Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Haematologica ; 106(10): 2613-2623, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32703790

RESUMEN

Transcriptional profiling of hematopoietic cell subpopulations has helped to characterize the developmental stages of the hematopoietic system and the molecular bases of malignant and non-malignant blood diseases. Previously, only the genes targeted by expression microarrays could be profiled genome-wide. High-throughput RNA sequencing, however, encompasses a broader repertoire of RNA molecules, without restriction to previously annotated genes. We analyzed the BLUEPRINT consortium RNA-sequencing data for mature hematopoietic cell types. The data comprised 90 total RNA-sequencing samples, each composed of one of 27 cell types, and 32 small RNA-sequencing samples, each composed of one of 11 cell types. We estimated gene and isoform expression levels for each cell type using existing annotations from Ensembl. We then used guided transcriptome assembly to discover unannotated transcripts. We identified hundreds of novel non-coding RNA genes and showed that the majority have cell type-dependent expression. We also characterized the expression of circular RNA and found that these are also cell type-specific. These analyses refine the active transcriptional landscape of mature hematopoietic cells, highlight abundant genes and transcriptional isoforms for each blood cell type, and provide a valuable resource for researchers of hematologic development and diseases. Finally, we made the data accessible via a web-based interface: https://blueprint.haem.cam.ac.uk/bloodatlas/.


Asunto(s)
ARN Largo no Codificante , Transcriptoma , Perfilación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , ARN Circular , ARN Largo no Codificante/genética , Análisis de Secuencia de ARN
2.
Circ Res ; 123(2): 301-308, 2018 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-29976694

RESUMEN

Heart failure (HF) is one of the leading causes of death worldwide and has reached epidemic proportions in most industrialized nations. Despite major improvements in the treatment and management of the disease, the prognosis for patients with HF remains poor with approximately only half of patients surviving for 5 years or longer after diagnosis. The poor prognosis of HF patients is in part because of irreparable damage to cardiac tissue and concomitant maladaptive changes associated with the disease. Cell-based therapies may have the potential to transform the treatment and prognosis of HF through regeneration or repair of damaged cardiac tissue. Accordingly, numerous phase I and II randomized clinical trials have tested the clinical benefits of cell transplant, mostly autologous bone marrow-derived mononuclear cells, in patients with HF, ischemic heart disease, and acute myocardial infarction. Although many of these trials were relatively small, meta-analyses of cell-based therapies have attempted to apply rigorous statistical methodology to assess the potential clinical benefits of the intervention. As a prelude to larger phase III trials, meta-analyses, therefore, remain the obvious means of evaluating the available clinical evidence. Here, we review the different meta-analyses of randomized clinical trials that evaluate the safety and potential beneficial effect of cell therapies in HF and acute myocardial infarction spanning nearly 2 decades since the first pioneering trials were conducted.


Asunto(s)
Estudios Clínicos como Asunto , Insuficiencia Cardíaca/terapia , Infarto del Miocardio/terapia , Trasplante de Células Madre/métodos , Animales , Humanos , Trasplante de Células Madre/efectos adversos , Investigación Biomédica Traslacional/métodos
3.
Circ Res ; 118(8): 1264-72, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-27081109

RESUMEN

Controversies from basic science, discrepancies from clinical trials, and divergent results from meta-analyses have recently arisen in the field of cell therapies for cardiovascular repair and regeneration. Noticeably, there are almost as many systematic reviews and meta-analyses published as there are well-conducted clinical studies. But how do we disentangle the confusion they have raised? This article addresses why results obtained from systematic reviews and meta-analyses of human cell-based cardiac regeneration therapies are still valid to inform the design of future clinical trials. It also addresses how meta-analyses are not free from limitations and how important it is to assess the quality of the evidence and the quality of the systematic reviews and finally how stronger conclusions can be drawn when several pieces of evidence converge.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Metaanálisis como Asunto , Isquemia Miocárdica/terapia , Regeneración/fisiología , Literatura de Revisión como Asunto , Animales , Tratamiento Basado en Trasplante de Células y Tejidos/tendencias , Humanos , Isquemia Miocárdica/diagnóstico , Isquemia Miocárdica/epidemiología , Ensayos Clínicos Controlados Aleatorios como Asunto
4.
Stem Cells ; 34(6): 1664-78, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26866290

RESUMEN

Hematopoietic stem/progenitor cells (HSPCs) reside in specialized bone marrow microenvironmental niches, with vascular elements (endothelial/mesenchymal stromal cells) and CXCR4-CXCL12 interactions playing particularly important roles for HSPC entry, retention, and maintenance. The functional effects of CXCL12 are dependent on its local concentration and rely on complex HSPC-niche interactions. Two Junctional Adhesion Molecule family proteins, Junctional Adhesion Molecule-B (JAM)-B and JAM-C, are reported to mediate HSPC-stromal cell interactions, which in turn regulate CXCL12 production by mesenchymal stromal cells (MSCs). Here, we demonstrate that another JAM family member, JAM-A, is most highly expressed on human hematopoietic stem cells with in vivo repopulating activity (p < .01 for JAM-A(high) compared to JAM-A(Int or Low) cord blood CD34(+) cells). JAM-A blockade, silencing, and overexpression show that JAM-A contributes significantly (p < .05) to the adhesion of human HSPCs to IL-1ß activated human bone marrow sinusoidal endothelium. Further studies highlight a novel association of JAM-A with CXCR4, with these molecules moving to the leading edge of the cell upon presentation with CXCL12 (p < .05 compared to no CXCL12). Therefore, we hypothesize that JAM family members differentially regulate CXCR4 function and CXCL12 secretion in the bone marrow niche. Stem Cells 2016;34:1664-1678.


Asunto(s)
Células Madre Hematopoyéticas/metabolismo , Molécula A de Adhesión de Unión/metabolismo , Receptores CXCR4/metabolismo , Antígeno AC133/metabolismo , Antígenos CD34/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Adhesión Celular/efectos de los fármacos , Quimiocina CXCL12/farmacología , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Sangre Fetal/citología , Técnicas de Silenciamiento del Gen , Células HL-60 , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Humanos , Células Jurkat , Unión Proteica/efectos de los fármacos , Nicho de Células Madre/efectos de los fármacos
5.
Circ Res ; 116(8): 1361-77, 2015 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-25632038

RESUMEN

RATIONALE: Cell-based therapies are a promising intervention for the treatment of heart failure (HF) secondary to ischemic and nonischemic cardiomyopathy. However, the clinical efficacy of such new treatment requires further evaluation. OBJECTIVE: To assess available clinical evidence on the safety and efficacy of cell-based therapies for HF. METHODS AND RESULTS: Electronic databases (CENTRAL, DARE, NHSEED & HTA, PubMed, MEDLINE, EMBASE, CINAHL, LILACS, KoreaMed, PakMediNet, IndMed, and the Transfusion Evidence Library) were searched for relevant randomized controlled trials to June 2014. Trials of participants with HF and where the administration of any dose of autologous cells by any delivery route was compared with no intervention or placebo were eligible for inclusion. Primary outcomes were defined as mortality and rehospitalization as a result of HF. Secondary outcomes included performance status, quality of life, incidence of arrhythmias, brain natriuretic peptide levels, left ventricular ejection fraction, myocardial perfusion, and adverse events. Thirty-one independent trials (1521 participants) were included. The treatment significantly reduced the risk of mortality and rehospitalization caused by HF. There was a significant improvement in favor of stem cell treatment in performance status and exercise capacity, left ventricular ejection fraction, and quality of life. The treatment was also associated with a reduction of brain natriuretic peptide levels and no increase in the incidence of arrhythmias. However, there was considerable risk of performance, selection, and reporting bias among the included trials. CONCLUSIONS: This study shows evidence that autologous cell therapy may be beneficial for patients having HF, but further evidence is required.


Asunto(s)
Insuficiencia Cardíaca/cirugía , Regeneración , Trasplante de Células Madre , Función Ventricular Izquierda , Distribución de Chi-Cuadrado , Supervivencia sin Enfermedad , Insuficiencia Cardíaca/mortalidad , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/fisiopatología , Humanos , Contracción Miocárdica , Oportunidad Relativa , Readmisión del Paciente , Calidad de Vida , Ensayos Clínicos Controlados Aleatorios como Asunto , Recuperación de la Función , Factores de Riesgo , Trasplante de Células Madre/mortalidad , Volumen Sistólico , Factores de Tiempo , Trasplante Autólogo , Resultado del Tratamiento
6.
Cochrane Database Syst Rev ; 12: CD007888, 2016 12 24.
Artículo en Inglés | MEDLINE | ID: mdl-28012165

RESUMEN

BACKGROUND: A promising approach to the treatment of chronic ischaemic heart disease and congestive heart failure is the use of stem cells. The last decade has seen a plethora of randomised controlled trials developed worldwide, which have generated conflicting results. OBJECTIVES: The critical evaluation of clinical evidence on the safety and efficacy of autologous adult bone marrow-derived stem/progenitor cells as a treatment for chronic ischaemic heart disease and congestive heart failure. SEARCH METHODS: We searched CENTRAL in the Cochrane Library, MEDLINE, Embase, CINAHL, LILACS, and four ongoing trial databases for relevant trials up to 14 December 2015. SELECTION CRITERIA: Eligible studies were randomised controlled trials comparing autologous adult stem/progenitor cells with no cells in people with chronic ischaemic heart disease and congestive heart failure. We included co-interventions, such as primary angioplasty, surgery, or administration of stem cell mobilising agents, when administered to treatment and control arms equally. DATA COLLECTION AND ANALYSIS: Two review authors independently screened all references for eligibility, assessed trial quality, and extracted data. We undertook a quantitative evaluation of data using random-effects meta-analyses. We evaluated heterogeneity using the I2 statistic and explored substantial heterogeneity (I2 greater than 50%) through subgroup analyses. We assessed the quality of the evidence using the GRADE approach. We created a 'Summary of findings' table using GRADEprofiler (GRADEpro), excluding studies with a high or unclear risk of selection bias. We focused our summary of findings on long-term follow-up of mortality, morbidity outcomes, and left ventricular ejection fraction measured by magnetic resonance imaging. MAIN RESULTS: We included 38 randomised controlled trials involving 1907 participants (1114 cell therapy, 793 controls) in this review update. Twenty-three trials were at high or unclear risk of selection bias. Other sources of potential bias included lack of blinding of participants (12 trials) and full or partial commercial sponsorship (13 trials).Cell therapy reduced the incidence of long-term mortality (≥ 12 months) (risk ratio (RR) 0.42, 95% confidence interval (CI) 0.21 to 0.87; participants = 491; studies = 9; I2 = 0%; low-quality evidence). Periprocedural adverse events associated with the mapping or cell/placebo injection procedure were infrequent. Cell therapy was also associated with a long-term reduction in the incidence of non-fatal myocardial infarction (RR 0.38, 95% CI 0.15 to 0.97; participants = 345; studies = 5; I2 = 0%; low-quality evidence) and incidence of arrhythmias (RR 0.42, 95% CI 0.18 to 0.99; participants = 82; studies = 1; low-quality evidence). However, we found no evidence that cell therapy affects the risk of rehospitalisation for heart failure (RR 0.63, 95% CI 0.36 to 1.09; participants = 375; studies = 6; I2 = 0%; low-quality evidence) or composite incidence of mortality, non-fatal myocardial infarction, and/or rehospitalisation for heart failure (RR 0.64, 95% CI 0.38 to 1.08; participants = 141; studies = 3; I2 = 0%; low-quality evidence), or long-term left ventricular ejection fraction when measured by magnetic resonance imaging (mean difference -1.60, 95% CI -8.70 to 5.50; participants = 25; studies = 1; low-quality evidence). AUTHORS' CONCLUSIONS: This systematic review and meta-analysis found low-quality evidence that treatment with bone marrow-derived stem/progenitor cells reduces mortality and improves left ventricular ejection fraction over short- and long-term follow-up and may reduce the incidence of non-fatal myocardial infarction and improve New York Heart Association (NYHA) Functional Classification in people with chronic ischaemic heart disease and congestive heart failure. These findings should be interpreted with caution, as event rates were generally low, leading to a lack of precision.


Asunto(s)
Insuficiencia Cardíaca/cirugía , Isquemia Miocárdica/cirugía , Trasplante de Células Madre/métodos , Células Madre Adultas/trasplante , Arritmias Cardíacas/epidemiología , Células de la Médula Ósea/citología , Enfermedad Crónica , Insuficiencia Cardíaca/mortalidad , Hospitalización/estadística & datos numéricos , Humanos , Infarto del Miocardio/epidemiología , Isquemia Miocárdica/mortalidad , Readmisión del Paciente , Ensayos Clínicos Controlados Aleatorios como Asunto , Trasplante de Células Madre/efectos adversos , Trasplante de Células Madre/mortalidad , Volumen Sistólico/fisiología
7.
Br J Haematol ; 169(4): 552-64, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25757087

RESUMEN

Murine models of bone marrow transplantation show that pre-conditioning regimens affect the integrity of the bone marrow endothelium and that the repair of this vascular niche is an essential pre-requisite for successful haematopoietic stem and progenitor cell engraftment. Little is known about the angiogenic pathways that play a role in the repair of the human bone marrow vascular niche. We therefore established an in vitro humanized model, composed of bone marrow stromal and endothelial cells and have identified several pro-angiogenic factors, VEGFA, ANGPT1, CXCL8 and CXCL16, produced by the stromal component of this niche. We demonstrate for the first time that addition of CXCL8 or inhibition of its receptor, CXCR2, modulates blood vessel formation in our bone marrow endothelial niche model. Compared to wild type, Cxcr2(-/-) mice displayed a reduction in bone marrow cellularity and delayed platelet and leucocyte recovery following myeloablation and bone marrow transplantation. The delay in bone marrow recovery correlated with impaired bone marrow vascular repair. Taken together, our data demonstrate that CXCR2 regulates bone marrow blood vessel repair/regeneration and haematopoietic recovery, and clinically may be a therapeutic target for improving bone marrow transplantation.


Asunto(s)
Trasplante de Médula Ósea , Médula Ósea/irrigación sanguínea , Hematopoyesis , Trasplante de Células Madre Hematopoyéticas , Neovascularización Fisiológica , Receptores de Interleucina-8B/metabolismo , Proteínas Angiogénicas/genética , Proteínas Angiogénicas/metabolismo , Animales , Línea Celular , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Ratones Noqueados , Receptores de Interleucina-8B/genética , Acondicionamiento Pretrasplante
8.
Circ J ; 79(2): 229-36, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25744736

RESUMEN

Heart failure (HF) is the major cause of mortality worldwide. For more than a decade, cell-based therapies have been developed as treatment for heart disease as an alternative to current therapies. Trials and systematic reviews have assessed the safety and efficacy of cell therapies in a diverse number of participants and clinical settings. The present study collated and synthesized evidence from all systematic reviews related to cell-based therapies and HF. A total of 11 systematic reviews were identified through searches of electronic databases up to June 2014. We set out to answer 2 key questions on the efficacy of cell therapies in HF: (1) What is the overall effect of cell therapies on primary outcomes such as left ventricular ejection fraction (LVEF) and mortality? (2) How important is it to define the clinical setting and length of follow-up when assessing cell therapies and HF? There seems to be enough evidence to suggest that cell therapies have a moderate, long-lasting effect on LVEF, but the reduction on the risk of mortality observed by some systematic reviews needs to be confirmed in larger, statistically powered clinical trials. Additionally, and in order to strengthen conclusions, it is important to assess clinical evidence for defined clinical settings and to standardize the length of follow-up when comparing outcome data across several trials and systematic reviews.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos , Bases de Datos Factuales , Insuficiencia Cardíaca/fisiopatología , Insuficiencia Cardíaca/terapia , Volumen Sistólico , Ensayos Clínicos como Asunto , Insuficiencia Cardíaca/mortalidad , Humanos
9.
Cochrane Database Syst Rev ; (9): CD006536, 2015 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-26419913

RESUMEN

BACKGROUND: Cell transplantation offers a potential therapeutic approach to the repair and regeneration of damaged vascular and cardiac tissue after acute myocardial infarction (AMI). This has resulted in multiple randomised controlled trials (RCTs) across the world. OBJECTIVES: To determine the safety and efficacy of autologous adult bone marrow stem cells as a treatment for acute myocardial infarction (AMI), focusing on clinical outcomes. SEARCH METHODS: This Cochrane review is an update of a previous version (published in 2012). We searched the Cochrane Central Register of Controlled Trials (CENTRAL 2015, Issue 2), MEDLINE (1950 to March 2015), EMBASE (1974 to March 2015), CINAHL (1982 to March 2015) and the Transfusion Evidence Library (1980 to March 2015). In addition, we searched several international and ongoing trial databases in March 2015 and handsearched relevant conference proceedings to January 2011. SELECTION CRITERIA: RCTs comparing autologous bone marrow-derived cells with no cells in patients diagnosed with AMI were eligible. DATA COLLECTION AND ANALYSIS: Two review authors independently screened all references, assessed the risk of bias of the included trials and extracted data. We conducted meta-analyses using random-effects models throughout. We analysed outcomes at short-term (less than 12 months) and long-term (12 months or more) follow-up. Dichotomous outcomes are reported as risk ratio (RR) and continuous outcomes are reported as mean difference (MD) or standardised MD (SMD). We performed sensitivity analyses to evaluate the results in the context of the risk of selection, performance and attrition bias. Exploratory subgroup analysis investigated the effects of baseline cardiac function (left ventricular ejection fraction, LVEF) and cell dose, type and timing of administration, as well as the use of heparin in the final cell solution. MAIN RESULTS: Forty-one RCTs with a total of 2732 participants (1564 cell therapy, 1168 controls) were eligible for inclusion. Cell treatment was not associated with any changes in the risk of all-cause mortality (34/538 versus 32/458; RR 0.93, 95% CI 0.58 to 1.50; 996 participants; 14 studies; moderate quality evidence), cardiovascular mortality (23/277 versus 18/250; RR 1.04, 95% CI 0.54 to 1.99; 527 participants; nine studies; moderate quality evidence) or a composite measure of mortality, reinfarction and re-hospitalisation for heart failure (24/262 versus 33/235; RR 0.63, 95% CI 0.36 to 1.10; 497 participants; six studies; moderate quality evidence) at long-term follow-up. Statistical heterogeneity was low (I(2) = 0% to 12%). Serious periprocedural adverse events were rare and were generally unlikely to be related to cell therapy. Additionally, cell therapy had no effect on morbidity, quality of life/performance or LVEF measured by magnetic resonance imaging. Meta-analyses of LVEF measured by echocardiography, single photon emission computed tomography and left ventricular angiography showed evidence of differences in mean LVEF between treatment groups although the mean differences ranged between 2% and 5%, which are accepted not to be clinically relevant. Results were robust to the risk of selection, performance and attrition bias from individual studies. AUTHORS' CONCLUSIONS: The results of this review suggest that there is insufficient evidence for a beneficial effect of cell therapy for AMI patients. However, most of the evidence comes from small trials that showed no difference in clinically relevant outcomes. Further adequately powered trials are needed and until then the efficacy of this intervention remains unproven.


Asunto(s)
Infarto del Miocardio/cirugía , Trasplante de Células Madre/métodos , Hospitalización/estadística & datos numéricos , Humanos , Infarto del Miocardio/mortalidad , Ensayos Clínicos Controlados Aleatorios como Asunto , Recurrencia , Trasplante de Células Madre/efectos adversos , Volumen Sistólico/fisiología
10.
Cochrane Database Syst Rev ; (4): CD007888, 2014 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-24777540

RESUMEN

BACKGROUND: A promising approach to the treatment of chronic ischaemic heart disease (IHD) and heart failure is the use of stem cells. The last decade has seen a plethora of randomised controlled trials (RCTs) developed worldwide which have generated conflicting results. OBJECTIVES: The critical evaluation of clinical evidence on the safety and efficacy of autologous adult bone marrow-derived stem cells (BMSC) as a treatment for chronic ischaemic heart disease (IHD) and heart failure. SEARCH METHODS: We searched the Cochrane Central Register of Controlled Trials (CENTRAL) (The Cochrane Library, 2013, Issue 3), MEDLINE (from 1950), EMBASE (from 1974), CINAHL (from 1982) and the Transfusion Evidence Library (from 1980), together with ongoing trial databases, for relevant trials up to 31st March 2013. SELECTION CRITERIA: Eligible studies included RCTs comparing autologous adult stem/progenitor cells with no autologous stem/progenitor cells in participants with chronic IHD and heart failure. Co-interventions such as primary angioplasty, surgery or administration of stem cell mobilising agents, were included where administered to treatment and control arms equally. DATA COLLECTION AND ANALYSIS: Two review authors independently screened all references for eligibility, assessed trial quality and extracted data. We undertook a quantitative evaluation of data using fixed-effect meta-analyses. We evaluated heterogeneity using the I² statistic; we explored considerable heterogeneity (I² > 75%) using a random-effects model and subgroup analyses. MAIN RESULTS: We include 23 RCTs involving 1255 participants in this review. Risk of bias was generally low, with the majority of studies reporting appropriate methods of randomisation and blinding, Autologous bone marrow stem cell treatment reduced the incidence of mortality (risk ratio (RR) 0.28, 95% confidence interval (CI) 0.14 to 0.53, P = 0.0001, 8 studies, 494 participants, low quality evidence) and rehospitalisation due to heart failure (RR 0.26, 95% CI 0.07 to 0.94, P = 0.04, 2 studies, 198 participants, low quality evidence) in the long term (≥12 months). The treatment had no clear effect on mortality (RR 0.68, 95% CI 0.32 to 1.41, P = 0.30, 21 studies, 1138 participants, low quality evidence) or rehospitalisation due to heart failure (RR 0.36, 95% CI 0.12 to 1.06, P = 0.06, 4 studies, 236 participants, low quality evidence) in the short term (< 12 months), which is compatible with benefit, no difference or harm. The treatment was also associated with a reduction in left ventricular end systolic volume (LVESV) (mean difference (MD) -14.64 ml, 95% CI -20.88 ml to -8.39 ml, P < 0.00001, 3 studies, 153 participants, moderate quality evidence) and stroke volume index (MD 6.52, 95% CI 1.51 to 11.54, P = 0.01, 2 studies, 62 participants, moderate quality evidence), and an improvement in left ventricular ejection fraction (LVEF) (MD 2.62%, 95% CI 0.50% to 4.73%, P = 0.02, 6 studies, 254 participants, moderate quality evidence), all at long-term follow-up. Overall, we observed a reduction in functional class (New York Heart Association (NYHA) class) in favour of BMSC treatment during short-term follow-up (MD -0.63, 95% CI -1.08 to -0.19, P = 0.005, 11 studies, 486 participants, moderate quality evidence) and long-term follow-up (MD -0.91, 95% CI -1.38 to -0.44, P = 0.0002, 4 studies, 196 participants, moderate quality evidence), as well as a difference in Canadian Cardiovascular Society score in favour of BMSC (MD -0.81, 95% CI -1.55 to -0.07, P = 0.03, 8 studies, 379 participants, moderate quality evidence). Of 19 trials in which adverse events were reported, adverse events relating to the BMSC treatment or procedure occurred in only four individuals. No long-term adverse events were reported. Subgroup analyses conducted for outcomes such as LVEF and NYHA class revealed that (i) route of administration, (ii) baseline LVEF, (iii) cell type, and (iv) clinical condition are important factors that may influence treatment effect. AUTHORS' CONCLUSIONS: This systematic review and meta-analysis found moderate quality evidence that BMSC treatment improves LVEF. Unlike in trials where BMSC were administered following acute myocardial infarction (AMI), we found some evidence for a potential beneficial clinical effect in terms of mortality and performance status in the long term (after at least one year) in people who suffer from chronic IHD and heart failure, although the quality of evidence was low.


Asunto(s)
Insuficiencia Cardíaca/cirugía , Isquemia Miocárdica/cirugía , Trasplante de Células Madre/métodos , Células de la Médula Ósea/citología , Enfermedad Crónica , Insuficiencia Cardíaca/mortalidad , Hospitalización/estadística & datos numéricos , Humanos , Isquemia Miocárdica/mortalidad , Ensayos Clínicos Controlados Aleatorios como Asunto , Trasplante de Células Madre/efectos adversos , Trasplante de Células Madre/mortalidad , Volumen Sistólico/fisiología
11.
Cochrane Database Syst Rev ; (2): CD006536, 2012 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-22336818

RESUMEN

BACKGROUND: Stem cell therapy offers a promising approach to the regeneration of damaged vascular and cardiac tissue after acute myocardial infarction (AMI). This has resulted in multiple randomised controlled trials (RCTs) worldwide. OBJECTIVES: To critically evaluate evidence from RCTs on the effectiveness of adult bone marrow-derived stem cells (BMSC) to treat acute myocardial infarction (AMI). SEARCH METHODS: This Cochrane review is an update of a previous one (published in 2008). MEDLINE (1950 to January 2011), EMBASE (1974 to January 2011), the Cochrane Central Register of Controlled Trials (CENTRAL) (Issue 1, 2011), CINAHL (1982 to January 2011) and the Transfusion Evidence Library (1980 to January 2011) were searched. In addition, several international and ongoing trial databases were searched and handsearching of relevant conference proceedings undertaken to January 2011. SELECTION CRITERIA: RCTs comparing autologous stem/progenitor cells with no autologous stem/progenitor cells in patients diagnosed with AMI were eligible. DATA COLLECTION AND ANALYSIS: Two authors independently screened all references, assessed trial quality and extracted data. Meta-analyses using a random-effects model were conducted and heterogeneity was explored for the primary outcome using sub-group analyses. MAIN RESULTS: Thirty-three RCTs (1765 participants) were eligible for inclusion. Stem/progenitor cell treatment was not associated with statistically significant changes in the incidence of mortality (RR 0.70, 95% CI 0.40 to 1.21) or morbidity (the latter measured by re-infarction, hospital re-admission, restenosis and target vessel revascularisation). A considerably high degree of heterogeneity has been observed among the included trials. In short-term follow up, stem cell treatment was observed to improve left ventricular ejection fraction (LVEF) significantly (WMD 2.87, 95% CI 2.00 to 3.73). This improvement in LVEF was maintained over long-term follow up of 12 to 61 months (WMD 3.75, 95% CI 2.57 to 4.93). With certain measurements and at certain times, stem cell treatment was observed to reduce left ventricular end systolic and end diastolic volumes (LVESV & LVEDV) and infarct size significantly in long-term follow up. There was a positive correlation between mononuclear cell dose infused and the effect on LVEF measured by magnetic resonance imaging. A correlation between timing of stem cell treatment and effect on LVEF measured by left ventricular angiography was also observed. AUTHORS' CONCLUSIONS: Despite the high degree of heterogeneity observed, the results of this systematic review suggest that moderate improvement in global heart function is significant and sustained long-term. However, because mortality rates after successful revascularization of the culprit arteries are very low, larger number of participants would be required to assess the full clinical effect of this treatment. Standardisation of methodology, cell dosing and cell product formulation, timing of cell transplantation and patient selection may also be required in order to reduce the substantial heterogeneity observed among the included studies.


Asunto(s)
Infarto del Miocardio/cirugía , Trasplante de Células Madre/métodos , Humanos , Infarto del Miocardio/mortalidad , Ensayos Clínicos Controlados Aleatorios como Asunto , Recurrencia , Trasplante de Células Madre/efectos adversos , Volumen Sistólico/fisiología
12.
J Clin Med ; 11(11)2022 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-35683592

RESUMEN

Individual patient data (IPD)-based meta-analysis (ACCRUE, meta-analysis of cell-based cardiac studies, NCT01098591) revealed an insufficient effect of intracoronary cell-based therapy in acute myocardial infarction. Patients with ischemic heart failure (iHF) have been treated with reparative cells using percutaneous endocardial, surgical, transvenous or intracoronary cell delivery methods, with variable effects in small randomized or cohort studies. The objective of this meta-analysis was to investigate the safety and efficacy of percutaneous transendocardial cell therapy in patients with iHF. Two investigators extracted the data. Individual patient data (IPD) (n = 8 studies) and publication-based (n = 10 studies) aggregate data were combined for the meta-analysis, including patients (n = 1715) with chronic iHF. The data are reported in accordance with PRISMA guidelines. The primary safety and efficacy endpoints were all-cause mortality and changes in global ejection fraction. The secondary safety and efficacy endpoints were major adverse events, hospitalization and changes in end-diastolic and end-systolic volumes. Post hoc analyses were performed using the IPD of eight studies to find predictive factors for treatment safety and efficacy. Cell therapy was significantly (p < 0.001) in favor of survival, major adverse events and hospitalization during follow-up. A forest plot analysis showed that cell therapy presents a significant benefit of increasing ejection fraction with a mean change of 2.51% (95% CI: 0.48; 4.54) between groups and of significantly decreasing end-systolic volume. The analysis of IPD data showed an improvement in the NYHA and CCS classes. Cell therapy significantly decreased the end-systolic volume in male patients; in patients with diabetes mellitus, hypertension or hyperlipidemia; and in those with previous myocardial infarction and baseline ejection fraction ≤ 45%. The catheter-based transendocardial delivery of regenerative cells proved to be safe and effective for improving mortality and cardiac performance. The greatest benefit was observed in male patients with significant atherosclerotic co-morbidities.

13.
Eur Heart J ; 29(15): 1807-18, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18523058

RESUMEN

AIMS: To provide systematic assessment of the safety and efficacy of autologous bone marrow-derived stem cell (BMSC) transplantation in acute myocardial infarction (AMI) based on clinical evidence. METHODS AND RESULTS: The search strategy included MEDLINE, EMBASE, the Cochrane Library, and Current Controlled Trials Register through to August 2007 for randomized controlled trials of BMSC treatment for AMI. Thirteen trials (14 comparisons) with a total of 811 participants were included. Data were analysed using a random effects model. Overall, stem cell therapy improved left ventricular ejection fraction (LVEF) by 2.99% [95% confidence interval (CI), 1.26-4.72%, P = 0.0007], significantly reduced left ventricular end-systolic volume (LVESV) by 4.74 mL (95% CI, -7.84 to -1.64 mL, P = 0.003), and myocardial lesion area by 3.51% (95% CI, -5.91 to -1.11%, P = 0.004) compared with controls. Subgroup analysis revealed that there was statistical significant difference in LEVF in favour of BMSCs when cells were infused within 7 days following AMI and when the BMSC dose administered was higher than 10(8) BMSCs. In addition, there were trends in favour of benefit for most clinical outcomes examined, although it should be acknowledged that the 95%CI included no significant difference. CONCLUSION: Stem cell treatment for AMI still holds promise. Clinically, these data suggest that improvement over conventional therapy can be achieved. Further, adequately powered trials using optimal dosing, longer term outcome assessments, more reliable, and more patient-centred outcomes are required.


Asunto(s)
Trasplante de Médula Ósea , Infarto del Miocardio/cirugía , Trasplante de Células Madre , Medicina Basada en la Evidencia , Femenino , Insuficiencia Cardíaca/etiología , Humanos , Masculino , Infarto del Miocardio/complicaciones , Ensayos Clínicos Controlados Aleatorios como Asunto , Resultado del Tratamiento
14.
Cochrane Database Syst Rev ; (4): CD006536, 2008 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-18843721

RESUMEN

BACKGROUND: Stem cell therapy offers a promising approach to the regeneration of damaged vascular and cardiac tissue after myocardial infarction (MI). This has resulted in multiple randomised controlled trials (RCTs) worldwide. OBJECTIVES: To critically evaluate evidence from RCTs on the effectiveness of adult bone marrow-derived stem cells (BMSC) to treat acute MI. SEARCH STRATEGY: MEDLINE (1950 to August 2007), EMBASE (1974 to August 2007), The Cochrane Library (Issue 3 2007), and CINAHL (1982 to August 2007) were searched. In addition LILACS, KOREAMED, INMED, Current Controlled Trials Register, the UK National Research Register and other handsearching was undertaken to August 2007. SELECTION CRITERIA: RCTs comparing autologous stem/progenitor cells with no autologous stem/progenitor cells in patients diagnosed with acute myocardial infarction (AMI) were eligible. DATA COLLECTION AND ANALYSIS: Two reviewers independently screened all references, assessed trial quality and extracted data. Meta-analyses using a random-effects model were conducted and heterogeneity was explored using sub-group analyses. MAIN RESULTS: Thirteen RCTs (811 participants) were included. There were insufficient events on clinical outcomes like mortality to draw clear conclusions. Stem/progenitor cell treatment does not appear to be associated with an increase in adverse events but again the data do not allow clear conclusions. Left ventricular ejection fraction (LVEF) was the outcome with most results and there was marked heterogeneity between trials. There was however a consistent pattern indicating that BMSC treatment generally improves short-term LVEF, with similar trends for left ventricular end systolic and end diastolic volumes (LVESV and LVEDV), infarct size or cardiac wall motion. There was a positive correlation between cell dose infused and the effect on LVEF measured by magnetic resonance imaging. AUTHORS' CONCLUSIONS: The results of this systematic review suggest that there is little evidence to assess the clinical effects of this treatment. Larger trials using optimal dosing and more reliable, patient-centred outcomes are required. Several trials are ongoing but is unclear whether these will overcome the limitations of the current evidence base.


Asunto(s)
Infarto del Miocardio/cirugía , Trasplante de Células Madre/métodos , Humanos , Ensayos Clínicos Controlados Aleatorios como Asunto
15.
Sci Rep ; 7(1): 11478, 2017 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-28904391

RESUMEN

Bactrocera oleae (Diptera: Tephritidae) remains a major pest of olive fruit production worldwide. Current pest management programs largely depend on chemical insecticides, resulting in high economic and environmental costs. Alternative pest control approaches are therefore highly desirable. We have created a conditional female-specific self-limiting strain of B. oleae (OX3097D-Bol) that could be applied for sustainable pest control. OX3097D-Bol olive fly carries a fluorescent marker (DsRed2) for identification and a self-limiting genetic trait that is repressed by tetracycline. In the absence of tetracycline, the tetracycline transactivator (tTAV) accumulates, resulting in female death at larvae and early pupal stages. The aim of this study was to evaluate the impact of genetically engineered OX3097D-Bol olive fly on three non-target organisms that either predate or parasitize olive flies, one from the guild of parasitoids (Psyttalia concolor) and two from the guild of predators (Pardosa spider species and the rove beetle Aleochara bilineata). No significant negative effect was observed on life history parameters, mortality and reproductive capacity of the non-target organisms studied. These results suggest that potential exposure to DsRed2 and tTAV gene products (e.g. mRNA and encoded proteins) would have a negligible impact on on-target organisms in the guilds or predators and parasitoids.


Asunto(s)
Animales Modificados Genéticamente , Interacciones Huésped-Parásitos , Tephritidae/genética , Animales , Escarabajos , Femenino , Larva , Olea , Control Biológico de Vectores , Conducta Predatoria , Arañas
16.
Sci Rep ; 7(1): 6312, 2017 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-28740084

RESUMEN

The Cardiomyopathy-associated gene 5 (Cmya5) encodes myospryn, a large tripartite motif (TRIM)-related protein found predominantly in cardiac and skeletal muscle. Cmya5 is an expression biomarker for a number of diseases affecting striated muscle and may also be a schizophrenia risk gene. To further understand the function of myospryn in striated muscle, we searched for additional myospryn paralogs. Here we identify a novel muscle-expressed TRIM-related protein minispryn, encoded by Fsd2, that has extensive sequence similarity with the C-terminus of myospryn. Cmya5 and Fsd2 appear to have originated by a chromosomal duplication and are found within evolutionarily-conserved gene clusters on different chromosomes. Using immunoaffinity purification and mass spectrometry we show that minispryn co-purifies with myospryn and the major cardiac ryanodine receptor (RyR2) from heart. Accordingly, myospryn, minispryn and RyR2 co-localise at the junctional sarcoplasmic reticulum of isolated cardiomyocytes. Myospryn redistributes RyR2 into clusters when co-expressed in heterologous cells whereas minispryn lacks this activity. Together these data suggest a novel role for the myospryn complex in the assembly of ryanodine receptor clusters in striated muscle.


Asunto(s)
Proteínas Portadoras/genética , Clonación Molecular/métodos , Proteínas Musculares/genética , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Animales , Células COS , Proteínas Portadoras/metabolismo , Chlorocebus aethiops , Cromatografía de Afinidad , Duplicación Cromosómica , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular , Espectrometría de Masas , Ratones , Proteínas Musculares/metabolismo , Retículo Sarcoplasmático/metabolismo
17.
Stem Cells Transl Med ; 6(5): 1399-1411, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28205406

RESUMEN

Cardiosphere-derived cell (CDC) infusion into damaged myocardium has shown some reparative effect; this could be improved by better selection of patients and cell subtype. CDCs isolated from patients with ischemic heart disease are able to support vessel formation in vitro but this ability varies between patients. The primary aim of our study was to investigate whether the vascular supportive function of CDCs impacts on their therapeutic potential, with the goal of improving patient stratification. A subgroup of patients produced CDCs which did not efficiently support vessel formation (poor supporter CDCs), had reduced levels of proliferation and increased senescence, despite them being isolated in the same manner and having a similar immunophenotype to CDCs able to support vessel formation. In a rodent model of myocardial infarction, poor supporter CDCs had a limited reparative effect when compared to CDCs which had efficiently supported vessel formation in vitro. This work suggests that not all patients provide cells which are suitable for cell therapy. Assessing the vascular supportive function of cells could be used to stratify which patients will truly benefit from cell therapy and those who would be better suited to an allogeneic transplant or regenerative preconditioning of their cells in a precision medicine fashion. This could reduce costs, culture times and improve clinical outcomes and patient prognosis. Stem Cells Translational Medicine 2017;6:1399-1411.


Asunto(s)
Enfermedad de la Arteria Coronaria/terapia , Isquemia Miocárdica/terapia , Células Madre/citología , Apoptosis/fisiología , Western Blotting , Movimiento Celular/fisiología , Citometría de Flujo , Humanos , Inmunohistoquímica
18.
J Neurosci ; 22(23): 10302-12, 2002 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-12451130

RESUMEN

Parkinson's disease (PD) is a neurodegenerative disorder characterized by the selective loss of dopaminergic neurons in the substantia nigra. This loss leads to complete dopamine depletion in the striatum and severe motor impairment. It has been demonstrated previously that a lentiviral vector system based on equine infectious anemia virus (EIAV) gives rise to highly efficient and sustained transduction of neurons in the rat brain. Therefore, a dopamine replacement strategy using EIAV has been investigated as a treatment in the 6-hydroxydopamine (6-OHDA) animal model of PD. A self-inactivating EIAV minimal lentiviral vector that expresses tyrosine hydroxylase (TH), aromatic amino acid dopa decarboxylase (AADC), and GTP cyclohydrolase 1 (CH1) in a single transcription unit has been generated. In cultured striatal neurons transduced with this vector, TH, AADC, and CH1 proteins can all be detected. After stereotactic delivery into the dopamine-denervated striatum of the 6-OHDA-lesioned rat, sustained expression of each enzyme and effective production of catecholamines were detected, resulting in significant reduction of apomorphine-induced motor asymmetry compared with control animals (p < 0.003). Expression of each enzyme in the striatum was observed for up to 5 months after injection. These data indicate that the delivery of three catecholaminergic synthetic enzymes by a single lentiviral vector can achieve functional improvement and thus open the potential for the use of this vector for gene therapy of late-stage PD patients.


Asunto(s)
Cuerpo Estriado/efectos de los fármacos , Dopamina/biosíntesis , Vectores Genéticos/administración & dosificación , Trastornos Parkinsonianos/terapia , Animales , Descarboxilasas de Aminoácido-L-Aromático/administración & dosificación , Descarboxilasas de Aminoácido-L-Aromático/biosíntesis , Descarboxilasas de Aminoácido-L-Aromático/genética , Catecolaminas/metabolismo , Células Cultivadas , Cuerpo Estriado/patología , Cuerpo Estriado/fisiopatología , Modelos Animales de Enfermedad , GTP Ciclohidrolasa/administración & dosificación , GTP Ciclohidrolasa/biosíntesis , GTP Ciclohidrolasa/genética , Expresión Génica/efectos de los fármacos , Técnicas de Transferencia de Gen , Genes/genética , Terapia Genética/métodos , Vectores Genéticos/genética , Humanos , Riñón/citología , Riñón/metabolismo , Lentivirus/genética , Masculino , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Oxidopamina , Trastornos Parkinsonianos/inducido químicamente , Trastornos Parkinsonianos/fisiopatología , Ratas , Ratas Wistar , Recuperación de la Función/efectos de los fármacos , Transgenes , Resultado del Tratamiento , Tirosina 3-Monooxigenasa/administración & dosificación , Tirosina 3-Monooxigenasa/biosíntesis , Tirosina 3-Monooxigenasa/genética
19.
Trends Pharmacol Sci ; 24(4): 178-83, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12707004

RESUMEN

Glycosylation is the most frequent modification of proteins and is important for many ligand-receptor interactions. Recently, defects in protein glycosylation have been linked to several forms of congenital muscular dystrophy that are frequently associated with brain abnormalities. Muscle-eye-brain disease and Walker-Warburg syndrome are caused by mutations in enzymes involved in O-mannosylation, whereas Fukuyama congenital muscular dystrophy and congenital muscular dystrophy type 1C are caused by mutations in genes that encode putative glycosyltransferases. The common factor in these disorders is defective processing and maturation of a protein called alpha-dystroglycan. This is thought to disrupt the link between alpha-dystroglycan and components of the extracellular matrix, and result in muscle disease and, in many cases, a neuronal-migration disorder.


Asunto(s)
Distrofias Musculares/congénito , Distrofias Musculares/metabolismo , Animales , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Distroglicanos , Terapia Genética , Glicosilación , Glicosiltransferasas/genética , Humanos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Distrofias Musculares/terapia , Mutación
20.
Trends Cardiovasc Med ; 12(5): 224-8, 2002 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12161077

RESUMEN

Intermediate filament (IF) proteins and the dystrophin-associated protein complex (DPC) play important roles in cardiac and skeletal muscle. Both systems are mutated in several different forms of inherited muscular dystrophy and cardiomyopathy. Recently two articles have been published that propose a physical link between the DPC and the IF network in muscle. Two novel IF proteins, syncoilin and desmuslin, have been identified as binding partners for the dystrophin-associated protein, alpha-dystrobrevin, in muscle. These novel interactions suggest that alpha-dystrobrevin may tether the IF protein network to the DPC. Mice lacking alpha-dystrobrevin develop muscular dystrophy without perturbing the assembly of the DPC at the muscle membrane, suggesting the involvement of other non-DPC proteins in the disease. The interaction between the DPC and the IF network may be disrupted in patients with Duchenne muscular dystrophy and in mice lacking alpha-dystrobrevin.


Asunto(s)
Proteínas del Citoesqueleto/fisiología , Distrofina/fisiología , Glicoproteínas de Membrana/fisiología , Distrofias Musculares/fisiopatología , Distroglicanos , Humanos , Proteínas de Filamentos Intermediarios/fisiología , Proteínas Musculares/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA