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












Base de datos
Intervalo de año de publicación
1.
J Neurooncol ; 2024 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-38842696

RESUMEN

PURPOSE: This study aimed to evaluate the prognostic performance of amino-acid PET in high-grade gliomas (HGG) patients at the time of temozolomide (TMZ) treatment discontinuation, after the Stupp protocol. METHODS: The analysis included consecutive HGG patients with dynamic [18F]FDOPA PET imaging within 3 months of the end of TMZ therapy, post-Stupp protocol. Static and dynamic PET parameters, responses to RANO criteria for MRI and clinical and histo-molecular factors were correlated to progression-free (PFS). RESULTS: Thirty-two patients (59.4 [54.0;67.6] years old, 13 (41%) women) were included. Static PET parameters peak tumor-to-background ratio and metabolic tumor volume (respective thresholds of 1.9 and 1.5 mL) showed the best 84% accuracies for predicting PFS at 6 months (p = 0.02). These static PET parameters were also independent predictor of PFS in multivariate analysis (p ≤ 0.05). CONCLUSION: In HGG patients having undergone a Stupp protocol, the absence of significant PET uptake after TMZ constitutes a favorable prognostic factor.

2.
Circulation ; 148(23): 1870-1886, 2023 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-37886847

RESUMEN

BACKGROUND: Microvasculature dysfunction is a common finding in pathologic remodeling of the heart and is thought to play an important role in the pathogenesis of hypertrophic cardiomyopathy (HCM), a disease caused by sarcomere gene mutations. We hypothesized that microvascular dysfunction in HCM was secondary to abnormal microvascular growth and could occur independent of ventricular hypertrophy. METHODS: We used multimodality imaging methods to track the temporality of microvascular dysfunction in HCM mouse models harboring mutations in the sarcomere genes Mybpc3 (cardiac myosin binding protein C3) or Myh6 (myosin heavy chain 6). We performed complementary molecular methods to assess protein quantity, interactions, and post-translational modifications to identify mechanisms regulating this response. We manipulated select molecular pathways in vivo using both genetic and pharmacological methods to validate these mechanisms. RESULTS: We found that microvascular dysfunction in our HCM models occurred secondary to reduced myocardial capillary growth during the early postnatal time period and could occur before the onset of myocardial hypertrophy. We discovered that the E3 ubiquitin protein ligase MDM2 (murine double minute 2) dynamically regulates the protein stability of both HIF1α (hypoxia-inducible factor 1 alpha) and HIF2α (hypoxia-inducible factor 2 alpha)/EPAS1 (endothelial PAS domain protein 1) through canonical and noncanonical mechanisms. The resulting HIF imbalance leads to reduced proangiogenic gene expression during a key period of myocardial capillary growth. Reducing MDM2 protein levels by genetic or pharmacological methods normalized HIF protein levels and prevented the development of microvascular dysfunction in both HCM models. CONCLUSIONS: Our results show that sarcomere mutations induce cardiomyocyte MDM2 signaling during the earliest stages of disease, and this leads to long-term changes in the myocardial microenvironment.


Asunto(s)
Cardiomiopatía Hipertrófica , Proteínas Proto-Oncogénicas c-mdm2 , Ratones , Animales , Proteínas Proto-Oncogénicas c-mdm2/genética , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Sarcómeros/metabolismo , Mutación , Hipertrofia , Cadenas Pesadas de Miosina/genética , Cadenas Pesadas de Miosina/metabolismo
3.
JAMA Cardiol ; 8(11): 1083-1088, 2023 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-37672268

RESUMEN

Importance: Valsartan has shown promise in attenuating cardiac remodeling in patients with early-stage sarcomeric hypertrophic cardiomyopathy (HCM). Genetic testing can identify individuals at risk of HCM in a subclinical stage who could benefit from therapies that prevent disease progression. Objective: To explore the potential for valsartan to modify disease development, and to characterize short-term phenotypic progression in subclinical HCM. Design, Setting, and Participants: The multicenter, double-blind, placebo-controlled Valsartan for Attenuating Disease Evolution in Early Sarcomeric Hypertrophic Cardiomyopathy (VANISH) randomized clinical trial was conducted from April 2014 to July 2019 at 17 sites in 4 countries (Brazil, Canada, Denmark, and the US), with 2 years of follow-up. The prespecified exploratory VANISH cohort studied here included sarcomere variant carriers with subclinical HCM and early phenotypic manifestations (reduced E' velocity, electrocardiographic abnormalities, or an increased left ventricular [LV] wall thickness [LVWT] to cavity diameter ratio) but no LV hypertrophy (LVH). Data were analyzed between March and December 2022. Interventions: Treatment with placebo or valsartan (80 mg/d for children weighing <35 kg, 160 mg/d for children weighing ≥35 kg, or 320 mg/d for adults aged ≥18 years). Main Outcomes and Measures: The primary outcome was a composite z score incorporating changes in 9 parameters of cardiac remodeling (LV cavity volume, LVWT, and LV mass; left atrial [LA] volume; E' velocity and S' velocity; and serum troponin and N-terminal prohormone of brain natriuretic peptide levels). Results: This study included 34 participants, with a mean (SD) age of 16 (5) years (all were White). A total of 18 participants (8 female [44%] and 10 male [56%]) were randomized to valsartan and 16 (9 female [56%] and 7 male [44%]) were randomized to placebo. No statistically significant effects of valsartan on cardiac remodeling were detected (mean change in composite z score compared with placebo: -0.01 [95% CI, -0.29 to 0.26]; P = .92). Overall, 2-year phenotypic progression was modest, with only a mild increase in LA volume detected (increased by 3.5 mL/m2 [95% CI, 1.4-6.0 mL/m2]; P = .002). Nine participants (26%) had increased LVWT, including 6 (18%) who developed clinically overt HCM. Baseline LA volume index (LAVI; 35 vs 28 mL/m2; P = .01) and average interventricular septum thickness (8.5 vs 7.0 mm; P = .009) were higher in participants who developed HCM. Conclusions and Relevance: In this exploratory cohort, valsartan was not proven to slow progression of subclinical HCM. Minimal changes in markers of cardiac remodeling were observed, although nearly one-fifth of patients developed clinically overt HCM. Transition to disease was associated with greater baseline interventricular septum thickness and LAVI. These findings highlight the importance of following sarcomere variant carriers longitudinally and the critical need to improve understanding of factors that drive disease penetrance and progression. Trial Registration: ClinicalTrials.gov Identifier: NCT01912534.


Asunto(s)
Cardiomiopatía Hipertrófica , Remodelación Ventricular , Adulto , Niño , Humanos , Masculino , Femenino , Adolescente , Predisposición Genética a la Enfermedad , Hipertrofia Ventricular Izquierda , Valsartán/uso terapéutico
5.
Cardiovasc Res ; 118(9): 2124-2138, 2022 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-34329394

RESUMEN

AIMS: The cardiac natriuretic peptides [atrial natriuretic peptide (ANP) and B-type natriuretic peptide (BNP)] are important regulators of cardiovascular physiology, with reduced natriuretic peptide (NP) activity linked to multiple human cardiovascular diseases. We hypothesized that deficiency of either ANP or BNP would lead to similar changes in left ventricular structure and function given their shared receptor affinities. METHODS AND RESULTS: We directly compared murine models deficient of ANP or BNP in the same genetic backgrounds (C57BL6/J) and environments. We evaluated control, ANP-deficient (Nppa-/-) or BNP-deficient (Nppb-/-) mice under unstressed conditions and multiple forms of pathological myocardial stress. Survival, myocardial structure, function and electrophysiology, tissue histology, and biochemical analyses were evaluated in the groups. In vitro validation of our findings was performed using human-derived induced pluripotent stem cell cardiomyocytes (iPS-CMs). In the unstressed state, both ANP- and BNP-deficient mice displayed mild ventricular hypertrophy which did not increase up to 1 year of life. NP-deficient mice exposed to acute myocardial stress secondary to thoracic aortic constriction (TAC) had similar pathological myocardial remodelling but a significant increase in sudden death. We discovered that the NP-deficient mice are more susceptible to stress-induced ventricular arrhythmias using both in vivo and ex vivo models. Mechanistically, deficiency of either ANP or BNP led to reduced myocardial cGMP levels and reduced phosphorylation of the cAMP response element-binding protein (CREBS133) transcriptional regulator. Selective CREB inhibition sensitized wild-type hearts to stress-induced ventricular arrhythmias. ANP and BNP regulate cardiomyocyte CREBS133 phosphorylation through a cGMP-dependent protein kinase 1 (PKG1) and p38 mitogen-activated protein kinase (p38 MAPK) signalling cascade. CONCLUSIONS: Our data show that ANP and BNP act in a non-redundant fashion to maintain myocardial cGMP levels to regulate cardiomyocyte p38 MAPK and CREB activity. Cardiac natriuretic peptide deficiency leads to a reduction in CREB signalling which sensitizes the heart to stress-induced ventricular arrhythmias.


Asunto(s)
Arritmias Cardíacas , Factor Natriurético Atrial , Péptido Natriurético Encefálico , Animales , Arritmias Cardíacas/genética , Arritmias Cardíacas/metabolismo , Factor Natriurético Atrial/genética , Factor Natriurético Atrial/metabolismo , GMP Cíclico , Ratones , Miocitos Cardíacos/metabolismo , Péptido Natriurético Encefálico/genética , Péptido Natriurético Encefálico/metabolismo , Péptidos Natriuréticos/metabolismo , Vasodilatadores , Proteínas Quinasas p38 Activadas por Mitógenos
6.
Nat Med ; 27(10): 1818-1824, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34556856

RESUMEN

Hypertrophic cardiomyopathy (HCM) is often caused by pathogenic variants in sarcomeric genes and characterized by left ventricular (LV) hypertrophy, myocardial fibrosis and increased risk of heart failure and arrhythmias. There are no existing therapies to modify disease progression. In this study, we conducted a multi-center, double-blind, placebo-controlled phase 2 clinical trial to assess the safety and efficacy of the angiotensin II receptor blocker valsartan in attenuating disease evolution in early HCM. In total, 178 participants with early-stage sarcomeric HCM were randomized (1:1) to receive valsartan (320 mg daily in adults; 80-160 mg daily in children) or placebo for 2 years ( NCT01912534 ). Standardized changes from baseline to year 2 in LV wall thickness, mass and volumes; left atrial volume; tissue Doppler diastolic and systolic velocities; and serum levels of high-sensitivity troponin T and N-terminal pro-B-type natriuretic protein were integrated into a single composite z-score as the primary outcome. Valsartan (n = 88) improved cardiac structure and function compared to placebo (n = 90), as reflected by an increase in the composite z-score (between-group difference +0.231, 95% confidence interval (+0.098, +0.364); P = 0.001), which met the primary endpoint of the study. Treatment was well-tolerated. These results indicate a key opportunity to attenuate disease progression in early-stage sarcomeric HCM with an accessible and safe medication.


Asunto(s)
Cardiomiopatía Hipertrófica/tratamiento farmacológico , Insuficiencia Cardíaca/tratamiento farmacológico , Corazón/efectos de los fármacos , Valsartán/administración & dosificación , Adolescente , Adulto , Cardiomiopatía Hipertrófica/fisiopatología , Método Doble Ciego , Femenino , Corazón/fisiopatología , Insuficiencia Cardíaca/fisiopatología , Humanos , Masculino , Persona de Mediana Edad , Valsartán/efectos adversos , Adulto Joven
7.
J Am Heart Assoc ; 10(15): e021768, 2021 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-34323119

RESUMEN

Background Sarcomere gene mutations lead to cardiomyocyte hypertrophy and pathological myocardial remodeling. However, there is considerable phenotypic heterogeneity at both the cellular and the organ level, suggesting modifiers regulate the effects of these mutations. We hypothesized that sarcomere dysfunction leads to cardiomyocyte genotoxic stress, and this modifies pathological ventricular remodeling. Methods and Results Using a murine model deficient in the sarcomere protein, Mybpc3-/- (cardiac myosin-binding protein 3), we discovered that there was a surge in cardiomyocyte nuclear DNA damage during the earliest stages of cardiomyopathy. This was accompanied by a selective increase in ataxia telangiectasia and rad3-related phosphorylation and increased p53 protein accumulation. The cause of the DNA damage and DNA damage pathway activation was dysregulated cardiomyocyte DNA synthesis, leading to replication stress. We discovered that selective inhibition of ataxia telangiectasia and rad3 related or cardiomyocyte deletion of p53 reduced pathological left ventricular remodeling and cardiomyocyte hypertrophy in Mybpc3-/- animals. Mice and humans harboring other types of sarcomere gene mutations also had evidence of activation of the replication stress response, and this was associated with cardiomyocyte aneuploidy in all models studied. Conclusions Collectively, our results show that sarcomere mutations lead to activation of the cardiomyocyte replication stress response, which modifies pathological myocardial remodeling in sarcomeric cardiomyopathy.


Asunto(s)
Miosinas Cardíacas/genética , Cardiomiopatías , Proteínas Portadoras , Daño del ADN , Miocitos Cardíacos/metabolismo , Sarcómeros , Remodelación Ventricular/genética , Animales , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Cardiomiopatías/genética , Cardiomiopatías/patología , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Modelos Animales de Enfermedad , Ratones , Ratones Noqueados , Mutación , Sarcómeros/genética , Sarcómeros/metabolismo
8.
Open Heart ; 8(1)2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33637569

RESUMEN

BACKGROUND: Despite advances in our understanding of the genetic causes of hypertrophic cardiomyopathy (HCM), a large portion of this patient population do not carry sarcomere gene mutations when screened. It remains largely unknown why patients without sarcomere mutations develop asymmetric myocardial hypertrophy. METHODS: We performed a retrospective analysis of probands with HCM who underwent genetic testing to determine if clinical phenotypes were different depending on sarcomere mutation status. A medical history, three generation family history and clinical phenotyping were performed on 127 probands with HCM. Genetic screening was performed using clinically available HCM genetic testing panels. RESULTS: We found that probands with HCM with pathogenic sarcomere mutations were over three times more likely to have a family history of HCM (66% vs 17%, p<0.0001) and were diagnosed with HCM at a much younger age (32 vs 51 years old, p<0.0001). In contrast, probands with HCM without sarcomere mutations were significantly more obese (body surface area p=0.003, body mass index p=0.04 adjusted for age) and were more likely to present with left ventricular outflow tract obstruction (p=0.0483). CONCLUSION: Patients with sarcomere mutation negative HCM present at an older age and are more obese compared with patients with sarcomere mutation positive HCM. The role of ageing and obesity in asymmetric myocardial hypertrophy warrants further investigation.


Asunto(s)
Envejecimiento/genética , Cardiomiopatía Hipertrófica/genética , Imagen por Resonancia Cinemagnética/métodos , Mutación , Miocardio/patología , Obesidad/complicaciones , Sarcómeros/genética , Adulto , Cardiomiopatía Hipertrófica/diagnóstico , Cardiomiopatía Hipertrófica/etiología , Proteínas Portadoras/genética , Femenino , Estudios de Seguimiento , Pruebas Genéticas , Humanos , Masculino , Persona de Mediana Edad , Fenotipo , Estudios Retrospectivos
9.
Circ Heart Fail ; 12(12): e006231, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31813281

RESUMEN

BACKGROUND: The VANISH trial (Valsartan for Attenuating Disease Evolution in Early Sarcomeric Hypertrophic Cardiomyopathy) targeted young sarcomeric gene mutation carriers with early-stage hypertrophic cardiomyopathy (HCM) to test whether valsartan can modify disease progression. We describe the baseline characteristics of the VANISH cohort and compare to previous trials evaluating angiotensin receptor blockers. METHODS: Applying a randomized, double-blinded, placebo-controlled design, 178 participants with nonobstructive HCM (age, 23.3±10.1 years; 61% men) were randomized in the primary cohort and 34 (age, 16.5±4.9 years; 50% men) in the exploratory cohort of sarcomeric mutation carriers without left ventricular hypertrophy. RESULTS: In the primary cohort, maximal left ventricular wall thickness was 17±4 mm for adults and Z score 7.0±4.5 for children. Nineteen percent had late gadolinium enhancement on cardiac magnetic resonance. Mean peak oxygen consumption was 33 mL/kg per minute, and 92% of participants were New York Heart Association functional class I. New York Heart Association class II was associated with older age, MYH7 variants, and more prominent imaging abnormalities. Six previous trials of angiotensin receptor blockers in HCM enrolled a median of 24 patients (range, 19-133) with mean age of 51.2 years; 42% of patients were in New York Heart Association class ≥II, and sarcomeric mutations were not required. CONCLUSIONS: The VANISH cohort is much larger, younger, less heterogeneous, and has less advanced disease than prior angiotensin receptor blocker trials in HCM. Participants had relatively normal functional capacity and mild HCM features. New York Heart Association functional class II symptoms were associated with older age, more prominent imaging abnormalities, and MYH7 variants, suggesting both phenotype and genotype contribute to disease manifestations. CLINICAL TRIAL REGISTRATION: URL: https://www.clinicaltrials.gov. Unique identifier: NCT01912534.


Asunto(s)
Bloqueadores del Receptor Tipo 1 de Angiotensina II/uso terapéutico , Cardiomiopatía Hipertrófica/tratamiento farmacológico , Mutación , Sarcómeros/genética , Valsartán/uso terapéutico , Adolescente , Adulto , Bloqueadores del Receptor Tipo 1 de Angiotensina II/efectos adversos , Brasil , Canadá , Cardiomiopatía Hipertrófica/diagnóstico , Cardiomiopatía Hipertrófica/genética , Cardiomiopatía Hipertrófica/fisiopatología , Niño , Dinamarca , Progresión de la Enfermedad , Método Doble Ciego , Femenino , Predisposición Genética a la Enfermedad , Humanos , Masculino , Persona de Mediana Edad , Fenotipo , Recuperación de la Función , Factores de Tiempo , Resultado del Tratamiento , Estados Unidos , Valsartán/efectos adversos , Adulto Joven
10.
Cardiovasc Res ; 115(5): 966-977, 2019 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-30629146

RESUMEN

AIMS: Tyrosine kinase inhibitors (TKIs) have revolutionized the treatment of chronic myelogenous leukaemia (CML). However, cardiotoxicity of these agents remains a serious concern. The underlying mechanism of these adverse cardiac effects is largely unknown. Delineation of the underlying mechanisms of TKIs associated cardiac dysfunction could guide potential prevention strategies, rescue approaches, and future drug design. This study aimed to determine the cardiotoxic potential of approved CML TKIs, define the associated signalling mechanism and identify potential alternatives. METHODS AND RESULTS: In this study, we employed a zebrafish transgenic BNP reporter line that expresses luciferase under control of the nppb promoter (nppb:F-Luciferase) to assess the cardiotoxicity of all approved CML TKIs. Our in vivo screen identified ponatinib as the most cardiotoxic agent among the approved CML TKIs. Then using a combination of zebrafish and isolated neonatal rat cardiomyocytes, we delineated the signalling mechanism of ponatinib-induced cardiotoxicity by demonstrating that ponatinib inhibits cardiac prosurvival signalling pathways AKT and extra-cellular-signal-regulated kinase (ERK), and induces cardiomyocyte apoptosis. As a proof of concept, we augmented AKT and ERK signalling by administration of Neuregulin-1ß (NRG-1ß), and this prevented ponatinib-induced cardiomyocyte apoptosis. We also demonstrate that ponatinib-induced cardiotoxicity is not mediated by inhibition of fibroblast growth factor signalling, a well-known target of ponatinib. Finally, our comparative profiling for the cardiotoxic potential of CML approved TKIs, identified asciminib (ABL001) as a potentially much less cardiotoxic treatment option for CML patients with the T315I mutation. CONCLUSION: Herein, we used a combination of in vivo and in vitro methods to systematically screen CML TKIs for cardiotoxicity, identify novel molecular mechanisms for TKI cardiotoxicity, and identify less cardiotoxic alternatives.


Asunto(s)
Antineoplásicos/toxicidad , Cardiopatías/inducido químicamente , Imidazoles/toxicidad , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Miocitos Cardíacos/efectos de los fármacos , Inhibidores de Proteínas Quinasas/toxicidad , Piridazinas/toxicidad , Transducción de Señal/efectos de los fármacos , Animales , Animales Modificados Genéticamente , Apoptosis/efectos de los fármacos , Cardiotoxicidad , Células Cultivadas , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Cardiopatías/metabolismo , Cardiopatías/patología , Cardiopatías/prevención & control , Leucemia Mielógena Crónica BCR-ABL Positiva/enzimología , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Péptido Natriurético Encefálico/genética , Péptido Natriurético Encefálico/metabolismo , Niacinamida/análogos & derivados , Niacinamida/toxicidad , Prueba de Estudio Conceptual , Proteínas Proto-Oncogénicas c-akt/metabolismo , Pirazoles/toxicidad , Ratas , Pez Cebra/embriología , Pez Cebra/genética , Pez Cebra/metabolismo
11.
FASEB J ; 33(1): 1138-1150, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30106602

RESUMEN

Raf1/c-Raf is a well-characterized serine/threonine-protein kinase that links Ras family members with the MAPK/ERK signaling cascade. We have identified a novel splice isoform of human Raf1 that causes protein truncation and loss of the C-terminal kinase domain (Raf1-tr). We found that Raf1-tr has increased nuclear localization compared with full-length Raf1, and this finding was secondary to reduced binding of Raf1-tr to the cytoplasmic chaperone FK506 binding protein 5. We show that Raf1-tr has increased binding to DNA-dependent protein kinase (DNA-PK), which inhibits DNA-PK function and causes amplification of irradiation- and bleomycin-induced DNA damage. We found that the human colorectal cancer cell line, HCT-116, displayed reduced expression of Raf1-tr, and reintroduction of Raf1-tr sensitized the cells to bleomycin-induced apoptosis. Furthermore, we identified differential Raf1-tr expression in breast cancer cell lines and showed that breast cancer cells with increased Raf1-tr expression become sensitized to bleomycin-induced apoptosis. Collectively, these results demonstrate a novel Raf1 isoform in humans that has a unique noncanonical role in regulating the double-stranded DNA damage response pathway through modulation of DNA-PK function.-Nixon, B. R., Sebag, S. C., Glennon, M. S., Hall, E. J., Kounlavong, E. S., Freeman, M. L., Becker, J. R. Nuclear localized Raf1 isoform alters DNA-dependent protein kinase activity and the DNA damage response.


Asunto(s)
Núcleo Celular/metabolismo , Daño del ADN , Proteína Quinasa Activada por ADN/metabolismo , Isoformas de Proteínas/metabolismo , Proteínas Proto-Oncogénicas c-raf/metabolismo , Empalme Alternativo , Antibióticos Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Bleomicina/farmacología , Línea Celular Tumoral , ADN/efectos de los fármacos , ADN/efectos de la radiación , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Células HEK293 , Humanos , Unión Proteica , Transducción de Señal , Proteínas de Unión a Tacrolimus/metabolismo , Proteínas ras/metabolismo
12.
Elife ; 72018 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-30540249

RESUMEN

The sarcomere is the contractile unit within cardiomyocytes driving heart muscle contraction. We sought to test the mechanisms regulating actin and myosin filament assembly during sarcomere formation. Therefore, we developed an assay using human cardiomyocytes to monitor sarcomere assembly. We report a population of muscle stress fibers, similar to actin arcs in non-muscle cells, which are essential sarcomere precursors. We show sarcomeric actin filaments arise directly from muscle stress fibers. This requires formins (e.g., FHOD3), non-muscle myosin IIA and non-muscle myosin IIB. Furthermore, we show short cardiac myosin II filaments grow to form ~1.5 µm long filaments that then 'stitch' together to form the stack of filaments at the core of the sarcomere (i.e., the A-band). A-band assembly is dependent on the proper organization of actin filaments and, as such, is also dependent on FHOD3 and myosin IIB. We use this experimental paradigm to present evidence for a unifying model of sarcomere assembly.


Asunto(s)
Fibras Musculares Esqueléticas/metabolismo , Miocitos Cardíacos/metabolismo , Sarcómeros/metabolismo , Fibras de Estrés/metabolismo , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Línea Celular , Línea Celular Tumoral , Forminas , Células HeLa , Humanos , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Microscopía Confocal , Proteínas Motoras Moleculares/genética , Proteínas Motoras Moleculares/metabolismo , Fibras Musculares Esqueléticas/citología , Miocitos Cardíacos/citología , Cadenas Pesadas de Miosina/genética , Cadenas Pesadas de Miosina/metabolismo , Miosina Tipo IIB no Muscular/genética , Miosina Tipo IIB no Muscular/metabolismo , Interferencia de ARN
13.
JCI Insight ; 2(4): e90656, 2017 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-28239655

RESUMEN

It remains unclear how perturbations in cardiomyocyte sarcomere function alter postnatal heart development. We utilized murine models that allowed manipulation of cardiac myosin-binding protein C (MYBPC3) expression at critical stages of cardiac ontogeny to study the response of the postnatal heart to disrupted sarcomere function. We discovered that the hyperplastic to hypertrophic transition phase of mammalian heart development was altered in mice lacking MYBPC3 and this was the critical period for subsequent development of cardiomyopathy. Specifically, MYBPC3-null hearts developed evidence of increased cardiomyocyte endoreplication, which was accompanied by enhanced expression of cell cycle stimulatory cyclins and increased phosphorylation of retinoblastoma protein. Interestingly, this response was self-limited at later developmental time points by an upregulation of the cyclin-dependent kinase inhibitor p21. These results provide valuable insights into how alterations in sarcomere protein function modify postnatal heart development and highlight the potential for targeting cell cycle regulatory pathways to counteract cardiomyopathic stimuli.


Asunto(s)
Proteínas Portadoras/genética , Aumento de la Célula , Proliferación Celular/genética , Regulación del Desarrollo de la Expresión Génica/genética , Corazón/crecimiento & desarrollo , Miocitos Cardíacos/metabolismo , Sarcómeros/metabolismo , Animales , Cardiomiopatías/genética , Cardiomiopatías/metabolismo , Ciclo Celular , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Ciclinas/metabolismo , Hiperplasia , Hipertrofia , Ratones , Miocitos Cardíacos/fisiología , Fosforilación , Proteína de Retinoblastoma/metabolismo , Sarcómeros/fisiología , Regulación hacia Arriba
14.
N Engl J Med ; 375(18): 1749-1755, 2016 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-27806233

RESUMEN

Immune checkpoint inhibitors have improved clinical outcomes associated with numerous cancers, but high-grade, immune-related adverse events can occur, particularly with combination immunotherapy. We report the cases of two patients with melanoma in whom fatal myocarditis developed after treatment with ipilimumab and nivolumab. In both patients, there was development of myositis with rhabdomyolysis, early progressive and refractory cardiac electrical instability, and myocarditis with a robust presence of T-cell and macrophage infiltrates. Selective clonal T-cell populations infiltrating the myocardium were identical to those present in tumors and skeletal muscle. Pharmacovigilance studies show that myocarditis occurred in 0.27% of patients treated with a combination of ipilimumab and nivolumab, which suggests that our patients were having a rare, potentially fatal, T-cell-driven drug reaction. (Funded by Vanderbilt-Ingram Cancer Center Ambassadors and others.).


Asunto(s)
Anticuerpos Monoclonales/efectos adversos , Inmunoterapia/efectos adversos , Miocarditis/etiología , Miocardio/patología , Anciano , Anticuerpos Monoclonales/uso terapéutico , Arritmias Cardíacas/inducido químicamente , Electrocardiografía/efectos de los fármacos , Resultado Fatal , Femenino , Glucocorticoides/uso terapéutico , Bloqueo Cardíaco/diagnóstico , Bloqueo Cardíaco/etiología , Humanos , Ipilimumab , Masculino , Melanoma/complicaciones , Melanoma/tratamiento farmacológico , Persona de Mediana Edad , Miocarditis/tratamiento farmacológico , Miocarditis/patología , Miositis/inducido químicamente , Nivolumab
15.
Appl Spectrosc ; 70(3): 467-73, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26810184

RESUMEN

Laser-induced breakdown spectroscopy (LIBS) holds potential advantages in special nuclear material (SNM) sensing and nuclear forensics, which require rapid analysis, minimal sample preparation, and stand-off distance capability. SNM, such as U, however, result in crowded emission spectra with LIBS, and characteristic emission lines are challenging to discern. It is well-known that double-pulse LIBS (DPLIBS) improves the signal intensity for analytes over conventional single-pulse LIBS (SPLIBS). This study investigates the U signal in a glass matrix using DPLIBS and compares it to signal obtained using SPLIBS. Double-pulse LIBS involves sequential firing of a 1.06 µm Nd:YAG pre-pulse and 10.6 µm TEA CO2 heating pulse in a near collinear geometry. Optimization of experimental parameters including inter-pulse delay and energy follows identification of characteristic lines for the bulk analyte Ca and the minor constituent analyte U for both DPLIBS and SPLIBS. Spatial and temporal coupling of the two pulses in the proposed DPLIBS technique yields improvements in analytical merits with a negligible increase in damage to the sample compared to SPLIBS. Subsequently, the study discusses optimum plasma emission conditions of U lines and relative figures of merit in both SPLIBS and DPLIBS. Investigation into plasma characteristics also addresses plausible mechanisms related to the observed U analyte signal variation between SPLIBS and DPLIBS.

16.
J Card Fail ; 22(4): 294-302, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26106805

RESUMEN

AIM: The aim of this work was to determine whether ethanol-associated myocardial apoptosis and cardiac dilation are related to myocardial telomere shortening in rats. METHODS AND RESULTS: Sprague-Dawley (SD) rats received either drinking water with (ethanol: n = 19) or without (control: n = 19) 5% (v/v) ethanol ad libitum, for 4 months. Left ventricular (LV) dimensions and function (echocardiography and isolated perfused heart preparations), cardiomyocyte apoptosis (terminal deoxynucleotide transferase-mediated dUTP nick-end labeling), and leukocyte and myocardial telomere length (real-time polymerase chain reaction) were determined at the end of the study. Ethanol administration resulted in a marked increase in cardiomyocyte apoptosis (ethanol 0.85 ± 0.13% vs control 0.36 ± 0.06%; P = .0021) and LV dilation (LV end-diastolic diameter: ethanol 8.20 ± 0.14 mm vs control 7.56 ± 0.11 mm [P = .0014]; volume intercept at 0 mm Hg (V0) of the LV end-diastolic pressure-volume relationship: ethanol 0.40 ± 0.03 mL vs control 0.31 ± 0.02 mL [P = .020]). However, there were no changes in systolic chamber function as indexed by LV endocardial fractional shortening or the slope of the LV systolic pressure-volume relationship (end systolic elastance). The percentage of myocardial apoptosis was correlated with the degree of LV dilation (% apoptosis vs LV EDD: r = 0.39; n = 38; P = .021; vs V0: r = 0.44; n = 19; P = .046). No differences in leukocyte or cardiac telomere length were noted between the ethanol and control groups. Furthermore, cardiac telomere length was not associated with indexes of LV dilation (LVEDD and V0) or cardiomyocyte apoptosis. CONCLUSIONS: Chronic ethanol-associated myocardial apoptosis and adverse remodeling occurs independently from changes in cardiac telomere length. Telomere shortening may not be a critical mechanism responsible for cardiomyocyte apoptosis and adverse cardiac remodeling.


Asunto(s)
Apoptosis/efectos de los fármacos , Etanol/toxicidad , Hipertrofia Ventricular Izquierda/inducido químicamente , Miocitos Cardíacos/efectos de los fármacos , Homeostasis del Telómero/efectos de los fármacos , Remodelación Ventricular/efectos de los fármacos , Animales , Apoptosis/fisiología , Etanol/administración & dosificación , Hipertrofia Ventricular Izquierda/patología , Masculino , Miocitos Cardíacos/patología , Miocitos Cardíacos/fisiología , Ratas , Ratas Sprague-Dawley , Homeostasis del Telómero/fisiología , Remodelación Ventricular/fisiología
17.
Development ; 141(2): 335-45, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24353062

RESUMEN

Organ development is a highly regulated process involving the coordinated proliferation and differentiation of diverse cellular populations. The pathways regulating cell proliferation and their effects on organ growth are complex and for many organs incompletely understood. In all vertebrate species, the cardiac natriuretic peptides (ANP and BNP) are produced by cardiomyocytes in the developing heart. However, their role during cardiogenesis is not defined. Using the embryonic zebrafish and neonatal mammalian cardiomyocytes we explored the natriuretic peptide signaling network during myocardial development. We observed that the cardiac natriuretic peptides ANP and BNP and the guanylate cyclase-linked natriuretic peptide receptors Npr1 and Npr2 are functionally redundant during early cardiovascular development. In addition, we demonstrate that low levels of the natriuretic peptides preferentially activate Npr3, a receptor with Gi activator sequences, and increase cardiomyocyte proliferation through inhibition of adenylate cyclase. Conversely, high concentrations of natriuretic peptides reduce cardiomyocyte proliferation through activation of the particulate guanylate cyclase-linked natriuretic peptide receptors Npr1 and Npr2, and activation of protein kinase G. These data link the cardiac natriuretic peptides in a complex hierarchy modulating cardiomyocyte numbers during development through opposing effects on cardiomyocyte proliferation mediated through distinct cyclic nucleotide signaling pathways.


Asunto(s)
Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Receptores del Factor Natriurético Atrial/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/embriología , Pez Cebra/metabolismo , Animales , Animales Modificados Genéticamente , Factor Natriurético Atrial/genética , Factor Natriurético Atrial/metabolismo , Proliferación Celular , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Técnicas de Silenciamiento del Gen , Corazón/embriología , Péptido Natriurético Encefálico/genética , Péptido Natriurético Encefálico/metabolismo , Receptores del Factor Natriurético Atrial/antagonistas & inhibidores , Receptores del Factor Natriurético Atrial/genética , Transducción de Señal , Pez Cebra/genética , Proteínas de Pez Cebra/antagonistas & inhibidores , Proteínas de Pez Cebra/genética
18.
Circ Cardiovasc Genet ; 6(4): 317-26, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23861363

RESUMEN

BACKGROUND: Whole exome sequencing is a powerful technique for Mendelian disease gene discovery. However, variant prioritization remains a challenge. We applied whole exome sequencing to identify the causal variant in a large family with familial dilated cardiomyopathy of unknown pathogenesis. METHODS AND RESULTS: A large family with autosomal dominant, familial dilated cardiomyopathy was identified. Exome capture and sequencing were performed in 3 remotely related, affected subjects predicted to share <0.1% of their genomes by descent. Shared variants were filtered for rarity, evolutionary conservation, and predicted functional significance, and remaining variants were filtered against 71 locally generated exomes. Variants were also prioritized using the Variant Annotation Analysis and Search Tool. Final candidates were validated by Sanger sequencing and tested for segregation. There were 664 shared heterozygous nonsense, missense, or splice site variants, of which 26 were rare (minor allele frequency ≤0.001 or not reported) in 2 public databases. Filtering against internal exomes reduced the number of candidates to 2, and of these, a single variant (c.1907 G>A) in RBM20, segregated with disease status and was absent in unaffected internal reference exomes. Bioinformatic prioritization with Variant Annotation Analysis and Search Tool supported this result. CONCLUSIONS: Whole exome sequencing of remotely related dilated cardiomyopathy subjects from a large, multiplex family, followed by systematic filtering, identified a causal RBM20 mutation without the need for linkage analysis.


Asunto(s)
Cardiomiopatía Dilatada/genética , Exoma , Proteínas de Unión al ARN/genética , Adolescente , Adulto , Alelos , Cardiomiopatía Dilatada/patología , Niño , Preescolar , Estudios de Cohortes , Biología Computacional , Bases de Datos Genéticas , Femenino , Frecuencia de los Genes , Ligamiento Genético , Genotipo , Heterocigoto , Humanos , Masculino , Persona de Mediana Edad , Mutación Missense , Linaje , Análisis de Secuencia de ADN , Adulto Joven
19.
Cardiovasc Res ; 93(3): 463-70, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22198505

RESUMEN

AIMS: Despite increased understanding of the fundamental biology regulating cardiomyocyte hypertrophy and heart failure, it has been challenging to find novel chemical or genetic modifiers of these pathways. Traditional cell-based methods do not model the complexity of an intact cardiovascular system and mammalian models are not readily adaptable to chemical or genetic screens. Our objective was to create an in vivo model suitable for chemical and genetic screens for hypertrophy and heart failure modifiers. METHODS AND RESULTS: Using the developing zebrafish, we established that the cardiac natriuretic peptide genes (nppa and nppb), known markers of cardiomyocyte hypertrophy and heart failure, were induced in the embryonic heart by pathological cardiac stimuli. This pathological induction was distinct from the developmental regulation of these genes. We created a luciferase-based transgenic reporter line that accurately modelled the pathological induction patterns of the zebrafish nppb gene. Utilizing this reporter line, we were able to show remarkable conservation of pharmacological responses between the larval zebrafish heart and adult mammalian models. CONCLUSION: By performing a focused screen of chemical agents, we were able to show a distinct response of a genetic model of hypertrophic cardiomyopathy to the histone deacetylase inhibitor, Trichostatin A, and the mitogen-activated protein kinase kinase 1/2 inhibitor, U0126. We believe this in vivo reporter line will offer a unique approach to the identification of novel chemical or genetic regulators of myocardial hypertrophy and heart failure.


Asunto(s)
Cardiomiopatía Hipertrófica/genética , Marcadores Genéticos/genética , Pruebas Genéticas/métodos , Luciferasas/genética , Péptidos Natriuréticos/genética , Pez Cebra/genética , Animales , Animales Modificados Genéticamente , Butadienos/farmacología , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Regulación del Desarrollo de la Expresión Génica/fisiología , Genes Reporteros/genética , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Ácidos Hidroxámicos/farmacología , Mamíferos , Péptidos Natriuréticos/metabolismo , Nitrilos/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Pez Cebra/embriología
20.
Dis Model Mech ; 4(3): 400-10, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21245263

RESUMEN

To assess the effects during cardiac development of mutations that cause human cardiomyopathy, we modeled a sarcomeric gene mutation in the embryonic zebrafish. We designed morpholino antisense oligonucleotides targeting the exon 13 splice donor site in the zebrafish cardiac troponin T (tnnt2) gene, in order to precisely recapitulate a human TNNT2 mutation that causes hypertrophic cardiomyopathy (HCM). HCM is a disease characterized by myocardial hypertrophy, myocyte and myofibrillar disarray, as well as an increased risk of sudden death. Similar to humans with HCM, the morphant zebrafish embryos displayed sarcomere disarray and there was a robust induction of myocardial hypertrophic pathways. Microarray analysis uncovered a number of shared transcriptional responses between this zebrafish model and a well-characterized mouse model of HCM. However, in contrast to adult hearts, these embryonic hearts developed cardiomyocyte hyperplasia in response to this genetic perturbation. The re-creation of a human disease-causing TNNT2 splice variant demonstrates that sarcomeric mutations can alter cardiomyocyte biology at the earliest stages of heart development with distinct effects from those observed in adult hearts despite shared transcriptional responses.


Asunto(s)
Cardiomiopatía Hipertrófica/genética , Cardiomiopatía Hipertrófica/patología , Corazón/embriología , Mutación/genética , Miocitos Cardíacos/patología , Organogénesis , Pez Cebra/embriología , Empalme Alternativo/efectos de los fármacos , Empalme Alternativo/genética , Secuencia de Aminoácidos , Animales , Calcio/metabolismo , Cardiomiopatía Hipertrófica/complicaciones , Cardiomiopatía Hipertrófica/fisiopatología , Embrión no Mamífero/efectos de los fármacos , Embrión no Mamífero/metabolismo , Embrión no Mamífero/patología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Corazón/efectos de los fármacos , Corazón/fisiopatología , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/patología , Ventrículos Cardíacos/fisiopatología , Humanos , Hiperplasia/complicaciones , Hiperplasia/patología , Ratones , Datos de Secuencia Molecular , Contracción Miocárdica/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Oligonucleótidos Antisentido/farmacología , Organogénesis/efectos de los fármacos , Sarcómeros/efectos de los fármacos , Sarcómeros/patología , Transcripción Genética/efectos de los fármacos , Troponina T/química , Troponina T/genética , Pez Cebra/genética , Proteínas de Pez Cebra/química , Proteínas de Pez Cebra/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...