Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
Mais filtros

País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
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
2.
Adv Exp Med Biol ; 1441: 481-493, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38884727

RESUMO

The relative simplicity of the clinical presentation and management of an atrial septal defect belies the complexity of the developmental pathogenesis. Here, we describe the anatomic development of the atrial septum and the venous return to the atrial chambers. Experimental models suggest how mutations and naturally occurring genetic variation could affect developmental steps to cause a defect within the oval fossa, the so-called secundum defect, or other interatrial communications, such as the sinus venosus defect or ostium primum defect.


Assuntos
Modelos Animais de Doenças , Comunicação Interatrial , Comunicação Interatrial/genética , Comunicação Interatrial/patologia , Comunicação Interatrial/fisiopatologia , Animais , Humanos , Mutação , Septo Interatrial/patologia , Transdução de Sinais/genética
3.
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
4.
Nature ; 520(7546): 230-3, 2015 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-25830876

RESUMO

Maternal age is a risk factor for congenital heart disease even in the absence of any chromosomal abnormality in the newborn. Whether the basis of this risk resides with the mother or oocyte is unknown. The impact of maternal age on congenital heart disease can be modelled in mouse pups that harbour a mutation of the cardiac transcription factor gene Nkx2-5 (ref. 8). Here, reciprocal ovarian transplants between young and old mothers establish a maternal basis for the age-associated risk in mice. A high-fat diet does not accelerate the effect of maternal ageing, so hyperglycaemia and obesity do not simply explain the mechanism. The age-associated risk varies with the mother's strain background, making it a quantitative genetic trait. Most remarkably, voluntary exercise, whether begun by mothers at a young age or later in life, can mitigate the risk when they are older. Thus, even when the offspring carry a causal mutation, an intervention aimed at the mother can meaningfully reduce their risk of congenital heart disease.


Assuntos
Envelhecimento/fisiologia , Cardiopatias/congênito , Cardiopatias/prevenção & controle , Idade Materna , Condicionamento Físico Animal/fisiologia , Prenhez/fisiologia , Idade de Início , Envelhecimento/genética , Animais , Animais Recém-Nascidos , Dieta Hiperlipídica , Feminino , Predisposição Genética para Doença , Coração/fisiologia , Coração/fisiopatologia , Cardiopatias/etiologia , Cardiopatias/genética , Proteína Homeobox Nkx-2.5 , Proteínas de Homeodomínio/genética , Hiperglicemia , Camundongos , Obesidade , Ovário/transplante , Fenótipo , Gravidez , Prenhez/genética , Locos de Características Quantitativas/genética , Risco , Fatores de Transcrição/genética
5.
Development ; 142(14): 2452-63, 2015 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-26062937

RESUMO

Although Notch1 and Notch2 are closely related paralogs and function through the same canonical signaling pathway, they contribute to different outcomes in some cell and disease contexts. To understand the basis for these differences, we examined in detail mice in which the Notch intracellular domains (N1ICD and N2ICD) were swapped. Our data indicate that strength (defined here as the ultimate number of intracellular domain molecules reaching the nucleus, integrating ligand-mediated release and nuclear translocation) and duration (half-life of NICD-RBPjk-MAML-DNA complexes, integrating cooperativity and stability dependent on shared sequence elements) are the factors that underlie many of the differences between Notch1 and Notch2 in all the contexts we examined, including T-cell development, skin differentiation and carcinogenesis, the inner ear, the lung and the retina. We were able to show that phenotypes in the heart, endothelium, and marginal zone B cells are attributed to haploinsufficiency but not to intracellular domain composition. Tissue-specific differences in NICD stability were most likely caused by alternative scissile bond choices by tissue-specific γ-secretase complexes following the intracellular domain swap. Reinterpretation of clinical findings based on our analyses suggests that differences in outcome segregating with Notch1 or Notch2 are likely to reflect outcomes dependent on the overall strength of Notch signals.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Regulação Neoplásica da Expressão Gênica , Receptor Notch1/metabolismo , Receptor Notch2/metabolismo , Alelos , Secretases da Proteína Precursora do Amiloide/metabolismo , Animais , Carcinogênese , Diferenciação Celular , Separação Celular , Orelha Interna/embriologia , Feminino , Citometria de Fluxo , Cardiopatias Congênitas , Homozigoto , Pulmão/embriologia , Masculino , Camundongos , Fenótipo , Estrutura Terciária de Proteína , Retina/embriologia , Transdução de Sinais , Pele/embriologia , Neoplasias Cutâneas/metabolismo , Linfócitos T/citologia , Transcriptoma
6.
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
7.
Gastroenterology ; 146(1): 157-165.e10, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24120474

RESUMO

BACKGROUND & AIMS: Infantile hypertrophic pyloric stenosis is a common birth anomaly characterized by obstruction of the pyloric lumen. A genome-wide association study implicated NKX2-5, which encodes a transcription factor that is expressed in embryonic heart and pylorus, in the pathogenesis of infantile hypertrophic pyloric stenosis. However, the function of the NKX2-5 in pyloric smooth muscle development has not been examined directly. We investigated the pattern of Nkx2-5 during the course of murine pyloric sphincter development and examined coexpression of Nkx2-5 with Gata3 and Sox9-other transcription factors with pyloric-specific mesenchymal expression. We also assessed pyloric sphincter development in mice with disruption of Nkx2-5 or Gata3. METHODS: We used immunofluorescence analysis to compare levels of NKX2-5, GATA3, and SOX9 in different regions of smooth muscle cells. Pyloric development was assessed in mice with conditional or germline deletion of Nkx2-5 or Gata3, respectively. RESULTS: Gata3, Nkx2-5, and Sox9 are coexpressed in differentiating smooth muscle cells of a distinct fascicle of the pyloric outer longitudinal muscle. Expansion of this fascicle coincides with development of the pyloric sphincter. Disruption of Nkx2-5 or Gata3 causes severe hypoplasia of this fascicle and alters pyloric muscle shape. Although expression of Sox9 requires Nkx2-5 and Gata3, there is no apparent hierarchical relationship between Nkx2-5 and Gata3 during pyloric outer longitudinal muscle development. CONCLUSIONS: Nkx2-5 and Gata3 are independently required for the development of a pyloric outer longitudinal muscle fascicle, which is required for pyloric sphincter morphogenesis in mice. These data indicate that regulatory changes that alter Nkx2-5 or Gata3 expression could contribute to pathogenesis of infantile hypertrophic pyloric stenosis.


Assuntos
Fator de Transcrição GATA3/metabolismo , Proteínas de Homeodomínio/metabolismo , Desenvolvimento Muscular/fisiologia , Músculo Liso/embriologia , Miócitos de Músculo Liso/metabolismo , Piloro/embriologia , Fatores de Transcrição SOX9/metabolismo , Fatores de Transcrição/metabolismo , Animais , Imunofluorescência , Proteína Homeobox Nkx-2.5 , Camundongos , Músculo Liso/metabolismo , Piloro/metabolismo
8.
J Pediatr ; 165(1): 194-6, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24721467

RESUMO

Tetralogy of Fallot and a complete atrioventricular septal defect are thought to arise by distinct mechanisms, yet their co-occurrence is a recognized association. Analysis of the prevalence of co-occurrence in Down syndrome suggests a common developmental basis. Trisomy 21 may perturb cardiac progenitor cells before they enter the heart tube.


Assuntos
Síndrome de Down/complicações , Insuficiência da Valva Mitral/complicações , Tetralogia de Fallot/complicações , Pré-Escolar , Síndrome de Down/fisiopatologia , Defeitos dos Septos Cardíacos , Humanos , Lactente , Insuficiência da Valva Mitral/epidemiologia , Insuficiência da Valva Mitral/fisiopatologia , Tetralogia de Fallot/epidemiologia , Tetralogia de Fallot/fisiopatologia
9.
Pediatr Cardiol ; 35(3): 411-8, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24061276

RESUMO

The Doppler Tei index is an independent predictor of outcomes in adult heart failure. Tissue Doppler imaging (TDI) may be a superior method to measure the Tei index in children because it is less affected by heart rate variability. We hypothesized that the TDI Tei index reflects severity of illness in pediatric heart failure. Twenty-five pediatric heart failure patients were prospectively enrolled. Listing for heart transplantation or death were the outcomes used to define severity of illness. Baseline demographics, brain natriuretic peptide (BNP), and standard echocardiographic and TDI-derived parameters were analyzed to determine outcome indicators. Ten of the 25 patients (40%) were listed for transplantation. There were no deaths. Multivariate analysis combining age, heart rate, standard echocardiographic parameters, and BNP resulted in shortening fraction (p = 0.002) as the best indicator of listing for transplantation (R(2) = 0.32). A second multivariate analysis combining age, heart rate, TDI parameters, and BNP resulted in age (p = 0.03) and septal Tei index (p = 0.03) as the best predictive model (R(2) = 0.36). The area under the receiver operating characteristic (ROC) curve for septal Tei index was 0.84 (95% confidence interval = 0.64-0.96,), and it was comparable with the ROC curve for shortening fraction, p = 0.76. Optimal values of sensitivity (100%) and specificity (60%) were obtained with septal Tei index values >0.51. The TDI septal Tei index is an indicator of disease severity in pediatric heart failure patients and offers potential advantages compared with standard echocardiographic measures of left-ventricular ejection.


Assuntos
Ecocardiografia Doppler , Insuficiência Cardíaca/diagnóstico por imagem , Insuficiência Cardíaca/fisiopatologia , Adolescente , Biomarcadores/sangue , Criança , Pré-Escolar , Feminino , Frequência Cardíaca/fisiologia , Humanos , Lactente , Masculino , Peptídeo Natriurético Encefálico/sangue , Valor Preditivo dos Testes , Estudos Prospectivos , Índice de Gravidade de Doença
10.
Physiol Genomics ; 45(2): 69-78, 2013 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-23170034

RESUMO

The physiological functions of a tissue in the body are carried out by its complement of expressed genes. Genes that execute a particular function should be more specifically expressed in tissues that perform the function. Given this premise, we mined public microarray expression data to build a database of genes ranked by their specificity of expression in multiple organs. The database permitted the accurate identification of genes and functions known to be specific to individual organs. Next, we used the database to predict transcriptional regulators of brown adipose tissue (BAT) and validated two candidate genes. Based upon hypotheses regarding pathways shared between combinations of BAT or white adipose tissue (WAT) and other organs, we identified genes that met threshold criteria for specific or counterspecific expression in each tissue. By contrasting WAT to the heart and BAT, the two most mitochondria-rich tissues in the body, we discovered a novel function for the transcription factor ESRRG in the induction of BAT genes in white adipocytes. Because the heart and other estrogen-related receptor gamma (ESRRG)-rich tissues do not express BAT markers, we hypothesized that an adipocyte co-regulator acts with ESRRG. By comparing WAT and BAT to the heart, brain, kidney and skeletal muscle, we discovered that an isoform of the transcription factor sterol regulatory element binding transcription factor 1 (SREBF1) induces BAT markers in C2C12 myocytes in the presence of ESRRG. The results demonstrate a straightforward bioinformatic strategy to associate genes with functions. The database upon which the strategy is based is provided so that investigators can perform their own screens.


Assuntos
Mineração de Dados , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Tecido Adiposo Marrom/metabolismo , Tecido Adiposo Branco/metabolismo , Algoritmos , Animais , Linhagem Celular , Feminino , Camundongos , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 1/genética , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo
11.
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.

12.
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
13.
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.

14.
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
15.
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
16.
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.

17.
J Mol Cell Cardiol ; 51(1): 120-8, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21549711

RESUMO

Lipin family proteins (lipin 1, 2, and 3) are bifunctional intracellular proteins that regulate metabolism by acting as coregulators of DNA-bound transcription factors and also dephosphorylate phosphatidate to form diacylglycerol [phosphatidate phosphohydrolase activity] in the triglyceride synthesis pathway. Herein, we report that lipin 1 is enriched in heart and that hearts of mice lacking lipin 1 (fld mice) exhibit accumulation of phosphatidate. We also demonstrate that the expression of the gene encoding lipin 1 (Lpin1) is under the control of the estrogen-related receptors (ERRs) and their coactivator the peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α). PGC-1α, ERRα, or ERRγ overexpression increased Lpin1 transcription in cultured ventricular myocytes and the ERRs were associated with response elements in the first intron of the Lpin1 gene. Concomitant RNAi-mediated knockdown of ERRα and ERRγ abrogated the induction of lipin 1 expression by PGC-1α overexpression. Consistent with these data, 3-fold overexpression of PGC-1α in intact myocardium of transgenic mice increased cardiac lipin 1 and ERRα/γ expression. Similarly, injection of the ß2-adrenergic agonist clenbuterol induced PGC-1α and lipin 1 expression, and the induction in lipin 1 after clenbuterol occurred in a PGC-1α-dependent manner. In contrast, expression of PGC-1α, ERRα, ERRγ, and lipin 1 was down-regulated in failing heart. Cardiac phosphatidic acid phosphohydrolase activity was also diminished, while cardiac phosphatidate content was increased, in failing heart. Collectively, these data suggest that lipin 1 is the principal lipin protein in the myocardium and is regulated in response to physiologic and pathologic stimuli that impact cardiac metabolism.


Assuntos
Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Proteínas Nucleares/metabolismo , Receptores de Estrogênio/metabolismo , Transativadores/metabolismo , Agonistas de Receptores Adrenérgicos beta 2/farmacologia , Animais , Animais Recém-Nascidos , Células Cultivadas , Imunoprecipitação da Cromatina , Clembuterol/farmacologia , Diglicerídeos/biossíntese , Insuficiência Cardíaca/metabolismo , Íntrons , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Proteínas Nucleares/biossíntese , Proteínas Nucleares/genética , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Fosfatidato Fosfatase/metabolismo , Ácidos Fosfatídicos/metabolismo , Interferência de RNA , RNA Interferente Pequeno , Ratos , Ratos Sprague-Dawley , Receptores de Estrogênio/biossíntese , Receptores de Estrogênio/genética , Elementos de Resposta , Fatores de Transcrição , Triglicerídeos/biossíntese , Receptor ERRalfa Relacionado ao Estrogênio
18.
Circulation ; 121(11): 1313-21, 2010 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-20212279

RESUMO

BACKGROUND: Mutations of the transcription factor Nkx2-5 cause pleiotropic heart defects with incomplete penetrance. This variability suggests that additional factors can affect or prevent the mutant phenotype. We assess here the role of genetic modifiers and their interactions. METHODS AND RESULTS: Heterozygous Nkx2-5 knockout mice in the inbred strain background C57Bl/6 frequently have atrial and ventricular septal defects. The incidences are substantially reduced in the Nkx2-5(+/-) progeny of first-generation (F1) outcrosses to the strains FVB/N or A/J. Defects recur in the second generation (F2) of the F1 X F1 intercross or backcrosses to the parental strains. Analysis of >3000 Nkx2-5(+/-) hearts from 5 F2 crosses demonstrates the profound influence of genetic modifiers on disease presentation. On the basis of their incidences and coincidences, anatomically distinct malformations have shared and unique modifiers. All 3 strains carry susceptibility alleles at different loci for atrial and ventricular septal defects. Relative to the other 2 strains, A/J carries polymorphisms that confer greater susceptibility to atrial septal defect and atrioventricular septal defects and C57Bl/6 to muscular ventricular septal defects. Segregation analyses reveal that > or = 2 loci influence membranous ventricular septal defect susceptibility, whereas > or = loci and at least 1 epistatic interaction affect muscular ventricular and atrial septal defects. CONCLUSIONS: Alleles of modifier genes can either buffer perturbations on cardiac development or direct the manifestation of a defect. In a genetically heterogeneous population, the predominant effect of modifier genes is health. (Circulation. 2010;121:1313-1321.)


Assuntos
Predisposição Genética para Doença/genética , Cardiopatias Congênitas/genética , Coração/embriologia , Animais , Modelos Animais de Doenças , Feminino , Cardiopatias Congênitas/epidemiologia , Comunicação Interatrial/epidemiologia , Comunicação Interatrial/genética , Comunicação Interventricular/epidemiologia , Comunicação Interventricular/genética , Proteína Homeobox Nkx-2.5 , Proteínas de Homeodomínio/genética , Incidência , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Knockout , Mutação/genética , Fenótipo , Fatores de Risco , Fatores de Transcrição/genética
19.
Biol Reprod ; 84(5): 1033-44, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21248289

RESUMO

Transcription factor GATA4 is expressed in granulosa cells and, to a lesser extent, in other ovarian cell types. Studies of mutant mice have shown that interactions between GATA4 and its cofactor, ZFPM2 (also termed FOG2), are required for proper development of the fetal ovary. The role of GATA4 in postnatal ovarian function, however, has remained unclear, in part because of prenatal lethality of homozygous mutations in the Gata4 gene in mice. To circumvent this limitation, we studied ovarian function in two genetically engineered mouse lines: C57BL/6 (B6) female mice heterozygous for a Gata4-null allele, and 129;B6 female mice in which Gata4 is deleted specifically in proliferating granulosa cells using the Cre-loxP recombination system and Amhr2-cre. Female B6 Gata4(+/-) mice had delayed puberty but normal estrous cycle lengths and litter size. Compared to wild-type mice, the ovaries of gonadotropin-stimulated B6 Gata4(+/-) mice were significantly smaller, released fewer oocytes, produced less estrogen, and expressed less mRNA for the putative GATA4 target genes Star, Cyp11a1, and Cyp19. Gata4 conditional knockout (cKO) mice had a more severe phenotype, including impaired fertility and cystic ovarian changes. Like Gata4(+/-) mice, the ovaries of gonadotropin-stimulated cKO mice released fewer oocytes and expressed less Cyp19 than those of control mice. Our findings, coupled with those of other investigators, support the premise that GATA4 is a key transcriptional regulator of ovarian somatic cell function in both fetal and adult mice.


Assuntos
Fator de Transcrição GATA4/fisiologia , Ovário/fisiologia , Animais , Aromatase/genética , Aromatase/metabolismo , Enzima de Clivagem da Cadeia Lateral do Colesterol/genética , Enzima de Clivagem da Cadeia Lateral do Colesterol/metabolismo , Estrogênios/metabolismo , Feminino , Fator de Transcrição GATA4/genética , Deleção de Genes , Expressão Gênica , Engenharia Genética/métodos , Heterozigoto , Infertilidade Feminina/genética , Infertilidade Feminina/metabolismo , Infertilidade Feminina/fisiopatologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oogênese , Tamanho do Órgão , Cistos Ovarianos/genética , RNA Mensageiro/metabolismo , Recombinação Genética , Maturidade Sexual
20.
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
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa