Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Gastroenterol Hepatol ; 38(8): 1325-1332, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37096760

RESUMEN

BACKGROUND AND AIM: Acute-on-chronic liver failure (ACLF) is distinct from acute decompensation (AD) of cirrhosis in its clinical presentation, pathophysiology, and prognosis. There are limited published Australian ACLF data. METHODS: We performed a single-center retrospective cohort study of all adults with cirrhosis admitted with a decompensating event to a liver transplantation (LT) centre between 2015 and 2020. ACLF was defined using the European Association for the Study of the Liver-Chronic Liver Failure (EASL-CLIF) definition while those who did not meet the definition were classified as AD. The primary outcome of interest was 90-day LT-free survival. RESULTS: A total of 615 patients had 1039 admissions for a decompensating event. On their index admission, 34% (209/615) of patients were classified as ACLF. Median admission model for end-stage liver disease (MELD) and MELD-Na scores were higher in ACLF patients compared with AD (21 vs 17 and 25 vs 20 respectively, both P < 0.001). Both the presence and severity of ACLF (grade ≥ 2) significantly predicted worse LT-free survival compared with patients with AD. The EASL-CLIF ACLF score (CLIF-C ACLF), MELD and MELD-Na scores performed similarly in predicting 90-day mortality. Patients with index ACLF had a higher risk of 28-day mortality (28.1% vs 5.1%, P < 0.001) and shorter times to readmission compared with those with AD. CONCLUSION: ACLF complicates over a third of hospital admissions for cirrhosis with decompensating events and is associated with a high short-term mortality. The presence and grade of ACLF predicts 90-day mortality and should be identified as those at greatest risk of poor outcome without intervention such as LT.


Asunto(s)
Insuficiencia Hepática Crónica Agudizada , Enfermedad Hepática en Estado Terminal , Adulto , Humanos , Insuficiencia Hepática Crónica Agudizada/diagnóstico , Insuficiencia Hepática Crónica Agudizada/epidemiología , Insuficiencia Hepática Crónica Agudizada/etiología , Estudios Retrospectivos , Enfermedad Hepática en Estado Terminal/complicaciones , Índice de Severidad de la Enfermedad , Australia/epidemiología , Cirrosis Hepática/complicaciones , Cirrosis Hepática/epidemiología , Pronóstico
2.
Int J Mol Sci ; 22(13)2021 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-34203369

RESUMEN

Hypertrophic cardiomyopathy (HCM) is an inherited cardiac disorder affecting one in 500 of the general population. Atrial fibrillation (AF) is the most common arrhythmia in patients with HCM. We sought to characterize the atrial electrophysiological and structural substrate in young and aging Gly203Ser cardiac troponin-I transgenic (HCM) mice. At 30 weeks and 50 weeks of age (n = 6 per strain each group), the left atrium was excised and placed on a multi-electrode array (MEA) for electrophysiological study; subsequent histological analyses and plasma samples were analyzed for biomarkers of extracellular matrix remodeling and cell adhesion and inflammation. Wild-type mice of matched ages were included as controls. Young HCM mice demonstrated significantly shortened atrial action potential duration (APD), increased conduction heterogeneity index (CHI), increased myocyte size, and increased interstitial fibrosis without changes in effective refractory periods (ERP), conduction velocity (CV), inflammatory infiltrates, or circulating markers of extracellular matrix remodeling and inflammation. Aging HCM mice demonstrated aggravated changes in atria electrophysiology and structural remodeling as well as increased circulating matrix metalloproteinases (MMP)-2, MMP-3, and VCAM-1 levels. This model of HCM demonstrates an underlying atrial substrate that progresses with age and may in part be responsible for the greater propensity for AF in HCM.


Asunto(s)
Fibrilación Atrial/metabolismo , Cardiomiopatía Hipertrófica/metabolismo , Atrios Cardíacos/metabolismo , Troponina I/metabolismo , Potenciales de Acción/fisiología , Animales , Fibrilación Atrial/genética , Remodelación Atrial/genética , Remodelación Atrial/fisiología , Presión Sanguínea/fisiología , Electrofisiología Cardíaca , Cardiomiopatía Hipertrófica/genética , Modelos Animales de Enfermedad , Electrofisiología , Femenino , Atrios Cardíacos/patología , Frecuencia Cardíaca/fisiología , Humanos , Masculino , Mutación , Troponina I/genética
3.
J Physiol ; 594(14): 4051-70, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-27062056

RESUMEN

KEY POINTS: Genetic mutations in cardiac troponin I (cTnI) are associated with development of hypertrophic cardiomyopathy characterized by myocyte remodelling, disorganization of cytoskeletal proteins and altered energy metabolism. The L-type Ca(2+) channel is the main route for calcium influx and is crucial to cardiac excitation and contraction. The channel also regulates mitochondrial function in the heart by a functional communication between the channel and mitochondria via the cytoskeletal network. We find that L-type Ca(2+) channel kinetics are altered in cTnI-G203S cardiac myocytes and that activation of the channel causes a significantly greater increase in mitochondrial membrane potential and metabolic activity in cTnI-G203S cardiac myocytes. These responses occur as a result of impaired communication between the L-type Ca(2+) channel and cytoskeletal protein F-actin, involving decreased movement of actin-myosin and block of the mitochondrial voltage-dependent anion channel, resulting in a 'hypermetabolic' mitochondrial state. We propose that L-type Ca(2+) channel antagonists, such as diltiazem, might be effective in reducing the cardiomyopathy by normalizing mitochondrial metabolic activity. ABSTRACT: Genetic mutations in cardiac troponin I (cTnI) account for 5% of families with hypertrophic cardiomyopathy. Hypertrophic cardiomyopathy is associated with disorganization of cytoskeletal proteins and altered energy metabolism. The L-type Ca(2+) channel (ICa-L ) plays an important role in regulating mitochondrial function. This involves a functional communication between the channel and mitochondria via the cytoskeletal network. We investigate the role of ICa-L in regulating mitochondrial function in 25- to 30-week-old cardiomyopathic mice expressing the human disease-causing mutation Gly203Ser in cTnI (cTnI-G203S). The inactivation rate of ICa-L is significantly faster in cTnI-G203S myocytes [cTnI-G203S: τ1  = 40.68 ± 3.22, n = 10 vs. wild-type (wt): τ1  = 59.05 ± 6.40, n = 6, P < 0.05]. Activation of ICa-L caused a greater increase in mitochondrial membrane potential (Ψm , 29.19 ± 1.85%, n = 15 vs. wt: 18.84 ± 2.01%, n = 10, P < 0.05) and metabolic activity (24.40 ± 6.46%, n = 8 vs. wt: 9.98 ± 1.57%, n = 9, P < 0.05). The responses occurred because of impaired communication between ICa-L and F-actin, involving lack of dynamic movement of actin-myosin and block of the mitochondrial voltage-dependent anion channel. Similar responses were observed in precardiomyopathic mice. ICa-L antagonists nisoldipine and diltiazem decreased Ψm to basal levels. We conclude that the Gly203Ser mutation leads to impaired functional communication between ICa-L and mitochondria, resulting in a 'hypermetabolic' state. This might contribute to development of cTnI-G203S cardiomyopathy because the response is present in young precardiomyopathic mice. ICa-L antagonists might be effective in reducing the cardiomyopathy by altering mitochondrial function.


Asunto(s)
Canales de Calcio Tipo L/fisiología , Cardiomiopatía Hipertrófica/fisiopatología , Mitocondrias Cardíacas/fisiología , Actinas/fisiología , Animales , Calcio/fisiología , Bloqueadores de los Canales de Calcio/farmacología , Cardiomiopatía Hipertrófica/genética , Citoesqueleto/fisiología , Diltiazem/farmacología , Modelos Animales de Enfermedad , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Mitocondrias Cardíacas/efectos de los fármacos , Mitocondrias Cardíacas/metabolismo , Mutación , Miocitos Cardíacos/fisiología , Nisoldipino/farmacología , Superóxidos/metabolismo , Troponina I/genética
4.
Clin Exp Pharmacol Physiol ; 43(1): 95-101, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26444142

RESUMEN

Hypertrophic cardiomyopathy (HCM) is a common heritable cardiac disorder with diverse clinical outcomes including sudden death, heart failure, and stroke. Depressed heart rate variability (HRV), a measure of cardiac autonomic regulation, has been shown to predict mortality in patients with cardiovascular disease. Cardiac autonomic remodelling in animal models of HCM are not well characterised. This study analysed Gly203Ser cardiac troponin-I transgenic (TG) male mice previously demonstrated to develop hallmarks of HCM by age 21 weeks. 33 mice aged 30 and 50 weeks underwent continuous electrocardiogram (ECG) recording for 30 min under anaesthesia. TG mice demonstrated prolonged P-wave duration (P < 0.001) and PR intervals (P < 0.001) compared to controls. Additionally, TG mice demonstrated depressed standard deviation of RR intervals (SDRR; P < 0.01), coefficient of variation of RR intervals (CVRR; P < 0.001) and standard deviation of heart rate (SDHR; P < 0.001) compared to controls. Additionally, total power was significantly reduced in TG mice (P < 0.05). No significant age-related difference in either strain was observed in ECG or HRV parameters. Mice with HCM developed slowed atrial and atrioventricular conduction and depressed HRV. These changes were conserved with increasing age. This finding may be indicative of atrial and ventricular hypertrophy or dysfunction, and perhaps an indication of worse clinical outcome in heart failure progression in HCM patients.


Asunto(s)
Cardiomiopatía Hipertrófica/fisiopatología , Atrios Cardíacos/fisiopatología , Sistema de Conducción Cardíaco/fisiopatología , Frecuencia Cardíaca , Ventrículos Cardíacos/fisiopatología , Animales , Modelos Animales de Enfermedad , Electrocardiografía , Femenino , Cinética , Masculino , Ratones , Ratones Endogámicos C57BL
5.
Physiol Genomics ; 47(3): 75-87, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25547111

RESUMEN

Intrinsic valvular degeneration and dysfunction is the most common complication of bicuspid aortic valve (BAV) disease. Phenotypically, it ranges from calcific aortic stenosis to redundant or prolapsing regurgitant leaflets. The underlying molecular mechanism underpinning phenotype heterogeneity of valvular degeneration in BAV is poorly understood. We used RNA sequencing (RNA-seq) to identify genes and pathways responsible for the development of valvular degeneration in BAV, compared with tricuspid aortic valve (TAV). Comparative transcriptome analysis was performed on total RNA of aortic valve tissues of patients with diseased BAV (n = 5) and calcified TAV (n = 3). RNA-seq findings were validated by RT-qPCR. A total of 59 and 177 genes were significantly up- and downregulated, respectively, in BAV compared with TAV. Hierarchical clustering indicated heterogeneity within the BAV group, separating those with heavy calcification (BAVc) from those with redundant leaflets and/or minimal calcification (BAVr). Interestingly, the gene expression profile of the BAVc group closely resembled the TAV, with shared up- and downregulation of inflammatory and NOTCH1 signaling pathways, respectively. Downregulation of matrix protease ADAMTS9 and protein aggrecan were observed in BAVr compared with TAV. Dysregulation of fetal gene programs were also present, with notable downregulation of SEMA6B and SEMA3F in BAVr and BAVc compared with TAV, respectively. Upregulation of TBX20 was observed exclusively in BAVr compared with BAVc. In conclusion, diverging molecular mechanisms underpin phenotype heterogeneity of valvular degeneration in BAV and data from the present study suggest that there may be shared mechanisms leading to calcification in BAV and TAV. Recognition of these pathways is fundamental to improve our understanding of the molecular basis of human BAV disease.


Asunto(s)
Válvula Aórtica/anomalías , Perfilación de la Expresión Génica , Enfermedades de las Válvulas Cardíacas/genética , Análisis de Secuencia de ARN/métodos , Anciano , Válvula Aórtica/metabolismo , Válvula Aórtica/patología , Enfermedad de la Válvula Aórtica Bicúspide , Estudios de Cohortes , Regulación hacia Abajo/genética , Ontología de Genes , Redes Reguladoras de Genes , Enfermedades de las Válvulas Cardíacas/metabolismo , Enfermedades de las Válvulas Cardíacas/patología , Humanos , Masculino , Persona de Mediana Edad , Reacción en Cadena en Tiempo Real de la Polimerasa , Reproducibilidad de los Resultados , Regulación hacia Arriba/genética
6.
J Mol Cell Cardiol ; 62: 164-78, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23756156

RESUMEN

Hypertrophic cardiomyopathy (HCM) is the most common inherited primary myocardial disorder. HCM is characterized by interstitial fibrosis and excessive accumulation of extracellular matrix (ECM) proteins. Fibrosis in HCM has been associated with impaired cardiac function and heart failure, and has been considered a key substrate for ventricular arrhythmias and sudden death. The molecular triggers underpinning ECM production are not well established. We have previously developed a double-mutant mouse model of HCM that recapitulates the phenotype seen in humans with multiple mutations, including earlier onset of the disease, progression to a dilated phenotype, severe heart failure and premature mortality. The present study investigated the expression of ECM-encoding genes in severe HCM and heart failure. Significant upregulation of structural Fn1, regulatory Mmp14, Timp1, Serpin3A, SerpinE1, SerpineE2, Tgfß1, and Tgfß2; and matricellular Ccn2, Postn, Spp1, Thbs1, Thbs4, and Tnc was evident from the early, pre-phenotype stage. Non-myocytes expressed ECM genes at higher levels than cardiomyocytes in normal and diseased hearts. Synchronous increase of secreted CCN2 and TIMP1 plasma levels and decrease of MMP3 levels were observed in end-stage disease. CCN2 protein expression was increased from early disease in double-mutant hearts and played an important role in ECM responses. It was a powerful modulator of ECM regulatory (Timp1 and SerpinE1) and matricellular protein-encoding (Spp1, Thbs1, Thbs4 and Tnc) gene expression in cardiomyocytes when added exogenously in vitro. Modulation of CCN2 (CTGF, connective tissue growth factor) and associated early ECM changes may represent a new therapeutic target in the treatment and prevention of heart failure in HCM.


Asunto(s)
Cardiomiopatía Hipertrófica/metabolismo , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Matriz Extracelular/metabolismo , Insuficiencia Cardíaca/metabolismo , Animales , Cardiomiopatía Hipertrófica/genética , Células Cultivadas , Factor de Crecimiento del Tejido Conjuntivo/genética , Ensayo de Inmunoadsorción Enzimática , Insuficiencia Cardíaca/genética , Ventrículos Cardíacos/metabolismo , Inmunohistoquímica , Masculino , Ratones , Miocitos Cardíacos/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
7.
J Biol Chem ; 286(2): 1576-87, 2011 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-21056972

RESUMEN

Excessive proliferation and stabilization of the microtubule (MT) array in cardiac myocytes can accompany pathological cardiac hypertrophy, but the molecular control of these changes remains poorly characterized. In this study, we examined MT stabilization in two independent murine models of heart failure and revealed increases in the levels of post-translationally modified stable MTs, which were closely associated with STAT3 activation. To explore the molecular signaling events contributing to control of the cardiac MT network, we stimulated cardiac myocytes with an α-adrenergic agonist phenylephrine (PE), and observed increased tubulin content without changes in detyrosinated (glu-tubulin) stable MTs. In contrast, the hypertrophic interleukin-6 (IL6) family cytokines increased both the glu-tubulin content and glu-MT density. When we examined a role for ERK in regulating cardiac MTs, we showed that the MEK/ERK-inhibitor U0126 increased glu-MT density in either control cardiac myocytes or following exposure to hypertrophic agents. Conversely, expression of an activated MEK1 mutant reduced glu-tubulin levels. Thus, ERK signaling antagonizes stabilization of the cardiac MT array. In contrast, inhibiting either JAK2 with AG490, or STAT3 signaling with Stattic or siRNA knockdown, blocked cytokine-stimulated increases in glu-MT density. Furthermore, the expression of a constitutively active STAT3 mutant triggered increased glu-MT density in the absence of hypertrophic stimulation. Thus, STAT3 activation contributes substantially to cytokine-stimulated glu-MT changes. Taken together, our results highlight the opposing actions of STAT3 and ERK pathways in the regulation of MT changes associated with cardiac myocyte hypertrophy.


Asunto(s)
Cardiomegalia/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Microtúbulos/metabolismo , Factor de Transcripción STAT3/metabolismo , Animales , Cardiomegalia/patología , Cardiomiopatía Hipertrófica/metabolismo , Cardiomiopatía Hipertrófica/patología , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Interleucina-6/metabolismo , Masculino , Ratones , Ratones Endogámicos , Ratones Transgénicos , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Procesamiento Proteico-Postraduccional/fisiología , ARN Interferente Pequeño , Ratas , Ratas Sprague-Dawley , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo
8.
DEN Open ; 2(1): e44, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35310703

RESUMEN

Objectives: Colonoscopy is the gold standard diagnostic test used to detect early colorectal lesions and prevent colorectal carcinoma. Narrow band imaging (NBI) is an imaging technique that provides improved image resolution of the mucosa during endoscopy. Whether NBI improves the detection of sessile serrated lesion (SSL) is controversial-our aim was to assess this during routine colonoscopy. Methods: We conducted a multicenter, prospective, randomized, controlled trial. Patients underwent colonoscopy for screening, surveillance, or symptoms. They were randomized to either high-definition white light (HD-WL) or NBI in a 1:1 ratio. The primary outcome was SSL detection rate. Secondary outcomes were adenoma detection rate (ADR) and polyp detection rate (PDR). Results: A total of 400 patients were randomized to NBI (N = 200) or HD-WL (N = 200). The total colonoscopy time was slightly longer in the NBI group compared to HD-WL (median time 14 vs. 12 min, p = 0.033). There were no statistically significant differences in SSL detection rate (7.5% NBI vs. 8.0% HD-WL; p = 0.852), ADR (41.0% NBI vs. 37.5% HD-WL; p = 0.531), or PDR (61.0% NBI vs. 54.0% HD-WL; p = 0.157) between the two groups. No significant predictors of SSL detection were found on univariable or multivariable analysis. Increasing age and increased withdrawal time were an independent predictors of polyp detection and increasing age was also an independent predictor of adenoma detection on multivariable analysis. Conclusion: In the hands of experienced colonoscopists, NBI does not improve SSL detection compared to HD-WL. Withdrawal time and patient age remain important factors for polyp and adenoma detection.

9.
Transplant Direct ; 8(7): e1346, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35706607

RESUMEN

Background: Controversy exists regarding the best predictive model of liver transplant waiting list (WL) mortality. Models for end-stage liver disease-glomerular filtration rate assessment in liver disease (MELD-GRAIL) and MELD-GRAIL-Na were recently described to provide better prognostication, particularly in females. We evaluated the performance of these scores compared to MELD and MELD-Na. Methods: Consecutive patients with cirrhosis waitlisted for liver transplant from 1998 to 2017 were examined in this single-center study. The primary outcome was 90-d WL mortality. MELD, MELD-Na, MELD-GRAIL, and MELD-GRAIL-Na at the time of WL registration were compared. Model discrimination was assessed with area under the receiver operating characteristic curves and Harrell's C-index after fitting Cox models. Model calibration was examined with Grønnesby and Borgan's modification of the Hosmer-Lemeshow formula and by comparing predicted/observed outcomes across model strata. Results: The study population comprised 1108 patients with a median age of 53.5 (interquartile range 48-59) y and male predominance (74.9%). All models had excellent areas under the receiver operating characteristic curves for the primary outcome (MELD 0.89, MELD-Na 0.91, MELD-GRAIL 0.89, MELD-GRAIL-Na 0.89; all comparisons P > 0.05). Youden index cutoffs for 90-d mortality were as follows: MELD, 19; MELD-Na, 22; MELD-GRAIL, 18; and MELD-GRAIL-Na, 17. Variables associated with 90-d mortality on multivariable Cox regression were sodium, bilirubin, creatinine, and international normalized ratio. There were no differences in model discrimination using Harrell's C-index. All models were well calibrated; however, divergence between observed and predicted mortality was noted with scores ≥25. Conclusion: There were no demonstrable differences in discrimination or calibration of GRAIL-based models compared with MELD or MELD-Na in our cohort. This suggests that GRAIL-based models may not have meaningful improvements in discriminatory ability when applied to other settings.

10.
J Mol Cell Cardiol ; 48(5): 1014-22, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-19715700

RESUMEN

Hypertrophic cardiomyopathy (HCM) is a genetic disorder caused by mutations in genes encoding sarcomere proteins. The mechanisms involved in the development of cardiac hypertrophy and heart failure remain poorly understood. Global proteomic profiling was used to study the cardiac proteome of mice predisposed to developing HCM. Hearts from three groups of mice (n=3 hearts per group) were studied: non-transgenic (NTG) and cardiac-specific transgenic models over-expressing either the normal (TnI(WT)) or a mutant cardiac troponin I gene (Gly203Ser; TnI(G203S)). Two-dimensional gel electrophoresis (2-DE) coupled with tandem mass spectrometry was used to identify proteins. Image analysis was performed using Progenesis SameSpots. A total of 34 proteins with at least a twofold change in the TnI(G203S) mouse model were identified. Alterations were detected in components involved in energy production, Ca(2+) handling, and cardiomyocyte structure. Expression level changes in cytoskeletal and contractile proteins were well represented in the study, including the intermediate filament protein desmin, which was further investigated in two additional physiological and pathological settings, i.e., exercise treatment, and severe heart failure in a novel double-mutant TnI-203/MHC-403 model of HCM. This study highlights the potential role of tissue proteomic profiling for mapping proteins, which may be critical in cardiac dysfunction and progression to heart failure in HCM.


Asunto(s)
Cardiomiopatía Hipertrófica/metabolismo , Regulación de la Expresión Génica , Miocardio/metabolismo , Proteínas/metabolismo , Animales , Western Blotting , Cromatografía Liquida , Electroforesis en Gel Bidimensional , Perfilación de la Expresión Génica , Ratones , Ratones Transgénicos , Espectrometría de Masas en Tándem , Troponina I/genética , Troponina I/metabolismo
11.
Am J Physiol Cell Physiol ; 297(6): C1490-500, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19625611

RESUMEN

Diabetic cardiomyopathy is characterized by interstitial fibrosis and cardiomyocyte hypertrophy and apoptosis. Also known as CCN2, connective tissue growth factor (CTGF) is implicated in the fibrosis; however, whether it contributes to cardiomyocytes changes and adverse effects of high glucose and lipids on these cells remains unknown. Hearts from streptozotocin-induced diabetic rats had elevated CTGF and changes of pathological myocardial hypertrophy, fibrosis, and cardiomyocyte apoptosis. Rat H9c2 cardiomyocytes were then treated with recombinant human (rh)CTGF, high glucose, or the saturated free fatty acid palmitate. Each reagent induced cell hypertrophy, as indicated by the ratio of total protein to cell number, cell size, and gene expression of cardiac hypertrophy marker genes atrial natriuretic peptide (ANP), and alpha-skeletal actin. Each treatment also caused apoptosis measured by increased caspase3/7 activity, apoptotic cells by transferase-mediated dUTP nick end labeling (TUNEL) assay, and lower viable cell number. Further studies showed CTGF mRNA was rapidly induced by high glucose and palmitate in H9c2 cells and in mouse neonatal cardiomyocyte primary cultures. small interfering RNA against CTGF blocked the high glucose and palmitate induction of hypertrophy and apoptosis. In addition, these CTGF effects were through the tyrosine kinase A (TrkA) receptor with tyrosine kinase activity, which has previously been implicated in CTGF signaling: TrkA was phosphorylated by CTGF, and a specific TrkA blocker abrogated CTGF-induced effects on hypertrophy and apoptosis. For the first time in any system, fatty acid is newly identified as a regulator of CTGF, and this work implicates autocrine CTGF as a mediator of adverse effects of high glucose and fatty acids in cardiomyocytes.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Glucosa/administración & dosificación , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Palmitatos/farmacología , Animales , Animales Recién Nacidos , Apoptosis , Cardiomegalia/etiología , Tamaño de la Célula/efectos de los fármacos , Células Cultivadas , Factor de Crecimiento del Tejido Conjuntivo/farmacología , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/fisiopatología , Relación Dosis-Respuesta a Droga , Humanos , Hipertrofia , Masculino , Ratones , Ratones Endogámicos C57BL , Miocardio/patología , Miocitos Cardíacos/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptor trkA/metabolismo , Proteínas Recombinantes/farmacología
12.
Circulation ; 117(14): 1820-31, 2008 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-18362229

RESUMEN

BACKGROUND: Familial hypertrophic cardiomyopathy (FHC) is characterized by genetic and clinical heterogeneity. Five percent of FHC families have 2 FHC-causing mutations, which results in earlier disease onset, increased cardiac dysfunction, and a higher incidence of sudden death events. These observations suggest a relationship between the number of gene mutations and phenotype severity in FHC. METHODS AND RESULTS: We sought to develop, characterize, and investigate the pathogenic mechanisms in a double-mutant murine model of FHC. This model (designated TnI-203/MHC-403) was generated by crossbreeding mice with the Gly203Ser cardiac troponin I (TnI-203) and Arg403Gln alpha-myosin heavy chain (MHC-403) FHC-causing mutations. The mortality rate in TnI-203/MHC-403 mice was 100% by age 21 days. At age 14 days, TnI-203/MHC-403 mice developed a significantly increased ratio of heart weight to body weight, marked interstitial myocardial fibrosis, and increased expression of atrial natriuretic factor and brain natriuretic peptide compared with nontransgenic, TnI-203, and MHC-403 littermates. By age 16 to 18 days, TnI-203/MHC-403 mice rapidly developed a severe dilated cardiomyopathy and heart failure, with inducibility of ventricular arrhythmias, which led to death by 21 days. Downregulation of mRNA levels of key regulators of Ca(2+) homeostasis in TnI-203/MHC-403 mice was observed. Increased levels of phosphorylated STAT3 were observed in TnI-203/MHC-403 mice and corresponded with the onset of disease, which suggests a possible cardioprotective response. CONCLUSIONS: TnI-203/MHC-403 double-mutant mice develop a severe cardiac phenotype characterized by heart failure and early death. The presence of 2 disease-causing mutations may predispose individuals to a greater risk of developing severe heart failure than human FHC caused by a single gene mutation.


Asunto(s)
Cardiomiopatía Dilatada/genética , Cardiomiopatía Hipertrófica Familiar/genética , Modelos Animales de Enfermedad , Mutación , Troponina I/genética , Miosinas Ventriculares/genética , Animales , Señalización del Calcio , Cardiomiopatía Dilatada/patología , Cardiomiopatía Hipertrófica Familiar/fisiopatología , Progresión de la Enfermedad , Femenino , Heterocigoto , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Mutación Missense , Comunicación Paracrina , Fenotipo , Sistema Renina-Angiotensina/fisiología , Factor de Transcripción STAT3/fisiología , Transducción de Señal , Miosinas Ventriculares/fisiología
13.
Clin Exp Pharmacol Physiol ; 35(11): 1349-57, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18761664

RESUMEN

1. Familial hypertrophic cardiomyopathy (FHC) is a primary cardiac disorder characterized by myocardial hypertrophy that demonstrates substantial diversity in both genetic causes and clinical manifestations. 2. Clinical heterogeneity can be explained by the causative gene (at least 13 have been identified to date), the position of the amino acid residue affected by a mutation within the protein (over 450 mutations have been reported to date) and modifying genetic and environmental factors. 3. Multiple mutations are found in up to 5% of human FHC cases, who typically present with a more severe phenotype compared with single-mutation carriers (i.e. earlier onset of disease, greater left ventricular hypertrophy and a higher incidence of sudden cardiac death events). 4. Multiple mutations usually involve MYH7, MYBPC3 and, to a lesser extent, TNNI2, reflecting the higher contribution of mutations in these genes to FHC. 5. Multiple-mutation mouse models appear to mimic the human multiple-mutation phenotype and, thus, will help improve our understanding of disease pathogenesis. The models provide a tool for future studies of disease mechanisms and signalling pathways in FHC and its sequelae (i.e. heart failure and sudden death), thereby allowing identification of novel targets for potential therapies and disease prevention strategies.


Asunto(s)
Cardiomiopatía Hipertrófica Familiar/diagnóstico , Cardiomiopatía Hipertrófica Familiar/genética , Insuficiencia Cardíaca/diagnóstico , Insuficiencia Cardíaca/genética , Mutación/genética , Animales , Cardiomiopatía Hipertrófica Familiar/patología , Insuficiencia Cardíaca/patología , Humanos , Índice de Severidad de la Enfermedad
14.
JACC Basic Transl Sci ; 1(1-2): 61-72, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-30167506

RESUMEN

Heterozygous mice (αMHC403/+ ) expressing the human disease-causing mutation Arg403Gln exhibit cardinal features of hypertrophic cardiomyopathy (HCM) including hypertrophy, myocyte disarray, and increased myocardial fibrosis. Treatment of αMHC403/+ mice with the L-type calcium channel (ICa-L) antagonist diltiazem has been shown to decrease left ventricular anterior wall thickness, cardiac myocyte hypertrophy, disarray, and fibrosis. However, the role of the ICa-L in the development of HCM is not known. In addition to maintaining cardiac excitation and contraction in myocytes, the ICa-L also regulates mitochondrial function through transmission of movement of ICa-L via cytoskeletal proteins to mitochondrial voltage-dependent anion channel. Here, the authors investigated the role of ICa-L in regulating mitochondrial function in αMHC403/+ mice. Whole-cell patch clamp studies showed that ICa-L current inactivation kinetics were significantly increased in αMHC403/+ cardiac myocytes, but that current density and channel expression were similar to wild-type cardiac myocytes. Activation of ICa-L caused a significantly greater increase in mitochondrial membrane potential and metabolic activity in αMHC403/+ . These increases were attenuated with ICa-L antagonists and following F-actin or ß-tubulin depolymerization. The authors observed increased levels of fibroblast growth factor-21 in αMHC403/+ mice, and altered mitochondrial DNA copy number consistent with altered mitochondrial activity and the development of cardiomyopathy. These studies suggest that the Arg403Gln mutation leads to altered functional communication between ICa-L and mitochondria that is associated with increased metabolic activity, which may contribute to the development of cardiomyopathy. ICa-L antagonists may be effective in reducing the cardiomyopathy in HCM by altering metabolic activity.

15.
Cell Res ; 13(1): 9-20, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12643345

RESUMEN

Major advances have been made over the last decade in our understanding of the molecular basis of several cardiac conditions. Hypertrophic cardiomyopathy (HCM) was the first cardiac disorder in which a genetic basis was identified and as such, has acted as a paradigm for the study of an inherited cardiac disorder. HCM can result in clinical symptoms ranging from no symptoms to severe heart failure and premature sudden death. HCM is the commonest cause of sudden death in those aged less than 35 years, including competitive athletes. At least ten genes have now been identified, defects in which cause HCM. All of these genes encode proteins which comprise the basic contractile unit of the heart, i.e. the sarcomere. While much is now known about which genes cause disease and the various clinical presentations, very little is known about how these gene defects cause disease, and what factors modify the expression of the mutant genes. Studies in both cell culture and animal models of HCM are now beginning to shed light on the signalling pathways involved in HCM, and the role of both environmental and genetic modifying factors. Understanding these mechanisms will ultimately improve our knowledge of the basic biology of heart muscle function, and will therefore provide new avenues for treating cardiovascular disease in man.


Asunto(s)
Cardiomiopatía Hipertrófica/genética , Cardiomiopatía Hipertrófica/patología , Animales , Cardiomiopatías/genética , Cardiomiopatía Restrictiva/patología , Genotipo , Humanos , Masculino , Ratones , Mutación , Fenotipo , Conejos , Sarcómeros/fisiología
16.
PLoS One ; 7(9): e44744, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23024758

RESUMEN

MicroRNAs (miRNAs) regulate post-transcriptional gene expression during development and disease. We have determined the miRNA expression levels of early- and end-stage hypertrophic cardiomyopathy (HCM) in a severe, transgenic mouse model of the disease. Five miRNAs were differentially expressed at an early stage of HCM development. Time-course analysis revealed that decreased expression of miR-1 and miR-133a commences at a pre-disease stage, and precedes upregulation of target genes causal of cardiac hypertrophy and extracellular matrix remodelling, suggesting a role for miR-1 and miR-133a in early disease development. At end-stage HCM, 16 miRNA are dysregulated to form an expression profile resembling that of other forms of cardiac hypertrophy, suggesting common responses. Analysis of the mRNA transcriptome revealed that miRNAs potentially target 15.7% upregulated and 4.8% downregulated mRNAs at end-stage HCM, and regulate mRNAs associated with cardiac hypertrophy and electrophysiology, calcium signalling, fibrosis, and the TGF-ß signalling pathway. Collectively, these results define the miRNA expression signatures during development and progression of severe HCM and highlight critical miRNA regulated gene networks that are involved in disease pathogenesis.


Asunto(s)
Cardiomiopatía Hipertrófica/genética , Perfilación de la Expresión Génica , Insuficiencia Cardíaca/genética , Ventrículos Cardíacos/metabolismo , MicroARNs/genética , Animales , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Ventrículos Cardíacos/patología , Masculino , Ratones , Ratones Transgénicos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reproducibilidad de los Resultados , Transcripción Genética
17.
Nat Commun ; 2: 287, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21505443

RESUMEN

The epithelial sodium channel (ENaC) is essential for sodium homoeostasis in many epithelia. ENaC activity is required for lung fluid clearance in newborn animals and for maintenance of blood volume and blood pressure in adults. In vitro studies show that the ubiquitin ligase Nedd4-2 ubiquitinates ENaC to regulate its cell surface expression. Here we show that knockout of Nedd4-2 in mice leads to increased ENaC expression and activity in embryonic lung. This increased ENaC activity is the likely reason for premature fetal lung fluid clearance in Nedd4-2(-/-) animals, resulting in a failure to inflate lungs and perinatal lethality. A small percentage of Nedd4-2(-/-) animals survive up to 22 days, and these animals also show increased ENaC expression and develop lethal sterile inflammation of the lung. Thus, we provide critical in vivo evidence that Nedd4-2 is essential for correct regulation of ENaC expression, fetal and postnatal lung function and animal survival.


Asunto(s)
Complejos de Clasificación Endosomal Requeridos para el Transporte/deficiencia , Canales Epiteliales de Sodio/metabolismo , Enfermedades Pulmonares/genética , Pulmón/patología , Ubiquitina-Proteína Ligasas/deficiencia , Animales , Presión Sanguínea , Northern Blotting , Lavado Broncoalveolar , Quimiocinas/metabolismo , Citocinas/metabolismo , Embrión de Mamíferos/metabolismo , Embrión de Mamíferos/patología , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Immunoblotting , Pulmón/embriología , Pulmón/metabolismo , Enfermedades Pulmonares/mortalidad , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal , Microscopía Fluorescente , Ubiquitina-Proteína Ligasas Nedd4 , Ubiquitina-Proteína Ligasas/metabolismo
18.
Int J Cardiol ; 119(2): 245-8, 2007 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-17067697

RESUMEN

Clinical outcome in familial hypertrophic cardiomyopathy (FHC) may be influenced by modifying factors such as exercise. Transgenic mice which overexpress the human disease-causing cTnI gene mutation, Gly203Ser (designated cTnI-G203S), develop all the characteristic phenotypic features of FHC. To study the modifying effect of exercise in early disease, mice underwent swimming exercise at an early age prior to the development of the FHC phenotype. In non-transgenic and cTnI-wt mice, swimming resulted in a significant increase in left ventricular wall thickness and contractility on echocardiography, consistent with a physiological hypertrophic response to exercise. In contrast, cTnI-G203S mice showed no increase in these parameters, indicating an abnormal response to exercise. The lack of a physiological response to exercise may indicate an important novel mechanistic insight into the role of exercise in triggering adverse events in FHC.


Asunto(s)
Cardiomiopatía Hipertrófica Familiar/fisiopatología , Natación/fisiología , Animales , Cardiomiopatía Hipertrófica Familiar/diagnóstico por imagen , Cardiomiopatía Hipertrófica Familiar/patología , Modelos Animales de Enfermedad , Ecocardiografía , Masculino , Ratones , Ratones Transgénicos , Mutación , Troponina I/fisiología
19.
Clin Exp Pharmacol Physiol ; 33(1-2): 139-45, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16445713

RESUMEN

1. Familial hypertrophic cardiomyopathy (FHC) is a primary disorder of the myocardium characterized by remarkable diversity in clinical presentations, ranging from no symptoms to severe heart failure and sudden cardiac death. 2. Over the past 15 years, at least 11 genes have been identified, defects of which cause FHC. Most of these genes encode proteins that comprise the basic contractile unit of the heart (i.e. the sarcomere). 3. Genetic studies are now beginning to have a major impact on the diagnosis in FHC, as well as in guiding treatment and preventative strategies. Although much is known about which genes cause disease, relatively little is known about the molecular steps leading from the gene defect to the clinical phenotype and what factors modify the expression of the mutant genes. 4. Concurrent studies in cell culture and animal models of FHC are now beginning to shed light on the signalling pathways involved in FHC and the role of both environmental and genetic modifying factors. Calcium dysregulation appears to be important in the pathogenesis of FHC. 5. Understanding these basic molecular mechanisms will ultimately improve our knowledge of the basic biology of heart muscle function and will therefore provide new avenues for diagnosis and treatment not only for FHC, but also for a range of human cardiovascular diseases.


Asunto(s)
Calcio/metabolismo , Cardiomiopatía Hipertrófica Familiar/patología , Predisposición Genética a la Enfermedad/genética , Animales , Cardiomiopatía Hipertrófica Familiar/genética , Cardiomiopatía Hipertrófica Familiar/metabolismo , Modelos Animales de Enfermedad , Genotipo , Humanos , Ratones , Fenotipo
20.
J Mol Cell Cardiol ; 41(4): 623-32, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16950368

RESUMEN

Gene mutations in cardiac troponin I (cTnI) account for up to 5% of genotyped families with familial hypertrophic cardiomyopathy (FHC). Little is known about how cTnI mutations cause disease. Five lines of transgenic mice were generated which overexpress the human disease-causing cTnI gene mutation, Gly203Ser (designated cTnI-G203S), in a cardiac-specific manner. Mice were compared to transgenic mice that overexpress normal cTnI (cTnI-wt) and non-transgenic littermates (NTG). cTnI-G203S mice developed all the characteristic features of FHC by age 21 weeks. Left ventricular hypertrophy was observed on echocardiography (1.25+/-0.05 mm vs. 0.86+/-0.02 mm in cTnI-wt, P<0.01), associated with a significant 4-fold increase in RNA markers of hypertrophy, ANF and BNP. Myocyte hypertrophy, myofiber disarray and interstitial fibrosis were observed in cTnI-G203S mice. Expression of the cTnI-G203S mutation in neonatal cardiomyocytes resulted in a significant increase in myocyte volume, and reduced interactions with both troponins T and C. Ca2+ cycling was abnormal in adult cardiomyocytes extracted from cTnI-G203S mice, with a prolonged decay constant in Ca2+ transients and a reduced decay constant in response to caffeine treatment. Mice with the cTnI-G203S gene mutation develop all the phenotypic features of human FHC. The cTnI-G203S mutation disrupts interactions with partner proteins, and results in intracellular Ca2+ dysregulation early in life, suggesting a pathogenic role in development of FHC.


Asunto(s)
Cardiomiopatía Hipertrófica Familiar/genética , Cardiomiopatía Hipertrófica Familiar/metabolismo , Modelos Animales de Enfermedad , ARN Mensajero/metabolismo , Troponina I/genética , Animales , Animales Recién Nacidos , Señalización del Calcio , Cardiomiopatía Hipertrófica Familiar/inducido químicamente , Cardiomiopatía Hipertrófica Familiar/fisiopatología , Tamaño de la Célula , Electrocardiografía , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Biológicos , Mutación , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/ultraestructura , Fenotipo , Troponina I/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA