Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Eur Heart J ; 42(3): 257-265, 2021 01 20.
Artigo em Inglês | MEDLINE | ID: mdl-33241418

RESUMO

AIMS: Somatic mutations of the epigenetic regulators DNMT3A and TET2 causing clonal expansion of haematopoietic cells (clonal haematopoiesis; CH) were shown to be associated with poor prognosis in chronic ischaemic heart failure (CHF). The aim of our analysis was to define a threshold of variant allele frequency (VAF) for the prognostic significance of CH in CHF. METHODS AND RESULTS: We analysed bone marrow and peripheral blood-derived cells from 419 patients with CHF by error-corrected amplicon sequencing. Cut-off VAFs were optimized by maximizing sensitivity plus specificity from a time-dependent receiver operating characteristic (ROC) curve analysis from censored data. 56.2% of patients were carriers of a DNMT3A- (N = 173) or a TET2- (N = 113) mutation with a VAF >0.5%, with 59 patients harbouring mutations in both genes. Survival ROC analyses revealed an optimized cut-off value of 0.73% for TET2- and 1.15% for DNMT3A-CH-driver mutations. Five-year-mortality was 18% in patients without any detected DNMT3A- or TET2 mutation (VAF < 0.5%), 29% with only one DNMT3A- or TET2-CH-driver mutations above the respective cut-off level and 42% in patients harbouring both DNMT3A- and TET2-CH-driver mutations above the respective cut-off levels. In carriers of a DNMT3A mutation with VAF ≥ 1.15%, 5-year mortality was 31%, compared with 18% mortality in those with VAF < 1.15% (P = 0.048). Likewise, in patients with TET2 mutations, 5-year mortality was 32% with VAF ≥ 0.73%, compared with 19% mortality with VAF < 0.73% (P = 0.029). CONCLUSION: The present study defines novel threshold levels for clone size caused by acquired somatic mutations in the CH-driver genes DNMT3A and TET2 that are associated with worse outcome in patients with CHF.


Assuntos
Hematopoiese Clonal , Insuficiência Cardíaca , Células Clonais , DNA (Citosina-5-)-Metiltransferases/genética , DNA Metiltransferase 3A , Proteínas de Ligação a DNA/genética , Dioxigenases , Humanos , Mutação , Prognóstico , Proteínas Proto-Oncogênicas/genética
2.
Circ Res ; 120(12): 1938-1946, 2017 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-28351842

RESUMO

RATIONALE: Cell-based therapies are a promising option in patients with chronic postinfarction heart failure (ischemic cardiomyopathy [ICM]). However, the responses after intracoronary infusion of autologous bone marrow-derived mononuclear cells (BMCs) are heterogeneous, which may be related to impaired cell retention in patients with ICM. Ischemic injury is associated with upregulation of prototypical chemoattractant cytokines mediating retention and homing of circulating cells. The development of ultrasensitive tests to measure high-sensitive troponin T (hs-TnT) serum levels revealed the presence of ongoing minute myocardial injury even in patients with stable ICM. OBJECTIVE: To test the hypothesis that serum levels of hs-TnT correlate with cell retention and determine the response to intracoronary BMC application in patients with ICM. METHODS AND RESULTS: About 157 patients with stable ICM and no substantial impairment of kidney function received intracoronary BMC administration. Immediately prior to cell application, hs-TnT levels to measure myocardial injury and NT-proBNP levels as marker of left ventricular wall stress were determined. Patients with elevated hs-TnT were older and had more severe heart failure. Importantly, only patients with elevated baseline hs-TnT≥15.19 pg/mL (upper tertile) demonstrated a significant (P=0.04) reduction in NT-proBNP serum levels (-250 [-1465; 33] pg/mL; relative reduction -24%) 4 months after BMC administration, whereas NT-proBNP levels remained unchanged in patients in the 2 lower hs-TnT tertiles. The absolute decrease in NT-proBNP at 4 months was inversely correlated with baseline hs-TnT (r=-0.27, P=0.001). Finally, retention of intracoronarily infused, 111Indium-labeled cells within the heart was closely associated with hs-TnT levels in patients with chronic ischemic heart failure (P=0.0008, n=10, triple measurements). CONCLUSIONS: The extent of ongoing myocardial injury as measured by serum levels of hs-TnT predicts the reduction of NT-proBNP serum levels at 4 months after intracoronary BMC administration in patients with ICM, suggesting that the beneficial effects of BMC application on LV remodeling and wall stress are confined to patients with ongoing minute myocardial injury. CLINICAL TRIAL REGISTRATION: URL: www.clinicaltrials.gov. Unique identifier: NCT00962364.


Assuntos
Transplante de Medula Óssea/tendências , Vasos Coronários/metabolismo , Insuficiência Cardíaca/sangue , Infarto do Miocárdio/sangue , Sistema de Registros , Troponina T/sangue , Idoso , Biomarcadores/sangue , Células da Medula Óssea/metabolismo , Doença Crônica , Feminino , Seguimentos , Insuficiência Cardíaca/diagnóstico , Insuficiência Cardíaca/terapia , Humanos , Infusões Intra-Arteriais , Masculino , Pessoa de Meia-Idade , Infarto do Miocárdio/diagnóstico , Infarto do Miocárdio/terapia , Miocárdio/metabolismo , Peptídeo Natriurético Encefálico/sangue , Fragmentos de Peptídeos/sangue , Projetos Piloto , Valor Preditivo dos Testes
3.
Stem Cells ; 34(8): 2236-48, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27145479

RESUMO

Administration of bone marrow-derived mononuclear cells (BMC) may increase cardiac function after myocardial ischemia. However, the functional capacity of BMC derived from chronic heart failure (CHF) patients is significantly impaired. As modulation of the energy metabolism allows cells to match the divergent demands of the environment, we examined the regulation of energy metabolism in BMC from patients and healthy controls (HC). The glycolytic capacity of CHF-derived BMC is reduced compared to HC, whereas BMC of metabolically activated bone marrow after acute myocardial infarction reveal increased metabolism. The correlation of metabolic pathways with the functional activity of cells indicates an influence of metabolism on cell function. Reducing glycolysis without profoundly affecting ATP-production reversibly reduces invasion as well as colony forming capacity and abolishes proliferation of CD34(+) CD38(-) lin(-) hematopoietic stem and progenitor cells (HSPC). Ex vivo inhibition of glycolysis further reduced the pro-angiogenic activity of transplanted cells in a hind limb ischemia model in vivo. In contrast, inhibition of respiration, without affecting total ATP production, leads to a compensatory increase in glycolytic capacity correlating with increased colony forming capacity. Isolated CD34(+) , CXCR4(+) , and CD14(+) cells showed higher glycolytic activity compared to their negative counterparts. Metabolic activity was profoundly modulated by the composition of media used to store or culture BMC. This study provides first evidence that metabolic alterations influence the functional activity of human HSPC and BMC independent of ATP production. Changing the balance between respiration and glycolysis might be useful to improve patient-derived cells for clinical cardiac cell therapy. Stem Cells 2016;34:2236-2248.


Assuntos
Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Insuficiência Cardíaca/terapia , Isquemia Miocárdica/terapia , Animais , Respiração Celular , Ensaio de Unidades Formadoras de Colônias , Meios de Cultura , Glicólise , Insuficiência Cardíaca/patologia , Membro Posterior/irrigação sanguínea , Membro Posterior/patologia , Humanos , Metabolômica , Camundongos Nus , MicroRNAs/metabolismo , Isquemia Miocárdica/patologia , Neovascularização Fisiológica , Fator de Transcrição STAT5/metabolismo
4.
Neurocrit Care ; 27(1): 68-74, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28054291

RESUMO

BACKGROUND: The neurological prognosis of patients after cardiopulmonary resuscitation (CPR) is difficult to assess. GFAP is an astrocytic intermediate filament protein released into bloodstream in case of cell death. We performed a prospective study aiming to compare the predictive potential of GFAP after resuscitation to the more widely used biomarker neuron-specific enolase (NSE). METHODS: One hundred patients were included at 48 h (tolerance interval ±12 h) after cardiac arrest. A serum sample was collected immediately after study inclusion. We determined serum levels of GFAP and NSE by means of immunoassays. Primary outcome was the modified Glasgow outcome scale at 4 weeks. Values below four were considered as a poor functional outcome. RESULTS: Median GFAP levels in poor outcome (n = 61) and good outcome (n = 39) patients were 0.03 µg/L (interquartile range 0.01-0.07 µg/L) and 0.02 µg/L (0.01-0.03 µg/L; p = 0.014), respectively. GFAP revealed a sensitivity of 60.7% and a specificity of 66.7% to predict a poor functional outcome. All patients having a GFAP level >0.08 µg/L had a poor functional outcome. For NSE, sensitivity was 44.3% and specificity was 100.0% for predicting a poor outcome. Multivariate regression analysis revealed GFAP, NSE, and the Karnofsky index to be independent predictors of outcome. CONCLUSIONS: The release patterns of GFAP and NSE after CPR show differences. GFAP levels above 0.08 µg/L were associated with a poor outcome in all cases, and patients with strongly elevated values (>3 µg/L) consistently had severe brain damage on brain imaging. Both biomarkers independently contribute to outcome prediction after CPR.


Assuntos
Reanimação Cardiopulmonar , Proteína Glial Fibrilar Ácida/sangue , Parada Cardíaca/terapia , Avaliação de Resultados em Cuidados de Saúde , Idoso , Idoso de 80 Anos ou mais , Biomarcadores/sangue , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos
6.
Circ Res ; 111(7): 854-62, 2012 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-22821930

RESUMO

RATIONALE: Cell therapy is a promising option for the treatment of acute or chronic myocardial ischemia. The intracoronary infusion of cells imposes the potential risk of cell clotting, which may be prevented by the addition of anticoagulants. However, a comprehensive analysis of the effects of anticoagulants on the function of the cells is missing. OBJECTIVE: Here, we investigated the effects of heparin and the thrombin inhibitor bivalirudin on bone marrow-derived mononuclear cell (BMC) functional activity and homing capacity. METHODS AND RESULTS: Heparin, but not bivalirudin profoundly and dose-dependently inhibited basal and stromal cell-derived factor 1 (SDF-1)-induced BMC migration. Incubation of BMCs with 20 U/mL heparin for 30 minutes abrogated SDF-1-induced BMC invasion (16±8% of control; P<0.01), whereas no effects on apoptosis or colony formation were observed (80±33% and 100±44% of control, respectively). Pretreatment of BMCs with heparin significantly reduced the homing of the injected cells in a mouse ear-wound model (69±10% of control; P<0.05). In contrast, bivalirudin did not inhibit in vivo homing of BMCs. Mechanistically, heparin binds to both, the chemoattractant SDF-1 and its receptor, chemokine receptor 4 (CXCR4), blocking CXCR4 internalization as well as SDF-1/CXCR4 signaling after SDF-1 stimulation. CONCLUSIONS: Heparin blocks SDF-1/CXCR4 signaling by binding to the ligand as well as the receptor, thereby interfering with migration and homing of BMCs. In contrast, the thrombin inhibitor bivalirudin did not interfere with BMC homing or SDF-1/CXCR4 signaling. These findings suggest that bivalirudin but not heparin might be recommended as an anticoagulant for intracoronary infusion of BMCs for cell therapy after cardiac ischemia.


Assuntos
Células da Medula Óssea/citologia , Terapia Baseada em Transplante de Células e Tecidos , Quimiocina CXCL12/efeitos dos fármacos , Heparina/farmacologia , Leucócitos Mononucleares/efeitos dos fármacos , Infarto do Miocárdio/terapia , Receptores CXCR4/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Animais , Anticoagulantes/farmacologia , Antitrombinas/farmacologia , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Células Cultivadas , Quimiocina CXCL12/fisiologia , Modelos Animais de Doenças , Feminino , Hirudinas/farmacologia , Humanos , Técnicas In Vitro , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/fisiologia , Camundongos , Camundongos Endogâmicos , Fragmentos de Peptídeos/farmacologia , Receptores CXCR4/fisiologia , Proteínas Recombinantes/farmacologia , Transdução de Sinais/fisiologia
7.
Arterioscler Thromb Vasc Biol ; 33(8): 1739-46, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23864723

RESUMO

MicroRNAs are small noncoding RNAs that posttranscriptionally control gene expression by targeting mRNAs. Distinct microRNAs regulate stem and progenitor cell functions, thereby modulating cell survival and homing or controlling differentiation and maturation. Experimental studies additionally show that microRNAs regulate endogenous repair and might potentially be useful to enhance the regeneration of the heart. This review summarizes the current studies that address the use of microRNAs to either improve cellular therapies or that might be targeted for enhancing endogenous tissue repair and regeneration after myocardial infarction.


Assuntos
Cardiopatias/genética , Cardiopatias/terapia , MicroRNAs/fisiologia , Medicina Regenerativa/métodos , Células-Tronco/fisiologia , Animais , Mobilização de Células-Tronco Hematopoéticas , Humanos , Processamento Pós-Transcricional do RNA/fisiologia
8.
Circulation ; 125(14): 1765-73, S1-7, 2012 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-22403243

RESUMO

BACKGROUND: Cell therapy with bone marrow-derived mononuclear cells (BMCs) can improve recovery of cardiac function after ischemia; however, the molecular mechanisms are not yet fully understood. MicroRNAs (miRNAs) are key regulators of gene expression and modulate the pathophysiology of cardiovascular diseases. METHODS AND RESULTS: We demonstrated that intramyocardial delivery of BMCs in infarcted mice regulates the expression of cardiac miRNAs and significantly downregulates the proapoptotic miR-34a. In vitro studies confirmed that the supernatant of BMC inhibited the expression of H(2)O(2)-induced miR-34a and cardiomyocytes apoptosis. These effects were blocked by neutralizing antibodies directed against insulin-like growth factor-1 (IGF-1). Indeed, IGF-1 significantly inhibited H(2)O(2)-induced miR-34a expression, and miR-34a overexpression abolished the antiapoptotic effect of IGF-1. Likewise, inhibition of IGF-1 signaling in vivo abolished the BMC-mediated inhibition of miR-34 expression and the protective effect on cardiac function and increased apoptosis and cardiac fibrosis. IGF-1 specifically blocked the expression of the precursor and the mature miR-34a, but did not interfere with the transcription of the primary miR-34a demonstrating that IGF-1 blocks the processing of miR-34a. CONCLUSIONS: Together, our data demonstrate that the paracrine regulation of cardiac miRNAs by transplanted BMCs contributes to the protective effects of cell therapy. BMCs release IGF-1, which inhibits the processing of miR-34a, thereby blocking cardiomyocyte apoptosis.


Assuntos
Células da Medula Óssea/fisiologia , Transplante de Medula Óssea , MicroRNAs/fisiologia , Infarto do Miocárdio/terapia , Miócitos Cardíacos/metabolismo , Apoptose/efeitos dos fármacos , Células Cultivadas , Humanos , Peróxido de Hidrogênio/farmacologia , Fator de Crescimento Insulin-Like I/fisiologia , Masculino
9.
Basic Res Cardiol ; 108(1): 313, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23184393

RESUMO

Bone marrow (BM) derived stem and progenitor cells contribute to cardiovascular homeostasis and are affected by cardiovascular risk factors. We devised a clinical data-driven approach to test candidate stem cell mobilizing mechanisms in pre-clinical models. We found that PB and BM CD34+ cell counts were directly correlated, and that most circulating CD34+ cells were viable, non-proliferating and derived from the BM. Thus, we analyzed PB and BM CD34+ cell levels as a two-compartment model in 72 patients with or without cardiovascular disease. Self-organizing maps showed that disturbed compartmentalization of CD34+ cells was associated with aging and cardiovascular risk factors especially diabetes. High activity of DPP-4, a regulator of the mobilizing chemokine SDF-1α, was associated with altered stem cell compartmentalization. For validation of these findings, we assessed the role of DPP-4 in the BM mobilization response of diabetic rats. Diabetes differentially affected DPP-4 activity in PB and BM and impaired stem/progenitor cell mobilization after ischemia or G-CSF administration. DPP-4 activity in the BM was required for the mobilizing effect of G-CSF, while in PB it blunted ischemia-induced mobilization. Indeed, DPP-4 deficiency restored ischemia (but not G-CSF)-induced stem cell mobilization and improved vascular recovery in diabetic animals. In conclusion, the analysis of stem cell compartmentalization in humans led us to discover mechanisms of BM unresponsiveness in diabetes determined by tissue-specific DPP-4 dysregulation.


Assuntos
Doenças Cardiovasculares/etiologia , Compartimento Celular , Diabetes Mellitus Experimental/patologia , Dipeptidil Peptidase 4/fisiologia , Mobilização de Células-Tronco Hematopoéticas , Células-Tronco/fisiologia , Animais , Antígenos CD34/análise , Células da Medula Óssea/fisiologia , Quimiocina CXCL12/metabolismo , Fator Estimulador de Colônias de Granulócitos/farmacologia , Humanos , Pessoa de Meia-Idade , Neovascularização Fisiológica , Ratos , Ratos Endogâmicos F344 , Fatores de Risco
10.
Circ Res ; 108(9): 1112-21, 2011 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-21393578

RESUMO

RATIONALE: Acquisition of a procalcific phenotype by resident or circulating cells is important for calcification of atherosclerotic plaques, which is common in diabetes. OBJECTIVE: We aim to identify and characterize circulating calcifying cells, and to delineate a pathophysiological role for these cells in type 2 diabetes. METHODS AND RESULTS: We demonstrate for the first time that a distinct subpopulation of circulating cells expressing osteocalcin and bone alkaline phosphatase (OC(+)BAP(+)) has procalcific activity in vitro and in vivo. The study of naïve patients with chronic myeloid leukemia indicated that OC(+)BAP(+) cells have a myeloid origin. Myeloid calcifying OC(+)BAP(+) cells (MCCs) could be differentiated from peripheral blood mononuclear cells, and generation of MCCs was closely associated with expression of the osteogenic transcription factor Runx2. In gender-mismatched bone marrow-transplanted humans, circulating MCCs had a much longer half-life compared with OC(-)BAP(-) cells, suggesting they belong to a stable cell repertoire. The percentage of MCCs was higher in peripheral blood and bone marrow of type 2 diabetic patients compared with controls but was lowered toward normal levels by optimization of glycemic control. Furthermore, diabetic carotid endoarterectomy specimens showed higher degree of calcification and amounts of cells expressing OC and BAP in the α-smooth muscle actin-negative areas surrounding calcified nodules, where CD68(+) macrophages colocalize. High glucose increased calcification by MCCs in vitro, and hypoxia may regulate MCC generation in vitro and in vivo. CONCLUSIONS: These data identify a novel type of blood-derived procalcific cells potentially involved in atherosclerotic calcification of diabetic patients.


Assuntos
Calcinose/patologia , Doenças das Artérias Carótidas/patologia , Diabetes Mellitus Tipo 2/patologia , Angiopatias Diabéticas/patologia , Células Mieloides/patologia , Fosfatase Alcalina/metabolismo , Animais , Biomarcadores/metabolismo , Transplante Ósseo , Doenças das Artérias Carótidas/cirurgia , Linhagem da Célula/fisiologia , Células Cultivadas , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Diabetes Mellitus Tipo 2/tratamento farmacológico , Endarterectomia das Carótidas , Feminino , Humanos , Hiperglicemia/tratamento farmacológico , Hiperglicemia/metabolismo , Hipoglicemiantes/uso terapêutico , Hipóxia/patologia , Insulina/uso terapêutico , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Leucemia Mielogênica Crônica BCR-ABL Positiva/terapia , Masculino , Camundongos , Camundongos Nus , Células Mieloides/metabolismo , Osteocalcina/metabolismo
11.
JAMA ; 309(15): 1622-31, 2013 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-23592107

RESUMO

IMPORTANCE: The modest effects of clinical studies using intracoronary administration of autologous bone marrow-derived mononuclear cells (BMCs) in patients with chronic postinfarction heart failure may be attributed to impaired homing of BMCs to the target area. Extracorporeal shock wave treatment has been experimentally shown to increase homing factors in the target tissue, resulting in enhanced retention of applied BMCs. OBJECTIVE: To test the hypothesis that targeted cardiac shock wave pretreatment with subsequent application of BMCs improves recovery of left ventricular ejection fraction (LVEF) in patients with chronic heart failure. DESIGN, SETTING, AND PARTICIPANTS: The CELLWAVE double-blind, randomized, placebo-controlled trial conducted among patients with chronic heart failure treated at Goethe University Frankfurt, Germany, between 2006 and 2011. INTERVENTIONS: Single-blind low-dose (n = 42), high-dose (n = 40), or placebo (n = 21) shock wave pretreatment targeted to the left ventricular anterior wall. Twenty-four hours later, patients receiving shock wave pretreatment were randomized to receive double-blind intracoronary infusion of BMCs or placebo, and patients receiving placebo shock wave received intracoronary infusion of BMCs. MAIN OUTCOMES AND MEASURES: Primary end point was change in LVEF from baseline to 4 months in the pooled groups shock wave + placebo infusion vs shock wave + BMCs; secondary end points included regional left ventricular function assessed by magnetic resonance imaging and clinical events. RESULTS: The primary end point was significantly improved in the shock wave + BMCs group (absolute change in LVEF, 3.2% [95% CI, 2.0% to 4.4%]), compared with the shock wave + placebo infusion group (1.0% [95% CI, -0.3% to 2.2%]) (P = .02). Regional wall thickening improved significantly in the shock wave + BMCs group (3.6% [95% CI, 2.0% to 5.2%]) but not in the shock wave + placebo infusion group (0.5% [95% CI, -1.2% to 2.1%]) (P = .01). Overall occurrence of major adverse cardiac events was significantly less frequent in the shock wave + BMCs group (n = 32 events) compared with the placebo shock wave + BMCs (n = 18) and shock wave + placebo infusion (n = 61) groups (hazard ratio, 0.58 [95% CI, 0.40-0.85]; P = .02). CONCLUSIONS AND RELEVANCE: Among patients with postinfarction chronic heart failure, shock wave-facilitated intracoronary administration of BMCs vs shock wave treatment alone resulted in a significant, albeit modest, improvement in LVEF at 4 months. Determining whether the increase in contractile function will translate into improved clinical outcomes requires confirmation in larger clinical end point trials. TRIAL REGISTRATION: clinicaltrials.gov Identifier: NCT00326989.


Assuntos
Transplante de Medula Óssea/métodos , Insuficiência Cardíaca/terapia , Ondas de Choque de Alta Energia/uso terapêutico , Idoso , Terapia Combinada , Angiografia Coronária , Método Duplo-Cego , Feminino , Insuficiência Cardíaca/etiologia , Humanos , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Infarto do Miocárdio/complicações , Método Simples-Cego , Volume Sistólico , Transplante Autólogo , Resultado do Tratamento , Disfunção Ventricular Esquerda
12.
Eur Heart J ; 32(3): 371-7, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20494899

RESUMO

AIMS: Coronary artery disease (CAD) patients have less circulating proangiogenic cells (PACs), formerly known as endothelial progenitor cells, which exhibit impaired neovascularization properties. Inverse correlations were also found between PAC function and risk factors like age. Krüppel-like factor 2 (KLF2) is expressed by mature endothelial cells (ECs), is induced by both shear stress and statins, and provokes endothelial functional differentiation. The aim of this study is to identify whether KLF2 can reverse negative effects of ageing on PAC function. METHODS AND RESULTS: We describe that progenitor cells in the bone marrow and PACs also express KLF2 at a comparable level to mature ECs and that senescence decreases KLF2 levels. To study the effects of ageing on KLF2 levels, we compared progenitor cells of 4 weeks and 16- to 18-month-old C57BL/6 mice. In addition to the three-fold reduction of circulating Sca1(+)/c-Kit(+)/Lin(-) progenitor cells and the 15% reduction of Sca1(+)/Flk1(+) endothelial-committed progenitor cells, the spleen-derived PACs and bone marrow-derived progenitor cells isolated from aged mice showed a lower level of KLF2 when compared with young mice. Lentiviral overexpression of KLF2 increased human PAC numbers and endothelial nitric oxide synthase expression by 60% during in vitro culture. Endothelial lineage-specific KLF2 overexpression in aged bone marrow-derived mononuclear cells strongly augments neovascularization in vivo in a murine hind-limb ischaemia model. CONCLUSION: These results imply that KLF2 is an attractive novel target to rejuvenate PACs before autologous administration to CAD patients.


Assuntos
Senescência Celular/fisiologia , Doença da Artéria Coronariana/fisiopatologia , Células Endoteliais/citologia , Endotélio Vascular/citologia , Fatores de Transcrição Kruppel-Like/metabolismo , Células-Tronco/citologia , Animais , Circulação Colateral/fisiologia , Membro Posterior/irrigação sanguínea , Isquemia/fisiopatologia , Leucócitos Mononucleares/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Fisiológica/fisiologia
13.
Circulation ; 121(18): 2001-11, 2010 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-20421519

RESUMO

BACKGROUND: Cell therapy is a promising option to improve functional recovery after ischemia. Several subsets of bone marrow-derived cells were shown to reduce infarct size and increase ejection fraction in experimental models of ischemia. The mechanisms underlying the functional improvement are diverse and have been shown to include paracrine effects of the injected cells, as well as a variable degree of differentiation to endothelial cells, pericytes, smooth muscle, and cardiac muscle. METHODS AND RESULTS: To elucidate the true nature of such plasticity and contribution to recovery, we engineered vectors that encoded inducible suicide genes under the control of endothelium (endothelial nitric oxide synthase)-, smooth muscle (SM22alpha)-, and cardiomyocyte (alpha-MHC)-specific promoters, thereby allowing selective depletion of the individual cell lineage acquired by the transplanted undifferentiated bone marrow-derived cells. Lentivirally delivered thymidine kinase, which converts the prodrug ganciclovir into a cytotoxic agent, was used to selectively eliminate cells 2 weeks after transplantation of bone marrow mononuclear cells in an acute myocardial infarction model. We demonstrate that elimination of transplanted endothelium-committed or SM22alpha-expressing cells, but not cardiac-committed cells, induced a significant deterioration of ejection fraction. Moreover, elimination of endothelial nitric oxide synthase-expressing cells 2 weeks after injection reduced capillary and arteriole density. CONCLUSIONS: This study demonstrates that elimination of bone marrow mononuclear cells reexpressing endothelial nitric oxide synthase particularly induced a deterioration of cardiac function, which indicates a functional contribution of the vascular cell fate decision of human bone marrow-derived mononuclear cells in vivo.


Assuntos
Células da Medula Óssea/citologia , Células da Medula Óssea/fisiologia , Transplante de Medula Óssea , Infarto do Miocárdio/patologia , Infarto do Miocárdio/terapia , Miócitos Cardíacos/citologia , Animais , Aorta/citologia , Linhagem da Célula/fisiologia , Células Endoteliais/citologia , Células Epiteliais/citologia , Células Epiteliais/fisiologia , Fibroblastos/citologia , Fibroblastos/fisiologia , Sobrevivência de Enxerto , Proteínas de Fluorescência Verde/genética , Humanos , Lentivirus/genética , Masculino , Camundongos , Camundongos Nus , Músculo Liso Vascular/citologia , Infarto do Miocárdio/fisiopatologia , Miócitos Cardíacos/fisiologia , Ratos , Recuperação de Função Fisiológica/fisiologia , Volume Sistólico/fisiologia , Timidina Quinase/genética , Transplante Heterólogo , Veias Umbilicais/citologia
14.
J Exp Med ; 201(11): 1825-35, 2005 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-15928198

RESUMO

The regulation of acetylation is central for the epigenetic control of lineage-specific gene expression and determines cell fate decisions. We provide evidence that the inhibition of histone deacetylases (HDACs) blocks the endothelial differentiation of adult progenitor cells. To define the mechanisms by which HDAC inhibition prevents endothelial differentiation, we determined the expression of homeobox transcription factors and demonstrated that HoxA9 expression is down-regulated by HDAC inhibitors. The causal involvement of HoxA9 in the endothelial differentiation of adult progenitor cells is supported by the finding that HoxA9 overexpression partially rescued the endothelial differentiation blockade induced by HDAC inhibitors. Knockdown and overexpression studies revealed that HoxA9 acts as a master switch to regulate the expression of prototypical endothelial-committed genes such as endothelial nitric oxide synthase, VEGF-R2, and VE-cadherin, and mediates the shear stress-induced maturation of endothelial cells. Consistently, HoxA9-deficient mice exhibited lower numbers of endothelial progenitor cells and showed an impaired postnatal neovascularization capacity after the induction of ischemia. Thus, HoxA9 is regulated by HDACs and is critical for postnatal neovascularization.


Assuntos
Diferenciação Celular/fisiologia , Células Endoteliais/fisiologia , Regulação da Expressão Gênica/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Histona Desacetilases/metabolismo , Proteínas de Homeodomínio/biossíntese , Animais , Antígenos CD , Caderinas/metabolismo , Células Cultivadas , Células Endoteliais/citologia , Sangue Fetal/citologia , Sangue Fetal/fisiologia , Células-Tronco Hematopoéticas/citologia , Proteínas de Homeodomínio/genética , Humanos , Isquemia/metabolismo , Camundongos , Camundongos Knockout , Neovascularização Fisiológica/genética , Neovascularização Fisiológica/fisiologia , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo II , Óxido Nítrico Sintase Tipo III , Estresse Mecânico , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
15.
Basic Res Cardiol ; 105(6): 703-12, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20652278

RESUMO

Diabetes is characterized by a chronic stage of hyperglycemia associated with endothelial progenitor cell dysfunction and reduced neovascularization in response to tissue ischemia. The underlying mechanisms are not entirely clear. The bone marrow niches provide the essential microenvironment for maintenance of stem cell function in the bone marrow. A disturbed stem cell niche might lead to stem cell dysfunction, thereby, impairing progenitor cell-dependent vascular repair. Therefore, we investigated the effects of streptozotocin-induced diabetes on the bone marrow stem cell niches and stem cell function in mice. Here, we show that long-term diabetes induced a reduction in Lin⁻Sca-1(+)c-kit(+) hematopoietic progenitor cells and reduced the repopulation capacity in a competitive engraftment experiment. Consistently, the expression of Bmi1, which prevents hematopoietic progenitor cell senescence, was significantly reduced in diabetic bone marrow cells. To address the mechanism underlying the progenitor cell dysfunction, we analyzed the composition of the stem cell niche and the cytokine environment. Although the morphology of the vascular and endosteal niche was not affected by diabetes, diabetic mice showed a significant deterioration of cytokine expression patterns in the bone marrow. In summary, these data indicate that diabetes imposes a long-term effect on the stem cell niche and affects important hematopoietic progenitor cell functions in mice.


Assuntos
Células da Medula Óssea/patologia , Proliferação de Células , Citocinas/metabolismo , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Tipo 1/patologia , Células-Tronco Hematopoéticas/patologia , Nicho de Células-Tronco , Animais , Antígenos Ly/metabolismo , Células da Medula Óssea/imunologia , Transplante de Medula Óssea , Diferenciação Celular , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/imunologia , Diabetes Mellitus Experimental/fisiopatologia , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/fisiopatologia , Células Endoteliais/imunologia , Células Endoteliais/patologia , Regulação da Expressão Gênica , Células-Tronco Hematopoéticas/imunologia , Membro Posterior , Isquemia/imunologia , Isquemia/patologia , Isquemia/fisiopatologia , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Músculo Esquelético/irrigação sanguínea , Neovascularização Fisiológica , Proteínas Nucleares/metabolismo , Osteoblastos/imunologia , Osteoblastos/patologia , Osteoclastos/imunologia , Osteoclastos/patologia , Complexo Repressor Polycomb 1 , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo , Proteínas Repressoras/metabolismo , Fatores de Tempo
16.
Basic Res Cardiol ; 105(3): 389-97, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-19911112

RESUMO

Initial trials suggest that bone marrow-derived vasculogenic cells augment neovascularization in patients after myocardial infarction. Moreover, in some experimental settings, they also provide an anti-atherosclerotic effect by maintaining the integrity of the endothelium. Risk factors for coronary artery disease were shown to reduce the number and functional activity of vasculogenic cells and increased cellular p38 MAP kinase activity. Inhibition of p38 MAP kinase increases the number and functional activity of proangiogenic cells in vitro and clinical trials are under way to examine the effect of p38 inhibition in patients with CAD. Here, we examined the effect of systemic p38 MAP kinase inhibition on vasculogenic cells and atherosclerotic disease progression in vivo. Treatment of ApoE(-/-) mice with the p38 inhibitor SB203580 significantly increased the number of pro-angiogenic cells such as Sca-1(+)Flk-1(+) as well as CD11b(low)Flk-1(+) cells and reduced the number of the inflammatory Gr1(+)CD45(+) cells. Moreover, invasion capacity of bone marrow-derived mononuclear cells under basal conditions as well as towards a gradient of SDF-1 was significantly augmented in ApoE-/- mice after p38 inhibition. Finally, treatment of ApoE(-/-) mice with SB203580 for 4 months reduced atheromatous lesion size by 51 +/- 3% (p < 0.05) without affecting the density of vasa vasorum in the plaques. In conclusion, this study demonstrates that systemic p38 MAP kinase inhibition with SB203580 improves the number and function of vasculogenic cells in an animal model of hypercholesterolemia, and reduces atherosclerotic disease progression in ApoE(-/-) mice.


Assuntos
Aterosclerose/prevenção & controle , Progressão da Doença , Inibidores Enzimáticos/farmacologia , Imidazóis/farmacologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Piridinas/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Animais , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Aterosclerose/etiologia , Modelos Animais de Doenças , Hipercolesterolemia/complicações , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/fisiologia , Camundongos , Camundongos Knockout , Neovascularização Fisiológica/efeitos dos fármacos , Neovascularização Fisiológica/fisiologia , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia
17.
Arterioscler Thromb Vasc Biol ; 29(11): 1802-9, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19696399

RESUMO

OBJECTIVE: Bone marrow-derived mononuclear cells (BMCs) improve the functional recovery after ischemia. However, BMCs comprise a heterogeneous mixture of cells, and it is not known which cell types are responsible for the induction of neovascularization after cell therapy. Because cell recruitment is critically dependent on the expression of the SDF-1-receptor CXCR4, we examined whether the expression of CXCR4 may identify a therapeutically active population of BMCs. METHODS AND RESULTS: Human CXCR4(+) and CXCR4(-) BMCs were sorted by magnetic beads. CXCR4(+) BMCs showed a significantly higher invasion capacity under basal conditions and after SDF-1 stimulation. Hematopoietic or mesenchymal colony-forming capacity did not differ between CXCR4(+) and CXCR4(-) BMCs. Injection of CXCR4(+) BMCs in mice after induction of hindlimb ischemia significantly improved the recovery of perfusion compared to injection of CXCR4(-) BMCs. Likewise, capillary density was significantly increased in CXCR4(+) BMC-treated mice. Because part of the beneficial effects of cell therapy were attributed to the release of paracrine effectors, we analyzed BMC supernatants for secreted factors. Importantly, supernatants of CXCR4(+) BMCs were enriched in the proangiogenic cytokines HGF and PDGF-BB. CONCLUSIONS: CXCR4(+) BMCs exhibit an increased therapeutic potential for blood flow recovery after acute ischemia. Mechanistically, their higher migratory capacity and their increased release of paracrine factors may contribute to enhanced tissue repair.


Assuntos
Células da Medula Óssea/metabolismo , Membro Posterior/irrigação sanguínea , Isquemia/terapia , Leucócitos Mononucleares/metabolismo , Neovascularização Fisiológica/fisiologia , Receptores CXCR4/metabolismo , Doença Aguda , Animais , Células da Medula Óssea/citologia , Células Cultivadas , Citocinas/metabolismo , Modelos Animais de Doenças , Regulação da Expressão Gênica , Membro Posterior/metabolismo , Humanos , Leucócitos Mononucleares/citologia , Camundongos , Camundongos Nus , Neovascularização Fisiológica/genética , Probabilidade , Distribuição Aleatória , Receptores CXCR4/genética , Fluxo Sanguíneo Regional/fisiologia , Estatísticas não Paramétricas , Regulação para Cima , Fator A de Crescimento do Endotélio Vascular/metabolismo
18.
Arch Med Sci Atheroscler Dis ; 5: e171-e177, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32832717

RESUMO

INTRODUCTION: Current studies suggest improved survival in patients with severe functional mitral regurgitation (FMR) treated successfully with the MitraClip (MC) compared to medical treatment alone, in addition to a significant reduction of FMR severity. Recently, the Carillon system (CS) has also been shown to significantly reduce FMR. However, whether this beneficial effect of CS also translates into a survival benefit comparable to the MC system has not been investigated so far. The aim of the study was to compare the course of FMR grade and mortality after MC or CS in a retrospective, non-randomized, single-center analysis. MATERIAL AND METHODS: A hundred and fifty-four patients with symptomatic FMR 2+ were included in this study (MC: n = 117, CS: n = 37). Baseline characteristics did not differ significantly between groups. RESULTS AND CONCLUSIONS: Initially, the degree of FMR was reduced in the MC group from 2.9 ±0.3 to 1.7 ±0.7 and from 2.7 ±0.5 to 2.1 ±0.7 in the CS group, p within and between groups < 0.01. Within 6 months, FMR remained reduced in the MC group (1.83 ±0.6) and CS group (2.1 ±0.7). One-year survival was 34.8% in the MC group and 54.8% in the CS group (p = 0.663). Median long-term survival was 1.66 years in the MC group and 3.92 years in the CS group, log rank p = 0.001.

19.
Circulation ; 118(14): 1425-32, 2008 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-18794392

RESUMO

BACKGROUND: Clinical trials indicate a beneficial effect of intracoronary infusion of progenitor cells on myocardial function in patients with ischemic heart disease. The extent and potential determinants of proangiogenic progenitor cell homing into the damaged myocardium after intracoronary infusion and the underlying mechanisms are still unknown. METHOD AND RESULTS: Circulating proangiogenic progenitor cells isolated from peripheral blood and cultivated for 3 days were labeled with radioactive indium oxine ((111)In-oxine). Radiolabeled proangiogenic progenitor cells (7.6+/-3.0 MBq, mean+/-SD) were administered to patients with previous myocardial infarction and a revascularized infarct vessel at various stages after infarction (5 days to 17 years). Viability of the infarcted myocardium was determined by (18)F-fluorodeoxyglucose-positron emission tomography and microcirculatory function by intracoronary Doppler measurements. One hour after application of progenitor cells, a mean of 6.9+/-4.7% (range, 1% to 19%; n=17) of total radioactivity was detected in the heart, which declined to 2+/-1% after 3 to 4 days. Average activity within the first 24 hours was highest among patients with acute myocardial infarction (14 days to 1 year; 4.5+/-3.2%; n=4) or a chronic stage (infarct age >1 year; 2.5+/-1.6%; n=5). Low viability of the infarcted myocardium and reduced coronary flow reserve were significant (P<0.05) predictors of proangiogenic progenitor cell homing. CONCLUSIONS: In patients after myocardial infarction undergoing intracoronary infusion of (111)In-oxine-labeled proangiogenic progenitor cells, a substantial amount of radioactivity is detected for several days in the heart, indicating homing of progenitor cells to the myocardium. The amount of proangiogenic progenitor cells retained in the heart decreased progressively with time after the acute myocardial infarction. Proangiogenic progenitor cells preferentially home to extensive acute myocardial infarcts characterized by low viability and reduced coronary flow reserve.


Assuntos
Infarto do Miocárdio/patologia , Infarto do Miocárdio/cirurgia , Miocárdio/citologia , Transplante de Células-Tronco/métodos , Células-Tronco/citologia , Adolescente , Células Cultivadas , Criança , Pré-Escolar , Feminino , Humanos , Lactente , Recém-Nascido , Masculino , Infarto do Miocárdio/metabolismo , Miocárdio/metabolismo , Projetos Piloto , Células-Tronco/metabolismo
20.
Biochem Biophys Res Commun ; 378(4): 826-31, 2009 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-19070587

RESUMO

We have recently shown that 3-deazaadenosine (c3Ado) inhibits atherogenesis in mice. We studied whether its anti-inflammatory capacity would also affect neointima-formation after balloon injury. Sprague Dawley rats underwent balloon angioplasty. C3Ado was administered orally, starting 5 days prior to the balloon injury and continued for 2 weeks. Fourteen days after balloon injury the intima/media ratio in the c3Ado-treated group was reduced by 67% (p<0.001) and luminal stenosis by 50% (p<0.001). Neointimal cellular density was decreased by 25% (p<0.001) and the induction of c-Jun and ki67 was markedly lower. The reduction of the intima/media ratio was still observed 3 months after balloon injury. Furthermore, a c3Ado-dependent inhibition of PDGF-mediated ERK-activation and proliferation could be demonstrated. Short-term administration of C3Ado inhibits neointima-formation in rats for at least 3 months after injury. The present findings implicate that c3Ado may be useful as an inhibitor of restenosis-formation after balloon angioplasty in humans.


Assuntos
Angioplastia com Balão/efeitos adversos , Reestenose Coronária/prevenção & controle , Tubercidina/administração & dosagem , Túnica Íntima/efeitos dos fármacos , Administração Oral , Animais , Medula Óssea/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Reestenose Coronária/patologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fígado/efeitos dos fármacos , Masculino , Fosforilação/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Tubercidina/efeitos adversos , Túnica Íntima/lesões , Túnica Íntima/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA