Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Cell Metab ; 31(2): 313-326.e5, 2020 02 04.
Article in English | MEDLINE | ID: mdl-31839488

ABSTRACT

Glucose is the essential energy source for the brain, whose deficit, triggered by energy deprivation or therapeutic agents, can be fatal. Increased appetite is the key behavioral defense against hypoglycemia; however, the central pathways involved are not well understood. Here, we describe a glucoprivic feeding pathway by tyrosine hydroxylase (TH)-expressing neurons from nucleus of solitary tract (NTS), which project densely to the hypothalamus and elicit feeding through bidirectional adrenergic modulation of agouti-related peptide (AgRP)- and proopiomelanocortin (POMC)-expressing neurons. Acute chemogenetic inhibition of arcuate nucleus (ARC)-projecting NTSTH neurons or their target, AgRP neurons, impaired glucoprivic feeding induced by 2-Deoxy-D-glucose (2DG) injection. Neuroanatomical tracing results suggested that ARC-projecting orexigenic NTSTH neurons are largely distinct from neighboring catecholamine neurons projecting to parabrachial nucleus (PBN) that promotes satiety. Collectively, we describe a circuit organization in which an ascending pathway from brainstem stimulates appetite through key hunger neurons in the hypothalamus in response to hypoglycemia.


Subject(s)
Agouti-Related Protein/metabolism , Appetite Regulation , Hypoglycemia/metabolism , Hypothalamus/metabolism , Neurons/metabolism , Solitary Nucleus/metabolism , Animals , Female , Hypothalamus/cytology , Male , Mice , Mice, Inbred C57BL , Neurons/cytology , Solitary Nucleus/cytology
2.
JCI Insight ; 52019 06 11.
Article in English | MEDLINE | ID: mdl-31184598

ABSTRACT

Patients with mutations in Cullin-3 (CUL3) exhibit severe early onset hypertension but the contribution of the smooth muscle remains unclear. Conditional genetic ablation of CUL3 in vascular smooth muscle (S-CUL3KO) causes progressive impairment in responsiveness to nitric oxide (NO), rapid development of severe hypertension, and increased arterial stiffness. Loss of CUL3 in primary aortic smooth muscle cells or aorta resulted in decreased expression of the NO receptor, soluble guanylate cyclase (sGC), causing a marked reduction in cGMP production and impaired vasodilation to cGMP analogues. Vasodilation responses to a selective large conductance Ca2+-activated K+-channel activator were normal suggesting that downstream signals which promote smooth muscle-dependent relaxation remained intact. We conclude that smooth muscle specific CUL3 ablation impairs both cGMP production and cGMP responses and that loss of CUL3 function selectively in smooth muscle is sufficient to cause severe hypertension by interfering with the NO-sGC-cGMP pathway. Our study provides compelling evidence for the sufficiency of vascular smooth muscle CUL3 as a major regulator of BP. CUL3 mutations cause severe vascular dysfunction, arterial stiffness and hypertension due to defects in vascular smooth muscle.


Subject(s)
Cullin Proteins/genetics , Cullin Proteins/metabolism , Genetic Predisposition to Disease/genetics , Hypertension/genetics , Hypertension/metabolism , Muscle, Smooth/metabolism , Animals , Aorta/metabolism , Aorta/pathology , Cyclic GMP/metabolism , Disease Models, Animal , Hypertension/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/metabolism , Mutation , Myocytes, Smooth Muscle/metabolism , Nitric Oxide , Soluble Guanylyl Cyclase/metabolism , Transcriptome , Vascular Stiffness , Vasodilation
3.
J Clin Invest ; 129(6): 2318-2332, 2019 03 21.
Article in English | MEDLINE | ID: mdl-30896450

ABSTRACT

Mice selectively expressing PPARγ dominant negative mutation in vascular smooth muscle exhibit RhoBTB1-deficiency and hypertension. Our rationale was to employ genetic complementation to uncover the mechanism of action of RhoBTB1 in vascular smooth muscle. Inducible smooth muscle-specific restoration of RhoBTB1 fully corrected the hypertension and arterial stiffness by improving vasodilator function. Notably, the cardiovascular protection occurred despite preservation of increased agonist-mediated contraction and RhoA/Rho kinase activity, suggesting RhoBTB1 selectively controls vasodilation. RhoBTB1 augmented the cGMP response to nitric oxide by restraining the activity of phosphodiesterase 5 (PDE5) by acting as a substrate adaptor delivering PDE5 to the Cullin-3 E3 Ring ubiquitin ligase complex for ubiquitination inhibiting PDE5. Angiotensin-II infusion also caused RhoBTB1-deficiency and hypertension which was prevented by smooth muscle specific RhoBTB1 restoration. We conclude that RhoBTB1 protected from hypertension, vascular smooth muscle dysfunction, and arterial stiffness in at least two models of hypertension.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , Hypertension/prevention & control , Muscle, Smooth, Vascular/metabolism , Vascular Stiffness , Vasodilation , rho GTP-Binding Proteins/metabolism , Angiotensin II/adverse effects , Angiotensin II/pharmacology , Animals , Cullin Proteins/genetics , Cullin Proteins/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 5/genetics , Disease Models, Animal , HEK293 Cells , Humans , Hypertension/chemically induced , Hypertension/genetics , Hypertension/metabolism , Mice , Mice, Transgenic , Muscle, Smooth, Vascular/pathology , Nitric Oxide/genetics , Nitric Oxide/metabolism , rho GTP-Binding Proteins/genetics , rho-Associated Kinases/genetics , rho-Associated Kinases/metabolism
4.
JCI Insight ; 2(6): e91738, 2017 03 23.
Article in English | MEDLINE | ID: mdl-28352663

ABSTRACT

Impaired PPARγ activity in endothelial cells causes oxidative stress and endothelial dysfunction which causes a predisposition to hypertension, but the identity of key PPARγ target genes that protect the endothelium remain unclear. Retinol-binding protein 7 (RBP7) is a PPARγ target gene that is essentially endothelium specific. Whereas RBP7-deficient mice exhibit normal endothelial function at baseline, they exhibit severe endothelial dysfunction in response to cardiovascular stressors, including high-fat diet and subpressor angiotensin II. Endothelial dysfunction was not due to differences in weight gain, impaired glucose homeostasis, or hepatosteatosis, but occurred through an oxidative stress-dependent mechanism which can be rescued by scavengers of superoxide. RNA sequencing revealed that RBP7 was required to mediate induction of a subset of PPARγ target genes by rosiglitazone in the endothelium including adiponectin. Adiponectin was selectively induced in the endothelium of control mice by high-fat diet and rosiglitazone, whereas RBP7 deficiency abolished this induction. Adiponectin inhibition caused endothelial dysfunction in control vessels, whereas adiponectin treatment of RBP7-deficient vessels improved endothelium-dependent relaxation and reduced oxidative stress. We conclude that RBP7 is required to mediate the protective effects of PPARγ in the endothelium through adiponectin, and RBP7 is an endothelium-specific PPARγ target and regulator of PPARγ activity.


Subject(s)
Adiponectin/metabolism , Antioxidants/metabolism , Endothelium, Vascular/metabolism , PPAR gamma/metabolism , Retinol-Binding Proteins, Cellular/metabolism , Adiponectin/genetics , Animals , Diet, High-Fat , Endothelium, Vascular/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidative Stress , PPAR gamma/genetics , RNA, Messenger/genetics , Retinol-Binding Proteins, Cellular/genetics
5.
JCI Insight ; 1(19): e91015, 2016 Nov 17.
Article in English | MEDLINE | ID: mdl-27882355

ABSTRACT

Cullin-3 (CUL3) mutations (CUL3Δ9) were previously identified in hypertensive patients with pseudohypoaldosteronism type-II (PHAII), but the mechanism causing hypertension and whether this is driven by renal tubular or extratubular mechanisms remains unknown. We report that selective expression of CUL3Δ9 in smooth muscle acts by interfering with expression and function of endogenous CUL3, resulting in impaired turnover of the CUL3 substrate RhoA, increased RhoA activity, and augmented RhoA/Rho kinase signaling. This caused vascular dysfunction and increased arterial pressure under baseline conditions and a marked increase in arterial pressure, collagen deposition, and vascular stiffness in response to a subpressor dose of angiotensin II, which did not cause hypertension in control mice. Inhibition of total cullin activity increased the level of CUL3 substrates cyclin E and RhoA, and expression of CUL3Δ9 decreased the level of the active form of endogenous CUL3 in human aortic smooth muscle cells. These data indicate that selective expression of the Cul3Δ9 mutation in vascular smooth muscle phenocopies the hypertension observed in Cul3Δ9 human subjects and suggest that mutations in CUL3 cause human hypertension in part through a mechanism involving smooth muscle dysfunction initiated by a loss of CUL3-mediated degradation of RhoA.


Subject(s)
Cullin Proteins/genetics , Hypertension/genetics , Muscle, Smooth, Vascular/physiopathology , Vascular Stiffness , Animals , Cells, Cultured , Humans , Male , Mice , Mice, Inbred C57BL , Mutation , Myocytes, Smooth Muscle/metabolism , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism
6.
Hypertension ; 68(6): 1385-1392, 2016 12.
Article in English | MEDLINE | ID: mdl-27754863

ABSTRACT

The renin-angiotensin system (RAS) in the brain is a critical determinant of blood pressure, but the mechanisms regulating RAS activity in the brain remain unclear. Expression of brain renin (renin-b) occurs from an alternative promoter-first exon. The predicted translation product is a nonsecreted enzymatically active renin whose function is unknown. We generated a unique mouse model by selectively ablating the brain-specific isoform of renin (renin-b) while preserving the expression and function of the classical isoform expressed in the kidney (renin-a). Preservation of renal renin was confirmed by measurements of renin gene expression and immunohistochemistry. Surprisingly, renin-b-deficient mice exhibited hypertension, increased sympathetic nerve activity to the kidney and heart, and impaired baroreflex sensitivity. Whereas these mice displayed decreased circulating RAS activity, there was a paradoxical increase in brain RAS activity. Physiologically, renin-b-deficient mice exhibited an exaggerated depressor response to intracerebroventricular administration of losartan, captopril, or aliskiren. At the molecular level, renin-b-deficient mice exhibited increased expression of angiotensin-II type 1 receptor in the paraventricular nucleus, which correlated with an increased renal sympathetic nerve response to leptin, which was dependent on angiotensin-II type 1 receptor activity. Interestingly, despite an ablation of renin-b expression, expression of renin-a was significantly increased in rostral ventrolateral medulla. These data support a new paradigm for the genetic control of RAS activity in the brain by a coordinated regulation of the renin isoforms, with expression of renin-b tonically inhibiting expression of renin-a under baseline conditions. Impairment of this control mechanism causes neurogenic hypertension.


