Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters










Publication year range
1.
Am J Physiol Heart Circ Physiol ; 324(4): H391-H410, 2023 04 01.
Article in English | MEDLINE | ID: mdl-36607797

ABSTRACT

This study reports a new methodology for right heart imaging by ultrasound in mice under right ventricular (RV) pressure overload. Pulmonary artery constriction (PAC) or sham surgeries were performed on C57BL/6 male mice at 8 wk of age. Ultrasound imaging was conducted at 2, 4, and 8 wk postsurgery using both classical and advanced ultrasound imaging modalities including electrocardiogram (ECG)-based kilohertz visualization, anatomical M-mode, and strain imaging. Based on pulsed Doppler, the PAC group demonstrated dramatically enhanced pressure gradient in the main pulmonary artery (MPA) as compared with the sham group. By the application of advanced imaging modalities in novel short-axis views of the ventricles, the PAC group demonstrated increased thickness of RV free wall, enlarged RV chamber, and reduced RV fractional shortening compared with the sham group. The PAC group also showed prolonged RV contraction, asynchronous interplay between RV and left ventricle (LV), and passive leftward motion of the interventricular septum (IVS) at early diastole. Consequently, the PAC group exhibited prolongation of LV isovolumic relaxation time, without change in LV wall thickness or systolic function. Significant correlations were found between the maximal pressure gradient in MPA measured by Doppler and the RV systolic pressure by catheterization, as well as the morphological and functional parameters of RV by ultrasound.NEW & NOTEWORTHY The established protocol overcomes the challenges in right heart imaging in mice, thoroughly elucidating the changes of RV, the dynamics of IVS, and the impact on LV and provides new insights into the pathophysiological mechanism of RV remodeling.


Subject(s)
Ventricular Dysfunction, Right , Ventricular Remodeling , Male , Animals , Mice , Mice, Inbred C57BL , Heart , Heart Ventricles/diagnostic imaging , Ultrasonography , Ventricular Dysfunction, Right/diagnostic imaging , Ventricular Dysfunction, Right/etiology , Ventricular Pressure/physiology , Ventricular Function, Right
2.
Cardiovasc Res ; 119(6): 1403-1415, 2023 06 13.
Article in English | MEDLINE | ID: mdl-36418171

ABSTRACT

AIMS: Circadian rhythms orchestrate important functions in the cardiovascular system: the contribution of microvascular rhythms to cardiovascular disease progression/severity is unknown. This study hypothesized that (i) myogenic reactivity in skeletal muscle resistance arteries is rhythmic and (ii) disrupting this rhythmicity would alter cardiac injury post-myocardial infarction (MI). METHODS AND RESULTS: Cremaster skeletal muscle resistance arteries were isolated and assessed using standard pressure myography. Circadian rhythmicity was globally disrupted with the ClockΔ19/Δ19 mutation or discretely through smooth muscle cell-specific Bmal1 deletion (Sm-Bmal1 KO). Cardiac structure and function were determined by echocardiographic, hemodynamic and histological assessments. Myogenic reactivity in cremaster muscle resistance arteries is rhythmic. This rhythm is putatively mediated by the circadian modulation of a mechanosensitive signalosome incorporating tumour necrosis factor and casein kinase 1. Following left anterior descending coronary artery ligation, myogenic responsiveness is locked at the circadian maximum, although circadian molecular clock gene expression cycles normally. Disrupting the molecular clock abolishes myogenic rhythmicity: myogenic tone is suspended at the circadian minimum and is no longer augmented by MI. The reduced myogenic tone in ClockΔ19/Δ19 mice and Sm-Bmal1 KO mice associates with reduced total peripheral resistance (TPR), improved cardiac function and reduced infarct expansion post-MI. CONCLUSIONS: Augmented microvascular constriction aggravates cardiac injury post-MI. Following MI, skeletal muscle resistance artery myogenic reactivity increases specifically within the rest phase, when TPR would normally decline. Disrupting the circadian clock interrupts the MI-induced augmentation in myogenic reactivity: therapeutics targeting the molecular clock, therefore, may be useful for improving MI outcomes.


Subject(s)
Heart Injuries , Myocardial Infarction , Mice , Animals , ARNTL Transcription Factors/genetics , Myocardial Infarction/metabolism , Heart , Hemodynamics , Vascular Resistance
3.
Matrix Biol Plus ; 12: 100085, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34693248

ABSTRACT

Arterial stiffening is a significant predictor of cardiovascular disease development and mortality. In elastic arteries, stiffening refers to the loss and fragmentation of elastic fibers, with a progressive increase in collagen fibers. Type VIII collagen (Col-8) is highly expressed developmentally, and then once again dramatically upregulated in aged and diseased vessels characterized by arterial stiffening. Yet its biophysical impact on the vessel wall remains unknown. The purpose of this study was to test the hypothesis that Col-8 functions as a matrix scaffold to maintain vessel integrity during extracellular matrix (ECM) development. These changes are predicted to persist into the adult vasculature, and we have tested this in our investigation. Through our in vivo and in vitro studies, we have determined a novel interaction between Col-8 and elastin. Mice deficient in Col-8 (Col8-/-) had reduced baseline blood pressure and increased arterial compliance, indicating an enhanced Windkessel effect in conducting arteries. Differences in both the ECM composition and VSMC activity resulted in Col8-/- carotid arteries that displayed increased crosslinked elastin and functional distensibility, but enhanced catecholamine-induced VSMC contractility. In vitro studies revealed that the absence of Col-8 dramatically increased tropoelastin mRNA and elastic fiber deposition in the ECM, which was decreased with exogenous Col-8 treatment. These findings suggest a causative role for Col-8 in reducing mRNA levels of tropoelastin and the presence of elastic fibers in the matrix. Moreover, we also found that Col-8 and elastin have opposing effects on VSMC phenotype, the former promoting a synthetic phenotype, whereas the latter confers quiescence. These studies further our understanding of Col-8 function and open a promising new area of investigation related to elastin biology.

4.
Biomedicines ; 9(8)2021 Aug 13.
Article in English | MEDLINE | ID: mdl-34440212

ABSTRACT

A number of diverse G-protein signaling pathways have been shown to regulate insulin secretion from pancreatic ß-cells. Accordingly, regulator of G-protein signaling (RGS) proteins have also been implicated in coordinating this process. One such protein, RGS4, is reported to show both positive and negative effects on insulin secretion from ß-cells depending on the physiologic context under which it was studied. We here use an RGS4-deficient mouse model to characterize previously unknown G-protein signaling pathways that are regulated by RGS4 during glucose-stimulated insulin secretion from the pancreatic islets. Our data show that loss of RGS4 results in a marked deficiency in glucose-stimulated insulin secretion during both phase I and phase II of insulin release in intact mice and isolated islets. These deficiencies are associated with lower cAMP/PKA activity and a loss of normal calcium surge (phase I) and oscillatory (phase II) kinetics behavior in the RGS4-deficient ß-cells, suggesting RGS4 may be important for regulation of both Gαi and Gαq signaling control during glucose-stimulated insulin secretion. Together, these studies add to the known list of G-protein coupled signaling events that are controlled by RGS4 during glucose-stimulated insulin secretion and highlight the importance of maintaining normal levels of RGS4 function in healthy pancreatic tissues.

5.
J Proteome Res ; 20(5): 2867-2881, 2021 05 07.
Article in English | MEDLINE | ID: mdl-33789425

ABSTRACT

Heart failure (HF) is associated with pathological remodeling of the myocardium, including the initiation of fibrosis and scar formation by activated cardiac fibroblasts (CFs). Although early CF-dependent scar formation helps prevent cardiac rupture by maintaining the heart's structural integrity, ongoing deposition of the extracellular matrix in the remote and infarct regions can reduce tissue compliance, impair cardiac function, and accelerate progression to HF. In our study, we conducted mass spectrometry (MS) analysis to identify differentially altered proteins and signaling pathways between CFs isolated from 7 day sham and infarcted murine hearts. Surprisingly, CFs from both the remote and infarct regions of injured hearts had a wide number of similarly altered proteins and signaling pathways that were consistent with fibrosis and activation into pathological myofibroblasts. Specifically, proteins enriched in CFs isolated from MI hearts were involved in pathways pertaining to cell-cell and cell-matrix adhesion, chaperone-mediated protein folding, and collagen fibril organization. These results, together with principal component analyses, provided evidence of global CF activation postinjury. Interestingly, however, direct comparisons between CFs from the remote and infarct regions of injured hearts identified 15 differentially expressed proteins between MI remote and MI infarct CFs. Eleven of these proteins (Gpc1, Cthrc1, Vmac, Nexn, Znf185, Sprr1a, Specc1, Emb, Limd2, Pawr, and Mcam) were higher in MI infarct CFs, whereas four proteins (Gstt1, Gstm1, Tceal3, and Inmt) were higher in MI remote CFs. Collectively, our study shows that MI injury induced global changes to the CF proteome, with the magnitude of change reflecting their relative proximity to the site of injury.


Subject(s)
Myocardial Infarction , Ventricular Remodeling , Animals , Disease Models, Animal , Fibroblasts/pathology , Fibrosis , LIM Domain Proteins , Mice , Microfilament Proteins , Myocardial Infarction/genetics , Myocardium/pathology , Myofibroblasts/pathology
6.
J Vasc Res ; 57(6): 325-340, 2020.
Article in English | MEDLINE | ID: mdl-32777783

ABSTRACT

We have shown that both insulin and resveratrol (RSV) decrease neointimal hyperplasia in chow-fed rodents via mechanisms that are in part overlapping and involve the activation of endothelial nitric oxide synthase (eNOS). However, this vasculoprotective effect of insulin is abolished in high-fat-fed insulin-resistant rats. Since RSV, in addition to increasing insulin sensitivity, can activate eNOS via pathways that are independent of insulin signaling, such as the activation of sirtuin 1 (SIRT1) and AMP-activated kinase (AMPK), we speculated that unlike insulin, the vasculoprotective effect of RSV would be retained in high-fat-fed rats. We found that high-fat feeding decreased insulin sensitivity and increased neointimal area and that RSV improved insulin sensitivity (p < 0.05) and decreased neointimal area in high-fat-fed rats (p < 0.05). We investigated the role of SIRT1 in the effect of RSV using two genetic mouse models. We found that RSV decreased neointimal area in high-fat-fed wild-type mice (p < 0.05), an effect that was retained in mice with catalytically inactive SIRT1 (p < 0.05) and in heterozygous SIRT1-null mice. In contrast, the effect of RSV was abolished in AMKPα2-null mice. Thus, RSV decreased neointimal hyperplasia after arterial injury in both high-fat-fed rats and mice, an effect likely not mediated by SIRT1 but by AMPKα2.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Carotid Artery Injuries/drug therapy , Carotid Artery, Common/drug effects , Diet, High-Fat , Femoral Artery/drug effects , Neointima , Resveratrol/pharmacology , Sirtuin 1/metabolism , Vascular System Injuries/drug therapy , AMP-Activated Protein Kinases/genetics , Animals , Carotid Artery Injuries/enzymology , Carotid Artery Injuries/pathology , Carotid Artery, Common/enzymology , Carotid Artery, Common/pathology , Disease Models, Animal , Femoral Artery/enzymology , Femoral Artery/injuries , Femoral Artery/pathology , Insulin Resistance , Mice, Knockout , Rats, Sprague-Dawley , Signal Transduction , Sirtuin 1/genetics , Vascular System Injuries/enzymology , Vascular System Injuries/pathology
7.
J Cell Sci ; 133(12)2020 06 24.
Article in English | MEDLINE | ID: mdl-32501280

ABSTRACT

Intracellular pools of the heterotrimeric G-protein α-subunit Gαi3 (encoded by GNAI3) have been shown to promote growth factor signaling, while at the same time inhibiting the activation of JNK and autophagic signaling following nutrient starvation. The precise molecular mechanisms linking Gαi3 to both stress and growth factor signaling remain poorly understood. Importantly, JNK-mediated phosphorylation of Bcl-2 was previously found to activate autophagic signaling following nutrient deprivation. Our data shows that activated Gαi3 decreases Bcl-2 phosphorylation, whereas inhibitors of Gαi3, such as RGS4 and AGS3 (also known as GPSM1), markedly increase the levels of phosphorylated Bcl-2. Manipulation of the palmitoylation status and intracellular localization of RGS4 suggests that Gαi3 modulates phosphorylated Bcl-2 levels and autophagic signaling from discreet TGN38 (also known as TGOLN2)-labeled vesicle pools. Consistent with an important role for these molecules in normal tissue responses to nutrient deprivation, increased Gαi signaling within nutrient-starved adrenal glands from RGS4-knockout mice resulted in a dramatic abrogation of autophagic flux, compared to wild-type tissues. Together, these data suggest that the activity of Gαi3 and RGS4 from discreet TGN38-labeled vesicle pools are critical regulators of autophagic signaling that act via their ability to modulate phosphorylation of Bcl-2.


Subject(s)
Autophagy , RGS Proteins , Animals , Intracellular Membranes , Lipoylation , Mice , Phosphorylation , Signal Transduction
8.
Front Physiol ; 11: 402, 2020.
Article in English | MEDLINE | ID: mdl-32477159

ABSTRACT

Subarachnoid hemorrhage (SAH) is a devastating cerebral event caused by an aneurysmal rupture. In addition to neurological injury, SAH has significant effects on cardiac function and the peripheral microcirculation. Since these peripheral complications may exacerbate brain injury, the prevention and management of these peripheral effects are important for improving the overall clinical outcome after SAH. In this investigation, we examined the effects of SAH on cardiac function and vascular reactivity in a well-characterized blood injection model of SAH. Standard echocardiographic and blood pressure measurement procedures were utilized to assess cardiac function and hemodynamic parameters in vivo; we utilized a pressure myography approach to assess vascular reactivity in cremaster skeletal muscle resistance arteries ex vivo. We observed that elevated catecholamine levels in SAH stun the myocardium, reduce cardiac output and augment myogenic vasoconstriction in isolated cremaster arteries. These cardiac and vascular effects are driven by beta- and alpha-adrenergic receptor signaling, respectively. Clinically utilized adrenergic receptor antagonists can prevent cardiac injury and normalize vascular function. We found that tumor necrosis factor (TNF) gene deletion prevents the augmentation of myogenic reactivity in SAH: since membrane-bound TNF serves as a mechanosensor in the arteries assessed, alpha-adrenergic signaling putatively augments myogenic vasoconstriction by enhancing mechanosensor activity.

9.
Sci Rep ; 9(1): 7867, 2019 05 27.
Article in English | MEDLINE | ID: mdl-31133706

ABSTRACT

Pathological cardiac hypertrophy is a debilitating condition characterized by deleterious thickening of the myocardium, dysregulated Ca2+ signaling within cardiomyocytes, and contractile dysfunction. Importantly, the nanoscale organization, localization, and patterns of expression of critical Ca2+ handling regulators including dihydropyridine receptor (DHPR), ryanodine receptor 2 (RyR2), phospholamban (PLN), and sarco/endoplasmic reticulum Ca2+-ATPase 2A (SERCA2A) remain poorly understood, especially during pathological hypertrophy disease progression. In the current study, we induced cardiac pathological hypertrophy via transverse aortic constriction (TAC) on 8-week-old CD1 mice, followed by isolation of cardiac ventricular myocytes. dSTORM super-resolution imaging was then used to visualize proteins at nanoscale resolution at two time points and we quantified changes in protein cluster properties using Voronoi tessellation and 2D Fast Fourier Transform analyses. We showed a decrease in the density of DHPR and RyR2 clusters with pressure-overload cardiac hypertrophy and an increase in the density of SERCA2A protein clusters. PLN protein clusters decreased in density in 2-week TAC but returned to sham levels by 4-week TAC. Furthermore, 2D-FFT analysis revealed changes in molecular organization during pathological hypertrophy, with DHPR and RyR2 becoming dispersed while both SERCA2A and PLN sequestered into dense clusters. Our work reveals molecular adaptations that occur in critical SR proteins at a single molecule during pressure overload-induced cardiomyopathy. Nanoscale alterations in protein localization and patterns of expression of crucial SR proteins within the cardiomyocyte provided insights into the pathogenesis of cardiac hypertrophy, and specific evidence that cardiomyocytes undergo significant structural remodeling during the progression of pathological hypertrophy.


Subject(s)
Cardiomegaly/pathology , Myocytes, Cardiac/pathology , Sarcoplasmic Reticulum/pathology , Animals , Calcium Channels, L-Type/analysis , Calcium-Binding Proteins/analysis , Cardiomegaly/diagnostic imaging , Cells, Cultured , Fourier Analysis , Mice , Microscopy , Optical Imaging , Pressure , Ryanodine Receptor Calcium Release Channel/analysis , Sarcoplasmic Reticulum Calcium-Transporting ATPases/analysis
10.
JACC Basic Transl Sci ; 4(8): 940-958, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31909302

ABSTRACT

Heart failure (HF) and subarachnoid hemorrhage (SAH) chronically reduce cerebral perfusion, which negatively affects clinical outcome. This work demonstrates a strong relationship between cerebral artery cystic fibrosis transmembrane conductance regulator (CFTR) expression and altered cerebrovascular reactivity in HF and SAH. In HF and SAH, CFTR corrector compounds (C18 or lumacaftor) normalize pathological alterations in cerebral artery CFTR expression, vascular reactivity, and cerebral perfusion, without affecting systemic hemodynamic parameters. This normalization correlates with reduced neuronal injury. Therefore, CFTR therapeutics have emerged as valuable clinical tools to manage cerebrovascular dysfunction, impaired cerebral perfusion, and neuronal injury.

11.
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
12.
Mol Metab ; 6(11): 1339-1349, 2017 11.
Article in English | MEDLINE | ID: mdl-29107282

ABSTRACT

OBJECTIVES: Glucagon-like peptide-1 (GLP-1) is secreted from enteroendocrine cells and exerts a broad number of metabolic actions through activation of a single GLP-1 receptor (GLP-1R). The cardiovascular actions of GLP-1 have garnered increasing attention as GLP-1R agonists are used to treat human subjects with diabetes and obesity that may be at increased risk for development of heart disease. Here we studied mechanisms linking GLP-1R activation to control of heart rate (HR) in mice. METHODS: The actions of GLP-1R agonists were examined on the control of HR in wild type mice (WT) and in mice with cardiomyocyte-selective disruption of the GLP-1R (Glp1rCM-/-). Complimentary studies examined the effects of GLP-1R agonists in mice co-administered propranolol or atropine. The direct effects of GLP-1R agonism on HR and ventricular developed pressure were examined in isolated perfused mouse hearts ex vivo, and atrial depolarization was quantified in mouse hearts following direct application of liraglutide to perfused atrial preparations ex vivo. RESULTS: Doses of liraglutide and lixisenatide that were equipotent for acute glucose control rapidly increased HR in WT and Glp1rCM-/- mice in vivo. The actions of liraglutide to increase HR were more sustained relative to lixisenatide, and diminished in Glp1rCM-/- mice. The acute chronotropic actions of GLP-1R agonists were attenuated by propranolol but not atropine. Neither native GLP-1 nor lixisenatide increased HR or developed pressure in perfused hearts ex vivo. Moreover, liraglutide had no direct effect on sinoatrial node firing rate in mouse atrial preparations ex vivo. Despite co-localization of HCN4 and GLP-1R in primate hearts, HCN4-directed Cre expression did not attenuate levels of Glp1r mRNA transcripts, but did reduce atrial Gcgr expression in the mouse heart. CONCLUSIONS: GLP-1R agonists increase HR through multiple mechanisms, including regulation of autonomic nervous system function, and activation of the atrial GLP-1R. Surprisingly, the isolated atrial GLP-1R does not transduce a direct chronotropic effect following exposure to GLP-1R agonists in the intact heart, or isolated atrium, ex vivo. Hence, cardiac GLP-1R circuits controlling HR require neural inputs and do not function in a heart-autonomous manner.


Subject(s)
Glucagon-Like Peptide-1 Receptor/physiology , Heart Rate/physiology , Animals , Autonomic Nervous System/physiology , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/metabolism , Heart Rate/drug effects , Liraglutide/pharmacology , Male , Mice , Mice, Inbred C57BL , Peptides/pharmacology
13.
Nat Commun ; 8: 14805, 2017 04 05.
Article in English | MEDLINE | ID: mdl-28378814

ABSTRACT

Tumour necrosis factor (TNF) is a ubiquitously expressed cytokine with functions beyond the immune system. In several diseases, the induction of TNF expression in resistance artery smooth muscle cells enhances microvascular myogenic vasoconstriction and perturbs blood flow. This pathological role prompted our hypothesis that constitutively expressed TNF regulates myogenic signalling and systemic haemodynamics under non-pathological settings. Here we show that acutely deleting the TNF gene in smooth muscle cells or pharmacologically scavenging TNF with etanercept (ETN) reduces blood pressure and resistance artery myogenic responsiveness; the latter effect is conserved across five species, including humans. Changes in transmural pressure are transduced into intracellular signals by membrane-bound TNF (mTNF) that connect to a canonical myogenic signalling pathway. Our data positions mTNF 'reverse signalling' as an integral element of a microvascular mechanosensor; pathologic or therapeutic perturbations of TNF signalling, therefore, necessarily affect microvascular tone and systemic haemodynamics.


Subject(s)
Blood Pressure/physiology , Muscle, Smooth, Vascular/metabolism , Tumor Necrosis Factor-alpha/physiology , Animals , Dogs , Etanercept/pharmacology , Female , Heart Failure/prevention & control , Humans , Male , Mesocricetus , Mice, Inbred C57BL , Mice, Knockout , Microcirculation , Muscle, Skeletal/metabolism , Signal Transduction/physiology , Species Specificity , Swine , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/metabolism , Vasoconstriction
14.
Article in English | MEDLINE | ID: mdl-26389115

ABSTRACT

Heterotrimeric G-protein signaling has been shown to modulate a wide variety of intracellular signaling pathways, including the mitogen-activated protein kinase (MAPK) family. The activity of one MAPK family class, c-Jun N-terminal kinases (JNKs), has been traditionally linked to the activation of G-protein coupled receptors (GPCRs) at the plasma membrane. Using a unique set of G-protein signaling tools developed in our laboratory, we show that subcellular domain-specific JNK activity is inhibited by the activation of Gαi3, the Gαi isoform found predominantly within intracellular membranes, such as the endoplasmic reticulum (ER)-Golgi interface, and their associated vesicle pools. Regulators of intracellular Gαi3, including activator of G-protein signaling 3 (AGS3) and the regulator of G-protein signaling protein 4 (RGS4), have a marked impact on the regulation of JNK activity. Together, these data support the existence of unique intracellular signaling complexes that control JNK activity deep within the cell. This work highlights some of the cellular pathways that are regulated by these intracellular complexes and identifies potential strategies for their regulation in mammalian cells.

15.
PLoS One ; 10(7): e0133274, 2015.
Article in English | MEDLINE | ID: mdl-26196737

ABSTRACT

Gradients of the fast transient outward K+ current (Ito,f) contribute to heterogeneity of ventricular repolarization in a number of species. Cardiac Ito,f levels and gradients change notably with heart disease. Human cardiac Ito,f appears to be encoded by the Kv4.3 pore-forming α-subunit plus the auxiliary KChIP2 ß-subunit while mouse cardiac Ito,f requires Kv4.2 and Kv4.3 α-subunits plus KChIP2. Regional differences in cardiac Ito,f are associated with expression differences in Kv4.2 and KChIP2. Although Ito,f was reported to be absent in mouse ventricular cardiomyocytes lacking the Kv4.2 gene (Kv4.2-/-) when short depolarizing voltage pulses were used to activate voltage-gated K+ currents, in the present study, we showed that the use of long depolarization steps revealed a heteropodatoxin-sensitive Ito,f (at ~40% of the wild-type levels). Immunohistological studies further demonstrated membrane expression of Kv4.3 in Kv4.2-/- cardiomyocytes. Transmural Ito,f gradients across the left ventricular wall were reduced by ~3.5-fold in Kv4.2-/- heart, compared to wild-type. The Ito,f gradient in Kv4.2-/- hearts was associated with gradients in KChIP2 mRNA expression while in wild-type there was also a gradient in Kv4.2 expression. In conclusion, we found that Kv4.3-based Ito,f exists in the absence of Kv4.2, although with a reduced transmural gradient. Kv4.2-/- mice may be a useful animal model for studying Kv4.3-based Ito,f as observed in humans.


Subject(s)
Action Potentials/physiology , Cell Membrane/physiology , Myocytes, Cardiac/physiology , Shal Potassium Channels/physiology , Action Potentials/drug effects , Action Potentials/genetics , Animals , Cell Membrane/drug effects , Cell Membrane/metabolism , Cells, Cultured , Fluorescent Antibody Technique , Gene Expression , Kv Channel-Interacting Proteins/genetics , Kv Channel-Interacting Proteins/metabolism , Kv Channel-Interacting Proteins/physiology , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Patch-Clamp Techniques , Potassium Channel Blockers/pharmacology , Protein Subunits/genetics , Protein Subunits/metabolism , Protein Subunits/physiology , Reverse Transcriptase Polymerase Chain Reaction , Shal Potassium Channels/genetics , Shal Potassium Channels/metabolism , Spider Venoms/pharmacology
16.
Atherosclerosis ; 241(1): 111-20, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25974101

ABSTRACT

In vitro, insulin has mitogenic effects on vascular smooth muscle cells (VSMC) but also has protective effects on endothelial cells by stimulating nitric oxide (NO) production and endothelial nitric oxide synthase (eNOS) expression. Furthermore, NOS inhibition attenuates the effect of insulin to inhibit VSMC migration in vitro. Using an in vivo model, we have previously shown that insulin decreases neointimal growth and cell migration and increases re-endothelialization after arterial injury in normal rats. Since insulin can stimulate NOS, and NO can decrease neointimal growth, we hypothesized that NOS, and more specifically eNOS was required for the effects of insulin in vivo. Rats were given subcutaneous insulin implants (3 U/day) alone or with the NOS inhibitor l-NAME (2 mg kg(-1) day(-1)) 3 days before arterial (carotid or aortic) balloon catheter injury. Insulin decreased both neointimal area (P < 0.01) and cell migration (P < 0.01), and increased re-endothelialization (P < 0.05). All of these effects were prevented by the co-administration of l-NAME. Insulin was found to decrease inducible NOS expression (P < 0.05) but increase eNOS phosphorylation (P < 0.05). These changes were also translated at the functional level where insulin improved endothelial-dependent vasorelaxation. To further study the NOS isoform involved in insulin action, s.c. insulin (0.1 U/day) was given to wild-type and eNOS knockout mice. We found that insulin was effective at decreasing neointimal formation in wild-type mice after wire injury of the femoral artery, whereas this effect of insulin was absent in eNOS knockout mice. These results show that the vasculoprotective effect of insulin after arterial injury is mediated by an eNOS-dependent mechanism.


Subject(s)
Carotid Artery Injuries/drug therapy , Insulin/administration & dosage , Neointima , Nitric Oxide Synthase Type III/metabolism , Vascular System Injuries/drug therapy , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/enzymology , Aorta, Thoracic/injuries , Aorta, Thoracic/pathology , Carotid Artery Injuries/enzymology , Carotid Artery Injuries/pathology , Carotid Artery Injuries/physiopathology , Carotid Artery, Common/drug effects , Carotid Artery, Common/enzymology , Carotid Artery, Common/pathology , Cell Movement/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Implants , Enzyme Inhibitors/pharmacology , Femoral Artery/drug effects , Femoral Artery/enzymology , Femoral Artery/injuries , Femoral Artery/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type III/antagonists & inhibitors , Nitric Oxide Synthase Type III/deficiency , Nitric Oxide Synthase Type III/genetics , Phosphorylation , Rats, Sprague-Dawley , Re-Epithelialization/drug effects , Signal Transduction/drug effects , Time Factors , Vascular System Injuries/enzymology , Vascular System Injuries/pathology , Vascular System Injuries/physiopathology , Vasodilation/drug effects , Vasodilator Agents/pharmacology
17.
J Recept Signal Transduct Res ; 34(6): 476-83, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24846582

ABSTRACT

CONTEXT: Regulator of G-protein signaling-2 (RGS2) inhibits Gq-mediated regulation of Ca(2+) signalling in vascular smooth muscle cells (VSMC). OBJECTIVE: RGS2 knockout (RGS2KO) mice are hypertensive and show arteriolar remodeling. VSMC proliferation modulates intracellular Ca(2+) concentration [Ca(2+)]i. RGS2 involvement in VSMC proliferation had not been examined. METHODS: Thymidine incorporation and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) conversion assays measured cell proliferation. Fura-2 ratiometric imaging quantified [Ca(2+)]i before and after UTP and thapsigargin. [(3)H]-labeled inositol was used for phosphoinositide hydrolysis. Quantitative RT-PCR and confocal immunofluorescence of select Ca(2+) transporters was performed in primary aortic VSMC. RESULTS AND DISCUSSION: Platelet-derived growth factor (PDGF) increased S-phase entry and proliferation in VSMC from RGS2KO mice to a greater extent than in VSMC from wild-type (WT) controls. Consistent with differential PDGF-induced changes in Ca(2+) homeostasis, RGS2KO VSMC showed lower resting [Ca(2+)]i but higher thapsigargin-induced [Ca(2+)]i as compared with WT. RGS2KO VSMC expressed lower mRNA levels of plasma membrane Ca(2+) ATPase-4 (PMCA4) and Na(+) Ca(2+) Exchanger (NCX), but higher levels of sarco-endoplasmic reticulum Ca(2+) ATPase-2 (SERCA2). Western blot and immunofluorescence revealed similar differences in PMCA4 and SERCA2 protein, while levels of NCX protein were not reduced in RGS2KO VSMC. Consistent with decreased Ca(2+) efflux activity, (45)Ca-extrusion rates were lower in RGS2KO VSMC. These differences were reversed by the PMCA inhibitor La(3+), but not by replacing extracellular Na(+) with choline, implicating differences in the activity of PMCA and not NCX. CONCLUSION: RGS2-deficient VSMC exhibit higher rates of proliferation and coordinate plasticity of Ca(2+)-handling mechanisms in response to PDGF stimulation.


Subject(s)
Calcium Signaling/physiology , Calcium/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/physiology , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/physiology , RGS Proteins/metabolism , Animals , Calcium Signaling/drug effects , Cell Proliferation/drug effects , Cell Proliferation/physiology , Cells, Cultured , Mice , Mice, Knockout , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Platelet-Derived Growth Factor/pharmacology , RGS Proteins/genetics
18.
Biol Psychiatry ; 76(4): 315-23, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24315552

ABSTRACT

BACKGROUND: The experience of early stress contributes to the etiology of several psychiatric disorders and can lead to lasting deficits in working memory and attention. These executive functions require activation of the prefrontal cortex (PFC) by muscarinic M1 acetylcholine (ACh) receptors. Such Gαq-protein coupled receptors trigger the release of calcium (Ca(2+)) from internal stores and elicit prolonged neuronal excitation. METHODS: In brain slices of rat PFC, we employed multiphoton imaging simultaneously with whole-cell electrophysiological recordings to examine potential interactions between ACh-induced Ca(2+) release and excitatory currents in adulthood, across postnatal development, and following the early stress of repeated maternal separation, a rodent model for depression. We also investigated developmental changes in related genes in these groups. RESULTS: Acetylcholine-induced Ca(2+) release potentiates ACh-elicited excitatory currents. In the healthy PFC, this potentiation of muscarinic excitation emerges in young adulthood, when executive function typically reaches maturity. However, the developmental consolidation of muscarinic ACh signaling is abolished in adults with a history of early stress, where ACh responses retain an adolescent phenotype. In prefrontal cortex, these rats show a disruption in the expression of multiple developmentally regulated genes associated with Gαq and Ca(2+) signaling. Pharmacologic and ionic manipulations reveal that the enhancement of muscarinic excitation in the healthy adult PFC arises via the electrogenic process of sodium/Ca(2+) exchange. CONCLUSIONS: This work illustrates a long-lasting disruption in ACh-mediated cortical excitation following early stress and raises the possibility that such cellular mechanisms may disrupt the maturation of executive function.


Subject(s)
Acetylcholine/metabolism , Calcium/metabolism , Maternal Deprivation , Prefrontal Cortex/growth & development , Prefrontal Cortex/physiopathology , Stress, Psychological/physiopathology , Animals , Depression , Disease Models, Animal , Gene Expression Regulation, Developmental/physiology , Male , Patch-Clamp Techniques , Prefrontal Cortex/drug effects , Pyramidal Cells/drug effects , Pyramidal Cells/growth & development , Pyramidal Cells/physiopathology , Random Allocation , Rats, Sprague-Dawley , Tissue Culture Techniques
19.
J Biol Chem ; 288(30): 21836-49, 2013 Jul 26.
Article in English | MEDLINE | ID: mdl-23733193

ABSTRACT

RGS4, a heterotrimeric G-protein inhibitor, localizes to plasma membrane (PM) and endosomal compartments. Here, we examined Rab-mediated control of RGS4 internalization and recycling. Wild type and constitutively active Rab5 decreased RGS4 PM levels while increasing its endosomal targeting. Rab5, however, did not appreciably affect the PM localization or function of the M1 muscarinic receptor (M1R)/Gq signaling cascade. RGS4-containing endosomes co-localized with subsets of Rab5-, transferrin receptor-, and Lamp1/Lysotracker-marked compartments suggesting RGS4 traffics through PM recycling or acidified endosome pathways. Rab7 activity promoted TGN association, whereas Rab7(dominant negative) trapped RGS4 in late endosomes. Furthermore, RGS4 was found to co-localize with an endosomal pool marked by Rab11, the protein that mediates recycling/sorting of proteins to the PM. The Cys-12 residue in RGS4 appeared important for its Rab11-mediated trafficking to the PM. Rab11(dominant negative) decreased RGS4 PM levels and increased the number of RGS4-containing endosomes. Inhibition of Rab11 activity decreased RGS4 function as an inhibitor of M1R activity without affecting localization and function of the M1R/Gq signaling complex. Thus, both Rab5 activation and Rab11 inhibition decreased RGS4 function in a manner that is independent from their effects on the localization and function of the M1R/Gq signaling complex. This is the first study to implicate Rab GTPases in the intracellular trafficking of an RGS protein. Thus, Rab GTPases may be novel molecular targets for the selective regulation of M1R-mediated signaling via their specific effects on RGS4 trafficking and function.


Subject(s)
Endosomes/metabolism , RGS Proteins/metabolism , rab GTP-Binding Proteins/metabolism , rab5 GTP-Binding Proteins/metabolism , Blotting, Western , Cell Membrane/metabolism , HEK293 Cells , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Microscopy, Confocal , Mutation , Protein Transport , RGS Proteins/genetics , Receptor, Muscarinic M1/metabolism , Signal Transduction , rab GTP-Binding Proteins/genetics , rab5 GTP-Binding Proteins/genetics , rab7 GTP-Binding Proteins
20.
Structure ; 21(3): 319-20, 2013 Mar 05.
Article in English | MEDLINE | ID: mdl-23473665

ABSTRACT

G protein-coupled receptors mediate a wide array of physiologic stimuli and, together with their regulators such as RGS2, are essential components of cellular signaling and function. RGS2 is a selective inhibitor of the Gαq class of α subunits. In this issue of Structure, Nance and colleagues provide structural insight into the features of RGS2 that mediate its potent and selective regulation of Gαq function.

SELECTION OF CITATIONS
SEARCH DETAIL
...