Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 14(1): 9795, 2024 04 29.
Artigo em Inglês | MEDLINE | ID: mdl-38684844

RESUMO

Cardiac fibrosis contributes to the development of heart failure, and is the response of cardiac fibroblasts (CFs) to pressure or volume overload. Limiting factors in CFs research are the poor availability of human cells and the tendency of CFs to transdifferentiate into myofibroblasts when cultured in vitro. The possibility to generate CFs from induced pluripotent stem cells (iPSC), providing a nearly unlimited cell source, opens new possibilities. However, the behaviour of iPSC-CFs under mechanical stimulation has not been studied yet. Our study aimed to assess the behaviour of iPSC-CFs under mechanical stretch and pro-fibrotic conditions. First, we confirm that iPSC-CFs are comparable to primary CFs at gene, protein and functional level. Furthermore, iPSC-derived CFs adopt a pro-fibrotic response to transforming growth factor beta (TGF-ß). In addition, mechanical stretch inhibits TGF-ß-induced fibroblast activation in iPSC-CFs. Thus, the responsiveness to cytokines and mechanical stimulation of iPSC-CFs demonstrates they possess key characteristics of primary CFs and may be useful for disease modelling.


Assuntos
Fibroblastos , Células-Tronco Pluripotentes Induzidas , Fator de Crescimento Transformador beta , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Humanos , Fibroblastos/metabolismo , Fibroblastos/citologia , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Estresse Mecânico , Células Cultivadas , Diferenciação Celular , Miocárdio/citologia , Miocárdio/metabolismo , Miofibroblastos/metabolismo , Miofibroblastos/citologia , Fibrose
2.
J Am Coll Cardiol ; 82(8): 704-717, 2023 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-37587582

RESUMO

BACKGROUND: Precapillary pulmonary hypertension (precPH) patients have altered right atrial (RA) function and right ventricular (RV) diastolic stiffness. OBJECTIVES: This study aimed to investigate RA function using pressure-volume (PV) loops, isolated cardiomyocyte, and histological analyses. METHODS: RA PV loops were constructed in control subjects (n = 9) and precPH patients (n = 27) using magnetic resonance and catheterization data. RA stiffness (pressure rise during atrial filling) and right atrioventricular coupling index (RA minimal volume / RV end-diastolic volume) were compared in a larger cohort of patients with moderate (n = 39) or severe (n = 41) RV diastolic stiffness. Cardiomyocytes were isolated from RA tissue collected from control subjects (n = 6) and precPH patients (n = 9) undergoing surgery. Autopsy material was collected from control subjects (n = 6) and precPH patients (n = 4) to study RA hypertrophy, capillarization, and fibrosis. RESULTS: RA PV loops showed 3 RA cardiac phases (reservoir, passive emptying, and contraction) with dilatation and elevated pressure in precPH. PrecPH patients with severe RV diastolic stiffness had increased RA stiffness and worse right atrioventricular coupling index. Cardiomyocyte cross-sectional area was increased 2- to 3-fold in precPH, but active tension generated by the sarcomeres was unaltered. There was no increase in passive tension of the cardiomyocytes, but end-stage precPH showed reduced number of capillaries per mm2 accompanied by interstitial and perivascular fibrosis. CONCLUSIONS: RA PV loops show increased RA stiffness and suggest atrioventricular uncoupling in patients with severe RV diastolic stiffness. Isolated RA cardiomyocytes of precPH patients are hypertrophied, without intrinsic sarcomeric changes. In end-stage precPH, reduced capillary density is accompanied by interstitial and perivascular fibrosis.


Assuntos
Apêndice Atrial , Fibrilação Atrial , Hipertensão Pulmonar , Humanos , Miócitos Cardíacos , Átrios do Coração/diagnóstico por imagem
3.
J Heart Lung Transplant ; 41(4): 445-457, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35039146

RESUMO

BACKGROUND: To investigate the association between altered sex hormone expression and long-term right ventricular (RV) adaptation and progression of right heart failure in a Dutch cohort of Pulmonary Arterial Hypertension (PAH)-patients across a wide range of ages. METHODS: In this study we included 279 PAH-patients, of which 169 females and 110 males. From 59 patients and 21 controls we collected plasma samples for sex hormone analysis. Right heart catheterization (RHC) and/or cardiac magnetic resonance (CMR) imaging was performed at baseline. For longitudinal data analysis, we selected patients that underwent a RHC and/or CMR maximally 1.5 years prior to an event (death or transplantation, N = 49). RESULTS: Dehydroepiandrosterone-sulfate (DHEA-S) levels were reduced in male and female PAH-patients compared to controls, whereas androstenedione and testosterone were only reduced in female patients. Interestingly, low DHEA-S and high testosterone levels were correlated to worse RV function in male patients only. Subsequently, we analyzed prognosis and RV adaptation in females stratified by age. Females ≤45years had best prognosis in comparison to females ≥55years and males. No differences in RV function at baseline were observed, despite higher pressure-overload in females ≤45years. Longitudinal data demonstrated a clear distinction in RV adaptation. Although females ≤45years had an event at a later time point, RV function was more impaired at end-stage disease. CONCLUSIONS: Sex hormones are differently associated with RV function in male and female PAH-patients. DHEA-S appeared to be lower in male and female PAH-patients. Females ≤45years could persevere pressure-overload for a longer time, but had a more severe RV phenotype at end-stage disease.


Assuntos
Hipertensão Arterial Pulmonar , Disfunção Ventricular Direita , Hipertensão Pulmonar Primária Familiar , Feminino , Hormônios Esteroides Gonadais , Ventrículos do Coração/diagnóstico por imagem , Humanos , Masculino , Função Ventricular Direita
4.
Front Cardiovasc Med ; 9: 1063967, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36588576

RESUMO

Background: Pulmonary arterial hypertension (PAH) is a progressive disease characterized by increased pulmonary artery pressure leading to right ventricular (RV) failure. While current PAH therapies improve patient outlook, they show limited benefit in attenuating RV dysfunction. Recent investigations demonstrated that the thromboxane (TX) A2 receptor (TP) antagonist NTP42 attenuates experimental PAH across key hemodynamic parameters in the lungs and heart. This study aimed to validate the efficacy of NTP42:KVA4, a novel oral formulation of NTP42 in clinical development, in preclinical models of PAH while also, critically, investigating its direct effects on RV dysfunction. Methods: The effects of NTP42:KVA4 were evaluated in the monocrotaline (MCT) and pulmonary artery banding (PAB) models of PAH and RV dysfunction, respectively, and when compared with leading standard-of-care (SOC) PAH drugs. In addition, the expression of the TP, the target for NTP42, was investigated in cardiac tissue from several other related disease models, and from subjects with PAH and dilated cardiomyopathy (DCM). Results: In the MCT-PAH model, NTP42:KVA4 alleviated disease-induced changes in cardiopulmonary hemodynamics, pulmonary vascular remodeling, inflammation, and fibrosis, to a similar or greater extent than the PAH SOCs tested. In the PAB model, NTP42:KVA4 improved RV geometries and contractility, normalized RV stiffness, and significantly increased RV ejection fraction. In both models, NTP42:KVA4 promoted beneficial RV adaptation, decreasing cellular hypertrophy, and increasing vascularization. Notably, elevated expression of the TP target was observed both in RV tissue from these and related disease models, and in clinical RV specimens of PAH and DCM. Conclusion: This study shows that, through antagonism of TP signaling, NTP42:KVA4 attenuates experimental PAH pathophysiology, not only alleviating pulmonary pathologies but also reducing RV remodeling, promoting beneficial hypertrophy, and improving cardiac function. The findings suggest a direct cardioprotective effect for NTP42:KVA4, and its potential to be a disease-modifying therapy in PAH and other cardiac conditions.

5.
Cardiovasc Res ; 118(12): 2688-2702, 2022 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-34550326

RESUMO

AIMS: Research on the pathophysiology of right ventricular (RV) failure has, in spite of the associated high mortality and morbidity, lagged behind compared to the left ventricle (LV). Previous work from our lab revealed that the embryonic basic helix-loop-helix transcription factor heart and neural crest derivatives expressed-2 (Hand2) is re-expressed in the adult heart and activates a 'foetal gene programme' contributing to pathological cardiac remodelling under conditions of LV pressure overload. As such, ablation of cardiac expression of Hand2 conferred protection to cardiac stress and abrogated the maladaptive effects that were observed upon increased expression levels. In this study, we aimed to understand the contribution of Hand2 to RV remodelling in response to pressure overload induced by pulmonary artery banding (PAB). METHODS AND RESULTS: In this study, Hand2F/F and MCM- Hand2F/F mice were treated with tamoxifen (control and knockout, respectively) and subjected to six weeks of RV pressure overload induced by PAB. Echocardiographic- and MRI-derived haemodynamic parameters as well as molecular remodelling were assessed for all experimental groups and compared to sham-operated controls. Six weeks after PAB, levels of Hand2 expression increased in the control-banded animals but, as expected, remained absent in the knockout hearts. Despite the dramatic differences in Hand2 expression, pressure overload resulted in impaired cardiac function independently of the genotype. In fact, Hand2 depletion seems to sensitize the RV to pressure overload as these mice develop more hypertrophy and more severe cardiac dysfunction. Higher expression levels of HAND2 were also observed in RV samples of human hearts from patients with pulmonary hypertension. In turn, the LV of RV pressure-overloaded hearts was also dramatically affected as reflected by changes in shape, decreased LV mass, and impaired cardiac function. RNA-sequencing revealed a distinct set of genes that are dysregulated in the pressure-overloaded RV, compared to the previously described pressure-overloaded LV. CONCLUSION: Cardiac-specific depletion of Hand2 is associated with severe cardiac dysfunction in conditions of RV pressure overload. While inhibiting Hand2 expression can prevent cardiac dysfunction in conditions of LV pressure overload, the same does not hold true for conditions of RV pressu re overload. This study highlights the need to better understand the molecular mechanisms driving pathological remodelling of the RV in contrast to the LV, in order to better diagnose and treat patients with RV or LV failure.


Assuntos
Insuficiência Cardíaca , Disfunção Ventricular Direita , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/metabolismo , Ventrículos do Coração/metabolismo , Humanos , Camundongos , RNA/metabolismo , Tamoxifeno/metabolismo , Fatores de Transcrição/metabolismo , Disfunção Ventricular Direita/genética , Disfunção Ventricular Direita/metabolismo , Função Ventricular Direita , Pressão Ventricular , Remodelação Ventricular
6.
Cells ; 10(12)2021 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-34944102

RESUMO

Pulmonary arterial hypertension (PAH) patients eventually die of right heart failure (RHF). Currently, there is no suitable pre-clinical model to study PAH. Therefore, we aim to develop a right heart dysfunction (RHD) model using the 3-dimensional engineered heart tissue (EHT) approach and cardiomyocytes derived from patient-induced pluripotent stem cells (iPSCs) to unravel the mechanisms that determine the fate of a pressure-overloaded right ventricle. iPSCs from PAH and healthy control subjects were differentiated into cardiomyocytes (iPSC-CMs), incorporated into the EHT, and maintained for 28 days. In comparison with control iPSC-CMs, PAH-derived iPSC-CMs exhibited decreased beating frequency and increased contraction and relaxation times. iPSC-CM alignment within the EHT was observed. PAH-derived EHTs exhibited higher force, and contraction and relaxation times compared with control EHTs. Increased afterload was induced using 2× stiffer posts from day 0. Due to high variability, there were no functional differences between normal and stiffer EHTs, and no differences in the hypertrophic gene expression. In conclusion, under baseline spontaneous conditions, PAH-derived iPSC-CMs and EHTs show prolonged contraction compared with controls, as observed clinically in PAH patients. Further optimization of the hypertrophic model and profound characterization may provide a platform for disease modelling and drug screening.


Assuntos
Coração/fisiopatologia , Imageamento Tridimensional , Modelos Cardiovasculares , Hipertensão Arterial Pulmonar/diagnóstico por imagem , Hipertensão Arterial Pulmonar/fisiopatologia , Adulto , Estudos de Casos e Controles , Diferenciação Celular , Feminino , Regulação da Expressão Gênica , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Miócitos Cardíacos/patologia , Hipertensão Arterial Pulmonar/genética , Engenharia Tecidual
8.
Clin Chest Med ; 42(1): 179-194, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33541611

RESUMO

The right ventricle is coupled to the low-pressure pulmonary circulation. In pulmonary vascular diseases, right ventricular (RV) adaptation is key to maintain ventriculoarterial coupling. RV hypertrophy is the first adaptation to diminish RV wall tension, increase contractility, and protect cardiac output. Unfortunately, RV hypertrophy cannot be sustained and progresses toward a maladaptive phenotype, characterized by dilation and ventriculoarterial uncoupling. The mechanisms behind the transition from RV adaptation to RV maladaptation and right heart failure are unraveled. Therefore, in this article, we explain the main traits of each phenotype, and how some early beneficial adaptations become prejudicial in the long-term.


Assuntos
Ventrículos do Coração/fisiopatologia , Hipertrofia Ventricular Direita/fisiopatologia , Disfunção Ventricular Direita/fisiopatologia , Animais , Humanos , Hipertrofia Ventricular Direita/complicações , Camundongos , Ratos , Disfunção Ventricular Direita/complicações
9.
Methods Cell Biol ; 157: 3-21, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32334718

RESUMO

The use of hydrogels derived from the extracellular matrix (ECM) in tissue engineering applications aims to overcome the conundrum of mimicking the complexity of ECM composition in vitro. In this chapter, we describe a method of decellularization and subsequent formation of an ECM-based hydrogel using porcine heart tissue. These decellularized ECM hydrogels could be used to create semi-interpenetrating networks or as bioinks in bioprinting applications to further enhance the bioactivity and increase the biomimicry degree of the biological cardiac constructs.


Assuntos
Matriz Extracelular , Ventrículos do Coração/citologia , Engenharia Tecidual/métodos , Alicerces Teciduais , Animais , Materiais Biomiméticos , Humanos , Hidrogéis , Infarto do Miocárdio/terapia , Suínos
10.
J Vis Exp ; (143)2019 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-30735196

RESUMO

Cardiovascular diseases are the leading cause of death in developed countries. Consequently, the demand for effective cardiac cell therapies has motivated researchers in the stem cell and bioengineering fields to develop in vitro high-fidelity human myocardium for both basic research and clinical applications. However, the immature phenotype of cardiac cells is a limitation on obtaining tissues that functionally mimic the adult myocardium, which is mainly characterized by mechanical and electrical signals. Thus, the purpose of this protocol is to prepare and mature the target cell population through electromechanical stimulation, recapitulating physiological parameters. Cardiac tissue engineering is evolving toward more biological approaches, and strategies based on biophysical stimuli, thus, are gaining momentum. The device developed for this purpose is unique and allows individual or simultaneous electrical and mechanical stimulation, carefully characterized and validated. In addition, although the methodology has been optimized for this stimulator and a specific cell population, it can easily be adapted to other devices and cell lines. The results here offer evidence of the increased cardiac commitment of the cell population after electromechanical stimulation. Electromechanically stimulated cells show an increased expression of main cardiac markers, including early, structural, and calcium-regulating genes. This cell conditioning could be useful for further regenerative cell therapy, disease modeling, and high-throughput drug screening.


Assuntos
Miócitos Cardíacos/fisiologia , Tecido Adiposo/citologia , Animais , Biomarcadores/metabolismo , Células Cultivadas , Estimulação Elétrica , Regulação da Expressão Gênica , Humanos , Miócitos Cardíacos/citologia , Células-Tronco/citologia , Interface Usuário-Computador
11.
Sci Rep ; 8(1): 6906, 2018 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-29703984

RESUMO

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has been fixed in the paper.

12.
Sci Rep ; 8(1): 499, 2018 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-29323152

RESUMO

Mechanical conditioning is incompletely characterized for stimulating therapeutic cells within the physiological range. We sought to unravel the mechanism of action underlying mechanical conditioning of adipose tissue-derived progenitor cells (ATDPCs), both in vitro and in silico. Cardiac ATDPCs, grown on 3 different patterned surfaces, were mechanically stretched for 7 days at 1 Hz. A custom-designed, magnet-based, mechanical stimulator device was developed to apply ~10% mechanical stretching to monolayer cell cultures. Gene and protein analyses were performed for each cell type and condition. Cell supernatants were also collected to analyze secreted proteins and construct an artificial neural network. Gene and protein modulations were different for each surface pattern. After mechanostimulation, cardiac ATDPCs increased the expression of structural genes and there was a rising trend on cardiac transcription factors. Finally, secretome analyses revealed upregulation of proteins associated with both myocardial infarction and cardiac regeneration, such as regulators of the immune response, angiogenesis or cell adhesion. To conclude, mechanical conditioning of cardiac ATDPCs enhanced the expression of early and late cardiac genes in vitro. Additionally, in silico analyses of secreted proteins showed that mechanical stimulation of cardiac ATDPCs was highly associated with myocardial infarction and repair.


Assuntos
Tecido Adiposo/citologia , Células-Tronco/metabolismo , Estresse Mecânico , Células Cultivadas , Conexina 43/metabolismo , Humanos , Fatores de Transcrição MEF2/metabolismo , Miocárdio/citologia , Miocárdio/metabolismo , Redes Neurais de Computação , Proteoma/metabolismo , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Células-Tronco/citologia , Fatores de Transcrição/metabolismo
13.
Tissue Eng Part C Methods ; 23(11): 736-744, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28699384

RESUMO

The combination of biomatrices and induced pluripotent stem cell (iPSC) derivatives to aid repair and myocardial scar formation may soon become a reality for cardiac regenerative medicine. However, the tumor risk associated with residual undifferentiated cells remains an important safety concern of iPSC-based therapies. This concern is not satisfactorily addressed in xenotransplantation, which requires immune suppression of the transplanted animal. In this study, we assessed the safety of transplanting undifferentiated iPSCs in an allogeneic setting. Given that swine are commonly used as large animal models in cardiac medicine, we used porcine iPSCs (p-iPSCs) in conjunction with bioengineered constructs that support recovery after acute myocardial infarction. Histopathology analyses found no evidence of p-iPSCs or p-iPSC-derived cells within the host myocardium or biomatrices after 30 and 90 days of follow-up. Consistent with the disappearance of the implanted cells, we could not observe functional benefit of these treatments in terms of left ventricular ejection fraction, cardiac output, ventricular volumes, or necrosis. We therefore conclude that residual undifferentiated iPSCs should pose no safety concern when used on immune-competent recipients in an allogeneic setting, at least in the context of cardiac regenerative medicine.


Assuntos
Células-Tronco Pluripotentes Induzidas/citologia , Infarto do Miocárdio/patologia , Infarto do Miocárdio/terapia , Transplante de Células-Tronco , Animais , Modelos Animais de Doenças , Testes de Função Cardíaca , Inflamação/patologia , Imageamento por Ressonância Magnética , Infarto do Miocárdio/fisiopatologia , Neovascularização Fisiológica , Reação em Cadeia da Polimerase , Sus scrofa , Engenharia Tecidual , Alicerces Teciduais/química , Transplante Homólogo , Cicatrização
14.
J Cell Mol Med ; 21(11): 3000-3009, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28557183

RESUMO

Idiopathic dilated cardiomyopathy (IDCM) is a frequent cause of heart transplantation. Potentially valuable blood markers are being sought, and low-density lipoprotein receptor-related protein 1 (LRP1) has been linked to the underlying molecular basis of the disease. This study compared circulating levels of soluble LRP1 (sLRP1) in IDCM patients and healthy controls and elucidated whether sLRP1 is exported out of the myocardium through extracellular vesicles (EVs) to gain a better understanding of the pathogenesis of the disease. LRP1 α chain expression was analysed in samples collected from the left ventricles of explanted hearts using immunohistochemistry. sLRP1 concentrations were determined in platelet-free plasma by enzyme-linked immunosorbent assay. Plasma-derived EVs were extracted by size-exclusion chromatography (SEC) and characterized by nanoparticle tracking analysis and cryo-transmission electron microscopy. The distributions of vesicular (CD9, CD81) and myocardial (caveolin-3) proteins and LRP1 α chain were assessed in SEC fractions by flow cytometry. LRP1 α chain was preferably localized to blood vessels in IDCM compared to control myocardium. Circulating sLRP1 was increased in IDCM patients. CD9- and CD81-positive fractions enriched with membrane vesicles with the expected size and morphology were isolated from both groups. The LRP1 α chain was not present in these SEC fractions, which were also positive for caveolin-3. The increase in circulating sLRP1 in IDCM patients may be clinically valuable. Although EVs do not contribute to higher sLRP1 levels in IDCM, a comprehensive analysis of EV content would provide further insights into the search for novel blood markers.


Assuntos
Cardiomiopatia Dilatada/sangue , Vesículas Extracelulares/química , Ventrículos do Coração/metabolismo , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/sangue , Miocárdio/metabolismo , Idoso , Biomarcadores/sangue , Cardiomiopatia Dilatada/diagnóstico , Cardiomiopatia Dilatada/genética , Cardiomiopatia Dilatada/cirurgia , Estudos de Casos e Controles , Caveolina 3/sangue , Caveolina 3/genética , Feminino , Regulação da Expressão Gênica , Transplante de Coração , Ventrículos do Coração/patologia , Humanos , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Masculino , Pessoa de Meia-Idade , Miocárdio/patologia , Tetraspanina 28/sangue , Tetraspanina 28/genética , Tetraspanina 29/sangue , Tetraspanina 29/genética
15.
Stem Cells Transl Med ; 6(3): 970-981, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28297585

RESUMO

Cardiac cells are subjected to mechanical and electrical forces, which regulate gene expression and cellular function. Therefore, in vitro electromechanical stimuli could benefit further integration of therapeutic cells into the myocardium. Our goals were (a) to study the viability of a tissue-engineered construct with cardiac adipose tissue-derived progenitor cells (cardiac ATDPCs) and (b) to examine the effect of electromechanically stimulated cardiac ATDPCs within a myocardial infarction (MI) model in mice for the first time. Cardiac ATDPCs were electromechanically stimulated at 2-millisecond pulses of 50 mV/cm at 1 Hz and 10% stretching during 7 days. The cells were harvested, labeled, embedded in a fibrin hydrogel, and implanted over the infarcted area of the murine heart. A total of 39 animals were randomly distributed and sacrificed at 21 days: groups of grafts without cells and with stimulated or nonstimulated cells. Echocardiography and gene and protein analyses were also carried out. Physiologically stimulated ATDPCs showed increased expression of cardiac transcription factors, structural genes, and calcium handling genes. At 21 days after implantation, cardiac function (measured as left ventricle ejection fraction between presacrifice and post-MI) increased up to 12% in stimulated grafts relative to nontreated animals. Vascularization and integration with the host blood supply of grafts with stimulated cells resulted in increased vessel density in the infarct border region. Trained cells within the implanted fibrin patch expressed main cardiac markers and migrated into the underlying ischemic myocardium. To conclude, synchronous electromechanical cell conditioning before delivery may be a preferred alternative when considering strategies for heart repair after myocardial infarction. Stem Cells Translational Medicine 2017;6:970-981.


Assuntos
Células-Tronco Adultas/transplante , Infarto do Miocárdio/fisiopatologia , Infarto do Miocárdio/terapia , Transplante de Células-Tronco , Células-Tronco Adultas/citologia , Células-Tronco Adultas/efeitos dos fármacos , Animais , Diferenciação Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Fibrina/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Testes de Função Cardíaca , Humanos , Camundongos SCID , Miocárdio/metabolismo , Miocárdio/patologia , Neovascularização Fisiológica/efeitos dos fármacos , Implantação de Prótese , Recuperação de Função Fisiológica/efeitos dos fármacos , Engenharia Tecidual , Remodelação Ventricular/efeitos dos fármacos
16.
Stem Cells Transl Med ; 6(2): 647-655, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28191775

RESUMO

Cardiac tissue engineering, which combines cells and biomaterials, is promising for limiting the sequelae of myocardial infarction (MI). We assessed myocardial function and scar evolution after implanting an engineered bioactive impedance graft (EBIG) in a swine MI model. The EBIG comprises a scaffold of decellularized human pericardium, green fluorescent protein-labeled porcine adipose tissue-derived progenitor cells (pATPCs), and a customized-design electrical impedance spectroscopy (EIS) monitoring system. Cardiac function was evaluated noninvasively by using magnetic resonance imaging (MRI). Scar healing was evaluated by using the EIS system within the implanted graft. Additionally, infarct size, fibrosis, and inflammation were explored by histopathology. Upon sacrifice 1 month after the intervention, MRI detected a significant improvement in left ventricular ejection fraction (7.5% ± 4.9% vs. 1.4% ± 3.7%; p = .038) and stroke volume (11.5 ± 5.9 ml vs. 3 ± 4.5 ml; p = .019) in EBIG-treated animals. Noninvasive EIS data analysis showed differences in both impedance magnitude ratio (-0.02 ± 0.04 per day vs. -0.48 ± 0.07 per day; p = .002) and phase angle slope (-0.18° ± 0.24° per day vs. -3.52° ± 0.84° per day; p = .004) in EBIG compared with control animals. Moreover, in EBIG-treated animals, the infarct size was 48% smaller (3.4% ± 0.6% vs. 6.5% ± 1%; p = .015), less inflammation was found by means of CD25+ lymphocytes (0.65 ± 0.12 vs. 1.26 ± 0.2; p = .006), and a lower collagen I/III ratio was detected (0.49 ± 0.06 vs. 1.66 ± 0.5; p = .019). An EBIG composed of acellular pericardium refilled with pATPCs significantly reduced infarct size and improved cardiac function in a preclinical model of MI. Noninvasive EIS monitoring was useful for tracking differential scar healing in EBIG-treated animals, which was confirmed by less inflammation and altered collagen deposit. Stem Cells Translational Medicine 2017;6:647-655.


Assuntos
Infarto do Miocárdio/cirurgia , Miócitos Cardíacos/transplante , Regeneração , Transplante de Células-Tronco/métodos , Células-Tronco , Engenharia Tecidual/métodos , Alicerces Teciduais , Função Ventricular Esquerda , Remodelação Ventricular , Animais , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Espectroscopia Dielétrica , Modelos Animais de Doenças , Humanos , Imageamento por Ressonância Magnética , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Fenótipo , Recuperação de Função Fisiológica , Células-Tronco/metabolismo , Volume Sistólico , Sus scrofa , Fatores de Tempo
17.
Tissue Eng Part C Methods ; 22(2): 113-124, 2016 02.
Artigo em Inglês | MEDLINE | ID: mdl-26741987

RESUMO

The prolonged ischemia after myocardial infarction leads to a high degree of cardiomyocyte death, which leads to a reduction of normal heart function. Valuable lessons can be learnt from human myocardium and stem cell biology that would help scientists to develop new, effective, safe, and affordable regenerative therapies. In vivo models are of high interest, but their high complexity limits the possibility to analyze specific factors. In vitro models permit analyzing specific factors of tissue physiology or pathophysiology providing accurate approaches that may guide the creation of three-dimensional (3D) engineered cell aggregates. These systems provide a simplistic way to examine individual factors as compared to animal models, and better mimic the reality than 2D models. In this sense, the objective of this work is to better understand the behavior of a human mesenchymal stem cell-like cell line (subcutaneous adipose tissue-derived progenitor cells [subATDPCs], susceptible to be used in cell therapies) when they are embedded in the 3D environment provided by RAD16-I self-assembling peptide (SAP). Specifically, we study the effect in subATDPCs viability, morphology, proliferation, and protein and gene expression of matrix composition (i.e., RGD motif and heparin polysaccharide modifications) in RAD16-I matrix under different media conditions. Results demonstrated that the 3D environment provided by RAD16-I SAP is able to maintain subATDPCs in this new milieu and at the same time its cardiac commitment. Additionally, it has been observed that chemical induction can induce upregulation of cardiac markers, such as TBX5, MEF2C, ACTN1, and GJA1. Therefore, we propose this 3D model as a promising platform to analyze the effect of specific cues that can help improve cell performance for future cell therapy.

18.
JACC Basic Transl Sci ; 1(5): 360-372, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-30167524

RESUMO

Myocardial infarction (MI) remains a dreadful disease around the world, causing irreversible sequelae that shorten life expectancy and reduce quality of life despite current treatment. Here, the authors engineered a cell-enriched myocardial graft, composed of a decellularized myocardial matrix refilled with adipose tissue-derived progenitor cells (EMG-ATDPC). Once applied over the infarcted area in the swine MI model, the EMG-ATDPC improved cardiac function, reduced infarct size, attenuated fibrosis progression, and promoted neovascularization of the ischemic myocardium. The beneficial effects exerted by the EMG-ATDPC and the absence of identified adverse side effects should facilitate its clinical translation as a novel MI therapy in humans.

19.
Stem Cell Res Ther ; 6: 108, 2015 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-26205795

RESUMO

Engineered bioimplants for cardiac repair require functional vascularization and innervation for proper integration with the surrounding myocardium. The aim of this work was to study nerve sprouting and neovascularization in an acellular pericardial-derived scaffold used as a myocardial bioimplant. To this end, 17 swine were submitted to a myocardial infarction followed by implantation of a decellularized human pericardial-derived scaffold. After 30 days, animals were sacrificed and hearts were analyzed with hematoxylin/eosin and Masson's and Gallego's modified trichrome staining. Immunohistochemistry was carried out to detect nerve fibers within the cardiac bioimplant by using ßIII tubulin and S100 labeling. Isolectin B4, smooth muscle actin, CD31, von Willebrand factor, cardiac troponin I, and elastin antibodies were used to study scaffold vascularization. Transmission electron microscopy was performed to confirm the presence of vascular and nervous ultrastructures. Left ventricular ejection fraction (LVEF), cardiac output (CO), stroke volume, end-diastolic volume, end-systolic volume, end-diastolic wall mass, and infarct size were assessed by using magnetic resonance imaging (MRI). Newly formed nerve fibers composed of several amyelinated axons as the afferent nerve endings of the heart were identified by immunohistochemistry. Additionally, neovessel formation occurred spontaneously as small and large isolectin B4-positive blood vessels within the scaffold. In summary, this study demonstrates for the first time the neoformation of vessels and nerves in cell-free cardiac scaffolds applied over infarcted tissue. Moreover, MRI analysis showed a significant improvement in LVEF (P = 0.03) and CO (P = 0.01) and a 43 % decrease in infarct size (P = 0.007).


Assuntos
Infarto do Miocárdio/terapia , Pericárdio/transplante , Animais , Vasos Coronários/fisiologia , Imuno-Histoquímica , Imageamento por Ressonância Magnética , Infarto do Miocárdio/patologia , Miocárdio/metabolismo , Neovascularização Patológica , Pericárdio/citologia , Pericárdio/metabolismo , Proteínas S100/metabolismo , Suínos , Alicerces Teciduais , Tubulina (Proteína)/metabolismo , Função Ventricular Esquerda/fisiologia
20.
Stem Cells Transl Med ; 4(8): 956-66, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26106218

RESUMO

Considerable research has been dedicated to restoring myocardial cell slippage and limiting ventricular remodeling after myocardial infarction (MI). We examined the ability of a three-dimensional (3D) engineered fibrin patch filled with human umbilical cord blood-derived mesenchymal stem cells (UCBMSCs) to induce recovery of cardiac function after MI. The UCBMSCs were modified to coexpress luciferase and fluorescent protein reporters, mixed with fibrin, and applied as an adhesive, viable construct (fibrin-cell patch) over the infarcted myocardium in mice (MI-UCBMSC group). The patch adhered well to the heart. Noninvasive bioluminescence imaging demonstrated early proliferation and differentiation of UCBMSCs within the construct in the postinfarct mice in the MI-UCBMSC group. The implanted cells also participated in the formation of new, functional microvasculature that connected the fibrin-cell patch to both the subjacent myocardial tissue and the host circulatory system. As revealed by echocardiography, the left ventricular ejection fraction and fractional shortening at sacrifice were improved in MI-UCBMSC mice and were markedly reduced in mice treated with fibrin alone and untreated postinfarction controls. In conclusion, a 3D engineered fibrin patch composed of UCBMSCs attenuated infarct-derived cardiac dysfunction when transplanted locally over a myocardial wound.


Assuntos
Fibrina/uso terapêutico , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Infarto do Miocárdio/terapia , Animais , Diferenciação Celular/genética , Sangue Fetal/citologia , Humanos , Camundongos , Infarto do Miocárdio/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...