Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
1.
J. card. fail ; 30(1): 134-134, jan. 2024.
Artigo em Inglês | CONASS, Sec. Est. Saúde SP, SESSP-IDPCPROD, Sec. Est. Saúde SP | ID: biblio-1532023

RESUMO

INTRODUCTION APOLLO-B is a Phase 3 study of patisiran in patients with transthyretin (ATTR) cardiac amyloidosis (NCT03997383), which demonstrated a significant benefit in functional capacity (6-MWT), and health status and quality of life (QoL) (KCCQ-OS) with patisiran vs placebo at Month (M) 12. HYPOTHESIS Patisiran improves health status and QoL in the daily lives of patients with ATTR cardiac amyloidosis vs placebo. METHODS Patients were 18-85 years old with ATTR amyloidosis and a medical history of heart failure (HF) due to ATTR cardiomyopathy, with ≥1 prior hospitalization for HF or current clinical evidence of HF. Patients were randomized (1:1) to intravenous patisiran 0.3 mg/kg or placebo every 3 weeks. These post-hoc analyses evaluated percentage of responders reporting ≥5-point improvement in KCCQ-OS, and change from baseline in 4 KCCQ domains and questions within the domains. RESULTS 359 patients received study drug (patisiran, N=181; placebo, N=178): median age (range), 76 (41, 85) years; male, 89%; wild-type ATTR, 80%; 25% were on tafamidis at baseline. At M12, patisiran showed significant benefit vs placebo in KCCQ-OS (LS mean [SEM] change from baseline: patisiran, 0.30 [1.26]; placebo, -3.41 [1.28]; LS mean [SEM] difference: 3.71 [1.80]; p=0.0397). A ≥ 5-point improvement in KCCQ at M12 was more frequent with patisiran vs placebo (34.1 vs 24.0%: difference [95% CI] 10.1% [0.7, 19.5]). Improvement vs placebo was consistent across domains, with LS mean differences [95% CI] in change from baseline (patisiran - placebo) in Physical Limitations (2.75 [-1.24, 6.74]), Total Symptoms (4.55 [0.75, 8.34]), QoL (4.27 [-0.12, 8.65]), and Social Limitations (2.76 [-2.21, 7.73]). Categorical changes from baseline to M12 demonstrated greater percentages of placebo-treated patients reporting worsening for questions in each domain, including activities requiring greater cardiometabolic demand. In patients with values at baseline and M12, notably greater percentages (>5%) of placebo- vs patisiran-treated patients reported worsening (percent difference; n=placebo/patisiran) for questions related to Walking 1 Block on Level Ground (10%; n=159/162), Frequency and Burden of Dyspnea (9.5% and 7.6%; n=164/170), Frequency of Orthopnea (9.6%; n=163/170), Feeling about Spending the Rest of Their Life with HF the Way It Is Right Now (6.4%; n=164/170), and Intimate Relationships (6.3%; n=88/86). Improvement from baseline was reported by greater percentages (>5%) of patisiran-treated patients (percent difference; n=patisiran/placebo) in Enjoyment of Life Limited Due to HF (12.8%; n=170/164) and Hobbies/Recreational Activities (6.0%; n=141/143). CONCLUSIONS In APOLLO-B, improvements in health status and QoL with patisiran vs placebo were apparent across all 4 KCCQ domains. Greater percentages of patisiran-treated patients had KCCQ-OS improved by ≥ 5 points at M12 and they more often reported improvements in QoL, and ability to enjoy life and perform hobbies/recreational activities. More placebo-treated patients reported worsening in walking on level ground, HF symptoms and QoL.


Assuntos
Qualidade de Vida , Pré-Albumina
2.
Circulation ; 148(Suppl.1)Nov. 7, 2023.
Artigo em Inglês | CONASS, Sec. Est. Saúde SP, SESSP-IDPCPROD, Sec. Est. Saúde SP | ID: biblio-1519637

RESUMO

INTRODUCTION: The Phase 3 APOLLO-B study evaluates patisiran in patients (pts) with transthyretin (ATTR) cardiac amyloidosis over a 12-month (M) double-blind (DB) period, followed by an open-label extension (OLE) period when all pts receive patisiran (NCT03997383). Hypothesis: Patisiran provides long-term benefit in pts with ATTR cardiac amyloidosis. Aims: Describe safety and efficacy of patisiran during the APOLLO-B OLE (18M+). METHODS: Pts (18-85 yrs) with ATTR cardiac amyloidosis and heart failure history were randomized 1:1 to patisiran or placebo (pbo). Pts completing DB period were eligible to receive patisiran in the OLE for ≤36M. Results summarized based on DB treatment arm. Exploratory assessments include change from study baseline (CFB) in 6-minute walk test (6MWT), KCCQ-OS, NT-proBNP, and troponin I. RESULTS: In the DB period, 359 pts (pbo n=178; patisiran n=181) received study drug (median [range] age, 76.0 [41, 85] yrs; male, 89%; wtATTR, 80%; tafamidis at baseline, 25%); 334 (93%) entered the OLE. In patisiran arm, M12 and M18 results, respectively, were similar for each endpoint: 6MWT and KCCQ-OS (mean [SEM] CFB) −8.09 [5.73] vs −9.21 [6.04] meters (m) and 0.60 [1.36] vs 0.22 [1.48]; NT-proBNP and troponin I (geometric mean fold-CFB [95%CI]) 1.10 [1.03, 1.17] vs 1.17 [1.07, 1.27] and 1.11 [1.05, 1.18] vs 1.09 [1.01, 1.17]). In pbo arm, patisiran initiation in OLE was associated with a slower rate of worsening or relative stability across endpoints; CFB at M12 vs M18, respectively: 6MWT, −25.43 [5.61] vs −31.08 [5.45] m; KCCQ-OS, −3.41 [1.33] vs −4.02 [1.49]; NT-proBNP, 1.39 [1.28, 1.51] vs 1.53 [1.38, 1.71]; and troponin I, 1.29 [1.21, 1.38] vs 1.21 [1.13, 1.30]. Patisiran had an acceptable safety profile; no new concerns. OLE analyses are ongoing; updated data to be presented. CONCLUSIONS: The M18 results provide evidence that beneficial effects observed in DB period on functional capacity, health status, and quality of life were maintained by continued treatment with patisiran during the OLE. Pbo-treated pts initiating patisiran at M12 showed slowed worsening or stabilization in most endpoints at M18. Early treatment initiation is important: pbo-treated pts did not recover functional capacity or health lost prior to initiating OLE patisiran.

3.
JCI Insight ; 8(22)2023 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-37815863

RESUMO

Ventricular arrhythmias (VAs) in heart failure are enhanced by sympathoexcitation. However, radiotracer studies of catecholamine uptake in failing human hearts demonstrate a proclivity for VAs in patients with reduced cardiac sympathetic innervation. We hypothesized that this counterintuitive finding is explained by heterogeneous loss of sympathetic nerves in the failing heart. In a murine model of dilated cardiomyopathy (DCM), delayed PET imaging of sympathetic nerve density using the catecholamine analog [11C]meta-Hydroxyephedrine demonstrated global hypoinnervation in ventricular myocardium. Although reduced, sympathetic innervation in 2 distinct DCM models invariably exhibited transmural (epicardial to endocardial) gradients, with the endocardium being devoid of sympathetic nerve fibers versus controls. Further, the severity of transmural innervation gradients was correlated with VAs. Transmural innervation gradients were also identified in human left ventricular free wall samples from DCM versus controls. We investigated mechanisms underlying this relationship by in silico studies in 1D, 2D, and 3D models of failing and normal human hearts, finding that arrhythmogenesis increased as heterogeneity in sympathetic innervation worsened. Specifically, both DCM-induced myocyte electrical remodeling and spatially inhomogeneous innervation gradients synergistically worsened arrhythmogenesis. Thus, heterogeneous innervation gradients in DCM promoted arrhythmogenesis. Restoration of homogeneous sympathetic innervation in the failing heart may reduce VAs.


Assuntos
Cardiomiopatia Dilatada , Humanos , Camundongos , Animais , Cardiomiopatia Dilatada/diagnóstico por imagem , Coração , Miocárdio , Arritmias Cardíacas/diagnóstico por imagem , Catecolaminas
4.
Biology (Basel) ; 12(10)2023 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-37887000

RESUMO

About 15% of congenital heart disease (CHD) patients have a known pathogenic copy number variant. The majority of their chromosomal microarray (CMA) tests are deemed normal. Diagnostic interpretation typically ignores microdeletions smaller than 100 kb. We hypothesized that unreported microdeletions are enriched for CHD genes. We analyzed "normal" CMAs of 1762 patients who were evaluated at a pediatric referral center, of which 319 (18%) had CHD. Using CMAs from monozygotic twins or replicates from the same individual, we established a size threshold based on probe count for the reproducible detection of small microdeletions. Genes in the microdeletions were sequentially filtered by their nominal association with a CHD diagnosis, the expression level in the fetal heart, and the deleteriousness of a loss-of-function mutation. The subsequent enrichment for CHD genes was assessed using the presence of known or potentially novel genes implicated by a large whole-exome sequencing study of CHD. The unreported microdeletions were modestly enriched for both known CHD genes and those of unknown significance identified using their de novo mutation in CHD patients. Our results show that readily available "normal" CMA data can be a fruitful resource for genetic discovery and that smaller deletions should receive more attention in clinical evaluation.

5.
N Engl J Med ; 389(17): 1553-1565, 2023 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-37888916

RESUMO

BACKGROUND: Transthyretin amyloidosis, also called ATTR amyloidosis, is associated with accumulation of ATTR amyloid deposits in the heart and commonly manifests as progressive cardiomyopathy. Patisiran, an RNA interference therapeutic agent, inhibits the production of hepatic transthyretin. METHODS: In this phase 3, double-blind, randomized trial, we assigned patients with hereditary, also known as variant, or wild-type ATTR cardiac amyloidosis, in a 1:1 ratio, to receive patisiran (0.3 mg per kilogram of body weight) or placebo once every 3 weeks for 12 months. A hierarchical procedure was used to test the primary and three secondary end points. The primary end point was the change from baseline in the distance covered on the 6-minute walk test at 12 months. The first secondary end point was the change from baseline to month 12 in the Kansas City Cardiomyopathy Questionnaire-Overall Summary (KCCQ-OS) score (with higher scores indicating better health status). The second secondary end point was a composite of death from any cause, cardiovascular events, and change from baseline in the 6-minute walk test distance over 12 months. The third secondary end point was a composite of death from any cause, hospitalizations for any cause, and urgent heart failure visits over 12 months. RESULTS: A total of 360 patients were randomly assigned to receive patisiran (181 patients) or placebo (179 patients). At month 12, the decline in the 6-minute walk distance was lower in the patisiran group than in the placebo group (Hodges-Lehmann estimate of median difference, 14.69 m; 95% confidence interval [CI], 0.69 to 28.69; P = 0.02); the KCCQ-OS score increased in the patisiran group and declined in the placebo group (least-squares mean difference, 3.7 points; 95% CI, 0.2 to 7.2; P = 0.04). Significant benefits were not observed for the second secondary end point. Infusion-related reactions, arthralgia, and muscle spasms occurred more often among patients in the patisiran group than among those in the placebo group. CONCLUSIONS: In this trial, administration of patisiran over a period of 12 months resulted in preserved functional capacity in patients with ATTR cardiac amyloidosis. (Funded by Alnylam Pharmaceuticals; APOLLO-B ClinicalTrials.gov number, NCT03997383.).


Assuntos
Amiloidose , Cardiomiopatias , Pré-Albumina , RNA Interferente Pequeno , Humanos , Cardiomiopatias/tratamento farmacológico , Cardiomiopatias/etiologia , Cardiomiopatias/genética , Cardiomiopatias/metabolismo , Pré-Albumina/genética , Pré-Albumina/metabolismo , RNA Interferente Pequeno/uso terapêutico , Amiloidose Familiar/complicações , Amiloidose Familiar/tratamento farmacológico , Amiloidose Familiar/genética , Fígado/metabolismo , Método Duplo-Cego , Amiloidose/complicações , Amiloidose/tratamento farmacológico , Amiloidose/genética
6.
Clin Pharmacokinet ; 62(10): 1509-1522, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37639169

RESUMO

BACKGROUND AND OBJECTIVE: Variants of the transthyretin (TTR) gene cause hereditary transthyretin-mediated (hATTR) amyloidosis, or ATTRv amyloidosis (v for variant), which results from deposition of misfolded TTR protein as amyloid in organs and tissues. Patisiran is an RNA interference (RNAi) therapeutic that suppresses the hepatic production of TTR protein. Patisiran improves multiple clinical manifestations of hATTR amyloidosis in patients without liver transplantation (LT). Because the liver is the predominant source of circulating TTR, LT has been prescribed to eliminate the production of the variant TTR. However, the continued production of wild-type TTR can contribute to disease progression after LT. Patisiran could potentially address an unmet need in these affected patients. This clinical trial was conducted to evaluate the safety, efficacy, and pharmacokinetics (PK) and pharmacodynamics (PD) of patisiran in patients with hATTR amyloidosis with polyneuropathy progression after LT. In this paper, we describe the PK/PD of patisiran in post-LT patients and compare it with prior patisiran studies in healthy subjects and patients without LT. METHODS: In an open-label study, patients (N = 23) with hATTR amyloidosis with polyneuropathy progression after LT received 0.3 mg/kg patisiran intravenously every 3 weeks (q3w) for 12 months. As a post hoc analysis, the PK and PD results from the current study were compared with prior patisiran studies in healthy volunteers from a Phase 1 study and in patients with hATTR amyloidosis without LT from Phase 2 and 3 studies. RESULTS: The PK profile of patisiran siRNA (ALN-18328) and its 2 lipid excipients, DLin-MC3-DMA and PEG2000-C-DMG, in hATTR amyloidosis patients after LT was consistent with prior patisiran studies in non-LT subjects. Plasma PK profiles of ALN-18328 and DLin-MC3-DMA exhibited 2 phases, the first characterized by a short distribution half-life and the second by a minor peak and relatively long elimination half-life. The plasma concentrations of PEG2000-C-DMG reached Cmax at the end of infusion and declined in a multiphasic manner. There was no appreciable accumulation at steady state. Consistent with prior studies in non-LT subjects, the post-LT patients showed a robust, and sustained TTR reduction; with median TTR reduction from baseline of 91% (average of Month 6 and Month 12). No anti-drug antibodies were observed in any patient. CONCLUSIONS: The consistency of patisiran PK and PD between patients with and without LT suggests that neither LT nor concomitantly administered immunosuppressants influence hepatic uptake or RNAi activity of patisiran. The patisiran dosing regimen of 0.3 mg/kg q3w is appropriate for hATTR amyloidosis patients with or without LT. CLINICAL TRIAL REGISTRATION NO: NCT03862807.


Assuntos
Amiloidose , Transplante de Fígado , Humanos , Transplante de Fígado/efeitos adversos , Pré-Albumina/genética , RNA Interferente Pequeno
7.
Elife ; 122023 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-37162194

RESUMO

The cell bodies of postganglionic sympathetic neurons innervating the heart primarily reside in the stellate ganglion (SG), alongside neurons innervating other organs and tissues. Whether cardiac-innervating stellate ganglionic neurons (SGNs) exhibit diversity and distinction from those innervating other tissues is not known. To identify and resolve the transcriptomic profiles of SGNs innervating the heart, we leveraged retrograde tracing techniques using adeno-associated virus (AAV) expressing fluorescent proteins (GFP or Td-tomato) with single cell RNA sequencing. We investigated electrophysiologic, morphologic, and physiologic roles for subsets of cardiac-specific neurons and found that three of five adrenergic SGN subtypes innervate the heart. These three subtypes stratify into two subpopulations; high (NA1a) and low (NA1b and NA1c) neuropeptide-Y (NPY) -expressing cells, exhibit distinct morphological, neurochemical, and electrophysiologic characteristics. In physiologic studies in transgenic mouse models modulating NPY signaling, we identified differential control of cardiac responses by these two subpopulations to high and low stress states. These findings provide novel insights into the unique properties of neurons responsible for cardiac sympathetic regulation, with implications for novel strategies to target specific neuronal subtypes for sympathetic blockade in cardiac disease.


Assuntos
Neurônios , Gânglio Estrelado , Camundongos , Animais , Neurônios/metabolismo , Gânglio Estrelado/metabolismo , Coração , Neuropeptídeo Y/metabolismo , Perfilação da Expressão Gênica
8.
bioRxiv ; 2023 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-36711942

RESUMO

The cell bodies of postganglionic sympathetic neurons innervating the heart primarily reside in the stellate ganglion (SG), alongside neurons innervating other organs and tissues. Whether cardiac-innervating stellate ganglionic neurons (SGNs) exhibit diversity and distinction from those innervating other tissues is not known. To identify and resolve the transcriptomic profiles of SGNs innervating the heart we leveraged retrograde tracing techniques using adeno-associated virus (AAV) expressing fluorescent proteins (GFP or Td-tomato) with single cell RNA sequencing. We investigated electrophysiologic, morphologic, and physiologic roles for subsets of cardiac-specific neurons and found that three of five adrenergic SGN subtypes innervate the heart. These three subtypes stratify into two subpopulations; high (NA1a) and low (NA1b and NA1c) Npy-expressing cells, exhibit distinct morphological, neurochemical, and electrophysiologic characteristics. In physiologic studies in transgenic mouse models modulating NPY signaling, we identified differential control of cardiac responses by these two subpopulations to high and low stress states. These findings provide novel insights into the unique properties of neurons responsible for cardiac sympathetic regulation, with implications for novel strategies to target specific neuronal subtypes for sympathetic blockade in cardiac disease.

9.
Cureus ; 14(10): e29994, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-36238417

RESUMO

Isolated congenital aneurysm of the muscular interventricular septum is rare. We present a patient with congenital aneurysm of the basal muscular ventricular septum, who also develops conduction abnormalities with first-degree heart block, right bundle branch block, and left posterior fascicular block. The case details the natural history of the aneurysm over a 10-year period follow-up during which the patient remained asymptomatic with evidence of regression of the aneurysm. Given the aneurysm's location close to the proximal right bundle and left posterior fascicle, we believe that the cause for the aneurysm also injured both fascicles resulting in bifascicular block. The diagnosis of bifascicular block was confirmed using the electrocardiogram-derived vectorcardiography loops. These conduction abnormalities have remained stable. The case illustrates the utility of vectorcardiography in diagnosing bundle branch conduction defects. The case also illustrates the importance of anatomical considerations when encountering congenital heart defects.

10.
Int. j. cardiovasc. sci. (Impr.) ; 35(5): 665-675, Sept.-Oct. 2022. tab, graf
Artigo em Inglês | LILACS-Express | LILACS | ID: biblio-1405193

RESUMO

Abstract Therapeutics that inhibit enzymes, receptors, ion channels, and cotransporters have long been the mainstay of cardiovascular medicine. Now, oligonucleotide therapeutics offer a modern variation on this paradigm of protein inhibition. Rather than target a protein, however, small interfering ribonucleic acids and antisense oligonucleotides target the messenger RNA (mRNA) from which a protein is translated. Endogenous, cellular mechanisms enable the oligonucleotides to bind a selected sequence on a target mRNA, leading to its degradation. The catalytic nature of the process confers an advantage over the stoichiometric binding of traditional small molecule therapeutics to their respective protein targets. Advances in nucleic acid chemistry and delivery have enabled development of oligonucleotide therapeutics against a wide range of diseases, including hyperlipidemias and hereditary transthyretin-mediated amyloidosis with polyneuropathy. While most of these therapeutics were initially designed for rare diseases, recent clinical trials highlight the potential impact of oligonucleotides on more common forms of cardiovascular disease.

11.
Am J Transplant ; 22(6): 1646-1657, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35213769

RESUMO

Hereditary transthyretin-mediated (hATTR) amyloidosis, or ATTRv amyloidosis, is a progressive disease, for which liver transplantation (LT) has been a long-standing treatment. However, disease progression continues post-LT. This Phase 3b, open-label trial evaluated efficacy and safety of patisiran in patients with ATTRv amyloidosis with polyneuropathy progression post-LT. Primary endpoint was median transthyretin (TTR) reduction from baseline. Twenty-three patients received patisiran for 12 months alongside immunosuppression regimens. Patisiran elicited a rapid, sustained TTR reduction (median reduction [Months 6 and 12 average], 91.0%; 95% CI: 86.1%-92.3%); improved neuropathy, quality of life, and autonomic symptoms from baseline to Month 12 (mean change [SEM], Neuropathy Impairment Score, -3.7 [2.7]; Norfolk Quality of Life-Diabetic Neuropathy questionnaire, -6.5 [4.9]; least-squares mean [SEM], Composite Autonomic Symptom Score-31, -5.0 [2.6]); and stabilized disability (Rasch-built Overall Disability Scale) and nutritional status (modified body mass index). Adverse events were mild or moderate; five patients experienced ≥1 serious adverse event. Most patients had normal liver function tests. One patient experienced transplant rejection consistent with inadequate immunosuppression, remained on patisiran, and completed the study. In conclusion, patisiran reduced serum TTR, was well tolerated, and improved or stabilized key disease impairment measures in patients with ATTRv amyloidosis with polyneuropathy progression post-LT (www.clinicaltrials.gov NCT03862807).


Assuntos
Neuropatias Amiloides Familiares , Transplante de Fígado , Polineuropatias , Neuropatias Amiloides Familiares/complicações , Neuropatias Amiloides Familiares/tratamento farmacológico , Neuropatias Amiloides Familiares/cirurgia , Humanos , Polineuropatias/tratamento farmacológico , Polineuropatias/etiologia , Pré-Albumina/uso terapêutico , Qualidade de Vida , RNA Interferente Pequeno
12.
Genes (Basel) ; 12(9)2021 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-34573350

RESUMO

In newborns, severe congenital heart defects are rarer than mild ones. This epidemiological relationship between heart defect severity and incidence lacks explanation. Here, an analysis of ~10,000 Nkx2-5+/- mice from two inbred strain crosses illustrates the fundamental role of epistasis. Modifier genes raise or lower the risk of specific defects via pairwise (G×GNkx) and higher-order (G×G×GNkx) interactions with Nkx2-5. Their effect sizes correlate with the severity of a defect. The risk loci for mild, atrial septal defects exert predominantly small G×GNkx effects, while the loci for severe, atrioventricular septal defects exert large G×GNkx and G×G×GNkx effects. The loci for moderately severe ventricular septal defects have intermediate effects. Interestingly, G×G×GNkx effects are three times more likely to suppress risk when the genotypes at the first two loci are from the same rather than different parental inbred strains. This suggests the genetic coadaptation of interacting G×G×GNkx loci, a phenomenon that Dobzhansky first described in Drosophila. Thus, epistasis plays dual roles in the pathogenesis of congenital heart disease and the robustness of cardiac development. The empirical results suggest a relationship between the fitness cost and genetic architecture of a disease phenotype and a means for phenotypic robustness to have evolved.


Assuntos
Aptidão Genética , Comunicação Interatrial/genética , Comunicação Interventricular/genética , Defeitos dos Septos Cardíacos/genética , Proteína Homeobox Nkx-2.5/genética , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Feminino , Loci Gênicos , Defeitos dos Septos Cardíacos/diagnóstico , Comunicação Interatrial/diagnóstico , Comunicação Interventricular/diagnóstico , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Índice de Gravidade de Doença
15.
Sci Rep ; 8(1): 6475, 2018 04 24.
Artigo em Inglês | MEDLINE | ID: mdl-29691457

RESUMO

GLUT transgenic and knockout mice have provided valuable insight into the role of facilitative glucose transporters (GLUTs) in cardiovascular and metabolic disease, but compensatory physiological changes can hinder interpretation of these models. To determine whether adaptations occur in response to GLUT inhibition in the failing adult heart, we chronically treated TG9 mice, a transgenic model of dilated cardiomyopathy and heart failure, with the GLUT inhibitor ritonavir. Glucose tolerance was significantly improved with chronic treatment and correlated with decreased adipose tissue retinol binding protein 4 (RBP4) and resistin. A modest improvement in lifespan was associated with decreased cardiomyocyte brain natriuretic peptide (BNP) expression, a marker of heart failure severity. GLUT1 and -12 protein expression was significantly increased in left ventricular (LV) myocardium in ritonavir-treated animals. Supporting a switch from fatty acid to glucose utilization in these tissues, fatty acid transporter CD36 and fatty acid transcriptional regulator peroxisome proliferator-activated receptor α (PPARα) mRNA were also decreased in LV and soleus muscle. Chronic ritonavir also increased cardiac output and dV/dt-d in C57Bl/6 mice following ischemia-reperfusion injury. Taken together, these data demonstrate compensatory metabolic adaptation in response to chronic GLUT blockade as a means to evade deleterious changes in the failing heart.


Assuntos
Cardiomiopatia Dilatada/metabolismo , Proteínas Facilitadoras de Transporte de Glucose/metabolismo , Glucose/metabolismo , Animais , Glicemia/metabolismo , Doença da Artéria Coronariana/metabolismo , Modelos Animais de Doenças , Proteínas de Transporte de Ácido Graxo/metabolismo , Ácidos Graxos/metabolismo , Proteínas Facilitadoras de Transporte de Glucose/fisiologia , Insuficiência Cardíaca/metabolismo , Ventrículos do Coração/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/fisiopatologia , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , PPAR alfa/metabolismo , Ritonavir/farmacologia
16.
Sci Rep ; 7: 46438, 2017 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-28406175

RESUMO

The muscular ventricular septum separates the flow of oxygenated and de-oxygenated blood in air-breathing vertebrates. Defects within it, termed muscular ventricular septal defects (VSDs), are common, yet less is known about how they arise than rarer heart defects. Mutations of the cardiac transcription factor NKX2-5 cause cardiac malformations, including muscular VSDs. We describe here a genetic interaction between Nkx2-5 and Sarcospan (Sspn) that affects the risk of muscular VSD in mice. Sspn encodes a protein in the dystrophin-glycoprotein complex. Sspn knockout (SspnKO) mice do not have heart defects, but Nkx2-5+/-/SspnKO mutants have a higher incidence of muscular VSD than Nkx2-5+/- mice. Myofibers in the ventricular septum follow a stereotypical pattern that is disrupted around a muscular VSD. Subendocardial myofibers normally run in parallel along the left ventricular outflow tract, but in the Nkx2-5+/-/SspnKO mutant they commonly deviate into the septum even in the absence of a muscular VSD. Thus, Nkx2-5 and Sspn act in a pathway that affects the alignment of myofibers during the development of the ventricular septum. The malalignment may be a consequence of a defect in the coalescence of trabeculae into the developing ventricular septum, which has been hypothesized to be the mechanistic basis of muscular VSDs.


Assuntos
Proteínas de Transporte/genética , Técnicas de Inativação de Genes , Comunicação Interventricular/genética , Proteína Homeobox Nkx-2.5/genética , Proteínas de Membrana/genética , Mutação , Proteínas de Neoplasias/genética , Animais , Proteínas de Transporte/química , Modelos Animais de Doenças , Comunicação Interventricular/epidemiologia , Comunicação Interventricular/patologia , Humanos , Incidência , Proteínas de Membrana/química , Camundongos , Miócitos Cardíacos/patologia , Proteínas de Neoplasias/química
17.
Circ J ; 81(5): 629-634, 2017 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-28381817

RESUMO

Twenty years ago, chromosomal abnormalities were the only identifiable genetic causes of a small fraction of congenital heart defects (CHD). Today, a de novo or inherited genetic abnormality can be identified as pathogenic in one-third of cases. We refer to them here as monogenic causes, insofar as the genetic abnormality has a readily detectable, large effect. What explains the other two-thirds? This review considers a complex genetic basis. That is, a combination of genetic mutations or variants that individually may have little or no detectable effect contribute to the pathogenesis of a heart defect. Genes in the embryo that act directly in cardiac developmental pathways have received the most attention, but genes in the mother that establish the gestational milieu via pathways related to metabolism and aging also have an effect. A growing body of evidence highlights the pathogenic significance of genetic interactions in the embryo and maternal effects that have a genetic basis. The investigation of CHD as guided by a complex genetic model could help estimate risk more precisely and logically lead to a means of prevention.


Assuntos
Cardiopatias Congênitas/genética , Coração/crescimento & desenvolvimento , Variação Genética , Coração/embriologia , Coração/fisiopatologia , Humanos , Mutação
18.
Thyroid ; 27(5): 732-737, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28314380

RESUMO

BACKGROUND: The thyroid hormone-inactivating enzyme type 3 deiodinase (D3) is induced during hypertrophic and ischemic cardiomyopathy, leading to a state of local cardiac hypothyroidism. Whether D3 induction occurs in dilated cardiomyopathy is unknown. METHODS: This study characterized changes in cardiac D3 and thyroid hormone signaling in a transgenic model of progressive dilated cardiomyopathy (TG9 mice). RESULTS: Cardiac D3 was dramatically induced 15-fold during the progression of dilated cardiomyopathy in TG9 mice. This D3 induction localized to cardiomyocytes and was associated with a decrease in myocardial thyroid hormone signaling. CONCLUSIONS: Cardiac D3 is induced in a mouse model of dilated cardiomyopathy, indicating that D3 induction may be a general response to diverse forms of cardiomyopathy.


Assuntos
Cardiomiopatia Dilatada/metabolismo , Iodeto Peroxidase/metabolismo , Miocárdio/metabolismo , Tiroxina/sangue , Tri-Iodotironina/sangue , Animais , Cardiomiopatia Dilatada/patologia , Modelos Animais de Doenças , Progressão da Doença , Camundongos , Camundongos Transgênicos , Miocárdio/patologia
19.
Mol Cell Endocrinol ; 435: 94-102, 2016 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-27555292

RESUMO

Despite decades of progress, congenital heart disease remains a major cause of mortality and suffering in children and young adults. Prevention would be ideal, but formidable biological and technical hurdles face any intervention that seeks to target the main causes, genetic mutations in the embryo. Other factors, however, significantly modify the total risk in individuals who carry mutations. Investigation of these factors could lead to an alternative approach to prevention. To define the risk modifiers, our group has taken an "experimental epidemiologic" approach via inbred mouse strain crosses. The original intent was to map genes that modify an individual's risk of heart defects caused by an Nkx2-5 mutation. During the analysis of >2000 Nkx2-5(+/-) offspring from one cross we serendipitously discovered a maternal-age associated risk, which also exists in humans. Reciprocal ovarian transplants between young and old mothers indicate that the incidence of heart defects correlates with the age of the mother and not the oocyte, which implicates a maternal pathway as the basis of the risk. The quantitative risk varies between strain backgrounds, so maternal genetic polymorphisms determine the activity of a factor or factors in the pathway. Most strikingly, voluntary exercise by the mother mitigates the risk. Therefore, congenital heart disease can in principle be prevented by targeting a maternal pathway even if the embryo carries a causative mutation. Further mechanistic insight is necessary to develop an intervention that could be implemented on a broad scale, but the physiology of maternal-fetal interactions, aging, and exercise are notoriously complex and undefined. This suggests that an unbiased genetic approach would most efficiently lead to the relevant pathway. A genetic foundation would lay the groundwork for human studies and clinical trials.


Assuntos
Modelos Animais de Doenças , Predisposição Genética para Doença , Cardiopatias Congênitas/etiologia , Cardiopatias Congênitas/prevenção & controle , Idade Materna , Polimorfismo de Nucleotídeo Único , Complicações na Gravidez/genética , Animais , Exercício Físico , Feminino , Cardiopatias Congênitas/genética , Humanos , Camundongos , Gravidez , Fatores de Risco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA