Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 294(46): 17395-17408, 2019 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-31586031

RESUMEN

Piezo1 is a mechanosensitive cation channel with widespread physiological importance; however, its role in the heart is poorly understood. Cardiac fibroblasts help preserve myocardial integrity and play a key role in regulating its repair and remodeling following stress or injury. Here we investigated Piezo1 expression and function in cultured human and mouse cardiac fibroblasts. RT-PCR experiments confirmed that Piezo1 mRNA in cardiac fibroblasts is expressed at levels similar to those in endothelial cells. The results of a Fura-2 intracellular Ca2+ assay validated Piezo1 as a functional ion channel that is activated by its agonist, Yoda1. Yoda1-induced Ca2+ entry was inhibited by Piezo1 blockers (gadolinium and ruthenium red) and was reduced proportionally by siRNA-mediated Piezo1 knockdown or in murine Piezo1+/- cells. Results from cell-attached patch clamp recordings on human cardiac fibroblasts established that they contain mechanically activated ion channels and that their pressure responses are reduced by Piezo1 knockdown. Investigation of Yoda1 effects on selected remodeling genes indicated that Piezo1 activation increases both mRNA levels and protein secretion of IL-6, a pro-hypertrophic and profibrotic cytokine, in a Piezo1-dependent manner. Moreover, Piezo1 knockdown reduced basal IL-6 expression from cells cultured on softer collagen-coated substrates. Multiplex kinase activity profiling combined with kinase inhibitor experiments and phosphospecific immunoblotting established that Piezo1 activation stimulates IL-6 secretion via the p38 mitogen-activated protein kinase downstream of Ca2+ entry. In summary, cardiac fibroblasts express mechanically activated Piezo1 channels coupled to secretion of the paracrine signaling molecule IL-6. Piezo1 may therefore be important in regulating cardiac remodeling.


Asunto(s)
Interleucina-6/genética , Canales Iónicos/genética , Miocardio/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Animales , Señalización del Calcio/genética , Endopeptidasas/genética , Células Endoteliales/química , Células Endoteliales/metabolismo , Fibroblastos/metabolismo , Regulación de la Expresión Génica/genética , Técnicas de Silenciamiento del Gen , Humanos , Interleucina-6/química , Canales Iónicos/química , Sistema de Señalización de MAP Quinasas/genética , Mecanotransducción Celular/genética , Ratones , Miocardio/química , Fosforilación/genética , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Transducción de Señal/genética , Tioléster Hidrolasas/genética , Proteínas Quinasas p38 Activadas por Mitógenos/química
2.
Europace ; 20(12): 1898-1909, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-29771311

RESUMEN

Synchronous ventricular electrical activation is a prerequisite for adequate left ventricular (LV) systolic function. Conduction abnormalities such as left bundle branch block, and ventricular pacing lead to a dyssynchronous electrical activation sequence, which may have deleterious consequences. The present review attempts to connect the various processes involved in the development of 'dyssynchronopathy', and its correction by cardiac resynchronization therapy (CRT). Abnormal electrical impulse conduction leads to abnormal contraction, characterized by regional differences in timing as well as shortening patterns and amount of external work performed. Early activated regions may show 'wasted work', which leads to inefficient action of the entire left ventricle. Moreover, both the development of heart failure (HF) in general and the regional differences in mechanical load lead to structural, electrical, and contractile remodelling processes. These have been demonstrated at the level of the myocardium (asymmetric hypertrophy, fibrosis, prolongation of activation and reduction in repolarization forces, decrease in LV ejection fraction), cell (gap junctional remodelling, derangement of the T-tubular structure), and molecule (under or overexpression of ion channels and contractile proteins subtypes and abnormal calcium handling). The myocardial adaptations to dyssynchrony are 'maladaptive'. This also explains why CRT, unlike most pharmacological treatments, continues to increase its therapeutic effect over time. Finally, better understanding of all processes involved in dyssynchrony and CRT may also lead to new pharmacological agents for treating HF and to novel pacing strategies.


Asunto(s)
Arritmias Cardíacas/terapia , Terapia de Resincronización Cardíaca , Insuficiencia Cardíaca/terapia , Contracción Miocárdica , Disfunción Ventricular Izquierda/terapia , Función Ventricular Izquierda , Remodelación Ventricular , Potenciales de Acción , Animales , Arritmias Cardíacas/fisiopatología , Terapia de Resincronización Cardíaca/efectos adversos , Sistema de Conducción Cardíaco/fisiopatología , Insuficiencia Cardíaca/fisiopatología , Frecuencia Cardíaca , Humanos , Recuperación de la Función , Factores de Tiempo , Resultado del Tratamiento , Disfunción Ventricular Izquierda/fisiopatología
3.
J Clin Med ; 12(2)2023 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-36675594

RESUMEN

Cardiac resynchronization therapy (CRT) has become a valuable addition to the treatment options for heart failure, in particular for patients with disturbances in electrical conduction that lead to regionally different contraction patterns (dyssynchrony). Dyssynchronous hearts show extensive molecular and cellular remodeling, which has primarily been investigated in experimental animals. Evidence showing that at least several miRNAs play a role in this remodeling is increasing. A comparison of results from measurements in plasma and myocardial tissue suggests that plasma levels of miRNAs may reflect the expression of these miRNAs in the heart. Because many miRNAs released in the plasma are included in extracellular vesicles (EVs), which protect them from degradation, measurement of myocardium-derived miRNAs in peripheral blood EVs may open new avenues to investigate and monitor (reverse) remodeling in dyssynchronous and resynchronized hearts of patients.

4.
Cells ; 11(19)2022 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-36230924

RESUMEN

Heart disease, as well as systemic metabolic alterations, can leave a 'fingerprint' of structural and functional changes in the atrial myocardium, leading to the onset of atrial cardiomyopathy. As demonstrated in various animal models, some of these changes, such as fibrosis, cardiomyocyte hypertrophy and fatty infiltration, can increase vulnerability to atrial fibrillation (AF), the most relevant manifestation of atrial cardiomyopathy in clinical practice. Atrial cardiomyopathy accompanying AF is associated with thromboembolic events, such as stroke. The interaction between AF and stroke appears to be far more complicated than initially believed. AF and stroke share many risk factors whose underlying pathological processes can reinforce the development and progression of both cardiovascular conditions. In this review, we summarize the main mechanisms by which atrial cardiomyopathy, preceding AF, supports thrombogenic events within the atrial cavity and myocardial interstitial space. Moreover, we report the pleiotropic effects of activated coagulation factors on atrial remodeling, which may aggravate atrial cardiomyopathy. Finally, we address the complex association between AF and stroke, which can be explained by a multidirectional causal relation between atrial cardiomyopathy and hypercoagulability.


Asunto(s)
Fibrilación Atrial , Cardiomiopatías , Accidente Cerebrovascular , Animales , Factores de Coagulación Sanguínea , Cardiomiopatías/patología , Atrios Cardíacos/metabolismo , Accidente Cerebrovascular/patología
5.
Bioengineering (Basel) ; 9(10)2022 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-36290519

RESUMEN

Isolation and culturing of cardiac fibroblasts (CF) induces rapid differentiation toward a myofibroblast phenotype, which is partly mediated by the high substrate stiffness of the culture plates. In the present study, a 3D model of Engineered Heart Matrix (EHM) of physiological stiffness (Youngs modulus ~15 kPa) was developed using primary adult rat CF and a natural hydrogel collagen type 1 matrix. CF were equally distributed, viable and quiescent for at least 13 days in EHM and the baseline gene expression of myofibroblast-markers alfa-smooth muscle actin (Acta2), and connective tissue growth factor (Ctgf) was significantly lower, compared to CF cultured in 2D monolayers. CF baseline gene expression of transforming growth factor-beta1 (Tgfß1) and brain natriuretic peptide (Nppb) was higher in EHM-fibers compared to the monolayers. EHM stimulation by 10% cyclic stretch (1 Hz) increased the gene expression of Nppb (3.0-fold), Ctgf (2.1-fold) and Tgfß1 (2.3-fold) after 24 h. Stimulation of EHM with TGFß1 (1 ng/mL, 24 h) induced Tgfß1 (1.6-fold) and Ctgf (1.6-fold). In conclusion, culturing CF in EHM of physiological stiffness reduced myofibroblast marker gene expression, while the CF response to stretch or TGFß1 was maintained, indicating that our novel EHM structure provides a good physiological model to study CF function and myofibroblast differentiation.

6.
Front Physiol ; 13: 782592, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35634163

RESUMEN

Introduction: Computational modeling of cardiac mechanics and hemodynamics in ischemic heart disease (IHD) is important for a better understanding of the complex relations between ischemia-induced heterogeneity of myocardial tissue properties, regional tissue mechanics, and hemodynamic pump function. We validated and applied a lumped two-compartment modeling approach for IHD integrated into the CircAdapt model of the human heart and circulation. Methods: Ischemic contractile dysfunction was simulated by subdividing a left ventricular (LV) wall segment into a hypothetical contractile and noncontractile compartment, and dysfunction severity was determined by the noncontractile volume fraction ( N C V F ). Myocardial stiffness was determined by the zero-passive stress length ( L s 0 , p a s ) and nonlinearity ( k E C M ) of the passive stress-sarcomere length relation of the noncontractile compartment. Simulated end-systolic pressure volume relations (ESPVRs) for 20% acute ischemia were qualitatively compared between a two- and one-compartment simulation, and parameters of the two-compartment model were tuned to previously published canine data of regional myocardial deformation during acute and prolonged ischemia and reperfusion. In six patients with myocardial infarction (MI), the N C V F was automatically estimated using the echocardiographic LV strain and volume measurements obtained acutely and 6 months after MI. Estimated segmental N C V F values at the baseline and 6-month follow-up were compared with percentage late gadolinium enhancement (LGE) at 6-month follow-up. Results: Simulation of 20% of N C V F shifted the ESPVR rightward while moderately reducing the slope, while a one-compartment simulation caused a leftward shift with severe reduction in the slope. Through tuning of the N C V F , L s 0 , p a s , and k E C M , it was found that manipulation of the N C V F alone reproduced the deformation during acute ischemia and reperfusion, while additional manipulations of L s 0 , p a s and k E C M were required to reproduce deformation during prolonged ischemia and reperfusion. Out of all segments with LGE>25% at the follow-up, the majority (68%) had higher estimated N C V F at the baseline than at the follow-up. Furthermore, the baseline N C V F correlated better with percentage LGE than N C V F did at the follow-up. Conclusion: We successfully used a two-compartment model for simulation of the ventricular pump and tissue mechanics in IHD. Patient-specific optimizations using regional myocardial deformation estimated the N C V F in a small cohort of MI patients in the acute and chronic phase after MI, while estimated N C V F values closely approximated the extent of the myocardial scar at the follow-up. In future studies, this approach can facilitate deformation imaging-based estimation of myocardial tissue properties in patients with cardiovascular diseases.

7.
PLoS One ; 17(7): e0271588, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35839240

RESUMEN

AIMS: Progressive changes to left atrial (LA) structure and function following mitral regurgitation (MR) remain incompletely understood. This study aimed to demonstrate potential underlying mechanisms using experimental canine models and computer simulations. METHODS: A canine model of MR was created by cauterization of mitral chordae followed by radiofrequency ablation-induced left bundle-branch block (LBBB) after 4 weeks (MR-LBBB group). Animals with LBBB alone served as control. Echocardiography was performed at baseline, acutely after MR induction, and at 4 and 20 weeks, and correlated with histology and computer simulations. RESULTS: Acute MR augmented LA reservoir and contractile strain (40±4 to 53±6% and -11±5 to -22±9% respectively, p<0.05). LA fractional area change increased significantly (47±4 to 56±4%, p<0.05) while LA end-systolic area remained unchanged (7.2±1.1 versus 7.9±1.1 cm2 respectively, p = 0.08). LA strain 'pseudonormalized' after 4 weeks and decompensated at 20 weeks with both strains decreasing to 25±6% and -3±2% respectively (p<0.05) together with a progressive increase in LA end-systolic area (7.2±1.1 to 14.0±6.3 cm2, p<0.05). In the LBBB-group, LA remodeling was less pronounced. Histology showed a trend towards increased interstitial fibrosis in the LA of the MR-LBBB group. Computer simulations indicated that the progressive changes in LA structure and function are a combination of progressive eccentric remodeling and fibrosis. CONCLUSION: MR augmented LA strain acutely to supranormal values without significant LA dilation. However, over time, LA strain gradually decreases (pseudornormal and decompensated) with LA dilation. Histology and computer simulations indicated a correlation to a varying degree of LA eccentric remodeling and fibrosis.


Asunto(s)
Remodelación Atrial , Insuficiencia de la Válvula Mitral , Animales , Bloqueo de Rama , Perros , Ecocardiografía , Fibrosis , Atrios Cardíacos , Insuficiencia de la Válvula Mitral/diagnóstico por imagen
8.
Cells ; 10(7)2021 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-34359915

RESUMEN

In response to stretch, cardiac tissue produces natriuretic peptides, which have been suggested to have beneficial effects in heart failure patients. In the present study, we explored the mechanism of stretch-induced brain natriuretic peptide (Nppb) expression in cardiac fibroblasts. Primary adult rat cardiac fibroblasts subjected to 4 h or 24 h of cyclic stretch (10% 1 Hz) showed a 6.6-fold or 3.2-fold (p < 0.05) increased mRNA expression of Nppb, as well as induction of genes related to myofibroblast differentiation. Moreover, BNP protein secretion was upregulated 5.3-fold in stretched cardiac fibroblasts. Recombinant BNP inhibited TGFß1-induced Acta2 expression. Nppb expression was >20-fold higher in cardiomyocytes than in cardiac fibroblasts, indicating that cardiac fibroblasts were not the main source of Nppb in the healthy heart. Yoda1, an agonist of the Piezo1 mechanosensitive ion channel, increased Nppb expression 2.1-fold (p < 0.05) and significantly induced other extracellular matrix (ECM) remodeling genes. Silencing of Piezo1 reduced the stretch-induced Nppb and Tgfb1 expression in cardiac fibroblasts. In conclusion, our study identifies Piezo1 as mediator of stretch-induced Nppb expression, as well as other remodeling genes, in cardiac fibroblasts.


Asunto(s)
Fibroblastos/metabolismo , Regulación de la Expresión Génica , Proteínas de la Membrana/metabolismo , Miocardio/citología , Receptores del Factor Natriurético Atrial/genética , Estrés Mecánico , Animales , Fibroblastos/efectos de los fármacos , Fibrosis , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas de la Membrana/genética , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Receptores del Factor Natriurético Atrial/metabolismo , Proteínas Recombinantes/farmacología
9.
Mol Metab ; 53: 101293, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34265467

RESUMEN

OBJECTIVE: The diabetic heart is characterized by extensive lipid accumulation which often leads to cardiac contractile dysfunction. The underlying mechanism involves a pivotal role for vacuolar-type H+-ATPase (v-ATPase, functioning as endosomal/lysosomal proton pump). Specifically, lipid oversupply to the heart causes disassembly of v-ATPase and endosomal deacidification. Endosomes are storage compartments for lipid transporter CD36. However, upon endosomal deacidification, CD36 is expelled to translocate to the sarcolemma, thereby inducing myocardial lipid accumulation, insulin resistance, and contractile dysfunction. Hence, the v-ATPase assembly may be a suitable target for ameliorating diabetic cardiomyopathy. Another function of v-ATPase involves the binding of anabolic master-regulator mTORC1 to endosomes, a prerequisite for the activation of mTORC1 by amino acids (AAs). We examined whether the relationship between v-ATPase and mTORC1 also operates reciprocally; specifically, whether AA induces v-ATPase reassembly in a mTORC1-dependent manner to prevent excess lipids from entering and damaging the heart. METHODS: Lipid overexposed rodent/human cardiomyocytes and high-fat diet-fed rats were treated with a specific cocktail of AAs (lysine/leucine/arginine). Then, v-ATPase assembly status/activity, cell surface CD36 content, myocellular lipid uptake/accumulation, insulin sensitivity, and contractile function were measured. To elucidate underlying mechanisms, specific gene knockdown was employed, followed by subcellular fractionation, and coimmunoprecipitation. RESULTS: In lipid-overexposed cardiomyocytes, lysine/leucine/arginine reinternalized CD36 to the endosomes, prevented/reversed lipid accumulation, preserved/restored insulin sensitivity, and contractile function. These beneficial AA actions required the mTORC1-v-ATPase axis, adaptor protein Ragulator, and endosomal/lysosomal AA transporter SLC38A9, indicating an endosome-centric inside-out AA sensing mechanism. In high-fat diet-fed rats, lysine/leucine/arginine had similar beneficial actions at the myocellular level as in vitro in lipid-overexposed cardiomyocytes and partially reversed cardiac hypertrophy. CONCLUSION: Specific AAs acting through v-ATPase reassembly reduce cardiac lipid uptake raising the possibility for treatment in situations of lipid overload and associated insulin resistance.


Asunto(s)
Aminoácidos/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , ATPasas de Translocación de Protón Vacuolares/metabolismo , Aminoácidos/administración & dosificación , Animales , Dieta Alta en Grasa , Suplementos Dietéticos , Endosomas/efectos de los fármacos , Endosomas/metabolismo , Resistencia a la Insulina , Lípidos/efectos adversos , Masculino , Contracción Miocárdica/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Ratas , Ratas Endogámicas Lew
10.
Res Pract Thromb Haemost ; 5(8): e12620, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34849448

RESUMEN

The goat model of atrial fibrillation (AF) allows investigation of the effect of AF on coagulation. However, assays for goat plasma are not available from commercial sources. Calibrated automated thrombography (CAT) provides a global view of the coagulation profile by assessing in vitro thrombin generation (TG). We describe the customization of the CAT assay in goat platelet-poor plasma (PPP) and in factor Xa (FXa)-inhibitor-anticoagulated PPP. TG was initiated in the presence of phospholipids and either (a) PPP reagent, reagent low, or reagent high; (b) goat brain protein extraction (GBP); or (c) Russell's viper venom-factor X activator (RVV-X). Contact activation was assessed by adding corn trypsin inhibitor. Different concentrations of prothrombin complex concentrate (PCC) were used to determine the sensitivity of both the GBP and RVV-X method. To obtain FXa-inhibitor anticoagulated plasma, rivaroxaban was added to plasma. TG settings with human reagents were not suitable for goat plasma. TG triggered with GBP increased peak height and ETP values. Similarly, the RVV-X method produced comparable TG curves and was more sensitive to PCC titration. Finally, both methods were able to detect the decrease in clotting potential induced by FXa inhibition. This is the first study that reports the customization of the CAT assay for goats. The GBP and RVV-X methods were comparable in triggering TG in goat plasma. The RVV-X method seemed to better discriminate changes in TG curves due to increases in clotting potential as well as to FXa inhibition by rivaroxaban in goat plasma.

11.
Cells ; 10(11)2021 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-34831181

RESUMEN

Coagulation factor (F) Xa induces proinflammatory responses through activation of protease-activated receptors (PARs). However, the effect of FXa on cardiac fibroblasts (CFs) and the contribution of PARs in FXa-induced cellular signalling in CF has not been fully characterised. To answer these questions, human and rat CFs were incubated with FXa (or TRAP-14, PAR-1 agonist). Gene expression of pro-fibrotic and proinflammatory markers was determined by qRT-PCR after 4 and 24 h. Gene silencing of F2R (PAR-1) and F2RL1 (PAR-2) was achieved using siRNA. MCP-1 protein levels were measured by ELISA of FXa-conditioned media at 24 h. Cell proliferation was assessed after 24 h of incubation with FXa ± SCH79797 (PAR-1 antagonist). In rat CFs, FXa induced upregulation of Ccl2 (MCP-1; >30-fold at 4 h in atrial and ventricular CF) and Il6 (IL-6; ±7-fold at 4 h in ventricular CF). Increased MCP-1 protein levels were detected in FXa-conditioned media at 24 h. In human CF, FXa upregulated the gene expression of CCL2 (>3-fold) and IL6 (>4-fold) at 4 h. Silencing of F2R (PAR-1 gene), but not F2RL1 (PAR-2 gene), downregulated this effect. Selective activation of PAR-1 by TRAP-14 increased CCL2 and IL6 gene expression; this was prevented by F2R (PAR-1 gene) knockdown. Moreover, SCH79797 decreased FXa-induced proliferation after 24 h. In conclusion, our study shows that FXa induces overexpression of proinflammatory genes in human CFs via PAR-1, which was found to be the most abundant PARs isoform in this cell type.


Asunto(s)
Factor Xa/metabolismo , Fibroblastos/patología , Inflamación/patología , Miocardio/metabolismo , Receptor PAR-1/metabolismo , Adulto , Animales , Bovinos , Proliferación Celular , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Fibroblastos/metabolismo , Atrios Cardíacos/patología , Humanos , Interleucina-6/genética , Interleucina-6/metabolismo , Ratas Wistar , Receptor PAR-1/agonistas , Receptor PAR-1/genética , Trombina/metabolismo , Regulación hacia Arriba/genética
12.
Am J Physiol Heart Circ Physiol ; 299(3): H780-7, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20639217

RESUMEN

Both mechanical and humoral triggers have been put forward to explain the hypertrophic response of the challenged cardiomyocyte. The aim of the present study was to investigate whether cyclic equibiaxial stretch is a direct stimulus for isolated adult rabbit cardiomyocytes to develop hypertrophy and to explore the potential involvement of the autocrine/paracrine factors ANG II, transforming growth factor (TGF)-beta(1), and IGF-I in this process. Isolated cardiomyocytes were exposed to 10% cyclic equibiaxial stretch (1 Hz) for up to 48 h or treated with ANG II (100 nM), TGF-beta(1) (5 ng/ml), IGF-I (100 ng/ml), ANG II type 1 (AT(1)) receptor blockers, or conditioned medium of stretched fibroblasts. Cyclic stretch significantly increased cell surface area (+3.1%), protein synthesis (+21%), and brain natriuretic peptide (BNP) mRNA expression (6-fold) in cardiomyocytes. TGF-beta(1) expression increased (+42%) transiently at 4 h, whereas cardiomyocyte IGF-I expression was not detectable under all experimental conditions. The AT(1) receptor blockers candesartan and irbesartan (100 nM) did not prevent the stretch-induced hypertrophic response. Direct exposure to ANG II, TGF-beta(1), or IGF-I did not enhance cardiomyocyte BNP expression. In cardiac fibroblasts, stretch elicited a significant approximately twofold increase in TGF-beta(1) and IGF-I expression. Conditioned medium of stretched fibroblasts increased BNP expression in cardiomyocytes ( approximately 2-fold, P = 0.07). This study clearly indicates that cyclic stretch is a strong, direct trigger to induce hypertrophy in fully differentiated rabbit cardiomyocytes. The present findings do not support the notion that stretch-mediated hypertrophy of adult rabbit cardiomyocytes involves autocrine/paracrine actions of ANG II, TGF-beta(1), or IGF-I.


Asunto(s)
Aumento de la Célula , Miocitos Cardíacos/metabolismo , Estrés Mecánico , Angiotensina II/genética , Angiotensina II/metabolismo , Angiotensina II/farmacología , Animales , Tamaño de la Célula , Células Cultivadas , Medios de Cultivo Condicionados , Factor I del Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/farmacología , Masculino , Miocitos Cardíacos/efectos de los fármacos , Péptido Natriurético Encefálico/genética , Péptido Natriurético Encefálico/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Conejos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta/farmacología
13.
J Am Soc Nephrol ; 19(11): 2098-107, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18632843

RESUMEN

In diabetic nephropathy, connective tissue growth factor (CTGF) is upregulated and bone morphogenetic protein 7 (BMP-7) is downregulated. CTGF is known to inhibit BMP-4, but similar cross-talk between BMP-7 and CTGF has not been studied. In this study, it was hypothesized that CTGF acts as an inhibitor of BMP-7 signaling activity in diabetic nephropathy. Compared with diabetic wild-type CTGF(+/+) mice, diabetic CTGF(+/-) mice had approximately 50% lower CTGF mRNA and protein, less severe albuminuria, no thickening of the glomerular basement membrane, and preserved matrix metalloproteinase (MMP) activity. Although the amount of BMP-7 mRNA was similar in the kidneys of diabetic CTGF(+/+) and CTGF(+/-) mice, phosphorylation of the BMP signal transduction protein Smad1/5 and expression of the BMP target gene Id1 were lower in diabetic CTGF(+/+) mice. Moreover, renal Id1 mRNA expression correlated with albuminuria (R = -0.86) and MMP activity (R = 0.76). In normoglycemic mice, intraperitoneal injection of CTGF led to a decrease of pSmad1/5 in the renal cortex. In cultured renal glomerular and tubulointerstitial cells, CTGF diminished BMP-7 signaling activity, evidenced by lower levels of pSmad1/5, Id1 mRNA, and BMP-responsive element-luciferase activity. Co-immunoprecipitation, solid-phase binding assay, and surface plasmon resonance analysis showed that CTGF binds BMP-7 with high affinity (Kd approximately 14 nM). In conclusion, upregulation of CTGF inhibits BMP-7 signal transduction in the diabetic kidney and contributes to altered gene transcription, reduced MMP activity, glomerular basement membrane thickening, and albuminuria, all of which are hallmarks of diabetic nephropathy.


Asunto(s)
Proteínas Morfogenéticas Óseas/fisiología , Nefropatías Diabéticas/fisiopatología , Proteínas Inmediatas-Precoces/fisiología , Péptidos y Proteínas de Señalización Intercelular/fisiología , Factor de Crecimiento Transformador beta/fisiología , Animales , Proteína Morfogenética Ósea 7 , Proteínas Morfogenéticas Óseas/deficiencia , Proteínas Morfogenéticas Óseas/genética , Factor de Crecimiento del Tejido Conjuntivo , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Experimental/fisiopatología , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/patología , Femenino , Expresión Génica , Proteínas Inmediatas-Precoces/deficiencia , Proteínas Inmediatas-Precoces/genética , Proteínas Inmediatas-Precoces/farmacología , Proteína 1 Inhibidora de la Diferenciación/genética , Péptidos y Proteínas de Señalización Intercelular/deficiencia , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/farmacología , Riñón/metabolismo , Riñón/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Unión Proteica , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Recombinantes/farmacología , Transducción de Señal , Proteína Smad1/metabolismo , Proteína Smad5/metabolismo , Factor de Crecimiento Transformador beta/deficiencia , Factor de Crecimiento Transformador beta/genética
14.
Cardiovasc Res ; 78(1): 79-89, 2008 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-18187461

RESUMEN

AIMS: Peroxisome proliferator-activated receptor-alpha (PPARalpha) is a nuclear receptor regulating cardiac metabolism that also has anti-inflammatory properties. Since the activation of inflammatory signalling pathways is considered to be important in cardiac hypertrophy and fibrosis, it is anticipated that PPARalpha modulates cardiac remodelling. Accordingly, in this study the hypothesis was tested that the absence of PPARalpha aggravates the cardiac hypertrophic response to pressure overload. METHODS AND RESULTS: Male PPARalpha-/- and wild-type mice were subjected to transverse aortic constriction (TAC) for 28 days. TAC resulted in a more pronounced increase in ventricular weight and left ventricular (LV) wall thickness in PPARalpha-/- than in wild-type mice. Compared with sham-operated mice, TAC did not affect cardiac function in wild-type mice, but significantly depressed LV ejection fraction and LV contractility in PPARalpha-/- mice. Moreover, after TAC mRNA levels of hypertrophic (atrial natriuretic factor, alpha-skeletal actin), fibrotic (collagen 1, matrix metalloproteinase-2), and inflammatory (interleukin-6, tumour necrosis factor-alpha, cyclo-oxygenase-2) marker genes were higher in PPARalpha-/- than in wild-type mice. The mRNA levels of genes involved in fatty acid metabolism (long-chain acyl-CoA synthetase, hydroxyacyl-CoA dehydrogenase) were decreased in PPARalpha-/- mice, but were not further compromised by TAC. CONCLUSION: The present findings show that the absence of PPARalpha results in a more pronounced hypertrophic growth response and cardiac dysfunction that are associated with an enhanced expression of markers of inflammation and extracellular matrix remodelling. These findings indicate that PPARalpha exerts salutary effects during cardiac hypertrophy.


Asunto(s)
Hipertrofia Ventricular Izquierda/metabolismo , Miocardio/metabolismo , PPAR alfa/metabolismo , Remodelación Ventricular , 3-Hidroxiacil-CoA Deshidrogenasas/metabolismo , Actinas/metabolismo , Animales , Aorta Torácica/cirugía , Factor Natriurético Atrial/metabolismo , Coenzima A Ligasas/metabolismo , Colágeno Tipo I/metabolismo , Ciclooxigenasa 2/metabolismo , Modelos Animales de Enfermedad , Fibrosis , Hipertrofia Ventricular Izquierda/diagnóstico por imagen , Hipertrofia Ventricular Izquierda/genética , Hipertrofia Ventricular Izquierda/fisiopatología , Interleucina-6/metabolismo , Ligadura , Masculino , Metaloproteinasa 2 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Contracción Miocárdica , Miocardio/enzimología , Miocardio/patología , PPAR alfa/deficiencia , PPAR alfa/genética , ARN Mensajero/metabolismo , Volumen Sistólico , Factor de Necrosis Tumoral alfa/metabolismo , Ultrasonografía , Función Ventricular Izquierda , Remodelación Ventricular/genética
15.
Hypertension ; 71(2): 280-288, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29255073

RESUMEN

Pressure overload causes cardiac fibroblast activation and transdifferentiation, leading to increased interstitial fibrosis formation and subsequently myocardial stiffness, diastolic and systolic dysfunction, and eventually heart failure. A better understanding of the molecular mechanisms underlying pressure overload-induced cardiac remodeling and fibrosis will have implications for heart failure treatment strategies. The microRNA (miRNA)-221/222 family, consisting of miR-221-3p and miR-222-3p, is differentially regulated in mouse and human cardiac pathology and inversely associated with kidney and liver fibrosis. We investigated the role of this miRNA family during pressure overload-induced cardiac remodeling. In myocardial biopsies of patients with severe fibrosis and dilated cardiomyopathy or aortic stenosis, we found significantly lower miRNA-221/222 levels as compared to matched patients with nonsevere fibrosis. In addition, miRNA-221/222 levels in aortic stenosis patients correlated negatively with the extent of myocardial fibrosis and with left ventricular stiffness. Inhibition of both miRNAs during AngII (angiotensin II)-mediated pressure overload in mice led to increased fibrosis and aggravated left ventricular dilation and dysfunction. In rat cardiac fibroblasts, inhibition of miRNA-221/222 derepressed TGF-ß (transforming growth factor-ß)-mediated profibrotic SMAD2 (mothers against decapentaplegic homolog 2) signaling and downstream gene expression, whereas overexpression of both miRNAs blunted TGF-ß-induced profibrotic signaling. We found that the miRNA-221/222 family may target several genes involved in TGF-ß signaling, including JNK1 (c-Jun N-terminal kinase 1), TGF-ß receptor 1 and TGF-ß receptor 2, and ETS-1 (ETS proto-oncogene 1). Our findings show that heart failure-associated downregulation of the miRNA-221/222 family enables profibrotic signaling in the pressure-overloaded heart.


Asunto(s)
Insuficiencia Cardíaca/metabolismo , MicroARNs/metabolismo , Miocardio/metabolismo , Animales , Estenosis de la Válvula Aórtica/complicaciones , Estenosis de la Válvula Aórtica/metabolismo , Cardiomiopatías/metabolismo , Fibroblastos/metabolismo , Fibrosis/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Miocardio/patología , Proto-Oncogenes Mas , Ratas , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo
16.
Diabetes Care ; 29(1): 83-8, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16373901

RESUMEN

OBJECTIVE: Levels of connective tissue growth factor (CTGF; CCN-2) in plasma are increased in various fibrotic disorders, including diabetic nephropathy. Recently, several articles have reported a strong increase of urinary CTGF excretion (U-CTGF) in patients with diabetic nephropathy. However, these studies addressed too small a number of patients to allow general conclusions to be drawn. Therefore, we evaluated U-CTGF in a large cross-sectional study of patients with type 1 diabetes. RESEARCH DESIGN AND METHODS: Subjects were 318 type 1 diabetic patients and 29 normoglycemic control subjects. U-CTGF was measured by sandwich enzyme-linked immunosorbent assay. Groups were compared by Kruskal-Wallis and Mann-Whitney analysis. The relation between U-CTGF and markers of diabetic nephropathy was determined by regression analysis. RESULTS: U-CTGF in patients with diabetic nephropathy (n = 89, median 155 pmol/24 h [interquartile range 96-258]) was significantly higher than in microalbuminuric (n = 79, 100 [65-78]) and normoalbuminuric (n = 150, 85 [48-127]) patients and control subjects (n = 29, 100 [78-114]). U-CTGF correlated with urinary albumin excretion (UAE) (R = 0.31) and glomerular filtration rate (R = -0.38) in patients with diabetic nephropathy. A standardized increase in U-CTGF was associated with diabetic nephropathy (odds ratio 2.3 [95% CI 1.7-3.1]), which was comparable with the odds ratios for diabetic nephropathy of increased HbA(1c) (2.0 [1.5-2.7]), and blood pressure (2.0 [1.5-2.6]). CONCLUSIONS: This is the first large cross-sectional study addressing U-CTGF in human type 1 diabetes. The observed association of U-CTGF with UAE and glomerular filtration rate might reflect a role of CTGF as progression promoter in diabetic nephropathy.


Asunto(s)
Diabetes Mellitus Tipo 1/orina , Nefropatías Diabéticas/orina , Proteínas Inmediatas-Precoces/orina , Péptidos y Proteínas de Señalización Intercelular/orina , Adulto , Biomarcadores/orina , Glucemia/metabolismo , Factor de Crecimiento del Tejido Conjuntivo , Estudios Transversales , Nefropatías Diabéticas/epidemiología , Ensayo de Inmunoadsorción Enzimática , Femenino , Tasa de Filtración Glomerular , Hemoglobina Glucada/análisis , Humanos , Masculino , Persona de Mediana Edad , Valores de Referencia , Análisis de Regresión
17.
ESC Heart Fail ; 4(3): 241-251, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28772031

RESUMEN

AIMS: Left bundle branch block (LBBB) creates considerable regional differences in mechanical load within the left ventricle (LV). We investigated expression of selected microRNAs (miRs) in relation to regional hypertrophy and fibrosis in LBBB hearts and their reversibility upon cardiac resynchronization therapy (CRT). METHODS AND RESULTS: Eighteen dogs were followed for 4 months after induction of LBBB, 10 of which received CRT after 2 months. Five additional dogs served as control. LV geometric changes were determined by echocardiography and myocardial strain by magnetic resonance imaging tagging. Expression levels of miRs, their target genes: connective tissue growth factor (CTGF), serum response factor (SRF), nuclear factor of activated T cells (NFATc4), and cardiomyocyte diameter and collagen deposition were measured in the septum and LV free wall (LVfw). In LBBB hearts, LVfw and septal systolic circumferential strain were 200% and 50% of control, respectively. This coincided with local hypertrophy in the LVfw. MiR-133a expression was reduced by 33% in the LVfw, which corresponded with a selective increase of CTGF expression in the LVfw (279% of control). By contrast, no change was observed in SRF and NFATc4 expression was decreased in LBBB hearts. CRT normalized strain patterns and reversed miR-133a and CTGF expression towards normal, expression of other miRs, related to remodelling, such as miR-199b and miR-155f, were not affected. CONCLUSIONS: In the clinically relevant large animal model of LBBB, a close inverse relation exists between local hypertrophy and miR-133a. Reduced miR-133a correlated with increased CTGF levels but not with SRF and NFATc4.

18.
Sci Rep ; 7(1): 16042, 2017 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-29167509

RESUMEN

Heart failure is accompanied by extracellular matrix (ECM) remodelling, often leading to cardiac fibrosis. In the present study we explored the significance of cartilage intermediate layer protein 1 (CILP1) as a novel mediator of cardiac ECM remodelling. Whole genome transcriptional analysis of human cardiac tissue samples revealed a strong association of CILP1 with many structural (e.g. COL1A2 r2 = 0.83) and non-structural (e.g. TGFB3 r2 = 0.75) ECM proteins. Gene enrichment analysis further underscored the involvement of CILP1 in human cardiac ECM remodelling and TGFß signalling. Myocardial CILP1 protein levels were significantly elevated in human infarct tissue and in aortic valve stenosis patients. CILP1 mRNA levels markedly increased in mouse heart after myocardial infarction, transverse aortic constriction, and angiotensin II treatment. Cardiac fibroblasts were found to be the primary source of cardiac CILP1 expression. Recombinant CILP1 inhibited TGFß-induced αSMA gene and protein expression in cardiac fibroblasts. In addition, CILP1 overexpression in HEK293 cells strongly (5-fold p < 0.05) inhibited TGFß signalling activity. In conclusion, our study identifies CILP1 as a new cardiac matricellular protein interfering with pro-fibrotic TGFß signalling, and as a novel sensitive marker for cardiac fibrosis.


Asunto(s)
Proteínas de la Matriz Extracelular/metabolismo , Matriz Extracelular/metabolismo , Miocardio/metabolismo , Pirofosfatasas/metabolismo , Animales , Proteínas de la Matriz Extracelular/genética , Fibroblastos/citología , Fibroblastos/metabolismo , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Miocardio/patología , Pirofosfatasas/genética , Factor de Crecimiento Transformador beta/metabolismo
19.
Arch Ophthalmol ; 124(10): 1457-62, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17030714

RESUMEN

OBJECTIVE: To investigate the expression of the profibrotic connective tissue growth factor (CTGF) in relation to severity of intraocular fibrosis and neovascularization in human vitreoretinal disorders for the identification of potential therapeutic targets to prevent fibrosis. METHODS: Concentrations of CTGF were measured by enzyme-linked immunosorbent assay in 119 vitreous samples from patients with proliferative diabetic retinopathy, proliferative vitreoretinopathy, epiretinal membrane, and macular hole. Clinical data, including degree of intraocular fibrosis and neovascularization, were collected using standardized forms. RESULTS: Multifactorial analysis revealed that only CTGF levels correlated highly significantly with degree of fibrosis in the various vitreoretinal disorders studied (P<.001; R2= 47.7%). Likewise, variation in degree of fibrosis was best predicted by CTGF levels (P<.001). CONCLUSION: The strong correlation between CTGF levels and degree of fibrosis in vitreoretinal disorders suggests that CTGF is an important factor in ocular fibrosis, similar to its role in pathologic fibrosis in other organs. CLINICAL RELEVANCE: Connective tissue growth factor may be a therapeutic target for prevention of sight-threatening vitreoretinal scarring in the eye.


Asunto(s)
Retinopatía Diabética/metabolismo , Membrana Epirretinal/metabolismo , Proteínas Inmediatas-Precoces/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Perforaciones de la Retina/metabolismo , Vitreorretinopatía Proliferativa/metabolismo , Cuerpo Vítreo/metabolismo , Factor de Crecimiento del Tejido Conjuntivo , Retinopatía Diabética/patología , Ensayo de Inmunoadsorción Enzimática , Membrana Epirretinal/patología , Femenino , Fibrosis/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Retina/patología , Perforaciones de la Retina/patología , Vitreorretinopatía Proliferativa/patología , Cuerpo Vítreo/patología
20.
Diabetes ; 52(12): 2975-83, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14633859

RESUMEN

Tubulointerstitial fibrosis is an important component in the development of diabetic nephropathy. Various renal cell types, including fibroblasts, contribute to the excessive matrix deposition in the kidney. Although transforming growth factor-beta (TGF-beta) has been thought to play a major role during fibrosis, other growth factors are also involved. Here we examined the effects of connective tissue growth factor (CTGF) and IGF-I on collagen type I and III production by human renal fibroblasts and their involvement in glucose-induced matrix accumulation. We have demonstrated that both CTGF and IGF-I expressions were increased in renal fibroblasts under hyperglycemic conditions, also in the absence of TGF-beta signaling. Although CTGF alone had no effect on collagen secretion, combined stimulation with IGF-I enhanced collagen accumulation. Furthermore, IGF-I also had a synergistic effect with glucose on the induction of collagens. Moreover, we observed a partial inhibition in glucose-induced collagen secretion with neutralizing anti-CTGF antibodies, thereby demonstrating for the first time the involvement of endogenous CTGF in glucose-induced effects in human renal fibroblasts. Therefore, the cooperation between CTGF and IGF-I might be involved in glucose-induced matrix accumulation in tubulointerstitial fibrosis and might contribute to the pathogenesis of diabetic nephropathy.


Asunto(s)
Colágeno Tipo III/biosíntesis , Colágeno Tipo I/biosíntesis , Fibroblastos/metabolismo , Glucosa/administración & dosificación , Proteínas Inmediatas-Precoces/fisiología , Factor I del Crecimiento Similar a la Insulina/fisiología , Péptidos y Proteínas de Señalización Intercelular/fisiología , Riñón/metabolismo , Células Cultivadas , Factor de Crecimiento del Tejido Conjuntivo , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Fibroblastos/efectos de los fármacos , Humanos , Proteínas Inmediatas-Precoces/biosíntesis , Proteínas Inmediatas-Precoces/farmacología , Factor I del Crecimiento Similar a la Insulina/biosíntesis , Factor I del Crecimiento Similar a la Insulina/farmacología , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Péptidos y Proteínas de Señalización Intercelular/farmacología , Proteínas Recombinantes/farmacología , Factor de Crecimiento Transformador beta/farmacología , Factor de Crecimiento Transformador beta1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA