Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
J Cardiothorac Vasc Anesth ; 24(5): 762-6, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20674392

RESUMO

OBJECTIVES: To compare cardiac output (CO) measurements from a novel endotracheal bioimpedance cardiac output monitor device (ECOM; ConMed, Irvine, CA) to simultaneous pulmonary artery thermodilution (TD) CO. DESIGN: Prospective study. SETTING: One academic hospital. PARTICIPANTS: Forty volunteer patients undergoing cardiac surgery. INTERVENTIONS: Intraoperative CO measurements. MEASUREMENTS AND MAIN RESULTS: Simultaneous comparative data points were collected from ECOM and TD at 4 periods: post-induction, post-sternotomy, post-cardiopulmonary bypass, and post-chest closure. The mean CO(TD) was compared with CO(ECOM) for each operative period then assessed for agreement by linear regression, Bland-Altman analysis, and percent error methods. There were 35 men (87.5%) with a mean age of 66 ± 10.7 years in the present study population. R values (p value) for the 4 time periods were 0.50 (0.002), 0.33 (0.035), 0.42 (0.007), and 0.48 (0.002). Bias and 95% limits of agreement in L/min were -0.11 (-2.40 to 2.18), 0.04 (-2.57 to 2.65), -0.06 (-2.86 to 2.74), and 0.02 (-2.42 to 2.45). Percent errors of the 4 time periods were 51%, 53%, 50%, and 48%. CONCLUSIONS: ECOM did not adequately agree with TD in patients undergoing cardiac surgery.


Assuntos
Débito Cardíaco/fisiologia , Procedimentos Cirúrgicos Cardíacos/métodos , Intubação Intratraqueal/métodos , Monitorização Intraoperatória/métodos , Idoso , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Termodiluição/métodos
2.
J Thorac Cardiovasc Surg ; 156(2): 556-565.e1, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29716728

RESUMO

OBJECTIVE: Reprogramming of fibroblasts into induced cardiomyocytes represents a potential new therapy for heart failure. We hypothesized that inactivation of p63, a p53 gene family member, may help overcome human cell resistance to reprogramming. METHODS: p63 Knockout (-/-) and knockdown murine embryonic fibroblasts (MEFs), p63-/- adult murine cardiac fibroblasts, and human cardiac fibroblasts were assessed for cardiomyocyte-specific feature changes, with or without treatment by the cardiac transcription factors Hand2-Myocardin (HM). RESULTS: Flow cytometry revealed that a significantly greater number of p63-/- MEFs expressed the cardiac-specific marker cardiac troponin T (cTnT) in culture compared with wild-type (WT) cells (38% ± 11% vs 0.9% ± 0.9%, P < .05). HM treatment of p63-/- MEFs increased cTnT expression to 74% ± 3% of cells but did not induce cTnT expression in wild-type murine embryonic fibroblasts. shRNA-mediated p63 knockdown likewise yielded a 20-fold increase in cTnT microRNA expression compared with untreated MEFs. Adult murine cardiac fibroblasts demonstrated a 200-fold increase in cTnT gene expression after inducible p63 knockout and expressed sarcomeric α-actinin as well as cTnT. These p63-/- adult cardiac fibroblasts exhibited calcium transients and electrically stimulated contractions when co-cultured with neonatal rat cardiomyocytes and treated with HM. Increased expression of cTnT and other marker genes was also observed in p63 knockdown human cardiac fibroblasts procured from patients undergoing procedures for heart failure. CONCLUSIONS: Downregulation of p63 facilitates direct cardiac cellular reprogramming and may help overcome the resistance of human cells to reprogramming.


Assuntos
Reprogramação Celular/genética , Fibroblastos/citologia , Inativação Gênica/fisiologia , Miócitos Cardíacos/citologia , Fosfoproteínas/genética , Transativadores/genética , Animais , Células Cultivadas , Humanos , Camundongos , Ratos , Troponina T/análise , Troponina T/metabolismo
3.
J Thorac Cardiovasc Surg ; 153(1): 128-130, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27726873

RESUMO

The prospect of genetically reprogramming cardiac fibroblasts into induced cardiomyocytes by using cardio-differentiating transcription factors represents a significant advantage over previous strategies involving stem cell implantation or the delivery of angiogenic factors. Remarkably, intramyocardial administration of cardio-differentiating factors consistently results in 20% to 30% improvements in postinfarct ejection fraction and nearly a 50% reduction in myocardial fibrosis in murine models. Despite these encouraging observations, few breakthroughs have been made in the reprogramming of human cells, which have more rigorous epigenetic constraints and gene regulatory networks that oppose reprogramming. As a potential solution to this challenge, Cao and colleagues used a cocktail of 9 chemicals capable of reprogramming human fibroblasts into contractile cardiomyocyte-like cells, albeit at a low efficiency. This strategy would obviate the concerns with viral vectors and appears to partially overcome the epigenetic constraints in human cells. Nevertheless, significant challenges, including drug-drug interactions, low reprogramming efficiency, and lack of in vivo data must be overcome before future clinical application.


Assuntos
Transdiferenciação Celular/efeitos dos fármacos , Técnicas de Reprogramação Celular , Reprogramação Celular/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Células Cultivadas , Fibroblastos/metabolismo , Humanos , Miócitos Cardíacos/metabolismo , Fenótipo
5.
J Thorac Cardiovasc Surg ; 153(2): 329-339.e3, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27773576

RESUMO

OBJECTIVE: The reprogramming of cardiac fibroblasts into induced cardiomyocyte-like cells improves ventricular function in myocardial infarction models. Only integrating persistent expression vectors have thus far been used to induce reprogramming, potentially limiting its clinical applicability. We therefore tested the reprogramming potential of nonintegrating, acute expression adenoviral (Ad) vectors. METHODS: Ad or lentivirus vectors encoding Gata4 (G), Mef2c (M), and Tbx5 (T) were validated in vitro. Sprague-Dawley rats then underwent coronary ligation and Ad-mediated administration of vascular endothelial growth factor to generate infarct prevascularization. Three weeks later, animals received Ad or lentivirus encoding G, M, or T (AdGMT or LentiGMT) or an equivalent dose of a null vector (n = 11, 10, and 10, respectively). Outcomes were analyzed by echocardiography, magnetic resonance imaging, and histology. RESULTS: Ad and lentivirus vectors provided equivalent G, M, and T expression in vitro. AdGMT and LentiGMT both likewise induced expression of the cardiomyocyte marker cardiac troponin T in approximately 6% of cardiac fibroblasts versus <1% cardiac troponin T expression in AdNull (adenoviral vector that does not encode a transgene)-treated cells. Infarcted myocardium that had been treated with AdGMT likewise demonstrated greater density of cells expressing the cardiomyocyte marker beta myosin heavy chain 7 compared with AdNull-treated animals. Echocardiography demonstrated that AdGMT and LentiGMT both increased ejection fraction compared with AdNull (AdGMT: 21% ± 3%, LentiGMT: 14% ± 5%, AdNull: -0.4% ± 2%; P < .05). CONCLUSIONS: Ad vectors are at least as effective as lentiviral vectors in inducing cardiac fibroblast transdifferentiation into induced cardiomyocyte-like cells and improving cardiac function in postinfarct rat hearts. Short-term expression Ad vectors may represent an important means to induce cardiac cellular reprogramming in humans.


Assuntos
Técnicas de Reprogramação Celular/métodos , Infarto do Miocárdio/patologia , Miócitos Cardíacos/patologia , Regeneração , Adenoviridae , Animais , Transdiferenciação Celular , Modelos Animais de Doenças , Fibroblastos/metabolismo , Fibroblastos/patologia , Técnicas de Transferência de Genes , Vetores Genéticos , Masculino , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Ratos , Ratos Sprague-Dawley , Fator A de Crescimento do Endotélio Vascular/farmacologia
6.
Artigo em Inglês | MEDLINE | ID: mdl-27554127

RESUMO

OPINION STATEMENT: Direct cardiac cellular reprogramming of endogenous cardiac fibroblasts directly into induced cardiomyocytes is a highly feasible, promising therapeutic option for patients with advanced heart failure. The most successful cardiac reprogramming strategy will likely be a multimodal approach involving an optimal combination of cardio-differentiating factors, suppression of fibroblast gene expression, and induction of angiogenic factors.

7.
J Am Heart Assoc ; 5(11)2016 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-27930352

RESUMO

BACKGROUND: Reprogramming of cardiac fibroblasts into induced cardiomyocyte-like cells represents a promising potential new therapy for treating heart disease, inducing significant improvements in postinfarct ventricular function in rodent models. Because reprogramming factors effective in transdifferentiating rodent cells are not sufficient to reprogram human cells, we sought to identify reprogramming factors potentially applicable to human studies. METHODS AND RESULTS: Lentivirus vectors expressing Gata4, Mef2c, and Tbx5 (GMT); Hand2 (H), Myocardin (My), or microRNA (miR)-590 were administered to rat, porcine, and human cardiac fibroblasts in vitro. induced cardiomyocyte-like cell production was then evaluated by assessing expression of the cardiomyocyte marker, cardiac troponin T (cTnT), whereas signaling pathway studies were performed to identify reprogramming factor targets. GMT administration induced cTnT expression in ≈6% of rat fibroblasts, but failed to induce cTnT expression in porcine or human cardiac fibroblasts. Addition of H/My and/or miR-590 to GMT administration resulted in cTNT expression in ≈5% of porcine and human fibroblasts and also upregulated the expression of the cardiac genes, MYH6 and TNNT2. When cocultured with murine cardiomyocytes, cTnT-expressing porcine cardiac fibroblasts exhibited spontaneous contractions. Administration of GMT plus either H/My or miR-590 alone also downregulated fibroblast genes COL1A1 and COL3A1. miR-590 was shown to directly suppress the zinc finger protein, specificity protein 1 (Sp1), which was able to substitute for miR-590 in inducing cellular reprogramming. CONCLUSIONS: These data support porcine studies as a surrogate for testing human cardiac reprogramming, and suggest that miR-590-mediated repression of Sp1 represents an alternative pathway for enhancing human cardiac cellular reprogramming.


Assuntos
Transdiferenciação Celular/genética , Técnicas de Reprogramação Celular/métodos , Fibroblastos/citologia , MicroRNAs/genética , Miócitos Cardíacos/citologia , Fator de Transcrição Sp1/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Citometria de Fluxo , Fator de Transcrição GATA4/genética , Vetores Genéticos , Humanos , Técnicas In Vitro , Fatores de Transcrição MEF2/genética , Proteínas Nucleares/genética , Ratos , Suínos , Proteínas com Domínio T/genética , Transativadores/genética
8.
J Thorac Dis ; 9(10): 3672-3673, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29268372
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA