Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 115
Filter
1.
Nat Commun ; 14(1): 3312, 2023 06 07.
Article in English | MEDLINE | ID: mdl-37286550

ABSTRACT

Mutations in thyroid hormone receptor α1 (TRα1) cause Resistance to Thyroid Hormone α (RTHα), a disorder characterized by hypothyroidism in TRα1-expressing tissues including the heart. Surprisingly, we report that treatment of RTHα patients with thyroxine to overcome tissue hormone resistance does not elevate their heart rate. Cardiac telemetry in male, TRα1 mutant, mice indicates that such persistent bradycardia is caused by an intrinsic cardiac defect and not due to altered autonomic control. Transcriptomic analyses show preserved, thyroid hormone (T3)-dependent upregulation of pacemaker channels (Hcn2, Hcn4), but irreversibly reduced expression of several ion channel genes controlling heart rate. Exposure of TRα1 mutant male mice to higher maternal T3 concentrations in utero, restores altered expression and DNA methylation of ion channels, including Ryr2. Our findings indicate that target genes other than Hcn2 and Hcn4 mediate T3-induced tachycardia and suggest that treatment of RTHα patients with thyroxine in high dosage without concomitant tachycardia, is possible.


Subject(s)
Thyroid Hormone Resistance Syndrome , Thyroxine , Male , Animals , Mice , Thyroxine/therapeutic use , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/genetics , Thyroid Hormone Resistance Syndrome/genetics , Thyroid Hormones , Thyroid Hormone Receptors alpha/genetics , Thyroid Hormone Receptors alpha/metabolism , Mutation , Tachycardia/genetics
2.
Int J Mol Sci ; 23(13)2022 Jun 23.
Article in English | MEDLINE | ID: mdl-35805966

ABSTRACT

Many cardiac insults causing atrial remodeling are linked to either stretch or tachycardia, but a comparative characterization of their effects on early remodeling events in human myocardium is lacking. Here, we applied isometric stretch or sustained tachycardia at 2.5 Hz in human atrial trabeculae for 6 h followed by microarray gene expression profiling. Among largely independent expression patterns, we found a small common fraction with the microRNA miR-1183 as the highest up-regulated transcript (up to 4-fold). Both, acute stretch and tachycardia induced down-regulation of the predicted miR-1183 target genes ADAM20 and PLA2G7. Furthermore, miR-1183 was also significantly up-regulated in chronically remodeled atrial samples from patients with persistent atrial fibrillation (3-fold up-regulation versus sinus rhythm samples), and in ventricular myocardium from dilative cardiomyopathy hearts (2-fold up-regulation) as compared to non-failing controls. In sum, although stretch and tachycardia show distinct transcriptomic signatures in human atrial myocardium, both cardiac insults consistently regulate the expression of miR-1183 and its downstream targets in acute and chronic remodeling. Thus, elevated expression of miR-1183 might serve as a tissue biomarker for atrial remodeling and might be of potential functional significance in cardiac disease.


Subject(s)
Atrial Fibrillation , Atrial Remodeling , MicroRNAs , Atrial Fibrillation/genetics , Atrial Fibrillation/metabolism , Atrial Fibrillation/pathology , Biomarkers/metabolism , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Myocardium/metabolism , Tachycardia/genetics , Tachycardia/metabolism
4.
JAMA Intern Med ; 181(9): 1185-1193, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34279564

ABSTRACT

Importance: The notion that caffeine increases the risk of cardiac arrhythmias is common. However, evidence that the consumption of caffeinated products increases the risk of arrhythmias remains poorly substantiated. Objective: To assess the association between consumption of common caffeinated products and the risk of arrhythmias. Design, Setting, and Participants: This prospective cohort study analyzed longitudinal data from the UK Biobank between January 1, 2006, and December 31, 2018. After exclusion criteria were applied, 386 258 individuals were available for analyses. Exposures: Daily coffee intake and genetic polymorphisms that affect caffeine metabolism. Main Outcomes and Measures: Any cardiac arrhythmia, including atrial fibrillation or flutter, supraventricular tachycardia, ventricular tachycardia, premature atrial complexes, and premature ventricular complexes. Results: A total of 386 258 individuals (mean [SD] age, 56 [8] years; 52.3% female) were assessed. During a mean (SD) follow-up of 4.5 (3.1) years, 16 979 participants developed an incident arrhythmia. After adjustment for demographic characteristics, comorbid conditions, and lifestyle habits, each additional cup of habitual coffee consumed was associated with a 3% lower risk of incident arrhythmia (hazard ratio [HR], 0.97; 95% CI, 0.96-0.98; P < .001). In analyses of each arrhythmia alone, statistically significant associations exhibiting a similar magnitude were observed for atrial fibrillation and/or flutter (HR, 0.97; 95% CI, 0.96-0.98; P < .001) and supraventricular tachycardia (HR, 0.96; 95% CI, 0.94-0.99; P = .002). Two distinct interaction analyses, one using a caffeine metabolism-related polygenic score of 7 genetic polymorphisms and another restricted to CYP1A2 rs762551 alone, did not reveal any evidence of effect modification. A mendelian randomization study that used these same genetic variants revealed no significant association between underlying propensities to differing caffeine metabolism and the risk of incident arrhythmia. Conclusions and Relevance: In this prospective cohort study, greater amounts of habitual coffee consumption were associated with a lower risk of arrhythmia, with no evidence that genetically mediated caffeine metabolism affected that association. Mendelian randomization failed to provide evidence that caffeine consumption was associated with arrhythmias.


Subject(s)
Caffeine/metabolism , Coffee/adverse effects , Cytochrome P-450 CYP1A2/genetics , Life Style , Mendelian Randomization Analysis/methods , Polymorphism, Genetic , Tachycardia/epidemiology , Adult , Aged , Cytochrome P-450 CYP1A2/metabolism , Female , Follow-Up Studies , Humans , Incidence , Male , Middle Aged , Prospective Studies , Risk Factors , Tachycardia/etiology , Tachycardia/genetics , Time Factors , United States/epidemiology
5.
BMC Med Genet ; 21(1): 211, 2020 10 21.
Article in English | MEDLINE | ID: mdl-33087052

ABSTRACT

BACKGROUND: Marfan syndrome (MFS) is a common autosomal dominant inherited disease, and the occurrence rate is around 0.1-0.2‰. The causative variant of FNB1 gene accounts for approximately 70-80% of all MFS cases. In this study, we found a heterozygous c.3217G > T (p.Glu1073*) nonsense variant in the FBN1 gene. This finding extended the variant spectrum of the FBN1 gene and will provide a solution for patients to bear healthy offspring by preimplantation genetic testing or prenatal diagnosis. CASE PRESENTATION: The patient was treated due to tachycardia during excitement in a hospital. Echocardiography showed dilatation of the ascending aorta and main pulmonary artery, mitral regurgitation (mild), tricuspid regurgitation (mild), and abnormal left ventricular filling. Electrocardiograph showed sinus rhythm. In addition, flutters of shadows in front of his eyes and vitreous opacity were present in the patient. Genomic DNA was extracted from peripheral blood samples from members of the family and 100 unrelated controls. Potential variants were screened out by next-generation sequencing and confirmed by MLPA & Sanger sequencing. Real-time fluorescence quantitative PCR (RT-qPCR) was performed to detect the relative mRNA quantitation in the patient. A heterozygous nonsense variant c.3217G > T of the FBN1 gene, which resulted in p. Glu1073Term, was identified in both patients. Only wild type bases were found in the cDNA sequence of the patient. Real-time fluorogenic quantitative PCR results showed that the relative expression level of FBN1 cDNA in the patient was only about 21% compared to that of normal individuals. This variant c.3217G > T of the FBN1 gene introduces a Stop codon in the cb-EGF12 domain. We speculated that a premature translational-termination codon (PTC) was located in the mRNA and the target mRNA was disintegrated through a process known as nonsense-mediated mRNA decay (NMD), which led to a significant decrease of the fibrillin-1 protein, eventually causing clinical symptoms in the patient. CONCLUSIONS: In this study, we found a heterozygous c.3217G > T (p.Glu1073*) nonsense variant in the FBN1 gene, which eventually led to Marfan syndrome in a Chinese family.


Subject(s)
Aortic Valve Insufficiency/genetics , Codon, Nonsense , Fibrillin-1/genetics , Marfan Syndrome/genetics , Mitral Valve Insufficiency/genetics , RNA, Messenger/genetics , Tachycardia/genetics , Adult , Aged , Aortic Valve Insufficiency/diagnosis , Aortic Valve Insufficiency/ethnology , Aortic Valve Insufficiency/pathology , Asian People , Base Sequence , Electrocardiography , Family , Female , Fibrillin-1/deficiency , Gene Expression , Genes, Dominant , Humans , Male , Marfan Syndrome/diagnosis , Marfan Syndrome/ethnology , Marfan Syndrome/pathology , Middle Aged , Mitral Valve Insufficiency/diagnosis , Mitral Valve Insufficiency/ethnology , Mitral Valve Insufficiency/pathology , Nonsense Mediated mRNA Decay , Pedigree , Tachycardia/diagnosis , Tachycardia/ethnology , Tachycardia/pathology
6.
J Clin Invest ; 130(7): 3671-3683, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32484458

ABSTRACT

The baroreceptor reflex is a powerful neural feedback that regulates arterial pressure (AP). Mechanosensitive channels transduce pulsatile AP to electrical signals in baroreceptors. Here we show that tentonin 3 (TTN3/TMEM150C), a cation channel activated by mechanical strokes, is essential for detecting AP changes in the aortic arch. TTN3 was expressed in nerve terminals in the aortic arch and nodose ganglion (NG) neurons. Genetic ablation of Ttn3 induced ambient hypertension, tachycardia, AP fluctuations, and impaired baroreflex sensitivity. Chemogenetic silencing or activation of Ttn3+ neurons in the NG resulted in an increase in AP and heart rate, or vice versa. More important, overexpression of Ttn3 in the NG of Ttn3-/- mice reversed the cardiovascular changes observed in Ttn3-/- mice. We conclude that TTN3 is a molecular component contributing to the sensing of dynamic AP changes in baroreceptors.


Subject(s)
Aorta, Thoracic , Blood Pressure , Membrane Proteins/metabolism , Neurons/metabolism , Nodose Ganglion , Pressoreceptors , Animals , Aorta, Thoracic/innervation , Aorta, Thoracic/metabolism , Aorta, Thoracic/physiopathology , HEK293 Cells , Humans , Hypertension/genetics , Hypertension/metabolism , Hypertension/physiopathology , Membrane Proteins/genetics , Mice , Mice, Knockout , Nodose Ganglion/metabolism , Nodose Ganglion/physiopathology , Pressoreceptors/metabolism , Pressoreceptors/physiopathology , Tachycardia/genetics , Tachycardia/metabolism , Tachycardia/physiopathology
7.
EBioMedicine ; 55: 102700, 2020 May.
Article in English | MEDLINE | ID: mdl-32192914

ABSTRACT

BACKGROUND: Duchenne muscular dystrophy (DMD) is caused by the loss of dystrophin. Severe and ultimately lethal, DMD progresses relatively slowly in that patients become wheelchair bound only around age twelve with a survival expectancy reaching the third decade of life. METHODS: The mildly-affected mdx mouse model of DMD, and transgenic DysΔMTB-mdx and Fiona-mdx mice expressing dystrophin or utrophin, respectively, were exposed to either mild (scruffing) or severe (subordination stress) stress paradigms and profiled for their behavioral and physiological responses. A subgroup of mdx mice exposed to subordination stress were pretreated with the beta-blocker metoprolol. FINDINGS: Subordination stress caused lethality in ∼30% of mdx mice within 24 h and ∼70% lethality within 48 h, which was not rescued by metoprolol. Lethality was associated with heart damage, waddling gait and hypo-locomotion, as well as marked up-regulation of the hypothalamus-pituitary-adrenocortical axis. A novel cardiovascular phenotype emerged in mdx mice, in that scruffing caused a transient drop in arterial pressure, while subordination stress caused severe and sustained hypotension with concurrent tachycardia. Transgenic expression of dystrophin or utrophin in skeletal muscle protected mdx mice from scruffing and social stress-induced responses including mortality. INTERPRETATION: We have identified a robust new stress phenotype in the otherwise mildly affected mdx mouse that suggests relatively benign handling may impact the outcome of behavioural experiments, but which should also expedite the knowledge-based therapy development for DMD. FUNDING: Greg Marzolf Jr. Foundation, Summer's Wish Fund, NIAMS, Muscular Dystrophy Association, University of Minnesota and John and Cheri Gunvalson Trust.


Subject(s)
Dystrophin/genetics , Gait Disorders, Neurologic/mortality , Heart Failure/mortality , Muscular Dystrophy, Duchenne/mortality , Stress, Psychological/mortality , Utrophin/genetics , Adrenergic beta-Antagonists/pharmacology , Animals , Arterial Pressure/drug effects , Disease Models, Animal , Dystrophin/metabolism , Gait Disorders, Neurologic/complications , Gait Disorders, Neurologic/genetics , Gait Disorders, Neurologic/physiopathology , Gene Expression , Heart Failure/complications , Heart Failure/genetics , Heart Failure/physiopathology , Humans , Hypotension/complications , Hypotension/genetics , Hypotension/mortality , Hypotension/physiopathology , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/metabolism , Hypothalamo-Hypophyseal System/physiopathology , Male , Metoprolol/pharmacology , Mice , Mice, Inbred mdx , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiopathology , Muscular Dystrophy, Duchenne/complications , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/physiopathology , Stress, Psychological/complications , Stress, Psychological/genetics , Stress, Psychological/physiopathology , Survival Analysis , Tachycardia/complications , Tachycardia/genetics , Tachycardia/mortality , Tachycardia/physiopathology , Transgenes , Utrophin/metabolism
9.
J Mol Cell Cardiol ; 135: 67-78, 2019 10.
Article in English | MEDLINE | ID: mdl-31419440

ABSTRACT

Atrial fibrillation (AF) is associated with oxidative stress and Ca2+-handling abnormalities in atrial myocytes. Our prior study has demonstrated the involvement of CD44, a membrane receptor for hyaluronan (HA), in the pathogenesis of AF. This study further evaluated whether CD44 and its related signaling mediate atrial tachycardia-induced oxidative stress and Ca2+-handling abnormalities. Tachypacing in atrium-derived myocytes (HL-1 cell line) induced the activation of CD44-related signaling, including HA and HA synthase (HAS) expression. Blocking HAS/HA/CD44 signaling attenuated tachypacing-induced oxidative stress (NADPH oxidase [NOX] 2/4 expression) and Ca2+-handling abnormalities (oxidized Ca2+/calmodulin-dependent protein kinase II [ox-CaMKII] and phospho-ryanodine receptor type 2 [p-RyR2] expression) in HL-1 myocytes. Furthermore, a direct association between CD44 and NOX4 was documented in tachy-paced HL-1 myocytes and atrial tissues from AF patients. In vitro, Ca2+ spark frequencies in atrial myocytes isolated from CD44-/- mice were lower than those from wild-type mice. Furthermore, administration of an anti-CD44 blocking antibody in atrial myocytes isolated from wild-type mice diminished the frequency of Ca2+ spark. Ex vivo tachypacing models of CD44-/- mice exhibited a lower degree of oxidative stress and expression of ox-CaMKII/p-RyR2 in their atria than those of wild-type mice. In vivo, burst atrial pacing stimulated a less inducibility of AF in CD44-/-mice than in wild-type mice. In conclusion, atrial tachypacing-induced Ca2+-handling abnormalities are mediated via CD44/NOX4 signaling, which provides a possible explanation for the development of AF.


Subject(s)
Atrial Fibrillation/genetics , Atrial Remodeling/genetics , Heart Atria/metabolism , NADPH Oxidase 4/genetics , Tachycardia/genetics , Animals , Atrial Fibrillation/metabolism , Atrial Fibrillation/pathology , Atrial Remodeling/physiology , Calcium Signaling/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Heart Atria/pathology , Humans , Hyaluronan Receptors/genetics , Hyaluronic Acid/genetics , Mice , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , NADPH Oxidase 2/genetics , Ryanodine Receptor Calcium Release Channel , Sarcoplasmic Reticulum/metabolism , Sarcoplasmic Reticulum/pathology , Signal Transduction/genetics , Tachycardia/pathology
10.
Thyroid ; 29(2): 258-267, 2019 02.
Article in English | MEDLINE | ID: mdl-30618332

ABSTRACT

BACKGROUND: Cyclic peptides derived from some cylindrical loops of the leucine-rich repeat domain (LRD) of the thyrotropin receptor (TSHR) have been shown to treat disease manifestations in a mouse model of Graves' disease during a long-term protocol of four-weekly immunizations with adenovirus coding for the TSHR A-subunit (Ad-TSHR289). METHODS: In a follow-up study, two additional cyclic peptides were tested, which were shortened in order to obtain additional information on the minimally involved epitopes and to enable easier production conditions. In addition, a linear peptide was tested, which mimics parts of three loops of the native TSHR LRD structure, and is potentially able to block the discontinuous epitopes of anti-TSHR antibodies. RESULTS: The novel peptides markedly reduced thyroid size, serum thyroxine levels, retro-orbital fibrosis, and tachycardia in Ad-TSHR289-immunized mice. In immunologically naïve mice, administration of the peptides did not induce any immune response. CONCLUSIONS: In summary, novel cyclic peptides mitigate many clinical findings in a mouse model of established Graves' disease and orbitopathy, and may therefore provide an additional therapeutic option compared to existing drugs or interventions.


Subject(s)
Graves Ophthalmopathy/immunology , Orbit/physiopathology , Peptides, Cyclic/therapeutic use , Adenoviridae , Animals , Disease Models, Animal , Epitopes/chemistry , Female , Fibrosis , Graves Ophthalmopathy/therapy , HEK293 Cells , Humans , Immunoglobulins, Thyroid-Stimulating , Mice , Mice, Inbred BALB C , Receptors, Thyrotropin/genetics , Tachycardia/genetics , Thyroxine/blood
11.
Circ Genom Precis Med ; 11(7): e001980, 2018 07.
Article in English | MEDLINE | ID: mdl-29987112

ABSTRACT

BACKGROUND: Tachycardia-induced cardiomyopathy (TIC) is a reversible cardiomyopathy induced by tachyarrhythmia, and the genetic background of the TIC is not well understood. The hyperpolarization-activated cyclic nucleotide-gated channel gene HCN4 is highly expressed in the conduction system where it is involved in heart rate control. We speculated that the HCN4 gene is associated with TIC. METHODS: We enrolled 930 Japanese patients with atrial fibrillation (AF) for screening, 350 Japanese patients with AF for replication, and 1635 non-AF controls. In the screening AF set, we compared HCN4 single-nucleotide polymorphism genotypes between AF subjects with TIC (TIC, n=73) and without TIC (non-TIC, n=857). Of 17 HCN4 gene-tag single-nucleotide polymorphisms, rs7172796, rs2680344, rs7164883, rs11631816, and rs12905211 were significantly associated with TIC. Among them, only rs7164883 was independently associated with TIC after conditional analysis (TIC versus non-TIC: minor allele frequency, 26.0% versus 9.7%; P=1.62×10-9; odds ratio=3.2). RESULTS: We confirmed this association of HCN4 single-nucleotide polymorphism rs7164883 with TIC in the replication set (TIC=41 and non-TIC=309; minor allele frequency, 28% versus 9.9%; P=1.94×10-6; odds ratio=3.6). The minor allele frequency of rs7164883 was similar in patients with AF and non-AF controls (11% versus 10.9%; P=0.908). CONCLUSIONS: The HCN4 gene single-nucleotide polymorphism rs7164883 may be a new genetic marker for TIC in patients with AF.


Subject(s)
Atrial Fibrillation , Cardiomegaly , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Muscle Proteins , Polymorphism, Genetic , Potassium Channels , Tachycardia , Atrial Fibrillation/complications , Atrial Fibrillation/genetics , Atrial Fibrillation/metabolism , Atrial Fibrillation/pathology , Cardiomegaly/etiology , Cardiomegaly/genetics , Cardiomegaly/metabolism , Cardiomegaly/pathology , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/genetics , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Muscle Proteins/genetics , Muscle Proteins/metabolism , Potassium Channels/genetics , Potassium Channels/metabolism , Tachycardia/complications , Tachycardia/genetics , Tachycardia/metabolism , Tachycardia/pathology
12.
Circ J ; 82(4): 965-973, 2018 03 23.
Article in English | MEDLINE | ID: mdl-29398686

ABSTRACT

BACKGROUND: Recent experimental studies have demonstrated that several microRNAs (miRNAs) expressed in atrial tissue promote a substrate of atrial fibrillation (AF). However, because it has not been fully elucidated whether these experimental data contribute to identifying circulating miRNAs as biomarkers for AF, we used a combined analysis of human serum and murine atrial samples with the aim of identifying these biomarkers for predicting AF.Methods and Results:Comprehensive analyses were performed to screen 733 miRNAs in serum from 10 AF patients and 5 controls, and 672 miRNAs in atrial tissue from 6 inducible atrial tachycardia model mice and 3 controls. We selected miRNAs for which expression was detected in both analyses, and their expression levels were changed in the human analyses, the murine analyses, or both. This screening identified 11 candidate miRNAs. Next, we quantified the selected miRNAs using a quantitative RT-PCR in 50 AF and 50 non-AF subjects. The individual assessment revealed that 4 miRNAs (miR-99a-5p, miR-192-5p, miR-214-3p, and miR-342-5p) were significantly upregulated in AF patients. A receiver-operating characteristics curve indicated that miR-214-3p and miR-342-5p had the highest accuracy. The combination of the 4 miRNAs modestly improved the predictive accuracy for AF (76% sensitivity, 80% specificity). CONCLUSIONS: Novel circulating miRNAs were upregulated in the serum of AF patients and might be potential biomarkers of AF.


Subject(s)
Atrial Fibrillation/diagnosis , Circulating MicroRNA/blood , Aged , Animals , Atrial Fibrillation/blood , Atrial Fibrillation/genetics , Biomarkers/blood , Case-Control Studies , Female , Humans , Male , Mice , MicroRNAs/blood , Middle Aged , ROC Curve , Sensitivity and Specificity , Tachycardia/blood , Tachycardia/genetics , Up-Regulation , Young Adult
13.
J Biomed Sci ; 25(1): 8, 2018 Jan 31.
Article in English | MEDLINE | ID: mdl-29382335

ABSTRACT

BACKGROUND: Intake of ethanol (alcohol) has been shown to influence cardiovascular function; the underlying brain mechanism remains unclear. Noting that nitric oxide (NO) system in the CNS is involved in the regulation of cardiovascular function, the present study examined the role of NO in medulla in ethanol-induced cardiovascular changes. METHODS: Ethanol was administered by oral gavage at dose of 3.2 g/kg once every day for 8 consecutive days. Changes in blood pressure (BP) and heart rate (HR) in response to ethanol were measured by radiotelemetry method in freely moving female Sprague-Dawley rats. NO modulators were applied by intracerebroventricular (ICV) injection. The protein levels of nitric oxide synthase (NOS) and NO content in rostroventral medulla were measured by Western blot and nitrate/nitrite colorimetric assay kit, respectively. RESULTS: Ethanol intake had little effects on basal BP and HR following 8 consecutive day treatments. A significant increase in HR but not BP following ethanol intake was observed at 6th and 8th, but not at 1st and 4th day treatments as compared with saline group. A decrease in the protein expression of neuronal NOS (nNOS) but not inducible NOS or endothelial NOS and a decline in the level of NO in the medulla 30 min after ethanol administration was observed at 8th day treatment. ICV treatment with NO donors attenuated ethanol-induced tachycardia effects at 8th day treatment. Ethanol produced significantly tachycardia responses when ICV nNOS inhibitors were given at 1st day treatment. CONCLUSION: Our results suggest that medulla nNOS/NO pathways play an important role in ethanol regulation of HR.


Subject(s)
Ethanol/adverse effects , Medulla Oblongata/drug effects , Nitric Oxide Synthase Type I/genetics , Nitric Oxide/metabolism , Tachycardia/genetics , Animals , Female , Medulla Oblongata/physiology , Nitric Oxide Synthase Type I/metabolism , Rats , Rats, Sprague-Dawley , Tachycardia/chemically induced , Tachycardia/physiopathology
14.
Proc Natl Acad Sci U S A ; 114(21): E4288-E4295, 2017 05 23.
Article in English | MEDLINE | ID: mdl-28490495

ABSTRACT

Alternative splicing changes the CaV1.2 calcium channel electrophysiological property, but the in vivo significance of such altered channel function is lacking. Structure-function studies of heterologously expressed CaV1.2 channels could not recapitulate channel function in the native milieu of the cardiomyocyte. To address this gap in knowledge, we investigated the role of alternative exon 33 of the CaV1.2 calcium channel in heart function. Exclusion of exon 33 in CaV1.2 channels has been reported to shift the activation potential -10.4 mV to the hyperpolarized direction, and increased expression of CaV1.2Δ33 channels was observed in rat myocardial infarcted hearts. However, how a change in CaV1.2 channel electrophysiological property, due to alternative splicing, might affect cardiac function in vivo is unknown. To address these questions, we generated mCacna1c exon 33-/--null mice. These mice contained CaV1.2Δ33 channels with a gain-of-function that included conduction of larger currents that reflects a shift in voltage dependence and a modest increase in single-channel open probability. This altered channel property underscored the development of ventricular arrhythmia, which is reflected in significantly more deaths of exon 33-/- mice from ß-adrenergic stimulation. In vivo telemetric recordings also confirmed increased frequencies in premature ventricular contractions, tachycardia, and lengthened QT interval. Taken together, the significant decrease or absence of exon 33-containing CaV1.2 channels is potentially proarrhythmic in the heart. Of clinical relevance, human ischemic and dilated cardiomyopathy hearts showed increased inclusion of exon 33. However, the possible role that inclusion of exon 33 in CaV1.2 channels may play in the pathogenesis of human heart failure remains unclear.


Subject(s)
Action Potentials/genetics , Calcium Channels, L-Type/genetics , Long QT Syndrome/genetics , Tachycardia/genetics , Ventricular Premature Complexes/genetics , Action Potentials/physiology , Alternative Splicing/genetics , Animals , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/metabolism , Cells, Cultured , Colforsin/pharmacology , Electrophysiological Phenomena/genetics , Heart Failure/genetics , Heart Failure/pathology , Isoproterenol/pharmacology , Long QT Syndrome/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Nifedipine/pharmacology , Rats , Sequence Deletion/genetics , Tachycardia/pathology , Ventricular Premature Complexes/pathology
15.
Int J Mol Sci ; 18(5)2017 May 10.
Article in English | MEDLINE | ID: mdl-28489021

ABSTRACT

The flow of intracellular calcium (Ca2+) is critical for the activation and regulation of important biological events that are required in living organisms. As the major Ca2+ repositories inside the cell, the endoplasmic reticulum (ER) and the sarcoplasmic reticulum (SR) of muscle cells are central in maintaining and amplifying the intracellular Ca2+ signal. The morphology of these organelles, along with the distribution of key calcium-binding proteins (CaBPs), regulatory proteins, pumps, and receptors fundamentally impact the local and global differences in Ca2+ release kinetics. In this review, we will discuss the structural and morphological differences between the ER and SR and how they influence localized Ca2+ release, related diseases, and the need for targeted genetically encoded calcium indicators (GECIs) to study these events.


Subject(s)
Calcium Signaling , Malignant Hyperthermia/genetics , Myotonia Congenita/genetics , Sarcoplasmic Reticulum/metabolism , Tachycardia/genetics , Animals , Calcium Channels/genetics , Calcium Channels/metabolism , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Humans , Malignant Hyperthermia/metabolism , Myotonia Congenita/metabolism , Sarcoplasmic Reticulum/genetics , Tachycardia/metabolism
16.
J Biomed Sci ; 23(1): 80, 2016 Nov 11.
Article in English | MEDLINE | ID: mdl-27835996

ABSTRACT

The goal of precision medicine is to separate patient populations into groups to ultimately provide customized care tailored to patients. In terms of precision medicine, ~540 million people in the world have a genetic variant of the aldehyde dehydrogenase 2 (ALDH2) enzyme causing a flushing response and tachycardia after alcohol consumption. The genetic variant is identified as ALDH2*2 and originates from East Asian descendants of the Han Chinese. The variant is particularly important to consider when discussing lifestyle choices with patients in terms of risk for developing specific diseases, preventative screening, and selection of medications for treatment. Here we provide examples why patients with an ALDH2*2 variant need more individualized medical management which is becoming a more standard practice in the precision medicine era.


Subject(s)
Alcohol Drinking/genetics , Aldehyde Dehydrogenase, Mitochondrial/genetics , Flushing/genetics , Tachycardia/genetics , Alcohol Drinking/pathology , Asian People/genetics , China , Flushing/pathology , Genetic Variation , Humans , Precision Medicine , Tachycardia/pathology
17.
Biochem J ; 473(21): 3951-3964, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27582498

ABSTRACT

Beat-to-beat alternations in the amplitude of the cytosolic Ca2+ transient (Ca2+ alternans) are thought to be the primary cause of cardiac alternans that can lead to cardiac arrhythmias and sudden death. Despite its important role in arrhythmogenesis, the mechanism underlying Ca2+ alternans remains poorly understood. Here, we investigated the role of cardiac ryanodine receptor (RyR2), the major Ca2+ release channel responsible for cytosolic Ca2+ transients, in cardiac alternans. Using a unique mouse model harboring a suppression-of-function (SOF) RyR2 mutation (E4872Q), we assessed the effect of genetically suppressing RyR2 function on Ca2+ and action potential duration (APD) alternans in intact hearts, and electrocardiogram (ECG) alternans in vivo We found that RyR2-SOF hearts displayed prolonged sarcoplasmic reticulum Ca2+ release refractoriness and enhanced propensity for Ca2+ alternans. RyR2-SOF hearts/mice also exhibited increased propensity for APD and ECG alternans. Caffeine, which enhances RyR2 activity and the propensity for catecholaminergic polymorphic ventricular tachycardia (CPVT), suppressed Ca2+ alternans in RyR2-SOF hearts, whereas carvedilol, a ß-blocker that suppresses RyR2 activity and CPVT, promoted Ca2+ alternans in these hearts. Thus, RyR2 function is an important determinant of Ca2+, APD, and ECG alternans. Our data also indicate that the activity of RyR2 influences the propensity for cardiac alternans and CPVT in an opposite manner. Therefore, overly suppressing or enhancing RyR2 function is pro-arrhythmic.


Subject(s)
Heart/physiopathology , Myocardium/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Animals , Disease Models, Animal , Electrocardiography , Heart/drug effects , Isoproterenol/pharmacology , Mice , Mutation/genetics , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Ryanodine Receptor Calcium Release Channel/genetics , Sarcoplasmic Reticulum/drug effects , Sarcoplasmic Reticulum/metabolism , Tachycardia/genetics , Tachycardia/metabolism , Tachycardia, Ventricular/genetics , Tachycardia, Ventricular/metabolism
18.
Medicine (Baltimore) ; 95(29): e4312, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27442679

ABSTRACT

Potassium calcium-activated channel subfamily N member 2 (KCNN2) encodes an integral membrane protein that forms small-conductance calcium-activated potassium (SK) channels. Recent studies in animal models show that SK channels are important in atrial and ventricular repolarization and arrhythmogenesis. However, the importance of SK channels in human arrhythmia remains unclear. The purpose of the present study was to test the association between genetic polymorphism of the SK2 channel and the occurrence of cardiac tachyarrhythmias in humans. We enrolled 327 Han Chinese, including 72 with clinically significant ventricular tachyarrhythmias (VTa) who had a history of aborted sudden cardiac death (SCD) or unexplained syncope, 98 with a history of atrial fibrillation (AF), and 144 normal controls. We genotyped 12 representative tag single nucleotide polymorphisms (SNPs) across a 141-kb genetic region containing the KCNN2 gene; these captured the full haplotype information. The rs13184658 and rs10076582 variants of KCNN2 were associated with VTa in both the additive and dominant models (odds ratio [OR] 2.89, 95% confidence interval [CI] = 1.505-5.545, P = 0.001; and OR 2.55, 95% CI = 1.428-4.566, P = 0.002, respectively). After adjustment for potential risk factors, the association remained significant. The population attributable risks of these 2 variants of VTa were 17.3% and 10.6%, respectively. One variant (rs13184658) showed weak but significant association with AF in a dominant model (OR 1.91, CI = 1.025-3.570], P = 0.042). There was a significant association between the KCNN2 variants and clinically significant VTa. These findings suggest an association between KCNN2 and VTa; it also appears that KCNN2 variants may be adjunctive markers for risk stratification in patients susceptible to SCD.


Subject(s)
Polymorphism, Single Nucleotide/genetics , Small-Conductance Calcium-Activated Potassium Channels/genetics , Tachycardia/genetics , Adult , Aged , Defibrillators, Implantable , Female , Gene Frequency/genetics , Genetic Predisposition to Disease/genetics , Genotype , Humans , Male , Middle Aged , Prospective Studies , Statistics as Topic , Tachycardia/therapy
19.
Am J Physiol Heart Circ Physiol ; 310(9): H1259-66, 2016 05 01.
Article in English | MEDLINE | ID: mdl-26945074

ABSTRACT

Cardiac pacemaker cell function is regulated by a coupled-clock system that integrates molecular cues on the cell-membrane surface (i.e., membrane clock) and on the sarcoplasmic reticulum (SR) (i.e., Ca(2+) clock). A recent study has shown that cotransfection of spontaneous beating cells (HEK293 cells and neonatal rat myocytes) with R524Q-mutant human hyperpolarization-activated cyclic nucleotide-gated molecules (the dominant component of funny channels) increases the funny channel's sensitivity to cAMP and leads to a decrease in spontaneous action potential (AP) cycle length (i.e., tachycardia). We hypothesize that in rabbit pacemaker cells, the same behavior is expected, and because of the coupled-clock system, the resultant steady-state decrease in AP cycle length will embody contributions from both clocks: the initial decrease in the spontaneous AP beating interval, arising from increased sensitivity of the f-channel to cAMP, will be accompanied by an increase in the adenylyl cyclase (AC)-cAMP-PKA-dependent phosphorylation activity, which will further decrease this interval. To test our hypothesis, we used the recently developed Yaniv-Lakatta pacemaker cell numerical model. This model predicts the cAMP signaling dynamics, as well as the kinetics and magnitude of protein phosphorylation in both normal and mutant pacemaker cells. We found that R524Q-mutant pacemaker cells have a shorter AP firing rate than that of wild-type cells and that gain in pacemaker function is the net effect of the R514Q mutation on the functioning of the coupled-clock system. Specifically, our results directly support the hypothesis that changes in Ca(2+)-activated AC-cAMP-PKA signaling are involved in the development of tachycardia in R524Q-mutant pacemaker cells.


Subject(s)
Biological Clocks , Cyclic AMP-Dependent Protein Kinases/metabolism , Heart Rate , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Models, Cardiovascular , Phosphorylation , Second Messenger Systems , Sinoatrial Node/enzymology , Tachycardia/enzymology , Action Potentials , Adenylyl Cyclases/metabolism , Animals , Calcium-Binding Proteins/metabolism , Computer Simulation , Cyclic AMP/metabolism , Genetic Predisposition to Disease , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/genetics , Kinetics , Mutation , Numerical Analysis, Computer-Assisted , Phenotype , Rabbits , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Sinoatrial Node/cytology , Tachycardia/genetics , Tachycardia/physiopathology
20.
Arterioscler Thromb Vasc Biol ; 36(2): 370-9, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26715683

ABSTRACT

OBJECTIVE: Palmitoylation, the reversible addition of the lipid palmitate to a cysteine, can alter protein localization, stability, and function. The ZDHHC family of protein acyl transferases catalyzes palmitoylation of numerous proteins. The role of ZDHHC enzymes in intact tissue and in vivo is largely unknown. Herein, we characterize vascular functions in a mouse that expresses a nonfunctional ZDHHC21 (F233Δ). APPROACH AND RESULTS: Physiological studies of isolated aortae and mesenteric arteries from F233Δ mice revealed an unexpected defect in responsiveness to phenylephrine, an α1 adrenergic receptor agonist. In vivo, F233Δ mice displayed a blunted response to infusion of phenylephrine, and they were found to have elevated catecholamine levels and elevated vascular α1 adrenergic receptor gene expression. Telemetry studies showed that the F233Δ mice were tachycardic and hypotensive at baseline, consistent with diminished vascular tone. In biochemical studies, ZDHHC21 was shown to palmitoylate the α1D adrenoceptor and to interact with it in a molecular complex, thus suggesting a possible molecular mechanism by which the receptor can be regulated by ZDHHC21. CONCLUSIONS: Together, the data support a model in which ZDHHC21 F233Δ diminishes the function of vascular α1 adrenergic receptors, leading to reduced vascular tone, which manifests in vivo as hypotension and tachycardia. This is to our knowledge the first demonstration of a ZDHHC isoform affecting vascular function in vivo and identifies a novel molecular mode of regulation of vascular tone and blood pressure.


Subject(s)
Acyltransferases/metabolism , Aorta/enzymology , Hemodynamics , Mesenteric Arteries/enzymology , Receptors, Adrenergic, alpha-1/metabolism , Acyltransferases/genetics , Adrenergic alpha-1 Receptor Agonists/pharmacology , Animals , Aorta/drug effects , Aorta/physiopathology , Blood Pressure , Dose-Response Relationship, Drug , Epinephrine/metabolism , Fibroblasts/drug effects , Fibroblasts/enzymology , Genotype , HEK293 Cells , Heart Rate , Hemodynamics/drug effects , Humans , Hypotension/enzymology , Hypotension/genetics , Hypotension/physiopathology , Lipoylation , Mesenteric Arteries/drug effects , Mesenteric Arteries/physiopathology , Mice, Inbred C57BL , Mice, Mutant Strains , Mutation , Norepinephrine/metabolism , Phenotype , Phenylephrine/pharmacology , Receptors, Adrenergic, alpha-1/drug effects , Receptors, Adrenergic, alpha-1/genetics , Signal Transduction , Tachycardia/enzymology , Tachycardia/genetics , Tachycardia/physiopathology , Time Factors , Transfection , Vasoconstriction
SELECTION OF CITATIONS
SEARCH DETAIL