Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Sheng Li Xue Bao ; 75(6): 779-787, 2023 Dec 25.
Artículo en Zh | MEDLINE | ID: mdl-38151343

RESUMEN

Atrial fibrillation (AF) is a cardiovascular epidemic that occurs primarily in the elderly with primary cardiovascular diseases, leading to severe consequences such as stroke and heart failure. The heart is an energy-consuming organ, which requires a high degree of metabolic flexibility to ensure a quick switch of metabolic substrates to meet its energy needs in response to physiological and pathological stimulation. Metabolism is closely related to the occurrence of AF, and AF patients manifest metabolic inflexibility, such as insulin resistance and the metabolic shift from aerobic metabolism to anaerobic glycolysis. Moreover, our research group and the others have shown that metabolic inflexibility is a crucial pathologic mechanism for AF. Energy metabolism is closely linked to the aging process and aging-related diseases, and impaired metabolic flexibility is considered as an essential driver of aging. Therefore, this review focuses on the alteration of metabolic flexibility in the elderly and reveals that impaired metabolic flexibility may be an important driver for the high prevalence of AF in the elderly, hoping to provide intervention strategies for the prevention and treatment of AF in the elderly.


Asunto(s)
Fibrilación Atrial , Insuficiencia Cardíaca , Accidente Cerebrovascular , Humanos , Anciano , Fibrilación Atrial/epidemiología , Anticoagulantes , Envejecimiento
2.
Pacing Clin Electrophysiol ; 44(11): 1817-1823, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-33973650

RESUMEN

BACKGROUND: It remains uncertain whether low-level electrical stimulation (LL-ES) of the ventricular ganglionated plexi (GP) improves heart function. This study investigated the anti-arrhythmic and anti-heart failure effects of LL-ES of the aortic root ventricular GP (ARVGP). METHODS: Thirty dogs were divided randomly into control, drug, and LL-ES groups after performing rapid right ventricular pacing to establish a heart failure (HF) model. The inducing rate of arrhythmia; levels of bioactive factors influencing HF, including angiotensin II type I receptor (AT-1R), transforming growth factor-beta (TGF-ß), matrix metalloproteinase (MMP), and phosphorylated extracellular signal-regulated kinase (p-ERK1/2); left ventricular stroke volume (LVSV), and left ventricular ejection fraction (LVEF)were measured after treatment with placebo, drugs, and LL-ES. RESULTS: The inducing rate of atrial arrhythmia decreased from 60% in the control group to 50% in the drug group and 10% in the LL-ES group (p = .033 vs. drug group) after 1 week of treatment. The ventricular effective refractory period was prolonged from 139 ± 8 ms in the drug group to 166 ± 13 ms in the LL-ES group (p = .001). Compared to the drug group, the expressions of AT-1R, TGF-ß, and MMP proteins were down-regulated in the LL-ES group, whereas that of p-ERK1/2 was significantly increased (all p = .001). Moreover, in the LL-ES group, LVSV increased markedly from 13.16 ± 0.22 to 16.86 ± 0.27 mL, relative to that in the drug group (p = .001), and LVEF increased significantly from 38.48% ± 0.53% to 48.94% ± 0.57% during the same time frame (p = .001). CONCLUSION: Short-term LL-ES of ARVGP had both anti-arrhythmic and anti-inflammatory effects and contributed to the treatment of tachycardia-induced HF and its associated arrhythmia.


Asunto(s)
Arritmias Cardíacas/prevención & control , Estimulación Eléctrica , Ganglios Autónomos/fisiología , Atrios Cardíacos/fisiopatología , Insuficiencia Cardíaca/prevención & control , Ventrículos Cardíacos/inervación , Ventrículos Cardíacos/fisiopatología , Animales , Arritmias Cardíacas/fisiopatología , Biomarcadores/sangre , Modelos Animales de Enfermedad , Perros , Insuficiencia Cardíaca/fisiopatología , Volumen Sistólico
3.
Zhongguo Zhong Yao Za Zhi ; 46(17): 4511-4521, 2021 Sep.
Artículo en Zh | MEDLINE | ID: mdl-34581057

RESUMEN

This study aims to explore the active components and molecular mechanism of Shenmai Injection in the treatment of atrial fibrillation(AF) based on the application of network pharmacology and molecular docking technology. The chemical components of single herbs of Shenmai Injection were collected from TCMSP and TCMID, with the standard chemical name and PubChem CID(referred to as CID) obtained from PubChem database. The active components were screened using SwissADME, and their targets were predicted using SwissTargetPrediction. Targets related to AF treatment were identified using GeneCards, OMIM, and other databases. Venn diagram was constructed using Venny 2.1 to obtain the intersection targets. The single herb-active component-potential target network was constructed using Cytoscape, and the clusterProfiler R function package was used to perform the gene ontology(GO) and Kyoto encyclopedia of genes and genomes(KEGG) pathway enrichment. The protein-protein interaction(PPI) network of intersection targets was generated based on the STRING database. The hub target protein was identified by visualization using Cytoscape, and then docked to its reverse-selected active components. The analysis showed that there were 65 active components with 681 corresponding targets in Shenmai Injection, 2 798 targets related to AF treatment, and 235 intersection targets involving 2 549 GO functions and 153 KEGG pathways. Finally, hub target proteins, including RAC-alpha serine/threonine-protein kinase(AKT1), phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha(PIK3 CA), and estrogen receptor 1(ESR1), were screened out by PPI network visualization. The molecular docking was performed for 39 active components screened out in reverse, among which 30 active components de-monstrated high affinity. Among them, homoisoflavanoids CID 10871974, CID 5319742, and CID 10361149 had stronger affinity docking with AKT1. This study preliminarily indicates that Shenmai Injection treats AF through multiple components, multiple targets, and multiple pathways. Homoisoflavonoids of Ophiopogon japonicus are its important active components, which target AKT1 to regulate metabolism, inflammation, and apoptosis in AF treatment.


Asunto(s)
Fibrilación Atrial , Medicamentos Herbarios Chinos , Fibrilación Atrial/tratamiento farmacológico , Combinación de Medicamentos , Humanos , Medicina Tradicional China , Simulación del Acoplamiento Molecular
4.
BMC Cell Biol ; 16: 22, 2015 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-26335746

RESUMEN

BACKGROUND: Environmental factors are important for stem cell lineage specification, and increasing evidence indicates that the nanoscale geometry/topography of the extracellular matrix (ECM) directs stem cell fate. Recently, many three-dimensional (3D) biomimetic nanofibrous scaffolds resembling many characteristics of the native ECM have been used in stem cell-based myocardial tissue engineering. However, the biophysical role and underlying mechanism of 3D nanofibrous scaffolds in cardiomyocyte differentiation of induced pluripotent stem cells (iPSCs) remain unclear. RESULTS: Here, we fabricated a 3D poly-(ε-caprolactone) (PCL) nanofibrous scaffold using the electrospinning method and verified its nanotopography and porous structure by scanning electron microscopy. We seeded murine iPSCs (miPSCs) directly on the 3D PCL nanofibrous scaffold and initiated non-directed, spontaneous differentiation using the monolayer method. After the 3D PCL nanofibrous scaffold was gelatin coated, it was suitable for monolayer miPSC cultivation and cardiomyocyte differentiation. At day 15 of differentiation, miPSCs differentiated into functional cardiomyocytes on the 3D PCL nanofibrous scaffold as evidenced by positive immunostaining of cardiac-specific proteins including cardiac troponin T (cTnT) and myosin light chain 2a (MLC2a). In addition, flow cytometric analysis of cTnT-positive cells and cardiac-specific gene and protein expression of cTnT and sarcomeric alpha actinin (α-actinin) demonstrated that the cardiomyocyte differentiation of miPSCs was more efficient on the 3D PCL nanofibrous scaffold than on normal tissue culture plates (TCPs). Furthermore, early inhibition of Wnt/ß-catenin signaling by the selective antagonist Dickkopf-1 significantly reduced the activity of Wnt/ß-catenin signaling and decreased the cardiomyocyte differentiation of miPSCs cultured on the 3D PCL nanofibrous scaffold, while the early activation of Wnt/ß-catenin signaling by CHIR99021 further increased the cardiomyocyte differentiation of miPSCs. CONCLUSION: These results indicated that the electrospun 3D PCL nanofibrous scaffolds directly promoted the cardiomyocyte differentiation of miPSCs, which was mediated by the activation of the Wnt/ß-catenin signaling during the early period of differentiation. These findings highlighted the biophysical role of 3D nanofibrous scaffolds during the cardiomyocyte differentiation of miPSCs and revealed its underlying mechanism involving Wnt/ß-catenin signaling, which will be helpful in guiding future stem cell- and scaffold-based myocardium bioengineering.


Asunto(s)
Diferenciación Celular , Células Madre Pluripotentes Inducidas/citología , Miocitos Cardíacos/citología , Nanofibras/química , Poliésteres/química , Ingeniería de Tejidos/métodos , Andamios del Tejido/química , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Animales , Células Cultivadas , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Miocitos Cardíacos/metabolismo , Transducción de Señal , Ingeniería de Tejidos/instrumentación , Proteínas Wnt/genética , beta Catenina/genética
5.
BMC Cell Biol ; 14: 5, 2013 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-23350814

RESUMEN

BACKGROUND: The interactions between stem cells and extracellular matrix (ECM) mediated by integrins play important roles in the processes that determine stem cell fate. However, the role of ECM/integrin interaction in the formation of embryoid bodies (EBs) during cardiogenesis from murine induced pluripotent stem cells (miPSCs) remains unclear. RESULTS: In the present study, collagen type I and ß(1) integrin were expressed and upregulated synergistically during the formation of miPSC-derived EBs, with a peak expression at day 3 of differentiation. The blockage of collagen/ß(1) integrin interaction by ß(1) integrin blocking antibody resulted in the production of defective EBs that were characterized by decreased size and the absence of a shell-like layer composed of primitive endoderm cells. The quantification of spontaneous beating activity, cardiac-specific gene expression and cardiac troponin T (cTnT) immunostaining showed that the cardiac differentiation of these defective miPSC-derived EBs was lower than that of control EBs. CONCLUSIONS: These findings indicate that collagen/ß(1) integrin interaction is required for the growth and cardiac differentiation of miPSC-derived EBs and will be helpful in future engineering of the matrix microenvironment within EBs to efficiently direct the cardiac fate of pluripotent stem cells to promote cardiovascular regeneration.


Asunto(s)
Colágeno Tipo I/metabolismo , Cuerpos Embrioides/citología , Células Madre Pluripotentes Inducidas/metabolismo , Integrina beta1/metabolismo , Animales , Anticuerpos/inmunología , Diferenciación Celular , Células Cultivadas , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Inmunohistoquímica , Células Madre Pluripotentes Inducidas/citología , Integrina beta1/inmunología , Ratones , Microscopía Electrónica de Rastreo , Miocardio/citología , Unión Proteica , Troponina T/metabolismo
6.
Mediators Inflamm ; 2012: 805149, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22778498

RESUMEN

Cardiac hypertrophy is an independent predictor of cardiovascular morbidity and mortality. In recent years, evidences suggest that high-mobility group box 1 (HMGB1) protein, an inflammatory cytokine, participates in cardiac remodeling; however, the involvement of HMGB1 in the pathogenesis of cardiac hypertrophy remains unknown. The aim of this study was to investigate whether HMGB1 is sufficient to induce cardiomyocyte hypertrophy and to identify the possible mechanisms underlying the hypertrophic response. Cardiomyocytes isolated from 1-day-old Sprague-Dawley rats were treated with recombinant HMGB1, at concentrations ranging from 50 ng/mL to 200 ng/mL. After 24 hours, cardiomyocytes were processed for the evaluation of atrial natriuretic peptide (ANP) and calcineurin A expression. Western blot and real-time RT-PCR was used to detect protein and mRNA expression levels, respectively. The activity of calcineurin was also evaluated using a biochemical enzyme assay. HMGB1 induced cardiomyocyte hypertrophy, characterized by enhanced expression of ANP, and increased protein synthesis. Meanwhile, increased calcineurin activity and calcineurin A protein expression were observed in cardiomyocytes preconditioned with HMGB1. Furthermore, cyclosporin A pretreatment partially inhibited the HMGB1-induced cardiomyocyte hypertrophy. Our findings suggest that HMGB1 leads to cardiac hypertrophy, at least in part through activating calcineurin.


Asunto(s)
Calcineurina/metabolismo , Cardiomegalia/metabolismo , Proteína HMGB1/farmacología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Animales , Animales Recién Nacidos , Factor Natriurético Atrial/metabolismo , Western Blotting , Calcineurina/genética , Células Cultivadas , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa
7.
J Cell Biochem ; 112(12): 3555-62, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21780160

RESUMEN

The cardiomyocyte (CM) differentiation of embryonic stem cells (ESCs) is routinely cultured as two-dimensional (2D) monolayer, which doesn't mimic in vivo physiological environment and may lead to low differentiated level of ESCs. Here, we develop a novel strategy that enhances CM differentiation of ESCs in collagen matrix three-dimensional (3D) culture combined with indirect cardiac fibroblasts co-culture. ESCs were cultured in hanging drops to form embryoid bodies (EBs) and then applied on collagen matrix. The EBs were indirectly co-cultured with cardiac fibroblasts by the hanging cell culture inserts (PET 1 µm). The molecular expressions and ultrastructural characteristics of ESC-derived CMs (ESCMs) were analyzed by real time RT-PCR, immunocytochemistry, and Transmission Electron Microscopy (TEM). We found that the percentage of beating EBs with cardiac fibroblasts co-culture was significantly higher than that without co-culture after differentiation period of 8 days. Type I collagen used as 3D substrates enhanced the late-stage CM differentiation of ESCs and had effect on ultrastructural mature of ESCMs in late-stage development. The combined effects of 3D and co-culture that mimic in vivo physiological environment further improved the efficiency of CM differentiation from ESCs, resulting in fiber-like structures of cardiac cells with organized sarcomeric structure in ESCMs. This novel 3D co-culture system emphasizes the fact that the ESC differentiation is actively responding to cues from their environment and those cues can drive phenotypic control, which provides a useful in vitro model to investigate CM differentiation of stem cells.


Asunto(s)
Diferenciación Celular , Células Madre Embrionarias/citología , Miocardio/citología , Animales , Secuencia de Bases , Técnicas de Cocultivo , Cartilla de ADN , Células Madre Embrionarias/ultraestructura , Inmunohistoquímica , Ratones , Microscopía Electrónica de Transmisión , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
8.
Biomarkers ; 16(8): 657-62, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21988056

RESUMEN

CONTEXT: Omentin-1, an adipokine secreted from visceral adipose tissue, has been reported to be associated with coronary artery disease (CAD) and metabolic disorders. OBJECTIVE: To clarify the relationship between serum omentin-1 levels and the presence and severity of CAD in patients with metabolic syndrome (MetS). METHODS: We measured serum omentin-1 levels in 175 consecutive patients with MetS and in 46 controls. RESULTS: Serum omentin-1 levels are inversely associated with the presence and angiographic severity of CAD in MetS patients. CONCLUSIONS: Serum omentin-1 might be a potential biomarker to predict the development and progression of CAD in MetS patients.


Asunto(s)
Enfermedad de la Arteria Coronaria/sangre , Citocinas/metabolismo , Lectinas/metabolismo , Síndrome Metabólico/sangre , Estudios de Casos y Controles , Enfermedad de la Arteria Coronaria/complicaciones , Proteínas Ligadas a GPI/metabolismo , Humanos , Síndrome Metabólico/complicaciones
9.
Bioessays ; 31(2): 246-52, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19204979

RESUMEN

Biological pacemakers can be achieved by various gene-based and cell-based approaches. Embryonic stem cells (ESCs)-derived pacemaker cells might be the most promising way to form biological pacemakers, but there are challenges as to how to control the differentiation of ESCs and to overcome the neoplasia, proarrhythmia, or immunogenicity resulting from the use of ESCs. As a potential approach to solve these difficult problems, tissue-engineering techniques may provide a precise control on the different cell components of multicellular aggregates and the forming of a construct with-defined architectures and functional properties. The combined interactions between ESC-derived pacemaker cells, supporting cells, and matrices may completely reproduce pacemaker properties and result in a steady functional unit to induce rhythmic electrical and contractile activities. As ESCs have a high capability for self-renewal, proliferation, and potential differentiation, we hypothesize that ESCs can be used as a source of pacemaker cells for tissue-engineering applications and the ambitious goal of biological cardiac pacemakers may ultimately be achieved with ESCs via tissue-engineering technology.


Asunto(s)
Relojes Biológicos , Células Madre Embrionarias/metabolismo , Ingeniería de Tejidos , Animales , Humanos
10.
Clin Exp Pharmacol Physiol ; 37(1): 40-5, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19515066

RESUMEN

1. The scaffolding protein Homer 1a is constitutively expressed in the myocardium, although its function in cardiomyocytes remains poorly understood. The aim of the present study was to investigate Homer 1a expression in hypertrophic cardiac cells and its role in angiotensin (Ang) II-induced cardiac hypertrophy. 2. After serum starvation for 24 h, cells were treated with 1 micromol/L simvastatin, 100 nmol/L angiotensin (Ang) II or their combination added to Dulbecco's modified Eagle's medium containing 0.5% serum. For combination treatment with AngII plus simvastatin, cells were exposed to simvastatin 12 h before the addition of AngII to the medium and cells were then incubated in the presence of both drugs for a further 24 h. Western blotting was used to determine Homer 1a protein expression. Hypertrophy was evaluated by determining the protein content per cell. 3. Homer 1a protein levels were upregulated following AngII-induced hypertrophy in H9C2 cells and neonatal rat cardiomyocytes, and these increases were augmented by simvastatin pretreatment. Concomitantly, simvastatin pretreatment inhibited extracellular signal-regulated kinase (ERK) 1/2 phosphorylation and AngII-induced hypertrophy. 4. The inhibitory effects of simvastatin against AngII-induced hypertrophy were attenuated by Homer 1a silencing, suggesting that simvastatin suppresses cardiac hypertrophy in a Homer 1a-dependent manner. Furthermore, AngII-induced hypertrophy and ERK1/2 phosphorylation in neonatal rat cardiomyocytes were significantly inhibited following the overexpression of Homer 1a using an adenovirus. 5. These results suggest a possible role for Homer 1a in inhibiting cardiac hypertrophy perhaps in part through inhibition of ERK1/2 activation.


Asunto(s)
Angiotensina II/antagonistas & inhibidores , Anticolesterolemiantes/farmacología , Cardiomegalia/fisiopatología , Proteínas Portadoras/fisiología , Miocitos Cardíacos/metabolismo , Simvastatina/farmacología , Angiotensina II/administración & dosificación , Angiotensina II/farmacología , Animales , Animales Recién Nacidos , Anticolesterolemiantes/administración & dosificación , Cardiomegalia/inducido químicamente , Cardiomegalia/tratamiento farmacológico , Cardiomegalia/metabolismo , Línea Celular , Interacciones Farmacológicas , Quimioterapia Combinada , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteínas de Andamiaje Homer , Fosforilación/efectos de los fármacos , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/farmacología , Ratas , Ratas Sprague-Dawley , Simvastatina/administración & dosificación , Transfección/métodos , Regulación hacia Arriba
11.
Cell Biol Int ; 33(10): 1123-4, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19545639

RESUMEN

This article gives a brief comment on the culture of human embryonic stem cells (hESCs) with the aim to maintain the potency of hESCs in well state and produce more homogenous cell clones.


Asunto(s)
Técnicas de Cultivo de Célula , Células Madre Embrionarias/fisiología , Células Clonales , Humanos
12.
Cell Biol Int ; 2009 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-19947942

RESUMEN

We studied the differentiation of embryonic stem cells (ESCs) and developed a novel protocol for generating functional cardiomyocytes (CMs) from ESCs by co-culturing these with live cardiac cells. We then evaluated the structural and functional properties of these ESC-derived CMs (ESCMs). An acellular matrix obtained from rabbit heart tissues was used as a scaffold. Then ESCMs were seeded onto the acellular matrix for preliminary tissue engineering applications. We found that by mimicking the cardiac microenvironment, the percentage of beating embryoid bodies (EBs) was much higher and the homogeneity of EBs were significantly improved over that seen in the control group (p<0.001). ESCMs in EBs acquired almost the same structural and functional properties as typical CMs. After implantation, the cells in the EBs rapidly grew and expanded in the extracellular matrix. These results indicate that the differentiation of ESCs can be controlled in a cardiac mimicking microenvironment and that ESCs can be used as an ideal cell source for large-scale tissue engineering applications for the procurement of cardiac muscle.

13.
Cardiology ; 112(3): 180-7, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-18679026

RESUMEN

BACKGROUND: Evidence has shown that autoantibodies against M2 muscarinic acetylcholine receptors may play a role in the development of atrial fibrillation. The goal of this study was to evaluate the effects of anti-M2 receptor autoantibodies on rabbit atria in vivo. METHODS: Rabbits were immunized monthly with a synthetic peptide corresponding to the M2 receptor. The atrial electrophysiology of the isolated perfused rabbit hearts was studied. Western blots and RT-PCR were performed to determine the expression of the atrial muscarinic receptor and the acetylcholine-activated potassium channel. Atrial tissue was stained with Masson's trichrome stain for fibrosis detection. RESULTS: Autoantibodies were persistently detected in immunized rabbits. M2 rabbits showed a significantly shorter atrial effective refractory period and a longer intra-atrial activation time than control rabbits. Electrical stimuli induced a significantly larger number of repetitive atrial responses in M2 rabbits. The protein levels of the M2 receptor and GIRK4 were upregulated in M2 rabbits. The mRNA levels of GIRK1 and GIRK4 were also upregulated. Histological examination revealed significantly increased diffuse fibrotic deposition in M2 rabbit atria compared with control rabbits. CONCLUSION: The M2 receptor autoantibody-positive rabbits showed altered atrial electrophysiology, overexpression of the M2 receptor-I(K,ACh) pathway and atrial fibrosis, which indicates that the autoantibodies against M2 receptors may participate in the induction and perpetuation of atrial fibrillation.


Asunto(s)
Fibrilación Atrial/inmunología , Autoanticuerpos/inmunología , Miocardio/inmunología , Receptor Muscarínico M2/inmunología , Animales , Fibrilación Atrial/patología , Fibrilación Atrial/fisiopatología , Estimulación Eléctrica , Ensayo de Inmunoadsorción Enzimática , Fibrosis , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/genética , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/inmunología , Atrios Cardíacos/inmunología , Atrios Cardíacos/metabolismo , Atrios Cardíacos/patología , Inmunización , Miocardio/metabolismo , Miocardio/patología , ARN Mensajero/metabolismo , Conejos , Receptor Muscarínico M2/genética , Receptor Muscarínico M2/metabolismo , Periodo Refractario Electrofisiológico/inmunología
15.
Med Hypotheses ; 71(3): 434-8, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18562127

RESUMEN

Dystrophin and its associated proteins form a scaffold underneath the cardiomyocyte membrane and connect the intracellular cytoskeleton to the extracellular matrix. Dystrophin localizes at the X chromosome, whose mutations might result in Duchenne muscular dystrophy, Becker muscular dystrophy and X-linked dilated cardiomyopathy. In addition to these genetic dilated cardiomyopathies, some acquired dilated cardiomyopathy like viral dilated cardiomyopathy is also related to dystrophin disruption or aberrant cleavage. In this review, we summarize the structure and distribution of dystrophin and researches of dystrophin in genetic and viral dilated cardiomyopathy. Moreover, we hypothesize that dystrophin play a critical role in ventricular remodeling in ischemic myocardium and treatment targeting restoration of dystrophin onto membrane could benefit for ischemic cardiomyopathy.


Asunto(s)
Cardiomiopatías/genética , Distrofina/metabolismo , Isquemia Miocárdica/genética , Remodelación Ventricular/fisiología , Distrofina/genética , Glicoproteínas/metabolismo , Humanos
16.
Sheng Li Xue Bao ; 60(3): 333-40, 2008 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-18560723

RESUMEN

Arginine vasopressin (AVP), a neurohormone and hemodynamic factor implicated in the pathophysiology of hypertension and congestive heart failure, can also act as a growth-stimulating factor. Our previous work demonstrated that AVP is a mitogen for neonatal rat cardiac fibroblasts (CFs). In the present study, we extended our investigations to adult rat CFs to explore whether AVP could induce adult rat CF proliferation and, if so, to identify the mechanism involved. Adult rat CFs were isolated, cultured and subjected to AVP treatment. DNA synthesis and cell cycle distribution were analyzed by [(3)H]-thymidine incorporation and flow cytometry. Cellular extracellular signal-regulated kinase 1/2 (ERK1/2) activity was measured by in vitro kinase assay using myelin basic protein (MBP) as a substrate. Protein expressions of total- and phospho-ERK1/2, p27(Kip1), cyclins D1, A, E were assessed by Western blot. The results showed that AVP stimulated DNA synthesis in adult rat CFs, and the effect was abolished by a V1 receptor antagonist, d(CH(2))(5)[Tyr(2)(Me), Arg(8)]-vasopressin (0.1 µmol/L), but not by a V2 receptor antagonist, desglycinamide-[d(CH(2))(5), D-Ile(2), Ile(4), Arg8]-vasopressin (0.1 µmol/L). AVP induced an activation of ERK1/2, which could be mimicked by the protein kinase C (PKC) activator, phorbol 12-myristate 13-acetate (PMA, 30 nmol/L, 5 min), but abolished by depletion of PKC via chronic PMA incubation (2.5 µmol/L, 24 h). In addition, AVP down-regulated protein expression of p27(Kip1), increased protein expressions of cyclins D1, A and E, and induced cell cycle progression from G(0)/G(1) into S stage. Inhibition of ERK1/2 activation by PD98059 (30 µmol/L) abolished the effect of AVP on DNA synthesis, protein expressions of p27(Kip1), cyclins D1, A and E as well as cell cycle progression. These results suggest that AVP is also a growth factor for adult rat CFs. The mitogenic effect of AVP is mediated via V1 receptors and PKC-ERK1/2 pathway. Moreover, AVP modulates the expressions of cell cycle regulatory proteins p27(Kip1) and cyclins D1, A and E, which lie downstream of ERK1/2 activation, and induces cell cycle progression in adult rat CFs.


Asunto(s)
Arginina Vasopresina/farmacología , Fibroblastos/efectos de los fármacos , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteína Quinasa C/metabolismo , Transducción de Señal , Animales , Antagonistas de los Receptores de Hormonas Antidiuréticas/farmacología , Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , Proliferación Celular , Fibroblastos/citología , Miocardio/citología , Fosforilación , Ratas , Acetato de Tetradecanoilforbol/farmacología
17.
Life Sci ; 81(4): 327-35, 2007 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-17604059

RESUMEN

Previous studies have shown that arginine vasopressin (AVP) promotes myocardial fibrosis (MF), whereas nitric oxide (NO) inhibits MF. Cardiac fibroblasts (CFs) are the main target cells of MF. However, the modulatory effect of AVP on NO production in CFs and the role of this effect in MF are still unknown. In the present study, CFs obtained from Sprague-Dawley rats were stimulated with or without AVP and pyrrolidine dithiocarbamate (PDTC), a specific inhibitor of nuclear factor kappa-B (NF-kappaB). NO production and NOS activity were detected with absorption spectrometry, inducible nitric oxide synthase (iNOS) protein with Western blot analysis, iNOS mRNA with real-time PCR, CF collagen synthesis with [(3)H]proline incorporation, and NF-kappaB activation with immunofluorescence staining and Western blot analysis. The results showed that AVP increased NO production in a dose- and time-dependent manner, with maximal effects at 10(-7) mol/l after 24-h stimulation. AVP also increased NOS activity, protein and mRNA levels of iNOS in a coincident manner. Furthermore, AVP also increased CF collagen synthesis in a dose- and time-dependent manner. In addition, it was found that NF-kappaB was activated by AVP, and that PDTC could inhibit NO production, NOS activity, protein and mRNA levels of iNOS stimulated by AVP in a dose-dependent manner. The inhibitory effects of PDTC on NF-kappaB translocation were coincident with the effects of PDTC on iNOS-NO system activity. It is suggested that AVP increases NO production via the regulation of iNOS gene expression, and the upregulation of iNOS gene expression stimulated by AVP is mediated through NF-kappaB activation. NO production induced by AVP may counteract the profibrotic effects of AVP, thus the development of MF perhaps depends on the balance between profibrotic AVP and antifibrotic NO effects on MF.


Asunto(s)
Arginina Vasopresina/metabolismo , Cardiomiopatías/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patología , FN-kappa B/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico/metabolismo , Animales , Arginina Vasopresina/farmacología , Cardiomiopatías/fisiopatología , Células Cultivadas , Colágeno/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Inducción Enzimática/genética , Inhibidores Enzimáticos/farmacología , Fibroblastos/ultraestructura , Fibrosis/metabolismo , Fibrosis/fisiopatología , Técnica del Anticuerpo Fluorescente , Regulación Enzimológica de la Expresión Génica , Técnicas In Vitro , Miocardio/citología , Miocardio/metabolismo , Miocardio/ultraestructura , FN-kappa B/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo II/genética , Pirrolidinas/farmacología , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , Espectrofotometría , Tiocarbamatos/farmacología
18.
Cell Prolif ; 50(1)2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27790820

RESUMEN

OBJECTIVES: Induced pluripotent stem cell (iPSC)-derived cardiomyocytes are a promising source of cells for regenerative heart disease therapies, but progress towards their use has been limited by their low differentiation efficiency and high cellular heterogeneity. Previous studies have demonstrated expression of adrenergic receptors (ARs) in stem cells after differentiation; however, roles of ARs in fate specification of stem cells, particularly in cardiomyocyte differentiation and development, have not been characterized. MATERIALS AND METHODS: Murine-induced pluripotent stem cells (miPSCs) were cultured in hanging drops to form embryoid bodies, cells of which were then differentiated into cardiomyocytes. To determine whether ARs regulated miPSC differentiation into cardiac lineages, effects of the AR agonist, epinephrine (EPI), on miPSC differentiation and underlying signalling mechanisms, were evaluated. RESULTS: Treatment with EPI, robustly enhanced miPSC cardiac differentiation, as indicated by increased expression levels of cardiac-specific markers, GATA4, Nkx2.5 and Tnnt2. Although ß-AR signalling is the foremost signalling pathway in cardiomyocytes, EPI-enhanced cardiac differentiation depended more on α-AR signalling than ß-AR signalling. In addition, selective activation of α1 -AR signalling with specific agonists induced vigorous cardiomyocyte differentiation, whereas selective activation of α2 - or ß-AR signalling induced no or less differentiation, respectively. EPI- and α1 -AR-dependent cardiomyocyte differentiation from miPSCs occurred through specific promotion of CPC proliferation via the MEK-ERK1/2 pathway and regulation of miPS cell-cycle progression. CONCLUSIONS: These results demonstrate that activation of ARs, particularly of α1 -ARs, promoted miPSC differentiation into cardiac lineages via MEK-ERK1/2 signalling.


Asunto(s)
Diferenciación Celular , Células Madre Pluripotentes Inducidas/citología , Miocitos Cardíacos/citología , Receptores Adrenérgicos alfa/metabolismo , Receptores Adrenérgicos beta/metabolismo , Agonistas Adrenérgicos beta/farmacología , Antagonistas Adrenérgicos beta/farmacología , Animales , Apoptosis , Puntos de Control del Ciclo Celular , Diferenciación Celular/efectos de los fármacos , Proliferación Celular , Células Cultivadas , Cuerpos Embrioides/metabolismo , Epinefrina/farmacología , Factor de Transcripción GATA4/metabolismo , Proteína Homeótica Nkx-2.5/metabolismo , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Microscopía Fluorescente , Miocitos Cardíacos/metabolismo , Receptores Adrenérgicos beta/química , Transducción de Señal/efectos de los fármacos , Troponina T/metabolismo
20.
Yao Xue Xue Bao ; 41(11): 1044-9, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17262945

RESUMEN

AIM: To investigate the effects of cyclosporin A (CsA) on growth and collagen synthesis of cardiac fibroblasts (CFs) induced by arginine vasopressin (AVP). METHODS: CFs of neonatal Sprague-Dawley rats were isolated by trypsinization and cultured; growth-arrested CFs were stimulated with 1 x 10(-7) mol x L(-1) AVP in the presence or absence of CsA (0.05, 0.5 and 5 micromol x L(-1)). MTT and flow cytometry techniques were adopted to measure cell number and analyze cell cycle respectively. Collagen synthesis was determined by measurement of hydroxyproline content in culture supernatant with colorimetry. Calcineurin activity was estimated by chemiluminescence. Trypan blue staining to test the viability of CFs. RESULTS: 0.05, 0.5 and 5 micromol x L(-1) CsA inhibited the increase of CFs number induced by 1 x 10(-7) mol x L(-1) AVP in a dose-dependent manner, with the inhibitory rates by 12%, 24% and 29%, respectively (P < 0.05). Furthermore, cell cycle analysis showed 0.5 micromol x L(-1) CsA decreased the S stage percentage and proliferation index of CFs stimulated by AVP (P < 0.05). In culture medium, the hydroxyproline content induced by AVP decreased by 0.5 and 5 micromol x L(-1) CsA (P < 0.05), with the inhibitory rates of 29% and 33%, respectively. CsA completely inhibited the increment of calcineurin activity induced by AVP (P < 0.01), but CsA itself had no effect on the baseline of calcineurin activity and CFs viability. CONCLUSION: CsA inhibits proliferation and collagen synthesis of CFs by virtue of blocking calcineurin signaling pathway and might provide a novel target for prevention and treatment to cardiac fibrosis.


Asunto(s)
Arginina Vasopresina/farmacología , Proliferación Celular/efectos de los fármacos , Colágeno/biosíntesis , Ciclosporina/farmacología , Fibroblastos/efectos de los fármacos , Animales , Animales Recién Nacidos , Calcineurina/metabolismo , Ciclo Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Ciclosporina/administración & dosificación , Relación Dosis-Respuesta a Droga , Fibroblastos/citología , Fibroblastos/metabolismo , Hidroxiprolina/metabolismo , Miocardio/citología , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA