Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Cardiology ; 146(2): 144-150, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33326980

RESUMO

The radial artery is the preferred access site for cardiac catheterization because of patient comfort, early ambulation, and improved survival in acute coronary syndromes, when compared to the femoral artery route. However, it is associated with a high radial artery occlusion (RAO) rate, and patent haemostasis which can reduce this is extremely hard to implement in a busy clinical practice. Smaller sized sheaths are associated with less RAO but are uncommonly used as they could limit procedural prowess and complexity. Alternatively, the distal radial artery (dRA) approach appears to be safer with observed RAO rates of well under 1 percent without compromising benefits offered by the radial artery access. Default dRA can be accessed by palpation alone in most cases with some practice, and this can be improved further with ultrasound guidance. There is a subset of patients, especially in the elderly, where dRA access can be particularly challenging. To mitigate this, we propose a two-step cannulation strategy and illustrate this with a few cases with difficult dRA and radial artery anatomies.


Assuntos
Síndrome Coronariana Aguda , Arteriopatias Oclusivas , Intervenção Coronária Percutânea , Idoso , Cateterismo Cardíaco , Angiografia Coronária , Humanos , Artéria Radial/diagnóstico por imagem , Artéria Radial/cirurgia , Resultado do Tratamento , Ultrassonografia
2.
JACC Clin Electrophysiol ; 5(8): 968-976, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31439299

RESUMO

OBJECTIVES: This meta-analysis examined the ability of pulmonary vein isolation (PVI) to prevent atrial fibrillation in randomized controlled trials (RCTs) in which the patients not receiving PVI nevertheless underwent a procedure. BACKGROUND: PVI is a commonly used procedure for the treatment of atrial fibrillation (AF), and its efficacy has usually been judged against therapy with anti-arrhythmic drugs in open-label trials. There have been several RCTs of AF ablation in which both arms received an ablation, but the difference between the treatment arms was inclusion or omission of PVI. These trials of an ablation strategy with PVI versus an ablation strategy without PVI may provide a more rigorous method for evaluating the efficacy of PVI. METHODS: Medline and Cochrane databases were searched for RCTs comparing ablation including PVI with ablation excluding PVI. The primary efficacy endpoint was freedom from atrial fibrillation (AF) and atrial tachycardia at 12 months. A random-effects meta-analysis was performed using the restricted maximum likelihood estimator. RESULTS: Overall, 6 studies (n = 610) met inclusion criteria. AF recurrence was significantly lower with an ablation including PVI than an ablation without PVI (RR: 0.54; 95% confidence interval [CI]: 0.33 to 0.89; p = 0.0147; I2 = 79.7%). Neither the type of AF (p = 0.48) nor the type of non-PVI ablation (p = 0.21) was a significant moderator of the effect size. In 3 trials the non-PVI ablation procedure was performed in both arms, whereas PVI was performed in only 1 arm. In these studies, AF recurrence was significantly lower when PVI was included (RR: 0.32; 95% CI: 0.14 to 0.73; p = 0.007, I2 78%). CONCLUSIONS: In RCTs where both arms received an ablation, and therefore an expectation amongst patients and doctors of benefit, being randomized to PVI had a striking effect, reducing AF recurrence by a half.


Assuntos
Fibrilação Atrial/cirurgia , Ablação por Cateter , Veias Pulmonares/cirurgia , Ablação por Cateter/efeitos adversos , Ablação por Cateter/métodos , Ablação por Cateter/estatística & dados numéricos , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Ensaios Clínicos Controlados Aleatórios como Assunto , Recidiva
3.
J Cardiovasc Electrophysiol ; 29(11): 1471-1479, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30230101

RESUMO

OBJECTIVE: We tested whether ablation methodology and study design can explain the varying outcomes in terms of atrial fibrillation (AF)-free survival at 1 year. BACKGROUND: There have been numerous paroxysmal AF ablation trials, which are heterogeneous in their use of different ablation techniques and study design. A useful approach to understanding how these factors influence outcome is to dismantle the trials into individual arms and reconstitute them as a large meta-regression. METHODS: Data were collected from 66 studies (6941 patients). With freedom from AF as the dependent variable, we performed meta-regression using the individual study arm as the unit. RESULTS: Success rates did not change regardless of the technique used to produce pulmonary vein isolation (PVI). Neither was adjunctive lesion sets associated with any improvement in outcome. Studies that included more males and fewer hypertensive patients were found more likely to report better outcomes. The electrocardiography method selected to assess outcome also plays an important role. Outcomes were worse in studies that used regular telemonitoring (by 23%; P < 0.001) or in patients who had implantable loop recorders (by 21%; P = 0.006), rather than those with the less thorough periodic Holter monitoring. CONCLUSIONS: Outcomes of AF ablation studies involving PVI are not affected by the technologies used to produce PVI. Neither do adjunctive lesion sets change the outcome. Achieving high success rates in these studies appears to be dependent more on patient mix and on the thoroughness of AF detection protocols. These should be carefully considered when quoting the success rates of AF ablation procedures that are derived from such studies.


Assuntos
Técnicas de Ablação/métodos , Fibrilação Atrial/cirurgia , Ablação por Cateter/métodos , Ensaios Clínicos Controlados Aleatórios como Assunto/métodos , Técnicas de Ablação/tendências , Fibrilação Atrial/diagnóstico , Fibrilação Atrial/mortalidade , Ablação por Cateter/tendências , Humanos , Seleção de Pacientes , Taxa de Sobrevida/tendências , Resultado do Tratamento
4.
Cardiovasc Res ; 114(5): 656-667, 2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29401264

RESUMO

Aims: The heart is constantly challenged with acute bouts of stretching or overload. Systolic adaptations to these challenges are known but adaptations in diastolic stiffness remain unknown. We evaluated adaptations in myocardial stiffness due to acute stretching and characterized the underlying mechanisms. Methods and results: Left ventricles (LVs) of intact rat hearts, rabbit papillary muscles and myocardial strips from cardiac surgery patients were stretched. After stretching, there was a sustained >40% decrease in end-diastolic pressure (EDP) or passive tension (PT) for 15 min in all species and experimental preparations. Stretching by volume loading in volunteers and cardiac surgery patients resulted in E/E' and EDP decreases, respectively, after sustained stretching. Stretched samples had increased myocardial cGMP levels, increased phosphorylated vasodilator-stimulated phosphoprotein phosphorylation, as well as, increased titin phosphorylation, which was reduced by prior protein kinase G (PKG) inhibition (PKGi). Skinned cardiomyocytes from stretched and non-stretched myocardia were studied. Skinned cardiomyocytes from stretched hearts showed decreased PT, which was abrogated by protein phosphatase incubation; whereas those from non-stretched hearts decreased PT after PKG incubation. Pharmacological studies assessed the role of nitric oxide (NO) and natriuretic peptides (NPs). PT decay after stretching was significantly reduced by combined NP antagonism, NO synthase inhibition and NO scavenging, or by PKGi. Response to stretching was remarkably reduced in a rat model of LV hypertrophy, which also failed to increase titin phosphorylation. Conclusions: We describe and translate to human physiology a novel adaptive mechanism, partly mediated by titin phosphorylation through cGMP-PKG signalling, whereby myocardial compliance increases in response to acute stretching. This mechanism may not function in the hypertrophic heart.


Assuntos
Hipertrofia Ventricular Esquerda/metabolismo , Mecanorreceptores/metabolismo , Contração Miocárdica , Miócitos Cardíacos/metabolismo , Músculos Papilares/metabolismo , Função Ventricular Esquerda , Adaptação Fisiológica , Animais , Estudos de Casos e Controles , Moléculas de Adesão Celular/metabolismo , Complacência (Medida de Distensibilidade) , Conectina/metabolismo , GMP Cíclico/metabolismo , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Modelos Animais de Doenças , Humanos , Hipertrofia Ventricular Esquerda/patologia , Hipertrofia Ventricular Esquerda/fisiopatologia , Preparação de Coração Isolado , Masculino , Mecanotransdução Celular , Proteínas dos Microfilamentos/metabolismo , Miócitos Cardíacos/patologia , Músculos Papilares/fisiopatologia , Fosfoproteínas/metabolismo , Fosforilação , Coelhos , Ratos Wistar , Sistemas do Segundo Mensageiro , Pressão Ventricular
5.
EBioMedicine ; 16: 224-237, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28163043

RESUMO

p53 is an important modulator of stem cell fate, but its role in cardiac progenitor cells (CPCs) is unknown. Here, we tested the effects of a single extra-copy of p53 on the function of CPCs in the presence of oxidative stress mediated by doxorubicin in vitro and type-1 diabetes in vivo. CPCs were obtained from super-p53 transgenic mice (p53-tg), in which the additional allele is regulated in a manner similar to the endogenous protein. Old CPCs with increased p53 dosage showed a superior ability to sustain oxidative stress, repair DNA damage and restore cell division. With doxorubicin, a larger fraction of CPCs carrying an extra-copy of the p53 allele recruited γH2A.X reestablishing DNA integrity. Enhanced p53 expression resulted in a superior tolerance to oxidative stress in vivo by providing CPCs with defense mechanisms necessary to survive in the milieu of the diabetic heart; they engrafted in regions of tissue injury and in three days acquired the cardiomyocyte phenotype. The biological advantage provided by the increased dosage of p53 in CPCs suggests that this genetic strategy may be translated to humans to increase cellular engraftment and growth, critical determinants of successful cell therapy for the failing heart.


Assuntos
Diabetes Mellitus Experimental/metabolismo , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Células-Tronco/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Western Blotting , Diferenciação Celular/genética , Proliferação de Células/genética , Células Cultivadas , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/metabolismo , Feminino , Expressão Gênica , Coração/fisiopatologia , Histonas/metabolismo , Humanos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia de Fluorescência , Miócitos Cardíacos/citologia , Miócitos Cardíacos/transplante , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transplante de Células-Tronco/métodos , Células-Tronco/citologia , Proteína Supressora de Tumor p53/genética
6.
Int J Cardiol ; 228: 465-480, 2017 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-27870978

RESUMO

Today there is an increasing demand for heart transplantations for patients diagnosed with heart failure. Though, shortage of donors as well as the large number of ineligible patients hurdle such treatment option. This, in addition to the considerable number of transplant rejections, has driven the clinical research towards the field of regenerative medicine. Nonetheless, to date, several stem cell therapies tested in animal models fall by the wayside and when they meet the criteria to clinical trials, subjects often exhibit modest improvements. A main issue slowing down the admission of such therapies in the domain of human trials is the lack of protocol standardization between research groups, which hampers comparison between different approaches as well as the lack of thought regarding the clinical translation. In this sense, given the large amount of reports on stem cell therapy studies in animal models reported in the last 3years, we sought to evaluate their advantages and limitations towards the clinical setting and provide some suggestions for the forthcoming investigations. We expect, with this review, to start a new paradigm on regenerative medicine, by evoking the debate on how to plan novel stem cell therapy studies with animal models in order to achieve more consistent scientific production and accelerate the admission of stem cell therapies in the clinical setting.


Assuntos
Terapia Genética/normas , Isquemia Miocárdica/terapia , Medicina Regenerativa/normas , Transplante de Células-Tronco/normas , Animais , Modelos Animais de Doenças , Humanos
7.
Clin Med (Lond) ; 15(5): 426-30, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26430179

RESUMO

Spontaneous bacterial peritonitis (SBP) in cirrhotic patients is a serious complication associated with a high mortality rate. A baseline audit of the acute medical take (AMT) at Northwick Park suggested a lack of awareness regarding management. A questionnaire based on contemporary SBP guidelines was circulated to all trainee doctors (FY1 to SpR). Ascitic fluid testing requests were analysed over a six-month period. The electronic requesting system was updated to include prompts and direct links to Trust SBP guidelines, and a one-hour lecture to all members of the AMT, supported by an educational booklet on SBP, was performed. Re-audit was carried out six months post-intervention, the AMT completed a second questionnaire and ascitic fluid testing requests were re-audited. In comparable pre- and post-intervention AMT cohorts, a clinical and educational intervention led to a significant improvement in understanding of when to investigate (p≤0.001), samples (p = 0.002) and containers (p≤0.001) required, urgency of obtaining results (p≤0.001), and initiation of treatment for suspected SBP (p = 0.007). Significantly more ascitic samples were sent, with specific suspicion of SBP more readily documented, crucial to expediting laboratory processing. Targeted education and production of a clinical algorithm has significantly improved the management of patients with SBP.


Assuntos
Infecções Bacterianas/terapia , Cirrose Hepática/complicações , Peritonite/terapia , Infecções Bacterianas/diagnóstico , Educação Médica Continuada , Humanos , Capacitação em Serviço/métodos , Peritonite/diagnóstico , Guias de Prática Clínica como Assunto , Inquéritos e Questionários
8.
Rev Port Cardiol ; 33(9): 493-9, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25240860

RESUMO

INTRODUCTION: The myocardial response to acute stretch consists of a two-phase increase in contractility: an acute increase by the Frank-Starling mechanism and a gradual and time-dependent increase in force generated known as the slow force response (SFR). The SFR is actively modulated by different signaling pathways, but the role of protein kinase G (PKG) signaling is unknown. In this study we aim to characterize the role of the PKG signaling pathway in the SFR under normal and ischemic conditions. METHODS: Rabbit papillary muscles were stretched from 92 to 100% of maximum length (Lmax) under basal conditions, in the absence (1) or presence of: a PKG agonist (2) and a PKG inhibitor (3); under ischemic conditions in the absence (4) or presence of: a PKG agonist (5); a nitric oxide (NO) donor (6) and a phosphodiesterase 5 (PDE5) inhibitor (7). RESULTS: Under normoxia, the SFR was significantly attenuated by inhibition of PKG and remained unaltered with PKG activation. Ischemia induced a progressive decrease in myocardial contractility after stretch. Neither the PKG agonist nor the NO donor altered the myocardial response to stretch under ischemic conditions. However, the use of a PDE5 inhibitor in ischemia partially reversed the progressive deterioration in contractility. CONCLUSIONS: PKG activity is essential for the SFR. During ischemia, a progressive decline in the force is observed in response to acute myocardial stretch. This dysfunctional response can be partially reversed by the use of PDE5 inhibitors.


Assuntos
Proteínas Quinases Dependentes de GMP Cíclico/fisiologia , Contração Miocárdica/fisiologia , Animais , Fenômenos Biomecânicos/fisiologia , Proteínas Quinases Dependentes de GMP Cíclico/antagonistas & inibidores , Coração/fisiologia , Miocárdio , Músculos Papilares/fisiologia , Coelhos , Transdução de Sinais
10.
Circulation ; 128(12): 1286-97, 2013 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-23983250

RESUMO

BACKGROUND: Little is known about the function of inositol 1,4,5-trisphosphate receptors (IP3Rs) in the adult heart experimentally. Moreover, whether these Ca(2+) release channels are present and play a critical role in human cardiomyocytes remains to be defined. IP3Rs may be activated after Gαq-protein-coupled receptor stimulation, affecting Ca(2+) cycling, enhancing myocyte performance, and potentially favoring an increase in the incidence of arrhythmias. METHODS AND RESULTS: IP3R function was determined in human left ventricular myocytes, and this analysis was integrated with assays in mouse myocytes to identify the mechanisms by which IP3Rs influence the electric and mechanical properties of the myocardium. We report that IP3Rs are expressed and operative in human left ventricular myocytes. After Gαq-protein-coupled receptor activation, Ca(2+) mobilized from the sarcoplasmic reticulum via IP3Rs contributes to the decrease in resting membrane potential, prolongation of the action potential, and occurrence of early afterdepolarizations. Ca(2+) transient amplitude and cell shortening are enhanced, and extrasystolic and dysregulated Ca(2+) elevations and contractions become apparent. These alterations in the electromechanical behavior of human cardiomyocytes are coupled with increased isometric twitch of the myocardium and arrhythmic events, suggesting that Gαq-protein-coupled receptor activation provides inotropic reserve, which is hampered by electric instability and contractile abnormalities. Additionally, our findings support the notion that increases in Ca(2+) load by IP3Rs promote Ca(2+) extrusion by forward-mode Na(+)/Ca(2+) exchange, an important mechanism of arrhythmic events. CONCLUSIONS: The Gαq-protein/coupled receptor/IP3R axis modulates the electromechanical properties of the human myocardium and its propensity to develop arrhythmias.


Assuntos
Potenciais de Ação/fisiologia , Sinalização do Cálcio/fisiologia , Insuficiência Cardíaca/fisiopatologia , Receptores de Inositol 1,4,5-Trifosfato/fisiologia , Miócitos Cardíacos/fisiologia , Adulto , Animais , Arritmias Cardíacas/fisiopatologia , Células Cultivadas , Feminino , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/fisiologia , Insuficiência Cardíaca/genética , Ventrículos do Coração/citologia , Humanos , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Contração Miocárdica/fisiologia , Miócitos Cardíacos/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/fisiologia , Retículo Sarcoplasmático/fisiologia , Transdução de Sinais/fisiologia
11.
Peptides ; 47: 77-84, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23856453

RESUMO

Acute myocardial stretch elicits a biphasic increase in contractility: an immediate increase, known as Frank-Starling mechanism (FSM), followed by a progressive increase, regarded as slow force response (SFR). In this study, we characterized the contractile response to acute stretch from 92 to 100% Lmax in rabbit papillary muscles (n=86) under normoxic and ischemic conditions, and its modulation by angiotensin II signaling pathway. Under normoxia, the FSM was independent of Na(+)/H(+)-exchanger, reverse mode of Na(+)/Ca(2+)-exchanger (r-NCX), AT1 receptor, AT2 receptor and PKC. Regarding the SFR, it was mediated by AT1 receptor activation and its downstream effectors PKC, Na(+)/H(+)-exchanger and r-NCX. Ischemia negatively impacted on the FSM and abolished the SFR, with the muscles exhibiting a time-dependent decline in contractility. Under ischemic conditions, FSM was not influenced by AT1 and AT2 receptors or PKC activation. AT1 receptor antagonism rescued the progressive deterioration in contractility, an effect partially dependent on AT2 receptor activation.


Assuntos
Angiotensina II/metabolismo , Contração Miocárdica/fisiologia , Isquemia Miocárdica/metabolismo , Miocárdio/metabolismo , Transdução de Sinais , Animais , Pressão Sanguínea , Masculino , Isquemia Miocárdica/fisiopatologia , Proteína Quinase C/metabolismo , Coelhos , Receptor Tipo 1 de Angiotensina/metabolismo , Receptor Tipo 2 de Angiotensina/metabolismo , Trocador de Sódio e Cálcio/metabolismo , Trocadores de Sódio-Hidrogênio/metabolismo , Estresse Mecânico , Estresse Fisiológico
12.
Circulation ; 126(15): 1869-81, 2012 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-22955965

RESUMO

BACKGROUND: Two opposite views of cardiac growth are currently held; one views the heart as a static organ characterized by a large number of cardiomyocytes that are present at birth and live as long as the organism, and the other views the heart a highly plastic organ in which the myocyte compartment is restored several times during the course of life. METHODS AND RESULTS: The average age of cardiomyocytes, vascular endothelial cells (ECs), and fibroblasts and their turnover rates were measured by retrospective (14)C birth dating of cells in 19 normal hearts 2 to 78 years of age and in 17 explanted failing hearts 22 to 70 years of age. We report that the human heart is characterized by a significant turnover of ventricular myocytes, ECs, and fibroblasts, physiologically and pathologically. Myocyte, EC, and fibroblast renewal is very high shortly after birth, decreases during postnatal maturation, remains relatively constant in the adult organ, and increases dramatically with age. From 20 to 78 years of age, the adult human heart entirely replaces its myocyte, EC, and fibroblast compartment ≈8, ≈6, and ≈8 times, respectively. Myocyte, EC, and fibroblast regeneration is further enhanced with chronic heart failure. CONCLUSIONS: The human heart is a highly dynamic organ that retains a remarkable degree of plasticity throughout life and in the presence of chronic heart failure. However, the ability to regenerate cardiomyocytes, vascular ECs, and fibroblasts cannot prevent the manifestations of myocardial aging or oppose the negative effects of ischemic and idiopathic dilated cardiomyopathy.


Assuntos
Insuficiência Cardíaca/fisiopatologia , Desenvolvimento Muscular/fisiologia , Miócitos Cardíacos/fisiologia , Adolescente , Adulto , Idoso , Envelhecimento , Criança , Pré-Escolar , Células Endoteliais/fisiologia , Fibroblastos/fisiologia , Coração/fisiologia , Humanos , Pessoa de Meia-Idade , Miócitos Cardíacos/citologia , Regeneração , Doadores de Tecidos , Adulto Jovem
13.
Circ Res ; 111(7): 894-906, 2012 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-22851539

RESUMO

RATIONALE: According to the immortal DNA strand hypothesis, dividing stem cells selectively segregate chromosomes carrying the old template DNA, opposing accumulation of mutations resulting from nonrepaired replication errors and attenuating telomere shortening. OBJECTIVE: Based on the premise of the immortal DNA strand hypothesis, we propose that stem cells retaining the old DNA would represent the most powerful cells for myocardial regeneration. METHODS AND RESULTS: Division of human cardiac stem cells (hCSCs) by nonrandom and random segregation of chromatids was documented by clonal assay of bromodeoxyuridine-tagged hCSCs. Additionally, their growth properties were determined by a series of in vitro and in vivo studies. We report that a small class of hCSCs retain during replication the mother DNA and generate 2 daughter cells, which carry the old and new DNA, respectively. hCSCs with immortal DNA form a pool of nonsenescent cells with longer telomeres and higher proliferative capacity. The self-renewal and long-term repopulating ability of these cells was shown in serial-transplantation assays in the infarcted heart; these cells created a chimeric organ, composed of spared rat and regenerated human cardiomyocytes and coronary vessels, leading to a remarkable restoration of cardiac structure and function. The documentation that hCSCs divide by asymmetrical and symmetrical chromatid segregation supports the view that the human heart is a self-renewing organ regulated by a compartment of resident hCSCs. CONCLUSIONS: The impressive recovery in ventricular hemodynamics and anatomy mediated by clonal hCSCs carrying the "mother" DNA underscores the clinical relevance of this stem cell class for the management of heart failure in humans.


Assuntos
Cromátides/fisiologia , Segregação de Cromossomos/fisiologia , Coração/fisiologia , Infarto do Miocárdio/terapia , Miocárdio/citologia , Regeneração/fisiologia , Transplante de Células-Tronco , Células-Tronco/citologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Bromodesoxiuridina , Proliferação de Células , Células Cultivadas , Criança , Pré-Escolar , Cromátides/ultraestrutura , DNA/fisiologia , Feminino , Humanos , Técnicas In Vitro , Lactente , Masculino , Pessoa de Meia-Idade , Modelos Animais , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Ratos , Ratos Endogâmicos F344 , Células-Tronco/fisiologia , Telômero/ultraestrutura , Adulto Jovem
14.
Circ Res ; 110(5): 701-15, 2012 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-22275487

RESUMO

RATIONALE: Embryonic and fetal myocardial growth is characterized by a dramatic increase in myocyte number, but whether the expansion of the myocyte compartment is dictated by activation and commitment of resident cardiac stem cells (CSCs), division of immature myocytes or both is currently unknown. OBJECTIVE: In this study, we tested whether prenatal cardiac development is controlled by activation and differentiation of CSCs and whether division of c-kit-positive CSCs in the mouse heart is triggered by spontaneous Ca(2+) oscillations. METHODS AND RESULTS: We report that embryonic-fetal c-kit-positive CSCs are self-renewing, clonogenic and multipotent in vitro and in vivo. The growth and commitment of c-kit-positive CSCs is responsible for the generation of the myocyte progeny of the developing heart. The close correspondence between values computed by mathematical modeling and direct measurements of myocyte number at E9, E14, E19 and 1 day after birth strongly suggests that the organogenesis of the embryonic heart is dependent on a hierarchical model of cell differentiation regulated by resident CSCs. The growth promoting effects of c-kit-positive CSCs are triggered by spontaneous oscillations in intracellular Ca(2+), mediated by IP3 receptor activation, which condition asymmetrical stem cell division and myocyte lineage specification. CONCLUSIONS: Myocyte formation derived from CSC differentiation is the major determinant of cardiac growth during development. Division of c-kit-positive CSCs in the mouse is promoted by spontaneous Ca(2+) spikes, which dictate the pattern of stem cell replication and the generation of a myocyte progeny at all phases of prenatal life and up to one day after birth.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Coração/embriologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/fisiologia , Proteínas Proto-Oncogênicas c-kit/metabolismo , Animais , Cálcio/metabolismo , Sinalização do Cálcio/fisiologia , Células Cultivadas , Técnicas de Cultura Embrionária , Receptores de Inositol 1,4,5-Trifosfato/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Animais , Modelos Teóricos , Organogênese/fisiologia , Proteínas Proto-Oncogênicas c-kit/genética
15.
N Engl J Med ; 364(19): 1795-806, 2011 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-21561345

RESUMO

BACKGROUND: Although progenitor cells have been described in distinct anatomical regions of the lung, description of resident stem cells has remained elusive. METHODS: Surgical lung-tissue specimens were studied in situ to identify and characterize human lung stem cells. We defined their phenotype and functional properties in vitro and in vivo. RESULTS: Human lungs contain undifferentiated human lung stem cells nested in niches in the distal airways. These cells are self-renewing, clonogenic, and multipotent in vitro. After injection into damaged mouse lung in vivo, human lung stem cells form human bronchioles, alveoli, and pulmonary vessels integrated structurally and functionally with the damaged organ. The formation of a chimeric lung was confirmed by detection of human transcripts for epithelial and vascular genes. In addition, the self-renewal and long-term proliferation of human lung stem cells was shown in serial-transplantation assays. CONCLUSIONS: Human lungs contain identifiable stem cells. In animal models, these cells participate in tissue homeostasis and regeneration. They have the undemonstrated potential to promote tissue restoration in patients with lung disease. (Funded by the National Institutes of Health.).


Assuntos
Pulmão/citologia , Células-Tronco/fisiologia , Adulto , Animais , Células Clonais , Feminino , Humanos , Pulmão/embriologia , Pulmão/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Pluripotentes , Proteínas Proto-Oncogênicas c-kit/análise , Regeneração , Transplante de Células-Tronco , Células-Tronco/química
16.
Circ Res ; 108(12): 1467-81, 2011 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-21546606

RESUMO

RATIONALE: Age and coronary artery disease may negatively affect the function of human cardiac stem cells (hCSCs) and their potential therapeutic efficacy for autologous cell transplantation in the failing heart. OBJECTIVE: Insulin-like growth factor (IGF)-1, IGF-2, and angiotensin II (Ang II), as well as their receptors, IGF-1R, IGF-2R, and AT1R, were characterized in c-kit(+) hCSCs to establish whether these systems would allow us to separate hCSC classes with different growth reserve in the aging and diseased myocardium. METHODS AND RESULTS: C-kit(+) hCSCs were collected from myocardial samples obtained from 24 patients, 48 to 86 years of age, undergoing elective cardiac surgery for coronary artery disease. The expression of IGF-1R in hCSCs recognized a young cell phenotype defined by long telomeres, high telomerase activity, enhanced cell proliferation, and attenuated apoptosis. In addition to IGF-1, IGF-1R(+) hCSCs secreted IGF-2 that promoted myocyte differentiation. Conversely, the presence of IGF-2R and AT1R, in the absence of IGF-1R, identified senescent hCSCs with impaired growth reserve and increased susceptibility to apoptosis. The ability of IGF-1R(+) hCSCs to regenerate infarcted myocardium was then compared with that of unselected c-kit(+) hCSCs. IGF-1R(+) hCSCs improved cardiomyogenesis and vasculogenesis. Pretreatment of IGF-1R(+) hCSCs with IGF-2 resulted in the formation of more mature myocytes and superior recovery of ventricular structure. CONCLUSIONS: hCSCs expressing only IGF-1R synthesize both IGF-1 and IGF-2, which are potent modulators of stem cell replication, commitment to the myocyte lineage, and myocyte differentiation, which points to this hCSC subset as the ideal candidate cell for the management of human heart failure.


Assuntos
Doença da Artéria Coronariana/metabolismo , Infarto do Miocárdio/metabolismo , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Receptor IGF Tipo 1/metabolismo , Regeneração , Células-Tronco/metabolismo , Angiotensina II/metabolismo , Diferenciação Celular , Doença da Artéria Coronariana/patologia , Doença da Artéria Coronariana/terapia , Feminino , Humanos , Fator de Crescimento Insulin-Like I/biossíntese , Fator de Crescimento Insulin-Like II/metabolismo , Masculino , Infarto do Miocárdio/patologia , Infarto do Miocárdio/terapia , Miocárdio/patologia , Miócitos Cardíacos/patologia , Receptor IGF Tipo 2/metabolismo , Transplante de Células-Tronco , Células-Tronco/patologia , Transplante Autólogo
17.
Circulation ; 123(12): 1287-96, 2011 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-21403094

RESUMO

BACKGROUND: Cardiac stem cells (CSCs) delivered to the infarcted heart generate a large number of small fetal-neonatal cardiomyocytes that fail to acquire the differentiated phenotype. However, the interaction of CSCs with postmitotic myocytes results in the formation of cells with adult characteristics. METHODS AND RESULTS: On the basis of results of in vitro and in vivo assays, we report that the commitment of human CSCs (hCSCs) to the myocyte lineage and the generation of mature working cardiomyocytes are influenced by microRNA-499 (miR-499), which is barely detectable in hCSCs but is highly expressed in postmitotic human cardiomyocytes. miR-499 traverses gap junction channels and translocates to structurally coupled hCSCs favoring their differentiation into functionally competent cells. Expression of miR-499 in hCSCs represses the miR-499 target genes Sox6 and Rod1, enhancing cardiomyogenesis in vitro and after infarction in vivo. Although cardiac repair was detected in all cell-treated infarcted hearts, the aggregate volume of the regenerated myocyte mass and myocyte cell volume were greater in animals injected with hCSCs overexpressing miR-499. Treatment with hCSCs resulted in an improvement in ventricular function, consisting of a better preservation of developed pressure and positive and negative dP/dt after infarction. An additional positive effect on cardiac performance occurred with miR-499, pointing to enhanced myocyte differentiation/hypertrophy as the mechanism by which miR-499 potentiated the restoration of myocardial mass and function in the infarcted heart. CONCLUSIONS: The recognition that miR-499 promotes the differentiation of hCSCs into mechanically integrated cardiomyocytes has important clinical implications for the treatment of human heart failure.


Assuntos
Células-Tronco Adultas/citologia , MicroRNAs/fisiologia , Infarto do Miocárdio/terapia , Miócitos Cardíacos/citologia , Transplante de Células-Tronco , Células-Tronco Adultas/fisiologia , Animais , Diferenciação Celular/fisiologia , Divisão Celular/fisiologia , Células Cultivadas , Técnicas de Cocultura , Modelos Animais de Doenças , Junções Comunicantes/fisiologia , Expressão Gênica/fisiologia , Humanos , Infarto do Miocárdio/patologia , Miócitos Cardíacos/fisiologia , Proteína de Ligação a Regiões Ricas em Polipirimidinas , Proteínas de Ligação a RNA/genética , Ratos , Regeneração/fisiologia , Fatores de Transcrição SOXD/genética
18.
Circ Res ; 108(9): 1071-83, 2011 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-21415392

RESUMO

RATIONALE: Understanding the mechanisms that regulate trafficking of human cardiac stem cells (hCSCs) may lead to development of new therapeutic approaches for the failing heart. OBJECTIVE: We tested whether the motility of hCSCs in immunosuppressed infarcted animals is controlled by the guidance system that involves the interaction of Eph receptors with ephrin ligands. METHODS AND RESULTS: Within the cardiac niches, cardiomyocytes expressed preferentially the ephrin A1 ligand, whereas hCSCs possessed the EphA2 receptor. Treatment of hCSCs with ephrin A1 resulted in the rapid internalization of the ephrin A1-EphA2 complex, posttranslational modifications of Src kinases, and morphological changes consistent with the acquisition of a motile cell phenotype. Ephrin A1 enhanced the motility of hCSCs in vitro, and their migration in vivo following acute myocardial infarction. At 2 weeks after infarction, the volume of the regenerated myocardium was 2-fold larger in animals injected with ephrin A1-activated hCSCs than in animals receiving control hCSCs; this difference was dictated by a greater number of newly formed cardiomyocytes and coronary vessels. The increased recovery in myocardial mass with ephrin A1-treated hCSCs was characterized by further restoration of cardiac function and by a reduction in arrhythmic events. CONCLUSIONS: Ephrin A1 promotes the motility of EphA2-positive hCSCs, facilitates their migration to the area of damage, and enhances cardiac repair. Thus, in situ stimulation of resident hCSCs with ephrin A1 or their ex vivo activation before myocardial delivery improves cell targeting to sites of injury, possibly providing a novel strategy for the management of the diseased heart.


Assuntos
Efrina-A1/genética , Efrina-A2/genética , Células-Tronco Hematopoéticas/citologia , Infarto do Miocárdio/fisiopatologia , Miócitos Cardíacos/citologia , Animais , Adesão Celular/fisiologia , Membrana Celular/metabolismo , Movimento Celular/fisiologia , Citoplasma/metabolismo , Efrina-A1/metabolismo , Efrina-A2/metabolismo , Expressão Gênica/fisiologia , Proteínas de Fluorescência Verde/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Infarto do Miocárdio/patologia , Infarto do Miocárdio/terapia , Ratos , Ratos Endogâmicos F344 , Taquicardia Ventricular/patologia , Taquicardia Ventricular/fisiopatologia , Taquicardia Ventricular/terapia
19.
J Biomed Biotechnol ; 2010: 807084, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20625419

RESUMO

Although systole was for long considered the core of cardiac function, hemodynamic performance is evenly dependent on appropriate systolic and diastolic functions. The recognition that isolated diastolic dysfunction is the major culprit for approximately fifty percent of all heart failure cases imposes a deeper understanding of its underlying mechanisms so that better diagnostic and therapeutic strategies can be designed. Risk factors leading to diastolic dysfunction affect myocardial relaxation and/or its material properties by disrupting the homeostasis of cardiomyocytes as well as their relation with surrounding matrix and vascular structures. As a consequence, slower ventricular relaxation and higher myocardial stiffness may result in higher ventricular filling pressures and in the risk of hemodynamic decompensation. Thus, determining the mechanisms of diastolic function and their implications in the pathophysiology of heart failure with normal ejection fraction has become a prominent field in basic and clinical research.


Assuntos
Diástole/fisiologia , Miocárdio/metabolismo , Animais , Matriz Extracelular/metabolismo , Insuficiência Cardíaca/fisiopatologia , Humanos , Miócitos Cardíacos/metabolismo , Fatores de Risco , Volume Sistólico/fisiologia
20.
Circ Res ; 107(2): 305-15, 2010 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-20522802

RESUMO

RATIONALE: The ability of the human heart to regenerate large quantities of myocytes remains controversial, and the extent of myocyte renewal claimed by different laboratories varies from none to nearly 20% per year. OBJECTIVE: To address this issue, we examined the percentage of myocytes, endothelial cells, and fibroblasts labeled by iododeoxyuridine in postmortem samples obtained from cancer patients who received the thymidine analog for therapeutic purposes. Additionally, the potential contribution of DNA repair, polyploidy, and cell fusion to the measurement of myocyte regeneration was determined. METHODS AND RESULTS: The fraction of myocytes labeled by iododeoxyuridine ranged from 2.5% to 46%, and similar values were found in fibroblasts and endothelial cells. An average 22%, 20%, and 13% new myocytes, fibroblasts, and endothelial cells were generated per year, suggesting that the lifespan of these cells was approximately 4.5, 5, and 8 years, respectively. The newly formed cardiac cells showed a fully differentiated adult phenotype and did not express the senescence-associated protein p16(INK4a). Moreover, measurements by confocal microscopy and flow cytometry documented that the human heart is composed predominantly of myocytes with 2n diploid DNA content and that tetraploid and octaploid nuclei constitute only a small fraction of the parenchymal cell pool. Importantly, DNA repair, ploidy formation, and cell fusion were not implicated in the assessment of myocyte regeneration. CONCLUSIONS: Our findings indicate that the human heart has a significant growth reserve and replaces its myocyte and nonmyocyte compartment several times during the course of life.


Assuntos
Proliferação de Células , Células Endoteliais/patologia , Fibroblastos/patologia , Desenvolvimento Muscular , Miocárdio/patologia , Miócitos Cardíacos/patologia , Neoplasias/patologia , Adulto , Fatores Etários , Idoso , Animais , Autopsia , Morte Celular , Fusão Celular , Núcleo Celular/patologia , Proliferação de Células/efeitos dos fármacos , Reparo do DNA , Células Endoteliais/efeitos dos fármacos , Feminino , Fibroblastos/efeitos dos fármacos , Citometria de Fluxo , Humanos , Idoxuridina/uso terapêutico , Imuno-Histoquímica , Masculino , Microscopia Confocal , Pessoa de Meia-Idade , Desenvolvimento Muscular/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Fenótipo , Poliploidia , Radiossensibilizantes/uso terapêutico , Ratos , Ratos Endogâmicos F344 , Regeneração , Fatores de Tempo , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...