Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 5.999
Filtrar
1.
Zhen Ci Yan Jiu ; 49(6): 558-565, 2024 Jun 25.
Artículo en Inglés, Chino | MEDLINE | ID: mdl-38897799

RESUMEN

OBJECTIVES: To observe the effect of electroacupuncture (EA) at "Neiguan"(PC6) on cardiac function, cardiac morphology and transient receptor potential channel (TRPC) protein expressions in myocardial tissue of mice with myocardial hypertrophy, so as to explore its mechanisms underlying improvement of myocardial hypertrophy. METHODS: Forty-five male C57BL/6 mice were randomly divided into control, model and EA groups (15 mice/group). The myocardial hypertrophy model was established by subcutaneous injection of isoproterenol hydrochloride (15 mg·kg-1·d-1) for 14 days. The mice of the control group received subcutaneous injection of same amount of normal saline. The mice of the EA group received EA stimulation (frequency of 2 Hz, intensity of 1 mA) of bilateral PC6 for 20 min each time, once a day for 14 consecutive days. After the intervention, the body weight, tibia length and heart weight were measured. The left ventricular ejection fraction (EF), fractional shortening index (FS), left ventricular end-systolic volume (LVEV), left ventricular end-systolic internal diameter (LVID) and left ventricular posterior wall thickness (LVPW) were measured by using echocardiography for evaluating the cardiac function. The mean number and surface area of myocardial cells was detected by wheat germ agglutinin (WGA) staining, and changes of the cardiac morphology were observed under light microscopy after HE staining. The expression levels of TRPC1, TRPC3, TRPC4 and TRPC6 (TRPC1/3/4/6) in the myocardial tissue were detected by real-time quantitative PCR (qPCR) and Western blot, separately. RESULTS: Compared with the control group, the heart-body weight ratio(P<0.05) and heart-weight-to-tibia-length ratio (P<0.01), LVEV and LVID levels, the relative surface area, left ventricular area ratio, and the expression levels of cardiac TRPC1/3/4/6 were significantly increased (P<0.01, P<0.05), while the EF, FS, LVPW, number of cardiomyocytes, and the left ventricular posterior wall ratio were obviously decreased (P<0.01, P<0.05) in the model group. In comparison with the model group, the heart/body weight ratio, heart-weight-to-tibia-length ratio, LVEV and LVID levels, relative surface area, left ventricular area ratio, and the expression levels of cardiac TRPC1/3/4/6 were significantly decreased (P<0.01, P<0.05), while the EF, FS, LVPW, number of cardiomyocytes and left ventricular posterior wall ratio were significantly increased (P<0.01, P<0.05) in the EA group. H.E. staining showed disordered arrangement of cardiomyocytes and obvious myocardial interstitial inflammatory cell infiltration in the model group, and evident reduction of degree of cardiac fibrosis and interstitial edema in the EA group. CONCLUSIONS: EA of PC6 can improve the cardiac function and cardiac morphology in mice with myocardial hypertrophy, which may be related to its functions in down-regulating the expression of transient receptor potential channels.


Asunto(s)
Electroacupuntura , Ratones Endogámicos C57BL , Miocardio , Animales , Ratones , Masculino , Humanos , Miocardio/metabolismo , Canales de Potencial de Receptor Transitorio/metabolismo , Canales de Potencial de Receptor Transitorio/genética , Cardiomegalia/metabolismo , Cardiomegalia/terapia , Cardiomegalia/genética , Cardiomegalia/fisiopatología , Puntos de Acupuntura , Canales Catiónicos TRPC/metabolismo , Canales Catiónicos TRPC/genética
2.
Clin Sci (Lond) ; 138(10): 573-597, 2024 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-38718356

RESUMEN

The three striatins (STRN, STRN3, STRN4) form the core of STRiatin-Interacting Phosphatase and Kinase (STRIPAK) complexes. These place protein phosphatase 2A (PP2A) in proximity to protein kinases thereby restraining kinase activity and regulating key cellular processes. Our aim was to establish if striatins play a significant role in cardiac remodelling associated with cardiac hypertrophy and heart failure. All striatins were expressed in control human hearts, with up-regulation of STRN and STRN3 in failing hearts. We used mice with global heterozygote gene deletion to assess the roles of STRN and STRN3 in cardiac remodelling induced by angiotensin II (AngII; 7 days). Using echocardiography, we detected no differences in baseline cardiac function or dimensions in STRN+/- or STRN3+/- male mice (8 weeks) compared with wild-type littermates. Heterozygous gene deletion did not affect cardiac function in mice treated with AngII, but the increase in left ventricle mass induced by AngII was inhibited in STRN+/- (but not STRN3+/-) mice. Histological staining indicated that cardiomyocyte hypertrophy was inhibited. To assess the role of STRN in cardiomyocytes, we converted the STRN knockout line for inducible cardiomyocyte-specific gene deletion. There was no effect of cardiomyocyte STRN knockout on cardiac function or dimensions, but the increase in left ventricle mass induced by AngII was inhibited. This resulted from inhibition of cardiomyocyte hypertrophy and cardiac fibrosis. The data indicate that cardiomyocyte striatin is required for early remodelling of the heart by AngII and identify the striatin-based STRIPAK system as a signalling paradigm in the development of pathological cardiac hypertrophy.


Asunto(s)
Angiotensina II , Cardiomegalia , Ratones Noqueados , Miocitos Cardíacos , Animales , Angiotensina II/farmacología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Cardiomegalia/genética , Cardiomegalia/patología , Cardiomegalia/metabolismo , Cardiomegalia/fisiopatología , Masculino , Humanos , Proteínas Musculares/metabolismo , Proteínas Musculares/genética , Remodelación Ventricular , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas de Unión a Calmodulina , Proteínas del Tejido Nervioso
3.
Med Sci Monit ; 30: e944114, 2024 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-38776271

RESUMEN

BACKGROUND Long-term right ventricular (RV) pacing has been linked to left atrial enlargement (LAE). The incidence and risk factors associated with significant LAE after RV pacing remain unknown. This retrospective study included 461 patients requiring RV pacing at 2 centers between 2012 and 2020 and aimed to evaluate the incidence, risk factors, outcomes, and complications of LAE. MATERIAL AND METHODS A total of 461 patients with normal-sized pre-implant left atrial dimension and dual-chamber pacing pacemaker implantation for complete atrioventricular block were enrolled. Patients were grouped based on a ≥20% increase from their baseline left atrial dimension by echocardiography, indicating significant LAE, and initial characteristics, echocardiographic data, and outcomes were compared. RESULTS During a mean 7.0±4.9 years follow-up period, 96 patients (20.8%) developed significant LAE, whereas 365 patients did not. In multivariate logistic regression analysis, smaller pre-implant left atrial dimension (OR, 0.776; 95% CI, 0.728-0.828; P<0.001), lower post-implant left ventricular ejection fraction (OR, 0.976; 95% CI, 0.957-0.995; P=0.014), post-implant development of moderate to severe mitral regurgitation (OR, 2.357; 95% CI, 1.172-4.740; P=0.016), and RV pacing duration ≥3.3 years (OR, 1.576; 95% CI, 1.039-2.646; P=0.045) were independent predictors of significant LAE after RV-dependent pacing. There was a significant difference in the incident stroke events between patients without and with significant LAE (9.9% vs 17.7%; log-rank P=0.047). CONCLUSIONS Long-term RV pacing was linked to significant LAE in 20.8% of patients with complete atrioventricular block, with those affected experiencing a higher stroke rate during follow-up.


Asunto(s)
Estimulación Cardíaca Artificial , Ecocardiografía , Atrios Cardíacos , Ventrículos Cardíacos , Humanos , Femenino , Masculino , Estudios Retrospectivos , Factores de Riesgo , Incidencia , Anciano , Atrios Cardíacos/fisiopatología , Estimulación Cardíaca Artificial/métodos , Estimulación Cardíaca Artificial/efectos adversos , Persona de Mediana Edad , Ventrículos Cardíacos/fisiopatología , Ecocardiografía/métodos , Bloqueo Atrioventricular/terapia , Bloqueo Atrioventricular/fisiopatología , Cardiomegalia/fisiopatología , Marcapaso Artificial , Resultado del Tratamiento , Anciano de 80 o más Años
4.
Expert Opin Investig Drugs ; 33(6): 543-547, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38702878

RESUMEN

INTRODUCTION: There are currently limited effective treatments available to improve lusitropy in patients suffering from heart failure with preserved ejection fraction. The role of PDE9A in diastolic dysfunction has been well-studied over recent years, with a special focus on its association with myocardial hypertrophy. Recent insights into PDE9A inhibition have brought to light the potential for reversal of cardiac remodeling, with multiple studies showing promising results in preclinical data. AREAS COVERED: This expert opinion provides an overview of the role of PDE9A in diastolic heart dysfunction along with the efficacy of PDE9A inhibitors in laboratory models of heart failure with preserved ejection fraction. EXPERT OPINION: The available data on PDE9A inhibition in preclinical studies suggest that there is potential for reversal of diastolic dysfunction and myocardial hypertrophy, however, conflicting data suggests that further studies are required before progressing to clinical trials.


Asunto(s)
3',5'-AMP Cíclico Fosfodiesterasas , Insuficiencia Cardíaca , Inhibidores de Fosfodiesterasa , Humanos , Animales , Insuficiencia Cardíaca/tratamiento farmacológico , Insuficiencia Cardíaca/fisiopatología , Inhibidores de Fosfodiesterasa/farmacología , 3',5'-AMP Cíclico Fosfodiesterasas/antagonistas & inhibidores , Cardiomegalia/tratamiento farmacológico , Cardiomegalia/fisiopatología , Remodelación Ventricular/efectos de los fármacos , Volumen Sistólico/efectos de los fármacos
5.
Physiology (Bethesda) ; 39(5): 0, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-38713091

RESUMEN

Insulin-like growth factor-1 (IGF-1) signaling has multiple physiological roles in cellular growth, metabolism, and aging. Myocardial hypertrophy, cell death, senescence, fibrosis, and electrical remodeling are hallmarks of various heart diseases and contribute to the progression of heart failure. This review highlights the critical role of IGF-1 and its cognate receptor in cardiac hypertrophy, aging, and remodeling.


Asunto(s)
Factor I del Crecimiento Similar a la Insulina , Transducción de Señal , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Animales , Transducción de Señal/fisiología , Receptor IGF Tipo 1/metabolismo , Miocardio/metabolismo , Envejecimiento/metabolismo , Envejecimiento/fisiología , Corazón/fisiología , Cardiomegalia/metabolismo , Cardiomegalia/fisiopatología
6.
Cells ; 13(10)2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38786079

RESUMEN

Heart failure is the common concluding pathway for a majority of cardiovascular diseases and is associated with cardiac dysfunction. Since heart failure is invariably preceded by adaptive or maladaptive cardiac hypertrophy, several biochemical mechanisms have been proposed to explain the development of cardiac hypertrophy and progression to heart failure. One of these includes the activation of different neuroendocrine systems for elevating the circulating levels of different vasoactive hormones such as catecholamines, angiotensin II, vasopressin, serotonin and endothelins. All these hormones are released in the circulation and stimulate different signal transduction systems by acting on their respective receptors on the cell membrane to promote protein synthesis in cardiomyocytes and induce cardiac hypertrophy. The elevated levels of these vasoactive hormones induce hemodynamic overload, increase ventricular wall tension, increase protein synthesis and the occurrence of cardiac remodeling. In addition, there occurs an increase in proinflammatory cytokines and collagen synthesis for the induction of myocardial fibrosis and the transition of adaptive to maladaptive hypertrophy. The prolonged exposure of the hypertrophied heart to these vasoactive hormones has been reported to result in the oxidation of catecholamines and serotonin via monoamine oxidase as well as the activation of NADPH oxidase via angiotensin II and endothelins to promote oxidative stress. The development of oxidative stress produces subcellular defects, Ca2+-handling abnormalities, mitochondrial Ca2+-overload and cardiac dysfunction by activating different proteases and depressing cardiac gene expression, in addition to destabilizing the extracellular matrix upon activating some metalloproteinases. These observations support the view that elevated levels of various vasoactive hormones, by producing hemodynamic overload and activating their respective receptor-mediated signal transduction mechanisms, induce cardiac hypertrophy. Furthermore, the occurrence of oxidative stress due to the prolonged exposure of the hypertrophied heart to these hormones plays a critical role in the progression of heart failure.


Asunto(s)
Cardiomegalia , Insuficiencia Cardíaca , Transducción de Señal , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/fisiopatología , Insuficiencia Cardíaca/patología , Humanos , Cardiomegalia/metabolismo , Cardiomegalia/fisiopatología , Cardiomegalia/patología , Animales , Angiotensina II/metabolismo , Estrés Oxidativo
7.
Pflugers Arch ; 476(7): 1109-1123, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38625371

RESUMEN

The myocardium is a highly oxidative tissue in which mitochondria are essential to supply the energy required to maintain pump function. When pathological hypertrophy develops, energy consumption augments and jeopardizes mitochondrial capacity. We explored the cardiac consequences of chronic swimming training, focusing on the mitochondrial network, in spontaneously hypertensive rats (SHR). Male adult SHR were randomized to sedentary or trained (T: 8-week swimming protocol). Blood pressure and echocardiograms were recorded, and hearts were removed at the end of the training period to perform molecular, imaging, or isolated mitochondria studies. Swimming improved cardiac midventricular shortening and decreased the pathological hypertrophic marker atrial natriuretic peptide. Oxidative stress was reduced, and even more interesting, mitochondrial spatial distribution, dynamics, function, and ATP were significantly improved in the myocardium of T rats. In the signaling pathway triggered by training, we detected an increase in the phosphorylation level of both AKT and glycogen synthase kinase-3 ß, key downstream targets of insulin-like growth factor 1 signaling that are crucially involved in mitochondria biogenesis and integrity. Aerobic exercise training emerges as an effective approach to improve pathological cardiac hypertrophy and bioenergetics in hypertension-induced cardiac hypertrophy.


Asunto(s)
Mitocondrias Cardíacas , Condicionamiento Físico Animal , Ratas Endogámicas SHR , Animales , Masculino , Ratas , Mitocondrias Cardíacas/metabolismo , Condicionamiento Físico Animal/métodos , Condicionamiento Físico Animal/fisiología , Cardiomegalia/metabolismo , Cardiomegalia/fisiopatología , Hipertensión/metabolismo , Hipertensión/fisiopatología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Natación/fisiología , Estrés Oxidativo , Transducción de Señal/fisiología , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Presión Sanguínea/fisiología , Factor Natriurético Atrial/metabolismo
8.
Am J Physiol Cell Physiol ; 326(5): C1437-C1450, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38525542

RESUMEN

Plasma apelin levels are reduced in aging and muscle wasting conditions. We aimed to investigate the significance of apelin signaling in cardiac and skeletal muscle responses to physiological stress. Apelin knockout (KO) and wild-type (WT) mice were subjected to high-intensity interval training (HIIT) by treadmill running. The effects of apelin on energy metabolism were studied in primary mouse skeletal muscle myotubes and cardiomyocytes. Apelin increased mitochondrial ATP production and mitochondrial coupling efficiency in myotubes and promoted the expression of mitochondrial genes both in primary myotubes and cardiomyocytes. HIIT induced mild concentric cardiac hypertrophy in WT mice, whereas eccentric growth was observed in the left ventricles of apelin KO mice. HIIT did not affect myofiber size in skeletal muscles of WT mice but decreased the myofiber size in apelin KO mice. The decrease in myofiber size resulted from a fiber type switch toward smaller slow-twitch type I fibers. The increased proportion of slow-twitch type I fibers in apelin KO mice was associated with upregulation of myosin heavy chain slow isoform expression, accompanied with upregulated expression of genes related to fatty acid transport and downregulated expression of genes related to glucose metabolism. Mechanistically, skeletal muscles of apelin KO mice showed defective induction of insulin-like growth factor-1 signaling in response to HIIT. In conclusion, apelin is required for proper skeletal and cardiac muscle adaptation to high-intensity exercise. Promoting apelinergic signaling may have benefits in aging- or disease-related muscle wasting conditions.NEW & NOTEWORTHY Apelin levels decline with age. This study demonstrates that in trained mice, apelin deficiency results in a switch from fast type II myofibers to slow oxidative type I myofibers. This is associated with a concomitant change in gene expression profile toward fatty acid utilization, indicating an aged-muscle phenotype in exercised apelin-deficient mice. These data are of importance in the design of exercise programs for aging individuals and could offer therapeutic target to maintain muscle mass.


Asunto(s)
Adaptación Fisiológica , Apelina , Ratones Noqueados , Músculo Esquelético , Condicionamiento Físico Animal , Animales , Apelina/metabolismo , Apelina/genética , Ratones , Condicionamiento Físico Animal/fisiología , Músculo Esquelético/metabolismo , Entrenamiento de Intervalos de Alta Intensidad/métodos , Masculino , Miocitos Cardíacos/metabolismo , Metabolismo Energético , Ratones Endogámicos C57BL , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares de Contracción Lenta/metabolismo , Cardiomegalia/metabolismo , Cardiomegalia/genética , Cardiomegalia/fisiopatología , Cardiomegalia/patología
9.
J Cardiovasc Pharmacol ; 83(5): 457-465, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38498600

RESUMEN

ABSTRACT: Angiotensin (Ang)-(1-7) is a cardioprotective peptide of the renin-angiotensin system. Prepuberty has been considered as a later susceptible window of development, and stressful factors in this life phase can induce chronic diseases in adulthood. We aimed to investigate whether the treatment with Ang-(1-7) during the prepuberty could attenuate the development of hypertension and cardiac injury in adult spontaneously hypertensive rats (SHRs). SHRs were treated with Ang-(1-7) (24 µg/kg/h) from age 4 to 7 weeks. Systolic blood pressure was measured by tail-cuff plethysmography up to 17th week. Thereafter, echocardiography was performed, and the rats were euthanized for the collection of tissues and blood. Ang-(1-7) did not change the systolic blood pressure but reduced the septal and posterior wall thickness, and cardiomyocyte hypertrophy and fibrosis in SHR. In addition, Ang-(1-7) reduced the gene expression of atrial natriuretic peptide and brain natriuretic peptide, increased the metalloproteinase 9 expression, and reduced the extracellular signal-regulated kinases 1/2 phosphorylation. Ang-(1-7) also prevented the reduction of Mas receptor but did not change the protein expression of angiotensin-converting enzyme, angiotensin-converting enzyme 2, AT1, and AT2. The treatment with Ang-(1-7) decreased the malondialdehyde (MDA) levels and increased superoxide dismutase-1 and catalase activities and protein expression of catalase. Our findings demonstrate that the treatment of SHR with Ang-(1-7) for 3 weeks early in life promotes beneficial effects in the heart later in life, even without altering blood pressure, through mechanisms involving the reduction of oxidative stress and ERK1/2 phosphorylation. In addition, this study supports the prepuberty as an important programming window.


Asunto(s)
Angiotensina I , Presión Sanguínea , Cardiomegalia , Hipertensión , Estrés Oxidativo , Fragmentos de Péptidos , Ratas Endogámicas SHR , Animales , Angiotensina I/farmacología , Fragmentos de Péptidos/farmacología , Masculino , Hipertensión/fisiopatología , Hipertensión/tratamiento farmacológico , Hipertensión/prevención & control , Cardiomegalia/prevención & control , Cardiomegalia/fisiopatología , Cardiomegalia/metabolismo , Cardiomegalia/tratamiento farmacológico , Cardiomegalia/patología , Estrés Oxidativo/efectos de los fármacos , Presión Sanguínea/efectos de los fármacos , Fibrosis , Modelos Animales de Enfermedad , Ratas , Fosforilación , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Miocitos Cardíacos/metabolismo , Péptido Natriurético Encefálico/metabolismo , Factores de Edad , Metaloproteinasa 9 de la Matriz/metabolismo , Factor Natriurético Atrial/metabolismo , Antihipertensivos/farmacología , Remodelación Ventricular/efectos de los fármacos
10.
Thyroid ; 34(6): 796-805, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38526409

RESUMEN

Background: Thyroid hormones regulate cardiac functions mainly through direct actions in the heart and by binding to the thyroid hormone receptor (TR) isoforms α1 and ß. While the role of the most abundantly expressed isoform, TRα1, is widely studied and well characterized, the role of TRß in regulating heart functions is still poorly understood, primarily due to the accompanying elevation of circulating thyroid hormone in TRß knockout mice (TRß-KO). However, their hyperthyroidism is ameliorated at thermoneutrality, which allows studying the role of TRß without this confounding factor. Methods: Here, we noninvasively monitored heart rate in TRß-KO mice over several days using radiotelemetry at different housing temperatures (22°C and 30°C) and upon 3,3',5-triiodothyronine (T3) administration in comparison to wild-type animals. Results: TRß-KO mice displayed normal average heart rate at both 22°C and 30°C with only minor changes in heart rate frequency distribution, which was confirmed by independent electrocardiogram recordings in freely-moving conscious mice. Parasympathetic nerve activity was, however, impaired in TRß-KO mice at 22°C, and only partly rescued at 30°C. As expected, oral treatment with pharmacological doses of T3 at 30°C led to tachycardia in wild-types, accompanied by broader heart rate frequency distribution and increased heart weight. The TRß-KO mice, in contrast, showed blunted tachycardia, as well as resistance to changes in heart rate frequency distribution and heart weight. At the molecular level, these observations were paralleled by a blunted cardiac mRNA induction of several important genes, including the pacemaker channels Hcn2 and Hcn4, as well as Kcna7. Conclusions: The phenotyping of TRß-KO mice conducted at thermoneutrality allows novel insights on the role of TRß in cardiac functions in the absence of the usual confounding hyperthyroidism. Even though TRß is expressed at lower levels than TRα1 in the heart, our findings demonstrate an important role for this isoform in the cardiac response to thyroid hormones.


Asunto(s)
Cardiomegalia , Frecuencia Cardíaca , Ratones Noqueados , Taquicardia , Receptores beta de Hormona Tiroidea , Triyodotironina , Animales , Receptores beta de Hormona Tiroidea/genética , Receptores beta de Hormona Tiroidea/metabolismo , Taquicardia/fisiopatología , Taquicardia/metabolismo , Ratones , Cardiomegalia/metabolismo , Cardiomegalia/fisiopatología , Cardiomegalia/genética , Triyodotironina/sangre , Masculino , Hormonas Tiroideas/metabolismo , Sistema Nervioso Parasimpático/fisiopatología , Temperatura , Electrocardiografía
11.
Am J Physiol Heart Circ Physiol ; 326(5): H1193-H1203, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38334973

RESUMEN

Pressure overload-induced hypertrophy compromises cardiac stretch-induced compliance (SIC) after acute volume overload (AVO). We hypothesized that SIC could be enhanced by physiological hypertrophy induced by pregnancy's chronic volume overload. This study evaluated SIC-cardiac adaptation in pregnant women with or without cardiovascular risk (CVR) factors. Thirty-seven women (1st trimester, 1stT) and a separate group of 31 (3rd trimester, 3rdT) women [healthy or with CVR factors (obesity and/or hypertension and/or with gestational diabetes)] underwent echocardiography determination of left ventricular end-diastolic volume (LVEDV) and E/e' before (T0), immediately after (T1), and 15 min after (T2; SIC) AVO induced by passive leg elevation. Blood samples for NT-proBNP quantification were collected before and after the AVO. Acute leg elevation significantly increased inferior vena cava diameter and stroke volume from T0 to T1 in both 1stT and 3rdT, confirming AVO. LVEDV and E/e' also increased immediately after AVO (T1) in both 1stT and 3rdT. SIC adaptation (T2, 15 min after AVO) significantly decreased E/e' in both trimesters, with additional expansion of LVEDV only in the 1stT. NT-pro-BNP increased slightly after AVO but only in the 1stT. CVR factors, but not parity or age, significantly impacted SIC cardiac adaptation. A distinct functional response to SIC was observed between 1stT and 3rdT, which was influenced by CVR factors. The LV of 3rdT pregnant women was hypertrophied, showing a structural limitation to dilate with AVO, whereas the lower LV filling pressure values suggest increased diastolic compliance.NEW & NOTEWORTHY The sudden increase of volume overload triggers an acute myocardial stretch characterized by an immediate rise in contractility by the Frank-Starling mechanism, followed by a progressive increase known as the slow force response. The present study is the first to characterize echocardiographically the stretch-induced compliance (SIC) mechanism in the context of physiological hypertrophy induced by pregnancy. A distinct functional adaptation to SIC was observed between first and third trimesters, which was influenced by cardiovascular risk factors.


Asunto(s)
Adaptación Fisiológica , Factores de Riesgo de Enfermedad Cardiaca , Humanos , Femenino , Embarazo , Adulto , Función Ventricular Izquierda , Cardiomegalia/fisiopatología , Cardiomegalia/diagnóstico por imagen , Cardiomegalia/etiología , Péptido Natriurético Encefálico/sangre , Fragmentos de Péptidos/sangre , Complicaciones Cardiovasculares del Embarazo/fisiopatología , Complicaciones Cardiovasculares del Embarazo/diagnóstico por imagen , Complicaciones Cardiovasculares del Embarazo/sangre , Volumen Sistólico , Tercer Trimestre del Embarazo , Diabetes Gestacional/fisiopatología , Adaptabilidad , Primer Trimestre del Embarazo , Obesidad/fisiopatología , Obesidad/complicaciones , Factores de Riesgo
12.
Am J Perinatol ; 41(S 01): e3413-e3419, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38266754

RESUMEN

OBJECTIVE: We aimed to determine whether exposure to severe maternal preeclampsia (PE) in very low birth weight (VLBW) infants is associated with hypertrophic cardiac changes and altered hemodynamics. STUDY DESIGN: Case-control study of VLBW infants born at Los Angeles General Medical Center from May 2015 to August 2023, who had an echocardiogram within the first 7 days of life. Cases were infants exposed to maternal PE and controls were infants not exposed to maternal PE matched by birth weight (BW) 1:1. Laboratory, placental pathology results, hemodynamic data and clinical outcomes were collected and compared between cases and control infants. RESULTS: A total of 43 cases matched by BW with control infants were studied. There were no significant anatomical cardiac changes by echocardiography between cases and control infants. Cases had significantly higher blood pressure within the first 72 hours of life and lower ejection fraction (EF), fractional shortening, and peak systolic flow velocity through their patent ductus arteriosus (PDA) within the first week of life. Cases were more likely to be smaller despite being born at a later gestational age (GA), as well as small for GA with placental weight less than 10th percentile compared to control infants. CONCLUSION: Our findings indicate that infants born to mothers with PE have higher systemic vascular resistance as evidenced by elevated blood pressure, and lower EF and shortening fraction and higher pulmonary vascular resistance as evidenced by lower peak flow velocity through the PDA. We did not observe hypertrophic cardiac changes in exposed infants. These findings should be considered in clinical decision-making during management of these infants. KEY POINTS: · VLBW infants exposed to severe PE have higher rate of Small for gestational age and smaller placentas.. · VLBW infants exposed to severe PE have higher systemic vascular resistance during transitional period and lower EF and fractional shortening.. · VLBW infants exposed to severe PE have higher pulmonary vascular resistance..


Asunto(s)
Ecocardiografía , Recién Nacido de muy Bajo Peso , Preeclampsia , Humanos , Femenino , Embarazo , Estudios de Casos y Controles , Preeclampsia/fisiopatología , Recién Nacido , Hemodinámica , Adulto , Masculino , Edad Gestacional , Conducto Arterioso Permeable/fisiopatología , Conducto Arterioso Permeable/diagnóstico por imagen , Presión Sanguínea/fisiología , Cardiomegalia/diagnóstico por imagen , Cardiomegalia/fisiopatología , Volumen Sistólico/fisiología
14.
Oxid Med Cell Longev ; 2022: 5044046, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35222798

RESUMEN

Cardiac lymphatic vessel growth (lymphangiogenesis) and integrity play an essential role in maintaining tissue fluid balance. Inhibition of lymphatic lymphangiogenesis is involved in cardiac edema and cardiac remodeling after ischemic injury or pressure overload. However, whether lymphatic vessel integrity is disrupted during angiotensin II- (Ang II-) induced cardiac remodeling remains to be investigated. In this study, cardiac remodeling models were established by Ang II (1000 ng/kg/min) in VEGFR-3 knockdown (Lyve-1Cre VEGFR-3f/-) and wild-type (VEGFR-3f/f) littermates. Our results indicated that Ang II infusion not only induced cardiac lymphangiogenesis and upregulation of VEGF-C and VEGFR-3 expression in the time-dependent manner but also enhanced proteasome activity, MKP5 and VE-cadherin degradation, p38 MAPK activation, and lymphatic vessel hyperpermeability. Moreover, VEGFR-3 knockdown significantly inhibited cardiac lymphangiogenesis in mice, resulting in exacerbation of tissue edema, hypertrophy, fibrosis superoxide production, inflammation, and heart failure (HF). Conversely, administration of epoxomicin (a selective proteasome inhibitor) markedly mitigated Ang II-induced cardiac edema, remodeling, and dysfunction; upregulated MKP5 and VE-cadherin expression; inactivated p38 MAPK; and reduced lymphatic vessel hyperpermeability in WT mice, indicating that inhibition of proteasome activity is required to maintain lymphatic endothelial cell (LEC) integrity. Our results show that both cardiac lymphangiogenesis and lymphatic barrier hyperpermeability are implicated in Ang II-induced adaptive hypertrophic remodeling and dysfunction. Proteasome-mediated hyperpermeability of LEC junctions plays a predominant role in the development of cardiac remodeling. Selective stimulation of lymphangiogenesis or inhibition of proteasome activity may be a potential therapeutic option for treating hypertension-induced cardiac remodeling.


Asunto(s)
Angiotensina II/metabolismo , Cardiomegalia/metabolismo , Edema Cardíaco/metabolismo , Vasos Linfáticos/metabolismo , Angiotensina II/administración & dosificación , Animales , Cardiomegalia/tratamiento farmacológico , Cardiomegalia/patología , Cardiomegalia/fisiopatología , Edema Cardíaco/tratamiento farmacológico , Edema Cardíaco/patología , Edema Cardíaco/fisiopatología , Células Endoteliales/metabolismo , Linfangiogénesis/efectos de los fármacos , Ratones , Ratones Noqueados , Miocardio/metabolismo , Miocardio/patología , Permeabilidad/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma/farmacología , Inhibidores de Proteasoma/uso terapéutico , Receptor 3 de Factores de Crecimiento Endotelial Vascular/deficiencia , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo
15.
Oxid Med Cell Longev ; 2022: 6603296, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35096272

RESUMEN

CD147, also known as EMMPRIN or basigin, is a transmembrane glycoprotein receptor that activates matrix metalloproteinases and promotes inflammation. CD147 function is regulated by posttranslational modifications of which glycosylation has attracted the most attention. In this study, we demonstrated that glycosylated CD147 was the dominant form in heart tissue, and its levels were markedly elevated in response to transverse aortic constriction (TAC). Adeno-associated virus 9-mediated, cardiac-specific overexpression of wild-type CD147 in mice significantly promoted pressure overload-induced pathological cardiac remodeling accompanied by augmented oxidative stress and ferroptosis. By contrast, mutations of CD147 glycosylation sites notably weakened these detrimental effects of CD147. Mechanistically, CD147 exacerbated TAC-induced pathological cardiac remodeling via direct binding with the adaptor molecule TRAF2 and subsequent activation of TAK1 signalling, which was dependent on glycosylation of CD147. Collectively, our findings provide the first evidence that CD147 promoted pathological cardiac remodeling and dysfunction in a glycosylation-dependent manner through binding the adaptor protein TRAF2 and activating the downstream TRAF2-TAK1 signalling pathway. Thus, glycosylation of CD147 may be a potent interventional target for heart failure treatment.


Asunto(s)
Basigina/efectos adversos , Cardiomegalia/fisiopatología , Animales , Glicosilación , Humanos , Masculino , Ratones
16.
Clin Sci (Lond) ; 136(2): 181-196, 2022 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-35048952

RESUMEN

Nicotinamide riboside kinase-2 (NRK-2) has recently emerged as a critical regulator of cardiac remodeling however, underlying molecular mechanisms is largely unknown. To explore the same, NRK2 knockout (KO) and littermate control mice were subjected to trans-aortic constriction (TAC) or sham surgeries and cardiac function was assessed by serial M-mode echocardiography. A mild cardiac contractile dysfunction was observed in the KOs at the early adaptive phase of remodeling followed by a significant deterioration during the maladaptive cardiac remodeling phase. Consistently, NRK2 KO hearts displayed increased cardiac hypertrophy and heart failure (HF) reflected by morphometric parameters as well as increased fetal genes, atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP) expressions. Histological assessment revealed an extensive left ventricular (LV) chamber dilatation accompanied by elevated cardiomyopathy (CM) and fibrosis in the KO hearts post-TAC. In a gain-of-function model, NRK-2 overexpressing in AC16 cardiomyocytes displayed significantly attenuated fetal genes ANP and BNP expression. Consistently, NRK-2 overexpression attenuated angiotensin II (Ang II)-induced cardiomyocyte death. Mechanistically, we identified NRK-2 as a regulator of c-jun N-terminal kinase (JNK) MAP kinase and mitochondrial function where NRK-2 overexpression in human cardiomyocytes markedly suppressed the Ang II-induced JNK activation and mitochondrial depolarization. Thus, our results demonstrate that NRK-2 plays protective roles in pressure overload (PO)-induced dilatative cardiac remodeling and, genetic ablation exacerbates dilated cardiomyopathy (DCM), interstitial collagen deposition, and cardiac dysfunction post-TAC due, in part, to increased JNK activation and mitochondrial dysfunction.


Asunto(s)
Cardiomiopatía Dilatada/fisiopatología , Sistema de Señalización de MAP Quinasas/fisiología , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Animales , Aorta , Cardiomegalia/fisiopatología , Línea Celular , Modelos Animales de Enfermedad , Insuficiencia Cardíaca/fisiopatología , Humanos , Masculino , Ratones , Ratones Noqueados , Contracción Miocárdica/fisiología , Miocitos Cardíacos/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética
17.
Cell Tissue Res ; 387(1): 111-121, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34669021

RESUMEN

Cardiac hypertrophy is considered as a common pathophysiological process in various cardiovascular diseases. CUG triplet repeat-binding protein 1 (CELF1) is an RNA-binding protein that has been shown to be an important post-transcription regulator and involved in several types of cancer, whereas its role in cardiac remodeling remains unclear. Herein, we found that the expression of CELF1 was significantly increased in pressure overload-induced hypertrophic hearts and angiotensin II (Ang II)-induced neonatal cardiomyocytes. Based on transverse aortic constriction-induced cardiac hypertrophy model, CELF1 deficiency markedly ameliorated cardiac hypertrophy, cardiac fibrosis, oxidative stress, and apoptosis. Accordingly, CELF1 deficiency alleviated the production of reactive oxygen species (ROS) and apoptosis of neonatal cardiomyocytes via inhibition of Raf1, TAK1, ERK1/2, and p38 phosphorylation. Mechanistically, depletion or overexpression of CELF1 negatively regulated the protein expression of phosphatidylethanolamine-binding protein 1 (PEBP1), while the mRNA expression of PEBP1 remained unchanged. RNA immunoprecipitation revealed that CELF1 directly interacted with PEBP1 mRNA. Biotin pull-down analysis and dual-luciferase assay showed that CELF1 directly bound to the fragment 1 within 3'UTR of PEBP1. Moreover, knockdown of PEBP1 partially enhanced the production of ROS and apoptosis of neonatal cardiomyocytes inhibited by CELF1 deficiency. In conclusion, CELF1 binds to the 3'UTR of PEBP1 and acts as an endogenous activator of MAPK signaling pathway. Inhibition of CELF1 attenuates pathological cardiac hypertrophy, oxidative stress, and apoptosis, thus could be a potential therapeutic strategy of pathological cardiac hypertrophy.


Asunto(s)
Proteínas CELF1/metabolismo , Cardiomegalia/genética , Ecocardiografía/métodos , Miocitos Cardíacos/metabolismo , Proteínas de Unión a Fosfatidiletanolamina/metabolismo , Proteínas de Unión al ARN/metabolismo , Animales , Cardiomegalia/fisiopatología , Humanos , Ratones , Transducción de Señal
18.
PLoS One ; 16(12): e0261388, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34914791

RESUMEN

Cardiac hypertrophy is a complex process induced by the activation of multiple signaling pathways. We previously reported that anacardic acid (AA), a histone acetyltransferase (HAT) inhibitor, attenuates phenylephrine (PE)-induced cardiac hypertrophy by downregulating histone H3 acetylation at lysine 9 (H3K9ac). Unfortunately, the related upstream signaling events remained unknown. The mitogen-activated protein kinase (MAPK) pathway is an important regulator of cardiac hypertrophy. In this study, we explored the role of JNK/MAPK signaling pathway in cardiac hypertrophy induced by PE. The mice cardiomyocyte hypertrophy model was successfully established by treating cells with PE in vitro. This study showed that p-JNK directly interacts with HATs (P300 and P300/CBP-associated factor, PCAF) and alters H3K9ac. In addition, both the JNK inhibitor SP600125 and the HAT inhibitor AA attenuated p-JNK overexpression and H3K9ac hyperacetylation by inhibiting P300 and PCAF during PE-induced cardiomyocyte hypertrophy. Moreover, we demonstrated that both SP600125 and AA attenuate the overexpression of cardiac hypertrophy-related genes (MEF2A, ANP, BNP, and ß-MHC), preventing cardiomyocyte hypertrophy and dysfunction. These results revealed a novel mechanism through which AA might protect mice from PE-induced cardiomyocyte hypertrophy. In particular, AA inhibits the effects of JNK signaling on HATs-mediated histone acetylation, and could therefore be used to prevent and treat pathological cardiac hypertrophy.


Asunto(s)
Ácidos Anacárdicos/farmacología , Cardiomegalia/fisiopatología , Sistema de Señalización de MAP Quinasas/fisiología , Acetilación , Ácidos Anacárdicos/metabolismo , Animales , Antracenos/farmacología , Cardiomegalia/metabolismo , China , Modelos Animales de Enfermedad , Femenino , Histona Acetiltransferasas/antagonistas & inhibidores , Histona Acetiltransferasas/metabolismo , Histonas/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Fenilefrina/efectos adversos , Fenilefrina/farmacología , Cultivo Primario de Células , Transducción de Señal/efectos de los fármacos , Factores de Transcripción p300-CBP
19.
Life Sci ; 287: 120106, 2021 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-34756930

RESUMEN

Cerebrovascular diseases such as ischemic stroke, brain hemorrhage, and subarachnoid hemorrhage provoke cardiac complications such as heart failure, neurogenic stress-related cardiomyopathy and Takotsubo cardiomyopathy. With regards to the pathophysiology of stroke-induced heart injury, several mechanisms have been postulated to contribute to this complex interaction between brain and heart, including damage from gut dysbiosis, immune and systematic inflammatory responses, microvesicle- and microRNA-mediated vascular injury and damage from a surge of catecholamines. All these cerebrovascular diseases may trigger pronounced catecholamine surges through diverse ways, including stimulation of hypothalamic-pituitary adrenal axis, dysregulation of autonomic system, and secretion of adrenocorticotropic hormone. Primary catecholamines involved in this pathophysiological response include norepinephrine (NE) and epinephrine. Both are important neurotransmitters that connect the nervous system with the heart, leading to cardiac damage via myocardial ischemia, calcium (Ca2+) overload, oxidative stress, and mitochondrial dysfunction. In this review, we will aim to summarize the molecular mechanisms behind catecholamine-induced cardiotoxicity including Ca2+ overload, oxidative stress, apoptosis, cardiac hypertrophy, interstitial fibrosis, and inflammation. In addition, we will focus on how synchronization among these pathways evokes cardiotoxicity.


Asunto(s)
Cardiomegalia/metabolismo , Cardiomegalia/fisiopatología , Catecolaminas/metabolismo , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/fisiopatología , Animales , Cardiomegalia/etiología , Cardiotoxicidad/etiología , Cardiotoxicidad/metabolismo , Cardiotoxicidad/fisiopatología , Catecolaminas/toxicidad , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/fisiopatología , Humanos , Estrés Oxidativo/fisiología , Accidente Cerebrovascular/complicaciones , Cardiomiopatía de Takotsubo/etiología , Cardiomiopatía de Takotsubo/metabolismo , Cardiomiopatía de Takotsubo/fisiopatología
20.
PLoS Biol ; 19(11): e3001447, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34758018

RESUMEN

During the first weeks of postnatal heart development, cardiomyocytes undergo a major adaptive metabolic shift from glycolytic energy production to fatty acid oxidation. This metabolic change is contemporaneous to the up-regulation and activation of the p38γ and p38δ stress-activated protein kinases in the heart. We demonstrate that p38γ/δ contribute to the early postnatal cardiac metabolic switch through inhibitory phosphorylation of glycogen synthase 1 (GYS1) and glycogen metabolism inactivation. Premature induction of p38γ/δ activation in cardiomyocytes of newborn mice results in an early GYS1 phosphorylation and inhibition of cardiac glycogen production, triggering an early metabolic shift that induces a deficit in cardiomyocyte fuel supply, leading to whole-body metabolic deregulation and maladaptive cardiac pathogenesis. Notably, the adverse effects of forced premature cardiac p38γ/δ activation in neonate mice are prevented by maternal diet supplementation of fatty acids during pregnancy and lactation. These results suggest that diet interventions have a potential for treating human cardiac genetic diseases that affect heart metabolism.


Asunto(s)
Glucógeno Sintasa/metabolismo , Proteína Quinasa 12 Activada por Mitógenos/metabolismo , Proteína Quinasa 13 Activada por Mitógenos/metabolismo , Miocardio/enzimología , Animales , Animales Recién Nacidos , Cardiomegalia/enzimología , Cardiomegalia/patología , Cardiomegalia/fisiopatología , Dieta Alta en Grasa , Activación Enzimática , Conducta Alimentaria , Femenino , Eliminación de Gen , Intolerancia a la Glucosa/enzimología , Glucógeno/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Resistencia a la Insulina , Metabolismo de los Lípidos , Sistema de Señalización de MAP Quinasas , Ratones Endogámicos C57BL , Miocitos Cardíacos/enzimología , Especificidad de Órganos , Fosforilación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA