Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
Neurochem Res ; 47(8): 2416-2430, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35716295

RESUMEN

Myocardial infraction (MI) is the principal risk factor for the onset of heart failure (HF). Investigations regarding the physiopathology of MI progression to HF have revealed the concerted engagement of other tissues, such as the autonomic nervous system and the medulla oblongata (MO), giving rise to systemic effects, important in the regulation of heart function. Cardiac sympathetic afferent denervation following application of resiniferatoxin (RTX) attenuates cardiac remodelling and restores cardiac function following MI. While the physiological responses are well documented in numerous species, the underlying molecular responses during the initiation and progression from MI to HF remains unclear. We obtained multi-tissue time course proteomics with a murine model of HF induced by MI in conjunction with RTX application. We isolated tissue sections from the left ventricle (LV), MO, cervical spinal cord and cervical vagal nerves at four time points over a 12-week study. Bioinformatic analyses consistently revealed a high statistical enrichment for metabolic pathways in all tissues and treatments, implicating a central role of mitochondria in the tissue-cellular response to both MI and RTX. In fact, the additional functional pathways found to be enriched in these tissues, involving the cytoskeleton, vesicles and signal transduction, could be downstream of responses initiated by mitochondria due to changes in neuronal pulse frequency after a shock such as MI or the modification of such frequency communication from the heart to the brain after RTX application. Development of future experiments, based on our proteomic results, should enable the dissection of more precise mechanisms whereby metabolic changes in neuronal and cardiac tissues can effectively ameliorate the negative physiological effects of MI via RTX application.


Asunto(s)
Insuficiencia Cardíaca , Infarto del Miocardio , Animales , Desnervación , Modelos Animales de Enfermedad , Redes y Vías Metabólicas , Ratones , Infarto del Miocardio/metabolismo , Proteómica , Transducción de Señal
2.
Mol Ther ; 26(3): 902-916, 2018 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-29249393

RESUMEN

Despite considerable advances in cardiovascular disease treatment, heart failure remains a public health challenge. In this context, gene therapy appears as an attractive approach, but clinical trials using single therapeutic molecules result in moderate benefit. With the objective of improving ischemic heart failure therapy, we designed a combined treatment, aimed to simultaneously stimulate angiogenesis, prevent cardiac remodeling, and restore contractile function. We have previously validated IRES-based vectors as powerful tools to co-express genes of interest. Mono- and multicistronic lentivectors expressing fibroblast growth factor 2 (angiogenesis), apelin (cardioprotection), and/or SERCA2a (contractile function) were produced and administrated by intramyocardial injection into a mouse model of myocardial infarction. Data reveal that combined treatment simultaneously improves vessel number, heart function parameters, and fibrosis prevention, due to FGF2, SERCA2a, and apelin, respectively. Furthermore, addition of SERCA2a in the combination decreases cardiomyocyte hypertrophy. Large-scale transcriptome analysis reveals that the triple treatment is the most efficient in restoring angiogenic balance as well as expression of genes involved in cardiac function and remodeling. Our study validates the concept of combined treatment of ischemic heart disease with apelin, FGF2, and SERCA2a and shows that such therapeutic benefit is mediated by a more effective recovery of gene network regulation.


Asunto(s)
Apelina/genética , Factor 2 de Crecimiento de Fibroblastos/genética , Expresión Génica , Redes Reguladoras de Genes , Isquemia Miocárdica/genética , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/genética , Animales , Cardiomegalia , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Fibrosis , Orden Génico , Técnicas de Transferencia de Gen , Terapia Genética , Vectores Genéticos/genética , Lentivirus/genética , Ratones , Isquemia Miocárdica/patología , Isquemia Miocárdica/terapia , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Transcriptoma , Transducción Genética
3.
Circulation ; 129(21): 2111-24, 2014 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-24657994

RESUMEN

BACKGROUND: The mechanisms by which the heart adapts to chronic pressure overload, producing compensated hypertrophy and eventually heart failure (HF), are still not well defined. We aimed to investigate the involvement of T cells in the progression to HF using a transverse aortic constriction (TAC) model. METHODS AND RESULTS: Chronic HF was associated with accumulation of T lymphocytes and activated/effector CD4(+) T cells within cardiac tissue. After TAC, enlarged heart mediastinal draining lymph nodes showed a high density of both CD4(+) and CD8(+) T-cell subsets. To investigate the role of T cells in HF, TAC was performed on mice deficient for recombination activating gene 2 expression (RAG2KO) lacking B and T lymphocytes. Compared with wild-type TAC mice, RAG2KO mice did not develop cardiac dilation and showed improved contractile function and blunted adverse remodeling. Reconstitution of the T-cell compartment into RAG2KO mice before TAC enhanced contractile dysfunction, fibrosis, collagen accumulation, and cross-linking. To determine the involvement of a specific T-cell subset, we performed TAC on mice lacking CD4(+) (MHCIIKO) and CD8(+) T-cell subsets (CD8KO). In contrast to CD8KO mice, MHCIIKO mice did not develop ventricular dilation and dysfunction. MHCIIKO mice also displayed very low fibrosis, collagen accumulation, and cross-linking within cardiac tissue. Interestingly, mice with transgenic CD4(+) T-cell receptor specific for ovalbumin failed to develop HF and adverse remodeling. CONCLUSIONS: These results demonstrate for the first time a crucial role of CD4(+) T cells and specific antigen recognition in the progression from compensated cardiac hypertrophy to HF.


Asunto(s)
Linfocitos T CD4-Positivos/patología , Cardiomegalia/patología , Progresión de la Enfermedad , Insuficiencia Cardíaca/patología , Animales , Linfocitos T CD4-Positivos/inmunología , Cardiomegalia/inmunología , Insuficiencia Cardíaca/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos
4.
Circ Res ; 110(5): 688-700, 2012 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-22302788

RESUMEN

RATIONALE: Cardiac tissue cohesion relying on highly ordered cardiomyocytes (CM) interactions is critical because most cardiomyopathies are associated with tissue remodeling and architecture alterations. OBJECTIVE: Eph/ephrin system constitutes a ubiquitous system coordinating cellular communications which recently emerged as a major regulator in adult organs. We examined if eph/ephrin could participate in cardiac tissue cyto-organization. METHODS AND RESULTS: We reported the expression of cardiac ephrin-B1 in both endothelial cells and for the first time in CMs where ephrin-B1 localized specifically at the lateral membrane. Ephrin-B1 knock-out (KO) mice progressively developed cardiac tissue disorganization with loss of adult CM rod-shape and sarcomeric and intercalated disk structural disorganization confirmed in CM-specific ephrin-B1 KO mice. CMs lateral membrane exhibited abnormal structure by electron microscopy and notably increased stiffness by atomic force microscopy. In wild-type CMs, ephrin-B1 interacted with claudin-5/ZO-1 complex at the lateral membrane, whereas the complex disappeared in KO/CM-specific ephrin-B1 KO mice. Ephrin-B1 deficiency resulted in decreased mRNA expression of CM basement membrane components and disorganized fibrillar collagen matrix, independently of classical integrin/dystroglycan system. KO/CM-specific ephrin-B1 KO mice exhibited increased left ventricle diameter and delayed atrioventricular conduction. Under pressure overload stress, KO mice were prone to death and exhibited striking tissue disorganization. Finally, failing CMs displayed downregulated ephrin-B1/claudin-5 gene expression linearly related to the ejection fraction. CONCLUSIONS: Ephrin-B1 is necessary for cardiac tissue architecture cohesion by stabilizing the adult CM morphology through regulation of its lateral membrane. Because decreased ephrin-B1 is associated with molecular/functional cardiac defects, it could represent a new actor in the transition toward heart failure.


Asunto(s)
Comunicación Celular/fisiología , Efrina-B1/fisiología , Proteínas de la Membrana/fisiología , Miocitos Cardíacos/fisiología , Animales , Membrana Celular/fisiología , Membrana Celular/ultraestructura , Células Cultivadas , Colágeno/fisiología , Colágeno/ultraestructura , Endotelio Vascular/citología , Endotelio Vascular/fisiología , Endotelio Vascular/ultraestructura , Efrina-B1/deficiencia , Efrina-B1/genética , Masculino , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Animales , Miocitos Cardíacos/citología , Miocitos Cardíacos/ultraestructura , Sarcómeros/diagnóstico por imagen , Sarcómeros/fisiología , Ultrasonografía
5.
Biochem Biophys Res Commun ; 438(2): 257-63, 2013 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-23872063

RESUMEN

Most end-stage renal disease kidneys display accumulation of extracellular matrix (ECM) in the renal tubular compartment (tubular interstitial fibrosis - TIF) which is strongly correlated with the future loss of renal function. Although inflammation is a key event in the development of TIF, it can also have a beneficial anti-fibrotic role depending in particular on the stage of the pathology. Chemokines play an important role in monocyte extravasation in the inflammatory process. CCL2 has already been shown to be involved in the development of TIF but CCL7, a close relative of CCL2 and able to bind to similar receptors, has not been studied in renal disease. We therefore studied chemokine CCL7 in a model of unilateral ureteral obstruction (UUO)-induced TIF. We observed that the role of CCL7 differs depending on the stage of the pathology. In early stages (0-8 days), CCL7 deficient (CCL7-KO) mice displayed attenuated TIF potentially involving two mechanisms: an early (0-3 days) decrease of inflammatory cell infiltration followed (3-8 days) by a decrease in tubular ECM production independent of inflammation. In contrast, during later stages of obstruction (10-14 days), CCL7-KO mice displayed increased TIF which was again associated with reduced inflammation. Interestingly, the switch between this anti- to profibrotic effect was accompanied by an increased influx of immunosuppressive regulatory T cells. In conclusion, these results highlight for the first time a dual role for CCL7 in the development of renal TIF, deleterious in early stages but beneficial during later stages.


Asunto(s)
Quimiocina CCL7/fisiología , Túbulos Renales/metabolismo , Animales , Línea Celular , Quimiocina CCL7/genética , Colágeno/metabolismo , Modelos Animales de Enfermedad , Fibrosis , Humanos , Inflamación/patología , Riñón/metabolismo , Túbulos Renales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Linfocitos T Reguladores/metabolismo , Factores de Tiempo
6.
Eur Heart J ; 33(18): 2360-9, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22028387

RESUMEN

AIMS: Activation of cardiac fibroblasts and their differentiation into myofibroblasts is a key event in the progression of cardiac fibrosis that leads to end-stage heart failure. Apelin, an adipocyte-derived factor, exhibits a number of cardioprotective properties; however, whether apelin is involved in cardiac fibroblast activation and myofibroblast formation remains unknown. The aim of this study was to determine the effects of apelin in activated cardiac fibroblasts, the potential related mechanisms and impact on cardiac fibrotic remodelling process. METHODS AND RESULTS: In vitro experiments were performed in mouse cardiac fibroblasts obtained from normal and pressure-overload hearts. Pretreatment of naive cardiac fibroblasts with apelin (1-100 nM) inhibited Transforming growth factor-ß (TGF-ß)-mediated expression of the myofibroblast marker α-smooth muscle actin (α-SMA) and collagen production. Furthermore, apelin decreased the spontaneous collagen production in cardiac fibroblasts isolated from hearts after aortic banding. Knockdown strategy and pharmacological inhibition revealed that prevention of collagen accumulation by apelin was mediated by a reduction in sphingosine kinase 1 (SphK1) activity. In vivo studies using the aortic banding model indicated that pretreatment with apelin attenuated the development of myocardial fibrotic remodelling and inhibited cardiac SphK1 activity and α-SMA expression. Moreover, administration of apelin 2 weeks after aortic banding prevented cardiac remodelling by inhibiting myocyte hypertrophy, cardiac fibrosis, and ventricular dysfunction. CONCLUSION: Our data provide the first evidence that apelin inhibits TGF-ß-stimulated activation of cardiac fibroblasts through a SphK1-dependent mechanism. We also demonstrated that the administration of apelin during the phase of reactive fibrosis prevents structural remodelling of the myocardium and ventricular dysfunction. These findings may have important implications for designing future therapies for myocardial performance during fibrotic remodelling, affecting the clinical management of patients with progressive heart failure.


Asunto(s)
Colágeno/biosíntesis , Fibroblastos/fisiología , Péptidos y Proteínas de Señalización Intercelular/fisiología , Miocitos Cardíacos/fisiología , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Proteínas Quinasas Activadas por AMP/farmacología , Adipoquinas , Animales , Apelina , Inhibidores Enzimáticos/farmacología , Hemodinámica/fisiología , Péptidos y Proteínas de Señalización Intercelular/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Distribución Aleatoria , Factor de Crecimiento Transformador beta/farmacología , Remodelación Ventricular/fisiología
7.
Life (Basel) ; 13(3)2023 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-36983999

RESUMEN

Whether in real or simulated microgravity, Humans or animals, the kinetics of cardiovascular adaptation and its regulation by the autonomic nervous system (ANS) remain controversial. In this study, we used hindlimb unloading (HU) in 10 conscious mice. Blood pressure (BP), heart rate (HR), temperature, and locomotor activity were continuously monitored with radio-telemetry, during 3 days of control, 5 days of HU, and 2 days of recovery. Six additional mice were used to assess core temperature. ANS activity was indirectly determined by analyzing both heart rate variability (HRV) and baroreflex sensitivity (BRS). Our study showed that HU induced an initial bradycardia, accompanied by an increase in vagal activity markers of HRV and BRS, together with a decrease in water intake, indicating the early adaptation to fluid redistribution. During HU, BRS was reduced; temperature and BP circadian rhythms were altered, showing a loss in day/night differences, a decrease in cycle amplitude, a drop in core body temperature, and an increase in day BP suggestive of a rise in sympathetic activity. Reloading induced resting tachycardia and a decrease in BP, vagal activity, and BRS. In addition to cardiovascular deconditioning, HU induces disruption in day/night rhythmicity of locomotor activity, temperature, and BP.

8.
Circulation ; 124(24): 2725-34, 2011 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-22082680

RESUMEN

BACKGROUND: Outcomes for organ transplantation are constantly improving because of advances in organ preservation, surgical techniques, immune clinical monitoring, and immunosuppressive treatment preventing acute transplant rejection. However, chronic rejection including transplant vasculopathy still limits long-term patient survival. Transplant vasculopathy is characterized by progressive neointimal hyperplasia leading to arterial stenosis and ischemic failure of the allograft. This work sought to decipher the manner in which the humoral immune response, mimicked by W6/32 anti-HLA antibody, contributes to transplant vasculopathy. METHODS AND RESULTS: Studies were performed in vitro on cultured human smooth muscle cells, ex vivo on human arterial segments, and in vivo in a model consisting of human arterial segments grafted into severe combined immunodeficiency/beige mice injected weekly with anti-HLA antibodies. We report that anti-HLA antibodies are mitogenic for smooth muscle cells through a signaling mechanism implicating matrix metalloproteinases (MMPs) (membrane type 1 MMP and MMP2) and neutral sphingomyelinase-2. This mitogenic signaling and subsequent DNA synthesis are blocked in smooth muscle cells silenced for MMP2 or for neutral sphingomyelinase-2 by small interfering RNAs, in smooth muscle cells transfected with a vector coding for a dominant-negative form of membrane type 1 MMP, and after treatment by pharmacological inhibitors of MMPs (Ro28-2653) or neutral sphingomyelinase-2 (GW4869). In vivo, Ro28-2653 and GW4869 reduced the intimal thickening induced by anti-HLA antibodies in human mesenteric arteries grafted into severe combined immunodeficiency/beige mice. CONCLUSIONS: These data highlight a crucial role for MMP2 and neutral sphingomyelinase-2 in vasculopathy triggered by a humoral immune response and open new perspectives for preventing transplant vasculopathy with the use of MMP and neutral sphingomyelinase inhibitors, in addition to conventional immunosuppression.


Asunto(s)
Anticuerpos Antiidiotipos/farmacología , Arterias/trasplante , Antígenos HLA/inmunología , Metaloproteinasa 14 de la Matriz/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , Esfingomielina Fosfodiesterasa/metabolismo , Enfermedades Vasculares/fisiopatología , Compuestos de Anilina/farmacología , Animales , Anticuerpos Antiidiotipos/efectos adversos , Arterias/patología , Arterias/fisiopatología , Compuestos de Bencilideno/farmacología , Células Cultivadas , Constricción Patológica/etiología , Constricción Patológica/fisiopatología , Modelos Animales de Enfermedad , Humanos , Hiperplasia/etiología , Hiperplasia/fisiopatología , Técnicas In Vitro , Metaloproteinasa 14 de la Matriz/efectos de los fármacos , Metaloproteinasa 2 de la Matriz/efectos de los fármacos , Inhibidores de la Metaloproteinasa de la Matriz , Ratones , Ratones SCID , Modelos Animales , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Neointima/patología , Neointima/fisiopatología , Piperazinas/farmacología , Pirimidinas/farmacología , ARN Interferente Pequeño/farmacología , Esfingomielina Fosfodiesterasa/antagonistas & inhibidores , Esfingomielina Fosfodiesterasa/efectos de los fármacos , Enfermedades Vasculares/etiología , Injerto Vascular
9.
Sci Rep ; 11(1): 16801, 2021 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-34413352

RESUMEN

The lymphatic network of mammalian heart is an important regulator of interstitial fluid compartment and immune cell trafficking. We observed a remodeling of the cardiac lymphatic vessels and a reduced lymphatic efficiency during heart hypertrophy and failure induced by transverse aortic constriction. The lymphatic endothelial cell number of the failing hearts was positively correlated with cardiac function and with a subset of cardiac macrophages. This macrophage population distinguished by LYVE-1 (Lymphatic vessel endothelial hyaluronic acid receptor-1) and by resident macrophage gene expression signature, appeared not replenished by CCR2 mediated monocyte infiltration during pressure overload. Isolation of macrophage subpopulations showed that the LYVE-1 positive subset sustained in vitro and in vivo lymphangiogenesis through the expression of pro-lymphangiogenic factors. In contrast, the LYVE-1 negative macrophage subset strongly expressed MMP12 and decreased the endothelial LYVE-1 receptors in lymphatic endothelial cells, a feature of cardiac lymphatic remodeling in failing hearts. The treatment of mice with a CCR2 antagonist during pressure overload modified the proportion of macrophage subsets within the pathological heart and preserved lymphatic network from remodeling. This study reports unknown and differential functions of macrophage subpopulations in the regulation of cardiac lymphatic during pathological hypertrophy and may constitute a key mechanism underlying the progression of heart failure.


Asunto(s)
Vasos Linfáticos/metabolismo , Macrófagos/metabolismo , Miocardio/patología , Presión , Animales , Benzoxazinas/farmacología , Células CHO , Polaridad Celular/efectos de los fármacos , Cricetulus , Electrocardiografía , Células Endoteliales/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Linfangiogénesis/efectos de los fármacos , Vasos Linfáticos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Monocitos/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores CCR2/metabolismo , Compuestos de Espiro/farmacología , Transcriptoma , Proteínas de Transporte Vesicular/metabolismo
10.
Acta Physiol (Oxf) ; 231(4): e13601, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33316126

RESUMEN

AIM: Impairments in cerebral structure and cognitive performance in chronic heart failure (CHF) are critical components of its comorbidity spectrum. Autonomic afferents that arise from cardiac sensory fibres show enhanced activity with CHF. Desensitization of these fibres by local application of resiniferatoxin (RTX) during myocardial infarction (MI) is known to prevent cardiac hypertrophy, sympathetic hyperactivity and CHF. Whether these afferents mediate cerebral allostasis is unknown. METHODS: CHF was induced by myocardial infarction. To evaluate if cardiac afferents contribute to cerebral allostasis, RTX was acutely applied to the pericardial space in controls (RTX) and in MI treated animals (MI/RTX). Subjects were then evaluated in a series of behavioural tests recapitulating different symptoms of depressive disorders. Proteomics of the frontal cortices (FC) was performed to identify contributing proteins and pathways responsible for behavioural allostasis. RESULTS: Desensitization of cardiac afferents relieves hallmarks of an anxio/depressive-like state in mice. Unique protein signatures and regulatory pathways in FCs isolated from each treatment reveal the degree of complexity inherent in the FC response to stresses originating in the heart. While cortices from the combined treatment (MI/RTX) did not retain protein signatures from the individual treatment groups, all three groups suffer dysregulation in circadian entrainment. CONCLUSION: CHF is comorbid with an anxio/depressive-like state and ablation of cardiac afferents relieves the despair phenotype. The strikingly different proteomic profiles observed in FCs suggest that MI and RTX lead to unique brain-signalling patterns and that the combined treatment, potentially through destructive interference mechanisms, most closely resembles controls.


Asunto(s)
Insuficiencia Cardíaca , Proteómica , Animales , Cardiomegalia , Corazón , Insuficiencia Cardíaca/tratamiento farmacológico , Ratones , Ratas , Ratas Sprague-Dawley
11.
Stem Cells ; 27(11): 2734-43, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19591227

RESUMEN

Recent studies showed that mesenchymal stem cells (MSCs) transplantation significantly decreased cardiac fibrosis; however, the mechanisms involved in these effects are still poorly understood. In this work, we investigated whether the antifibrotic properties of MSCs involve the regulation of matrix metalloproteinases (MMPs) and matrix metalloproteinase endogenous inhibitor (TIMP) production by cardiac fibroblasts. In vitro experiments showed that conditioned medium from MSCs decreased viability, alpha-smooth muscle actin expression, and collagen secretion of cardiac fibroblasts. These effects were concomitant with the stimulation of MMP-2/MMP-9 activities and membrane type 1 MMP expression. Experiments performed with fibroblasts from MMP2-knockout mice demonstrated that MMP-2 plays a preponderant role in preventing collagen accumulation upon incubation with conditioned medium from MSCs. We found that MSC-conditioned medium also decreased the expression of TIMP2 in cardiac fibroblasts. In vivo studies showed that intracardiac injection of MSCs in a rat model of postischemic heart failure induced a significant decrease in ventricular fibrosis. This effect was associated with the improvement of morphological and functional cardiac parameters. In conclusion, we showed that MSCs modulate the phenotype of cardiac fibroblasts and their ability to degrade extracellular matrix. These properties of MSCs open new perspectives for understanding the mechanisms of action of MSCs and anticipate their potential therapeutic or side effects.


Asunto(s)
Colagenasas/metabolismo , Fibroblastos/metabolismo , Fibrosis/prevención & control , Células Madre Mesenquimatosas/fisiología , Infarto del Miocardio/patología , Actinas/metabolismo , Animales , Western Blotting , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Colágeno/metabolismo , Medios de Cultivo Condicionados/farmacología , Ecocardiografía , Fibroblastos/efectos de los fármacos , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/patología , Inmunohistoquímica , Metaloproteinasa 1 de la Matriz/genética , Metaloproteinasa 1 de la Matriz/metabolismo , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Noqueados , Infarto del Miocardio/metabolismo , Infarto del Miocardio/terapia , Reacción en Cadena de la Polimerasa , Puntos Cuánticos , Ratas , Inhibidor Tisular de Metaloproteinasa-2/metabolismo
12.
Acta Biomater ; 108: 261-272, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32165191

RESUMEN

For some years now, gadolinium oxysulfide nanoparticles (NPs) appear as strong candidates for very efficient multimodal in vivo imaging by: 1) Magnetic Resonance (MRI), 2) X-ray Computed Tomography (CT) and 3) photoluminescence imaging. In this paper, we present a selection of results centered on the evaluation of physico-chemical stability, toxicity, bio-distribution and excretion mechanisms of Gd2O2S:Ln3+ nanoparticles intravenously injected in rats. Two formulations are here tested with a common matrix and different dopants: Gd2O2S:Eu3+5% and Gd2O2S:Yb3+4%/Tm3+0.1%. The NPs appear to be almost insoluble in pure water and human plasma but corrosion/degradation phenomenon appears in acidic conditions classically encountered in cell lysosomes. Whole body in vivo distribution, excretion and toxicity evaluation revealed a high tolerance of nanoparticles with a long-lasting imaging signal associated with a slow hepatobiliary clearance and very weak urinary excretion. The results show that the majority of the injected product (>60%) has been excreted through the feces after five months. Experiments have evidenced that the NPs mainly accumulate in macrophage-rich organs, that is mainly liver and spleen and to a lesser extent lungs and bones (mainly marrow). No significant amounts have been detected in other organs such as heart, kidneys, brain, intestine and skin. Gd2O2S:Ln3+ NPs appeared to be very well tolerated up to 400 mg/kg when administered intravenously. STATEMENT OF SIGNIFICANCE: Since 2011, we have focused our work on Gd2O2S nanoparticles (NPs) for multimodal bioimaging using fluorescence, Magnetic Resonance Imaging (MRI) and Computed Tomography with very efficient results already published. However, since the European Medicines Agency has concluded its review of gadolinium contrast agents, confirming recommendations to restrict the use of some linear gadolinium agents used in MRI, a particular attention must be paid to any new contrast media containing gadolinium. Therefore, we present in this paper a compilation of studies about toxicity, bio-distribution and excretion mechanisms of Gd2O2S:Ln3+ NPs intravenously injected into rats. We also present an in vitro kinetic study of NPs degradation in aqueous and biological media to provide some information on chemical and biological stability.


Asunto(s)
Gadolinio , Nanopartículas , Animales , Medios de Contraste/toxicidad , Gadolinio/toxicidad , Imagen por Resonancia Magnética , Nanopartículas/toxicidad , Ratas , Distribución Tisular
13.
J Mol Cell Cardiol ; 46(4): 587-95, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19162038

RESUMEN

The potential role of serotonin (5-HT) in cardiac function has generated much interest in recent years. In particular, the need for a tight regulation of 5-HT to maintain normal cardiovascular activity has been demonstrated in different experimental models. However, it remains unclear how increased levels of 5-HT could contribute to the development of cardiac hypertrophy. Availability of 5-HT depends on the mitochondrial enzyme monoamine oxidase A (MAO-A). Therefore, we investigated the consequences of MAO-A deletion on ventricular remodeling in the model of aortic banding in mice. At baseline, MAO-A deletion was associated with an increase in whole blood 5-HT (39.4+/-1.9 microM vs. 24.0+/-0.9 microM in KO and WT mice, respectively). Cardiac 5-HT(2A), but not 5-HT(2B) receptors were overexpressed in MAO-A KO mice, as demonstrated by real-time PCR and Western-blot experiments. After aortic banding, MAO-A KO mice demonstrated greater increase in heart wall thickness, heart to body weight ratios, cardiomyocyte cross-section areas, and myocardial fibrosis compared to WT. Exacerbation of hypertrophy in KO mice was associated with increased amounts of 5-HT in the heart. In order to determine the role of 5-HT and 5-HT(2A) receptors in ventricular remodeling in MAO-A KO mice, we administered the 5-HT(2A) receptor antagonists ketanserin (1 mg/kg/day) or M100907 (0.1 mg/kg/day) during 4 weeks of aortic banding. Chronic administration of these antagonists strongly prevented exacerbation of ventricular hypertrophy in MAO-A KO mice. These results show for the first time that regulation of peripheral 5-HT by MAO-A plays a role in ventricular remodeling via activation of 5-HT(2A) receptors.


Asunto(s)
Cardiomegalia/enzimología , Cardiomegalia/patología , Eliminación de Gen , Ventrículos Cardíacos/patología , Monoaminooxidasa/genética , Presión , Serotonina/metabolismo , Estrés Fisiológico , Animales , Aorta/patología , Presión Sanguínea/efectos de los fármacos , Cardiomegalia/diagnóstico por imagen , Cardiomegalia/fisiopatología , Fibrosis , Fluorobencenos/administración & dosificación , Fluorobencenos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Ventrículos Cardíacos/diagnóstico por imagen , Ventrículos Cardíacos/enzimología , Ventrículos Cardíacos/fisiopatología , Ketanserina/administración & dosificación , Ketanserina/farmacología , Ratones , Ratones Noqueados , Monoaminooxidasa/metabolismo , Miocardio/enzimología , Miocardio/patología , Piperidinas/administración & dosificación , Piperidinas/farmacología , Receptores de Serotonina/genética , Receptores de Serotonina/metabolismo , Serotonina/sangre , Estrés Fisiológico/efectos de los fármacos , Ultrasonografía
14.
Stem Cells ; 26(7): 1749-57, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18467662

RESUMEN

Bone marrow mesenchymal stem cells (MSCs) have shown great potential in cell therapy of solid organs. Approaches to improving the ability of grafted MSCs to survive and secrete paracrine factors represent one of the challenges for the further development of this novel therapy. In the present study, we designed a strategy of ex vivo pretreatment with the pineal hormone melatonin to improve survival, paracrine activity, and efficiency of MSCs. Using a rat model of acute renal failure, we showed that melatonin pretreatment strongly increased survival of MSCs after intraparenchymal injection. This effect was concomitant with overstimulation of angiogenesis, proliferation of renal cells, and accelerated recovery of renal function. To gain insight into the mechanisms involved in the effects observed in vivo, melatonin was tested in vitro on cultured MSCs. Our results show that through stimulation of specific melatonin receptors, melatonin induced an overexpression of the antioxidant enzyme catalase and superoxide dismutase-1 and increased the resistance of MSCs to hydrogen peroxide-dependent apoptosis. Compared with untreated cells, MSCs incubated with melatonin displayed a higher expression of basic fibroblast growth factor and hepatocyte growth factor. In addition, conditioned culture media from melatonin-treated MSCs stimulated tube formation by endothelial progenitor cells and proliferation of proximal tubule cells in culture. In conclusion, our results show that melatonin behaves as a preconditioning agent increasing survival, paracrine activity, and efficiency of MSCs. The use of this molecule for pretreatment of stem cells may represent a novel and safe approach to improving the beneficial effects of cell therapy of solid organs.


Asunto(s)
Células de la Médula Ósea/citología , Supervivencia Celular/efectos de los fármacos , Isquemia/patología , Riñón/efectos de los fármacos , Riñón/metabolismo , Melatonina/metabolismo , Células Madre Mesenquimatosas/citología , Animales , Proliferación Celular , Factor 2 de Crecimiento de Fibroblastos/biosíntesis , Factor de Crecimiento de Hepatocito/biosíntesis , Humanos , Neovascularización Patológica , Ratas , Ratas Endogámicas Lew , Daño por Reperfusión/metabolismo
15.
Circ Res ; 100(1): 41-9, 2007 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-17158340

RESUMEN

The mitochondrial enzyme monoamine oxidase (MAO), its isoform MAO-A, plays a major role in reactive oxygen species-dependent cardiomyocyte apoptosis and postischemic cardiac damage. In the current study, we investigated whether sphingolipid metabolism can account for mediating MAO-A- and reactive oxygen species-dependent cardiomyocyte apoptosis. In H9c2 cardiomyoblasts, MAO-A-dependent reactive oxygen species generation led to mitochondria-mediated apoptosis, along with sphingosine kinase-1 (SphK1) inhibition. These phenomena were associated with generation of proapoptotic ceramide and decrease in prosurvival sphingosine 1-phosphate. These events were mimicked by inhibition of SphK1 with either pharmacological inhibitor or small interfering RNA, as well as by extracellular addition of C(2)-ceramide or H(2)O(2). In contrast, enforced expression of SphK1 protected H9c2 cells from serotonin- or H(2)O(2)-induced apoptosis. Analysis of cardiac tissues from wild-type mice subjected to ischemia/reperfusion revealed significant upregulation of ceramide and inhibition of SphK1. It is noteworthy that SphK1 inhibition, ceramide accumulation, and concomitantly infarct size and cardiomyocyte apoptosis were significantly decreased in MAO-A-deficient animals. In conclusion, we show for the first time that the upregulation of ceramide/sphingosine 1-phosphate ratio is a critical event in MAO-A-mediated cardiac cell apoptosis. In addition, we provide the first evidence linking generation of reactive oxygen species with SphK1 inhibition. Finally, we propose sphingolipid metabolites as key mediators of postischemic/reperfusion cardiac injury.


Asunto(s)
Apoptosis/fisiología , Monoaminooxidasa/metabolismo , Miocitos Cardíacos/fisiología , Estrés Oxidativo/fisiología , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Animales , Apoptosis/efectos de los fármacos , Células Cultivadas , Ceramidas/metabolismo , Ceramidas/farmacología , Regulación hacia Abajo , Resistencia a Medicamentos/fisiología , Peróxido de Hidrógeno/farmacología , Lisofosfolípidos/metabolismo , Ratones , Ratones Noqueados , Mitocondrias Cardíacas/fisiología , Monoaminooxidasa/deficiencia , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/enzimología , Oxidantes/farmacología , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Serotonina/farmacología , Esfingolípidos/metabolismo , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Regulación hacia Arriba
16.
FASEB Bioadv ; 1(4): 227-245, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32123829

RESUMEN

Alarmins and damage-associated molecular patterns (DAMPs) are powerful inflammatory mediators, capable of initiating and maintaining sterile inflammation during acute or chronic tissue injury. Recent evidence suggests that alarmins/DAMPs may also trigger tissue regeneration and repair, suggesting a potential contribution to tissue fibrogenesis. High mobility group B1 (HMGB1), a bona fide alarmin/DAMP, may be released passively by necrotic cells or actively secreted by innate immune cells. Macrophages can release large amounts of HMGB1 and play a key role in wound healing and regeneration processes. Here, we hypothesized that macrophages may be a key source of HMGB1 and thereby contribute to wound healing and fibrogenesis. Surprisingly, cell-specific deletion approaches, demonstrated that macrophage-derived HMGB1 is not involved in tissue fibrogenesis in multiple organs with different underlying pathologies. Compared to control HMGB1Flox mice, mice with macrophage-specific HMGB1 deletion (HMGB1ΔMac) do not display any modification of fibrogenesis in the liver after CCL4 or thioacetamide treatment and bile duct ligation; in the kidney following unilateral ureter obstruction; and in the heart after transverse aortic constriction. Of note, even under thermoneutral housing, known to exacerbate inflammation and fibrosis features, HMGB1ΔMac mice do not show impairment of fibrogenesis. In conclusion, our study clearly establishes that macrophage-derived HMGB1 does not contribute to tissue repair and fibrogenesis.

17.
PLoS One ; 14(12): e0225729, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31815963

RESUMEN

Restricted and controlled drug delivery to the heart remains a challenge giving frequent off-target effects as well as limited retention of drugs in the heart. There is a need to develop and optimize tools to allow for improved design of drug candidates for treatment of heart diseases. Over the last decade, novel drug platforms and nanomaterials were designed to confine bioactive materials to the heart. Yet, the research remains in its infancy, not only in the development of tools but also in the understanding of effects of these materials on cardiac function and tissue integrity. Upconverting nanoparticles are nanomaterials that recently accelerated interest in theranostic nanomedicine technologies. Their unique photophysical properties allow for sensitive in vivo imaging that can be combined with spatio-temporal control for targeted release of encapsulated drugs. Here we synthesized upconverting NaYF4:Yb,Tm nanoparticles and show for the first time their innocuity in the heart, when injected in the myocardium or in the pericardial space in mice. Nanoparticle retention and upconversion in the cardiac region did not alter heart rate variability, nor cardiac function as determined over a 15-day time course ensuing the sole injection. Altogether, our nanoparticles show innocuity primarily in the pericardial region and can be safely used for controlled spatiotemporal drug delivery. Our results support the use of upconverting nanoparticles as potential theranostics tools overcoming some of the key limitations associated with conventional experimental cardiology.


Asunto(s)
Cardiopatías/diagnóstico , Cardiopatías/terapia , Nanopartículas/uso terapéutico , Nanomedicina Teranóstica , Animales , Peso Corporal , Cardiopatías/fisiopatología , Pruebas de Función Cardíaca , Masculino , Ratones Endogámicos C57BL , Nanopartículas/ultraestructura
18.
Methods Mol Biol ; 456: 23-38, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18516550

RESUMEN

The importance and the role of adipose tissues are now largely expanded not only because the very high occurrence of obesity but also because the emerging view that adipose tissue could be a reservoir of therapeutic cells. A critical examination of the adipose tissue features according to their location shows that sampling is not as easy as previously thought and needs special attention to heterogeneity and differences. We discussed here these different points and give precise protocols to sample the different adipose tissues and manipulate them.


Asunto(s)
Tejido Adiposo/anatomía & histología , Tejido Adiposo/cirugía , Distribución de la Grasa Corporal , Técnicas de Laboratorio Clínico , Adipocitos/citología , Tejido Adiposo/metabolismo , Animales , Humanos , Ratones , Obesidad , Ratas
19.
J Am Soc Nephrol ; 18(12): 3110-8, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18003779

RESUMEN

Tubulointerstitial fibrosis in chronic renal disease is strongly associated with progressive loss of renal function. We studied the potential involvement of lysophosphatidic acid (LPA), a growth factor-like phospholipid, and its receptors LPA(1-4) in the development of tubulointerstitial fibrosis (TIF). Renal fibrosis was induced in mice by unilateral ureteral obstruction (UUO) for up to 8 d, and kidney explants were prepared from the distal poles to measure LPA release into conditioned media. After obstruction, the extracellular release of LPA increased approximately 3-fold. Real-time reverse transcription PCR (RT-PCR) analysis demonstrated significant upregulation in the expression of the LPA(1) receptor subtype, downregulation of LPA3, and no change of LPA2 or LPA4. TIF was significantly attenuated in LPA1 (-/-) mice compared to wild-type littermates, as measured by expression of collagen III, alpha-smooth muscle actin (alpha-SMA), and F4/80. Furthermore, treatment of wild-type mice with the LPA1 antagonist Ki16425 similarly reduced fibrosis and significantly attenuated renal expression of the profibrotic cytokines connective tissue growth factor (CTGF) and transforming growth factor beta (TGFbeta). In vitro, LPA induced a rapid, dose-dependent increase in CTGF expression that was inhibited by Ki16425. In conclusion, LPA, likely acting through LPA1, is involved in obstruction-induced TIF. Therefore, the LPA1 receptor might be a pharmaceutical target to treat renal fibrosis.


Asunto(s)
Fibrosis/metabolismo , Enfermedades Renales/patología , Lisofosfolípidos/metabolismo , Receptores del Ácido Lisofosfatídico/metabolismo , Animales , Medios de Cultivo Condicionados/metabolismo , Humanos , Isoxazoles/farmacología , Riñón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosfolípidos/química , Propionatos/farmacología , Factor de Crecimiento Transformador beta/metabolismo , Uréter/embriología
20.
Cardiovasc Res ; 114(1): 123-137, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29136112

RESUMEN

Aims: Tenascin-C (TNC) is an endogenous danger signal molecule strongly associated with inflammatory diseases and with poor outcome in patients with cardiomyopathies. Its function within pathological cardiac tissue during pressure overload remains poorly understood. Methods and results: We showed that TNC accumulates after 1 week of transverse aortic constriction (TAC) in the heart of 12-week-old male mice. By cross bone marrow transplantation experiments, we determined that TNC deposition relied on cardiac cells and not on haematopoietic cells. The expression of TNC induced by TAC, or by administration of a recombinant lentivector coding for TNC, triggered a pro-inflammatory cardiac microenvironment, monocyte/macrophage (MO/MΦ) accumulation, and systolic dysfunction. TNC modified macrophage polarization towards the pro-inflammatory phenotype and stimulated RhoA/Rho-associated protein kinase (ROCK) pathways to promote mesenchymal to amoeboid transition that enhanced macrophage migration into fibrillar collagen matrices. The amplification of inflammation and MO/MΦ recruitment by TNC was abrogated by genetic invalidation of TNC in knockout mice. These mice showed less ventricular remodelling and an improved cardiac function after TAC as compared with wild-type mice. Conclusions: By promoting a pro-inflammatory microenvironment and macrophage migration, TNC appears to be a key factor to enable the MO/MΦ accumulation within fibrotic hearts leading to cardiac dysfunction. As TNC is highly expressed during inflammation and sparsely during the steady state, its inhibition could be a promising therapeutic strategy to control inflammation and immune cell infiltration in heart disease.


Asunto(s)
Movimiento Celular , Hipertrofia Ventricular Izquierda/metabolismo , Macrófagos/metabolismo , Monocitos/metabolismo , Miocardio/metabolismo , Tenascina/metabolismo , Disfunción Ventricular Izquierda/metabolismo , Función Ventricular Izquierda , Remodelación Ventricular , Animales , Microambiente Celular , Quimiocinas/metabolismo , Modelos Animales de Enfermedad , Fibrosis , Hipertrofia Ventricular Izquierda/genética , Hipertrofia Ventricular Izquierda/patología , Hipertrofia Ventricular Izquierda/fisiopatología , Mediadores de Inflamación/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Miocardio/patología , Fenotipo , Transducción de Señal , Tenascina/genética , Disfunción Ventricular Izquierda/genética , Disfunción Ventricular Izquierda/patología , Disfunción Ventricular Izquierda/fisiopatología , Proteínas de Unión al GTP rho/metabolismo , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda