Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters










Publication year range
1.
J Cell Mol Med ; 25(20): 9837-9850, 2021 10.
Article in English | MEDLINE | ID: mdl-34528389

ABSTRACT

The inflammation of adipose tissue is one of the most common secondary pathological changes in atherosclerosis, which in turn influences the process of atherosclerosis. Natriuretic peptides have been revealed important effect in regulating adipose metabolism. However, the relationship between natriuretic peptide receptor C and inflammation of adipose tissue in atherosclerosis remains unknown. This study aims to explore the effect natriuretic peptide receptor C exerts on the regulation of the adipose inflammation in atherosclerotic mice induced by western-type diet and its overlying mechanisms. To clarify the importance of NPRC of adipose inflammation in atherosclerotic mice, NPRC expression was measured in mice fed with chow diet and western-type diet for 12 weeks and we found a considerable increase in adipose tissue of atherosclerotic mice. Global NPRC knockout in mice was bred onto ApoE-/- mice to generate NPRC-/- ApoE-/- mice, which displayed remarked increase in browning of white adipose tissue and lipolysis of adipose tissue and decrease in adipose inflammation manifested by decreased macrophage invasion to form less CLS (crown-like structure), reduced oxidative stress and alleviated expression of TNFα, IL-6, IL-1ß and MCP1, but increased expression of adiponectin in adipose tissue. Moreover, our study showed that white adipose tissue browning in NPRC-/- ApoE-/- atherosclerotic mice was associated with decreased inflammatory response through cAMP/PKA signalling activation. These results identify NPRC as a novel regulator for adipose inflammation in atherosclerotic mice by modulating white adipose tissue browning.


Subject(s)
Apolipoproteins E/deficiency , Hypercholesterolemia/complications , Panniculitis/etiology , Panniculitis/metabolism , Receptors, Atrial Natriuretic Factor/deficiency , Animals , Biomarkers , Cyclic AMP , Cyclic AMP-Dependent Protein Kinases/metabolism , Disease Models, Animal , Disease Susceptibility , Gene Expression Regulation , Hypercholesterolemia/genetics , Hypercholesterolemia/metabolism , Immunohistochemistry , Inflammasomes/metabolism , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Oxidative Stress , Panniculitis/pathology , Signal Transduction
2.
Mol Cell Proteomics ; 20: 100072, 2021.
Article in English | MEDLINE | ID: mdl-33812089

ABSTRACT

Atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP) are important biological markers and cardiac function regulators. Natriuretic peptide receptor A (NPRA) binds to an ANP or BNP ligand and induces transmembrane signal transduction by elevating the intracellular cyclic guanosine monophosphate (cGMP) levels. However, the metabolic phenotype and related mechanisms induced by NPRA deletion remain ambiguous. Here, we constructed myocardial-specific NPRA deletion mice and detected the heart functional and morphological characteristics by histological analysis and explored the altered metabolic pattern and the expression patterns of proteins by liquid chromatography-mass spectrometry (LC-MS)-based omics technology. NPRA deficiency unexpectedly did not result in significant cardiac remodeling or dysfunction. However, compared with the matched littermates, NPRA-deficient mice had significant metabolic differences. Metabolomic analysis showed that the metabolite levels varied in cardiac tissues and plasma. In total, 33 metabolites were identified in cardiac tissues and 54 were identified in plasma. Compared with control mice, NPRA-deficient mice had 20 upregulated and six downregulated metabolites in cardiac tissues and 25 upregulated and 23 downregulated metabolites in plasma. Together, NPRA deficiency resulted in increased nucleotide biosynthesis and histidine metabolism only in heart tissues and decreased creatine metabolism only in plasma. Further proteomic analysis identified 136 differentially abundant proteins in cardiac tissues, including 54 proteins with higher abundance and 82 proteins with lower abundance. Among them, cytochrome c oxidase subunit 7c and 7b (Cox7c, Cox7b), ATP synthase, H+ transporting, mitochondrial Fo complex subunit F2 (ATP5J2), ubiquinol-cytochrome c reductase, complex III subunit X (Uqcr10), and myosin heavy chain 7 (Myh7) were mainly involved in related metabolic pathways. These results revealed the essential role of NPRA in metabolic profiles and may elucidate new underlying pathophysiological mechanisms of NPRA in cardiovascular diseases.


Subject(s)
Myocardium/metabolism , Receptors, Atrial Natriuretic Factor/deficiency , Animals , Metabolomics , Mice, Knockout , Phenotype , Protein Interaction Maps , Proteomics , RNA, Messenger/metabolism , Receptors, Atrial Natriuretic Factor/genetics , Receptors, Atrial Natriuretic Factor/metabolism
3.
Hypertension ; 77(3): 868-881, 2021 03 03.
Article in English | MEDLINE | ID: mdl-33486984

ABSTRACT

Genome-wide association studies have identified that NPR-C (natriuretic peptide receptor-C) variants are associated with elevation of blood pressure. However, the mechanism underlying the relationship between NPR-C and blood pressure regulation remains elusive. Here, we investigate whether NPR-C regulates Ang II (angiotensin II)-induced hypertension through sodium transporters activity. Wild-type mice responded to continuous Ang II infusion with an increased renal NPR-C expression. Global NPR-C deficiency attenuated Ang II-induced increased blood pressure both in male and female mice associated with more diuretic and natriuretic responses to a saline challenge. Interestingly, Ang II increased both total and phosphorylation of NCC (NaCl cotransporter) abundance involving in activation of WNK4 (with-no-lysine kinase 4)/SPAK (Ste20-related proline/alanine-rich kinase) which was blunted by NPR-C deletion. NCC inhibitor, hydrochlorothiazide, failed to induce natriuresis in NPR-C knockout mice. Moreover, low-salt and high-salt diets-induced changes of total and phosphorylation of NCC expression were normalized by NPR-C deletion. Importantly, tubule-specific deletion of NPR-C also attenuated Ang II-induced elevated blood pressure, total and phosphorylation of NCC expression. Mechanistically, in distal convoluted tubule cells, Ang II dose and time-dependently upregulated WNK4/SPAK/NCC kinase pathway and NPR-C/Gi/PLC/PKC signaling pathway mediated NCC activation. These results demonstrate that NPR-C signaling regulates NCC function contributing to sodium retention-mediated elevated blood pressure, which suggests that NPR-C is a promising candidate for the treatment of sodium retention-related hypertension.


Subject(s)
Blood Pressure/physiology , Hypertension/physiopathology , Kidney/metabolism , Receptors, Atrial Natriuretic Factor/deficiency , Solute Carrier Family 12, Member 3/metabolism , Angiotensin II , Animals , Blood Pressure/genetics , Cells, Cultured , Female , Hypertension/chemically induced , Hypertension/genetics , Kidney Tubules, Distal/cytology , Kidney Tubules, Distal/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptors, Atrial Natriuretic Factor/genetics , Renin-Angiotensin System/genetics , Renin-Angiotensin System/physiology , Signal Transduction/genetics , Sodium/blood , Sodium/urine , Solute Carrier Family 12, Member 3/genetics
4.
Circulation ; 141(7): 571-588, 2020 02 18.
Article in English | MEDLINE | ID: mdl-31665900

ABSTRACT

BACKGROUND: The maternal circulatory system and hormone balance both change dynamically during pregnancy, delivery, and the postpartum period. Although atrial natriuretic peptides and brain natriuretic peptides produced in the heart control circulatory homeostasis through their common receptor, NPR1, the physiologic and pathophysiologic roles of endogenous atrial natriuretic peptide/brain natriuretic peptide in the perinatal period are not fully understood. METHODS: To clarify the physiologic and pathophysiologic roles of the endogenous atrial natriuretic peptide/brain natriuretic peptide-NPR1 system during the perinatal period, the phenotype of female wild-type and conventional or tissue-specific Npr1-knockout mice during the perinatal period was examined, especially focusing on maternal heart weight, blood pressure, and cardiac function. RESULTS: In wild-type mice, lactation but not pregnancy induced reversible cardiac hypertrophy accompanied by increases in fetal cardiac gene mRNAs and ERK1/2 (extracellular signaling-regulated kinase) phosphorylation. Npr1-knockout mice exhibited significantly higher plasma aldosterone level than did wild-type mice, severe cardiac hypertrophy accompanied by fibrosis, and left ventricular dysfunction in the lactation period. Npr1-knockout mice showed a high mortality rate over consecutive pregnancy-lactation cycles. In the hearts of Npr1-knockout mice during or after the lactation period, an increase in interleukin-6 mRNA expression, phosphorylation of signal transducer and activator of transcription 3, and activation of the calcineurin-nuclear factor of the activated T cells pathway were observed. Pharmacologic inhibition of the mineralocorticoid receptor or neuron-specific deletion of the mineralocorticoid receptor gene significantly ameliorated cardiac hypertrophy in lactating Npr1-knockout mice. Anti-interleukin-6 receptor antibody administration tended to reduce cardiac hypertrophy in lactating Npr1-knockout mice. CONCLUSIONS: These results suggest that the characteristics of lactation-induced cardiac hypertrophy in wild-type mice are different from exercise-induced cardiac hypertrophy, and that the endogenous atrial natriuretic peptide/brain natriuretic peptide-NPR1 system plays an important role in protecting the maternal heart from interleukin-6-induced inflammation and remodeling in the lactation period, a condition mimicking peripartum cardiomyopathy.


Subject(s)
Atrial Natriuretic Factor/deficiency , Cardiomegaly/metabolism , Lactation , MAP Kinase Signaling System , Peripartum Period , Receptors, Atrial Natriuretic Factor/deficiency , Animals , Cardiomegaly/genetics , Cardiomegaly/pathology , Female , Mice , Mice, Knockout
5.
Circ Arrhythm Electrophysiol ; 12(1): e006863, 2019 01.
Article in English | MEDLINE | ID: mdl-30636477

ABSTRACT

BACKGROUND: Atrial fibrillation (AF) commonly occurs in hypertension and in association with elevated Ang II (angiotensin II) levels. The specific mechanisms underlying Ang II-mediated AF are unclear, and interventions to prevent the effects of Ang II are lacking. NPs (natriuretic peptides), which elicit their effects through specific NP receptors, including NPR-C (natriuretic peptide receptor-C), are cardioprotective hormones that affect cardiac structure and function. METHODS: This study used wild-type and NPR-C knockout (NPR-C-/-) mice to investigate the effects of Ang II (3 mg/kg per day for 3 weeks) on AF susceptibility and atrial function using in vivo electrophysiology, high-resolution optical mapping, patch clamping, and molecular biology. In some experiments, wild-type mice were cotreated with Ang II and the NPR-C agonist cANF (0.07-0.14 mg/kg per day) for 3 weeks. RESULTS: In wild-type mice, Ang II increased susceptibility to AF in association with a prolongation of P-wave duration, increased atrial refractory period, and slowed atrial conduction. These effects were exacerbated in Ang II-treated NPR-C-/- mice. Ang II prolonged action potential duration and reduced action potential upstroke velocity (Vmax). These effects were greater in left atrial myocytes from Ang II-treated NPR-C-/- mice. Ang II also increased fibrosis in both atria in wild-type mice, whereas Ang II-treated NPR-C-/- mice exhibited substantially higher fibrosis throughout the atria. Fibrotic responses were associated with changes in expression of profibrotic genes, including TGFß and TIMP1. Cotreating wild-type mice with Ang II and the NPR-C agonist cANF dose dependently reduced AF inducibility by preventing some of the Ang II-induced changes in atrial myocyte electrophysiology and preventing fibrosis throughout the atria. CONCLUSIONS: NPR-C may represent a new target for the prevention of Ang II-induced AF via protective effects on atrial electrical and structural remodeling.


Subject(s)
Angiotensin II , Atrial Fibrillation/metabolism , Atrial Remodeling , Heart Atria/metabolism , Myocytes, Cardiac/metabolism , Receptors, Atrial Natriuretic Factor/metabolism , Action Potentials , Animals , Atrial Fibrillation/chemically induced , Atrial Fibrillation/genetics , Atrial Fibrillation/physiopathology , Disease Models, Animal , Disease Progression , Fibrosis , Heart Atria/physiopathology , Heart Rate , Hypertension/chemically induced , Hypertension/metabolism , Hypertension/physiopathology , Male , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/pathology , Receptors, Atrial Natriuretic Factor/deficiency , Receptors, Atrial Natriuretic Factor/genetics , Time Factors
6.
Cardiovasc Res ; 115(2): 357-372, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30239604

ABSTRACT

Aims: TGF-ß1 plays an important role in atrial fibrosis and atrial fibrillation (AF); previous studies have shown that the atria are more susceptible to TGF-ß1 mediated fibrosis than the ventricles. Natriuretic peptides (NPs) play an important role in cardiac remodelling and fibrosis, but the role of natriuretic peptide clearance (NPR-C) receptor is largely unknown. We investigated the role of NPR-C in modulating TGF-ß1 signalling in the atria. Methods and results: MHC-TGF-ß1 transgenic (TGF-ß1-Tx) mice, which develop isolated atrial fibrosis and AF, were cross-bred with NPR-C knock-out mice (NPR-C-KO). Transverse aortic constriction (TAC) was performed in wild type (Wt) and NPR-C knockout mice to study. Atrial fibrosis and AF inducibility in a pathophysiologic model. Electrophysiology, molecular, and histologic studies were performed in adult mice. siRNA was used to interrogate the interaction between TGF-ß1 and NP signalling pathways in isolated atrial and ventricular fibroblasts/myofibroblasts. NPR-C expression level was 17 ± 5.8-fold higher in the atria compared with the ventricle in Wt mice (P = 0.009). Cross-bred mice demonstrated markedly decreased pSmad2 and collagen expression, atrial fibrosis, and AF compared with TGF-ß1-Tx mice with intact NPR-C. There was a marked reduction in atrial fibrosis gene expression and AF inducibility in the NPR-C-KO-TAC mice compared with Wt-TAC. In isolated fibroblasts, knockdown of NPR-C resulted in a marked reduction of pSmad2 (56 ± 4% and 24 ± 14% reduction in atrial and ventricular fibroblasts, respectively) and collagen (76 ± 15% and 35 ± 23% reduction in atrial and ventricular fibroblasts/myofibroblasts, respectively) in response to TGF-ß1 stimulation. This effect was reversed by simultaneously knocking down NPR-A but not with simultaneous knock down of PKG-1. Conclusion: The differential response to TGF-ß1 stimulated fibrosis between the atria and ventricle are in part mediated by the abundance of NPR-C receptors in the atria.


Subject(s)
Atrial Fibrillation/prevention & control , Atrial Remodeling , Cardiomyopathies/prevention & control , Heart Atria/metabolism , Heart Rate , Receptors, Atrial Natriuretic Factor/deficiency , Transforming Growth Factor beta1/metabolism , Action Potentials , Animals , Atrial Fibrillation/genetics , Atrial Fibrillation/metabolism , Atrial Fibrillation/physiopathology , Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Collagen/metabolism , Disease Models, Animal , Fibrosis , Heart Atria/pathology , Heart Atria/physiopathology , Heart Ventricles/metabolism , Heart Ventricles/pathology , Heart Ventricles/physiopathology , Mice, Knockout , Myofibroblasts/metabolism , Myofibroblasts/pathology , Phosphorylation , Receptors, Atrial Natriuretic Factor/genetics , Signal Transduction , Smad2 Protein/metabolism , Transforming Growth Factor beta1/genetics
7.
Front Neural Circuits ; 12: 65, 2018.
Article in English | MEDLINE | ID: mdl-30275816

ABSTRACT

Sensory axon T-like branching (bifurcation) in neurons from dorsal root ganglia and cranial sensory ganglia depends on the molecular signaling cascade involving the secreted factor C-type natriuretic peptide, the natriuretic peptide receptor guanylyl cyclase B (GC-B; also known as Npr2) and cGMP-dependent protein kinase I (cGKI, also known as PKGI). The bifurcation of cranial nerves is suggested to be important for information processing by second-order neurons in the hindbrain or spinal cord. Indeed, mice with a spontaneous GC-B loss of function mutation (Npr2cn/cn ) display an impaired bifurcation of auditory nerve (AN) fibers. However, these mice did not show any obvious sign of impaired basal hearing. Here, we demonstrate that mice with a targeted inactivation of the GC-B gene (Npr2 lacZ/lacZ , GC-B KO mice) show an elevation of audiometric thresholds. In the inner ear, the cochlear hair cells in GC-B KO mice were nevertheless similar to those from wild type mice, justified by the typical expression of functionally relevant marker proteins. However, efferent cholinergic feedback to inner and outer hair cells was reduced in GC-B KO mice, linked to very likely reduced rapid efferent feedback. Sound-evoked AN responses of GC-B KO mice were elevated, a feature that is known to occur when the efferent axo-dendritic feedback on AN is compromised. Furthermore, late sound-evoked brainstem responses were significantly delayed in GC-B KO mice. This delay in sound response was accompanied by a weaker sensitivity of the auditory steady state response to amplitude-modulated sound stimuli. Finally, the acoustic startle response (ASR) - one of the fastest auditory responses - and the prepulse inhibition of the ASR indicated significant changes in temporal precision of auditory processing. These findings suggest that GC-B-controlled axon bifurcation of spiral ganglion neurons is important for proper activation of second-order neurons in the hindbrain and is a prerequisite for proper temporal auditory processing likely by establishing accurate efferent top-down control circuits. These data hypothesize that the bifurcation pattern of cranial nerves is important to shape spatial and temporal information processing for sensory feedback control.


Subject(s)
Auditory Perception/physiology , Auditory Threshold/physiology , Axons/physiology , Cochlear Nerve/physiology , Cranial Nerves/physiology , Evoked Potentials, Auditory/physiology , Prepulse Inhibition/physiology , Receptors, Atrial Natriuretic Factor/physiology , Reflex, Startle/physiology , Spiral Ganglion/physiology , Animals , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Atrial Natriuretic Factor/deficiency
8.
Cardiovasc Diabetol ; 17(1): 103, 2018 07 17.
Article in English | MEDLINE | ID: mdl-30016962

ABSTRACT

BACKGROUND: The cardiac hormones atrial (ANP) and B-type natriuretic peptides (BNP) moderate arterial blood pressure and improve energy metabolism as well as insulin sensitivity via their shared cGMP-producing guanylyl cyclase-A (GC-A) receptor. Obesity is associated with impaired NP/GC-A/cGMP signaling, which possibly contributes to the development of type 2 diabetes and its cardiometabolic complications. In vitro, synthetic ANP, via GC-A, stimulates glucose-dependent insulin release from cultured pancreatic islets and ß-cell proliferation. However, the relevance for systemic glucose homeostasis in vivo is not known. To dissect whether the endogenous cardiac hormones modulate the secretory function and/or proliferation of ß-cells under (patho)physiological conditions in vivo, here we generated a novel genetic mouse model with selective disruption of the GC-A receptor in ß-cells. METHODS: Mice with a floxed GC-A gene were bred to Rip-CreTG mice, thereby deleting GC-A selectively in ß-cells (ß GC-A KO). Weight gain, glucose tolerance, insulin sensitivity, and glucose-stimulated insulin secretion were monitored in normal diet (ND)- and high-fat diet (HFD)-fed mice. ß-cell size and number were measured by immunofluorescence-based islet morphometry. RESULTS: In vitro, the insulinotropic and proliferative actions of ANP were abolished in islets isolated from ß GC-A KO mice. Concordantly, in vivo, infusion of BNP mildly enhanced baseline plasma insulin levels and glucose-induced insulin secretion in control mice. This effect of exogenous BNP was abolished in ß GC-A KO mice, corroborating the efficient inactivation of the GC-A receptor in ß-cells. Despite this under physiological, ND conditions, fasted and fed insulin levels, glucose-induced insulin secretion, glucose tolerance and ß-cell morphology were similar in ß GC-A KO mice and control littermates. However, HFD-fed ß GC-A KO animals had accelerated glucose intolerance and diminished adaptative ß-cell proliferation. CONCLUSIONS: Our studies of ß GC-A KO mice demonstrate that the cardiac hormones ANP and BNP do not modulate ß-cell's growth and secretory functions under physiological, normal dietary conditions. However, endogenous NP/GC-A signaling improves the initial adaptative response of ß-cells to HFD-induced obesity. Impaired ß-cell NP/GC-A signaling in obese individuals might contribute to the development of type 2 diabetes.


Subject(s)
Atrial Natriuretic Factor/metabolism , Blood Glucose/metabolism , Gene Deletion , Glucose Intolerance/etiology , Insulin-Secreting Cells/enzymology , Obesity/complications , Receptors, Atrial Natriuretic Factor/deficiency , Animals , Cell Proliferation , Disease Models, Animal , Disease Progression , Genetic Predisposition to Disease , Glucose Intolerance/enzymology , Glucose Intolerance/genetics , Glucose Intolerance/pathology , Insulin/blood , Insulin-Secreting Cells/pathology , Mice, Knockout , Natriuretic Peptide, Brain/metabolism , Obesity/enzymology , Obesity/genetics , Phenotype , Receptors, Atrial Natriuretic Factor/genetics , Signal Transduction , Tissue Culture Techniques
9.
Circulation ; 138(5): 494-508, 2018 07 31.
Article in English | MEDLINE | ID: mdl-29626067

ABSTRACT

BACKGROUND: Peripheral vascular resistance has a major impact on arterial blood pressure levels. Endothelial C-type natriuretic peptide (CNP) participates in the local regulation of vascular tone, but the target cells remain controversial. The cGMP-producing guanylyl cyclase-B (GC-B) receptor for CNP is expressed in vascular smooth muscle cells (SMCs). However, whereas endothelial cell-specific CNP knockout mice are hypertensive, mice with deletion of GC-B in vascular SMCs have unaltered blood pressure. METHODS: We analyzed whether the vasodilating response to CNP changes along the vascular tree, ie, whether the GC-B receptor is expressed in microvascular types of cells. Mice with a floxed GC-B ( Npr2) gene were interbred with Tie2-Cre or PDGF-Rß-Cre ERT2 lines to develop mice lacking GC-B in endothelial cells or in precapillary arteriolar SMCs and capillary pericytes. Intravital microscopy, invasive and noninvasive hemodynamics, fluorescence energy transfer studies of pericyte cAMP levels in situ, and renal physiology were combined to dissect whether and how CNP/GC-B/cGMP signaling modulates microcirculatory tone and blood pressure. RESULTS: Intravital microscopy studies revealed that the vasodilatatory effect of CNP increases toward small-diameter arterioles and capillaries. CNP consistently did not prevent endothelin-1-induced acute constrictions of proximal arterioles, but fully reversed endothelin effects in precapillary arterioles and capillaries. Here, the GC-B receptor is expressed both in endothelial and mural cells, ie, in pericytes. It is notable that the vasodilatatory effects of CNP were preserved in mice with endothelial GC-B deletion, but abolished in mice lacking GC-B in microcirculatory SMCs and pericytes. CNP, via GC-B/cGMP signaling, modulates 2 signaling cascades in pericytes: it activates cGMP-dependent protein kinase I to phosphorylate downstream targets such as the cytoskeleton-associated vasodilator-activated phosphoprotein, and it inhibits phosphodiesterase 3A, thereby enhancing pericyte cAMP levels. These pathways ultimately prevent endothelin-induced increases of pericyte calcium levels and pericyte contraction. Mice with deletion of GC-B in microcirculatory SMCs and pericytes have elevated peripheral resistance and chronic arterial hypertension without a change in renal function. CONCLUSIONS: Our studies indicate that endothelial CNP regulates distal arteriolar and capillary blood flow. CNP-induced GC-B/cGMP signaling in microvascular SMCs and pericytes is essential for the maintenance of normal microvascular resistance and blood pressure.


Subject(s)
Arterial Pressure/drug effects , Endothelial Cells/drug effects , Hypertension/metabolism , Microcirculation/drug effects , Microvessels/drug effects , Natriuretic Peptide, C-Type/pharmacology , Pericytes/metabolism , Vasodilation/drug effects , Vasodilator Agents/pharmacology , Animals , Biosensing Techniques , Calcium Signaling/drug effects , Cells, Cultured , Cyclic GMP/metabolism , Endothelial Cells/metabolism , Fluorescence Resonance Energy Transfer , Genetic Predisposition to Disease , Hypertension/genetics , Hypertension/physiopathology , Mice, Inbred C57BL , Mice, Knockout , Microvessels/metabolism , Microvessels/physiopathology , Natriuretic Peptide, C-Type/metabolism , Paracrine Communication/drug effects , Phenotype , Receptor, Platelet-Derived Growth Factor beta/deficiency , Receptor, Platelet-Derived Growth Factor beta/genetics , Receptors, Atrial Natriuretic Factor/deficiency , Receptors, Atrial Natriuretic Factor/genetics
10.
Circ Res ; 122(3): 405-416, 2018 02 02.
Article in English | MEDLINE | ID: mdl-29273600

ABSTRACT

RATIONALE: Aortic valve disease is a cell-mediated process without effective pharmacotherapy. CNP (C-type natriuretic peptide) inhibits myofibrogenesis and osteogenesis of cultured valve interstitial cells and is downregulated in stenotic aortic valves. However, it is unknown whether CNP signaling regulates aortic valve health in vivo. OBJECTIVE: The aim of this study is to determine whether a deficient CNP signaling axis in mice causes accelerated progression of aortic valve disease. METHODS AND RESULTS: In cultured porcine valve interstitial cells, CNP inhibited pathological differentiation via the guanylate cyclase NPR2 (natriuretic peptide receptor 2) and not the G-protein-coupled clearance receptor NPR3 (natriuretic peptide receptor 3). We used Npr2+/- and Npr2+/-;Ldlr-/- mice and wild-type littermate controls to examine the valvular effects of deficient CNP/NPR2 signaling in vivo, in the context of both moderate and advanced aortic valve disease. Myofibrogenesis in cultured Npr2+/- fibroblasts was insensitive to CNP treatment, whereas aged Npr2+/- and Npr2+/-;Ldlr-/- mice developed cardiac dysfunction and ventricular fibrosis. Aortic valve function was significantly impaired in Npr2+/- and Npr2+/-;Ldlr-/- mice versus wild-type littermates, with increased valve thickening, myofibrogenesis, osteogenesis, proteoglycan synthesis, collagen accumulation, and calcification. 9.4% of mice heterozygous for Npr2 had congenital bicuspid aortic valves, with worse aortic valve function, fibrosis, and calcification than those Npr2+/- with typical tricuspid aortic valves or all wild-type littermate controls. Moreover, cGK (cGMP-dependent protein kinase) activity was downregulated in Npr2+/- valves, and CNP triggered synthesis of cGMP and activation of cGK1 (cGMP-dependent protein kinase 1) in cultured porcine valve interstitial cells. Finally, aged Npr2+/-;Ldlr-/- mice developed dilatation of the ascending aortic, with greater aneurysmal progression in Npr2+/- mice with bicuspid aortic valves than those with tricuspid valves. CONCLUSIONS: Our data establish CNP/NPR2 signaling as a novel regulator of aortic valve development and disease and elucidate the therapeutic potential of targeting this pathway to arrest disease progression.


Subject(s)
Aortic Aneurysm/genetics , Aortic Valve/abnormalities , Heart Valve Diseases/genetics , Natriuretic Peptide, C-Type/physiology , Receptors, Atrial Natriuretic Factor/deficiency , Ventricular Dysfunction, Left/genetics , Animals , Aorta/pathology , Aortic Aneurysm/physiopathology , Aortic Valve/physiopathology , Aortic Valve Stenosis/genetics , Aortic Valve Stenosis/physiopathology , Bicuspid Aortic Valve Disease , Calcinosis/genetics , Calcinosis/physiopathology , Cells, Cultured , Collagen/biosynthesis , Cyclic GMP/physiology , Cyclic GMP-Dependent Protein Kinase Type I/metabolism , Extracellular Matrix/pathology , Hyperlipidemias/complications , Hyperlipidemias/genetics , Mice , Mice, Knockout , Myofibroblasts/cytology , Natriuretic Peptide, C-Type/pharmacology , Osteogenesis , Proteoglycans/biosynthesis , Receptors, Atrial Natriuretic Factor/physiology , Receptors, LDL/deficiency , Receptors, LDL/genetics , Swine , Ventricular Dysfunction, Left/physiopathology
11.
Sci Rep ; 7(1): 17564, 2017 12 14.
Article in English | MEDLINE | ID: mdl-29242602

ABSTRACT

Natriuretic peptides (NPs) play essential roles in the regulation of cardiovascular function. NP effects are mediated by receptors known as NPR-A, NPR-B or NPR-C. NPs have potent effects on regulation of heart rate (HR) by the autonomic nervous system (ANS), but the role of NPR-C in these effects has not been investigated. Accordingly, we have used telemetric ECG recordings in awake, freely moving wildtype and NPR-C knockout (NPR-C-/-) mice and performed heart rate variability (HRV) analysis to assess alterations in sympatho-vagal balance on the heart following loss of NPR-C. Our novel data demonstrate that NPR-C-/- mice are characterized by elevations in HR, reductions in circadian changes in HR and enhanced occurrence of sinus pauses, indicating increased arrhythmogenesis and a loss of HRV. Time domain and frequency domain analyses further demonstrate that HRV is reduced in NPR-C-/- mice in association with a reduction in parasympathetic activity. Importantly, the low frequency to high frequency ratio was increased in NPR-C-/- mice indicating that sympathetic activity is also enhanced. These changes in autonomic regulation were confirmed using atropine and propranolol to antagonize the ANS. These findings illustrate that loss of NPR-C reduces HRV due to perturbations in the regulation of the heart by the ANS.


Subject(s)
Autonomic Nervous System/physiology , Heart Rate , Receptors, Atrial Natriuretic Factor/deficiency , Animals , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/physiopathology , Autonomic Nervous System/physiopathology , Gene Knockout Techniques , Mice , Receptors, Atrial Natriuretic Factor/genetics
12.
Am J Physiol Renal Physiol ; 313(3): F781-F795, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28566502

ABSTRACT

Guanylyl cyclase/natriuretic peptide receptor-A (GC-A/NPRA) plays a critical role in the regulation of blood pressure and fluid volume homeostasis. Mice lacking functional Npr1 (coding for GC-A/NPRA) exhibit hypertension and congestive heart failure. However, the underlying mechanisms remain largely less clear. The objective of the present study was to determine the physiological efficacy and impact of all-trans-retinoic acid (ATRA) and sodium butyrate (NaBu) in ameliorating the renal fibrosis, inflammation, and hypertension in Npr1 gene-disrupted haplotype (1-copy; +/-) mice (50% expression levels of NPRA). Both ATRA and NaBu, either alone or in combination, decreased the elevated levels of renal proinflammatory and profibrotic cytokines and lowered blood pressure in Npr1+/- mice compared with untreated controls. The treatment with ATRA-NaBu facilitated the dissociation of histone deacetylase (HDAC) 1 and 2 from signal transducer and activator of transcription 1 (STAT1) and enhanced its acetylation in the kidneys of Npr1+/- mice. The acetylated STAT1 formed a complex with nuclear factor-κB (NF-κB) p65, thereby inhibiting its DNA-binding activity and downstream proinflammatory and profibrotic signaling cascades. The present results demonstrate that the treatment of the haplotype Npr1+/- mice with ATRA-NaBu significantly lowered blood pressure and reduced the renal inflammation and fibrosis involving the interactive roles of HDAC, NF-κB (p65), and STAT1. The current findings will help in developing the molecular therapeutic targets and new treatment strategies for hypertension and renal dysfunction in humans.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Butyric Acid/pharmacology , Haplotypes , Histone Deacetylase 1/metabolism , Histone Deacetylase 2/metabolism , Histone Deacetylase Inhibitors/pharmacology , Kidney/drug effects , Nephritis/prevention & control , Receptors, Atrial Natriuretic Factor/deficiency , STAT1 Transcription Factor/metabolism , Transcription Factor RelA/metabolism , Tretinoin/pharmacology , Acetylation , Animals , Blood Pressure/drug effects , Cytokines/metabolism , Disease Models, Animal , Fibrosis , Genetic Predisposition to Disease , Inflammation Mediators/metabolism , Kidney/enzymology , Kidney/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Nephritis/enzymology , Nephritis/genetics , Nephritis/pathology , Phenotype , Receptors, Atrial Natriuretic Factor/genetics , Signal Transduction/drug effects
13.
Eur J Pharmacol ; 782: 44-58, 2016 Jul 05.
Article in English | MEDLINE | ID: mdl-27108789

ABSTRACT

Natriuretic peptide receptor-C (NPR-C) is considered as a clearance receptor that maintains the circulatory levels of natriuretic peptides. It has been suggested that augmented expression of NPR-C as a cause for the diminished anti-hypertrophic action of natriuretic peptides in the failing heart. Hence, we sought to determine the level of Npr3 gene (coding for NPR-C) expression in the Isoproterenol (ISO) treated Wistar rats. In addition, we studied the effect of Npr3 gene silencing on the hypertrophic growth. A significant increase in heart weight-to-body weight ratio (HW/BW-24%,P<0.01), an indicator of cardiac hypertrophic growth was observed in the ISO (10mg/kg BW/day,i.p for 7 days) treated rats. As expected, the cardiac NPR-C protein expression was significantly increased by 4 fold as compared to control rats. In parallel, the circulatory atrial natriuretic peptide (ANP) level was significantly decreased (2 fold) in ISO treated rats. Upon treatment with siRNA-Npr3, a significant decrease in the cardiac NPR-C protein expression (70%,P<0.01), HW/BW ratio (70%,P<0.01) and hypertrophic marker genes (α-Sk, ß-MHC, c-fos, P<0.01, respectively) mRNA expression were observed. Interestingly, the circulatory ANP level was increased by 1.5 fold in the siRNA-Npr3 treated rats as compared to ISO treated rats. Moreover, the cardiac collagen content, matrixmetalloprotinases-2 (MMP-2) and enzymatic antioxidant status (P<0.01, respectively) were found to be restored back to near normal upon siRNA-Npr3 treatment. Taken together, the results of this study indicates that specific down-regulation of Npr3 gene improves the circulatory levels of ANP and antioxidant system and there by attenuates the ß-adrenoceptor over-activation mediated cardiac hypertrophic growth in experimental rats.


Subject(s)
Atrial Natriuretic Factor/blood , Gene Silencing , Receptors, Adrenergic, beta/metabolism , Receptors, Atrial Natriuretic Factor/deficiency , Receptors, Atrial Natriuretic Factor/genetics , Animals , Biomarkers/blood , Cell Line , Collagen/metabolism , Down-Regulation/genetics , Heart Ventricles/metabolism , Heart Ventricles/pathology , Male , Matrix Metalloproteinase 2/metabolism , RNA, Small Interfering/genetics , Rats , Reactive Oxygen Species/metabolism
14.
Endocrinology ; 157(1): 358-67, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26517044

ABSTRACT

Atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP) bind to the receptor guanylyl cyclase (GC)-A, leading to diuresis, natriuresis, and blood vessel dilation. In addition, ANP and BNP have various angiogenic properties in ischemic tissue. When breeding mice devoid of GC-A, we noted significant skewing of the Mendelian ratio in the offspring, suggesting embryonic lethality due to knockout of GC-A. Consequently, we here investigated the roles of endogenous ANP and BNP in embryonic neovascularization and organ morphogenesis. Embryos resulting from GC-A(-/-) × GC-A(+/-) crosses developed hydrops fetalis (HF) beginning at embryonic day (E)14.5. All embryos with HF had the genotype GC-A(-/-). At E17.5, 33.3% (12 of 36) of GC-A(-/-) embryos had HF, and all GC-A(-/-) embryos with HF were dead. Beginning at E16.0, HF-GC-A(-/-) embryos demonstrated poorly developed superficial vascular vessels and sc hemorrhage, the fetal side of the placenta appeared ischemic, and vitelline vessels on the yolk sac were poorly developed. Furthermore, HF-GC-A(-/-) embryos also showed abnormal constriction of umbilical cord vascular vessels, few cardiac trabeculae and a thin compact zone, hepatic hemorrhage, and poor bone development. Electron microscopy of E16.5 HF-GC-A(-/-) embryos revealed severe vacuolar degeneration in endothelial cells, and the expected 3-layer structure of the smooth muscle wall of the umbilical artery was indistinct. These data demonstrate the importance of the endogenous ANP/BNP-GC-A system not only in the neovascularization of ischemic tissues but also in embryonic vascular development and organ morphogenesis.


Subject(s)
Atrial Natriuretic Factor/metabolism , Embryo, Mammalian/metabolism , Gene Expression Regulation, Developmental , Natriuretic Peptide, Brain/metabolism , Neovascularization, Physiologic , Organogenesis , Receptors, Atrial Natriuretic Factor/metabolism , Animals , Atrial Natriuretic Factor/genetics , Cells, Cultured , Crosses, Genetic , Embryo, Mammalian/cytology , Embryo, Mammalian/pathology , Embryo, Mammalian/ultrastructure , Female , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/ultrastructure , Humans , Hydrops Fetalis/genetics , Hydrops Fetalis/pathology , Hydrops Fetalis/veterinary , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Mice, Knockout , Microscopy, Electron, Transmission , Natriuretic Peptide, Brain/genetics , Pregnancy , Receptors, Atrial Natriuretic Factor/agonists , Receptors, Atrial Natriuretic Factor/deficiency , Receptors, Atrial Natriuretic Factor/genetics , Signal Transduction
15.
Histochem Cell Biol ; 144(4): 365-75, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26059418

ABSTRACT

Guanylyl cyclase A (GC-A), the receptor for atrial and B-type natriuretic peptides, is implicated in the regulation of blood pressure and cardiac growth. We used design-based stereological methods to examine the effect of GC-A inactivation on cardiomyocyte volume, number and subcellular composition in postnatal mice at day P2. In mice with global, systemic GC-A deletion, the cardiomyocyte number was significantly increased, demonstrating that hyperplasia is the main cause for the increase in ventricle weight in these early postnatal animals. In contrast, conditional, cardiomyocyte-restricted inactivation of GC-A had no significant effect on ventricle weight or cardiomyocyte number. The mean volume of cardiomyocytes and the myocyte-related volumes of the four major cell organelles (myofibrils, mitochondria, nuclei and sarcoplasm) were similar between genotypes. Taken together, systemic GC-A deficiency induces cardiac enlargement based on a higher number of normally composed and sized cardiomyocytes early after birth, whereas cardiomyocyte-specific GC-A abrogation is not sufficient to induce cardiac enlargement and has no effect on number, size and composition of cardiomyocytes. We conclude that postnatal cardiac hyperplasia in mice with global GC-A inactivation is provoked by systemic alterations, e.g., arterial hypertension. Direct GC-A-mediated effects in cardiomyocytes seem not to be involved in the regulation of myocyte proliferation at this early stage.


Subject(s)
Cardiomegaly/enzymology , Gene Deletion , Myocytes, Cardiac/enzymology , Receptors, Atrial Natriuretic Factor/deficiency , Animals , Animals, Newborn , Cardiomegaly/genetics , Cardiomegaly/pathology , Cell Count , Cell Proliferation , Genotype , Hyperplasia , Mice, Knockout , Myocytes, Cardiac/pathology , Phenotype , Receptors, Atrial Natriuretic Factor/genetics
16.
Sci Rep ; 5: 10554, 2015 May 27.
Article in English | MEDLINE | ID: mdl-26014585

ABSTRACT

Recent studies revealed C-type natriuretic peptide (CNP) and its receptor, guanylyl cyclase-B (GC-B) are potent stimulators of endochondral bone growth. As they exist ubiquitously in body, we investigated the physiological role of the local CNP/GC-B in the growth plate on bone growth using cartilage-specific knockout mice. Bones were severely shorter in cartilage-specific CNP or GC-B knockout mice and the extent was almost the same as that in respective systemic knockout mice. Cartilage-specific GC-B knockout mice were shorter than cartilage-specific CNP knockout mice. Hypertrophic chondrocyte layer of the growth plate was drastically reduced and proliferative chondrocyte layer, along with the proliferation of chondrocytes there, was moderately reduced in either cartilage-specific knockout mice. The survival rate of cartilage-specific CNP knockout mice was comparable to that of systemic CNP knockout mice. The local CNP/GC-B system in growth plate is responsible for physiological endochondral bone growth and might further affect mortality via unknown mechanisms.


Subject(s)
Bone Development/physiology , Growth Plate/metabolism , Natriuretic Peptide, C-Type/metabolism , Receptors, Atrial Natriuretic Factor/metabolism , Animals , Bone and Bones/diagnostic imaging , Bone and Bones/pathology , Cartilage/metabolism , Growth Plate/pathology , In Situ Hybridization , Mice , Mice, Inbred C57BL , Mice, Knockout , Natriuretic Peptide, C-Type/deficiency , Natriuretic Peptide, C-Type/genetics , Phenotype , Radiography , Receptors, Atrial Natriuretic Factor/deficiency , Receptors, Atrial Natriuretic Factor/genetics , Survival Rate
17.
Clin Exp Nephrol ; 19(2): 197-207, 2015 Apr.
Article in English | MEDLINE | ID: mdl-24845230

ABSTRACT

BACKGROUND AND AIM: The infusion of chronic angiotensin II (Ang II) has been shown to promote renal interstitial fibrosis. To evaluate the pathophysiological significance of the natriuretic peptide-GC-A system, we infused Ang II (1.0 mg/kg/day) in GC-A-deficient mice (GC-A-KO). METHODS: We used 5 groups (Wild-Saline n = 12, Wild-Ang II n = 14, GC-A-KO-Saline n = 11, GC-A-KO-Ang II n = 13, and GC-A-KO-Ang II-Hydralazine n = 10). Saline or Ang II was infused subcutaneously using an osmotic minipump for 3 weeks. Hydralazine was administered orally (0.05 g/L in drinking water). RESULTS: Systolic blood pressure was significantly higher in the GC-A-KO-Saline group (130 ± 12 mmHg) than in the Wild-Saline group (105 ± 30 mmHg), and was similar to that in the Wild-Ang II (141 ± 17 mmHg) and GC-A-KO-Ang II-Hydralazine (140 ± 20 mmHg) groups. Systolic blood pressure was significantly higher in the GC-A-KO-Ang II group (159 ± 21 mmHg) than in the 4 other groups. Renal tubular atrophy and interstitial fibrosis were significantly more severe in the GC-A-KO-Ang II group (atrophy 13.4 %, fibrosis 12.0 %) than in the Wild-Saline (0, 2.0 %), Wild-Ang II (2.9, 4.4 %), and GC-A-KO-Saline (0, 2.6 %) groups. Hydralazine could not inhibit this aggravation (GC-A-KO-Ang II-Hydralazine 13.5, 11.3 %). The expression of monocyte chemotactic protein-1 in tubular cells, and F4/80 and alpha-smooth muscle actin in the interstitium was clearly detected in the Ang II-infused wild and GC-A-KO groups and was associated with renal tubular atrophy and interstitial fibrosis. The expression of E-cadherin in tubular cells was absent in the Ang II-infused wild and GC-A-KO groups and was associated with renal tubular atrophy. CONCLUSIONS: The natriuretic peptide-GC-A system may play an inhibitory role in Ang II-induced renal tubular atrophy, interstitial fibrosis, and phenotypic transformation in renal tubular cells and fibroblasts.


Subject(s)
Angiotensin II/pharmacology , Antihypertensive Agents/pharmacology , Hydralazine/pharmacology , Kidney Tubules/pathology , Receptors, Atrial Natriuretic Factor/deficiency , Vasoconstrictor Agents/pharmacology , Actins/analysis , Animals , Antigens, Differentiation/analysis , Antigens, Differentiation/genetics , Atrophy , Blood Pressure/drug effects , Blood Pressure/genetics , Cadherins/analysis , Chemokine CCL2/analysis , Fibrosis , Gene Expression/drug effects , Intracellular Signaling Peptides and Proteins/genetics , Kidney Tubules/chemistry , Male , Membrane Proteins/genetics , Mice , Mice, Knockout , Osteopontin/genetics , RNA, Messenger/metabolism , Receptors, Atrial Natriuretic Factor/genetics , Sodium Chloride/pharmacology
18.
J Clin Invest ; 124(6): 2464-71, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24837433

ABSTRACT

Inhibition of cGMP-specific phosphodiesterase 5 (PDE5) ameliorates pathological cardiac remodeling and has been gaining attention as a potential therapy for heart failure. Despite promising results in males, the efficacy of the PDE5 inhibitor sildenafil in female cardiac pathologies has not been determined and might be affected by estrogen levels, given the hormone's involvement in cGMP synthesis. Here, we determined that the heart-protective effect of sildenafil in female mice depends on the presence of estrogen via a mechanism that involves myocyte eNOS-dependent cGMP synthesis and the cGMP-dependent protein kinase Iα (PKGIα). Sildenafil treatment failed to exert antiremodeling properties in female pathological hearts from Gαq-overexpressing or pressure-overloaded mice after ovary removal; however, estrogen replacement restored the effectiveness of sildenafil in these animals. In females, sildenafil-elicited myocardial PKG activity required estrogen, which stimulated tonic cardiomyocyte cGMP synthesis via an eNOS/soluble guanylate cyclase pathway. In contrast, eNOS activation, cGMP synthesis, and sildenafil efficacy were not estrogen dependent in male hearts. Estrogen and sildenafil had no impact on pressure-overloaded hearts from animals expressing dysfunctional PKGIα, indicating that PKGIα mediates antiremodeling effects. These results support the importance of sex differences in the use of PDE5 inhibitors for treating heart disease and the critical role of estrogen status when these agents are used in females.


Subject(s)
Estrogens/metabolism , Heart Diseases/drug therapy , Heart Diseases/metabolism , Phosphodiesterase 5 Inhibitors/pharmacology , Animals , Cardiotonic Agents/pharmacology , Cyclic GMP/biosynthesis , Cyclic GMP-Dependent Protein Kinase Type I/metabolism , Disease Models, Animal , Estradiol/administration & dosage , Female , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Guanylate Cyclase/metabolism , Heart Failure/drug therapy , Heart Failure/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type III/deficiency , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Ovariectomy , Piperazines/pharmacology , Purines/pharmacology , Receptors, Atrial Natriuretic Factor/deficiency , Receptors, Atrial Natriuretic Factor/genetics , Receptors, Atrial Natriuretic Factor/metabolism , Sex Characteristics , Sildenafil Citrate , Sulfones/pharmacology , Treatment Outcome
19.
Am J Physiol Renal Physiol ; 306(12): F1489-98, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24717731

ABSTRACT

The renin-angiotensin-aldosterone system and cardiac natriuretic peptides [atrial natriuretic peptide (ANP) and B-type natriuretic peptide (BNP)] are opposing control mechanisms for arterial blood pressure. Accordingly, an inverse relationship between plasma renin concentration (PRC) and ANP exists in most circumstances. However, PRC and ANP levels are both elevated in renovascular hypertension. Because ANP can directly suppress renin release, we used ANP knockout (ANP(-/-)) mice to investigate whether high ANP levels attenuate the increase in PRC in response to renal hypoperfusion, thus buffering renovascular hypertension. ANP(-/-) mice were hypertensive and had reduced PRC compared with that in wild-type ANP(+/+) mice under control conditions. Unilateral renal artery stenosis (2-kidney, 1-clip) for 1 wk induced similar increases in blood pressure and PRC in both genotypes. Unexpectedly, plasma BNP concentrations in ANP(-/-) mice significantly increased in response to two-kidney, one-clip treatment, potentially compensating for the lack of ANP. In fact, in mice lacking guanylyl cyclase A (GC-A(-/-) mice), which is the common receptor for both ANP and BNP, renovascular hypertension was markedly augmented compared with that in wild-type GC-A(+/+) mice. However, the higher blood pressure in GC-A(-/-) mice was not caused by disinhibition of the renin system because PRC and renal renin synthesis were significantly lower in GC-A(-/-) mice than in GC-A(+/+) mice. Thus, natriuretic peptides buffer renal vascular hypertension via renin-independent effects, such as vasorelaxation. The latter possibility is supported by experiments in isolated perfused mouse kidneys, in which physiological concentrations of ANP and BNP elicited renal vasodilatation and attenuated renal vasoconstriction in response to angiotensin II.


Subject(s)
Atrial Natriuretic Factor/metabolism , Hypertension, Renovascular/metabolism , Hypertension, Renovascular/physiopathology , Natriuretic Peptide, Brain/metabolism , Renin/metabolism , Animals , Atrial Natriuretic Factor/deficiency , Atrial Natriuretic Factor/genetics , Blood Pressure/physiology , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Atrial Natriuretic Factor/deficiency , Receptors, Atrial Natriuretic Factor/genetics , Receptors, Atrial Natriuretic Factor/metabolism , Renin-Angiotensin System/physiology , Surgical Instruments , Vasoconstriction/physiology , Vasodilation/physiology
20.
J Neurosci ; 34(3): 737-47, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24431432

ABSTRACT

Axonal branching is a prerequisite for the establishment of complex neuronal circuits and their capacity for parallel information processing. Previously, we have identified a cGMP signaling pathway composed of the ligand C-type natriuretic peptide (CNP), its receptor, the guanylyl cyclase natriuretic peptide receptor 2 (Npr2), and the cGMP-dependent kinase Iα (cGKIα) that regulates axon bifurcation of dorsal root ganglion (DRG) neurons in the spinal cord. Now we asked whether this cascade also controls axon bifurcation elsewhere in the nervous system. An Npr2-lacZ reporter mouse line was generated to clarify the pattern of the CNP receptor expression. It was found that during the period of axonal outgrowth, Npr2 and cGKIα were strongly labeled in neurons of all cranial sensory ganglia (gV, gVII, gVIII, gIX, and gX). In addition, strong complementary expression of CNP was detected in the hindbrain at the entry zones of sensory afferents. To analyze axon branching in individual Npr2-positive neurons, we generated a mouse mutant expressing a tamoxifen-inducible variant of Cre recombinase expressed under control of the Npr2-promoter (Npr2-CreER(T2)). After crossing this strain with conditional reporter mouse lines, we revealed that the complete absence of Npr2 activity indeed prohibited the bifurcation of cranial sensory axons in their entrance region. Consequently, axons only turned in either an ascending or descending direction, while collateral formation and growth of the peripheral arm was not affected. These findings indicate that in neurons of the cranial sensory ganglia, as in DRG neurons, cGMP signals are necessary for the execution of an axonal bifurcation program.


Subject(s)
Axons/chemistry , Cranial Nerves/chemistry , Cyclic GMP/genetics , Receptors, Atrial Natriuretic Factor/deficiency , Sensory Receptor Cells/chemistry , Signal Transduction/genetics , Animals , Axons/physiology , Cranial Nerves/pathology , Cranial Nerves/physiology , Cyclic GMP/physiology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/pathology , Embryonic Stem Cells/physiology , Female , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Mutant Strains , Mice, Transgenic , Receptors, Atrial Natriuretic Factor/genetics , Receptors, Atrial Natriuretic Factor/physiology , Sensory Receptor Cells/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...