Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Circ Res ; 127(6): 796-810, 2020 08 28.
Article in English | MEDLINE | ID: mdl-32507058

ABSTRACT

RATIONALE: Cardiotoxic ß1 adrenergic receptor (ß1AR)-CaMKII (calmodulin-dependent kinase II) signaling is a major and critical feature associated with development of heart failure. SAP97 (synapse-associated protein 97) is a multifunctional scaffold protein that binds directly to the C-terminus of ß1AR and organizes a receptor signalosome. OBJECTIVE: We aim to elucidate the dynamics of ß1AR-SAP97 signalosome and its potential role in chronic cardiotoxic ß1AR-CaMKII signaling that contributes to development of heart failure. METHODS AND RESULTS: The integrity of cardiac ß1AR-SAP97 complex was examined in heart failure. Cardiac-specific deletion of SAP97 was developed to examine ß1AR signaling in aging mice, after chronic adrenergic stimulation, and in pressure overload hypertrophic heart failure. We show that the ß1AR-SAP97 signaling complex is reduced in heart failure. Cardiac-specific deletion of SAP97 yields an aging-dependent cardiomyopathy and exacerbates cardiac dysfunction induced by chronic adrenergic stimulation and pressure overload, which are associated with elevated CaMKII activity. Loss of SAP97 promotes PKA (protein kinase A)-dependent association of ß1AR with arrestin2 and CaMKII and turns on an Epac (exchange protein directly activated by cAMP)-dependent activation of CaMKII, which drives detrimental functional and structural remodeling in myocardium. Moreover, we have identified that GRK5 (G-protein receptor kinase-5) is necessary to promote agonist-induced dissociation of SAP97 from ß1AR. Cardiac deletion of GRK5 prevents adrenergic-induced dissociation of ß1AR-SAP97 complex and increases in CaMKII activity in hearts. CONCLUSIONS: These data reveal a critical role of SAP97 in maintaining the integrity of cardiac ß1AR signaling and a detrimental cardiac GRK5-CaMKII axis that can be potentially targeted in heart failure therapy. Graphical Abstract: A graphical abstract is available for this article.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Discs Large Homolog 1 Protein/metabolism , G-Protein-Coupled Receptor Kinase 5/metabolism , Heart Failure/enzymology , Myocytes, Cardiac/enzymology , Receptors, Adrenergic, beta-1/metabolism , Animals , Apoptosis , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/metabolism , Discs Large Homolog 1 Protein/genetics , Disease Models, Animal , Excitation Contraction Coupling , G-Protein-Coupled Receptor Kinase 5/genetics , Guanine Nucleotide Exchange Factors/metabolism , Heart Failure/genetics , Heart Failure/pathology , Heart Failure/physiopathology , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Myocardial Contraction , Myocytes, Cardiac/pathology , beta-Arrestin 1/metabolism
2.
Arterioscler Thromb Vasc Biol ; 38(10): 2371-2381, 2018 10.
Article in English | MEDLINE | ID: mdl-30354206

ABSTRACT

Objective- This study examined whether caveolae position CaV3.2 (T-type Ca2+ channel encoded by the α-3.2 subunit) sufficiently close to RyR (ryanodine receptors) for extracellular Ca2+ influx to trigger Ca2+ sparks and large-conductance Ca2+-activated K+ channel feedback. Approach and Results- Using smooth muscle cells from mouse mesenteric arteries, the proximity ligation assay confirmed that CaV3.2 reside within 40 nm of caveolin 1, a key caveolae protein. Methyl-ß-cyclodextrin, a cholesterol depleting agent that disrupts caveolae, suppressed CaV3.2 activity along with large-conductance Ca2+-activated K+-mediated spontaneous transient outward currents in cells from C57BL/6 but not CaV3.2-/- mice. Genetic deletion of caveolin 1, a perturbation that prevents caveolae formation, also impaired spontaneous transient outward current production but did so without impairing Ca2+ channel activity, including CaV3.2. These observations indicate a mistargeting of CaV3.2 in caveolin 1-/- mice, a view supported by a loss of Ni2+-sensitive Ca2+ spark generation and colocalization signal (CaV3.2-RyR) from the proximity ligation assay. Vasomotor and membrane potential measurements confirmed that cellular disruption of the CaV3.2-RyR axis functionally impaired the ability of large-conductance Ca2+-activated K+ to set tone in pressurized caveolin 1-/- arteries. Conclusions- Caveolae play a critical role in protein targeting and preserving the close structural relationship between CaV3.2 and RyR needed to drive negative feedback control in resistance arteries.


Subject(s)
Calcium Channels, T-Type/metabolism , Calcium Signaling , Caveolae/metabolism , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Animals , Calcium Channels, T-Type/deficiency , Calcium Channels, T-Type/genetics , Caveolin 1/genetics , Caveolin 1/metabolism , Feedback, Physiological , Male , Membrane Potentials , Mesenteric Arteries/metabolism , Mice, Inbred C57BL , Mice, Knockout , Ryanodine Receptor Calcium Release Channel/metabolism , Vasoconstriction , Vasodilation
3.
Am J Physiol Regul Integr Comp Physiol ; 314(3): R433-R446, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29167165

ABSTRACT

Long-term hypoxia (LTH) has a profound effect on pulmonary arterial vasoconstriction in the fetus and adult. Dysregulation in Ca2+ signaling is important during the development of LTH-induced pulmonary hypertension. In the present study, we tested the hypothesis that L-type Ca2+ channels (CaL), which are voltage dependent and found in smooth, skeletal, and cardiac muscle, are important in the adaptation of pulmonary arterial contractions in postnatal maturation and in response to LTH. Pulmonary arteries were isolated from fetal or adult sheep maintained at low or high altitude (3,801 m) for >100 days. The effects were measured using an L-type Ca2+ channel opener FPL 64176 (FPL) in the presence or absence of an inhibitor, Nifedipine (NIF) on arterial contractions, intracellular Ca2+ oscillations, and ryanodine receptor-driven Ca2+ sparks. FPL induced pulmonary arterial contractions in all groups were sensitive to NIF. However, when compared with 125 mM K+, FPL contractions were greater in fetuses than in adults. FPL reduced Ca2+ oscillations in myocytes of adult but not fetal arteries, independently of altitude. The FPL effects on Ca2+ oscillations were reversed by NIF in myocytes of hypoxic but not normoxic adults. FPL failed to enhance Ca2+ spark frequency and had little impact on spatiotemporal firing characteristics. These data suggest that CaL-dependent contractions are largely uncoupled from intracellular Ca2+ oscillations and the development of Ca2+ sparks. This raises questions regarding the coupling of pulmonary arterial contractility to membrane depolarization, attendant CaL facilitation, and the related associations with the activation of Ca2+ oscillations and Ca2+ sparks.


Subject(s)
Altitude Sickness/metabolism , Calcium Channels, L-Type/metabolism , Calcium Signaling , Calcium/metabolism , Pulmonary Artery/metabolism , Vasoconstriction , Altitude Sickness/physiopathology , Animals , Calcium Channel Agonists/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/drug effects , Calcium Signaling/drug effects , Disease Models, Animal , Female , Fetal Heart/metabolism , Fetal Heart/physiopathology , Gestational Age , Membrane Potentials , Myocytes, Cardiac/metabolism , Pregnancy , Pulmonary Artery/drug effects , Pulmonary Artery/embryology , Pulmonary Artery/physiopathology , Sheep, Domestic , Time Factors , Vasoconstriction/drug effects
4.
J Physiol ; 595(4): 1111-1126, 2017 02 15.
Article in English | MEDLINE | ID: mdl-27805790

ABSTRACT

KEY POINTS: Distinct Ca2+ channels work in a coordinated manner to grade Ca2+ spark/spontaneous transient outward currents (STOCs) in rat cerebral arteries. The relative contribution of each Ca2+ channel to Ca2+ spark/STOC production depends upon their biophysical properties and the resting membrane potential of smooth muscle. Na+ /Ca2+ exchanger, but not TRP channels, can also facilitate STOC production. ABSTRACT: Ca2+ sparks are generated in a voltage-dependent manner to initiate spontaneous transient outward currents (STOCs), events that moderate arterial constriction. In this study, we defined the mechanisms by which membrane depolarization increases Ca2+ sparks and subsequent STOC production. Using perforated patch clamp electrophysiology and rat cerebral arterial myocytes, we monitored STOCs in the presence and absence of agents that modulate Ca2+ entry. Beginning with CaV 3.2 channel inhibition, Ni2+ was shown to decrease STOC frequency in cells held at hyperpolarized (-40 mV) but not depolarized (-20 mV) voltages. In contrast, nifedipine, a CaV 1.2 inhibitor, markedly suppressed STOC frequency at -20 mV but not -40 mV. These findings aligned with the voltage-dependent profiles of L- and T-type Ca2+ channels. Furthermore, computational and experimental observations illustrated that Ca2+ spark production is intimately tied to the activity of both conductances. Intriguingly, this study observed residual STOC production at depolarized voltages that was independent of CaV 1.2 and CaV 3.2. This residual component was insensitive to TRPV4 channel modulation and was abolished by Na+ /Ca2+ exchanger blockade. In summary, our work highlights that the voltage-dependent triggering of Ca2+ sparks/STOCs is not tied to a single conductance but rather reflects an interplay among multiple Ca2+ permeable pores with distinct electrophysiological properties. This integrated orchestration enables smooth muscle to grade Ca2+ spark/STOC production and thus precisely tune negative electrical feedback.


Subject(s)
Calcium Signaling , Cerebral Arteries/metabolism , Myocytes, Smooth Muscle/metabolism , Animals , Calcium Channels/metabolism , Cells, Cultured , Cerebral Arteries/cytology , Cerebral Arteries/physiology , Female , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Rats , Rats, Sprague-Dawley , Sodium-Calcium Exchanger/metabolism , TRPV Cation Channels/metabolism
5.
J Mol Cell Cardiol ; 98: 62-72, 2016 09.
Article in English | MEDLINE | ID: mdl-27318036

ABSTRACT

Diastolic calcium (Ca) leak via cardiac ryanodine receptors (RyR2) can cause arrhythmias and heart failure (HF). Ca/calmodulin (CaM)-dependent kinase II (CaMKII) is upregulated and more active in HF, promoting RyR2-mediated Ca leak by RyR2-Ser2814 phosphorylation. Here, we tested a mechanistic hypothesis that RyR2 phosphorylation by CaMKII increases Ca leak by promoting a pathological RyR2 conformation with reduced CaM affinity. Acute CaMKII activation in wild-type RyR2, and phosphomimetic RyR2-S2814D (vs. non-phosphorylatable RyR2-S2814A) knock-in mouse myocytes increased SR Ca leak, reduced CaM-RyR2 affinity, and caused a pathological shift in RyR2 conformation (detected via increased access of the RyR2 structural peptide DPc10). This same trio of effects was seen in myocytes from rabbits with pressure/volume-overload induced HF. Excess CaM quieted leak and restored control conformation, consistent with negative allosteric coupling between CaM affinity and DPc10 accessible conformation. Dantrolene (DAN) also restored CaM affinity, reduced DPc10 access, and suppressed RyR2-mediated Ca leak and ventricular tachycardia in RyR2-S2814D mice. We propose that a common pathological RyR2 conformational state (low CaM affinity, high DPc10 access, and elevated leak) may be caused by CaMKII-dependent phosphorylation, oxidation, and HF. Moreover, DAN (or excess CaM) can shift this pathological gating state back to the normal physiological conformation, a potentially important therapeutic approach.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Protein Conformation , Ryanodine Receptor Calcium Release Channel/chemistry , Ryanodine Receptor Calcium Release Channel/metabolism , Animals , Calcium/metabolism , Calcium Signaling , Calmodulin/metabolism , Dantrolene/pharmacology , Disease Models, Animal , Heart Failure/metabolism , Heart Failure/pathology , Ion Channel Gating , Mice , Myocytes, Cardiac/metabolism , Permeability , Phosphorylation , Protein Binding , Protein Conformation/drug effects , Rabbits , Sarcoplasmic Reticulum/metabolism , Tachycardia, Ventricular/drug therapy , Tachycardia, Ventricular/etiology , Tachycardia, Ventricular/metabolism , Tachycardia, Ventricular/physiopathology , Tacrolimus Binding Proteins/metabolism
6.
Circ Res ; 114(4): 607-15, 2014 Feb 14.
Article in English | MEDLINE | ID: mdl-24323672

ABSTRACT

RATIONALE: Increased contractility of arterial myocytes and enhanced vascular tone during hyperglycemia and diabetes mellitus may arise from impaired large-conductance Ca(2+)-activated K(+) (BKCa) channel function. The scaffolding protein A-kinase anchoring protein 150 (AKAP150) is a key regulator of calcineurin (CaN), a phosphatase known to modulate the expression of the regulatory BKCa ß1 subunit. Whether AKAP150 mediates BKCa channel suppression during hyperglycemia and diabetes mellitus is unknown. OBJECTIVE: To test the hypothesis that AKAP150-dependent CaN signaling mediates BKCa ß1 downregulation and impaired vascular BKCa channel function during hyperglycemia and diabetes mellitus. METHODS AND RESULTS: We found that AKAP150 is an important determinant of BKCa channel remodeling, CaN/nuclear factor of activated T-cells c3 (NFATc3) activation, and resistance artery constriction in hyperglycemic animals on high-fat diet. Genetic ablation of AKAP150 protected against these alterations, including augmented vasoconstriction. d-glucose-dependent suppression of BKCa channel ß1 subunits required Ca(2+) influx via voltage-gated L-type Ca(2+) channels and mobilization of a CaN/NFATc3 signaling pathway. Remarkably, high-fat diet mice expressing a mutant AKAP150 unable to anchor CaN resisted activation of NFATc3 and downregulation of BKCa ß1 subunits and attenuated high-fat diet-induced elevation in arterial blood pressure. CONCLUSIONS: Our results support a model whereby subcellular anchoring of CaN by AKAP150 is a key molecular determinant of vascular BKCa channel remodeling, which contributes to vasoconstriction during diabetes mellitus.


Subject(s)
A Kinase Anchor Proteins/metabolism , Diabetes Mellitus, Experimental/metabolism , Hyperglycemia/metabolism , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/metabolism , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Vasoconstriction/physiology , A Kinase Anchor Proteins/genetics , Animals , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/physiopathology , Dietary Fats/pharmacology , Gene Knock-In Techniques , Hyperglycemia/genetics , Hyperglycemia/physiopathology , Hypertension/genetics , Hypertension/metabolism , Hypertension/physiopathology , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/genetics , Large-Conductance Calcium-Activated Potassium Channels/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Mutant Strains , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/physiology , NFATC Transcription Factors/metabolism , Peptides/pharmacology , Signal Transduction/physiology , Toxins, Biological/pharmacology , Vasoconstriction/drug effects
7.
Arterioscler Thromb Vasc Biol ; 35(8): 1843-51, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26069238

ABSTRACT

OBJECTIVE: In resistance arteries, there is an emerging view that smooth muscle CaV3.2 channels restrain arterial constriction through a feedback response involving the large-conductance Ca(2+)-activated K(+) channel (BKCa). Here, we used wild-type and CaV3.2 knockout (CaV3.2(-/-)) mice to definitively test whether CaV3.2 moderates myogenic tone in mesenteric arteries via the CaV3.2-ryanodine receptor-BKCa axis and whether this regulatory mechanism influences blood pressure regulation. APPROACH AND RESULTS: Using pressurized vessel myography, CaV3.2(-/-) mesenteric arteries displayed enhanced myogenic constriction to pressure but similar K(+)-induced vasoconstriction compared with wild-type C57BL/6 arteries. Electrophysiological and myography experiments subsequently confirmed the inability of micromolar Ni(2+), a CaV3.2 blocker, to either constrict arteries or suppress T-type currents in CaV3.2(-/-) smooth muscle cells. The frequency of BKCa-induced spontaneous transient outward K(+) currents dropped in wild-type but not in knockout arterial smooth muscle cells upon the pharmacological suppression of CaV3.2 channel. Line scan analysis performed on en face arteries loaded with Fluo-4 revealed the presence of Ca(2+) sparks in all arteries, with the subsequent application of Ni(2+) only affecting wild-type arteries. Although CaV3.2 channel moderated myogenic constriction of resistance arteries, the blood pressure measurements of CaV3.2(-/-) and wild-type animals were similar. CONCLUSIONS: Overall, our findings establish a negative feedback mechanism of the myogenic response in which CaV3.2 channel modulates downstream ryanodine receptor-BKCa to hyperpolarize and relax arteries.


Subject(s)
Calcium Channels, T-Type/deficiency , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Muscle, Smooth, Vascular/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Vasoconstriction , Vasodilation , Animals , Arterial Pressure , Calcium Channel Blockers/pharmacology , Calcium Channels, T-Type/drug effects , Calcium Channels, T-Type/genetics , Calcium Signaling , Feedback, Physiological , Female , Male , Mesenteric Arteries/metabolism , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/metabolism , Vasoconstriction/drug effects , Vasodilation/drug effects
8.
J Mol Cell Cardiol ; 81: 162-75, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25724724

ABSTRACT

A five-state model of myofilament contraction was integrated into a well-established rabbit ventricular myocyte model of ion channels, Ca(2+) transporters and kinase signaling to analyze the relative contribution of different phosphorylation targets to the overall mechanical response driven by ß-adrenergic stimulation (ß-AS). ß-AS effect on sarcoplasmic reticulum Ca(2+) handling, Ca(2+), K(+) and Cl(-) currents, and Na(+)/K(+)-ATPase properties was included based on experimental data. The inotropic effect on the myofilaments was represented as reduced myofilament Ca(2+) sensitivity (XBCa) and titin stiffness, and increased cross-bridge (XB) cycling rate (XBcy). Assuming independent roles of XBCa and XBcy, the model reproduced experimental ß-AS responses on action potentials and Ca(2+) transient amplitude and kinetics. It also replicated the behavior of force-Ca(2+), release-restretch, length-step, stiffness-frequency and force-velocity relationships, and increased force and shortening in isometric and isotonic twitch contractions. The ß-AS effect was then switched off from individual targets to analyze their relative impact on contractility. Preventing ß-AS effects on L-type Ca(2+) channels or phospholamban limited Ca(2+) transients and contractile responses in parallel, while blocking phospholemman and K(+) channel (IKs) effects enhanced Ca(2+) and inotropy. Removal of ß-AS effects from XBCa enhanced contractile force while decreasing peak Ca(2+) (due to greater Ca(2+) buffering), but had less effect on shortening. Conversely, preventing ß-AS effects on XBcy preserved Ca(2+) transient effects, but blunted inotropy (both isometric force and especially shortening). Removal of titin effects had little impact on contraction. Finally, exclusion of ß-AS from XBCa and XBcy while preserving effects on other targets resulted in preserved peak isometric force response (with slower kinetics) but nearly abolished enhanced shortening. ß-AS effects on XBCa and XBcy have greater impact on isometric and isotonic contraction, respectively.


Subject(s)
Adrenergic Agents/pharmacology , Calcium/metabolism , Isoproterenol/pharmacology , Models, Cardiovascular , Myocardial Contraction/drug effects , Myofibrils/drug effects , Propranolol/pharmacology , Action Potentials/drug effects , Action Potentials/physiology , Animals , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Computer Simulation , Connectin/genetics , Connectin/metabolism , Gene Expression Regulation , Heart Ventricles/cytology , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Myocardial Contraction/physiology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myofibrils/physiology , Rabbits , Sarcoplasmic Reticulum/drug effects , Sarcoplasmic Reticulum/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Signal Transduction , Sodium-Potassium-Exchanging ATPase/genetics , Sodium-Potassium-Exchanging ATPase/metabolism , Software
9.
Arch Biochem Biophys ; 552-553: 50-9, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24480308

ABSTRACT

Chronic increases in myofilament Ca(2+)-sensitivity in the heart are known to alter gene expression potentially modifying Ca(2+)-homeostasis and inducing arrhythmias. We tested age-dependent effects of a chronic increase in myofilament Ca(2+)-sensitivity on induction of altered alter gene expression and activity of Ca(2+) transport systems in cardiac myocytes. Our approach was to determine the relative contributions of the major mechanisms responsible for restoring Ca(2+) to basal levels in field stimulated ventricular myocytes. Comparisons were made from ventricular myocytes isolated from non-transgenic (NTG) controls and transgenic mice expressing the fetal, slow skeletal troponin I (TG-ssTnI) in place of cardiac TnI (cTnI). Replacement of cTnI by ssTnI induces an increase in myofilament Ca(2+)-sensitivity. Comparisons included myocytes from relatively young (5-7months) and older mice (11-13months). Employing application of caffeine in normal Tyrode and in 0Na(+) 0Ca(2+) solution, we were able to dissect the contribution of the sarcoplasmic reticulum Ca(2+) pump (SR Ca(2+)-ATPase), the Na(+)/Ca(2+) exchanger (NCX), and "slow mechanisms" representing the activity of the sarcolemmal Ca(2+) pump and the mitochondrial Ca(2+) uniporter. The relative contribution of the SR Ca(2+)-ATPase to restoration of basal Ca(2+) levels in younger TG-ssTnI myocytes was lower than in NTG (81.12±2.8% vs 92.70±1.02%), but the same in the older myocytes. Younger and older NTG myocytes demonstrated similar contributions from the SR Ca(2+)-ATPase and NCX to restoration of basal Ca(2+). However, the slow mechanisms for Ca(2+) removal were increased in the older NTG (3.4±0.3%) vs the younger NTG myocytes (1.4±0.1%). Compared to NTG, younger TG-ssTnI myocytes demonstrated a significantly bigger contribution of the NCX (16±2.7% in TG vs 6.9±0.9% in NTG) and slow mechanisms (3.3±0.4% in TG vs 1.4±0.1% in NTG). In older TG-ssTnI myocytes the contributions were not significantly different from NTG (NCX: 4.9±0.6% in TG vs 5.5±0.7% in NTG; slow mechanisms: 2.5±0.3% in TG vs 3.4±0.3% in NTG). Our data indicate that constitutive increases in myofilament Ca(2+)-sensitivity alter the relative significance of the NCX transport system involved in Ca(2+)-homeostasis only in a younger group of mice. This modification may be of significance in early changes in altered gene expression and electrical stability hearts with increased myofilament Ca-sensitivity.


Subject(s)
Calcium Signaling , Calcium/metabolism , Myocytes, Cardiac/cytology , Myofibrils/metabolism , Animals , Caffeine/pharmacology , Calcium Signaling/drug effects , Cells, Cultured , Central Nervous System Stimulants/pharmacology , Female , Gene Expression , Male , Mice , Mice, Transgenic , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Troponin I/genetics , Troponin I/metabolism
10.
J Womens Health (Larchmt) ; 33(4): 453-466, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38112561

ABSTRACT

Purpose: Pregnant women are vulnerable to Coronavirus Disease 2019 (COVID-19) complications, yet may hesitate to get vaccinated. It is important to identify racial/ethnic and other individual characteristics associated with COVID-19 vaccine acceptance in the United States during pregnancy. Methods: We searched PubMed, Embase, and Web of Science for articles published through January 2023 for keywords/terms related to immunization, COVID-19, and pregnancy, and performed a systematic review and meta-analysis to examine characteristics associated with vaccine acceptance. Results: Of 1,592 articles, 23 met inclusion criteria (focused on pregnant women in the United States, and their willingness or hesitation to vaccinate). Twenty-two of the studies examined receipt of ≥1 COVID-19 vaccine dose and/or intention to vaccinate, while one examined vaccine hesitancy. Vaccine acceptance rates ranged from 7% to 78.3%. Meta-analyses demonstrated that compared with Whites, Hispanics (odds ratios [OR] 0.72; 95% confidence interval [CI] 0.58-0.91) and Blacks (OR 0.44; 95% CI 0.30-0.63) had less COVID-19 vaccine acceptance, while Asians (OR 1.78; 95% CI 1.10-2.88) had greater vaccine acceptance. College graduation or more (OR 3.25; 95% CI 2.53-4.17), receipt or intention to receive the influenza vaccine (OR 3.46; 95% CI 2.22-5.41), and at least part-time employment (OR 2.12; 95% CI 1.66-2.72) were significantly associated with vaccine acceptance. Conclusions: COVID-19 vaccine nonacceptance in pregnant women is associated with Hispanic ethnicity and Black race, while acceptance is associated with Asian race, college education or more, at least part-time employment, and acceptance of the influenza vaccine. Future COVID-19 vaccination campaigns can target identified subgroups of pregnant women who are less likely to accept vaccination.


Subject(s)
COVID-19 Vaccines , COVID-19 , Pregnant Women , Vaccination Hesitancy , Humans , Pregnancy , Female , COVID-19 Vaccines/administration & dosage , United States , COVID-19/prevention & control , Pregnant Women/psychology , Pregnant Women/ethnology , Vaccination Hesitancy/statistics & numerical data , Vaccination Hesitancy/psychology , Vaccination/statistics & numerical data , Vaccination/psychology , SARS-CoV-2 , Patient Acceptance of Health Care/ethnology , Patient Acceptance of Health Care/statistics & numerical data , Patient Acceptance of Health Care/psychology , Pregnancy Complications, Infectious/prevention & control , Adult
11.
J Mol Cell Cardiol ; 58: 153-61, 2013 May.
Article in English | MEDLINE | ID: mdl-23481579

ABSTRACT

Sympathetic stimulation regulates cardiac excitation-contraction coupling in hearts but can also trigger ventricular arrhythmias caused by early afterdepolarizations (EADs) in pathological conditions. Isoproterenol (ISO) stimulation can transiently cause EADs which could result from differential kinetics of L-type Ca current (ICaL) vs. delayed rectifier potassium current (IKs) effects, but multiple PKA targets complicate mechanistic analysis. Utilizing a biophysically detailed model integrating Ca and ß-adrenergic signaling, we investigate how different phosphorylation kinetics and targets influence ß-adrenergic-induced transient EADs. We found that: 1) The faster time course of ICaL vs. IKs increases recapitulates experimentally observed ISO-induced transient EADs (which are due to ICaL reactivation). These EADs disappear at steady state ISO and do not occur during more gradual ISO application. 2) This ICaL vs. IKs kinetic mismatch with ISO can also induce transient EADs due to spontaneous sarcoplasmic reticulum (SR) Ca release and Na/Ca exchange current. The increased ICaL, SR Ca uptake and action potential duration (APD) raise SR Ca to cause spontaneous SR Ca release, but eventual IKs activation and APD shortening abolish these EADs. 3) Phospholemman (PLM) phosphorylation decreases both types of EADs by increasing outward Na/K-ATPase current (INaK) for ICaL-mediated EADs, and reducing intracellular Na and Ca loading for SR Ca-release-mediated EADs. Slowing PLM phosphorylation kinetics abolishes this protective effect. 4) Blocking phospholamban (PLB) phosphorylation has little effect on ICaL-mediated transient EADs, but abolishes SR Ca-release-mediated transient EADs by limiting SR Ca loading. 5) RyR phosphorylation has little effect on either transient EAD type. Our study emphasizes the importance of understanding non-steady state kinetics of several systems in mediating ß-adrenergic-induced EADs and arrhythmias.


Subject(s)
Arrhythmias, Cardiac/metabolism , Calcium Channels, L-Type/metabolism , Calcium/metabolism , Receptors, Adrenergic, beta/metabolism , Animals , Arrhythmias, Cardiac/pathology , Calcium Signaling/drug effects , Excitation Contraction Coupling/physiology , Humans , Isoproterenol/administration & dosage , Kinetics , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Phosphorylation/drug effects , Rabbits
12.
Adv Physiol Educ ; 37(1): 28-34, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23471245

ABSTRACT

The force-frequency relationship has intrigued researchers since its discovery by Bowditch in 1871. Many attempts have been made to construct mathematical descriptions of this phenomenon, beginning with the simple formulation of Koch-Wesser and Blinks in 1963 to the most sophisticated ones of today. This property of cardiac muscle is amplified by ß-adrenergic stimulation, and, in a coordinated way, the neurohumoral state alters both frequency (acting on the sinoatrial node) as well as force generation (modifying ventricular myocytes). This synchronized tuning is needed to meet new metabolic demands. Cardiac modelers have already linked mechanical and electrical activity in their formulations and showed how those activities feedback on each other. However, now it is necessary to include neurological control to have a complete description of heart performance, especially when changes in frequency are involved. Study of arrhythmias (or antiarrhythmic drugs) based on mathematical models should incorporate this effect to make useful predictions or point out potential pharmaceutical targets.


Subject(s)
Heart Rate/physiology , Models, Cardiovascular , Animals , Exercise/physiology , Humans
13.
AJPM Focus ; 2(3): 100112, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37790675

ABSTRACT

Introduction: Ensuring that people at risk of overdosing on opioids have easy access to naloxone is an essential part of the fight against the opioid crisis. This study evaluates the impact of the 2016 California law (CA AB1535) permitting pharmacies to dispense this life-saving medication without a physician's prescription. Methods: California counties were categorized on the basis of population density (rural, suburban, urban), rate of opioid-related deaths by population density (high, medium, low), and rate of opioid prescriptions by population density (high, medium, low). Ten diverse pharmacies from each category were selected for inclusion. In a brief 1-minute interview conducted between July and August 2021, pharmacists from 146 California pharmacies were surveyed regarding their knowledge of CA AB1535, their practice of dispensing naloxone without a physician's prescription, and whether they normally stock naloxone. Chi-square tests were used to compare responses. Results: Although almost all pharmacies interviewed (94%) were aware of the law and most of them (64%) dispensed naloxone without a physician's prescription, few statistically significant differences were found between surveyed categories. There were no significant relationships between naloxone availability at pharmacies and overdose death rates. Conclusions: Our results suggest that the number of California pharmacies dispensing naloxone without a physician's prescription has continued to increase since the implementation of CA AB1535. However, despite increased access to naloxone at pharmacies, opioid overdose rates have continued to rise since 2016, indicating the need for a multifaceted harm reduction approach.

14.
Adv Physiol Educ ; 36(4): 319-24, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23209014

ABSTRACT

The Einthoven triangle is central to the field of electrocardiography, but the concept of cardiac vectors is often a difficult notion for students to grasp. To illustrate this principle, we constructed a device that recreates the conditions of an ECG reading using a battery to simulate the electrical vector of the heart and three voltmeters for the main electrocardiographic leads. Requiring minimal construction with low cost, this device provides hands-on practice that enables students to rediscover the principles of the Einthoven triangle, namely, that the direction of the cardiac dipole can be predicted from the deflections in any two leads and that lead I + lead III = lead II independent of the position of heart's electrical vector. We built a total of 6 devices for classes of 30 students and tested them in the first-year Human Physiology course at the University of California-Davis School of Medicine. Combined with traditional demonstrations with ECG machines, this equipment demonstrated its ability to help medical students obtain a solid foundation of the basic principles of electrocardiography.


Subject(s)
Curriculum , Electrocardiography/instrumentation , Physiology/education , Physiology/instrumentation , Students, Medical , Electrocardiography/methods , Heart/physiology , Humans , Physiology/methods , Vectorcardiography/instrumentation , Vectorcardiography/methods
15.
JCI Insight ; 7(22)2022 11 22.
Article in English | MEDLINE | ID: mdl-36509290

ABSTRACT

Sinoatrial node (SAN) cells are the heart's primary pacemaker. Their activity is tightly regulated by ß-adrenergic receptor (ß-AR) signaling. Adenylyl cyclase (AC) is a key enzyme in the ß-AR pathway that catalyzes the production of cAMP. There are current gaps in our knowledge regarding the dominant AC isoforms and the specific roles of Ca2+-activated ACs in the SAN. The current study tests the hypothesis that distinct AC isoforms are preferentially expressed in the SAN and compartmentalize within microdomains to orchestrate heart rate regulation during ß-AR signaling. In contrast to atrial and ventricular myocytes, SAN cells express a diverse repertoire of ACs, with ACI as the predominant Ca2+-activated isoform. Although ACI-KO (ACI-/-) mice exhibit normal cardiac systolic or diastolic function, they experience SAN dysfunction. Similarly, SAN-specific CRISPR/Cas9-mediated gene silencing of ACI results in sinus node dysfunction. Mechanistically, hyperpolarization-activated cyclic nucleotide-gated 4 (HCN4) channels form functional microdomains almost exclusively with ACI, while ryanodine receptor and L-type Ca2+ channels likely compartmentalize with ACI and other AC isoforms. In contrast, there were no significant differences in T-type Ca2+ and Na+ currents at baseline or after ß-AR stimulation between WT and ACI-/- SAN cells. Due to its central characteristic feature as a Ca2+-activated isoform, ACI plays a unique role in sustaining the rise of local cAMP and heart rates during ß-AR stimulation. The findings provide insights into the critical roles of the Ca2+-activated isoform of AC in sustaining SAN automaticity that is distinct from contractile cardiomyocytes.


Subject(s)
Adenylyl Cyclases , Sinoatrial Node , Animals , Mice , Sinoatrial Node/metabolism , Adenylyl Cyclases/genetics , Adenylyl Cyclases/metabolism , Calcium/metabolism , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Protein Isoforms/metabolism
16.
Am J Physiol Heart Circ Physiol ; 300(2): H617-26, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21131481

ABSTRACT

Endothelin-1 (ET-1) and activation of protein kinase C (PKC) have been implicated in alterations of myocyte function in cardiac hypertrophy and heart failure. Changes in cellular Ca2+ handling and electrophysiological properties also occur in these states and may contribute to mechanical dysfunction and arrhythmias. While ET-1 or PKC stimulation induces cellular hypertrophy in cultured neonatal rat ventricular myocytes (NRVMs), a system widely used in studies of hypertrophic signaling, there is little data about electrophysiological changes. Here we studied the effects of ET-1 (100 nM) or the PKC activator phorbol 12-myristate 13-acetate (PMA, 1 µM) on ionic currents in NRVMs. The acute effects of PMA or ET-1 (≤30 min) were small or insignificant. However, PMA or ET-1 exposure for 48-72 h increased cell capacitance by 100 or 25%, respectively, indicating cellular hypertrophy. ET-1 also slightly increased Ca2+ current density (T and L type). Na+/Ca2+ exchange current was increased by chronic pretreatment with either PMA or ET-1. In contrast, transient outward and delayed rectifier K+ currents were strongly downregulated by PMA or ET-1 pretreatment. Inward rectifier K+ current tended toward a decrease at larger negative potential, but time-independent outward K+ current was unaltered by either treatment. The enhanced inward and reduced outward currents also result in action potential prolongation after PMA or ET-1 pretreatment. We conclude that chronic PMA or ET-1 exposure in cultured NRVMs causes altered functional expression of cardiac ion currents, which mimic electrophysiological changes seen in whole animal and human hypertrophy and heart failure.


Subject(s)
Calcium Channels/biosynthesis , Endothelin-1/pharmacology , Myocytes, Cardiac/metabolism , Potassium Channels/biosynthesis , Sodium-Calcium Exchanger/biosynthesis , Tetradecanoylphorbol Acetate/pharmacology , Action Potentials/drug effects , Animals , Animals, Newborn , Blotting, Western , Calcium Channels/drug effects , Cell Size , Cells, Cultured , Electrophysiological Phenomena , In Vitro Techniques , Myocytes, Cardiac/drug effects , Patch-Clamp Techniques , Phosphorylation , Potassium Channels/drug effects , Protein Kinase C/metabolism , Rats , Rats, Sprague-Dawley
17.
Front Physiol ; 12: 760176, 2021.
Article in English | MEDLINE | ID: mdl-35115953

ABSTRACT

Cerebral arterial vasoreactivity is vital to the regulation of cerebral blood flow. Depolarization of arterial myocytes elicits whole-cell Ca2+ oscillations as well as subcellular Ca2+ sparks due to activation of ryanodine receptors on the sarcoplasmic reticulum. Previous evidence illustrates that contraction of cerebral arteries from sheep and underlying Ca2+ signaling pathways are modified by age and that long-term hypoxia (LTH) causes aberrations in Ca2+ signaling pathways and downstream effectors impacting vasoregulation. We hypothesize that age and LTH affect the influence of membrane depolarization on whole-cell intracellular Ca2+ oscillations and sub-cellular Ca2+ spark activity in cerebral arteries. To test this hypothesis, we examined Ca2+ oscillatory and spark activities using confocal fluorescence imaging techniques of Fluo-4 loaded basilar arterial myocytes of low- and high-altitude term fetal (∼145 days of gestation) and adult sheep, where high-altitude pregnant and non-pregnant sheep were placed at 3,801 m for >100 days. Ca2+ oscillations and sparks were recorded using an in situ preparation evaluated in the absence or presence of 30 mM K+ (30K) to depolarize myocytes. Myocytes from adult animals tended to have a lower basal rate of whole-cell Ca2+ oscillatory activity and 30K increased the activity within cells. LTH decreased the ability of myocytes to respond to depolarization independent of age. These observations illustrate that both altitude and age play a role in affecting whole-cell and localized Ca2+ signaling, which are important to arterial vasoreactivity and cerebral blood flow.

18.
Biophys J ; 93(11): 3835-47, 2007 Dec 01.
Article in English | MEDLINE | ID: mdl-17704163

ABSTRACT

Ca-calmodulin-dependent protein kinase II (CaMKII) was recently shown to alter Na(+) channel gating and recapitulate a human Na(+) channel genetic mutation that causes an unusual combined arrhythmogenic phenotype in patients: simultaneous long QT syndrome and Brugada syndrome. CaMKII is upregulated in heart failure where arrhythmias are common, and CaMKII inhibition can reduce arrhythmias. Thus, CaMKII-dependent channel modulation may contribute to acquired arrhythmic disease. We developed a Markovian Na(+) channel model including CaMKII-dependent changes, and incorporated it into a comprehensive myocyte action potential (AP) model with Na(+) and Ca(2+) transport. CaMKII shifts Na(+) current (I(Na)) availability to more negative voltage, enhances intermediate inactivation, and slows recovery from inactivation (all loss-of-function effects), but also enhances late noninactivating I(Na) (gain of function). At slow heart rates, with long diastolic time for I(Na) recovery, late I(Na) is the predominant effect, leading to AP prolongation (long QT syndrome). At fast heart rates, where recovery time is limited and APs are shorter, there is little effect on AP duration, but reduced availability decreases I(Na), AP upstroke velocity, and conduction (Brugada syndrome). CaMKII also increases cardiac Ca(2+) and K(+) currents (I(Ca) and I(to)), complicating CaMKII-dependent AP changes. Incorporating I(Ca) and I(to) effects individually prolongs and shortens AP duration. Combining I(Na), I(Ca), and I(to) effects results in shortening of AP duration with CaMKII. With transmural heterogeneity of I(to) and I(to) downregulation in heart failure, CaMKII may accentuate dispersion of repolarization. This provides a useful initial framework to consider pathways by which CaMKII may contribute to arrhythmogenesis.


Subject(s)
Action Potentials/physiology , Calcium Channels/physiology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/physiology , Calcium/metabolism , Ion Channel Gating/physiology , Myocytes, Cardiac/physiology , Sodium Channels/physiology , Animals , Calcium Signaling/physiology , Computer Simulation , Heart Ventricles/cytology , Markov Chains , Models, Cardiovascular , Rabbits , Ventricular Function
19.
Circ Res ; 95(12): 1216-24, 2004 Dec 10.
Article in English | MEDLINE | ID: mdl-15528464

ABSTRACT

The KCNQ1-G589D gene mutation, associated with a long-QT syndrome, has been shown to disrupt yotiao-mediated targeting of protein kinase A and protein phosphatase-1 to the I(Ks) channel. To investigate how this defect may lead to ventricular arrhythmia during sympathetic stimulation, we use integrative computational models of beta-adrenergic signaling, myocyte excitation-contraction coupling, and action potential propagation in a rabbit ventricular wedge. Paradoxically, we find that the KCNQ1-G589D mutation alone does not prolong the QT interval. But when coupled with beta-adrenergic stimulation in a whole-cell model, the KCNQ1-G589D mutation induced QT prolongation and transient afterdepolarizations, known cellular mechanisms for arrhythmogenesis. These cellular mechanisms amplified tissue heterogeneities in a three-dimensional rabbit ventricular wedge model, elevating transmural dispersion of repolarization and creating other T-wave abnormalities on simulated electrocardiograms. Increasing heart rate protected both single myocyte and the coupled myocardium models from arrhythmic consequences. These findings suggest that the KCNQ1-G589D mutation disrupts a critical link between beta-adrenergic signaling and myocyte electrophysiology, creating both triggers of cardiac arrhythmia and a myocardial substrate vulnerable to such electrical disturbances.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Substitution , Computer Simulation , Cytoskeletal Proteins/metabolism , Long QT Syndrome/etiology , Models, Cardiovascular , Models, Molecular , Mutation, Missense , Myocytes, Cardiac/metabolism , Point Mutation , Potassium Channels, Voltage-Gated/genetics , Receptors, Adrenergic, beta-1/physiology , Action Potentials/drug effects , Adrenergic beta-1 Receptor Agonists , Animals , Binding Sites , Computational Biology , Electrocardiography , Heart Ventricles/cytology , Ion Transport/drug effects , Isoproterenol/pharmacology , KCNQ Potassium Channels , KCNQ1 Potassium Channel , Long QT Syndrome/genetics , Long QT Syndrome/physiopathology , Myocardial Contraction , Potassium/metabolism , Potassium Channels, Voltage-Gated/chemistry , Potassium Channels, Voltage-Gated/metabolism , Protein Binding , Protein Conformation , Protein Interaction Mapping , Rabbits , Structure-Activity Relationship
20.
Prog Biophys Mol Biol ; 85(2-3): 163-78, 2004.
Article in English | MEDLINE | ID: mdl-15142742

ABSTRACT

Computer modeling of cardiac myocytes has flourished in recent years. Models have evolved from mathematical descriptions of ionic channels alone to more sophisticated formulations that include calcium transport mechanisms, ATP production and metabolic pathways. The increased complexity is fueled by the new data available in the field. The continuous production of experimental data has led to the evolution of increasingly refined descriptions of the phenomena by modelers. Integrating the numerous systems involved in cardiac myocyte homeostasis makes the use of computer models necessary due to the unreliability of intuitive approaches. However the complexity of the model should not imply a cumbersome operation of the program. As with any tool, computer models have to be easy to operate or their strength will be diminished and potential users will not benefit fully from them. The contribution of the computer modeler to their respective biological fields will be more successful and enduring if modelers devote sufficient time to implement their equations into a model with user-friendly characteristics.


Subject(s)
Action Potentials/physiology , Calcium Signaling/physiology , Cell Membrane/physiology , Ion Channels/physiology , Membrane Potentials/physiology , Models, Cardiovascular , Myocardial Contraction/physiology , Myocytes, Cardiac/physiology , Animals , Computer Simulation , Humans
SELECTION OF CITATIONS
SEARCH DETAIL