Subject(s)
Gene Deletion , Hypertension/physiopathology , Paraventricular Hypothalamic Nucleus/metabolism , Renin-Angiotensin System/genetics , Renin/genetics , Analysis of Variance , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Biomarkers/metabolism , Disease Models, Animal , Gene Expression Regulation , Hypertension/drug therapy , Hypertension/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Paraventricular Hypothalamic Nucleus/physiopathology , Protein Isoforms/metabolism , Random Allocation , Renin-Angiotensin System/drug effects , Sensitivity and Specificity
7.
Hypertension ; 67(1): 214-22, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26597823

ABSTRACT

Loss of peroxisome proliferator-activated receptor-γ (PPARγ) function causes hypertension, whereas its activation lowers blood pressure. Evidence suggests that these effects may be attributable to PPARγ activity in the vasculature. However, the specific transcriptional targets of PPARγ in vessels remain largely unidentified. In this study, we examined the role of smooth muscle PPARγ during salt-sensitive hypertension and investigated its transcriptional targets and functional effect. Transgenic mice expressing dominant-negative PPARγ (S-P467L) in smooth muscle cells were more prone to deoxycorticosterone acetate-salt-induced hypertension and mesenteric arterial dysfunction compared with nontransgenic controls. Despite similar morphometry at baseline, vascular remodeling in conduit and small arteries was enhanced in S-P467L after deoxycorticosterone acetate-salt treatment. Gene expression profiling in aorta and mesenteric arteries revealed significantly decreased expression of tissue inhibitor of metalloproteinase-4 (TIMP-4) in S-P467L. Expression of TIMP-4 was increased by deoxycorticosterone acetate-salt treatment, but this increase was ablated in S-P467L. Interference with PPARγ activity either by treatment with a PPARγ inhibitor, GW9662, or by expressing P467L PPARγ markedly suppressed TIMP-4 in primary smooth muscle cells. PPARγ binds to a PPAR response element (PPRE) in chromatin close to the TIMP-4 gene in smooth muscle cells, suggesting that TIMP-4 is a novel target of PPARγ. The interference with PPARγ and decrease in TIMP-4 were accompanied by an increase in total matrix metalloproteinase activity. PPARγ-mediated loss of TIMP-4 increased, whereas overexpression of TIMP-4 decreased smooth muscle cell migration in a scratch assay. Our findings highlight a protective mechanism induced by PPARγ in deoxycorticosterone acetate-salt treatment, establishing a novel mechanistic link between PPARγ and TIMP-4.


Subject(s)
DNA/genetics , Gene Expression Regulation , Hypertension/genetics , Muscle, Smooth, Vascular/metabolism , PPAR gamma/genetics , Tissue Inhibitor of Metalloproteinases/genetics , Animals , Blood Pressure/physiology , Desoxycorticosterone Acetate/toxicity , Disease Models, Animal , Hypertension/metabolism , Hypertension/physiopathology , Mice , Mice, Transgenic , Muscle, Smooth, Vascular/physiopathology , PPAR gamma/metabolism , Tissue Inhibitor of Metalloproteinases/antagonists & inhibitors , Vasoconstriction , Tissue Inhibitor of Metalloproteinase-4
8.
Physiol Genomics ; 48(2): 124-34, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26534936

ABSTRACT

The ligand activated nuclear receptor peroxisome proliferator-activated receptor γ (PPARγ) in the endothelium regulates vascular function and blood pressure (BP). We previously reported that transgenic mice (E-V290M) with selectively targeted endothelial-specific expression of dominant negative PPARγ exhibited endothelial dysfunction when treated with a high-fat diet, and exhibited an augmented pressor response to angiotensin II (ANG II). We hypothesize that interference with endothelial PPARγ would exacerbate ANG II-induced endothelial dysfunction. Endothelial function was examined in E-V290M mice infused with a subpressor dose of ANG II (120 ng·kg(-1)·min(-1)) or saline for 2 wk. ANG II infusion significantly impaired the responses to the endothelium-dependent agonist acetylcholine both in basilar and carotid arteries from E-V290M but not NT mice. This impairment was not due to increased BP, which was not significantly different in ANG II-infused E-V290M compared with NT mice. Superoxide levels, and expression of the pro-oxidant Nox2 gene was elevated, whereas expression of the anti-oxidant genes Catalase and SOD3 decreased in carotid arteries from ANG II-infused E-V290M mice. Increased p65 and decreased Iκ-Bα suggesting increased NF-κB activity was also observed in aorta from ANG II-infused E-V290M mice. The responses to acetylcholine were significantly improved both in basilar and carotid arteries after treatment with Tempol (1 mmol/l), a scavenger of superoxide. These findings provide evidence that interference with endothelial PPARγ accelerates ANG II-mediated endothelial dysfunction both in cerebral and conduit arteries through an oxidative stress-dependent mechanism, suggesting a role for endothelial PPARγ in protecting against ANG II-induced endothelial dysfunction.


Subject(s)
Angiotensin II/metabolism , Endothelium, Vascular/metabolism , PPAR gamma/metabolism , Acetylcholine/metabolism , Animals , Antioxidants/metabolism , Blood Pressure , Carotid Arteries/pathology , Catalase/metabolism , Diet, High-Fat , Endothelium, Vascular/pathology , Heart Rate , Ligands , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , NF-kappa B/metabolism , Oxidative Stress , PPAR gamma/antagonists & inhibitors , PPAR gamma/genetics , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Spectrometry, Fluorescence , Superoxide Dismutase/metabolism , Superoxides/metabolism
9.
Physiol Rep ; 3(1)2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25602015

ABSTRACT

Preeclampsia is a cardiovascular disorder of late pregnancy that is, commonly characterized by hypertension, renal structural damage and dysfunction, and fetal growth restriction. Prevailing etiologic models of this disorder include T-cell dysfunction as an initiating cause of preeclampsia. Indoleamine 2,3-dioxygenase (IDO), an enzyme that mediates the conversion of tryptophan to kynurenine, has been linked to preeclampsia in humans, and is known to regulate T-cell activity and an endothelial-derived relaxing factor. To test the hypothesis that IDO is causally involved in the pathogenesis of preeclampsia, mice deficient for IDO (IDO-KO) were generated on a C57BL/6 background. IDO-KO and wild-type C57BL/6 mice were bred, and preeclampsia phenotypes were evaluated during pregnancy. Pregnant IDO-KO mice exhibited pathognomonic renal glomerular endotheliosis, proteinuria, pregnancy-specific endothelial dysfunction, intrauterine growth restriction, and mildly elevated blood pressure compared to wild-type mice. Together these findings highlight an important role for IDO in the generation of phenotypes typical of preeclampsia. Loss of IDO function may represent a risk factor for the development of preeclampsia. By extension, increased IDO activity, reductions in IDO reactants, or increases in IDO products may represent novel therapeutic approaches for this disorder.

10.
PLoS One ; 9(8): e103786, 2014.
Article in English | MEDLINE | ID: mdl-25122005

ABSTRACT

Peroxisome proliferator activated receptor γ (PPARγ) has been reported to play a protective role in the vasculature; however, the underlying mechanisms involved are not entirely known. We previously showed that vascular smooth muscle-specific overexpression of a dominant negative human PPARγ mutation in mice (S-P467L) leads to enhanced myogenic tone and increased angiotensin-II-dependent vasoconstriction. S-P467L mice also exhibit increased arterial blood pressure. Here we tested the hypotheses that a) mesenteric smooth muscle cells isolated from S-P467L mice exhibit enhanced angiotensin-II AT1 receptor signaling, and b) the increased arterial pressure of S-P467L mice is angiotensin-II AT1 receptor dependent. Phosphorylation of mitogen-activated protein/extracellular signal-regulated kinase (ERK1/2) was robustly increased in mesenteric artery smooth muscle cell cultures from S-P467L in response to angiotensin-II. The increase in ERK1/2 activation by angiotensin-II was blocked by losartan, a blocker of AT1 receptors. Angiotensin-II-induced ERK1/2 activation was also blocked by Tempol, a scavenger of reactive oxygen species, and correlated with increased Nox4 protein expression. To investigate whether endogenous renin-angiotensin system activity contributes to the elevated arterial pressure in S-P467L, non-transgenic and S-P467L mice were treated with the AT1 receptor blocker, losartan (30 mg/kg per day), for 14-days and arterial pressure was assessed by radiotelemetry. At baseline S-P467L mice showed a significant increase of systolic arterial pressure (142.0 ± 10.2 vs 129.1 ± 3.0 mmHg, p<0.05). Treatment with losartan lowered systolic arterial pressure in S-P467L (132.2 ± 6.9 mmHg) to a level similar to untreated non-transgenic mice. Losartan also lowered arterial pressure in non-transgenic (113.0 ± 3.9 mmHg) mice, such that there was no difference in the losartan-induced depressor response between groups (-13.53 ± 1.39 in S-P467L vs -16.16 ± 3.14 mmHg in non-transgenic). Our results suggest that interference with PPARγ in smooth muscle: a) causes enhanced angiotensin-II AT1 receptor-mediated ERK1/2 activation in resistance vessels, b) and may elevate arterial pressure through both angiotensin-II AT1 receptor-dependent and -independent mechanisms.


Subject(s)
Angiotensin II/metabolism , Hypertension/metabolism , Muscle, Smooth, Vascular/metabolism , PPAR gamma/metabolism , Receptor, Angiotensin, Type 1/metabolism , Animals , Arterial Pressure/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Hypertension/drug therapy , Losartan/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Reactive Oxygen Species/metabolism , Renin-Angiotensin System/drug effects , Signal Transduction/drug effects , Vasoconstriction/drug effects
11.
Am J Physiol Regul Integr Comp Physiol ; 307(4): R376-86, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24965793

ABSTRACT

Increased activity of the renin-angiotensin system within the brain elevates fluid intake, blood pressure, and resting metabolic rate. Renin and angiotensinogen are coexpressed within the same cells of the subfornical organ, and the production and action of ANG II through the ANG II type 1 receptor in the subfornical organ (SFO) are necessary for fluid intake due to increased activity of the brain renin-angiotensin system. We generated an inducible model of ANG II production by breeding transgenic mice expressing human renin in neurons controlled by the synapsin promoter with transgenic mice containing a Cre-recombinase-inducible human angiotensinogen construct. Adenoviral delivery of Cre-recombinase causes SFO-selective induction of human angiotensinogen expression. Selective production of ANG II in the SFO results in increased water intake but did not change blood pressure or resting metabolic rate. The increase in water intake was ANG II type 1 receptor-dependent. When given a choice between water and 0.15 M NaCl, these mice increased total fluid and sodium, but not water, because of an increased preference for NaCl. When provided a choice between water and 0.3 M NaCl, the mice exhibited increased fluid, water, and sodium intake, but no change in preference for NaCl. The increase in fluid intake was blocked by an inhibitor of PKC, but not ERK, and was correlated with increased phosphorylated cyclic AMP response element binding protein in the subfornical organ. Thus, increased production and action of ANG II specifically in the subfornical organ are sufficient on their own to mediate an increase in drinking through PKC.


Subject(s)
Angiotensinogen/metabolism , Drinking , Renin-Angiotensin System , Renin/metabolism , Subfornical Organ/enzymology , Angiotensinogen/genetics , Animals , Behavior, Animal , Blood Pressure , CREB-Binding Protein/metabolism , Drinking/drug effects , Drinking Behavior , Energy Metabolism , Female , Humans , Integrases/genetics , Luminescent Proteins/biosynthesis , Luminescent Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phosphorylation , Promoter Regions, Genetic , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Protein Kinase Inhibitors/pharmacology , Renin/genetics , Renin-Angiotensin System/drug effects , Renin-Angiotensin System/genetics , Signal Transduction , Sodium Chloride/administration & dosage , Subfornical Organ/drug effects , Synapsins/genetics , Time Factors
12.
Am J Physiol Regul Integr Comp Physiol ; 304(10): R818-28, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23535460

ABSTRACT

An indispensable role for the brain renin-angiotensin system (RAS) has been documented in most experimental animal models of hypertension. To identify the specific efferent pathway activated by the brain RAS that mediates hypertension, we examined the hypothesis that elevated arginine vasopressin (AVP) release is necessary for hypertension in a double-transgenic model of brain-specific RAS hyperactivity (the "sRA" mouse model). sRA mice experience elevated brain RAS activity due to human angiotensinogen expression plus neuron-specific human renin expression. Total daily loss of the 4-kDa AVP prosegment (copeptin) into urine was grossly elevated (≥8-fold). Immunohistochemical staining for AVP was increased in the supraoptic nucleus of sRA mice (~2-fold), but no quantitative difference in the paraventricular nucleus was observed. Chronic subcutaneous infusion of a nonselective AVP receptor antagonist conivaptan (YM-087, Vaprisol, 22 ng/h) or the V(2)-selective antagonist tolvaptan (OPC-41061, 22 ng/h) resulted in normalization of the baseline (~15 mmHg) hypertension in sRA mice. Abdominal aortas and second-order mesenteric arteries displayed AVP-specific desensitization, with minor or no changes in responses to phenylephrine and endothelin-1. Mesenteric arteries exhibited substantial reductions in V(1A) receptor mRNA, but no significant changes in V(2) receptor expression in kidney were observed. Chronic tolvaptan infusion also normalized the (5 mmol/l) hyponatremia of sRA mice. Together, these data support a major role for vasopressin in the hypertension of mice with brain-specific hyperactivity of the RAS and suggest a primary role of V(2) receptors.


Subject(s)
Blood Pressure/physiology , Brain/metabolism , Hypertension/metabolism , Renin-Angiotensin System/physiology , Vasopressins/metabolism , Animals , Antidiuretic Hormone Receptor Antagonists , Benzazepines/pharmacology , Blood Pressure/drug effects , Brain/drug effects , Gene Expression/drug effects , Hypertension/genetics , Hypothalamus/drug effects , Hypothalamus/metabolism , Mice , Mice, Transgenic , Receptors, Vasopressin/genetics , Receptors, Vasopressin/metabolism , Renin-Angiotensin System/drug effects , Tolvaptan , Vasopressins/genetics
13.
Hypertension ; 61(3): 716-22, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23266541

ABSTRACT

Although elevated renin-angiotensin system activity and angiotensinergic signaling within the brain are required for hypertension, polydipsia, and increased metabolic rate induced by deoxycorticosterone acetate (DOCA)-salt, the contribution of specific receptor subtypes and brain nuclei mediating these responses remains poorly defined. We hypothesized that angiotensin type 1a receptors (AT(1a)R) within the subfornical organ (SFO) mediate these responses. Transgenic mice carrying a conditional allele of the endogenous AT(1a)R (AT(1a)R(flox)) were administered an adenovirus encoding Cre-recombinase and enhanced green fluorescent protein (eGFP) or adenovirus encoding eGFP alone into the lateral cerebral ventricle. Adenovirus encoding Cre-recombinase reduced AT(1a)R mRNA and induced recombination in AT(1a)R(flox) genomic DNA specifically in the SFO, without significant effect in the paraventricular or arcuate nuclei, and also induced SFO-specific recombination in ROSA(TdTomato) reporter mice. The effect of SFO-targeted ablation of endogenous AT(1a)R was evaluated in AT(1a)R(flox) mice at 3 time points: (1) baseline, (2) 1 week after virus injection but before DOCA-salt, and (3) after 3 weeks of DOCA-salt. DOCA-salt-treated mice with deletion of AT(1a)R in SFO exhibited a blunted increase in arterial pressure. Increased sympathetic cardiac modulation and urine copeptin, a marker of vasopressin release, were both significantly reduced in DOCA-salt mice when AT(1a)R was deleted in the SFO. Additionally, deletion of AT(1a)R in the SFO significantly attenuated the polydipsia, polyuria, and sodium intake in response to DOCA-salt. Together, these data highlight the contribution of AT(1a)R in the SFO to arterial pressure regulation potentially through changes on sympathetic cardiac modulation, vasopressin release, and hydromineral balance in the DOCA-salt model of hypertension.


Subject(s)
Desoxycorticosterone/adverse effects , Hypertension/chemically induced , Mineralocorticoids/adverse effects , Receptor, Angiotensin, Type 1/physiology , Subfornical Organ/drug effects , Subfornical Organ/physiopathology , Animals , Arterial Pressure/drug effects , Biomarkers/urine , Glycopeptides/urine , Heart/drug effects , Heart/innervation , Male , Mice , Mice, Transgenic , Polydipsia/chemically induced , Polyuria/chemically induced , Receptor, Angiotensin, Type 1/genetics , Recombination, Genetic , Sodium/metabolism , Sympathetic Nervous System/drug effects
14.
Hypertension ; 59(6): 1212-9, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22547438

ABSTRACT

A single-nucleotide polymorphism (C/A) located within an E-box at the -20 position of the human angiotensinogen (AGT) promoter may regulate transcriptional activation through differential recruitment of the transcription factors upstream stimulatory factor (USF) 1 and 2. To study the contribution of USF1 on AGT gene expression, mice carrying a (-20C) human AGT (hAGT) transgene were bred with mice harboring a USF1 gene trap allele designed to knock down USF1 expression. USF1 mRNA was reduced relative to controls in liver (9 ± 1%), perigenital adipose (16 ± 3%), kidney (17 ± 1%), and brain (34 ± 2%) in double-transgenic mice. This decrease was confirmed by electrophoretic mobility shift assay. Chromatin immunoprecipitation analyses revealed a decrease in USF1, with retention of USF2 binding at the hAGT promoter in the liver of male mice. hAGT expression was reduced in the liver and other tissues of female but not male mice. The decrease in endogenous AGT expression was insufficient to alter systolic blood pressure at baseline but caused reduced systolic blood pressure in female USF1 gene trap mice fed a high-fat diet. Treatment of USF1 knockdown males with intravenous adenoviral short hairpin RNA targeting USF2 resulted in reduced expression of USF1, USF2, and hAGT protein. Our data from chromatin immunoprecipitation assays suggests that this decrease in hAGT is attributed to decreased USF2 binding to the hAGT promoter. In conclusion, both USF1 and USF2 are essential for AGT transcriptional regulation, and distinct sex-specific and tissue-specific mechanisms are involved in the activities of these transcription factors in vivo.


Subject(s)
Angiotensinogen/metabolism , Gene Knockdown Techniques/methods , Upstream Stimulatory Factors/metabolism , Angiotensinogen/genetics , Animals , Blood Pressure/drug effects , Blood Pressure/genetics , Blood Pressure/physiology , Blotting, Western , Chromatin Immunoprecipitation , Diet, High-Fat , Dietary Fats/administration & dosage , Electrophoretic Mobility Shift Assay , Female , Gene Expression Regulation , Humans , Hypertension/etiology , Hypertension/genetics , Hypertension/physiopathology , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Polymorphism, Single Nucleotide , Promoter Regions, Genetic/genetics , Protein Binding , RNA Interference , Sex Factors , Upstream Stimulatory Factors/genetics
15.
Am J Physiol Regul Integr Comp Physiol ; 301(4): R1067-77, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21753145

ABSTRACT

All components of the renin angiotensin system necessary for ANG II generation and action have been reported to be present in renal proximal convoluted tubules. Given the close relationship between renal sodium handling and blood pressure regulation, we hypothesized that modulating the action of ANG II specifically in the renal proximal tubules would alter the chronic level of blood pressure. To test this, we used a proximal tubule-specific, androgen-dependent, promoter construct (KAP2) to generate mice with either overexpression of a constitutively active angiotensin type 1A receptor transgene or depletion of endogenous angiotensin type 1A receptors. Androgen administration to female transgenic mice caused a robust induction of the transgene in the kidney and increased baseline blood pressure. In the receptor-depleted mice, androgen administration to females resulted in a Cre recombinase-mediated deletion of angiotensin type 1A receptors in the proximal tubule and reduced blood pressure. In contrast to the changes observed at baseline, there was no difference in the blood pressure response to a pressor dose of ANG II in either experimental model. These data, from two separate mouse models, provide evidence that ANG II signaling via the type 1A receptor in the renal proximal tubule is a regulator of systemic blood pressure under baseline conditions.


Subject(s)
Blood Pressure/physiology , Kidney Tubules, Proximal/metabolism , Receptor, Angiotensin, Type 1/metabolism , Androgens/pharmacology , Angiotensin II/pharmacology , Animals , Blood Pressure/drug effects , Female , Integrases/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Models, Animal , Receptor, Angiotensin, Type 1/drug effects , Receptor, Angiotensin, Type 1/genetics , Signal Transduction/drug effects , Signal Transduction/physiology
16.
Hypertension ; 57(3): 600-7, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21263123

ABSTRACT

Low-renin hypertension accounts for ≈ 25% of essential hypertensive patients. It is modeled in animals by chronic delivery of deoxycorticosterone acetate and excess dietary sodium (the DOCA-salt model). Previous studies have demonstrated that DOCA-salt hypertension is mediated through activation of the brain renin-angiotensin system. Here, we demonstrate robust metabolic phenotypes of DOCA-salt treatment. Male C57BL/6J mice (6 to 8 weeks old) received a subcutaneous pellet of DOCA (50 mg for 21 days) and were offered a 0.15 mol/L NaCl drink solution in addition to regular chow and tap water. Treatment resulted in mild hypertension, a blunting of weight gain, gross polydipsia, polyuria, and sodium intake, alterations in urinary sodium and potassium turnover, and serum sodium retention. Most strikingly, DOCA-salt mice exhibited no difference in food intake but did exhibited a large elevation in basal metabolic rate. Normalization of blood pressure by hydralazine (500 mg/L in drink solutions) attenuated the hydromineral phenotypes and renal renin suppression effects of DOCA-salt but had no effect on the elevated metabolic rate. In contrast, intracerebroventricular infusion of the angiotensin II type 1 receptor antagonist losartan (5 µg/h) attenuated the elevation in metabolic rate with DOCA-salt treatment. Together, these data illustrate the necessity of angiotensinergic signaling within the brain, independent of blood pressure alterations, in the metabolic consequences of DOCA-salt treatment.


Subject(s)
Angiotensins/metabolism , Basal Metabolism/drug effects , Brain/metabolism , Desoxycorticosterone/pharmacology , Hypertension/metabolism , Renin-Angiotensin System/physiology , Analysis of Variance , Animals , Basal Metabolism/physiology , Blood Pressure/drug effects , Blood Pressure/physiology , Brain/drug effects , Calorimetry , Desoxycorticosterone/metabolism , Eating/drug effects , Eating/physiology , Hypertension/chemically induced , Male , Mice , Mice, Inbred C57BL , Renin/metabolism , Renin-Angiotensin System/drug effects , Signal Transduction , Sodium, Dietary/administration & dosage , Sodium, Dietary/metabolism , Statistics, Nonparametric
17.
Physiol Genomics ; 43(6): 286-94, 2011 Mar 29.
Article in English | MEDLINE | ID: mdl-21189370

ABSTRACT

The renin-angiotensin system (RAS), known for its roles in cardiovascular, metabolic, and developmental regulation, is present in both the circulation and in many individual tissues throughout the body. Substantial evidence supports the existence of a brain RAS, though quantification and localization of brain renin have been hampered by its low expression levels. We and others have previously determined that there are two isoforms of renin expressed in the brain. The classical isoform encoding secreted renin (sREN) and a novel isoform encoding intracellular renin (icREN), the product of an alternative promoter and first exon (exon 1b). The differential role that these two isoforms play in cardiovascular and metabolic regulation remains unclear. Here we examined the physiological consequences of neuron- and glia-specific knockouts of sREN by crossing mice in which the sREN promoter and isoform-specific first exon (exon-1a) is flanked by LoxP sequences (sREN(flox) mice) with mice expressing Cre-recombinase controlled by either the neuron-specific Nestin promoter or the glia-specific GFAP promoter. Resulting offspring exhibited selective knockout of sREN in either neurons or glia, while preserving expression of icREN. Consistent with a hypothesized role of icREN in the brain RAS, neuron- and glia-specific knockout of sREN had no effect on blood pressure or heart rate; food, water, or sodium intake; renal function; or metabolic rate. These data demonstrate that sREN is dispensable within the brain for normal physiological regulation of cardiovascular, hydromineral, and metabolic regulation, and thereby indirectly support the importance of icREN in brain RAS function.


Subject(s)
Blood Pressure/physiology , Brain/metabolism , Kidney/metabolism , Neuroglia/metabolism , Neurons/metabolism , Renin-Angiotensin System/physiology , Renin/physiology , Animals , Basal Metabolism , Brain/cytology , Exons , Mice , Mice, Knockout , Mice, Transgenic , Renin/genetics
18.
Hypertension ; 56(5): 981-7, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20823378

ABSTRACT

Genetic and functional data support a role for angiotensinogen in blood pressure control, and many population studies have suggested that polymorphisms in the angiotensinogen gene contribute to hypertension. Two common haplotypes of the human angiotensinogen gene are -6A/235T and -6G/235M. To study their contributions to blood pressure regulation in a controlled model system, we developed triple-transgenic mice expressing either -6A/235T or -6G/235M human angiotensinogen, expressing either an overexpressed and poorly regulated (REN9) or a tightly regulated (PAC160) human renin, and all carrying a null mutation in the endogenous murine angiotensinogen gene. These humanized mice were then examined for blood pressure differences at baseline and after a high-salt diet, changes in cardiovascular organ weight, and differences in angiotensinogen and renin gene expression. Mice expressing the -6G/235M haplotype on the PAC160 background exhibited increased blood pressure and cardiac hypertrophy at baseline. In contrast, all of the mice with the REN9 background had equivalent baseline blood pressures. On the REN9 background, there was a greater increase in blood pressure in -6A/235T in response to a high-salt diet, providing evidence it may be a susceptibility allele. There were no differences in angiotensinogen expression between haplotypes on either background strain. The data suggest that the impact of angiotensinogen haplotypes on cardiovascular end points may be dependent on renin status and environmental influences, such as dietary sodium. These insights may help explain the discrepancies among observational studies that have examined roles for the -6A/235T and -6G/235M angiotensinogen haplotypes in varied human populations.


Subject(s)
Angiotensinogen/genetics , Blood Pressure/genetics , Alleles , Angiotensinogen/metabolism , Animals , Genetic Variation , Genotype , Haplotypes , Hypertension/genetics , Hypertension/metabolism , Mice , Mice, Transgenic , Renin/genetics , Renin/metabolism , Sodium, Dietary , Telemetry
19.
Circ Res ; 103(9): 940-7, 2008 Oct 24.
Article in English | MEDLINE | ID: mdl-18802024

ABSTRACT

Among naturally occurring polymorphisms in the 5' flanking region of the human angiotensinogen (AGT) gene, the -20 and -217 polymorphisms have the strongest effects on AGT regulation in AGT-expressing cells derived from liver, kidney, brain, and fat. These polymorphisms may affect allele-specific transcription factor binding, and the high-expressing alleles are both relatively common. We show herein that the -20C allele has higher transcriptional activity than -20A, and the -20A allele confers no additional transactivation potential beyond that of a mutated vector. Gel-shift assays show that upstream stimulatory factor (USF)1 and USF2 preferentially bind the -20C allele, whereas the -20A allele retains a low affinity USF binding site. Plasmid immunoprecipitation assays confirmed preferential association of USF1 with the -20C allele in transfected HepG2 cells. Chromatin immunoprecipitation confirmed that USF1 binds to the endogenous AGT -20C allele in CCF cells, the only cell line tested that carries the -20C allele, and to the human AGT promoter in liver and adipose tissue from transgenic mice. Transduction of AGT-expressing cells with short hairpin RNAs specifically targeting USF1 or USF2, resulted in cell- and allele-specific attenuation of AGT promoter activity. In vivo, knockdown of USF expression in the liver of transgenic mice expressing the -20C allele of AGT resulted in lower AGT expression, a decrease in circulating human AGT protein but no change in expression of GAPDH or hepatocyte nuclear factor-4alpha. We conclude that USF1 functionally and differentially regulates AGT expression via the -20 polymorphism and that the differential expression exhibited by -20 can be accounted for by differential association with USF1.


Subject(s)
5' Flanking Region , Angiotensinogen/metabolism , Polymorphism, Genetic , Promoter Regions, Genetic , Transcription, Genetic , Transcriptional Activation , Upstream Stimulatory Factors/metabolism , 3T3-L1 Cells , Adipose Tissue/metabolism , Angiotensinogen/genetics , Animals , Cell Line, Tumor , Chromatin Immunoprecipitation , Electrophoretic Mobility Shift Assay , Humans , Liver/metabolism , Mice , Mice, Transgenic , RNA Interference , RNA, Small Interfering/metabolism , Transfection , Upstream Stimulatory Factors/genetics
20.
Am J Physiol Renal Physiol ; 294(6): F1481-6, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18385272

ABSTRACT

To facilitate the study of renal proximal tubules, we generated a transgenic mouse strain expressing an improved Cre recombinase (iCre) under the control of the kidney androgen-regulated protein (KAP) promoter. The transgene was expressed in the kidney of male mice but not in female mice. Treatment of female transgenic mice with androgen induced robust expression of the transgene in the kidney. We confirmed the presence of Cre recombinase activity and the cell specificity by breeding the KAP2-iCRE mice with ROSA26 reporter mice. X-Gal staining of kidney sections from male double transgenic mice showed robust staining in the epithelial cells of renal proximal tubules. beta-Gal staining in female mice became evident in proximal tubules after administration of androgen. This model of inducible Cre recombinase in the renal proximal tubule should provide a novel useful tool for studying the physiological significance of genes expressed in the renal proximal tubule. This has advantages over other current models where Cre recombinase expression is constitutive, not inducible.


Subject(s)
Integrases/genetics , Kidney Tubules, Proximal/physiology , Mice, Transgenic/physiology , Proteins/genetics , Transgenes/genetics , Animals , Breeding , Female , Gene Expression Regulation/genetics , Genes, Reporter , Male , Mice , Sex Characteristics
SELECTION OF CITATIONS
SEARCH DETAIL
...