15 results on '"Wang DW"'
Search Results
2. Cardiac sodium channel dysfunction in sudden infant death syndrome.
- Author
-
Wang DW, Desai RR, Crotti L, Arnestad M, Insolia R, Pedrazzini M, Ferrandi C, Vege A, Rognum T, Schwartz PJ, and George AL Jr
- Published
- 2007
3. Prevalence of long-QT syndrome gene variants in sudden infant death syndrome.
- Author
-
Arnestad M, Crotti L, Rognum TO, Insolia R, Pedrazzini M, Ferrandi C, Vege A, Wang DW, Rhodes TE, George AL Jr, and Schwartz PJ
- Published
- 2007
4. Sustained whole-body functional rescue in congestive heart failure and muscular dystrophy hamsters by systemic gene transfer.
- Author
-
Zhu T, Zhou L, Mori S, Wang Z, McTiernan CF, Qiao C, Chen C, Wang DW, Li J, and Xiao X
- Published
- 2005
5. LncRNA DCRT Protects Against Dilated Cardiomyopathy by Preventing NDUFS2 Alternative Splicing by Binding to PTBP1.
- Author
-
Du H, Zhao Y, Wen J, Dai B, Hu G, Zhou Y, Yin Z, Ding N, Li H, Fan J, Nie X, Wang F, Liu Q, Wen Z, Xu G, Wang DW, and Chen C
- Subjects
- Animals, Mice, Humans, Electron Transport Complex I metabolism, Electron Transport Complex I genetics, Myocytes, Cardiac metabolism, Myocytes, Cardiac pathology, Male, Mitochondria, Heart metabolism, Mitochondria, Heart pathology, Mitochondria, Heart genetics, Mice, Transgenic, Cardiomyopathy, Dilated genetics, Cardiomyopathy, Dilated metabolism, Cardiomyopathy, Dilated pathology, Polypyrimidine Tract-Binding Protein genetics, Polypyrimidine Tract-Binding Protein metabolism, RNA, Long Noncoding genetics, RNA, Long Noncoding metabolism, Alternative Splicing, Mice, Knockout, Heterogeneous-Nuclear Ribonucleoproteins genetics, Heterogeneous-Nuclear Ribonucleoproteins metabolism
- Abstract
Background: Dilated cardiomyopathy is characterized by left ventricular dilation and continuous systolic dysfunction. Mitochondrial impairment is critical in dilated cardiomyopathy; however, the underlying mechanisms remain unclear. Here, we explored the cardioprotective role of a heart-enriched long noncoding RNA, the dilated cardiomyopathy repressive transcript (DCRT), in maintaining mitochondrial function., Methods: The DCRT knockout (DCRT
-/- ) mice and DCRT knockout cells were developed using CRISPR-Cas9 technology. Cardiac-specific DCRT transgenic mice were generated using α-myosin heavy chain promoter. Chromatin coimmunoprecipitation, RNA immunoprecipitation, Western blot, and isoform sequencing were performed to investigate the underlying mechanisms., Results: We found that the long noncoding RNA DCRT was highly enriched in the normal heart tissues and that its expression was significantly downregulated in the myocardium of patients with dilated cardiomyopathy. DCRT-/- mice spontaneously developed cardiac dysfunction and enlargement with mitochondrial impairment. DCRT transgene or overexpression with the recombinant adeno-associated virus system in mice attenuated cardiac dysfunction induced by transverse aortic constriction treatment. Mechanistically, DCRT inhibited the third exon skipping of NDUFS2 (NADH dehydrogenase ubiquinone iron-sulfur protein 2) by directly binding to PTBP1 (polypyrimidine tract binding protein 1) in the nucleus of cardiomyocytes. Skipping of the third exon of NDUFS2 induced mitochondrial dysfunction by competitively inhibiting mitochondrial complex I activity and binding to PRDX5 (peroxiredoxin 5) and suppressing its antioxidant activity. Furthermore, coenzyme Q10 partially alleviated mitochondrial dysfunction in cardiomyocytes caused by DCRT reduction., Conclusions: Our study revealed that the loss of DCRT contributed to PTBP1-mediated exon skipping of NDUFS2, thereby inducing cardiac mitochondrial dysfunction during dilated cardiomyopathy development, which could be partially treated with coenzyme Q10 supplementation., Competing Interests: None.- Published
- 2024
- Full Text
- View/download PDF
6. AGO2 Protects Against Diabetic Cardiomyopathy by Activating Mitochondrial Gene Translation.
- Author
-
Zhan J, Jin K, Xie R, Fan J, Tang Y, Chen C, Li H, and Wang DW
- Subjects
- Mice, Animals, Genes, Mitochondrial, Mitochondria genetics, Myocytes, Cardiac metabolism, Diabetic Cardiomyopathies, Sirtuin 3 genetics, MicroRNAs genetics, Diabetes Mellitus metabolism
- Abstract
Background: Diabetes is associated with cardiovascular complications. microRNAs translocate into subcellular organelles to modify genes involved in diabetic cardiomyopathy. However, functional properties of subcellular AGO2 (Argonaute2), a core member of miRNA machinery, remain elusive., Methods: We elucidated the function and mechanism of subcellular localized AGO2 on mouse models for diabetes and diabetic cardiomyopathy. Recombinant adeno-associated virus type 9 was used to deliver AGO2 to mice through the tail vein. Cardiac structure and functions were assessed by echocardiography and catheter manometer system., Results: AGO2 was decreased in mitochondria of diabetic cardiomyocytes. Overexpression of mitochondrial AGO2 attenuated diabetes-induced cardiac dysfunction. AGO2 recruited TUFM , a mitochondria translation elongation factor, to activate translation of electron transport chain subunits and decrease reactive oxygen species. Malonylation, a posttranslational modification of AGO2, reduced the importing of AGO2 into mitochondria in diabetic cardiomyopathy. AGO2 malonylation was regulated by a cytoplasmic-localized short isoform of SIRT3 through a previously unknown demalonylase function., Conclusions: Our findings reveal that the SIRT3 -AGO2- CYTB axis links glucotoxicity to cardiac electron transport chain imbalance, providing new mechanistic insights and the basis to develop mitochondria targeting therapies for diabetic cardiomyopathy., Competing Interests: Disclosures None.
- Published
- 2024
- Full Text
- View/download PDF
7. Ganglioside GM3 Protects Against Abdominal Aortic Aneurysm by Suppressing Ferroptosis.
- Author
-
Zhang F, Li K, Zhang W, Zhao Z, Chang F, Du J, Zhang X, Bao K, Zhang C, Shi L, Liu Z, Dai X, Chen C, Wang DW, Xian Z, Jiang H, and Ai D
- Subjects
- Humans, Mice, Animals, G(M3) Ganglioside metabolism, Proteomics, Muscle, Smooth, Vascular metabolism, Iron, Myocytes, Smooth Muscle metabolism, Disease Models, Animal, Ferroptosis, Aortic Aneurysm, Abdominal genetics, Aortic Aneurysm, Abdominal prevention & control, Aortic Aneurysm, Abdominal metabolism
- Abstract
Background: Abdominal aortic aneurysm (AAA) is a potentially life-threatening vascular condition, but approved medical therapies to prevent AAA progression and rupture are currently lacking. Sphingolipid metabolism disorders are associated with the occurrence and development of AAA. It has been discovered that ganglioside GM3, a sialic acid-containing type of glycosphingolipid, plays a protective role in atherosclerosis, which is an important risk factor for AAA; however, the potential contribution of GM3 to AAA development has not been investigated., Methods: We performed a metabolomics study to evaluated GM3 level in plasma of human patients with AAA. We profiled GM3 synthase (ST3GAL5) expression in the mouse model of aneurysm and human AAA tissues through Western blotting and immunofluorescence staining. RNA sequencing, affinity purification and mass spectrometry, proteomic analysis, surface plasmon resonance analysis, and functional studies were used to dissect the molecular mechanism of GM3-regulating ferroptosis. We conditionally deleted and overexpressed St3gal5 in smooth muscle cells (SMCs) in vivo to investigate its role in AAA., Results: We found significantly reduced plasma levels of GM3 in human patients with AAA. GM3 content and ST3GAL5 expression were decreased in abdominal aortic vascular SMCs in patients with AAA and an AAA mouse model. RNA sequencing analysis showed that ST3GAL5 silencing in human aortic SMCs induced ferroptosis. We showed that GM3 interacted directly with the extracellular domain of TFR1 (transferrin receptor 1), a cell membrane protein critical for cellular iron uptake, and disrupted its interaction with holo-transferrin. SMC-specific St3gal5 knockout exacerbated iron accumulation at lesion sites and significantly promoted AAA development in mice, whereas GM3 supplementation suppressed lipid peroxidation, reduced iron deposition in aortic vascular SMCs, and markedly decreased AAA incidence., Conclusions: Together, these results suggest that GM3 dysregulation promotes ferroptosis of vascular SMCs in AAA. Furthermore, GM3 may constitute a new therapeutic target for AAA., Competing Interests: Disclosures None.
- Published
- 2024
- Full Text
- View/download PDF
8. Glucose-Sensitive Myokine/Cardiokine MG53 Regulates Systemic Insulin Response and Metabolic Homeostasis.
- Author
-
Wu HK, Zhang Y, Cao CM, Hu X, Fang M, Yao Y, Jin L, Chen G, Jiang P, Zhang S, Song R, Peng W, Liu F, Guo J, Tang L, He Y, Shan D, Huang J, Zhou Z, Wang DW, Lv F, and Xiao RP
- Subjects
- Adult, Animals, Antibodies, Monoclonal pharmacology, Antigens, CD metabolism, Biomarkers blood, Blood Glucose drug effects, Case-Control Studies, Diabetes Mellitus drug therapy, Diabetes Mellitus enzymology, Diabetes Mellitus immunology, Disease Models, Animal, Female, HEK293 Cells, Homeostasis, Humans, Hypoglycemic Agents pharmacology, Male, Membrane Proteins antagonists & inhibitors, Membrane Proteins genetics, Membrane Proteins immunology, Mice, Mice, Inbred C57BL, Mice, Knockout, Muscle Proteins metabolism, Muscle, Skeletal enzymology, Myocardium enzymology, Rats, Sprague-Dawley, Rats, Zucker, Receptor, Insulin metabolism, Signal Transduction, Tripartite Motif Proteins metabolism, Vesicular Transport Proteins metabolism, Blood Glucose metabolism, Diabetes Mellitus blood, Energy Metabolism drug effects, Insulin Resistance, Membrane Proteins metabolism
- Abstract
Background: Mitsugumin 53 (MG53 or TRIM72), a striated muscle-specific E3 ligase, promotes ubiquitin-dependent degradation of the insulin receptor and insulin receptor substrate-1 and subsequently induces insulin resistance, resulting in metabolic syndrome and type 2 diabetes mellitus (T2DM). However, it is unknown how MG53 from muscle regulates systemic insulin response and energy metabolism. Increasing evidence demonstrates that muscle secretes proteins as myokines or cardiokines that regulate systemic metabolic processes. We hypothesize that MG53 may act as a myokine/cardiokine, contributing to interorgan regulation of insulin sensitivity and metabolic homeostasis., Methods: Using perfused rodent hearts or skeletal muscle, we investigated whether high glucose, high insulin, or their combination (conditions mimicking metabolic syndrome or T2DM) alters MG53 protein concentration in the perfusate. We also measured serum MG53 levels in rodents and humans in the presence or absence of metabolic diseases, particularly T2DM. The effects of circulating MG53 on multiorgan insulin response were evaluated by systemic delivery of recombinant MG53 protein to mice. Furthermore, the potential involvement of circulating MG53 in the pathogenesis of T2DM was assessed by neutralizing blood MG53 with monoclonal antibodies in diabetic db/db mice. Finally, to delineate the mechanism underlying the action of extracellular MG53 on insulin signaling, we analyzed the potential interaction of MG53 with extracellular domain of insulin receptor using coimmunoprecipitation and surface plasmon resonance assays., Results: Here, we demonstrate that MG53 is a glucose-sensitive myokine/cardiokine that governs the interorgan regulation of insulin sensitivity. First, high glucose or high insulin induces MG53 secretion from isolated rodent hearts and skeletal muscle. Second, hyperglycemia is accompanied by increased circulating MG53 in humans and rodents with diabetes mellitus. Third, systemic delivery of recombinant MG53 or cardiac-specific overexpression of MG53 causes systemic insulin resistance and metabolic syndrome in mice, whereas neutralizing circulating MG53 with monoclonal antibodies has therapeutic effects in T2DM db/db mice. Mechanistically, MG53 binds to the extracellular domain of the insulin receptor and acts as an allosteric blocker., Conclusions: Thus, MG53 has dual actions as a myokine/cardiokine and an E3 ligase, synergistically inhibiting the insulin signaling pathway. Targeting circulating MG53 opens a new therapeutic avenue for T2DM and its complications.
- Published
- 2019
- Full Text
- View/download PDF
9. Response by Zhou et al to Letter Regarding Article, "Cardioprotective Role of Myeloid-Derived Suppressor Cells in Heart Failure".
- Author
-
Zhou L, Li Z, and Wang DW
- Subjects
- Humans, Heart Failure, Myeloid-Derived Suppressor Cells
- Published
- 2019
- Full Text
- View/download PDF
10. Cystathionine γ Lyase Sulfhydrates the RNA Binding Protein Human Antigen R to Preserve Endothelial Cell Function and Delay Atherogenesis.
- Author
-
Bibli SI, Hu J, Sigala F, Wittig I, Heidler J, Zukunft S, Tsilimigras DI, Randriamboavonjy V, Wittig J, Kojonazarov B, Schürmann C, Siragusa M, Siuda D, Luck B, Abdel Malik R, Filis KA, Zografos G, Chen C, Wang DW, Pfeilschifter J, Brandes RP, Szabo C, Papapetropoulos A, and Fleming I
- Subjects
- Aged, Aged, 80 and over, Animals, Atherosclerosis genetics, Atherosclerosis pathology, Atherosclerosis prevention & control, Carotid Arteries pathology, Carotid Artery Diseases genetics, Carotid Artery Diseases pathology, Carotid Artery Diseases prevention & control, Cathepsins metabolism, Cell Adhesion, Cell Adhesion Molecules genetics, Cell Adhesion Molecules metabolism, Cystathionine gamma-Lyase deficiency, Cystathionine gamma-Lyase genetics, Disease Models, Animal, Disease Progression, ELAV-Like Protein 1 genetics, Endothelial Cells pathology, Female, HEK293 Cells, Humans, Male, Mice, Inbred C57BL, Mice, Knockout, ApoE, Middle Aged, Monocytes metabolism, Monocytes pathology, Phosphorylation, Protein Processing, Post-Translational, Signal Transduction, Atherosclerosis enzymology, Carotid Arteries enzymology, Carotid Artery Diseases enzymology, Cystathionine gamma-Lyase metabolism, ELAV-Like Protein 1 metabolism, Endothelial Cells enzymology, Hydrogen Sulfide metabolism, Plaque, Atherosclerotic
- Abstract
Background: Hydrogen sulfide (H
2 S), generated by cystathionine γ lyase (CSE), is an important endogenous regulator of vascular function. The aim of the present study was to investigate the control and consequences of CSE activity in endothelial cells under physiological and proatherogenic conditions., Methods: Endothelial cell CSE knockout mice were generated, and lung endothelial cells were studied in vitro (gene expression, protein sulfhydration, and monocyte adhesion). Mice were crossed onto the apolipoprotein E-deficient background, and atherogenesis (partial carotid artery ligation) was monitored over 21 days. CSE expression, H2 S bioavailability, and amino acid profiling were also performed with human material., Results: The endothelial cell-specific deletion of CSE selectively increased the expression of CD62E and elevated monocyte adherence in the absence of an inflammatory stimulus. Mechanistically, CD62E mRNA was more stable in endothelial cells from CSE-deficient mice, an effect attributed to the attenuated sulfhydration and dimerization of the RNA-binding protein human antigen R. CSE expression was upregulated in mice after partial carotid artery ligation and in atheromas from human subjects. Despite the increase in CSE protein, circulating and intraplaque H2 S levels were reduced, a phenomenon that could be attributed to the serine phosphorylation (on Ser377) and inhibition of the enzyme, most likely resulting from increased interleukin-1β. Consistent with the loss of H2 S, human antigen R sulfhydration was attenuated in atherosclerosis and resulted in the stabilization of human antigen R-target mRNAs, for example, CD62E and cathepsin S, both of which are linked to endothelial cell activation and atherosclerosis. The deletion of CSE from endothelial cells was associated with the accelerated development of endothelial dysfunction and atherosclerosis, effects that were reversed on treatment with a polysulfide donor. Finally, in mice and humans, plasma levels of the CSE substrate l-cystathionine negatively correlated with vascular reactivity and H2 S levels, indicating its potential use as a biomarker for vascular disease., Conclusions: The constitutive S-sulfhydration of human antigen R (on Cys13) by CSE-derived H2 S prevents its homodimerization and activity, which attenuates the expression of target proteins such as CD62E and cathepsin S. However, as a consequence of vascular inflammation, the beneficial actions of CSE-derived H2 S are lost owing to the phosphorylation and inhibition of the enzyme.- Published
- 2019
- Full Text
- View/download PDF
11. Cardioprotective Role of Myeloid-Derived Suppressor Cells in Heart Failure.
- Author
-
Zhou L, Miao K, Yin B, Li H, Fan J, Zhu Y, Ba H, Zhang Z, Chen F, Wang J, Zhao C, Li Z, and Wang DW
- Subjects
- Aged, Animals, Cell Proliferation, Cells, Cultured, Coculture Techniques, Cytokines blood, Disease Models, Animal, Disease Progression, Female, Gene Expression Regulation, Heart Failure chemically induced, Humans, Immune Tolerance, Isoproterenol, Male, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Middle Aged, Rats, Heart Failure immunology, Myeloid-Derived Suppressor Cells physiology, T-Lymphocytes immunology
- Abstract
Background: Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of cells that expand in cancer, inflammation, and infection and negatively regulate inflammation and the immune response. Heart failure (HF) is a complex clinical syndrome wherein inflammation induction and incomplete resolution can potentially contribute to HF development and progression. However, the role of MDSCs in HF remains unclear., Methods: The percentage of MDSCs in patients with HF and in mice with pressure overload-induced HF using isoproterenol infusion or transverse aortic constriction (TAC) was detected by flow cytometry. The effects of MDSCs on isoproterenol- or TAC-induced HF were observed on depleting MDSCs with 5-fluorouracil (50 mg/kg) or gemcitabine (120 mg/kg), transferring purified MDSCs, or enhancing endogenous MDSCs with rapamycin (2 mg·kg
-1 ·d-1 ). Hypertrophic markers and inflammatory factors were detected by ELISA, real-time polymerase chain reaction, or Western blot. Cardiac functions were determined by echocardiography and hemodynamic analysis., Results: The percentage of human leukocyte antigen-D-related (HLA-DR)- CD33+ CD11b+ MDSCs in the blood of patients with HF was significantly increased and positively correlated with disease severity and increased plasma levels of cytokines, including interleukin-6, interleukin-10, and transforming growth factor-β. Furthermore, MDSCs derived from patients with HF inhibited T-cell proliferation and interferon-γ secretion. Similar results were observed in TAC- and isoproterenol-induced HF in mice. Pharmaceutical depletion of MDSCs significantly exacerbated isoproterenol- and TAC-induced pathological cardiac remodeling and inflammation, whereas adoptive transfer of MDSCs prominently rescued isoproterenol- and TAC-induced HF. Consistently, administration of rapamycin significantly increased endogenous MDSCs by suppressing their differentiation and improved isoproterenol- and TAC-induced HF, but MDSC depletion mostly blocked beneficial rapamycin-mediated effects. Mechanistically, MDSC-secreted molecules suppressed isoproterenol-induced hypertrophy and proinflammatory gene expression in cardiomyocytes in a coculture system. Neutralization of interleukin-10 blunted both monocytic MDSC- and granulocytic MDSC-mediated anti-inflammatory and antihypertrophic effects, but treatment with a nitric oxide inhibitor only partially blocked the antihypertrophic effect of monocytic MDSCs., Conclusions: Our findings revealed a cardioprotective role of MDSCs in HF by their antihypertrophic effects on cardiomyocytes and anti-inflammatory effects through interleukin-10 and nitric oxide. Pharmacological targeting of MDSCs by rapamycin constitutes a promising therapeutic strategy for HF., (© 2018 The Authors.)- Published
- 2018
- Full Text
- View/download PDF
12. MicroRNA-21 Lowers Blood Pressure in Spontaneous Hypertensive Rats by Upregulating Mitochondrial Translation.
- Author
-
Li H, Zhang X, Wang F, Zhou L, Yin Z, Fan J, Nie X, Wang P, Fu XD, Chen C, and Wang DW
- Subjects
- Animals, Blood Pressure drug effects, Hypertension drug therapy, Male, MicroRNAs genetics, MicroRNAs pharmacology, Mitochondria drug effects, Mitochondria genetics, Rats, Rats, Inbred SHR, Rats, Wistar, Reactive Oxygen Species metabolism, Transcriptional Activation drug effects, Up-Regulation drug effects, Blood Pressure physiology, Hypertension metabolism, MicroRNAs therapeutic use, Mitochondria metabolism, Transcriptional Activation physiology, Up-Regulation physiology
- Abstract
Background: Excessive reactive oxygen species generated in mitochondria has been implicated as a causal event in hypertensive cardiomyopathy. Multiple recent studies suggest that microRNAs (miRNAs) are able to translocate to mitochondria to modulate mitochondrial activities, but the medical significance of such a new miRNA function has remained unclear. Here, we characterized spontaneous hypertensive rats (SHRs) in comparison with Wistar rats, finding that micro RNA-21 (miR-21) was dramatically induced in SHRs relative to Wistar rats. We designed a series of experiments to determine whether miR-21 is involved in regulating reactive oxygen species generation in mitochondria, and if so, how induced miR-21 may either contribute to hypertensive cardiomyopathy or represent a compensatory response., Methods: Western blotting was used to compare the expression of key nuclear genome (nDNA)-encoded and mitochondrial genome (mtDNA)-encoded genes involved in reactive oxygen species production in SHRs and Wistar rats. Bioinformatics was used to predict miRNA targets followed by biochemical validation using quantitative real-time polymerase chain reaction and Ago2 immunoprecipitation. The direct role of miRNA in mitochondria was determined by GW182 dependence, which is required for miRNA to function in the cytoplasm, but not in mitochondria. Recombinant adeno-associated virus (type 9) was used to deliver miRNA mimic to rats via tail vein, and blood pressure was monitored with a photoelectric tail-cuff system. Cardiac structure and functions were assessed by echocardiography and catheter manometer system., Results: We observed a marked reduction of mtDNA-encoded cytochrome b (mt-Cytb) in the heart of SHRs. Downregulation of mt-Cytb by small interfering RNA in mitochondria recapitulates some key disease features, including elevated reactive oxygen species production. Computational prediction coupled with biochemical analysis revealed that miR-21 directly targeted mt-Cytb to positively modulate mt-Cytb translation in mitochondria. Circulating miR-21 levels in hypertensive patients were significantly higher than those in controls, showing a positive correlation between miR-21 expression and blood pressure. Remarkably, recombinant adeno-associated virus-mediated delivery of miR-21 was sufficient to reduce blood pressure and attenuate cardiac hypertrophy in SHRs., Conclusions: Our findings reveal a positive function of miR-21 in mitochondrial translation, which is sufficient to reduce blood pressure and alleviate cardiac hypertrophy in SHRs. This observation indicates that induced miR-21 is part of the compensatory program and suggests a novel theoretical ground for developing miRNA-based therapeutics against hypertension., (© 2016 American Heart Association, Inc.)
- Published
- 2016
- Full Text
- View/download PDF
13. Clinical, genetic, and biophysical characterization of SCN5A mutations associated with atrioventricular conduction block.
- Author
-
Wang DW, Viswanathan PC, Balser JR, George AL Jr, and Benson DW
- Subjects
- Action Potentials, Adult, Amino Acid Sequence, Cell Line, Child, Computer Simulation, Electrocardiography, Female, Heart Block diagnosis, Heart Block physiopathology, Humans, Kinetics, Male, Middle Aged, Molecular Sequence Data, NAV1.5 Voltage-Gated Sodium Channel, Patch-Clamp Techniques, Protein Structure, Tertiary, Sequence Homology, Amino Acid, Sodium Channels chemistry, Sodium Channels metabolism, Atrioventricular Node physiopathology, Heart Block genetics, Mutation, Sodium Channels genetics
- Abstract
Background: Three distinct cardiac arrhythmia disorders, the long-QT syndrome, Brugada syndrome, and conduction system disease, have been associated with heterozygous mutations in the cardiac voltage-gated sodium channel alpha-subunit gene (SCN5A). We present clinical, genetic, and biophysical features of 2 new SCN5A mutations that result in atrioventricular (AV) conduction block. Methods and Results- SCN5A was used as a candidate gene in 2 children with AV block. Molecular genetic studies revealed G to A transition mutations that resulted in the substitution of serine for glycine (G298S) in the domain I S5-S6 loop and asparagine for aspartic acid (D1595N) within the S3 segment of domain IV. The functional consequences of G298S and D1595N were assessed by whole-cell patch clamp recording of recombinant mutant channels coexpressed with the beta1 subunit in a cultured cell line (tsA201). Both mutations impair fast inactivation but do not exhibit sustained non-inactivating currents. The mutations also reduce sodium current density and enhance slower inactivation components. Action potential simulations predict that this combination of biophysical abnormalities will significantly slow myocardial conduction velocity., Conclusions: A distinct pattern of biophysical abnormalities not previously observed for any other SCN5A mutant have been recognized in association with AV block. These data provide insight into the distinct clinical phenotypes resulting from mutation of a single ion channel.
- Published
- 2002
- Full Text
- View/download PDF
14. Cardiac Na(+) channel dysfunction in Brugada syndrome is aggravated by beta(1)-subunit.
- Author
-
Makita N, Shirai N, Wang DW, Sasaki K, George AL Jr, Kanno M, and Kitabatake A
- Subjects
- Amino Acid Substitution, Animals, Cell Membrane physiology, Chromosome Mapping, Humans, Ion Channel Gating, Kinetics, Long QT Syndrome physiopathology, Macromolecular Substances, Membrane Potentials, Myocardium metabolism, Oocytes physiology, Protein Structure, Secondary, Recombinant Proteins chemistry, Recombinant Proteins metabolism, Sodium Channels chemistry, Syndrome, Ventricular Fibrillation physiopathology, Xenopus, Chromosomes, Human, Pair 3, Heart physiopathology, Long QT Syndrome genetics, Sodium Channels genetics, Sodium Channels physiology, Ventricular Fibrillation genetics
- Abstract
Background: Mutations in the gene encoding the human cardiac Na(+) channel alpha-subunit (hH1) are responsible for chromosome 3-linked congenital long-QT syndrome (LQT3) and idiopathic ventricular fibrillation (IVF). An auxiliary beta(1)-subunit, widely expressed in excitable tissues, shifts the voltage dependence of steady-state inactivation toward more negative potentials and restores normal gating kinetics of brain and skeletal muscle Na(+) channels expressed in Xenopus oocytes but has little if any functional effect on the cardiac isoform. Here, we characterize the altered effects of a human beta(1)-subunit (hbeta(1)) on the heterologously expressed hH1 mutation (T1620M) previously associated with IVF., Methods and Results: When expressed alone in Xenopus oocytes, T1620M exhibited no persistent currents, in contrast to the LQT3 mutant channels, but the midpoint of steady-state inactivation (V(1/2)) was significantly shifted toward more positive potentials than for wild-type hH1. Coexpression of hbeta(1) did not significantly alter current decay or recovery from inactivation of wild-type hH1; however, it further shifted the V(1/2) and accelerated the recovery from inactivation of T1620M. Oocyte macropatch analysis revealed that the activation kinetics of T1620M were normal., Conclusions: It is suggested that coexpression of hbeta(1) exposes a more severe functional defect that results in a greater overlap in the relationship between channel inactivation and activation (window current) in T1620M, which is proposed to be a potential pathophysiological mechanism of IVF in vivo. One possible explanation for our finding is an altered alpha-/beta(1)-subunit association in the mutant.
- Published
- 2000
- Full Text
- View/download PDF
15. Congenital long-QT syndrome caused by a novel mutation in a conserved acidic domain of the cardiac Na+ channel.
- Author
-
Wei J, Wang DW, Alings M, Fish F, Wathen M, Roden DM, and George AL Jr
- Subjects
- Adolescent, Animals, Base Sequence, Cloning, Molecular, Conserved Sequence, DNA Primers, Death, Sudden, Electrocardiography, Electrophysiology, Female, Humans, Long QT Syndrome diagnosis, Male, Membrane Potentials drug effects, Membrane Potentials physiology, Molecular Sequence Data, Mutagenesis, Site-Directed, Myocardium chemistry, NAV1.5 Voltage-Gated Sodium Channel, Oocytes physiology, Pedigree, Polymorphism, Single-Stranded Conformational, Protein Structure, Tertiary, Sequence Homology, Amino Acid, Sodium Channels chemistry, Sodium Channels metabolism, Structure-Activity Relationship, Tetrodotoxin pharmacology, Xenopus, Long QT Syndrome congenital, Long QT Syndrome genetics, Point Mutation, Sodium Channels genetics
- Abstract
Background: Congenital long-QT syndrome (LQTS) is an inherited condition of abnormal cardiac excitability characterized clinically by an increased risk of ventricular tachyarrhythmias. One form, LQT3, is caused by mutations in the cardiac voltage-dependent sodium channel gene, SCN5A. Only 5 SCN5A mutations have been associated with LQTS, and more work is needed to improve correlations between SCN5A genotypes and associated clinical syndromes., Methods and Results: We researched a 3-generation white family with autosomal dominant LQTS who exhibited a wide clinical spectrum from mild bradycardia to sudden death. Molecular genetic studies revealed a single nucleotide substitution in SCN5A exon 28 that caused the substitution of Glu1784 by Lys (E1784K). The mutation occurs in a highly conserved domain within the C-terminus of the cardiac sodium channel containing multiple, negatively charged amino acids. Two-electrode voltage-clamp recordings of a recombinant E1784K mutant channel expressed in Xenopus oocytes revealed a defect in fast inactivation characterized by a small, persistent current during long membrane depolarizations. Coexpression of the mutant with the human sodium channel beta1-subunit did not affect the persistent current, even though we did observe shifts in the voltage dependence of steady-state inactivation. Neutralizing multiple, negatively charged residues in the same region of the sodium channel C-terminus did not cause a more severe functional defect., Conclusions: We characterized the genetics and molecular pathophysiology of a novel SCN5A sodium channel mutation, E1784K. The functional defect exhibited by the mutant channel causes delayed myocardial repolarization, and our data on the effects of multiple charge neutralizations in this region of the C-terminus suggest that the molecular mechanism of channel dysfunction involves an allosteric rather than a direct effect on channel gating.
- Published
- 1999
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.