Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 215
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
BMC Biol ; 19(1): 124, 2021 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-34134693

RESUMEN

BACKGROUND: Doxorubicin (Dox) is an anti-cancer anthracycline drug that causes double-stranded DNA breaks. It is highly effective against several types of tumours; however, it also has adverse effects on regenerative populations of normal cells, such as human cardiac mesenchymal progenitor cells (hCmPCs), and its clinical use is limited by cardiotoxicity. Another known effect of Dox is nucleolar disruption, which triggers the ubiquitously expressed nucleolar phosphoprotein Nucleophosmin (NPM) to be released from the nucleolus into the cell, where it participates in the orchestration of cellular stress responses. NPM has also been observed in the extracellular space in response to different stress stimuli; however, the mechanism behind this and its functional implications are as yet largely unexplored. The aim of this study was to establish whether Dox could elicit NPM secretion in the extracellular space and to elucidate the mechanism of secretion and the effect of extracellular NPM on hCmPCs. RESULTS: We found that following the double-strand break formation in hCmPCs caused by Dox, NPM was rapidly secreted in the extracellular space by an active mechanism, in the absence of either apoptosis or necrosis. Extracellular release of NPM was similarly seen in response to ultraviolet radiation (UV). Furthermore, we observed an increase of NPM levels in the plasma of Dox-treated mice; thus, NPM release also occurred in vivo. The treatment of hCmPCs with extracellular recombinant NPM induced a decrease of cell proliferation and a response mediated through the Toll-like receptor (TLR)4. We demonstrated that NPM binds to TLR4, and via TLR4, and nuclear factor kappa B (NFkB) activation/nuclear translocation, exerts proinflammatory functions by inducing IL-6 and COX-2 gene expression. Finally, we found that in hCmPCs, NPM secretion could be driven by an autophagy-dependent unconventional mechanism that requires TLR4, since TLR4 inhibition dramatically reduced Dox-induced secretion. CONCLUSIONS: We hypothesise that the extracellular release of NPM could be a general response to DNA damage since it can be elicited by either a chemical agent such as Dox or a physical genotoxic stressor such as UV radiation. Following genotoxic stress, NPM acts similarly to an alarmin in hCmPCs, being rapidly secreted and promoting cell cycle arrest and a TLR4/NFκB-dependent inflammatory response.


Asunto(s)
Células Madre Mesenquimatosas , Alarminas , Animales , Apoptosis , Comunicación Autocrina , Doxorrubicina/efectos adversos , Corazón , Humanos , Ratones , FN-kappa B , Proteínas Nucleares/genética , Nucleofosmina , Comunicación Paracrina , Receptor Toll-Like 4/genética , Rayos Ultravioleta
2.
Cell Mol Life Sci ; 76(2): 211-229, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30306212

RESUMEN

High mobility group box 1 (HMGB1) is a ubiquitous nuclear protein involved in transcription regulation, DNA replication and repair and nucleosome assembly. HMGB1 is passively released by necrotic tissues or actively secreted by stressed cells. Extracellular HMGB1 acts as a damage-associated molecular pattern (DAMPs) molecule and gives rise to several redox forms that by binding to different receptors and interactors promote a variety of cellular responses, including tissue inflammation or regeneration. Inhibition of extracellular HMGB1 in experimental models of myocardial ischemia/reperfusion injury, myocarditis, cardiomyopathies induced by mechanical stress, diabetes, bacterial infection or chemotherapeutic drugs reduces inflammation and is protective. In contrast, administration of HMGB1 after myocardial infarction induced by permanent coronary artery ligation ameliorates cardiac performance by promoting tissue regeneration. HMGB1 decreases contractility and induces hypertrophy and apoptosis in cardiomyocytes, stimulates cardiac fibroblast activities, and promotes cardiac stem cell proliferation and differentiation. Interestingly, maintenance of appropriate nuclear HMGB1 levels protects cardiomyocytes from apoptosis by preventing DNA oxidative stress, and mice with HMGB1cardiomyocyte-specific overexpression are partially protected from cardiac damage. Finally, higher levels of circulating HMGB1 are associated to human heart diseases. Hence, during cardiac injury, HMGB1 elicits both harmful and beneficial responses that may in part depend on the generation and stability of the diverse redox forms, whose specific functions in this context remain mostly unexplored. This review summarizes recent findings on HMGB1 biology and heart dysfunctions and discusses the therapeutic potential of modulating its expression, localization, and oxidative-dependent activities.


Asunto(s)
Proteína HMGB1/metabolismo , Cardiopatías/patología , Alarminas/metabolismo , Animales , Biomarcadores/metabolismo , Cardiopatías/metabolismo , Humanos , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Miocarditis/metabolismo , Miocarditis/patología , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo
3.
Arterioscler Thromb Vasc Biol ; 38(9): 2079-2090, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30026277

RESUMEN

Objective- Vascular calcification (VC) is age dependent and a risk factor for cardiovascular and all-cause mortality. VC involves the senescence-induced transdifferentiation of vascular smooth muscle cells (SMCs) toward an osteochondrogenic lineage resulting in arterial wall mineralization. miR-34a increases with age in aortas and induces vascular SMC senescence through the modulation of its target SIRT1 (sirtuin 1). In this study, we aimed to investigate whether miR-34a regulates VC. Approach and Results- We found that miR-34a and Runx2 (Runt-related transcription factor 2) expression correlates in young and old mice. Mir34a+/+ and Mir34a-/- mice were treated with vitamin D, and calcium quantification revealed that Mir34a deficiency reduces soft tissue and aorta medial calcification and the upregulation of the VC Sox9 (SRY [sex-determining region Y]-box 9) and Runx2 and the senescence p16 and p21 markers. In this model, miR-34a upregulation was transient and preceded aorta mineralization. Mir34a-/- SMCs were less prone to undergo senescence and under osteogenic conditions deposited less calcium compared with Mir34a+/+ cells. Furthermore, unlike in Mir34a+/+ SMC, the known VC inhibitors SIRT1 and Axl (AXL receptor tyrosine kinase) were only partially downregulated in calcifying Mir34a-/- SMC. Strikingly, constitutive miR-34a overexpression to senescence-like levels in human aortic SMCs increased calcium deposition and enhanced Axl and SIRT1 decrease during calcification. Notably, we also showed that miR-34a directly decreased Axl expression in human aortic SMC, and restoration of its levels partially rescued miR-34a-dependent growth arrest. Conclusions- miR-34a promotes VC via vascular SMC mineralization by inhibiting cell proliferation and inducing senescence through direct Axl and SIRT1 downregulation, respectively. This miRNA could be a good therapeutic target for the treatment of VC.


Asunto(s)
Senescencia Celular/fisiología , MicroARNs/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Sirtuina 1/metabolismo , Calcificación Vascular , Adulto , Envejecimiento/patología , Animales , Aorta/metabolismo , Proliferación Celular , Células Cultivadas , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Regulación hacia Abajo , Humanos , Masculino , Ratones , Ratones Noqueados , Músculo Liso Vascular/citología , Factor de Transcripción SOX9/metabolismo , Regulación hacia Arriba , Adulto Joven , Tirosina Quinasa del Receptor Axl
4.
J Cell Sci ; 129(5): 1003-17, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26795563

RESUMEN

The role of Ras in human skin tumorigenesis induction is still ambiguous. Overexpression of oncogenic Ras causes premature senescence in cultured human cells and hyperplasia in transgenic mice. Here, we investigated whether the oncogenic insult outcome might depend on the nature of the founding keratinocyte. We demonstrate that overexpression of the constitutively active Ras-V12 induces senescence in primary human keratinocyte cultures, but that some cells escape senescence and proliferate indefinitely. Ras overexpression in transient-amplifying- or stem-cell-enriched cultures shows that p16 (encoded by CDKN2A) levels are crucial for the final result. Indeed, transient-amplifying keratinocytes expressing high levels of p16 are sensitive to Ras-V12-induced senescence, whereas cells with high proliferative potential, but that do not display p16, are resistant. The subpopulation that sustains the indefinite culture growth exhibits stem cell features. Bypass of senescence correlates with inhibition of the pRb (also known as RB1) pathway and resumption of telomerase reverse transcriptase (TERT) activity. Immortalization is also sustained by activation of the ERK1 and ERK2 (ERK1/2, also known as MAPK3 and MAPK1) and Akt pathways. Moreover, only transduced cultures originating from cultures bearing stem cells induce tumors in nude mice. Our findings demonstrate that the Ras overexpression outcome depends on the clonogenic potential of the recipient keratinocyte and that only the stem cell compartment is competent to initiate tumorigenesis.


Asunto(s)
Queratinocitos/enzimología , Proteínas Proto-Oncogénicas p21(ras)/genética , Neoplasias Cutáneas/genética , Animales , Proliferación Celular , Transformación Celular Neoplásica/genética , Senescencia Celular , Técnicas de Cocultivo , Humanos , Ratones , Ratones Desnudos , Células 3T3 NIH , Trasplante de Neoplasias , Células Madre Neoplásicas/fisiología , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Transducción de Señal , Neoplasias Cutáneas/patología
5.
Clin Sci (Lond) ; 132(22): 2423-2436, 2018 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-30389857

RESUMEN

Early recognition of vulnerable carotid plaques could help in identifying patients at high stroke risk, who may benefit from earlier revascularisation. Nowadays, different biomarkers of plaque instability have been unravelled, among these miRNAs are promising tools for the diagnosis and treatment of atherosclerosis. Inflammation, reactive oxygen species (ROS) and endothelial dysfunction play a key role in unstable plaques genesis. We showed that miR-200c induces endothelial dysfunction, ROS production and a positive mechanism among miR-200c and miR-33a/b, two miRNAs involved in atherosclerosis progression. The goal of the present study was to determine whether miR-200c could be an atherosclerosis biomarker. Carotid plaques of patients that underwent carotid endarterectomy (CEA) were assayed for miR-200c expression. miR-200c was up-regulated in carotid plaques (n=22) and its expression was higher in unstable (n=12) compared with stable (n=10) plaques. miR-200c positively correlated with instability biomarkers (i.e. monocyte chemoattractant protein-1, cicloxigenase-2 (COX2), interleukin 6 (IL6), metalloproteinase (MMP) 1 (MMP1), 9 (MMP9)) and miR-33a/b. Moreover, miR-200c negatively correlated with stability biomarkers (i.e. zinc finger E-box binding homoeobox 1 (ZEB1), endothelial nitric oxide (NO) synthase (eNOS), forkhead boxO1 (FOXO1) and Sirtuin1 (SIRT1)) (stable plaques = 15, unstable plaques = 15). Circulating miR-200c was up-regulated before CEA in 24 patients, correlated with miR-33a/b and decreased 1 day after CEA. Interestingly, 1 month after CEA, circulating miR-200c is low in patients with stable plaques (n=11) and increased to control levels, in patients with unstable plaques (n=13). Further studies are needed to establish whether miR-200c represents a circulating biomarker of plaque instability. Our results show that miR-200c is an atherosclerotic plaque progression biomarker and suggest that it may be clinically useful to identify patients at high embolic risk.


Asunto(s)
Arterias Carótidas/patología , Estenosis Carotídea/genética , MicroARNs/genética , Placa Aterosclerótica , Anciano , Arterias Carótidas/diagnóstico por imagen , Arterias Carótidas/cirugía , Estenosis Carotídea/diagnóstico por imagen , Estenosis Carotídea/patología , Estenosis Carotídea/cirugía , Endarterectomía Carotidea , Femenino , Regulación de la Expresión Génica , Marcadores Genéticos , Humanos , Masculino , MicroARNs/sangre , Valor Predictivo de las Pruebas , Medición de Riesgo , Factores de Riesgo , Rotura Espontánea , Factores de Tiempo , Resultado del Tratamiento , Ultrasonografía
6.
Biochim Biophys Acta Mol Basis Dis ; 1863(11): 2693-2704, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28716707

RESUMEN

Myocardial infarction (MI) is a major health burden worldwide. Extracellular High mobility group box 1 (HMGB1) regulates tissue healing after injuries. The reduced form of HMGB1 (fr-HMGB1) exerts chemotactic activity by binding CXCL12 through CXCR4, while the disulfide form, (ds-HMGB1), induces cytokines expression by TLR4. Here, we assessed the role of HMGB1 redox forms and the non-oxidizable mutant (3S) on human cardiac fibroblast (hcFbs) functions and cardiac remodeling after infarction. Among HMGB1 receptors, hcFbs express CXCR4. Fr-HMGB1 and 3S, but not ds-HMGB1, promote hcFbs migration through Src activation, while none of HMGB1 redox forms induces proliferation or inflammatory mediators. 3S is more effective than fr-HMGB1 in stimulating hcFbs migration and Src phosphorylation being active at lower concentrations and in oxidizing conditions. Notably, chemotaxis toward both proteins is CXCR4-dependent but, in contrast to fr-HMGB1, 3S does not require CXCL12 since hcFbs migration persists in the presence of the CXCL12/CXCR4 inhibitor AMD3100 or an anti-CXCL12 antibody. Interestingly, 3S interacts with CXCR4 and induces a different receptor conformation than CXCL12. Mice undergoing MI and receiving 3S exhibit adverse LV remodeling owing to an excessive collagen deposition promoted by a higher number of myofibroblasts. On the contrary, fr-HMGB1 ameliorates cardiac performance enhancing neoangiogenesis and reducing the infarcted area and fibrosis. Altogether, our results demonstrate that non-oxidizable HMGB1 induce a sustained cardiac fibroblasts migration despite the redox state of the environment and by altering CXCL12/CXCR4 axis. This affects proper cardiac remodeling after an infarction.


Asunto(s)
Movimiento Celular , Quimiocina CXCL12/metabolismo , Fibroblastos/metabolismo , Proteína HMGB1/metabolismo , Infarto del Miocardio/metabolismo , Miocardio/metabolismo , Receptores CXCR4/metabolismo , Femenino , Fibroblastos/patología , Humanos , Masculino , Infarto del Miocardio/patología , Miocardio/patología , Oxidación-Reducción
7.
Clin Sci (Lond) ; 131(18): 2397-2408, 2017 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-28811385

RESUMEN

Hypercholesterolaemia provokes reactive oxygen species (ROS) increase and is a major risk factor for cardiovascular disease (CVD) development. We previously showed that circulating miR-33a/b expression levels were up-regulated in children with familial hypercholesterolaemia (FH). miR-33a/b control cholesterol homoeostasis and recently miR-33b has been demonstrated to directly target the transcription factor zinc finger E-box-binding homeobox 1 (ZEB1). The latter acts in a negative feedback loop with the miR-200 family. Our previous studies showed that the ROS-dependent miR-200c up-regulation induces endothelial dysfunction and provokes a ZEB1-dependent apoptosis and senescence. In the present study, we aimed to verify whether circulating miR-200c was induced in FH children, and whether a correlation existed with miR-33a/b Total RNA was extracted from plasma of 28 FH children and 25 age-matched healthy subjects (HS) and miR-200c levels were measured. We found that miR-200c was up-regulated in FH compared with HS (4.00 ± 0.48-fold increase, P<0.05) and exhibited a positive correlation with miR-33a/b. miR-200c did not correlate with plasma lipids, but correlated with C-reactive protein (CRP) plasma levels and glycaemia (GLI). Ordinary least squares (OLS) regression analysis revealed that miR-200c was significantly affected by GLI and by miR-33a (P<0.01; P<0.001 respectively). Moreover, we found that miR-33 overexpression, in different cell lines, decreased ZEB1 expression and up-regulated both the intracellular and the extracellular miR-200c expression levels. In conclusion, circulating miR-200c is up-regulated in FH, probably due to oxidative stress and inflammation and via a miR-33a/b-ZEB1-dependent mechanism. The present study could provide the first evidence to point to the use of miR-33a/b and miR-200c, as early biomarkers of CVD, in paediatric FH.


Asunto(s)
Hiperlipoproteinemia Tipo II/metabolismo , MicroARNs/metabolismo , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/fisiología , Adolescente , Glucemia/análisis , Proteína C-Reactiva/análisis , Estudios de Casos y Controles , Niño , Preescolar , Femenino , Humanos , Hiperlipoproteinemia Tipo II/genética , Masculino , MicroARNs/sangre , Especies Reactivas de Oxígeno/metabolismo , Regulación hacia Arriba
8.
Aesthet Surg J ; 37(5): 591-602, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28052909

RESUMEN

Background: Fat grafts enriched with cells of the stromal vascular fraction (SVF), especially adipose-derived stromal cells (ASCs), exhibit significantly improved retention over non enriched, plain fat. Different types of liposuction cannulae may yield lipoaspirates with different subpopulations of cells. Moreover, preparation of adipose tissue for transplantation typically involves centrifugation, which creates a density gradient of fat. Objectives: The authors sought to determine whether liposuction with a barbed or smooth cannula altered the enrichment of the SVF, and specifically ASCs, in low-density (LD) and high-density (HD) fractions of centrifuged adipose tissue. Methods: Fat was harvested from 2 abdominal sites of 5 healthy women with a barbed or smooth multihole blunt-end cannula. After centrifugation, LD and HD fat fractions were digested with collagenase and analyzed by polychromatic flow cytometry to identify and enumerate distinct populations of cells. Results: Overall cell yield and the number of immune cells were consistently higher in HD fractions than in LD fractions, regardless of the cannula employed. More living cells, and specifically more ASCs, populated the HD fractions of lipoaspirates obtained with a barbed cannula than with a smooth cannula. Conclusions: In this study, lipoaspiration with a barbed cannula and isolation of the HD layer of centrifuged adipose tissue yielded maximal amounts of SVF cells, including ASCs.


Asunto(s)
Tejido Adiposo/citología , Tejido Adiposo/trasplante , Separación Celular/métodos , Lipectomía/instrumentación , Recolección de Tejidos y Órganos/instrumentación , Trasplantes/citología , Adulto , Cánula , Centrifugación , Femenino , Citometría de Flujo/métodos , Humanos , Lipectomía/métodos , Persona de Mediana Edad , Células del Estroma/trasplante , Recolección de Tejidos y Órganos/métodos
9.
Cell Mol Life Sci ; 72(9): 1725-40, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25575564

RESUMEN

Cardiovascular disease is the leading cause of morbidity and mortality in the developed world. Although ongoing therapeutic strategies ameliorate symptoms and prolong life for patients with cardiovascular diseases, they do not solve the critical issue related to the loss of cardiac tissue. Accordingly, stem/progenitor cell therapy has emerged as a paramount approach for cardiac repair and regeneration. In this regard, c-kit(+) cells have animated much interest and controversy. These cells are self-renewing, clonogenic, and multipotent and display a noteworthy potential to differentiate into all cardiovascular lineages. However, their functional contribution to cardiomyocyte turnover is one of the centrally debated issues concerning their regenerative potential. Regardless, plentiful preclinical and clinical studies have been conducted which provide evidence for the capacity of c-kit(+) cells to improve cardiac function. The purpose of this review is to give a comprehensive, impartial, critical description and evaluation of the literature on c-kit(+) cells from bench to bedside in order to address their true potential, benefits and controversies.


Asunto(s)
Enfermedades Cardiovasculares/terapia , Corazón/fisiología , Miocitos Cardíacos/citología , Proteínas Proto-Oncogénicas c-kit/metabolismo , Regeneración , Células Madre/citología , Animales , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/fisiopatología , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Corazón/fisiopatología , Humanos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Proteínas Proto-Oncogénicas c-kit/análisis , Células Madre/metabolismo , Células Madre/patología
10.
J Mol Cell Cardiol ; 83: 21-31, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25724723

RESUMEN

DNA and histone methylation are well characterized epigenetic marks that are altered during the aging process. In aged cells and tissues, DNA cytosine tagging by methylation undergoes the so-called "epigenetic drift", in parallel with a change in the methylated histone profile. Despite the large body of knowledge regarding age-dependent epigenetic changes, there are few reports related to this topic in the cardiovascular field. This review summarizes age-dependent changes in DNA and histone methylation with a specific focus on age-related cardiovascular diseases (CVDs).


Asunto(s)
Envejecimiento/metabolismo , Arritmias Cardíacas/metabolismo , Aterosclerosis/metabolismo , Cromatina/metabolismo , Epigénesis Genética , Insuficiencia Cardíaca/metabolismo , Envejecimiento/genética , Animales , Arritmias Cardíacas/genética , Arritmias Cardíacas/patología , Aterosclerosis/genética , Aterosclerosis/patología , Cromatina/química , Metilación de ADN , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/patología , Histonas/genética , Histonas/metabolismo , Humanos , Miocardio/metabolismo , Miocardio/patología , Proteínas del Grupo Polycomb/genética , Proteínas del Grupo Polycomb/metabolismo , Transducción de Señal
11.
J Mol Cell Cardiol ; 81: 62-70, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25640160

RESUMEN

Age-associated cardiovascular diseases are at least partially ascribable to vascular cell senescence. Replicative senescence (RS) and stress-induced premature senescence (SIPS) are provoked respectively by endogenous (telomere erosion) and exogenous (H2O2, UV) stimuli resulting in cell cycle arrest in G1 and G2 phases. In both scenarios, mitochondria-derived ROS are important players in senescence initiation. We aimed to define whether a mtDNA-transcribed long-non-coding-RNA (lncRNA), ASncmtRNA-2, has a role in vascular aging and senescence. Aortas of old mice, characterized by increased senescence, showed an increment in ASncmtRNA-2 expression. In vitro analysis of Endothelial Cells (EC) and Vascular Smooth Muscle Cells (VSMC) established that ASncmtRNA-2 is induced in EC, but not in VSMC, during RS. Surprisingly, ASncmtRNA-2 is not upregulated in two different EC SIPS scenarios, treated with H2O2 and UV. The p16 gene displayed similar ASncmtRNA-2 expression patterns, suggesting a possible co-regulation of the two genes. Interestingly, the expression of two miRNAs, hsa-miR-4485 and hsa-miR-1973, with perfect homology to the double strand region of ASncmtRNA-2 and originating at least in part from a mitochondrial transcript, was induced in RS, opening to the possibility that this lncRNA functions as a non-canonical precursor of these miRNAs. Cell cycle analysis of EC transiently over-expressing ASncmtRNA-2 revealed an accumulation of EC in the G2/M phase, but not in the G1 phase. We propose that ASncmtRNA-2 in EC might be involved in the RS establishment by participating in the cell cycle arrest in G2/M phase, possibly through the production of hsa-miR-4485 and hsa-miR-1973. This article is part of a Special Issue entitled: Mitochondria.


Asunto(s)
Envejecimiento/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Mitocondrias/metabolismo , Miocitos del Músculo Liso/metabolismo , ARN Largo no Codificante/genética , ARN/genética , Envejecimiento/genética , Animales , Aorta/citología , Aorta/metabolismo , Secuencia de Bases , Senescencia Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de la radiación , Regulación de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/efectos de la radiación , Humanos , Peróxido de Hidrógeno/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , MicroARNs/metabolismo , Mitocondrias/genética , Datos de Secuencia Molecular , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/efectos de la radiación , ARN/metabolismo , ARN Largo no Codificante/metabolismo , ARN Mitocondrial , Transducción de Señal , Rayos Ultravioleta
12.
J Mol Cell Cardiol ; 87: 54-64, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26264759

RESUMEN

Communication between cardiomyocytes depends upon gap junctions (GJ). Previous studies have demonstrated that electrical stimulation induces GJ remodeling and modifies histone acetylase (HAT) and deacetylase (HDAC) activities, although these two results have not been linked. The aim of this work was to establish whether electrical stimulation modulates GJ-mediated cardiac cell-cell communication by acetylation-dependent mechanisms. Field stimulation of HL-1 cardiomyocytes at 0.5 Hz for 24 h significantly reduced connexin43 (Cx43) expression and cell-cell communication. HDAC activity was down-regulated whereas HAT activity was not modified resulting in increased acetylation of Cx43. Consistent with a post-translational mechanism, we did not observe a reduction in Cx43 mRNA in electrically stimulated cells, while the proteasomal inhibitor MG132 maintained Cx43 expression. Further, the treatment of paced cells with the HAT inhibitor Anacardic Acid maintained both the levels of Cx43 and cell-cell communication. Finally, we observed increased acetylation of Cx43 in the left ventricles of dogs subjected to chronic tachypacing as a model of abnormal ventricular activation. In conclusion, our findings suggest that altered electrical activity can regulate cardiomyocyte communication by influencing the acetylation status of Cx43.


Asunto(s)
Comunicación Celular/genética , Conexina 43/biosíntesis , Uniones Comunicantes/genética , Ventrículos Cardíacos/metabolismo , Miocitos Cardíacos/metabolismo , Acetilación/efectos de los fármacos , Ácidos Anacárdicos/administración & dosificación , Animales , Conexina 43/genética , Perros , Estimulación Eléctrica , Uniones Comunicantes/patología , Ventrículos Cardíacos/patología , Histona Acetiltransferasas/antagonistas & inhibidores , Histona Acetiltransferasas/metabolismo , Histona Desacetilasa 1/antagonistas & inhibidores , Histona Desacetilasa 1/metabolismo , Humanos , Ratones , Miocitos Cardíacos/patología , ARN Mensajero/biosíntesis
13.
Am Heart J ; 170(4): 652-658.e7, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26386788

RESUMEN

BACKGROUND: Granulocyte-colony stimulating factor (G-CSF) has been clinically tested in ST-elevation myocardial infarction (STEMI) with mixed results. Our 3-year follow-up data from STEM-AMI trial documented a sustained benefit of G-CSF on adverse ventricular remodeling after large anterior STEMI, when administered early and at a high-dose in patients with left ventricular (LV) dysfunction. The Aim of the present trial is to establish whether G-CSF improves hard clinical long-term outcomes. METHODS: The STEM-AMI OUTCOME is a prospective, multicenter, randomized, open-label, phase III trial. It will include 1,530 patients with anterior STEMI undergoing primary percutaneous coronary intervention 2 to 24 hours after symptoms onset and with LV ejection fraction ≤45% after successful reperfusion. Patients will be randomized 1:1 to G-CSF and/or standard treatment. The primary end point is a reduced occurrence of all-cause death, recurrence of myocardial infarction, or hospitalization due to heart failure in G-CSF-treated patients. Left ventricular remodeling will be assessed via cardiac ultrasound and a substudy with cardiac magnetic resonance will be carried out in 120 subjects. Accrual and follow-up periods will last 3 and 2 years, respectively. CONCLUSIONS: The STEM-AMI OUTCOME study is designed to be a rigorous controlled phase III trial with adequate statistical power to conclusively assess efficacy of G-CSF treatment in STEMI.


Asunto(s)
Electrocardiografía , Factor Estimulante de Colonias de Granulocitos/administración & dosificación , Infarto del Miocardio/terapia , Intervención Coronaria Percutánea/métodos , Anciano , Angiografía Coronaria , Femenino , Estudios de Seguimiento , Humanos , Inyecciones Intraarteriales , Imagen por Resonancia Cinemagnética , Masculino , Persona de Mediana Edad , Infarto del Miocardio/diagnóstico , Infarto del Miocardio/fisiopatología , Estudios Prospectivos , Factores de Tiempo , Resultado del Tratamiento , Función Ventricular Izquierda , Remodelación Ventricular
14.
Cytotherapy ; 17(9): 1302-13, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26038175

RESUMEN

BACKGROUND AIMS: The Pall Celeris system is a filtration-based point-of-care device designed to obtain a high concentrate of peripheral blood total nucleated cells (PB-TNCs). We have characterized the Pall Celeris-derived TNCs for their in vitro and in vivo angiogenic potency. METHODS: PB-TNCs isolated from healthy donors were characterized through the use of flow cytometry and functional assays, aiming to assess migratory capacity, ability to form capillary-like structures, endothelial trans-differentiation and paracrine factor secretion. In a hind limb ischemia mouse model, we evaluated perfusion immediately and 7 days after surgery, along with capillary, arteriole and regenerative fiber density and local bio-distribution. RESULTS: Human PB-TNCs isolated by use of the Pall Celeris filtration system were shown to secrete a panel of angiogenic factors and migrate in response to vascular endothelial growth factor and stromal-derived factor-1 stimuli. Moreover, after injection in a mouse model of hind limb ischemia, PB-TNCs induced neovascularization by increasing capillary, arteriole and regenerative fiber numbers, with human cells detected in murine tissue up to 7 days after ischemia. CONCLUSIONS: The Pall Celeris system may represent a novel, effective and reliable point-of-care device to obtain a PB-derived cell product with adequate potency for therapeutic angiogenesis.


Asunto(s)
Isquemia/terapia , Neovascularización Fisiológica , Enfermedad Arterial Periférica/terapia , Sistemas de Atención de Punto , Animales , Eliminación de Componentes Sanguíneos , Diferenciación Celular , Movimiento Celular , Separación Celular/métodos , Quimiocina CXCL12/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/citología , Filtración , Citometría de Flujo , Miembro Posterior/irrigación sanguínea , Humanos , Leucocitos/inmunología , Ratones , Reperfusión , Factor A de Crecimiento Endotelial Vascular/metabolismo
15.
J Biol Chem ; 288(16): 11004-12, 2013 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-23463510

RESUMEN

In a mouse model of skin repair we found that the class I-IIa histone deacetylase inhibitor trichostatin A accelerated tissue regeneration. Unexpectedly, this effect was suppressed by Sirtinol, a class III histone deacetylase (HDAC) (sirtuin)-selective inhibitor. The role of sirtuins (SIRTs) was then investigated by using resveratrol and a novel SIRT1-2-3 activator, the MC2562 compound we synthesized recently. Both resveratrol and MC2562 were effective in accelerating wound repair. The local administration of natural or synthetic SIRT activators, in fact, significantly accelerated skin regeneration by increasing keratinocyte proliferation. In vitro experiments revealed that the activation of SIRTs stimulated keratinocyte proliferation via endothelial NO synthase phosphorylation and NO production. In this condition, the class I member HDAC2 was found S-nitrosylated on cysteine, a post-transduction modification associated with loss of activity and DNA binding capacity. After deacetylase inhibitor or SIRT activator treatment, ChIP showed, in fact, a significant HDAC2 detachment from the promoter region of insulin growth factor I (IGF-I), fibroblast growth factor 10 (FGF-10), and Epithelial Growth Factor (EGF), which may be the final recipients and effectors of the SIRT-NO-HDAC signaling cascade. Consistently, the effect of SIRT activators was reduced in the presence of NG-nitro-L-arginine methyl ester (L-NAME), a general inhibitor of NO synthesis. In conclusion, the NO-dependent cross-talk among class III and I histone deacetylases suggests an unprecedented signaling pathway important for skin repair.


Asunto(s)
Histona Desacetilasas del Grupo III/metabolismo , Histona Desacetilasa 2/metabolismo , Óxido Nítrico/metabolismo , Piel/enzimología , Piel/lesiones , Cicatrización de Heridas/fisiología , Animales , Línea Celular Transformada , Activadores de Enzimas/farmacología , Inhibidores Enzimáticos/farmacología , Factor 10 de Crecimiento de Fibroblastos/metabolismo , Histona Desacetilasas del Grupo III/antagonistas & inhibidores , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Masculino , Ratones , NG-Nitroarginina Metil Éster/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Cicatrización de Heridas/efectos de los fármacos
16.
J Biol Chem ; 288(32): 22915-29, 2013 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-23836913

RESUMEN

Histone deacetylase inhibitors (DIs) are promising drugs for the treatment of several pathologies including ischemic and failing heart where they demonstrated efficacy. However, adverse side effects and cardiotoxicity have also been reported. Remarkably, no information is available about the effect of DIs during tissue regeneration following acute peripheral ischemia. In this study, mice made ischemic by femoral artery excision were injected with the DIs MS275 and MC1568, selective for class I and IIa histone deacetylases (HDACs), respectively. In untreated mice, soon after damage, class IIa HDAC phosphorylation and nuclear export occurred, paralleled by dystrophin and neuronal nitric-oxide synthase (nNOS) down-regulation and decreased protein phosphatase 2A activity. Between 14 and 21 days after ischemia, dystrophin and nNOS levels recovered, and class IIa HDACs relocalized to the nucleus. In this condition, the MC1568 compound increased the number of newly formed muscle fibers but delayed their terminal differentiation, whereas MS275 abolished the early onset of the regeneration process determining atrophy and fibrosis. The selective DIs had differential effects on the vascular compartment: MC1568 increased arteriogenesis whereas MS275 inhibited it. Capillarogenesis did not change. Chromatin immunoprecipitations revealed that class IIa HDAC complexes bind promoters of proliferation-associated genes and of class I HDAC1 and 2, highlighting a hierarchical control between class II and I HDACs during tissue regeneration. Our findings indicate that class-selective DIs interfere with normal mouse ischemic hindlimb regeneration and suggest that their use could be limited by alteration of the regeneration process in peripheral ischemic tissues.


Asunto(s)
Benzamidas/efectos adversos , Miembro Posterior/irrigación sanguínea , Inhibidores de Histona Desacetilasas/efectos adversos , Ácidos Hidroxámicos/efectos adversos , Isquemia , Músculo Esquelético , Piridinas/efectos adversos , Pirroles/efectos adversos , Regeneración/efectos de los fármacos , Animales , Benzamidas/farmacología , Distrofina/metabolismo , Miembro Posterior/metabolismo , Miembro Posterior/patología , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/metabolismo , Ácidos Hidroxámicos/farmacología , Isquemia/tratamiento farmacológico , Isquemia/metabolismo , Isquemia/patología , Masculino , Ratones , Músculo Esquelético/irrigación sanguínea , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Óxido Nítrico Sintasa de Tipo I/metabolismo , Proteína Fosfatasa 2/metabolismo , Piridinas/farmacología , Pirroles/farmacología , Factores de Tiempo
17.
RNA ; 18(3): 472-84, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22282338

RESUMEN

In order to understand the role of microRNAs (miRNAs) in vascular physiopathology, we took advantage of deep-sequencing techniques to accurately and comprehensively profile the entire miRNA population expressed by endothelial cells exposed to hypoxia. SOLiD sequencing of small RNAs derived from human umbilical vein endothelial cells (HUVECs) exposed to 1% O2 or normoxia for 24 h yielded more than 22 million reads per library. A customized bioinformatic pipeline identified more than 400 annotated microRNA/microRNA* species with a broad abundance range: miR-21 and miR-126 totaled almost 40% of all miRNAs. A complex repertoire of isomiRs was found, displaying also 5' variations, potentially affecting target recognition. High-stringency bioinformatic analysis identified microRNA candidates, whose predicted pre-miRNAs folded into a stable hairpin. Validation of a subset by qPCR identified 18 high-confidence novel miRNAs as detectable in independent HUVEC cultures and associated to the RISC complex. The expression of two novel miRNAs was significantly down-modulated by hypoxia, while miR-210 was significantly induced. Gene ontology analysis of their predicted targets revealed a significant association to hypoxia-inducible factor signaling, cardiovascular diseases, and cancer. Overexpression of the novel miRNAs in hypoxic endothelial cells affected cell growth and confirmed the biological relevance of their down-modulation. In conclusion, deep-sequencing accurately profiled known, variant, and novel microRNAs expressed by endothelial cells in normoxia and hypoxia.


Asunto(s)
Células Endoteliales/metabolismo , Secuenciación de Nucleótidos de Alto Rendimiento , MicroARNs/análisis , MicroARNs/química , Carboxipeptidasas/metabolismo , Hipoxia de la Célula , Proliferación Celular , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Biblioteca de Genes , Células HEK293 , Humanos , MicroARNs/metabolismo , Anotación de Secuencia Molecular , Conformación de Ácido Nucleico , ARN Bicatenario , Análisis de Secuencia de ARN , Transducción de Señal
18.
Rev Cardiovasc Med ; 15(1): 31-7, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24762464

RESUMEN

A challenge of modern cardiovascular medicine is to find new, effective treatments for patients with refractory angina pectoris, a clinical condition characterized by severe angina despite optimal medical therapy. These patients are not candidates for surgical or percutaneous revascularization. Herein we review the most up-to-date information regarding the modern approach to the patient with refractory angina pectoris, from conventional medical management to new medications and shock wave therapy, focusing on the use of endothelial precursor cells (EPCs) in the treatment of this condition. Clinical limitations of the efficiency of conventional approaches justify the search for new therapeutic options. Regenerative medicine is considered the next step in the evolution of organ replacement therapy. It is driven largely by the same health needs as transplantation and replacement therapies, but it aims further than traditional approaches, such as cell-based therapy. Increasing knowledge of the role of circulating cells derived from bone marrow (EPCs) on cardiovascular homeostasis in physiologic and pathologic conditions has prompted the clinical use of these cells to relieve ischemia. The current state of therapeutic angiogenesis still leaves many questions unanswered. It is of paramount importance that the treatment is delivered safely. Direct intramyocardial and intracoronary administration has demonstrated acceptable safety profiles in early trials, and may represent a major advance over surgical thoracotomy. The combined efforts of bench and clinical researchers will ultimately answer the question of whether cell therapy is a suitable strategy for treatment of patients with refractory angina.


Asunto(s)
Acetanilidas/uso terapéutico , Angina de Pecho/terapia , Células Endoteliales/trasplante , Ondas de Choque de Alta Energía/uso terapéutico , Piperazinas/uso terapéutico , Bloqueadores de los Canales de Sodio/uso terapéutico , Trasplante de Células Madre , Acetanilidas/efectos adversos , Angina de Pecho/diagnóstico , Angina de Pecho/patología , Angina de Pecho/fisiopatología , Animales , Humanos , Neovascularización Fisiológica , Piperazinas/efectos adversos , Ranolazina , Regeneración , Bloqueadores de los Canales de Sodio/efectos adversos , Trasplante de Células Madre/efectos adversos , Resultado del Tratamiento
20.
Arterioscler Thromb Vasc Biol ; 33(12): 2867-76, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24072696

RESUMEN

OBJECTIVE: Dystrophin, the missing or defective protein in Duchenne muscular dystrophy, is expressed not only in muscle cells but also in vascular endothelial cells (ECs). In this study, we assessed the effects of dystrophin deficiency on the angiogenic capacities of ECs. APPROACH AND RESULTS: We isolated vascular ECs from mdx mice, the murine equivalent of Duchenne muscular dystrophy in humans, and wild-type controls, and we found that mdx-derived ECs have impaired angiogenic properties, in terms of migration, proliferation, and tube formation. They also undergo increased apoptosis in vitro compared with wild-type cells and have increased senescence-associated ß-galactosidase activity. Mdx-derived ECs also display reduced ability to support myoblast proliferation when cocultured with satellite cell-derived primary myoblasts. These endothelial defects are mirrored by systemic impairment of angiogenesis in vivo, both on induction of ischemia, stimulation with growth factors in the corneal model and matrigel plug assays, and tumor growth. We also found that dystrophin forms a complex with endothelial NO synthase and caveolin-1 in ECs, and that NO production and cGMP formation are compromised in ECs isolated from mdx mice. Interestingly, treatment with aspirin enhances production of both cGMP and NO in dystrophic ECs, whereas low-dose aspirin improves the dystrophic phenotype of mdx mice in vivo, in terms of resistance to physical exercise, muscle fiber permeability, and capillary density. CONCLUSIONS: These findings demonstrate that impaired angiogenesis is a novel player and potential therapeutic target in Duchenne muscular dystrophy.


Asunto(s)
Distrofina/metabolismo , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Distrofia Muscular de Duchenne/metabolismo , Neovascularización Fisiológica , Animales , Apoptosis , Aspirina/farmacología , Carcinoma Pulmonar de Lewis/irrigación sanguínea , Carcinoma Pulmonar de Lewis/metabolismo , Carcinoma Pulmonar de Lewis/patología , Caveolina 1/metabolismo , Movimiento Celular , Proliferación Celular , Células Cultivadas , Senescencia Celular , Técnicas de Cocultivo , Neovascularización de la Córnea/metabolismo , Neovascularización de la Córnea/patología , Neovascularización de la Córnea/fisiopatología , GMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Distrofina/genética , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/patología , Endotelio Vascular/fisiopatología , Isquemia/metabolismo , Isquemia/patología , Isquemia/fisiopatología , Ratones , Ratones Endogámicos mdx , Distrofia Muscular de Duchenne/tratamiento farmacológico , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patología , Distrofia Muscular de Duchenne/fisiopatología , Mutación , Mioblastos Esqueléticos/metabolismo , Mioblastos Esqueléticos/patología , Neovascularización Patológica , Neovascularización Fisiológica/efectos de los fármacos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA