Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 137
Filter
2.
Circ Arrhythm Electrophysiol ; 17(1): e012072, 2024 01.
Article in English | MEDLINE | ID: mdl-38099441

ABSTRACT

Although there is consensus on the management of patients with Brugada Syndrome with high risk for sudden cardiac arrest, asymptomatic or intermediate-risk patients present clinical management challenges. This document explores the management opinions of experts throughout the world for patients with Brugada Syndrome who do not fit guideline recommendations. Four real-world clinical scenarios were presented with commentary from small expert groups for each case. All authors voted on case-specific questions to evaluate the level of consensus among the entire group in nuanced diagnostic and management decisions relevant to each case. Points of agreement, points of controversy, and gaps in knowledge are highlighted.


Subject(s)
Brugada Syndrome , Heart Arrest , Humans , Brugada Syndrome/diagnosis , Brugada Syndrome/therapy , Electrocardiography , Heart Arrest/diagnosis , Heart Arrest/therapy , Death, Sudden, Cardiac/etiology , Death, Sudden, Cardiac/prevention & control , Consensus
3.
J Electrocardiol ; 82: 131-135, 2024.
Article in English | MEDLINE | ID: mdl-38128158

ABSTRACT

This non-technical review introduces key concepts in personalized ECG monitoring (pECG), which aims to optimize the detection of clinical events and their warning signs as well as the selection of alarm thresholds. We review several pECG methods, including anomaly detection and adaptive machine learning (ML), in which learning is performed sequentially as new data are collected. We describe a distributed-network multiscale pECG system to show how the computational load and time associated with adaptive ML could be optimized. In this architecture, the limited analysis of ECG waveforms is performed locally (e.g., on a smart phone) to determine a small number of clinically important ECG elements, and an adaptive ML engine is located on a remote server (Internet cloud) to determine an individual's "fingerprint" basis patterns and to detect anomalies in those patterns.


Subject(s)
Electrocardiography , Machine Learning , Humans , Electrocardiography/methods , Smartphone
4.
Nat Commun ; 14(1): 5595, 2023 09 11.
Article in English | MEDLINE | ID: mdl-37696839

ABSTRACT

Downregulation of endothelial Sirtuin1 (Sirt1) in insulin resistant states contributes to vascular dysfunction. Furthermore, Sirt1 deficiency in skeletal myocytes promotes insulin resistance. Here, we show that deletion of endothelial Sirt1, while impairing endothelial function, paradoxically improves skeletal muscle insulin sensitivity. Compared to wild-type mice, male mice lacking endothelial Sirt1 (E-Sirt1-KO) preferentially utilize glucose over fat, and have higher insulin sensitivity, glucose uptake, and Akt signaling in fast-twitch skeletal muscle. Enhanced insulin sensitivity of E-Sirt1-KO mice is transferrable to wild-type mice via the systemic circulation. Endothelial Sirt1 deficiency, by inhibiting autophagy and activating nuclear factor-kappa B signaling, augments expression and secretion of thymosin beta-4 (Tß4) that promotes insulin signaling in skeletal myotubes. Thus, unlike in skeletal myocytes, Sirt1 deficiency in the endothelium promotes glucose homeostasis by stimulating skeletal muscle insulin sensitivity through a blood-borne mechanism, and augmented secretion of Tß4 by Sirt1-deficient endothelial cells boosts insulin signaling in skeletal muscle cells.


Subject(s)
Insulin Resistance , Sirtuin 1 , Animals , Male , Mice , Endothelial Cells , Endothelium , Glucose , Insulin , Muscle, Skeletal , Secretome , Sirtuin 1/genetics
5.
Heart Rhythm ; 20(11): 1548-1557, 2023 11.
Article in English | MEDLINE | ID: mdl-37543305

ABSTRACT

BACKGROUND: Decreased peak sodium current (INa) and increased late sodium current (INa,L), through the cardiac sodium channel NaV1.5 encoded by SCN5A, cause arrhythmias. Many NaV1.5 posttranslational modifications have been reported. A recent report concluded that acute hypoxia increases INa,L by increasing a small ubiquitin-like modifier (SUMOylation) at K442-NaV1.5. OBJECTIVE: The purpose of this study was to determine whether and by what mechanisms SUMOylation alters INa, INa,L, and cardiac electrophysiology. METHODS: SUMOylation of NaV1.5 was detected by immunoprecipitation and immunoblotting. INa was measured by patch clamp with/without SUMO1 overexpression in HEK293 cells expressing wild-type (WT) or K442R-NaV1.5 and in neonatal rat cardiac myocytes (NRCMs). SUMOylation effects were studied in vivo by electrocardiograms and ambulatory telemetry using Scn5a heterozygous knockout (SCN5A+/-) mice and the de-SUMOylating protein SENP2 (AAV9-SENP2), AAV9-SUMO1, or the SUMOylation inhibitor anacardic acid. NaV1.5 trafficking was detected by immunofluorescence. RESULTS: NaV1.5 was SUMOylated in HEK293 cells, NRCMs, and human heart tissue. HyperSUMOylation at NaV1.5-K442 increased INa in NRCMs and in HEK cells overexpressing WT but not K442R-Nav1.5. SUMOylation did not alter other channel properties including INa,L. AAV9-SENP2 or anacardic acid decreased INa, prolonged QRS duration, and produced heart block and arrhythmias in SCN5A+/- mice, whereas AAV9-SUMO1 increased INa and shortened QRS duration. SUMO1 overexpression enhanced membrane localization of NaV1.5. CONCLUSION: SUMOylation of K442-Nav1.5 increases peak INa without changing INa,L, at least in part by altering membrane abundance. Our findings do not support SUMOylation as a mechanism for changes in INa,L. Nav1.5 SUMOylation may modify arrhythmic risk in disease states and represents a potential target for pharmacologic manipulation.


Subject(s)
Myocytes, Cardiac , Sumoylation , Animals , Humans , Mice , Rats , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/metabolism , HEK293 Cells , Myocytes, Cardiac/metabolism , NAV1.5 Voltage-Gated Sodium Channel/genetics , NAV1.5 Voltage-Gated Sodium Channel/metabolism , Sodium/metabolism , Sodium Channels/metabolism
6.
J Am Heart Assoc ; 12(6): e025368, 2023 03 21.
Article in English | MEDLINE | ID: mdl-36926933

ABSTRACT

Background Cardiac adrenergic receptor gene polymorphisms have the potential to influence risk of developing ventricular fibrillation (VF) during ST-segment-elevation myocardial infarction, but no previous study has comprehensively investigated those most likely to alter norepinephrine release, signal transduction, or biased signaling. Methods and Results In a case-control study, we recruited 953 patients with ST-segment-elevation myocardial infarction without previous cardiac history, 477 with primary VF, and 476 controls without VF, and genotyped them for ADRB1 Arg389Gly and Ser49Gly, ADRB2 Gln27Glu and Gly16Arg, and ADRA2C Ins322-325Del. Within each minor allele-containing genotype, haplotype, or 2-genotype combination, patients with incident VF were compared with non-VF controls by odds ratios (OR) of variant frequencies referenced against major allele homozygotes. Of 156 investigated genetic constructs, 19 (12.2%) exhibited significantly (P<0.05) reduced association with incident VF, and none was associated with increased VF risk except for ADRB1 Gly389 homozygotes in the subset of patients not receiving ß-blockers. ADRB1 Gly49 carriers (prevalence 23.0%) had an OR (95% CI) of 0.70 (0.49-0.98), and the ADRA2C 322-325 deletion (Del) carriers (prevalence 13.5%) had an OR of 0.61 (0.39-0.94). When present in genotype combinations (8 each), both ADRB1 Gly49 carriers (OR, 0.67 [0.56-0.80]) and ADRA2C Del carriers (OR, 0.57 [0.45- 0.71]) were associated with reduced VF risk. Conclusions In ST-segment-elevation myocardial infarction, the adrenergic receptor minor alleles ADRB1 Gly49, whose encoded receptor undergoes enhanced agonist-mediated internalization and ß-arrestin interactions leading to cardioprotective biased signaling, and ADRA2C Del322-325, whose receptor causes disinhibition of norepinephrine release, are associated with a lower incidence of VF. Registration URL: https://clinicaltrials.gov; Unique identifier: NCT00859300.


Subject(s)
ST Elevation Myocardial Infarction , Ventricular Fibrillation , Humans , Ventricular Fibrillation/diagnosis , Ventricular Fibrillation/epidemiology , Ventricular Fibrillation/genetics , Case-Control Studies , Polymorphism, Genetic , Receptors, Adrenergic/genetics , Norepinephrine
7.
J Am Heart Assoc ; 11(24): e028938, 2022 Dec 20.
Article in English | MEDLINE | ID: mdl-36515234
8.
J Am Heart Assoc ; 11(23): e027288, 2022 12 06.
Article in English | MEDLINE | ID: mdl-36453633

ABSTRACT

Background We compared cardiac outcomes for surgery-eligible patients with stage III non-small-cell lung cancer treated adjuvantly or neoadjuvantly with chemotherapy versus chemo-radiation therapy in the Surveillance, Epidemiology and End Results-Medicare database. Methods and Results Patients were age 66+, had stage IIIA/B resectable non-small-cell lung cancer diagnosed between 2007 and 2015, and received adjuvant or neoadjuvant chemotherapy or chemo-radiation within 121 days of diagnosis. Patients having chemo-radiation and chemotherapy only were propensity-score matched and followed from day 121 to first cardiac outcome, noncardiac death, radiation initiation by patients who received chemotherapy only, fee-for-service enrollment interruption, or December 31, 2016. Cause-specific hazard ratios (HRs) and competing risks subdistribution HRs were estimated. The primary outcome was the first of these severe cardiac events: acute myocardial infarction, other hospitalized ischemic heart disease, hospitalized heart failure, percutaneous coronary intervention/coronary artery bypass graft, cardiac death, or urgent/inpatient care for pericardial disease, conduction abnormality, valve disorder, or ischemic heart disease. With median follow-up of 13 months, 70 of 682 patients who received chemo-radiation (10.26%) and 43 of 682 matched patients who received chemotherapy only (6.30%) developed a severe cardiac event (P=0.008) with median time to first event 5.45 months. Chemo-radiation increased the rate of severe cardiac events (cause-specific HR: 1.62 [95% CI, 1.11-2.37] and subdistribution HR: 1.41 [95% CI, 0.97-2.04]). Cancer severity appeared greater among patients who received chemo-radiation (noncardiac death cause-specific HR, 2.53 [95% CI, 1.93-3.33] and subdistribution HR, 2.52 [95% CI, 1.90-3.33]). Conclusions Adding radiation therapy to chemotherapy is associated with an increased risk of severe cardiac events among patients with resectable stage III non-small-cell lung cancer for whom survival benefit of radiation therapy is unclear.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Cardiovascular Diseases , Lung Neoplasms , Myocardial Ischemia , United States/epidemiology , Humans , Aged , Infant , Carcinoma, Non-Small-Cell Lung/epidemiology , Carcinoma, Non-Small-Cell Lung/therapy , Medicare , Lung Neoplasms/epidemiology , Lung Neoplasms/therapy
9.
Clin Transl Med ; 12(7): e954, 2022 07.
Article in English | MEDLINE | ID: mdl-35872650

