Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 771
Filtrar
1.
J Mol Cell Cardiol ; 187: 90-100, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38331557

RESUMEN

Cardiac regenerative therapy using human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) is expected to become an alternative to heart transplantation for severe heart failure. It is now possible to produce large numbers of human pluripotent stem cells (hPSCs) and eliminate non-cardiomyocytes, including residual undifferentiated hPSCs, which can cause teratoma formation after transplantation. There are two main strategies for transplanting hPSC-CMs: injection of hPSC-CMs into the myocardium from the epicardial side, and implantation of hPSC-CM patches or engineered heart tissues onto the epicardium. Transplantation of hPSC-CMs into the myocardium of large animals in a myocardial infarction model improved cardiac function. The engrafted hPSC-CMs matured, and microvessels derived from the host entered the graft abundantly. Furthermore, as less invasive methods using catheters, injection into the coronary artery and injection into the myocardium from the endocardium side have recently been investigated. Since transplantation of hPSC-CMs alone has a low engraftment rate, various methods such as transplantation with the extracellular matrix or non-cardiomyocytes and aggregation of hPSC-CMs have been developed. Post-transplant arrhythmias, imaging of engrafted hPSC-CMs, and immune rejection are the remaining major issues, and research is being conducted to address them. The clinical application of cardiac regenerative therapy using hPSC-CMs has just begun and is expected to spread widely if its safety and efficacy are proven in the near future.


Asunto(s)
Insuficiencia Cardíaca , Células Madre Pluripotentes Inducidas , Células Madre Pluripotentes , Animales , Humanos , Diferenciación Celular , Miocardio , Miocitos Cardíacos/trasplante , Insuficiencia Cardíaca/terapia
2.
J Mol Cell Cardiol ; 183: 14-21, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37595498

RESUMEN

Despite the dramatic improvements in the management of patients with chronic heart failure which have occurred over the last decades, some of them still exhaust conventional drug-based therapies without being eligible for more aggressive options like heart transplantation or implantation of a left ventricular assist device. Cell therapy has thus emerged as a possible means of filling this niche. Multiple cell types have now been tested both in the laboratory but also in the clinics and it is fair to acknowledge that none of the clinical trials have yet conclusively proven the efficacy of cell-based approaches. These clinical studies, however, have entailed the use of cells from various sources but of non-cardiac lineage origins. Although this might not be the main reason for their failures, the discovery of pluripotent stem cells capable of generating cardiomyocytes now raises the hope that such cardiac-committed cells could be therapeutically more effective. In this review, we will first describe where we currently are with regard to the clinical trials using PSC-differentiated cells and discuss the main issues which remain to be addressed. In parallel, because the capacity of cells to stably engraft in the recipient heart has increasingly been questioned, it has been hypothesized that a major mechanism of action could be the cell-triggered release of biomolecules that foster host-associated reparative pathways. Thus, in the second part of this review, we will discuss the rationale, clinically relevant advantages and pitfalls associated with the use of these PSC "products".


Asunto(s)
Procedimientos Quirúrgicos Cardíacos , Insuficiencia Cardíaca , Células Madre Pluripotentes , Humanos , Células Madre Embrionarias , Insuficiencia Cardíaca/terapia , Miocitos Cardíacos/trasplante , Trasplante de Células Madre
3.
J Mol Cell Cardiol ; 182: 75-85, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37482238

RESUMEN

Cardiovascular disease is the leading cause of mortality and morbidity worldwide. Despite improvements in the standard of care for patients with heart diseases, including innovation in pharmacotherapy and surgical interventions, none have yet been proven effective to prevent the progression to heart failure. Cardiac transplantation is the last resort for patients with severe heart failure, but donor shortages remain a roadblock. Cardiac regenerative strategies include cell-based therapeutics, gene therapy, direct reprogramming of non-cardiac cells, acellular biologics, and tissue engineering methods to restore damaged hearts. Significant advancements have been made over the past several decades within each of these fields. This review focuses on the advancements of: 1) cell-based cardiac regenerative therapies, 2) the use of noncoding RNA to induce endogenous cell proliferation, and 3) application of bioengineering methods to promote retention and integration of engrafted cells. Different cell sources have been investigated, including adult stem cells derived from bone marrow and adipose cells, cardiosphere-derived cells, skeletal myoblasts, and pluripotent stem cells. In addition to cell-based transplantation approaches, there have been accumulating interest over the past decade in inducing endogenous CM proliferation for heart regeneration, particularly with the use of noncoding RNAs such as miRNAs and lncRNAs. Bioengineering applications have focused on combining cell-transplantation approaches with fabrication of a porous, vascularized scaffold using biomaterials and advanced bio-fabrication techniques that may offer enhanced retention of transplanted cells, with the hope that these cells would better engraft with host tissue to improve cardiac function. This review summarizes the present status and future challenges of cardiac regenerative therapies.


Asunto(s)
Enfermedades Cardiovasculares , Cardiopatías , Insuficiencia Cardíaca , Adulto , Humanos , Miocitos Cardíacos/trasplante , Trasplante de Células Madre/métodos , Cardiopatías/genética
4.
Circulation ; 146(15): 1159-1169, 2022 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-36073365

RESUMEN

BACKGROUND: Transplantation of pluripotent stem cell-derived cardiomyocytes represents a promising therapeutic strategy for cardiac regeneration, and the first clinical studies in patients with heart failure have commenced. Yet, little is known about the mechanism of action underlying graft-induced benefits. Here, we explored whether transplanted cardiomyocytes actively contribute to heart function. METHODS: We injected cardiomyocytes with an optogenetic off-on switch in a guinea pig cardiac injury model. RESULTS: Light-induced inhibition of engrafted cardiomyocyte contractility resulted in a rapid decrease of left ventricular function in ≈50% (7/13) animals that was fully reversible with the offset of photostimulation. CONCLUSIONS: Our optogenetic approach demonstrates that transplanted cardiomyocytes can actively participate in heart function, supporting the hypothesis that the delivery of new force-generating myocardium can serve as a regenerative therapeutic strategy.


Asunto(s)
Miocitos Cardíacos , Células Madre Pluripotentes , Animales , Diferenciación Celular/fisiología , Cobayas , Miocardio , Miocitos Cardíacos/trasplante , Células Madre Pluripotentes/fisiología , Función Ventricular Izquierda
5.
Stem Cell Reports ; 17(5): 1170-1182, 2022 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-35427484

RESUMEN

A rotating wall vessel (RWV) bioreactor was constructed for growing massive functional cardiac constructs to recover the function of a distressed rat heart. Three-dimensional cardiac tissues were engineered by seeding human-induced pluripotent stem cell-derived cardiomyocytes on poly(lactic-co-glycolic acid) fiber sheets (3D-hiPSC-CTs) and cultured in the RWV bioreactor (RWV group) or under static conditions (control group). The tissues were transplanted into a myocardial infarction nude rat model, and cardiac performance was evaluated. In the RWV group, cell viability and contractile and electrical properties significantly improved, mature cardiomyocytes were observed, and mechanical stress-related mediators of mammalian target of rapamycin signaling were upregulated compared with those of the control. Four weeks post-transplantation, tissue survival and left ventricular ejection fraction significantly improved in the RWV group. Hence, dynamic culture in an RWV bioreactor could provide a superior culture environment for improved performance of 3D-hiPSC-CTs, providing a means for functional cardiomyogenesis in myocyte-loss heart failure.


Asunto(s)
Infarto del Miocardio , Función Ventricular Izquierda , Animales , Reactores Biológicos , Mamíferos , Infarto del Miocardio/terapia , Miocitos Cardíacos/trasplante , Ratas , Ratas Desnudas , Volumen Sistólico , Ingeniería de Tejidos/métodos
6.
J Mol Cell Cardiol ; 166: 1-10, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35081367

RESUMEN

Myocardial injury leads to an irreversible loss of cardiomyocytes (CM). The implantation of human engineered heart tissue (EHT) has become a promising regenerative approach. Previous studies exhibited beneficial, dose-dependent effects of human induced pluripotent stem cell (hiPSC)-derived EHT patch transplantation in a guinea pig model in the subacute phase of myocardial injury. Yet, advanced heart failure often results from a chronic remodeling process. Therefore, from a clinical standpoint it is worthwhile to explore the ability to repair the chronically injured heart. In this study human EHT patches were generated from hiPSC-derived CMs (15 × 106 cells) and implanted epicardially four weeks after injury in a guinea pig cryo-injury model. Cardiac function was evaluated by echocardiography after a follow-up period of four weeks. Hearts revealed large transmural myocardial injuries amounting to 27% of the left ventricle. EHT recipient hearts demonstrated compact muscle islands of human origin in the scar region, as indicated by a positive staining for human Ku80 and dystrophin, remuscularizing 5% of the scar area. Echocardiographic analysis demonstrated no significant functional difference between animals that received EHT patches and animals in the cell-free control group (fractional area change 36% vs. 34%). Thus, EHT patches engrafted in the chronically injured heart but in contrast to the subacute model, grafts were smaller and EHT patch transplantation did not improve left ventricular function, highlighting the difficulties for a regenerative approach.


Asunto(s)
Células Madre Pluripotentes Inducidas , Animales , Cicatriz , Cobayas , Ventrículos Cardíacos , Humanos , Miocitos Cardíacos/trasplante , Ingeniería de Tejidos/métodos
7.
J Mol Cell Cardiol ; 162: 97-109, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34487753

RESUMEN

Acute engraftment arrhythmias (EAs) remain a serious complication of remuscularization therapy. Preliminary evidence suggests that a focal source underlies these EAs stemming from the automaticity of immature pluripotent stem cell-derived cardiomyocytes (PSC-CMs) in nascent myocardial grafts. How these EAs arise though during early engraftment remains unclear. In a series of in silico experiments, we probed the origin of EAs-exploring aspects of altered impulse formation and altered impulse propagation within nascent PSC-CM grafts and at the host-graft interface. To account for poor gap junctional coupling during early PSC-CM engraftment, the voltage dependence of gap junctions and the possibility of ephaptic coupling were incorporated. Inspired by cardiac development, we also studied the contributions of another feature of immature PSC-CMs, circumferential sodium channel (NaCh) distribution in PSC-CMs. Ectopic propagations emerged from nascent grafts of immature PSC-CMs at a rate of <96 bpm. Source-sink effects dictated this rate and contributed to intermittent capture between host and graft. Moreover, ectopic beats emerged from dynamically changing sites along the host-graft interface. The latter arose in part because circumferential NaCh distribution in PSC-CMs contributed to preferential conduction slowing and block of electrical impulses from host to graft myocardium. We conclude that additional mechanisms, in addition to focal ones, contribute to EAs and recognize that their relative contributions are dynamic across the engraftment process.


Asunto(s)
Miocitos Cardíacos , Células Madre Pluripotentes , Diferenciación Celular , Simulación por Computador , Miocardio , Miocitos Cardíacos/trasplante
8.
Pathol Res Pract ; 229: 153701, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34872024

RESUMEN

Treating cardiovascular diseases with cardiac stem cells (CSCs) is a valid treatment among various stem cell-based therapies. With supplying the physiological need for cardiovascular cells as their main function, under pathological circumstances, CSCs can also reproduce the myocardial cells. Although studies have identified many of CSCs' functions, our knowledge of molecular pathways that regulate these functions is not complete enough. Either physiological or pathological studies have shown, stem cells proliferation and differentiation could be regulated by microRNAs (miRNAs). How miRNAs regulate CSC behavior is an interesting area of research that can help us study and control the function of these cells in vitro; an achievement that may be beneficial for patients with cardiovascular diseases. The secretome of stem and progenitor cells has been studied and it has been determined that exosomes are the main source of their secretion which are very small vesicles at the nanoscale and originate from endosomes, which are secreted into the extracellular space and act as key signaling organelles in intercellular communication. Mesenchymal stem cells, cardiac-derived progenitor cells, embryonic stem cells, induced pluripotent stem cells (iPSCs), and iPSC-derived cardiomyocytes release exosomes that have been shown to have cardioprotective, immunomodulatory, and reparative effects. Herein, we summarize the regulation roles of miRNAs and exosomes in cardiac stem cells.


Asunto(s)
Enfermedades Cardiovasculares/cirugía , Exosomas/fisiología , Cardiopatías/cirugía , MicroARNs/fisiología , Miocitos Cardíacos/trasplante , Trasplante de Células Madre , Animales , Humanos , Miocitos Cardíacos/citología
9.
J Am Coll Cardiol ; 78(21): 2092-2105, 2021 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-34794691

RESUMEN

This paper aims to provide an important update on the recent preclinical and clinical trials using cell therapy strategies and engineered heart tissues for the treatment of postinfarction left ventricular remodeling and heart failure. In addition to the authors' own works and opinions on the roadblocks of the field, they discuss novel approaches for cardiac remuscularization via the activation of proliferative mechanisms in resident cardiomyocytes or direct reprogramming of somatic cells into cardiomyocytes. This paper's main mindset is to present current and future strategies in light of their implications for the design of future patient trials with the ultimate objective of facilitating the translation of discoveries in regenerative myocardial therapies to the clinic.


Asunto(s)
Insuficiencia Cardíaca/terapia , Infarto del Miocardio/terapia , Regeneración/fisiología , Medicina Regenerativa/métodos , Investigación Biomédica Traslacional/métodos , Remodelación Ventricular/fisiología , Animales , Prótesis Vascular/tendencias , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Tratamiento Basado en Trasplante de Células y Tejidos/tendencias , Insuficiencia Cardíaca/fisiopatología , Humanos , Infarto del Miocardio/fisiopatología , Miocitos Cardíacos/fisiología , Miocitos Cardíacos/trasplante , Medicina Regenerativa/tendencias , Literatura de Revisión como Asunto , Investigación Biomédica Traslacional/tendencias
10.
Sci Rep ; 11(1): 22155, 2021 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-34772964

RESUMEN

The epicardial administration of therapeutics via the pericardial sac offers an attractive route, since it is minimally invasive and carries no risks of coronary embolization. The aim of this study was to assess viability, safety and effectiveness of cardiosphere-derived cells (CDCs), their extracellular vesicles (EVs) or placebo administered via a mini-thoracotomy 72 h after experimental infarction in swine. The epicardial administration was completed successfully in all cases in a surgery time (knife-to-skin) below 30 min. No significant differences between groups were found in cardiac function parameters evaluated using magnetic resonance imaging before therapy and at the end of the study, despite a trend towards improved function in CDC-treated animals. Moreover, infarct size at 10 weeks was smaller in treated animals, albeit not significantly. Arrhythmia inducibility did not differ between groups. Pathological examination showed no differences, nor were there any pericardial adhesions evidenced in any case 10 weeks after surgery. These results show that the epicardial delivery of CDCs or their EVs is safe and technically easy 3 days after experimental myocardial infarction in swine, but it does not appear to have any beneficial effect on cardiac function. Our results do not support clinical translation of these therapies as implemented in this work.


Asunto(s)
Vesículas Extracelulares , Infarto del Miocardio/terapia , Miocitos Cardíacos/citología , Animales , Modelos Animales de Enfermedad , Femenino , Imagen por Resonancia Magnética , Infarto del Miocardio/diagnóstico por imagen , Infarto del Miocardio/patología , Miocitos Cardíacos/trasplante , Pericardio/patología , Esferoides Celulares , Sus scrofa , Toracotomía , Trasplante Autólogo
11.
Stem Cell Reports ; 16(10): 2473-2487, 2021 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-34506727

RESUMEN

Heart failure remains a significant cause of morbidity and mortality following myocardial infarction. Cardiac remuscularization with transplantation of human pluripotent stem cell-derived cardiomyocytes is a promising preclinical therapy to restore function. Recent large animal data, however, have revealed a significant risk of engraftment arrhythmia (EA). Although transient, the risk posed by EA presents a barrier to clinical translation. We hypothesized that clinically approved antiarrhythmic drugs can prevent EA-related mortality as well as suppress tachycardia and arrhythmia burden. This study uses a porcine model to provide proof-of-concept evidence that a combination of amiodarone and ivabradine can effectively suppress EA. None of the nine treated subjects experienced the primary endpoint of cardiac death, unstable EA, or heart failure compared with five out of eight (62.5%) in the control cohort (hazard ratio = 0.00; 95% confidence interval: 0-0.297; p = 0.002). Pharmacologic treatment of EA may be a viable strategy to improve safety and allow further clinical development of cardiac remuscularization therapy.


Asunto(s)
Amiodarona/uso terapéutico , Arritmias Cardíacas/tratamiento farmacológico , Ivabradina/uso terapéutico , Infarto del Miocardio/tratamiento farmacológico , Miocitos Cardíacos/trasplante , Trasplante de Células Madre/efectos adversos , Taquicardia/tratamiento farmacológico , Animales , Antiarrítmicos/uso terapéutico , Línea Celular , Tratamiento Basado en Trasplante de Células y Tejidos/efectos adversos , Modelos Animales de Enfermedad , Combinación de Medicamentos , Humanos , Masculino , Células Madre Pluripotentes/trasplante , Porcinos
12.
Med Sci Monit ; 27: e931063, 2021 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-34381009

RESUMEN

Stem cells have significant potential use in tissue regeneration, especially for treating cardiac diseases because of their multi-directional differentiation capability. By mimicking the in vivo physiological environment of native cardiomyocytes during their development and maturation, researchers have been able to induce pluripotent stem cell-derived cardiomyocytes (PSC-CMs) at high purity. However, the phenotype of these PSC-CMs is immature compared with that of adult cardiomyocytes. Various strategies have been explored to improve the maturity of PSC-CMs, such as long-term culturing, mechanical stimuli, chemical stimuli, and combinations of these strategies. Among these strategies, mechanical stretch as a key mechanical stimulus plays an important role in PSC-CM maturation. In this review, the optimal parameters of mechanical stretch, the effects of mechanical stretch on maturation of PSC-CMs, underlying molecular mechanisms as well as existing problems are discussed. Mechanical stretch is a powerful approach to promote the maturation of SC-CMs in terms of morphology, structure, and functionality. Nonetheless, further research efforts are needed to reach a satisfactory standard for clinical applications of PSC-CMs in treating cardiac diseases.


Asunto(s)
Diferenciación Celular/fisiología , Miocitos Cardíacos/fisiología , Células Madre Pluripotentes/fisiología , Técnicas de Cultivo de Célula/métodos , Técnicas de Cultivo de Célula/tendencias , Cardiopatías/terapia , Humanos , Miocitos Cardíacos/trasplante , Estrés Mecánico
13.
Proc Natl Acad Sci U S A ; 118(28)2021 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-34244428

RESUMEN

The emerging field of regenerative cell therapy is still limited by the few cell types that can reliably be differentiated from pluripotent stem cells and by the immune hurdle of commercially scalable allogeneic cell therapeutics. Here, we show that gene-edited, immune-evasive cell grafts can survive and successfully treat diseases in immunocompetent, fully allogeneic recipients. Transplanted endothelial cells improved perfusion and increased the likelihood of limb preservation in mice with critical limb ischemia. Endothelial cell grafts transduced to express a transgene for alpha1-antitrypsin (A1AT) successfully restored physiologic A1AT serum levels in mice with genetic A1AT deficiency. This cell therapy prevented both structural and functional changes of emphysematous lung disease. A mixture of endothelial cells and cardiomyocytes was injected into infarcted mouse hearts, and both cell types orthotopically engrafted in the ischemic areas. Cell therapy led to an improvement in invasive hemodynamic heart failure parameters. Our study supports the development of hypoimmune, universal regenerative cell therapeutics for cost-effective treatments of major diseases.


Asunto(s)
Enfermedades Cardiovasculares/inmunología , Enfermedades Cardiovasculares/terapia , Inmunocompetencia , Células Madre Pluripotentes Inducidas/inmunología , Enfermedades Pulmonares/inmunología , Enfermedades Pulmonares/terapia , Trasplante de Células Madre , Animales , Células Endoteliales/trasplante , Insuficiencia Cardíaca/terapia , Miembro Posterior/irrigación sanguínea , Miembro Posterior/patología , Isquemia/patología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Miocitos Cardíacos/trasplante , Trasplante Homólogo , alfa 1-Antitripsina/metabolismo
14.
Cell Death Dis ; 12(7): 677, 2021 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-34226515

RESUMEN

Muscular dystrophies are debilitating neuromuscular disorders for which no cure exists. As this disorder affects both cardiac and skeletal muscle, patients would benefit from a cellular therapy that can simultaneously regenerate both tissues. The current protocol to derive bipotent mesodermal progenitors which can differentiate into cardiac and skeletal muscle relies on the spontaneous formation of embryoid bodies, thereby hampering further clinical translation. Additionally, as skeletal muscle is the largest organ in the human body, a high myogenic potential is necessary for successful regeneration. Here, we have optimized a protocol to generate chemically defined human induced pluripotent stem cell-derived mesodermal progenitors (cdMiPs). We demonstrate that these cells contribute to myotube formation and differentiate into cardiomyocytes, both in vitro and in vivo. Furthermore, the addition of valproic acid, a clinically approved small molecule, increases the potential of the cdMiPs to contribute to myotube formation that can be prevented by NOTCH signaling inhibitors. Moreover, valproic acid pre-treated cdMiPs injected in dystrophic muscles increase physical strength and ameliorate the functional performances of transplanted mice. Taken together, these results constitute a novel approach to generate mesodermal progenitors with enhanced myogenic potential using clinically approved reagents.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Mesodermo/efectos de los fármacos , Desarrollo de Músculos/efectos de los fármacos , Fibras Musculares Esqueléticas/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Receptores Notch/metabolismo , Ácido Valproico/farmacología , Animales , Linaje de la Célula , Células Cultivadas , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Femenino , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/trasplante , Masculino , Mesodermo/citología , Mesodermo/metabolismo , Mesodermo/trasplante , Ratones , Ratones Noqueados , Contracción Muscular , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/trasplante , Fuerza Muscular , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiopatología , Distrofias Musculares/genética , Distrofias Musculares/metabolismo , Distrofias Musculares/fisiopatología , Distrofias Musculares/cirugía , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/trasplante , Fenotipo , Ratas , Transducción de Señal
15.
Methods Mol Biol ; 2320: 285-293, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34302665

RESUMEN

Myocardial infarction is caused by a lack of oxygen due to blockage of a coronary artery and is a common cause of heart failure. Despite therapeutic advances, the prognosis of patients with heart failure is poor. One of the reasons is that present therapeutic approaches do not restore the loss of cardiac tissue. Stem cell-based therapies have the potential to regenerate the myocardium, and numerous studies using stem cells have shown improved cardiac function and reduced infarct size. In this chapter, we describe our methodology for transplanting human induced pluripotent stem cell-derived cardiomyocytes into immunodeficient mouse hearts with myocardial infarction.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Miocitos Cardíacos/trasplante , Animales , Modelos Animales de Enfermedad , Corazón/fisiología , Humanos , Inyecciones Intramusculares , Ligadura , Masculino , Ratones , Ratones Endogámicos NOD , Infarto del Miocardio/terapia , Regeneración , Respiración Artificial/métodos , Respiración Artificial/veterinaria , Toracotomía/métodos , Toracotomía/veterinaria
16.
Methods Mol Biol ; 2320: 295-302, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34302666

RESUMEN

Recent evidence has provided exciting proof of concepts for the use of pluripotent stem cell-derived cardiomyocytes (PSC-CMs) for cardiac repair; however, large animal studies, which better reflect human disease, are required for clinical application. Here, we describe how to create myocardial infarction in cynomolgus monkey followed by transplantation of PSC-CMs. This method ensures the establishment of a myocardial infarction model and enables reliable PSC-CM transplantation.


Asunto(s)
Modelos Animales de Enfermedad , Células Madre Pluripotentes Inducidas/citología , Macaca fascicularis , Infarto del Miocardio/terapia , Miocitos Cardíacos/trasplante , Anestesia por Inhalación/métodos , Anestesia por Inhalación/veterinaria , Animales , Atropina/uso terapéutico , Bradicardia/tratamiento farmacológico , Bradicardia/prevención & control , Células Cultivadas , Complicaciones Intraoperatorias/tratamiento farmacológico , Complicaciones Intraoperatorias/prevención & control , Ligadura
17.
J Heart Lung Transplant ; 40(8): 767-777, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34108109

RESUMEN

BACKGROUND: Although induced pluripotent stem (iPS) cell-derived cardiac constructs may have a potential in cardiomyogenesis of a distressed myocardium, obtaining polarity in cardiac constructs, such as via myocyte alignment, may be crucial to achieve a maximum contractile force for better clinical outcomes. We herein hypothesized that transplantation of an aligned cardiac tissue derived from iPS cells has therapeutic effects in a porcine ischemic cardiomyopathy model as a preclinical trial. METHODS: Aligned cardiac tissues were developed by culturing high-purity iPS cell-derived cardiomyocytes in xeno-free conditions and transplanting them into infarct porcine hearts (iPS-CM group, n = 7; control, n = 6). Three months after treatment, therapeutic efficacy was evaluated functionally and histologically. RESULTS: In vitro assessment revealed that the aligned cardiac tissue containing high purity cardiomyocytes contracted homogeneously and had excellent mechanical properties. In the in vivo study, the left ventricular ejection fraction of the iPS-CM group was significantly greater than that of the control group, 3 months after transplantation (37.8% ± 2.3% vs 28.3% ± 2.5%, p < 0.05). Pathologically, attenuated interstitial fibrosis, attenuation of hypertrophied cardiomyocytes, and an increased capillary density were also prominent in the iPS-CM group. A limited amount of engraftment of the transplanted tissue maintaining tissue alignment was observed at 2 weeks after transplantation. CONCLUSIONS: The creation of large-scale functional aligned cardiac tissue was feasible, and the transplantation of the aligned tissue improved cardiac function with angiogenesis and antifibrotic effects in a porcine cardiomyopathy model.


Asunto(s)
Cardiomiopatías/terapia , Células Madre Pluripotentes Inducidas/trasplante , Isquemia Miocárdica/terapia , Miocitos Cardíacos/trasplante , Ingeniería de Tejidos/métodos , Remodelación Ventricular/fisiología , Animales , Cardiomiopatías/diagnóstico , Cardiomiopatías/fisiopatología , Diferenciación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Isquemia Miocárdica/diagnóstico , Isquemia Miocárdica/fisiopatología , Porcinos
18.
Nat Commun ; 12(1): 3155, 2021 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-34039977

RESUMEN

Compact cardiomyocytes that make up the ventricular wall of the adult heart represent an important therapeutic target population for modeling and treating cardiovascular diseases. Here, we established a differentiation strategy that promotes the specification, proliferation and maturation of compact ventricular cardiomyocytes from human pluripotent stem cells (hPSCs). The cardiomyocytes generated under these conditions display the ability to use fatty acids as an energy source, a high mitochondrial mass, well-defined sarcomere structures and enhanced contraction force. These ventricular cells undergo metabolic changes indicative of those associated with heart failure when challenged in vitro with pathological stimuli and were found to generate grafts consisting of more mature cells than those derived from immature cardiomyocytes following transplantation into infarcted rat hearts. hPSC-derived atrial cardiomyocytes also responded to the maturation cues identified in this study, indicating that the approach is broadly applicable to different subtypes of the heart. Collectively, these findings highlight the power of recapitulating key aspects of embryonic and postnatal development for generating therapeutically relevant cell types from hPSCs.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Insuficiencia Cardíaca/terapia , Infarto del Miocardio/terapia , Miocitos Cardíacos/trasplante , Células Madre Pluripotentes/fisiología , Animales , Diferenciación Celular , Línea Celular , Proliferación Celular , Modelos Animales de Enfermedad , Embrión de Mamíferos , Desarrollo Embrionario/fisiología , Atrios Cardíacos/citología , Atrios Cardíacos/embriología , Insuficiencia Cardíaca/patología , Ventrículos Cardíacos/citología , Ventrículos Cardíacos/embriología , Ventrículos Cardíacos/patología , Humanos , Infarto del Miocardio/complicaciones , Infarto del Miocardio/patología , Miocitos Cardíacos/fisiología , Ratas
19.
Cell Prolif ; 54(6): e13041, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33942933

RESUMEN

OBJECTIVES: Myocardial infarction (MI) is the most predominant type of cardiovascular diseases with high mortality and morbidity. Stem cell therapy, especially cardiac progenitor cell therapy, has been proposed as a promising approach for cardiac regeneration and MI treatment. Previously, we have successfully generated cardiac progenitor-like cells, induced cardiosphere (iCS), via somatic reprogramming. However, the genome integration characteristic of virus-based reprogramming approach hampered their therapeutic applications due to the risk of tumour formation. In the current study, we aim to establish a safer iCS generation strategy with transgene-free approaches. MATERIALS AND METHODS: Four transgene-free approaches for somatic reprogramming, including episome, minicircle, self-replicative RNA, and sendai virus, were compared, from the perspective of cardiac progenitor marker expression, iCS formation, and cardiac differentiation. The therapeutic effects were assessed in the mouse model of MI, from the perspective of survival rate, cardiac function, and structural alterations. RESULTS: The self-replicative RNA approach produced more iCS, which had cardiomyocyte differentiation ability and therapeutic effects on the mouse model of MI with comparable levels with endogenous cardiospheres and iCS generated with retrovirus. In addition, the CXCR4 (C-X-C chemokine receptor 4) positive subpopulation of iCS derived cells (iCSDC) delivered by intravenous injection was found to have similar therapeutic effects with intramyocardial injection on the mouse model of MI, representing a safer delivery approach. CONCLUSION: Thus, the optimized strategy for iCS generation is safer and has more therapeutic potentials.


Asunto(s)
Técnicas de Reprogramación Celular , Infarto del Miocardio/terapia , Miocitos Cardíacos/citología , Receptores CXCR4/análisis , Trasplante de Células Madre , Animales , Diferenciación Celular , Células Cultivadas , Reprogramación Celular , Fibroblastos/citología , Ratones , Miocitos Cardíacos/trasplante , Células Madre/citología
20.
Sci Rep ; 11(1): 8666, 2021 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-33883598

RESUMEN

Cell therapy limits ischemic injury following myocardial infarction (MI) by preventing cell death, modulating the immune response, and promoting tissue regeneration. The therapeutic efficacy of cardiosphere-derived cells (CDCs) and mesenchymal stem cells (MSCs) is associated with extracellular vesicle (EV) release. Prior head-to-head comparisons have shown CDCs to be more effective than MSCs in MI models. Despite differences in cell origin, it is unclear why EVs from different adult stem cell populations elicit differences in therapeutic efficacy. Here, we compare EVs derived from multiple human MSC and CDC donors using diverse in vitro and in vivo assays. EV membrane protein and non-coding RNA composition are highly specific to the parent cell type; for example, miR-10b is enriched in MSC-EVs relative to CDC-EVs, while Y RNA fragments follow the opposite pattern. CDC-EVs enhance the Arg1/Nos2 ratio in macrophages in vitro and reduce MI size more than MSC-EVs and suppress inflammation during acute peritonitis in vivo. Thus, CDC-EVs are distinct from MSC-EVs, confer immunomodulation, and protect the host against ischemic myocardial injury and acute inflammation.


Asunto(s)
Vesículas Extracelulares/metabolismo , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Miocitos Cardíacos/metabolismo , ARN no Traducido/metabolismo , Adulto , Animales , Niño , Preescolar , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Persona de Mediana Edad , Infarto del Miocardio/terapia , Miocitos Cardíacos/trasplante , Reacción en Cadena en Tiempo Real de la Polimerasa , Resultado del Tratamiento , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...