuated in the LNSV-AT(1)R-AS-treated rats. Chronic infusion of low-dose Ang II (55 ng. kg(-)(1). min(-)(1)) in LNSV-alone-treated rats caused a modest increase in BP, profound increase in cardiac hypertrophy, and increased vascular contractility. In contrast, the LNSV-AT(1)R-AS-treated rats were protected from developing these changes after Ang II infusion. These data establish that LNSV-AT(1)R-AS pretreatment protects healthy rats from developing Ang II-dependent hypertension.


Subject(s)
Blood Pressure/physiology , Hypertension/prevention & control , Oligonucleotides, Antisense/metabolism , Receptors, Angiotensin/genetics , Angiotensin II/administration & dosage , Angiotensin II/pharmacology , Animals , Blood Pressure/drug effects , Cardiomegaly/chemically induced , Cardiomegaly/prevention & control , Dose-Response Relationship, Drug , Drinking/drug effects , Female , Gene Transfer Techniques , Oligonucleotides, Antisense/genetics , Rats , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Reference Values , Renal Circulation/drug effects , Time Factors , Vasoconstriction/drug effects
6.
Circ Res ; 85(10): e44-50, 1999 Nov 12.
Article in English | MEDLINE | ID: mdl-10559146

ABSTRACT

Our previous studies have demonstrated that the introduction of angiotensin II type I receptor antisense (AT(1)R-AS) cDNA by a retrovirally mediated delivery system prevents the development of hypertension in the spontaneously hypertensive rat (SHR), an animal model for primary hypertension in humans. These results have led us to propose the hypothesis that an interruption of the renin-angiotensin system (RAS) activity at a genetic level would prevent hypertension on a permanent basis. F(1) and F(2) generations of offspring from a retroviral vector, LNSV- and LNSV-AT(1)R-AS-treated SHR, were generated, and various physiological parameters indicative of hypertension were studied and compared with those of their parents to investigate this hypothesis. Both F(1) and F(2) generations of LNSV-AT(1)R-AS-treated SHR expressed a persistently lower blood pressure, decreased cardiac hypertrophy and fibrosis, decreased medial thickness, and normalization of renal artery excitation-contraction coupling, Ca(2+) current, and [Ca(2+)](i) when compared with offspring derived from the LNSV-treated SHR. In fact, the magnitude of the prevention of these pathophysiological alterations was similar to that observed in the LNSV-AT(1)R-AS-treated SHR parent. The prevention of cardiovascular pathophysiology and expression of normotensive phenotypes are, at least in part, a result of integration and subsequent transmission of AT(1)R-AS from the SHR parents to offspring. These data demonstrate that a single intracardiac injection of LNSV-AT(1)R-AS causes a permanent cardiovascular protection against hypertension as a result of a genomic integration and germ line transmission of the AT(1)R-AS in the SHR offspring.


Subject(s)
DNA, Antisense/therapeutic use , Hypertension/genetics , Hypertension/prevention & control , Receptors, Angiotensin/genetics , Animals , Animals, Newborn , Antihypertensive Agents/pharmacology , Aorta/pathology , Blood Pressure/drug effects , Blood Pressure/genetics , Fibrosis/genetics , Genetic Therapy , Kidney/blood supply , Kidney/physiopathology , Losartan/pharmacology , Myocardium/pathology , Organ Size/genetics , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Transfection
7.
Mol Endocrinol ; 6(12): 2009-17, 1992 Dec.
Article in English | MEDLINE | ID: mdl-1491687

ABSTRACT

Angiotensin-II (AII) stimulates plasminogen activator inhibitor-1 (PAI-1) gene transcription, translation, and protein secretion from astroglial cells derived from normotensive [Wistar-Kyoto (WKY)] rat brain, an effect mediated by AII type 1 (AT1) receptors. Since abnormal expression of the brain AII system has been demonstrated in spontaneously hypertensive (SH) rats, we investigated the regulation of PAI-1 gene expression by AII in astroglial cells from the brains of these animals. AII caused an increase in PAI-1 gene expression in SH rat astroglia in a manner similar to that observed in WKY-derived cultures. However, both the basal and AII-stimulated levels of PAI-1 mRNA in SH rat astroglia were only 20% of those observed in WKY rat astroglial cultures. Consequently, there was a significant reduction in the de novo synthesis and secretion of PAI-1 from astroglia of SH rat brain. The reduced synthesis and secretion of PAI-1 from SH rat brain astroglia was associated with lower numbers of AT1 receptors in these cells. However, the steady state levels of AT1 receptor mRNA were comparable in both WKY and SH rat astroglia. This reduction in AII-modulated PAI-1 levels in SH rat astroglia is consistent with a proposed role of these interactions in the development of hypertension in these animals.


Subject(s)
Angiotensin II/pharmacology , Astrocytes/drug effects , Gene Expression Regulation/drug effects , Hypertension/metabolism , Plasminogen Activator Inhibitor 1/biosynthesis , Rats, Inbred SHR/metabolism , Renin-Angiotensin System/physiology , Animals , Astrocytes/metabolism , Brain/cytology , Cells, Cultured , Plasminogen Activator Inhibitor 1/genetics , Plasminogen Activator Inhibitor 1/metabolism , Rats , Rats, Inbred WKY/metabolism , Rats, Sprague-Dawley/metabolism , Receptors, Angiotensin/drug effects
8.
Braz J Med Biol Res ; 48(12): 1109-14, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26421871

ABSTRACT

Diabetic retinopathy (DR) is a serious complication of diabetes mellitus that may result in blindness. We evaluated the effects of activation of endogenous angiotensin converting enzyme (ACE) 2 on the early stages of DR. Rats were administered an intravenous injection of streptozotocin to induce hyperglycemia. The ACE2 activator 1-[[2-(dimethylamino) ethyl] amino]-4-(hydroxymethyl)-7-[[(4-methylphenyl) sulfonyl] oxy]-9H-xanthone 9 (XNT) was administered by daily gavage. The death of retinal ganglion cells (RGC) was evaluated in histological sections, and retinal ACE2, caspase-3, and vascular endothelial growth factor (VEGF) expressions were analyzed by immunohistochemistry. XNT treatment increased ACE2 expression in retinas of hyperglycemic (HG) rats (control: 13.81±2.71 area%; HG: 14.29±4.30 area%; HG+XNT: 26.87±1.86 area%; P<0.05). Importantly, ACE2 activation significantly increased the RCG number in comparison with HG animals (control: 553.5±14.29; HG: 530.8±10.3 cells; HG+XNT: 575.3±16.5 cells; P<0.05). This effect was accompanied by a reduction in the expression of caspase-3 in RGC of the HG+XNT group when compared with untreated HG rats (control: 18.74±1.59; HG: 38.39±3.39 area%; HG+XNT: 27.83±2.80 area%; P<0.05). Treatment with XNT did not alter the VEGF expression in HG animals (P>0.05). Altogether, these findings indicate that activation of ACE2 reduced the death of retinal ganglion cells by apoptosis in HG rats.


Subject(s)
Hyperglycemia/complications , Peptidyl-Dipeptidase A/metabolism , Retinal Diseases/etiology , Retinal Diseases/prevention & control , Secondary Prevention/methods , Administration, Oral , Angiotensin-Converting Enzyme 2 , Animals , Apoptosis , Caspase 3/metabolism , Cell Proliferation/physiology , Cell Survival/physiology , Diabetes Mellitus, Experimental/metabolism , Enzyme Activation , Hyperglycemia/chemically induced , Immunohistochemistry , Male , Peptidyl-Dipeptidase A/drug effects , Rats, Wistar , Retinal Diseases/metabolism , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , Streptozocin , Vascular Endothelial Growth Factor A/metabolism , Xanthones/administration & dosage
9.
Br J Pharmacol ; 172(9): 2219-31, 2015 May.
Article in English | MEDLINE | ID: mdl-25522140

ABSTRACT

BACKGROUND AND PURPOSE: Pulmonary hypertension (PH) is a devastating disease characterized by increased pulmonary arterial pressure, which progressively leads to right-heart failure and death. A dys-regulated renin angiotensin system (RAS) has been implicated in the development and progression of PH. However, the role of the angiotensin AT2 receptor in PH has not been fully elucidated. We have taken advantage of a recently identified non-peptide AT2 receptor agonist, Compound 21 (C21), to investigate its effects on the well-established monocrotaline (MCT) rat model of PH. EXPERIMENTAL APPROACH: A single s.c. injection of MCT (50 mg·kg(-1) ) was used to induce PH in 8-week-old male Sprague Dawley rats. After 2 weeks of MCT administration, a subset of animals began receiving either 0.03 mg·kg(-1) C21, 3 mg·kg(-1) PD-123319 or 0.5 mg·kg(-1) A779 for an additional 2 weeks, after which right ventricular haemodynamic parameters were measured and tissues were collected for gene expression and histological analyses. KEY RESULTS: Initiation of C21 treatment significantly attenuated much of the pathophysiology associated with MCT-induced PH. Most notably, C21 reversed pulmonary fibrosis and prevented right ventricular fibrosis. These beneficial effects were associated with improvement in right heart function, decreased pulmonary vessel wall thickness, reduced pro-inflammatory cytokines and favourable modulation of the lung RAS. Conversely, co-administration of the AT2 receptor antagonist, PD-123319, or the Mas antagonist, A779, abolished the protective actions of C21. CONCLUSIONS AND IMPLICATIONS: Taken together, our results suggest that the AT2 receptor agonist, C21, may hold promise for patients with PH.


Subject(s)
Cardiovascular Agents/pharmacology , Hypertension, Pulmonary/prevention & control , Hypertrophy, Right Ventricular/prevention & control , Lung/drug effects , Myocardium , Pulmonary Fibrosis/prevention & control , Receptor, Angiotensin, Type 2/agonists , Ventricular Dysfunction, Right/prevention & control , Angiotensin II/analogs & derivatives , Angiotensin II/pharmacology , Angiotensin II Type 2 Receptor Blockers/pharmacology , Animals , Disease Models, Animal , Fibrosis , Hemodynamics/drug effects , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/physiopathology , Hypertrophy, Right Ventricular/chemically induced , Hypertrophy, Right Ventricular/metabolism , Hypertrophy, Right Ventricular/pathology , Imidazoles/pharmacology , Lung/metabolism , Lung/pathology , Male , Monocrotaline , Myocardium/metabolism , Myocardium/pathology , Peptide Fragments/pharmacology , Proto-Oncogene Mas , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/metabolism , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Pyridines/pharmacology , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 2/metabolism , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects , Vascular Remodeling/drug effects , Ventricular Dysfunction, Right/chemically induced , Ventricular Dysfunction, Right/metabolism , Ventricular Dysfunction, Right/pathology , Ventricular Dysfunction, Right/physiopathology , Ventricular Function, Right/drug effects , Ventricular Remodeling/drug effects
10.
Physiol Genomics ; 7(2): 79-94, 2001 Dec 21.
Article in English | MEDLINE | ID: mdl-11773594

ABSTRACT

This article is based on an Experimental Biology symposium held in April 2001 and presents the current status of gene therapy for cardiovascular diseases in experimental studies and clinical trials. Evidence for the use of gene therapy to limit neointimal hyperplasia and confer myocardial protection was presented, and it was found that augmenting local nitric oxide (NO) production using gene transfer (GT) of NO synthase or interruption of cell cycle progression through a genetic transfer of cell cycle regulatory genes limited vascular smooth muscle hyperplasia in animal models and infra-inguinal bypass patients. The results of application of vascular endothelial growth factor (VEGF) GT strategies for therapeutic angiogenesis in critical limb and myocardial ischemia in pilot clinical trials was reviewed. In addition, experimental evidence was presented that genetic manipulation of peptide systems (i.e., the renin-angiotensin II system and the kallikrein-kinin system) was effective in the treatment of systemic cardiovascular diseases such as hypertension, heart failure, and renal failure. Although, as of yet, there are no well controlled human trials proving the clinical benefits of gene therapy for cardiovascular diseases, the data presented here in animal models and in human subjects show that genetic targeting is a promising and encouraging modality, not only for the treatment and long-term control of cardiovascular diseases, but for their prevention as well.


Subject(s)
Cardiovascular Diseases/therapy , Gene Targeting/methods , Genetic Therapy/methods , Animals , Cardiovascular Diseases/genetics , Cell Cycle Proteins/biosynthesis , Cell Cycle Proteins/genetics , Cell Cycle Proteins/therapeutic use , Clinical Trials as Topic , Congresses as Topic , Endothelial Growth Factors/genetics , Endothelial Growth Factors/therapeutic use , Gene Targeting/trends , Genetic Therapy/adverse effects , Genetic Therapy/trends , Graft Occlusion, Vascular/prevention & control , Humans , Lymphokines/genetics , Lymphokines/therapeutic use , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Neovascularization, Physiologic/drug effects , Nitric Oxide Synthase/biosynthesis , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/therapeutic use , Renin-Angiotensin System/drug effects , Renin-Angiotensin System/physiology , Treatment Outcome , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
11.
Endocrinology ; 142(8): 3502-11, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11459796

ABSTRACT

Interaction of angiotensin II with the neuronal angiotensin type 1 receptor stimulates the PI3K signaling pathway. Our objective in this study was to investigate the hypothesis that the PI3K cascade regulates the neurotropic actions of angiotensin II in rat brain neurons. We followed growth associated protein-43 expression and neurite extension as markers of neurotropic activity. Angiotensin II, through its interaction with the angiotensin type 1 receptor, increased growth associated protein-43 expression and neurite extension. These effects were abolished by pretreatment of neurons with wortmannin and rapamycin, but not by PD 98059. Antisense oligonucleotides specific for p70(S6) kinase also inhibited angiotensin II-stimulated neurotropic activity. These data confirm the involvement of PI3K and p70(S6) kinase in angiotensin II-mediated neurotropic action. Further support for this was provided by the observation that angiotensin II caused a time-dependent stimulation of p70(S6) kinase by an angiotensin type 1 receptor-mediated process. We also found that the neurotropic actions of angiotensin II are mediated by plasminogen activator inhibitor-1. Evidence for this includes 1) angiotensin II-stimulated neuronal plasminogen activator inhibitor-1 gene expression, 2) potent neurotropic action of exogenous plasminogen activator inhibitor-1, and 3) inhibitory neurotropic effect of angiotensin II by antisense oligonucleotide-mediated depletion of plasminogen activator inhibitor-1. Finally, we found that the neurotropic action of plasminogen activator inhibitor-1 is not blocked by either angiotensin type 1 receptor antagonist or inhibitors of PI3K or p70(S6) kinase, indicating that the plasminogen activator inhibitor-1 step is downstream from the p70(S6) kinase. These observations demonstrate that angiotensin II is a neurotropic hormone that engages a distinct PI3K-p70(S6) kinase-plasminogen activator inhibitor-1 signaling pathway for this action.


Subject(s)
Angiotensin II/physiology , Brain/physiology , Nerve Growth Factors/physiology , Neurons/physiology , Signal Transduction/physiology , Angiotensin II/pharmacology , Animals , Brain/cytology , Brain/drug effects , Cells, Cultured , GAP-43 Protein/metabolism , Neurons/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Plasminogen Activator Inhibitor 1/pharmacology , Plasminogen Activator Inhibitor 1/physiology , Rats , Rats, Inbred WKY , Ribosomal Protein S6 Kinases/metabolism
12.
Endocrinology ; 132(4): 1715-22, 1993 Apr.
Article in English | MEDLINE | ID: mdl-8462471

ABSTRACT

In this study we compared the expression of angiotensin II type 1 (AT1) receptor messenger RNA (mRNA) and AT1 receptors in neurons cultured from Wistar-Kyoto (WKY) and spontaneously hypertensive (SH) rat brains. Neuronal cultures from the hypothalamus and brain-stem of 1-day-old SH rats exhibited approximately 4-fold higher steady-state levels of AT1 receptor mRNA than the corresponding WKY cultures. This was attributable to greater levels of both AT1A and AT1B receptor mRNA subtypes in SH rat neuronal cultures compared with WKY rat neurons. SH rat neuronal cultures also exhibited increased numbers (approximately 2.3-fold) of binding sites for [3H]DuP753, an AT1 receptor selective ligand, and enhanced (approximately 3.4-fold) stimulation of inositol phospholipid hydrolysis by angiotensin II compared with WKY neurons. By contrast, cultured astroglia from SH and WKY rat brain exhibited no significant differences in either the levels of AT1 receptor mRNA or the specific binding of [3H]DuP753. These data suggest that in SH rat neurons, AT1 receptor transcription and translation is increased, compared with neurons from WKY rats.


Subject(s)
Gene Expression , Neurons/physiology , Receptors, Angiotensin/genetics , Amino Acid Sequence , Angiotensin II/pharmacology , Animals , Biphenyl Compounds/metabolism , Cells, Cultured , Hydrolysis/drug effects , Imidazoles/metabolism , Inositol/metabolism , Losartan , Molecular Sequence Data , RNA, Messenger/metabolism , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Tetrazoles/metabolism
13.
Endocrinology ; 107(5): 1652-5, 1980 Nov.
Article in English | MEDLINE | ID: mdl-7000499

ABSTRACT

Investigation of mechanisms responsible for the decreased numbers of insulin receptors observed in obesity and diabetes has been facilitated by the development of cell culture systems permitting study of cellular events independent of fluctuating hormone levels and multiple endocrine interactions present in the whole organism. With such a system, we have found that cells cultured from the skin of diabetic mice have 45-48% fewer receptors for insulin than those from nondiabetic littermates. This difference is maintained in culture over many generations, suggesting that the decreased expression of insulin receptors in these cells is related to the genetic trait for diabetes.


Subject(s)
Diabetes Mellitus/metabolism , Receptor, Insulin/metabolism , Skin/cytology , Animals , Cell Division , Cells, Cultured , Fibroblasts/metabolism , Insulin/metabolism , Male , Mice , Mice, Inbred C57BL
14.
Endocrinology ; 137(8): 3566-76, 1996 Aug.
Article in English | MEDLINE | ID: mdl-8754788

ABSTRACT

In the present study we investigated the regulation of tyrosine hydroxylase (TH) by angiotensin II (Ang II) in an attempt to provide cellular and molecular evidence that this hormone has increased neuromodulatory actions in the spontaneously hypertensive (SH) rat brain. Neuronal cells in primary culture from the hypothalamus-brain stem of both normotensive [Wistar-Kyoto (WKY)] and SH rats have been used. These cultures mimic in vivo situations. Ang II caused a time-dependent increase in TH activity in WKY rat brain neurons. A maximal increase of 2.5-fold was observed with 100 nM Ang II in an actinomycin- and cycloheximide-dependent process. In addition, Ang II caused a parallel increase in TH messenger RNA (mRNA) levels, with a maximal stimulation of 5-fold in 4 h by 100 nM Ang II in WKY rat brain neurons. The stimulation of TH mRNA was mediated by the AT1 receptor subtype, resulted from an increase in its transcription, and involved activation of phospholipase C and protein kinase C. Antisense oligonucleotide for c-fos attenuated Ang II stimulation of TH mRNA in a time- and dose-dependent fashion, indicating an involvement of c-fos as a putative third messenger in Ang II stimulation of TH. Ang II also caused stimulation of TH activity and its mRNA levels in neuronal cultures of SH rat brain by a mechanism similar to that observed for neuronal cultures of WKY rat brain, involving AT1 receptors, protein kinase C, and c-fos. However, the stimulation of TH activity and that of TH mRNA were approximately 30% and 80% higher, respectively, in the SH rat brain neurons than those in the WKY rat brain neurons. In vivo experiments have been carried out to validate the elevated response of TH gene expression to Ang II in SH rat brain neuronal cultures. Ang II stimulated both TH activity and TH mRNA levels in the hypothalami and brain stems of adult WKY and SH rats. The level of stimulation in the brain of the SH rat was significantly higher than that in the WKY rat. These observations are consistent with an increase in AT1, receptor gene expression and suggest that increased TH gene expression could be the cellular/molecular basis for the greater neuromodulatory action of Ang II in the SH rat brain.


Subject(s)
Angiotensin II/pharmacology , Gene Expression/drug effects , Neurons/enzymology , Rats, Inbred SHR/genetics , Tyrosine 3-Monooxygenase/genetics , Animals , Base Sequence , Brain/cytology , Brain/metabolism , Cells, Cultured , Molecular Sequence Data , Oligonucleotide Probes/genetics , RNA, Messenger/metabolism , Rats , Rats, Inbred WKY/genetics , Rats, Sprague-Dawley , Reference Values , Tyrosine 3-Monooxygenase/metabolism
15.
Endocrinology ; 142(3): 1009-16, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11181513

ABSTRACT

Angiotensin (Ang II) activates neuronal AT(1) receptors located in the hypothalamus and the brainstem and stimulates noradrenergic neurons that are involved in the control of blood pressure and fluid intake. In this study we used complementary DNA microarrays for high throughput gene expression profiling to reveal unique genes that are linked to the neuromodulatory actions of Ang II in neuronal cultures from newborn rat hypothalamus and brainstem. Of several genes that were regulated, we focused on calmodulin and synapsin I. Ang II (100 nM; 1-24 h) elicited respective increases and decreases in the levels of calmodulin and synapsin I messenger RNAs, effects mediated by AT(1) receptors. This was associated with similar changes in calmodulin and synapsin protein expression. The actions of Ang II on calmodulin expression involve an intracellular pathway that includes activation of phospholipase C, increased intracellular calcium, and stimulation of protein kinase C. Taken together with studies that link calmodulin and synapsin I to axonal transport and exocytotic processes, the data suggest that Ang II regulates these two proteins via a Ca(2+)-dependent pathway, and that this may contribute to longer term or slower neuromodulatory actions of this peptide.


Subject(s)
Angiotensin II/physiology , Brain/physiology , Calmodulin/metabolism , Gene Expression , Neurons/physiology , Synapsins/metabolism , Angiotensin II/pharmacology , Animals , Brain/cytology , Brain/drug effects , Calcium/physiology , Calmodulin/physiology , Cells, Cultured , Dopamine beta-Hydroxylase/metabolism , Neurons/drug effects , Neurotransmitter Agents/physiology , Protein Kinase C/physiology , Rats , Rats, Sprague-Dawley , Type C Phospholipases/physiology , Tyrosine 3-Monooxygenase/metabolism
16.
Endocrinology ; 139(1): 365-75, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9421435

ABSTRACT

Angiotensin II (Ang II) interaction with the neuronal AT1 receptor results in a chronic stimulation of neuromodulation that involves the expression of norepinephrine transporter (NET) and tyrosine hydroxylase (TH). In view of this unique property and the presence of putative nuclear localization signal (NLS) consensus sequence in the AT1 receptor, this study was conducted to investigate the hypothesis that Ang II would induce nuclear sequestration of this G protein-coupled receptor and that the sequestration may have implications on Ang II-induced expression of NET and TH genes. Incubation of neuronal cultures with Ang II caused a time- and dose-dependent increase in the levels of AT1 receptor immunoreactivity in the nucleus. A 6.7-fold increase was observed with 100 nM Ang II, in 15 min, that was blocked by losartan, an AT1 receptor-specific antagonist. Ang II-induced nuclear sequestration was specific for AT1 receptor, because Ang II failed to produce a similar effect on neuronal AT2 receptors. The presence of the putative NLS sequence in the cytoplasmic tail of the AT1 receptor seems to be the key in nuclear targeting because: 1) nuclear targeting was attenuated by a peptide of the AT1 receptor that contained the putative NLS sequence; and 2) Ang II failed to cause nuclear translocation of the AT2 receptor, which does not contain the putative NLS. Ang II also caused a time- and dose-dependent stimulation of P62 phosphorylation, a glycoprotein of the nuclear pore complex. A 6-fold stimulation of phosphorylation was observed with 100 nM Ang II, in 15 min, that was completely blocked by losartan and not by PD123,319, an AT2 receptor specific antagonist. Preloading of neurons with p62-pep (a peptide containing consenses of mitogen-activated protein kinase in p62) resulted in a loss of Ang II-induced p62 phosphorylation and stimulation of NET and TH messenger RNA levels. In conclusion, these data demonstrate that Ang II induces nuclear sequestration of AT1 receptor involving NLS in the AT1 receptor and p62 of the nuclear pore complex in brain neurons. A possible role of such a nuclear targeting of the AT1 receptor on chronic neuromodulatory actions of Ang II has been discussed.


Subject(s)
Angiotensin II/pharmacology , Cell Nucleus/metabolism , Receptors, Angiotensin/metabolism , Amino Acid Sequence , Animals , Brain/metabolism , Molecular Sequence Data , Neurons/metabolism , Rats , Rats, Inbred WKY
17.
Endocrinology ; 130(3): 1255-62, 1992 Mar.
Article in English | MEDLINE | ID: mdl-1537291

ABSTRACT

In this study, we investigated the mechanism of angiotensin II (Ang II) induced secretion of plasminogen activator inhibitor-1 (PAI-1) from astroglial cells prepared from 21-day-old rat brain. Competition-inhibition experiments with the use of selective antagonists for Ang II receptor subtypes indicated that astroglial cells contain chiefly Ang II type 1 (AT1) receptors. The interaction of Ang II with AT1 receptors resulted in a time- and concentration-dependent stimulation of PAI-1 gene expression. A maximal, 20-fold induction of PAI-1 messenger RNA (mRNA) steady-state levels was observed with 10 nM Ang II. This effect of Ang II was blocked by DuP753, an AT1 receptor antagonist, but not by PD123177, an AT2 receptor antagonist. Raise in PAI-1 mRNA levels was followed by an elevation in PAI-1 concentration in culture media reaching its maximum after 24 h. Interaction of Ang II with AT1 receptors also resulted in a time- and concentration-dependent stimulation of inositol phospholipid (IP) hydrolysis. A maximal, 3- to 5-fold stimulation of IP hydrolysis was observed with 10 nM Ang II. The time course experiments indicated that Ang II-induced stimulation of IP hydrolysis precedes the stimulation of PAI-1 mRNA. This suggested that activation of phospholipase C, IP hydrolysis system and possibly protein kinase C (PKC) may mediate Ang II's effect on PAI-1 mRNA. Direct stimulation of PKC by phorbol ester, phorbol 12,13-dibutyrate (PDB), resulted in a time- and concentration-dependent elevation of PAI-1 mRNA levels, similar to that caused by Ang II (maximal stimulation of 20-fold with 100 nM PDB for 4 h). This effect was totally blocked by the protein kinase C inhibitor, H7. In addition, Ang II stimulation of PAI-1 mRNA was also blocked by H7. In contrast, Ang II did not elevate PAI-1 mRNA levels in astroglial cultures from neonatal rat brains. However, treatment of neonatal cultures with PDB increased levels of this mRNA species. These observations indicate that the coupling of AT1 receptors with IP hydrolysis and PKC activation may be important for Ang II stimulation of PAI-1 gene expression. The lack of Ang II's effect on PAI-1 mRNA in neonatal astroglia may be explained either by a low coupling efficiency between AT1 receptors and the second messenger system, or by a low AT1 to AT2 receptor level ratio.


Subject(s)
Angiotensin II/pharmacology , Astrocytes/chemistry , Gene Expression/drug effects , Plasminogen Inactivators/analysis , Animals , Antineoplastic Agents/pharmacology , Astrocytes/ultrastructure , Biphenyl Compounds/pharmacology , Blotting, Western , Brain Chemistry/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Gene Expression/genetics , Polyenes/pharmacology , RNA, Messenger/analysis , RNA, Messenger/genetics , Rats , Receptors, Angiotensin/analysis , Time Factors
18.
Endocrinology ; 129(2): 1066-74, 1991 Aug.
Article in English | MEDLINE | ID: mdl-1713158

ABSTRACT

Brain astrocytes were established in primary culture from postnatal and adult rats to characterize the developmental expression of secreted proteins. Astrocytes cultured from 21-day rat brain, but not 1-day rat brain, secreted a distinct group of proteins with Mr of 35,000 as determined by analysis of [35S]methionine-labeled proteins using two-dimensional sodium dodecyl sulfate-polyacrylamide gel electrophoresis. N-terminal amino acid sequence analysis of this protein group showed 100% identity to rat insulin-like growth factor binding protein-2 (rIGFBP-2), the BRL-3A IGFBP purified from a fetal rat liver cell line. An antiserum was generated against this astrocyte 35,000 Mr protein, and immunoblot analysis revealed a dramatic increase in rIGFBP-2 secretion in astrocytes cultured from 14-day, 21-day, and adult rat brain compared to astrocytes from 1-day and 7-day rat brain. Similar analysis of neonatal rat brain neurons in culture failed to show immunoreactive rIGFBP-2 in cell lysates or secreted protein. Ligand Western blot analysis demonstrated [125I]IGF-II binding to a single protein band which comigrated with a prominant rIGFBP-2 immunoreactive species in nonreduced conditioned medium from 21-day astrocytes. In comparison [125I]IGF-II binding proteins were detected only at low levels in medium from astrocytes cultured from 1-day rat brain and were undetectable in neuron-conditioned media. Northern blot analysis using a rIGFBP-2 complementary DNA revealed 5-fold greater messenger RNA levels in astrocytes from 21-day rat brain compared with astrocytes from 1-day brain, whereas neonate neurons showed no transcripts. Thus, rIGFBP-2 exhibits a pattern of developmental and cell-specific expression in cultured rat brain cells.


Subject(s)
Aging/metabolism , Astrocytes/metabolism , Brain/growth & development , Carrier Proteins/biosynthesis , Amino Acid Sequence , Animals , Animals, Newborn/metabolism , Brain/cytology , Carrier Proteins/chemistry , Carrier Proteins/genetics , Electrophoresis, Polyacrylamide Gel , Immunoblotting , Insulin-Like Growth Factor Binding Protein 2 , Insulin-Like Growth Factor II/metabolism , Isoelectric Point , Molecular Sequence Data , Molecular Weight , Nucleic Acid Hybridization , RNA, Messenger/analysis , Rats , Rats, Inbred Strains
19.
Endocrinology ; 128(5): 2548-57, 1991 May.
Article in English | MEDLINE | ID: mdl-1850355

ABSTRACT

Astrocytic glial cells from 1- and 21-day-old rat brains were established in primary culture to study the expression of insulin-like growth factor-I (IGF-I) receptors and IGF-I-stimulated glucose transporter (Glut-1). Astrocytes from both age groups expressed specific high affinity IGF-I receptors, whose relative affinities for IGF-I, IGF-II, and insulin were comparable. However, the total number of binding sites and IGF-I receptor mRNA levels were 148% and 240% higher in astrocytes from 21-day-old compared with 1-day-old brains. IGF-I caused a dose-dependent stimulation of [3H]2-deoxy-D-glucose [( 3H]dGlc) uptake in astrocytes from 1-day-old brains. This was associated with increases in Glut-1 protein and mRNA levels. In contrast, astrocytes from 21-day-old brains exhibited a 58% decrease in the binding capacity and a 77% decrease in the steady state levels of Glut-1 protein and its mRNA. In addition, IGF-I failed to stimulate the Glut-1 system in these cells. This lack of IGF-I effect is not due to an alteration inherent to the Glut-1 system, since 12-O-tetradecanoyl-phorbol-13-acetate stimulated [3H]dGlc uptake and Glut-1 protein and its mRNA levels. These observations suggest that changes in basal and IGF-I-stimulated Glut-1 system in brain astrocytes may be developmentally regulated.


Subject(s)
Astrocytes/metabolism , Brain/metabolism , Insulin-Like Growth Factor I/pharmacology , Monosaccharide Transport Proteins/metabolism , Animals , Animals, Newborn/growth & development , Animals, Newborn/metabolism , Brain/cytology , RNA, Messenger/metabolism , Rats , Receptors, Cell Surface/genetics , Receptors, Somatomedin , Somatomedins/metabolism , Tetradecanoylphorbol Acetate/pharmacology
20.
Endocrinology ; 137(2): 763-72, 1996 Feb.
Article in English | MEDLINE | ID: mdl-8593828

ABSTRACT

Brain angiotensin II (Ang II) plays a key role in blood pressure control in part by interacting with catecholamines (CA) and by stimulation of sympathetic pathways. The significance of Ang-CA interaction is further heightened by the presence of a hyperactive brain Ang II system in spontaneously hypertensive (SH) rat, a genetic model for essential hypertension. Neuronal cells in primary culture from the hypothalamus-brainstem that mimic in vivo situations in so far as many cellular actions of Ang II are concerned, have been used in the present study to elucidate Ang II regulation of CA by determining its cellular action on the norepinephrine transporter (NET) system. Ang II causes both acute and chronic stimulation of [3H]-norepinephrine (NE) uptake in neuronal cultures of Wistar Kyoto (WKY) rat brain. Acute stimulation begins as early as 5 min, reaches maximal levels in about 30 min in the presence of 100 nM Ang II, and is blocked by losartan, a specific antagonist for AT1 receptor subtype. In addition, this acute stimulation appears to be a posttranscriptional event and does not involve protein kinase C (PKC) or NET gene transcription. Chronic stimulation of [3H]-NE uptake by Ang II persists throughout the duration of Ang II incubation (24 h), is dose dependent, and is also mediated by AT1 receptor subtype. However, chronic stimulation of [3H]-NE uptake involves PKC, cfos, and NET gene transcription. Ang II also stimulates [3H]-NE uptake in neuronal cultures of SH rat brain, both acutely and chronically, by mechanisms similar to those observed in neuronal cultures of WKY rat brain. The stimulation of NET by Ang II is 2-fold higher than that seen in WKY and is consistent with increased AT1 receptor gene transcription and increased functional AT1 receptors in SH rat brain neurons compared with WKY rat brain neurons. The Ang II stimulation of the NET system is also higher in adult SH compared with WKY rats in vivo. These observations show that 1) Ang II stimulates the NET system both acutely and chronically, the former involving activation of preexisting transporters and the latter involving NET gene transcription and translation; and 2) Ang II stimulation of the NET system is elevated in SH rat brain neurons.


Subject(s)
Angiotensin II/pharmacology , Brain/cytology , Carrier Proteins/metabolism , Hypertension/metabolism , Neurons/metabolism , Symporters , Animals , Autoradiography , Base Sequence , Brain/metabolism , Brain/pathology , Carrier Proteins/genetics , Cells, Cultured , Hypertension/pathology , Kinetics , Molecular Probes/genetics , Molecular Sequence Data , Norepinephrine Plasma Membrane Transport Proteins , Proto-Oncogene Proteins c-fos/physiology , RNA, Messenger/metabolism , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Reference Values
SELECTION OF CITATIONS
SEARCH DETAIL