ABSTRACT

BACKGROUND: Mice with deletion of complex I subunit Ndufs4 develop mitochondrial encephalomyopathy resembling Leigh syndrome (LS). The metabolic derangement and underlying mechanisms of cardio-encephalomyopathy in LS remains incompletely understood. METHODS: We performed echocardiography, electrophysiology, confocal microscopy, metabolic and molecular/morphometric analysis of the mice lacking Ndufs4. HEK293 cells, human iPS cells-derived cardiomyocytes and neurons were used to determine the mechanistic role of mitochondrial complex I deficiency. RESULTS: LS mice develop severe cardiac bradyarrhythmia and diastolic dysfunction. Human-induced pluripotent stem cell-derived cardiomyocytes (iPS-CMs) with Ndufs4 deletion recapitulate LS cardiomyopathy. Mechanistically, we demonstrate a direct link between complex I deficiency, decreased intracellular (nicotinamide adenine dinucleotide) NAD+ /NADH and bradyarrhythmia, mediated by hyperacetylation of the cardiac sodium channel NaV 1.5, particularly at K1479 site. Neuronal apoptosis in the cerebellar and midbrain regions in LS mice was associated with hyperacetylation of p53 and activation of microglia. Targeted metabolomics revealed increases in several amino acids and citric acid cycle intermediates, likely due to impairment of NAD+ -dependent dehydrogenases, and a substantial decrease in reduced Glutathione (GSH). Metabolic rescue by nicotinamide riboside (NR) supplementation increased intracellular NAD+ / NADH, restored metabolic derangement, reversed protein hyperacetylation through NAD+ -dependent Sirtuin deacetylase, and ameliorated cardiomyopathic phenotypes, concomitant with improvement of NaV 1.5 current and SERCA2a function measured by Ca2+ -transients. NR also attenuated neuronal apoptosis and microglial activation in the LS brain and human iPS-derived neurons with Ndufs4 deletion. CONCLUSIONS: Our study reveals direct mechanistic explanations of the observed cardiac bradyarrhythmia, diastolic dysfunction and neuronal apoptosis in mouse and human induced pluripotent stem cells (iPSC) models of LS.


Subject(s)
Cardiomyopathies , Induced Pluripotent Stem Cells , Leigh Disease , Animals , Bradycardia/metabolism , Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Electron Transport Complex I/deficiency , Electron Transport Complex I/metabolism , HEK293 Cells , Humans , Induced Pluripotent Stem Cells/metabolism , Leigh Disease/genetics , Leigh Disease/metabolism , Mice , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Diseases , NAD/metabolism
10.
J Am Heart Assoc ; 11(13): e025687, 2022 07 05.
Article in English | MEDLINE | ID: mdl-35730644

ABSTRACT

Background Sorbin and SH3 domain containing 2 (Sorbs2) protein is a cytoskeletal adaptor with an emerging role in cardiac biology and disease; yet, its potential relevance to adult-onset cardiomyopathies remains underexplored. Sorbs2 global knockout mice display lethal arrhythmogenic cardiomyopathy; however, the causative mechanisms remain unclear. Herein, we examine Sorbs2 dysregulation in heart failure, characterize novel Sorbs2 cardiomyocyte-specific knockout mice (Sorbs2-cKO), and explore associations between Sorbs2 genetic variations and human cardiovascular disease. Methods and Results Bioinformatic analyses show myocardial Sorbs2 mRNA is consistently upregulated in humans with adult-onset cardiomyopathies and in heart failure models. We generated Sorbs2-cKO mice and report that they develop progressive systolic dysfunction and enlarged cardiac chambers, and they die with congestive heart failure at about 1 year old. After 3 months, Sorbs2-cKO mice begin to show atrial enlargement and P-wave anomalies, without dysregulation of action potential-associated ion channel and gap junction protein expressions. After 6 months, Sorbs2-cKO mice exhibit impaired contractility in dobutamine-treated hearts and skinned myofibers, without dysregulation of contractile protein expressions. From our comprehensive survey of potential mechanisms, we found that within 4 months, Sorbs2-cKO hearts have defective microtubule polymerization and compensatory upregulation of structural cytoskeletal and adapter proteins, suggesting that this early intracellular structural remodeling is responsible for contractile dysfunction. Finally, we identified genetic variants that associate with decreased Sorbs2 expression and human cardiac phenotypes, including conduction abnormalities, atrial enlargement, and dilated cardiomyopathy, consistent with Sorbs2-cKO mice phenotypes. Conclusions Our studies show that Sorbs2 is essential for maintaining structural integrity in cardiomyocytes, likely through strengthening the interactions between microtubules and other cytoskeletal proteins at cross-link sites.


Subject(s)
Adaptor Proteins, Signal Transducing , Cardiomyopathy, Dilated , Heart Failure , RNA-Binding Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adult , Animals , Disease Models, Animal , Humans , Infant , Mice , Mice, Knockout , Myocytes, Cardiac/metabolism , RNA-Binding Proteins/genetics , src Homology Domains
11.
Clin Transl Med ; 12(1): e693, 2022 01.
Article in English | MEDLINE | ID: mdl-35060347

ABSTRACT

BACKGROUND: MicroRNAs regulate cardiac hypertrophy development, which precedes and predicts the risk of heart failure. microRNA-204-5p (miR-204) is well expressed in cardiomyocytes, but its role in developing cardiac hypertrophy and cardiac dysfunction (CH/CD) remains poorly understood. METHODS: We performed RNA-sequencing, echocardiographic, and molecular/morphometric analysis of the heart of mice lacking or overexpressing miR-204 five weeks after trans-aortic constriction (TAC). The neonatal rat cardiomyocytes, H9C2, and HEK293 cells were used to determine the mechanistic role of miR-204. RESULTS: The stretch induces miR-204 expression, and miR-204 inhibits the stretch-induced hypertrophic response of H9C2 cells. The mice lacking miR-204 displayed a higher susceptibility to CH/CD during pressure overload, which was reversed by the adeno-associated virus serotype-9-mediated cardioselective miR-204 overexpression. Bioinformatic analysis of the cardiac transcriptomics of miR-204 knockout mice following pressure overload suggested deregulation of apelin-receptor (APJ) signalling. We found that the stretch-induced extracellular signal-regulated kinase 1/2 (ERK1/2) activation and hypertrophy-related genes expression depend on the APJ, and both of these effects are subject to miR-204 levels. The dynamin inhibitor dynasore inhibited both stretch-induced APJ endocytosis and ERK1/2 activation. In contrast, the miR-204-induced APJ endocytosis was neither inhibited by dynamin inhibitors (dynasore and dyngo) nor associated with ERK1/2 activation. We find that the miR-204 increases the expression of ras-associated binding proteins (e.g., Rab5a, Rab7) that regulate cellular endocytosis. CONCLUSIONS: Our results show that miR-204 regulates trafficking of APJ and confers resistance to pressure overload-induced CH/CD, and boosting miR-204 can inhibit the development of CH/CD.


Subject(s)
Apelin Receptors/antagonists & inhibitors , Cardiomegaly/prevention & control , MicroRNAs/pharmacology , Animals , Apelin Receptors/metabolism , Cardiomegaly/drug therapy , Disease Models, Animal , Heart Diseases/drug therapy , Heart Diseases/prevention & control , MicroRNAs/metabolism , Rats , Signal Transduction/drug effects
12.
J Am Heart Assoc ; 10(15): e021475, 2021 08 03.
Article in English | MEDLINE | ID: mdl-34278832

Subject(s)
Peer Review , Publishing
14.
J Am Heart Assoc ; 9(20): e019307, 2020 10 20.
Article in English | MEDLINE | ID: mdl-32941090

ABSTRACT

We, the Editors of the Journal of the American Heart Association, sincerely regret the publication of the article "Diversity, Inclusion, and Equity: Evolution of Race and Ethnicity Considerations for the Cardiology Workforce in the United States of America From 1969 to 2019".1 We are aware that the publication of this flawed and biased article has caused a great deal of unnecessary pain and anguish to a number of parties, and reflects extremely poorly on us. We fully support the retraction of this article.

15.
J Am Heart Assoc ; 9(17): e014592, 2020 09.
Article in English | MEDLINE | ID: mdl-32805189
16.
J Am Heart Assoc ; 9(12): e017013, 2020 06 16.
Article in English | MEDLINE | ID: mdl-32347144

ABSTRACT

Coronavirus Disease 2019 (COVID-19) has infected more than 3.0 million people worldwide and killed more than 200,000 as of April 27, 2020. In this White Paper, we address the cardiovascular co-morbidities of COVID-19 infection; the diagnosis and treatment of standard cardiovascular conditions during the pandemic; and the diagnosis and treatment of the cardiovascular consequences of COVID-19 infection. In addition, we will also address various issues related to the safety of healthcare workers and the ethical issues related to patient care in this pandemic.


Subject(s)
Betacoronavirus , Cardiovascular Diseases/epidemiology , Coronavirus Infections/epidemiology , Pandemics , Pneumonia, Viral/epidemiology , COVID-19 , Comorbidity , Global Health , Humans , Incidence , SARS-CoV-2
17.
J Neuroimmunol ; 343: 577227, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32247877

ABSTRACT

During neuroinflammation, the shaker type potassium channel Kv1.4 is re-expressed in oligodendrocytes (Ol), but not immune cells. Here, we analyze the role of endogenous Kv1.4 in two demyelinating animal models of multiple sclerosis. While Kv1.4 deficiency in primary murine Ol led to a decreased proliferation rate in vitro, it did not exert an effect on Ol proliferation or on the extent of de- or remyelination in the cuprizone model in vivo. However, in experimental autoimmune encephalomyelitis, Kv1.4-/- mice exhibited a milder disease course and reduced Th1 responses. These data argue for an indirect effect of Kv1.4 on immune cells, possibly via glial cells.


Subject(s)
Demyelinating Diseases/metabolism , Kv1.4 Potassium Channel/metabolism , Remyelination/physiology , Animals , Cell Proliferation/physiology , Chelating Agents/toxicity , Cuprizone/toxicity , Demyelinating Diseases/chemically induced , Demyelinating Diseases/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligodendroglia/metabolism , Th1 Cells/immunology
18.
J Mol Cell Cardiol ; 141: 70-81, 2020 04.
Article in English | MEDLINE | ID: mdl-32209328

ABSTRACT

RATIONALE: The cardiac sodium channel NaV1.5, encoded by SCN5A, produces the rapidly inactivating depolarizing current INa that is responsible for the initiation and propagation of the cardiac action potential. Acquired and inherited dysfunction of NaV1.5 results in either decreased peak INa or increased residual late INa (INa,L), leading to tachy/bradyarrhythmias and sudden cardiac death. Previous studies have shown that increased cellular NAD+ and NAD+/NADH ratio increase INa through suppression of mitochondrial reactive oxygen species and PKC-mediated NaV1.5 phosphorylation. In addition, NAD+-dependent deacetylation of NaV1.5 at K1479 by Sirtuin 1 increases NaV1.5 membrane trafficking and INa. The role of NAD+ precursors in modulating INa remains unknown. OBJECTIVE: To determine whether and by which mechanisms the NAD+ precursors nicotinamide riboside (NR) and nicotinamide (NAM) affect peak INa and INa,Lin vitro and cardiac electrophysiology in vivo. METHODS AND RESULTS: The effects of NAD+ precursors on the NAD+ metabolome and electrophysiology were studied using HEK293 cells expressing wild-type and mutant NaV1.5, rat neonatal cardiomyocytes (RNCMs), and mice. NR increased INa in HEK293 cells expressing NaV1.5 (500 µM: 51 ± 18%, p = .02, 5 mM: 59 ± 22%, p = .03) and RNCMs (500 µM: 60 ± 26%, p = .02, 5 mM: 74 ± 39%, p = .03) while reducing INa,L at the higher concentration (RNCMs, 5 mM: -45 ± 11%, p = .04). NR (5 mM) decreased NaV1.5 K1479 acetylation but increased INa in HEK293 cells expressing a mutant form of NaV1.5 with disruption of the acetylation site (NaV1.5-K1479A). Disruption of the PKC phosphorylation site abolished the effect of NR on INa. Furthermore, NAM (5 mM) had no effect on INa in RNCMs or in HEK293 cells expressing wild-type NaV1.5, but increased INa in HEK293 cells expressing NaV1.5-K1479A. Dietary supplementation with NR for 10-12 weeks decreased QTc in C57BL/6 J mice (0.35% NR: -4.9 ± 2.0%, p = .14; 1.0% NR: -9.5 ± 2.8%, p = .01). CONCLUSIONS: NAD+ precursors differentially regulate NaV1.5 via multiple mechanisms. NR increases INa, decreases INa,L, and warrants further investigation as a potential therapy for arrhythmic disorders caused by NaV1.5 deficiency and/or dysfunction.


Subject(s)
Ion Channel Gating , Myocardium/metabolism , NAD/metabolism , NAV1.5 Voltage-Gated Sodium Channel/metabolism , Acetylation/drug effects , Animals , Dietary Supplements , HEK293 Cells , Humans , Ion Channel Gating/drug effects , Lysine/metabolism , Metabolome , Mice, Inbred C57BL , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Niacinamide/analogs & derivatives , Niacinamide/chemistry , Niacinamide/pharmacology , Phosphorylation/drug effects , Pyridinium Compounds/chemistry , Pyridinium Compounds/pharmacology , Rats, Sprague-Dawley
20.
J Am Heart Assoc ; 8(21): e013685, 2019 11 05.
Article in English | MEDLINE | ID: mdl-31668118

ABSTRACT

Background Contemporary outcomes of transcatheter aortic valve replacement (TAVR) in nonagenarians are unknown. Methods and Results We identified 13 544 nonagenarians (aged 90-100 years) who underwent TAVR between 2012 and 2016 using Medicare claims. Generalized estimating equations were used to study the change in short-term outcomes among nonagenarians over time. We compared outcomes between nonagenarians and non-nonagenarians undergoing TAVR in 2016. A mixed-effect multivariable logistic regression was performed to determine predictors of 30-day mortality in nonagenarians in 2016. A center was defined as a high-volume center if it performed ≥100 TAVR procedures per year. After adjusting for changes in patients' characteristics, risk-adjusted 30-day mortality declined in nonagenarians from 9.8% in 2012 to 4.4% in 2016 (P<0.001), whereas mortality for patients <90 years decreased from 6.4% to 3.5%. In 2016, 35 712 TAVR procedures were performed, of which 12.7% were in nonagenarians. Overall, in-hospital mortality in 2016 was higher in nonagenarians compared with younger patients (2.4% versus 1.7%, P<0.05) but did not differ in analysis limited to high-volume centers (2.2% versus 1.7%; odds ratio: 1.33; 95% CI, 0.97-1.81; P=0.07). Important predictors of 30-day mortality in nonagenarians included in-hospital stroke (adjusted odds ratio [aOR]: 8.67; 95% CI, 5.03-15.00), acute kidney injury (aOR: 4.11; 95% CI, 2.90-5.83), blood transfusion (aOR: 2.66; 95% CI, 1.81-3.90), respiratory complications (aOR: 2.96; 95% CI, 1.52-5.76), heart failure (aOR: 1.86; 95% CI, 1.04-3.34), coagulopathy (aOR: 1.59; 95% CI, 1.12-2.26; P<0.05 for all). Conclusions Short-term outcomes after TAVR have improved in nonagenarians. Several procedural complications were associated with increased 30-day mortality among nonagenarians.


Subject(s)
Transcatheter Aortic Valve Replacement , Age Factors , Aged , Aged, 80 and over , Female , Humans , Male , Retrospective Studies , Time Factors , Transcatheter Aortic Valve Replacement/mortality , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...