Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Adv Funct Mater ; 28(1)2018 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-30473658

RESUMEN

The last decade has seen impressive progress in human embryonic stem cell-derived cardiomyocytes (hESC-CMs) that makes them ideal tools to repair injured hearts. To achieve an optimal outcome, advanced molecular imaging methods are essential to accurately track these transplanted cells in the heart. Herein, we demonstrate for the first time that a class of photoacoustic nanoparticles (PANPs) incorporating semiconducting polymers (SPs) as contrast agents can be used in the photoacoustic imaging (PAI) of transplanted hESC-CMs in living mouse hearts. This is achieved by virtue of two benefits of PANPs. First, strong PA signals and specific spectral features of SPs allow PAI to sensitively detect and distinguish a small number of PANP-labeled cells (2,000) from background tissues in vivo. Second, the PANPs show a high efficiency for hESC-CM labeling without adverse effects on cell structure, function, and gene expression. Assisted by ultrasound imaging, the delivery and engraftment of hESC-CMs in living mouse hearts can be assessed by PANP-based PAI with high spatial resolution (~100 µm). In summary, this study explores and validates a novel application of SPs as a PA contrast agent to track labeled cells with high sensitivity and accuracy in vivo, highlighting the advantages of integrating PAI and PANPs to advance cardiac regenerative therapies.

2.
Stem Cells ; 35(8): 1994-2000, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28600830

RESUMEN

Human pluripotent stem cells, including human embryonic stem cells (hESCs) and human induced PSCs (hiPSCs), have great potential as an unlimited donor source for cell-based therapeutics. The risk of teratoma formation from residual undifferentiated cells, however, remains a critical barrier to the clinical application of these cells. Herein, we describe external beam radiation therapy (EBRT) as an attractive option for the treatment of this iatrogenic growth. We present evidence that EBRT is effective in arresting growth of hESC-derived teratomas in vivo at day 28 post-implantation by using a microCT irradiator capable of targeted treatment in small animals. Within several days of irradiation, teratomas derived from injection of undifferentiated hESCs and hiPSCs demonstrated complete growth arrest lasting several months. In addition, EBRT reduced reseeding potential of teratoma cells during serial transplantation experiments, requiring irradiated teratomas to be seeded at 1 × 103 higher doses to form new teratomas. We demonstrate that irradiation induces teratoma cell apoptosis, senescence, and growth arrest, similar to established radiobiology mechanisms. Taken together, these results provide proof of concept for the use of EBRT in the treatment of existing teratomas and highlight a strategy to increase the safety of stem cell-based therapies. Stem Cells 2017;35:1994-2000.


Asunto(s)
Células Madre Pluripotentes/patología , Radiación Ionizante , Teratoma/radioterapia , Apoptosis/efectos de la radiación , Diferenciación Celular/efectos de la radiación , Proliferación Celular/efectos de la radiación , Humanos , Células Madre Pluripotentes/efectos de la radiación , Teratoma/patología
3.
Proc Natl Acad Sci U S A ; 112(1): 94-9, 2015 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-25535360

RESUMEN

There is a high mortality in patients with diabetes and severe pressure ulcers. For example, chronic pressure sores of the heels often lead to limb loss in diabetic patients. A major factor underlying this is reduced neovascularization caused by impaired activity of the transcription factor hypoxia inducible factor-1 alpha (HIF-1α). In diabetes, HIF-1α function is compromised by a high glucose-induced and reactive oxygen species-mediated modification of its coactivator p300, leading to impaired HIF-1α transactivation. We examined whether local enhancement of HIF-1α activity would improve diabetic wound healing and minimize the severity of diabetic ulcers. To improve HIF-1α activity we designed a transdermal drug delivery system (TDDS) containing the FDA-approved small molecule deferoxamine (DFO), an iron chelator that increases HIF-1α transactivation in diabetes by preventing iron-catalyzed reactive oxygen stress. Applying this TDDS to a pressure-induced ulcer model in diabetic mice, we found that transdermal delivery of DFO significantly improved wound healing. Unexpectedly, prophylactic application of this transdermal delivery system also prevented diabetic ulcer formation. DFO-treated wounds demonstrated increased collagen density, improved neovascularization, and reduction of free radical formation, leading to decreased cell death. These findings suggest that transdermal delivery of DFO provides a targeted means to both prevent ulcer formation and accelerate diabetic wound healing with the potential for rapid clinical translation.


Asunto(s)
Deferoxamina/uso terapéutico , Complicaciones de la Diabetes/tratamiento farmacológico , Complicaciones de la Diabetes/prevención & control , Diabetes Mellitus Experimental/tratamiento farmacológico , Presión/efectos adversos , Úlcera/tratamiento farmacológico , Administración Cutánea , Animales , Apoptosis/efectos de los fármacos , Deferoxamina/administración & dosificación , Deferoxamina/farmacología , Dermis/irrigación sanguínea , Dermis/efectos de los fármacos , Dermis/patología , Complicaciones de la Diabetes/patología , Diabetes Mellitus Experimental/patología , Sistemas de Liberación de Medicamentos , Ratones Endogámicos C57BL , Necrosis , Neovascularización Fisiológica/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Estrés Fisiológico/efectos de los fármacos , Úlcera/patología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Cicatrización de Heridas/efectos de los fármacos
4.
Circ Res ; 117(8): 720-30, 2015 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-26291556

RESUMEN

RATIONALE: Tissue engineering approaches may improve survival and functional benefits from human embryonic stem cell-derived cardiomyocyte transplantation, thereby potentially preventing dilative remodeling and progression to heart failure. OBJECTIVE: Assessment of transport stability, long-term survival, structural organization, functional benefits, and teratoma risk of engineered heart muscle (EHM) in a chronic myocardial infarction model. METHODS AND RESULTS: We constructed EHMs from human embryonic stem cell-derived cardiomyocytes and released them for transatlantic shipping following predefined quality control criteria. Two days of shipment did not lead to adverse effects on cell viability or contractile performance of EHMs (n=3, P=0.83, P=0.87). One month after ischemia/reperfusion injury, EHMs were implanted onto immunocompromised rat hearts to simulate chronic ischemia. Bioluminescence imaging showed stable engraftment with no significant cell loss between week 2 and 12 (n=6, P=0.67), preserving ≤25% of the transplanted cells. Despite high engraftment rates and attenuated disease progression (change in ejection fraction for EHMs, -6.7±1.4% versus control, -10.9±1.5%; n>12; P=0.05), we observed no difference between EHMs containing viable and nonviable human cardiomyocytes in this chronic xenotransplantation model (n>12; P=0.41). Grafted cardiomyocytes showed enhanced sarcomere alignment and increased connexin 43 expression at 220 days after transplantation. No teratomas or tumors were found in any of the animals (n=14) used for long-term monitoring. CONCLUSIONS: EHM transplantation led to high engraftment rates, long-term survival, and progressive maturation of human cardiomyocytes. However, cell engraftment was not correlated with functional improvements in this chronic myocardial infarction model. Most importantly, the safety of this approach was demonstrated by the lack of tumor or teratoma formation.


Asunto(s)
Células Madre Embrionarias/trasplante , Supervivencia de Injerto , Trasplante de Corazón/métodos , Infarto del Miocardio/cirugía , Miocitos Cardíacos/trasplante , Músculos Papilares/trasplante , Ingeniería de Tejidos/métodos , Animales , Biomarcadores/metabolismo , Diferenciación Celular , Línea Celular , Supervivencia Celular , Conexina 43/metabolismo , Modelos Animales de Enfermedad , Células Madre Embrionarias/inmunología , Células Madre Embrionarias/metabolismo , Trasplante de Corazón/efectos adversos , Xenoinjertos , Humanos , Inmunosupresores/farmacología , Masculino , Contracción Miocárdica , Infarto del Miocardio/inmunología , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocitos Cardíacos/inmunología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Músculos Papilares/inmunología , Músculos Papilares/metabolismo , Músculos Papilares/patología , Músculos Papilares/fisiopatología , Ratas Desnudas , Ratas Sprague-Dawley , Volumen Sistólico , Factores de Tiempo , Transfección
5.
Transpl Int ; 30(11): 1181-1189, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28672061

RESUMEN

Cardiac allograft vasculopathy (CAV) affects approximately 30% of cardiac transplant patients at 5 years post-transplantation. To date, there are few CAV treatment or prevention options, none of which are highly effective. The aim of the study was to investigate the effect of thalidomide on the development of CAV. The effect of thalidomide treatment on chronic rejection was assessed in rat orthotopic aortic transplants in allogeneic F344 or syngeneic Lew rats (n = 6 per group). Animals were left untreated or received thalidomide for 30 days post-transplant, and evidence of graft CAV was determined by histology (trichrome and immunohistochemistry) and intragraft cytokine measurements. Animals that received thalidomide treatment post-transplant showed markedly reduced luminal obliteration, with concomitant rescue of smooth muscle cells (SMCs) in the aortic media of grafts. Thalidomide counteracted neointimal hyperplasia by preventing dedifferentiation of vascular SMCs. Measurement of intragraft cytokine levels after thalidomide treatment revealed downregulation of matrix metalloproteinase 8 and monocyte chemotactic protein 1, cytokines involved in tissue remodelling and inflammation, respectively. Importantly, no negative side effects of thalidomide were observed. Thalidomide treatment prevents CAV development in a rodent model and is therefore potentially useful in clinical applications to prevent post-transplant heart rejection.


Asunto(s)
Aorta Torácica/trasplante , Enfermedad de la Arteria Coronaria/prevención & control , Rechazo de Injerto/prevención & control , Inmunosupresores/uso terapéutico , Talidomida/uso terapéutico , Animales , Aorta Torácica/efectos de los fármacos , Aorta Torácica/metabolismo , Enfermedad Crónica , Enfermedad de la Arteria Coronaria/etiología , Citocinas/metabolismo , Evaluación Preclínica de Medicamentos , Rechazo de Injerto/complicaciones , Inmunosupresores/farmacología , Linfocitos/efectos de los fármacos , Masculino , Miocitos del Músculo Liso/efectos de los fármacos , Ratas Endogámicas F344 , Ratas Endogámicas Lew , Talidomida/farmacología , Túnica Media/efectos de los fármacos
6.
Proc Natl Acad Sci U S A ; 111(22): 7974-9, 2014 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-24843161

RESUMEN

The ability to implant electronic systems in the human body has led to many medical advances. Progress in semiconductor technology paved the way for devices at the scale of a millimeter or less ("microimplants"), but the miniaturization of the power source remains challenging. Although wireless powering has been demonstrated, energy transfer beyond superficial depths in tissue has so far been limited by large coils (at least a centimeter in diameter) unsuitable for a microimplant. Here, we show that this limitation can be overcome by a method, termed midfield powering, to create a high-energy density region deep in tissue inside of which the power-harvesting structure can be made extremely small. Unlike conventional near-field (inductively coupled) coils, for which coupling is limited by exponential field decay, a patterned metal plate is used to induce spatially confined and adaptive energy transport through propagating modes in tissue. We use this method to power a microimplant (2 mm, 70 mg) capable of closed-chest wireless control of the heart that is orders of magnitude smaller than conventional pacemakers. With exposure levels below human safety thresholds, milliwatt levels of power can be transferred to a deep-tissue (>5 cm) microimplant for both complex electronic function and physiological stimulation. The approach developed here should enable new generations of implantable systems that can be integrated into the body at minimal cost and risk.


Asunto(s)
Electrónica Médica/instrumentación , Electrónica Médica/métodos , Miniaturización/métodos , Modelos Teóricos , Prótesis e Implantes , Tecnología Inalámbrica/instrumentación , Animales , Corteza Cerebral , Suministros de Energía Eléctrica , Campos Electromagnéticos , Diseño de Equipo , Ventrículos Cardíacos , Humanos , Óptica y Fotónica/instrumentación , Óptica y Fotónica/métodos , Conejos , Semiconductores , Piel , Porcinos
8.
Small ; 8(23): 3573-8, 2012 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-22888073

RESUMEN

Amphiphilic bovine serum albumin-poly(methyl methacrylate) conjugate forms nanoparticles with the uniform size of ~100 nm by self-assembling. Loaded with the hydrophobic anti-tumor drug camptothecin, the nanoparticle efficiently delivers drugs into cancer cells, and thus inhibits ~79% of tumor growth in animals compared with free drug.


Asunto(s)
Antineoplásicos/química , Nanopartículas/química , Polimetil Metacrilato/química , Albúmina Sérica Bovina/química , Animales , Antineoplásicos/farmacología , Camptotecina/farmacología , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos , Interacciones Hidrofóbicas e Hidrofílicas , Tamaño de la Partícula , Polimetil Metacrilato/farmacología , Albúmina Sérica Bovina/farmacología
9.
JCI Insight ; 6(7)2021 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-33830086

RESUMEN

Human pluripotent stem cells (PSCs), which are composed of embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), provide an opportunity to advance cardiac cell therapy-based clinical trials. However, an important hurdle that must be overcome is the risk of teratoma formation after cell transplantation due to the proliferative capacity of residual undifferentiated PSCs in differentiation batches. To tackle this problem, we propose the use of a minimal noncardiotoxic doxorubicin dose as a purifying agent to selectively target rapidly proliferating stem cells for cell death, which will provide a purer population of terminally differentiated cardiomyocytes before cell transplantation. In this study, we determined an appropriate in vitro doxorubicin dose that (a) eliminates residual undifferentiated stem cells before cell injection to prevent teratoma formation after cell transplantation and (b) does not cause cardiotoxicity in ESC-derived cardiomyocytes (CMs) as demonstrated through contractility analysis, electrophysiology, topoisomerase activity assay, and quantification of reactive oxygen species generation. This study establishes a potentially novel method for tumorigenic-free cell therapy studies aimed at clinical applications of cardiac cell transplantation.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Doxorrubicina/administración & dosificación , Células Madre Embrionarias/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Células Madre Pluripotentes/citología , Animales , Apoptosis/efectos de los fármacos , Cardiotoxicidad/etiología , Cardiotoxicidad/prevención & control , Muerte Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Tratamiento Basado en Trasplante de Células y Tejidos/efectos adversos , Relación Dosis-Respuesta a Droga , Doxorrubicina/farmacología , Células Madre Embrionarias/trasplante , Regulación de la Expresión Génica/efectos de los fármacos , Células Madre Embrionarias Humanas/citología , Células Madre Embrionarias Humanas/efectos de los fármacos , Humanos , Ratones SCID , Especies Reactivas de Oxígeno/metabolismo , Teratoma/prevención & control
10.
Stem Cells ; 27(1): 266-74, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18974212

RESUMEN

Evolving evidence suggests a possible role for adipose stromal cells (ASCs) in adult neovascularization, although the specific cues that stimulate their angiogenic behavior are poorly understood. We evaluated the effect of hypoxia, a central mediator of new blood vessel development within ischemic tissue, on proneovascular ASC functions. Murine ASCs were exposed to normoxia (21% oxygen) or hypoxia (5%, 1% oxygen) for varying lengths of time. Vascular endothelial growth factor (VEGF) secretion by ASCs increased as an inverse function of oxygen tension, with progressively higher VEGF expression at 21%, 5%, and 1% oxygen, respectively. Greater VEGF levels were also associated with longer periods in culture. ASCs were able to migrate towards stromal cell-derived factor (SDF)-1, a chemokine expressed by ischemic tissue, with hypoxia augmenting ASC expression of the SDF-1 receptor (CXCR4) and potentiating ASC migration. In vivo, ASCs demonstrated the capacity to proliferate in response to a hypoxic insult remote from their resident niche, and this was supported by in vitro studies showing increasing ASC proliferation with greater degrees of hypoxia. Hypoxia did not significantly alter the expression of endothelial surface markers by ASCs. However, these cells did assume an endothelial phenotype as evidenced by their ability to tubularize when seeded with differentiated endothelial cells on Matrigel. Taken together, these data suggest that ASCs upregulate their proneovascular activity in response to hypoxia, and may harbor the capacity to home to ischemic tissue and function cooperatively with existing vasculature to promote angiogenesis.


Asunto(s)
Tejido Adiposo/citología , Neovascularización Fisiológica , Células del Estroma/metabolismo , Animales , Hipoxia de la Célula/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Quimiocina CXCL12/farmacología , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Citometría de Flujo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ratones , Ratones Endogámicos C57BL , Neovascularización Fisiológica/efectos de los fármacos , Fenotipo , Células del Estroma/citología , Células del Estroma/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/metabolismo
11.
Circ Heart Fail ; 13(3): e006298, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32160771

RESUMEN

BACKGROUND: MicroRNAs are small, noncoding RNAs that play a key role in gene expression. Accumulating evidence suggests that aberrant microRNA expression contributes to the heart failure (HF) phenotype; however, the underlying molecular mechanisms are not well understood. A better understanding of the mechanisms of action of microRNAs could potentially lead to targeted therapies that could halt the progression or even reverse HF. METHODS AND RESULTS: We found that microRNA-152 (miR-152) expression was upregulated in the failing human heart and experimental animal models of HF. Transgenic mice with cardiomyocyte-specific miR-152 overexpression developed systolic dysfunction (mean difference, -38.74% [95% CI, -45.73% to -31.74%]; P<0.001) and dilated cardiomyopathy. At the cellular level, miR-152 overexpression perturbed mitochondrial ultrastructure and dysregulated key genes involved in cardiomyocyte metabolism and inflammation. Mechanistically, we identified Glrx5 (glutaredoxin 5), a critical regulator of mitochondrial iron homeostasis and iron-sulfur cluster synthesis, as a direct miR-152 target. Finally, a proof-of-concept of the therapeutic efficacy of targeting miR-152 in vivo was obtained by utilizing a locked nucleic acid-based inhibitor of miR-152 (LNA 152) in a murine model of HF subjected to transverse aortic constriction. We demonstrated that animals treated with LNA-152 (n=10) showed preservation of systolic function when compared with locked nucleic acid-control treated animals (n=9; mean difference, 18.25% [95% CI, 25.10% to 11.39%]; P<0.001). CONCLUSIONS: The upregulation of miR-152 expression in the failing myocardium contributes to HF pathophysiology. Preclinical evidence suggests that miR-152 inhibition preserves cardiac function in a model of pressure overload-induced HF. These findings offer new insights into the pathophysiology of HF and point to miR-152-Glrx5 axis as a potential novel therapeutic target.


Asunto(s)
Antagomirs/administración & dosificación , Silenciador del Gen , Insuficiencia Cardíaca/prevención & control , MicroARNs/metabolismo , Miocitos Cardíacos/metabolismo , Animales , Aorta/fisiopatología , Aorta/cirugía , Estudios de Casos y Controles , Modelos Animales de Enfermedad , Glutarredoxinas/genética , Glutarredoxinas/metabolismo , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/fisiopatología , Humanos , Ligadura , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , MicroARNs/genética , Mitocondrias Cardíacas/genética , Mitocondrias Cardíacas/metabolismo , Mitocondrias Cardíacas/ultraestructura , Miocitos Cardíacos/ultraestructura , Prueba de Estudio Conceptual , Volumen Sistólico , Función Ventricular Izquierda
12.
Dev Cell ; 54(6): 694-709.e9, 2020 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-32763147

RESUMEN

Transposable elements (TEs) comprise nearly half of the human genome and are often transcribed or exhibit cis-regulatory properties with unknown function in specific processes such as heart development. In the case of endogenous retroviruses (ERVs), a TE subclass, experimental interrogation is constrained as many are primate-specific or human-specific. Here, we use primate pluripotent stem-cell-derived cardiomyocytes that mimic fetal cardiomyocytes in vitro to discover hundreds of ERV transcripts from the primate-specific MER41 family, some of which are regulated by the cardiogenic transcription factor TBX5. The most significant of these are located within BANCR, a long non-coding RNA (lncRNA) exclusively expressed in primate fetal cardiomyocytes. Functional studies reveal that BANCR promotes cardiomyocyte migration in vitro and ventricular enlargement in vivo. We conclude that recently evolved TE loci such as BANCR may represent potent de novo developmental regulatory elements that can be interrogated with species-matching pluripotent stem cell models.


Asunto(s)
Retrovirus Endógenos/genética , Miocitos Cardíacos/metabolismo , ARN Largo no Codificante/genética , Factores de Transcripción/genética , Animales , Elementos Transponibles de ADN/genética , Evolución Molecular , Regulación de la Expresión Génica/genética , Genoma Humano , Humanos , Primates/genética , Especificidad de la Especie
13.
Int J Cardiol ; 281: 8-14, 2019 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-30739802

RESUMEN

BACKGROUND: Manganese-enhanced MRI (MEMRI) detects viable cardiomyocytes based on the intracellular manganese uptake via L-type calcium-channels. This study aimed to quantify myocardial viability based on manganese uptake by viable myocardium in the infarct core (IC), peri-infarct region (PIR) and remote myocardium (RM) using T1 mapping before and after MEMRI and assess their association with cardiac function and arrhythmogenesis. METHODS: Fifteen female swine had a 60-minute balloon ischemia-reperfusion injury in the LAD. MRI (Signa 3T, GE Healthcare) and electrophysiological study (EPS) were performed 4 weeks later. MEMRI and delayed gadolinium-enhanced MRI (DEMRI) were acquired on LV short axis. The DEMRI positive total infarct area was subdivided into the regions of MEMRI-negative non-viable IC and MEMRI-positive viable PIR. T1 mapping was performed to evaluate native T1, post-MEMRI T1, and delta R1 (R1post-R1pre, where R1 equals 1/T1) of each territory. Their correlation with LV function and EPS data was assessed. RESULTS: PIR was characterized by intermediate native T1 (1530.5 ±â€¯75.2 ms) compared to IC (1634.7 ±â€¯88.4 ms, p = 0.001) and RM (1406.4 ±â€¯37.9 ms, p < 0.0001). Lower post-MEMRI T1 of PIR (1136.3 ±â€¯99.6 ms) than IC (1262.6 ±â€¯126.8 ms, p = 0.005) and higher delta R1 (0.23 ±â€¯0.08 s-1) of PIR than IC (0.18 ±â€¯0.09 s-1, p = 0.04) indicated higher myocardial manganese uptake of PIR compared to IC. Post-MEMRI T1 (r = -0.57, p = 0.02) and delta R1 (r = 0.51, p = 0.04) of PIR correlated significantly with LVEF. CONCLUSIONS: PIR is characterized by higher manganese uptake compared to the infarct core. In the subacute phase post-IR, PIR viability measured by post-MEMRI T1 correlates with cardiac function.


Asunto(s)
Imagen por Resonancia Magnética/métodos , Infarto del Miocardio/diagnóstico por imagen , Infarto del Miocardio/fisiopatología , Miocardio/patología , Disfunción Ventricular Izquierda/diagnóstico por imagen , Disfunción Ventricular Izquierda/fisiopatología , Animales , Femenino , Porcinos
14.
Stem Cell Reports ; 12(6): 1232-1241, 2019 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-31105048

RESUMEN

Chagas disease (ChD) is one of the most neglected tropical diseases, with cardiomyopathy being the main cause of death in Trypanosoma cruzi-infected patients. As the parasite actively replicates in cardiomyocytes (CMs), the heart remains a key target organ in the pathogenesis of ChD. Here we modeled ChD using human induced pluripotent stem cell-derived CMs (iPSC-CMs) to understand the complex interplay between the parasite and host cells. We showed that iPSC-CMs can get infected with the T. cruzi Y strain and that all parasite cycle stages can be identified in our model system. Importantly, characterization of T. cruzi-infected iPSC-CMs showed significant changes in their gene expression profile, cell contractility, and distribution of key cardiac markers. Moreover, these infected iPSC-CMs exhibited a pro-inflammatory profile as indicated by significantly elevated cytokine levels and cell-trafficking regulators. We believe our iPSC-CM model is a valuable platform to explore new treatment strategies for ChD.


Asunto(s)
Cardiomiopatía Chagásica/metabolismo , Células Madre Pluripotentes Inducidas , Modelos Biológicos , Miocitos Cardíacos , Trypanosoma cruzi/metabolismo , Cardiomiopatía Chagásica/patología , Cardiomiopatía Chagásica/terapia , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/parasitología , Células Madre Pluripotentes Inducidas/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/parasitología , Miocitos Cardíacos/patología
15.
Stem Cell Reports ; 10(2): 422-435, 2018 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-29398480

RESUMEN

Non-human primates (NHPs) can serve as a human-like model to study cell therapy using induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs). However, whether the efficacy of NHP and human iPSC-CMs is mechanistically similar remains unknown. To examine this, RNU rats received intramyocardial injection of 1 × 107 NHP or human iPSC-CMs or the same number of respective fibroblasts or PBS control (n = 9-14/group) at 4 days after 60-min coronary artery occlusion-reperfusion. Cardiac function and left ventricular remodeling were similarly improved in both iPSC-CM-treated groups. To mimic the ischemic environment in the infarcted heart, both cultured NHP and human iPSC-CMs underwent 24-hr hypoxia in vitro. Both cells and media were collected, and similarities in transcriptomic as well as metabolomic profiles were noted between both groups. In conclusion, both NHP and human iPSC-CMs confer similar cardioprotection in a rodent myocardial infarction model through relatively similar mechanisms via promotion of cell survival, angiogenesis, and inhibition of hypertrophy and fibrosis.


Asunto(s)
Células Madre Pluripotentes Inducidas/trasplante , Infarto del Miocardio/terapia , Miocitos Cardíacos/trasplante , Trasplante de Células Madre , Animales , Diferenciación Celular , Hipoxia de la Célula/fisiología , Supervivencia Celular/fisiología , Modelos Animales de Enfermedad , Humanos , Células Madre Pluripotentes Inducidas/citología , Infarto del Miocardio/fisiopatología , Miocitos Cardíacos/citología , Primates , Ratas
16.
J Nucl Med ; 58(10): 1679-1684, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28490473

RESUMEN

6″-18F-fluoromaltotriose is a PET tracer that can potentially be used to image and localize most bacterial infections, much like 18F-FDG has been used to image and localize most cancers. However, unlike 18F-FDG, 6″-18F-fluoromaltotriose is not taken up by inflammatory lesions and appears to be specific to bacterial infections by targeting the maltodextrin transporter that is expressed in gram-positive and gram-negative strains of bacteria. Methods: 6″-18F-fluoromaltotriose was synthesized with high radiochemical purity and evaluated in several clinically relevant bacterial strains in cultures and in living mice. Results: 6″-18F-fluoromaltotriose was taken up in both gram-positive and gram-negative bacterial strains. 6″-18F-fluoromaltotriose was also able to detect Pseudomonas aeruginosa in a clinically relevant mouse model of wound infection. The utility of 6″-18F-fluoromaltotriose to help monitor antibiotic therapies was also evaluated in rats. Conclusion: 6″-18F-fluoromaltotriose is a promising new tracer that has significant diagnostic utility, with the potential to change the clinical management of patients with infectious diseases of bacterial origin.


Asunto(s)
Infecciones Bacterianas/diagnóstico por imagen , Infecciones Bacterianas/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Polisacáridos/metabolismo , Tomografía de Emisión de Positrones/métodos , Trisacáridos , Animales , Transporte Biológico , Ratones , Ratones Desnudos , Trazadores Radiactivos , Infección de Heridas/diagnóstico por imagen , Infección de Heridas/metabolismo
17.
JAMA Cardiol ; 1(8): 953-962, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27579998

RESUMEN

Importance: Although progress continues to be made in the field of stem cell regenerative medicine for the treatment of cardiovascular disease, significant barriers to clinical implementation still exist. Objectives: To summarize the current barriers to the clinical implementation of stem cell therapy in patients with cardiovascular disease and to discuss potential strategies to overcome them. Evidence Review: Information for this review was obtained through a search of PubMed and the Cochrane database for English-language studies published between January 1, 2000, and July 25, 2016. Ten randomized clinical trials and 8 systematic reviews were included. Findings: One of the major clinical barriers facing the routine implementation of stem cell therapy in patients with cardiovascular disease is the limited and inconsistent benefit observed thus far. Reasons for this finding are unclear but may be owing to poor cell retention and survival, as suggested by numerous preclinical studies and a small number of human studies incorporating imaging to determine cell fate. Additional studies in humans using imaging to determine cell fate are needed to understand how these factors contribute to the limited efficacy of stem cell therapy. Treatment strategies to address poor cell retention and survival are under investigation and include the following: coadministration of immunosuppressive and prosurvival agents, delivery of cardioprotective factors packaged in exosomes rather than the cells themselves, and use of tissue-engineering strategies to provide structural support for cells. If larger grafts are achieved using these strategies, it will be imperative to carefully monitor for the potential risks of tumorigenicity, immunogenicity, and arrhythmogenicity. Conclusions and Relevance: Despite important achievements to date, stem cell therapy is not yet ready for routine clinical implementation. Significant research is still needed to address the clinical barriers outlined herein before the next wave of large clinical trials is under way.


Asunto(s)
Enfermedades Cardiovasculares/terapia , Tratamiento Basado en Trasplante de Células y Tejidos , Medicina Regenerativa/tendencias , Investigación con Células Madre , Supervivencia Celular , Humanos , Trasplante de Células Madre Mesenquimatosas , Trasplante de Células Madre , Ingeniería de Tejidos/tendencias
18.
Stem Cell Res Ther ; 7(1): 84, 2016 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-27296220

RESUMEN

BACKGROUND: Acute myocardial infarction (MI) leads to an irreversible loss of proper cardiac function. Application of stem cell therapy is an attractive option for MI treatment. Adipose tissue has proven to serve as a rich source of stem cells (ADSCs). Taking into account the different morphogenesis, anatomy, and physiology of adipose tissue, we hypothesized that ADSCs from different adipose tissue depots may exert a diverse multipotency and cardiogenic potential. METHODS: The omental, pericardial, and epicardial adipose tissue samples were obtained from organ donors and patients undergoing heart transplantation at our institution. Human foreskin fibroblasts were used as the control group. Isolated ADSCs were analyzed for adipogenic and osteogenic differentiation capacity and proliferation potential. The immunophenotype and constitutive gene expression of alkaline phosphatase (ALP), GATA4, Nanog, and OCT4 were analyzed. DNA methylation inhibitor 5-azacytidine was exposed to the cells to stimulate the cardiogenesis. Finally, reprogramming towards cardiomyocytes was initiated with exogenous overexpression of seven transcription factors (ESRRG, GATA4, MEF2C, MESP1, MYOCD, TBX5, ZFPM2) previously applied successfully for fibroblast transdifferentiation toward cardiomyocytes. Expression of cardiac troponin T (cTNT) and alpha-actinin (Actn2) was analyzed 3 weeks after initiation of the cardiac differentiation. RESULTS: The multipotent properties of isolated plastic adherent cells were confirmed with expression of CD29, CD44, CD90, and CD105, as well as successful differentiation toward adipocytes and osteocytes; with the highest osteogenic and adipogenic potential for the epicardial and omental ADSCs, respectively. Epicardial ADSCs demonstrated a lower doubling time as compared with the pericardium and omentum-derived cells. Furthermore, epicardial ADSCs revealed higher constitutive expression of ALP and GATA4. Increased Actn2 and cTNT expression was observed after the transduction of seven reprogramming factors, with the highest expression in the epicardial ADSCs, as compared with the other ADSC subtypes and fibroblasts. CONCLUSIONS: Human epicardial ADSCs revealed a higher cardiomyogenic potential as compared with the pericardial and omental ADSC subtypes as well as the fibroblast counterparts. Epicardial ADSCs may thus serve as the valuable subject for further studies on more effective methods of adult stem cell differentiation toward cardiomyocytes.


Asunto(s)
Adipocitos/citología , Epiplón/citología , Pericardio/citología , Células Madre/citología , Actinina/genética , Actinina/metabolismo , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Adulto , Anciano , Fosfatasa Alcalina/genética , Fosfatasa Alcalina/metabolismo , Antígenos CD/genética , Antígenos CD/metabolismo , Azacitidina/farmacología , Biomarcadores/metabolismo , Transdiferenciación Celular , Metilación de ADN/efectos de los fármacos , Femenino , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Factor de Transcripción GATA4/genética , Factor de Transcripción GATA4/metabolismo , Expresión Génica , Trasplante de Corazón , Humanos , Masculino , Persona de Mediana Edad , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Proteína Homeótica Nanog/genética , Proteína Homeótica Nanog/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Epiplón/efectos de los fármacos , Epiplón/metabolismo , Osteocitos/citología , Osteocitos/efectos de los fármacos , Osteocitos/metabolismo , Pericardio/efectos de los fármacos , Pericardio/metabolismo , Cultivo Primario de Células , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Factores de Transcripción/farmacología , Troponina T/genética , Troponina T/metabolismo
19.
Transplantation ; 100(5): 1022-31, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26910327

RESUMEN

BACKGROUND: The efficacy of selective Janus kinase 1/3 inhibitor R507 to prevent obliterative airway disease was analyzed in preclinical airway transplantation models. METHODS: Orthotopic trachea transplantations were performed between Lewis donors and Brown Norway rat recipients. Oral everolimus (4 mg/kg once per day) or oral respective inhaled R507 (60 mg/kg twice per day, each) was used for immunosuppression. Grafts were retrieved after 6 or 60 days. Toxicity and anti-inflammatory effects of R507 were analyzed on human airway epithelial cells. RESULTS: In 6-day animals, oral and inhaled R507 more potently diminished mononuclear graft infiltration than everolimus and preserved ciliated pseudostratified columnar respiratory epithelium. Everolimus and R507 similarly suppressed systemic cellular and humoral immune activation. In untreated rats, marked obliterative airway disease developed over 60 days. Oral and inhaled R507 was significantly more effective in reducing airway obliteration and preserved the morphology of the airway epithelium. Luciferase-positive donors revealed that a substantial amount of smooth muscle cells within the obliterative tissue was of donor origin. Only everolimus but not R507, adversely altered kidney function and lipid profiles. The R507 aerosol did not show airway toxicity in vitro but effectively suppressed activation of inflammatory signaling pathways induced by IL-1ß. CONCLUSIONS: The Janus kinase 1/3 inhibitor R507 is a very well-tolerated immunosuppressant that similarly diminished obliterative airway disease with systemic or inhaled administration.


Asunto(s)
Bronquiolitis Obliterante/prevención & control , Inmunosupresores/administración & dosificación , Janus Quinasa 1/antagonistas & inhibidores , Janus Quinasa 3/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/administración & dosificación , Tráquea/trasplante , Administración por Inhalación , Administración Oral , Aerosoles/química , Animales , Células Cultivadas , Células Epiteliales/metabolismo , Everolimus/administración & dosificación , Humanos , L-Lactato Deshidrogenasa/metabolismo , Microscopía Fluorescente , Análisis de Secuencia por Matrices de Oligonucleótidos , Ratas , Ratas Endogámicas Lew , Transducción de Señal , Resultado del Tratamiento
20.
Stem Cell Reports ; 6(2): 176-87, 2016 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-26777057

RESUMEN

The use of cells derived from pluripotent stem cells (PSCs) for regenerative therapies confers a considerable risk for neoplastic growth and teratoma formation. Preclinical and clinical assessment of such therapies will require suitable monitoring strategies to understand and mitigate these risks. Here we generated human-induced pluripotent stem cells (iPSCs), selected clones that continued to express reprogramming factors after differentiation into cardiomyocytes, and transplanted these cardiomyocytes into immunocompromised rat hearts post-myocardial infarction. We compared magnetic resonance imaging (MRI), cardiac ultrasound, and serum biomarkers for their ability to delineate teratoma formation and growth. MRI enabled the detection of teratomas with a volume >8 mm(3). A combination of three plasma biomarkers (CEA, AFP, and HCG) was able to detect teratomas with a volume >17 mm(3) and with a sensitivity of more than 87%. Based on our findings, a combination of serum biomarkers with MRI screening may offer the highest sensitivity for teratoma detection and tracking.


Asunto(s)
Biomarcadores de Tumor/sangre , Imagen por Resonancia Magnética/métodos , Células Madre Pluripotentes/patología , Teratoma/sangre , Teratoma/diagnóstico , Animales , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Gadolinio , Corazón/fisiopatología , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Inflamación/patología , Lentivirus/metabolismo , Masculino , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Miocitos Cardíacos/trasplante , Fenotipo , Células Madre Pluripotentes/efectos de los fármacos , Ratas Desnudas , Teratoma/irrigación sanguínea , Carga Tumoral/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA