Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Circ Res ; 121(1): 71-80, 2017 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-28533209

RESUMEN

RATIONALE: Stem cell therapy has increased the therapeutic armamentarium in the fight against ischemic heart disease and heart failure. The administration of exogenous stem cells has been investigated in patients suffering an acute myocardial infarction, with the final aim of salvaging jeopardized myocardium and preventing left ventricular adverse remodeling and functional deterioration. However, phase I and II clinical trials with autologous and first-generation stem cells have yielded inconsistent benefits and mixed results. OBJECTIVE: In the search for new and more efficient cellular regenerative products, interesting cardioprotective, immunoregulatory, and cardioregenerative properties have been demonstrated for human cardiac stem cells. On the other hand, allogeneic cells show several advantages over autologous sources: they can be produced in large quantities, easily administered off-the-shelf early after an acute myocardial infarction, comply with stringent criteria for product homogeneity, potency, and quality control, and may exhibit a distinctive immunologic behavior. METHODS AND RESULTS: With a promising preclinical background, CAREMI (Cardiac Stem Cells in Patients With Acute Myocardial Infarction) has been designed as a double-blind, 2:1 randomized, controlled, and multicenter clinical trial that will evaluate the safety, feasibility, and efficacy of intracoronary delivery of allogeneic human cardiac stem cell in 55 patients with large acute myocardial infarction, left ventricular dysfunction, and at high risk of developing heart failure. CONCLUSIONS: This phase I/II clinical trial represents a novel experience in humans with allogeneic cardiac stem cell in a rigorously imaging-based selected group of acute myocardial infarction patients, with detailed safety immunologic assessments and magnetic resonance imaging-based efficacy end points. CLINICAL TRIAL REGISTRATION: URL: http://www.clinicaltrials.gov. Unique identifier: NCT02439398.


Asunto(s)
Vasos Coronarios , Infarto del Miocardio/terapia , Miocitos Cardíacos/trasplante , Trasplante de Células Madre/métodos , Disfunción Ventricular Izquierda/terapia , Vasos Coronarios/fisiología , Método Doble Ciego , Estudios de Factibilidad , Estudios de Seguimiento , Humanos , Infusiones Intraarteriales/métodos , Infarto del Miocardio/diagnóstico , Trasplante Homólogo/métodos , Resultado del Tratamiento , Disfunción Ventricular Izquierda/diagnóstico
2.
Arterioscler Thromb Vasc Biol ; 38(9): 2160-2173, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29930004

RESUMEN

Objective- Cardiac progenitor cells reside in the heart in adulthood, although their physiological relevance remains unknown. Here, we demonstrate that after myocardial infarction, adult Bmi1+ (B lymphoma Mo-MLV insertion region 1 homolog [PCGF4]) cardiac cells are a key progenitor-like population in cardiac neovascularization during ventricular remodeling. Approach and Results- These cells, which have a strong in vivo differentiation bias, are a mixture of endothelial- and mesenchymal-related cells with in vitro spontaneous endothelial cell differentiation capacity. Genetic lineage tracing analysis showed that heart-resident Bmi1+ progenitor cells proliferate after acute myocardial infarction and differentiate to generate de novo cardiac vasculature. In a mouse model of induced myocardial infarction, genetic ablation of these cells substantially deteriorated both heart angiogenesis and the ejection fraction, resulting in an ischemic-dilated cardiac phenotype. Conclusions- These findings imply that endothelial-related Bmi1+ progenitor cells are necessary for injury-induced neovascularization in adult mouse heart and highlight these cells as a suitable therapeutic target for preventing dysfunctional left ventricular remodeling after injury.


Asunto(s)
Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocitos Cardíacos/patología , Miocitos Cardíacos/fisiología , Neovascularización Patológica , Complejo Represivo Polycomb 1/fisiología , Células Madre/patología , Células Madre/fisiología , Remodelación Ventricular , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Transgénicos , Proteínas Proto-Oncogénicas c-kit/metabolismo , Factores de Transcripción/metabolismo
3.
J Mol Cell Cardiol ; 99: 162-173, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27545043

RESUMEN

MicroRNAs (miR) have considerable potential as therapeutic tools in cardiac diseases. Alterations in atrial miR are involved in the development of atrial fibrillation (AF), but the molecular mechanism underlying their contribution to atrial remodeling in chronic atrial fibrillation (CAF) is only partially understood. Here we used miR array to analyze the miR profile of atrial biopsies from sinus rhythm (SR) and CAF patients. qRT-PCR identified a distinctive CAF-miR signature and described conserved miR-208b upregulation in human and ovine AF atrial tissue. We used bioinformatics analysis to predict genes and signaling pathways as putative miR-208b targets, which highlighted genes from the cardiac muscle gene program and from canonical WNT, gap-junction and Ca2+ signaling networks. Results from analysis of miR-208b-overexpressing HL-1 atrial myocytes and from myocytes isolated from CAF patients showed that aberrant miR-208b levels reduced the expression and function of L-type Ca2+ channel subunits (CACNA1C and CACNB2) as well as the sarcoplasmic reticulum-Ca2+ pump SERCA2. These findings clearly pointed to CAF-specific upregulated miR-208b as an important mediator in Ca2+ handling impairment during atrial remodeling.


Asunto(s)
Fibrilación Atrial/genética , Fibrilación Atrial/metabolismo , Calcio/metabolismo , Atrios Cardíacos/citología , Atrios Cardíacos/metabolismo , MicroARNs/genética , Miocitos Cardíacos/metabolismo , Regiones no Traducidas 3' , Animales , Fibrilación Atrial/fisiopatología , Remodelación Atrial , Secuencia de Bases , Canales de Calcio Tipo L/genética , Canales de Calcio Tipo L/metabolismo , Línea Celular , Enfermedad Crónica , Conexina 43/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Atrios Cardíacos/fisiopatología , Humanos , Miosinas/genética , Unión Proteica , Interferencia de ARN , ARN Mensajero/genética , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Ovinos , Proteínas Wnt/metabolismo
4.
Proteomics ; 15(7): 1332-7, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25504917

RESUMEN

Human cardiac stem cells (hCSC) express a portfolio of plasma membrane receptors that are involved in the regulatory auto/paracrine feedback loop mechanism of activation of these cells, and consequently contribute to myocardial regeneration. In order to attain a comprehensive description of hCSC receptome and overcoming the inability demonstrated by other technologies applied in receptor identification, mainly due to the transmembrane nature, high hydrophobic character and relative low concentration of these proteins, we have exploited and improved a proteomics workflow. This approach was based on the enrichment of hCSC plasma membrane fraction and addition of prefractionation steps prior to MS analysis. More than 100 plasma membrane receptors were identified. The data reported herein constitute a valuable source of information to further understand cardiac stem cells activation mechanisms and the subsequent cardiac repair process. All MS data have been deposited in the ProteomeXchange with identifier PXD001117 (http://proteomecentral.proteomexchange.org/dataset/PXD001117).


Asunto(s)
Células Madre Adultas/química , Proteoma/química , Receptores de Superficie Celular/química , Células Madre Adultas/metabolismo , Células Cultivadas , Cromatografía por Intercambio Iónico , Humanos , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Miocardio/patología , Proteoma/aislamiento & purificación , Proteoma/metabolismo , Proteómica , Receptores de Superficie Celular/aislamiento & purificación , Receptores de Superficie Celular/metabolismo , Regeneración , Espectrometría de Masas en Tándem
5.
Stem Cells ; 32(8): 2229-44, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24648336

RESUMEN

MicroRNAs, small noncoding RNAs, regulate gene expression primarily at the posttranscriptional level. We previously found that miR-335 is critically involved in the regulation and differentiation capacity of human mesenchymal stem cells (hMSCs) in vitro. In this study, we investigated the significance of miR-335 for the therapeutic potential of hMSCs. Analysis of hMSCs in ex vivo culture demonstrated a significant and progressive increase in miR-335 that is prevented by telomerase. Expression levels of miR-335 were also positively correlated with donor age of hMSCs, and were increased by stimuli that induce cell senescence, such as γ-irradiation and standard O2 concentration. Forced expression of miR-335 resulted in early senescence-like alterations in hMSCs, including: increased SA-ß-gal activity and cell size, reduced cell proliferation capacity, augmented levels of p16 protein, and the development of a senescence-associated secretory phenotype. Furthermore, overexpression of miR-335 abolished the in vivo chondro-osseous potential of hMSCs, and disabled their immunomodulatory capacity in a murine experimental model of lethal endotoxemia. These effects were accompanied by a severely reduced capacity for cell migration in response to proinflammatory signals and a marked reduction in Protein Kinase D1 phosphorylation, resulting in a pronounced decrease of AP-1 activity. Our results demonstrate that miR-335 plays a key role in the regulation of reparative activities of hMSCs and suggests that it might be considered a marker for the therapeutic potency of these cells in clinical applications.


Asunto(s)
Diferenciación Celular/genética , Senescencia Celular/genética , Células Madre Mesenquimatosas/fisiología , MicroARNs/genética , Factor de Transcripción AP-1/metabolismo , Western Blotting , Movimiento Celular/genética , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción Genética
6.
Stem Cells ; 32(7): 1865-77, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24496748

RESUMEN

Mesenchymal stem cells (MSCs) possess unique paracrine and immunosuppressive properties, which make them useful candidates for cellular therapy. Here, we address how cellular senescence influences the therapeutic potential of human MSCs (hMSCs). Senescence was induced in bone marrow-derived hMSC cultures with gamma irradiation. Control and senescent cells were tested for their immunoregulatory activity in vitro and in vivo, and an extensive molecular characterization of the phenotypic changes induced by senescence was performed. We also compared the gene expression profiles of senescent hMSCs with a collection of hMSCs used in an ongoing clinical study of Graft Versus Host disease (GVHD). Our results show that senescence induces extensive phenotypic changes in hMSCs and abrogates their protective activity in a murine model of LPS-induced lethal endotoxemia. Although senescent hMSCs retain an ability to regulate the inflammatory response on macrophages in vitro, and, in part retain their capacity to significantly inhibit lymphocyte proliferation, they have a severely impaired migratory capacity in response to proinflammatory signals, which is associated with an inhibition of the AP-1 pathway. Additionally, expression analysis identified PLEC, C8orf48, TRPC4, and ZNF14, as differentially regulated genes in senescent hMSCs that were similarly regulated in those hMSCs which failed to produce a therapeutic effect in a GVHD trial. All the observed phenotypic alterations were confirmed in replicative-senescent hMSCs. In conclusion, this study highlights important changes in the immunomodulatory phenotype of senescent hMSCs and provides candidate gene signatures which may be useful to evaluate the therapeutic potential of hMSCs used in future clinical studies.


Asunto(s)
Senescencia Celular , Endotoxemia/terapia , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/fisiología , Animales , Movimiento Celular , Proliferación Celular , Células Cultivadas , Endotoxemia/inmunología , Humanos , Inmunomodulación , Lipopolisacáridos/farmacología , Linfocitos/inmunología , Masculino , Ratones Endogámicos BALB C , Plectina/genética , Plectina/metabolismo , Canales Catiónicos TRPC/genética , Canales Catiónicos TRPC/metabolismo , Factor de Transcripción AP-1/metabolismo , Transcriptoma
7.
Biochem J ; 463(2): 201-13, 2014 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-25084525

RESUMEN

MAL2 (myelin and lymphocyte protein 2) is thought to regulate at least two steps in the hepatic apical transcytotic pathway. As vesicle budding and delivery at each step are driven by complex machineries, we predicted that MAL2 participates in several large protein complexes with multiple binding partners. To identify novel MAL2 interactors, we performed split-ubiquitin yeast two-hybrid assays and identified STK16 (serine/threonine kinase 16) as a putative interactor which we verified morphologically and biochemically. As STK16 is a Golgi-associated constitutively active kinase implicated in regulating secretion and because of the massive constitutive secretory capacity of hepatic cells, we tested whether MAL2 and STK16 function in secretion. Expression of a dominant-negative kinase-dead STK16 mutant (E202A) or knockdown of MAL2 impaired secretion that correlated with decreased expression of albumin and haptoglobin. By using 19°C temperature blocks and lysosome deacidification, we determined that E202A expression or MAL2 knockdown did not interfere with albumin synthesis or processing, but led to albumin lysosomal degradation. We conclude that MAL2 and the constitutively active STK16 function to sort secretory soluble cargo into the constitutive secretory pathway at the TGN (trans-Golgi network) in polarized hepatocytes.


Asunto(s)
Hepatocitos/metabolismo , Proteínas Proteolipídicas Asociadas a Mielina y Linfocito/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Factores de Transcripción/metabolismo , Albúminas/metabolismo , Hepatocitos/enzimología , Humanos , Lisosomas/enzimología , Lisosomas/metabolismo , Proteínas Proteolipídicas Asociadas a Mielina y Linfocito/genética , Unión Proteica , Proteínas Serina-Treonina Quinasas/genética , Transporte de Proteínas , Factores de Transcripción/genética , Técnicas del Sistema de Dos Híbridos , Red trans-Golgi/enzimología , Red trans-Golgi/metabolismo
8.
Cells ; 12(18)2023 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-37759522

RESUMEN

Oxidative stress-induced myocardial apoptosis and necrosis are critically involved in ischemic infarction, and several sources of extracellular vesicles appear to be enriched in therapeutic activities. The central objective was to identify and validate the differential exosome miRNA repertoire in human cardiac progenitor cells (CPC). CPC exosomes were first analyzed by LC-MS/MS and compared by RNAseq with exomes of human mesenchymal stromal cells and human fibroblasts to define their differential exosome miRNA repertoire (exo-miRSEL). Proteomics demonstrated a highly significant representation of cardiovascular development functions and angiogenesis in CPC exosomes, and RNAseq analysis yielded about 350 different miRNAs; among the exo-miRSEL population, miR-935 was confirmed as the miRNA most significantly up-regulated; interestingly, miR-935 was also found to be preferentially expressed in mouse primary cardiac Bmi1+high CPC, a population highly enriched in progenitors. Furthermore, it was found that transfection of an miR-935 antagomiR combined with oxidative stress treatment provoked a significant increment both in apoptotic and necrotic populations, whereas transfection of a miR-935 mimic did not modify the response. Conclusion. miR-935 is a highly differentially expressed miRNA in exo-miRSEL, and its expression reduction promotes oxidative stress-associated apoptosis. MiR-935, together with other exosomal miRNA members, could counteract oxidative stress-related apoptosis, at least in CPC surroundings.

9.
Arterioscler Thromb Vasc Biol ; 31(11): 2455-63, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21885849

RESUMEN

OBJECTIVE: Genetic ablation of the growth suppressor p27(Kip1) (p27) in the mouse aggravates atherosclerosis coinciding with enhanced arterial cell proliferation. However, it is unknown whether molecular mechanisms that limit p27's protective function contribute to atherosclerosis development and whether p27 exerts proliferation-independent activities in the arterial wall. This study aims to provide insight into both questions by investigating the role in atherosclerosis of p27 phosphorylation at serine 10 (p27-phospho-Ser10), a major posttranslational modification of this protein. METHODS AND RESULTS: Immunoblotting studies revealed a marked reduction in p27-phospho-Ser10 in atherosclerotic arteries from apolipoprotein E-null mice, and expression of the nonphosphorylatable mutant p27Ser10Ala, either global or restricted to bone marrow, accelerated atherosclerosis. p27Ser10Ala expression did not affect cell proliferation in early and advanced atheroma but activated RhoA/Rho-associated coiled-coil containing protein kinase (ROCK) signaling and promoted macrophage foam cell formation in a ROCK-dependent manner. Supporting the clinical relevance of these findings, human atherosclerotic coronary arteries exhibited a prominent reduction in p27-phospho-Ser10 and increased ezrin/radixin/moesin protein phosphorylation, a marker of RhoA/ROCK activation. CONCLUSION: Scarce phosphorylation of p27 at Ser10 is a hallmark of human and mouse atherosclerosis and promotes disease progression in mice. This proatherogenic effect is mediated by a proliferation-independent mechanism that involves augmented foam cell formation owing to increased RhoA/ROCK activity. These findings unveil a new atheroprotective action of p27 and identify p27-phospho-Ser10 as an attractive target for the treatment of atherosclerosis.


Asunto(s)
Aterosclerosis/metabolismo , Aterosclerosis/patología , Proliferación Celular , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/deficiencia , Células Espumosas/patología , Serina/metabolismo , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Arterias/metabolismo , Arterias/patología , Estudios de Casos y Controles , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Transducción de Señal , Proteínas de Unión al GTP rho , Quinasas Asociadas a rho , Proteína de Unión al GTP rhoA
10.
Adv Exp Med Biol ; 741: 103-20, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22457106

RESUMEN

Stem cells are characterized by their unlimited ability to divide specifically; a stem cell is capable of making an immense number of copies of itself, maintaining the same characteristics. Moreover, these cells are able to generate several of the cell lineages which make up the body, including cells from the heart, liver, kidney, neurons, and muscles. Investigation of the mechanisms through which this differentiation occurs, the genes involved and the possibility of increasing the efficiency with which stem cells can be isolated and/or characterized are currently among the most important fields in biology and biomedicine.To date, stems cells have been identified from four different sources: Embryonic stem cells (ESC), germinal stem cells, and those derived from embryonic carcinomas (teratocarcinomas) and from somatic tissues (somatic stem cells). The latter are called adult stem cells (ASC) when they are found in postnatal tissues. We now know that there is a great diversity among ASC, with some tissues, such as the bone marrow, containing more than one type of ASC. Adult stem cells have several characteristics that make them to be the main players in current regenerative medicine and are being investigated as potential therapeutic agents for a wide variety of diseases. Specifically, HSC and MSC are being assessed in increasing numbers of clinical trials.


Asunto(s)
Células Madre Adultas/fisiología , Adulto , Células Madre Adultas/citología , División Celular/fisiología , Linaje de la Célula , Ensayos Clínicos como Asunto , Expresión Génica , Humanos , Transducción de Señal/fisiología , Nicho de Células Madre
11.
PLoS Genet ; 5(2): e1000389, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19229323

RESUMEN

Polymerase micro (Polmicro) is an error-prone, DNA-directed DNA polymerase that participates in non-homologous end-joining (NHEJ) repair. In vivo, Polmicro deficiency results in impaired Vkappa-Jkappa recombination and altered somatic hypermutation and centroblast development. In Polmicro(-/-) mice, hematopoietic development was defective in several peripheral and bone marrow (BM) cell populations, with about a 40% decrease in BM cell number that affected several hematopoietic lineages. Hematopoietic progenitors were reduced both in number and in expansion potential. The observed phenotype correlates with a reduced efficiency in DNA double-strand break (DSB) repair in hematopoietic tissue. Whole-body gamma-irradiation revealed that Polmicro also plays a role in DSB repair in non-hematopoietic tissues. Our results show that Polmicro function is required for physiological hematopoietic development with an important role in maintaining early progenitor cell homeostasis and genetic stability in hematopoietic and non-hematopoietic tissues.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN , ADN Polimerasa Dirigida por ADN/metabolismo , Hematopoyesis , Animales , Células Cultivadas , ADN Polimerasa Dirigida por ADN/genética , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/enzimología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados
12.
Antioxidants (Basel) ; 11(5)2022 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-35624750

RESUMEN

Research on cardiac progenitor cell populations has generated expectations about their potential for cardiac regeneration capacity after acute myocardial infarction and during physiological aging; however, the endogenous capacity of the adult mammalian heart is limited. The modest efficacy of exogenous cell-based treatments can guide the development of new approaches that, alone or in combination, can be applied to boost clinical efficacy. The identification and manipulation of the adult stem cell environment, termed niche, will be critical for providing new evidence on adult stem cell populations and improving stem-cell-based therapies. Here, we review and discuss the state of our understanding of the interaction of adult cardiac progenitor cells with other cardiac cell populations, with a focus on the description of the B-CPC progenitor population (Bmi1+ cardiac progenitor cell), which is a strong candidate progenitor for all main cardiac cell lineages, both in the steady state and after cardiac damage. The set of all interactions should be able to define the vascular cardiac stem cell niche, which is associated with low oxidative stress domains in vasculature, and whose manipulation would offer new hope in the cardiac regeneration field.

13.
Sci Rep ; 12(1): 146, 2022 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-34997006

RESUMEN

Clinical trials evaluating cardiac progenitor cells (CPC) demonstrated feasibility and safety, but no clear functional benefits. Therefore a deeper understanding of CPC biology is warranted to inform strategies capable to enhance their therapeutic potential. Here we have defined, using a label-free proteomic approach, the differential cytoplasmic and nuclear compartments of human CPC (hCPC). Global analysis of cytoplasmic repertoire in hCPC suggested an important hypoxia response capacity and active collagen metabolism. In addition, comparative analysis of the nuclear protein compartment identified a significant regulation of a small number of proteins in hCPC versus human mesenchymal stem cells (hMSC). Two proteins significantly upregulated in the hCPC nuclear compartment, IL1A and IMP3, showed also a parallel increase in mRNA expression in hCPC versus hMSC, and were studied further. IL1A, subjected to an important post-transcriptional regulation, was demonstrated to act as a dual-function cytokine with a plausible role in apoptosis regulation. The knockdown of the mRNA binding protein (IMP3) did not negatively impact hCPC viability, but reduced their proliferation and migration capacity. Analysis of a panel of putative candidate genes identified HMGA2 and PTPRF as IMP3 targets in hCPC. Therefore, they are potentially involved in hCPC proliferation/migration regulation.


Asunto(s)
Núcleo Celular/metabolismo , Citoplasma/metabolismo , Fibroblastos/metabolismo , Células Madre Mesenquimatosas/metabolismo , Miocitos Cardíacos/metabolismo , Proteoma , Proteómica , Movimiento Celular , Proliferación Celular , Células Cultivadas , Regulación de la Expresión Génica , Proteína HMGA2/genética , Proteína HMGA2/metabolismo , Humanos , Interleucina-1alfa/genética , Interleucina-1alfa/metabolismo , Estrés Oxidativo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Proteínas Tirosina Fosfatasas Clase 2 Similares a Receptores/genética , Proteínas Tirosina Fosfatasas Clase 2 Similares a Receptores/metabolismo , Transducción de Señal
15.
Antioxidants (Basel) ; 11(10)2022 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-36290678

RESUMEN

Anthracycline-induced cardiotoxicity is the most severe collateral effect of chemotherapy originated by an excess of oxidative stress in cardiomyocytes that leads to cardiac dysfunction. We assessed clinical data from patients with breast cancer receiving anthracyclines and searched for discriminating microRNAs between patients that developed cardiotoxicity (cases) and those that did not (controls), using RNA sequencing and regression analysis. Serum levels of 25 microRNAs were differentially expressed in cases versus controls within the first year after anthracycline treatment, as assessed by three different regression models (elastic net, Robinson and Smyth exact negative binomial test and random forest). MiR-4732-3p was the only microRNA identified in all regression models and was downregulated in patients that experienced cardiotoxicity. MiR-4732-3p was also present in neonatal rat cardiomyocytes and cardiac fibroblasts and was modulated by anthracycline treatment. A miR-4732-3p mimic was cardioprotective in cardiac and fibroblast cultures, following doxorubicin challenge, in terms of cell viability and ROS levels. Notably, administration of the miR-4732-3p mimic in doxorubicin-treated rats preserved cardiac function, normalized weight loss, induced angiogenesis, and decreased apoptosis, interstitial fibrosis and cardiac myofibroblasts. At the molecular level, miR-4732-3p regulated genes of TGFß and Hippo signaling pathways. Overall, the results indicate that miR-4732-3p is a novel biomarker of cardiotoxicity that has therapeutic potential against anthracycline-induced heart damage.

16.
Nat Med ; 10(9): 917-9, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15322538

RESUMEN

Several non-hypercalcemic analogs of 1alpha,25-dihydroxyvitamin D3 (1,25(OH)(2)D(3)) show antitumor activity in a subset of cancer patients. High vitamin D receptor (VDR) expression, which is associated with good prognosis but is lost during tumor progression. We show that the SNAIL transcription factor represses VDR gene expression in human colon cancer cells and blocks the antitumor action of EB1089, a 1,25(OH)(2)D(3) analog, in xenografted mice. In human colon cancers, elevated SNAIL expression correlates with downregulation of VDR.


Asunto(s)
Calcitriol/análogos & derivados , Calcitriol/antagonistas & inhibidores , Neoplasias del Colon/metabolismo , Proteínas de Unión al ADN/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Receptores de Calcitriol/metabolismo , Factores de Transcripción/farmacología , Animales , Antineoplásicos/antagonistas & inhibidores , Cadherinas/metabolismo , Relación Dosis-Respuesta a Droga , Ensayo de Cambio de Movilidad Electroforética , Humanos , Inmunoprecipitación , Ratones , Regiones Promotoras Genéticas/genética , Receptores de Calcitriol/genética , Factores de Transcripción de la Familia Snail
17.
Cells ; 10(10)2021 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-34685551

RESUMEN

Human cardiac progenitor cells (hCPC) are considered a good candidate in cell therapy for ischemic heart disease, demonstrating capacity to improve functional recovery after myocardial infarction (MI), both in small and large preclinical animal models. However, improvements are required in terms of cell engraftment and efficacy. Based on previously published reports, insulin-growth factor 1 (IGF-1) and hepatocyte growth factor (HGF) have demonstrated substantial cardioprotective, repair and regeneration activities, so they are good candidates to be evaluated in large animal model of MI. We have validated porcine cardiac progenitor cells (pCPC) and lentiviral vectors to overexpress IGF-1 (co-expressing eGFP) and HGF (co-expressing mCherry). pCPC were transduced and IGF1-eGFPpos and HGF-mCherrypos populations were purified by cell sorting and further expanded. Overexpression of IGF-1 has a limited impact on pCPC expression profile, whereas results indicated that pCPC-HGF-mCherry cultures could be counter selecting high expresser cells. In addition, pCPC-IGF1-eGFP showed a higher cardiogenic response, evaluated in co-cultures with decellularized extracellular matrix, compared with native pCPC or pCPC-HGF-mCherry. In vivo intracoronary co-administration of pCPC-IGF1-eGFP and pCPC-HFG-mCherry (1:1; 40 × 106/animal), one week after the induction of an MI model in swine, revealed no significant improvement in cardiac function.


Asunto(s)
Factor de Crecimiento de Hepatocito/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Infarto del Miocardio/terapia , Miocitos Cardíacos/metabolismo , Células Madre/metabolismo , Animales , Modelos Animales de Enfermedad , Femenino , Infarto del Miocardio/fisiopatología , Porcinos
18.
Stem Cells ; 27(5): 1164-72, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19418448

RESUMEN

A major unanswered question in autologous cell therapy is the appropriate timing for cell isolation. Many of the putative target diseases arise with old age and previous evidence, mainly from animal models, suggests that the stem/progenitor cell pool decreases steadily with age. Studies with human cells have been generally hampered to date by poor sample availability. In recent years, several laboratories have reported on the existence, both in rodents and humans, of skin-derived precursor (SKP) cells with the capacity to generate neural and mesodermal progenies. This easily obtainable multipotent cell population has raised expectations for their potential use in cell therapy of neurodegeneration. However, we still lack a clear understanding of the spatiotemporal abundance and phenotype of human SKPs. Here we show an analysis of human SKP abundance and in vitro differentiation potential, by using SKPs isolated from four distinct anatomic sites (abdomen, breast, foreskin, and scalp) from 102 healthy subjects aged 8 months to 85 years. Human SKP abundance and differentiation potential decrease sharply with age, being extremely difficult to isolate, expand, and differentiate when obtained from the elderly. Our data suggest preserving human SKP cell banks early in life would be desirable for use in clinical protocols in the aging population.


Asunto(s)
Envejecimiento/fisiología , Piel/citología , Células Madre/citología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores/metabolismo , Biopsia , Diferenciación Celular , Proliferación Celular , Separación Celular , Niño , Preescolar , Dermis/citología , Prepucio/citología , Humanos , Masculino , Persona de Mediana Edad , Fenotipo , Nicho de Células Madre/citología , Donantes de Tejidos
19.
Structure ; 16(1): 115-24, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18184589

RESUMEN

The activation segment of protein kinases is structurally highly conserved and central to regulation of kinase activation. Here we report an atypical activation segment architecture in human MPSK1 comprising a beta sheet and a large alpha-helical insertion. Sequence comparisons suggested that similar activation segments exist in all members of the MPSK1 family and in MAST kinases. The consequence of this nonclassical activation segment on substrate recognition was studied using peptide library screens that revealed a preferred substrate sequence of X-X-P/V/I-phi-H/Y-T*-N/G-X-X-X (phi is an aliphatic residue). In addition, we identified the GTPase DRG1 as an MPSK1 interaction partner and specific substrate. The interaction domain in DRG1 was mapped to the N terminus, leading to recruitment and phosphorylation at Thr100 within the GTPase domain. The presented data reveal an atypical kinase structural motif and suggest a role of MPSK1 regulating DRG1, a GTPase involved in regulation of cellular growth.


Asunto(s)
Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/metabolismo , Factores de Transcripción/química , Factores de Transcripción/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia Conservada , Activación Enzimática , Humanos , Cinética , Modelos Moleculares , Datos de Secuencia Molecular , Conformación Proteica , Estaurosporina/metabolismo , Especificidad por Sustrato
20.
Stem Cells Int ; 2020: 8872009, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33101423

RESUMEN

Human bone marrow mesenchymal stem cells (BM-MSCs) and cardiac progenitor/stem cells (CPCs) have been extensively studied as a potential therapeutic treatment for myocardial infarction (MI). Previous reports suggest that lower doses of CPCs are needed to improve cardiac function relative to their bone marrow counterparts. Here, we confirmed this observations and investigated the surface protein expression profile that might explain this effect. Myocardial infarction was performed in nude rats by permanent ligation of the left coronary artery. Cardiac function and infarct size before and after cell transplantation were evaluated by echocardiography and morphometry, respectively. The CPC and BM-MSC receptome were analyzed by proteomic analysis of biotin-labeled surface proteins. Rats transplanted with CPCs showed a greater improvement in cardiac function after MI than those transplanted with BM-MSCs, and this was associated with a smaller infarct size. Analysis of the receptome of CPCs and BM-MSCs showed that gene ontology biological processes and KEGG pathways associated with adhesion mechanisms were upregulated in CPCs compared with BM-MSCs. Moreover, the membrane protein interactome in CPCs showed a strong relationship with biological processes related to cell adhesion whereas the BM-MSCs interactome was more related to immune regulation processes. We conclude that the stronger capacity of CPCs over BM-MSCs to engraft in the infarcted area is likely linked to a more pronounced cell adhesion expression program.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA