Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 644
Filtrar
1.
Nat Commun ; 15(1): 3366, 2024 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-38684678

RESUMEN

Autologous skin grafting is a standard treatment for skin defects such as burns. No artificial skin substitutes are functionally equivalent to autologous skin grafts. The cultured epidermis lacks the dermis and does not engraft deep wounds. Although reconstituted skin, which consists of cultured epidermal cells on a synthetic dermal substitute, can engraft deep wounds, it requires the wound bed to be well-vascularized and lacks skin appendages. In this study, we successfully generate complete skin grafts with pluripotent stem cell-derived epidermis with appendages on p63 knockout embryos' dermis. Donor pluripotent stem cell-derived keratinocytes encroach the embryos' dermis by eliminating p63 knockout keratinocytes based on cell-extracellular matrix adhesion mediated cell competition. Although the chimeric skin contains allogenic dermis, it is engraftable as long as autologous grafts. Furthermore, we could generate semi-humanized skin segments by human keratinocytes injection into the amnionic cavity of p63 knockout mice embryos. Niche encroachment opens the possibility of human skin graft production in livestock animals.


Asunto(s)
Dermis , Queratinocitos , Ratones Noqueados , Trasplante de Piel , Animales , Trasplante de Piel/métodos , Queratinocitos/citología , Queratinocitos/trasplante , Humanos , Dermis/citología , Dermis/trasplante , Ratones , Epidermis/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/trasplante , Piel Artificial , Células Epidérmicas/trasplante , Células Epidérmicas/citología , Matriz Extracelular/metabolismo , Piel/citología
2.
Stem Cell Reports ; 19(2): 254-269, 2024 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-38181785

RESUMEN

Pluripotent stem cell-based therapy for retinal degenerative diseases is a promising approach to restoring visual function. A clinical study using retinal organoid (RO) sheets was recently conducted in patients with retinitis pigmentosa. However, the graft preparation currently requires advanced skills to identify and excise suitable segments from the transplantable area of the limited number of suitable ROs. This remains a challenge for consistent clinical implementations. Herein, we enabled the enrichment of wild-type (non-reporter) retinal progenitor cells (RPCs) from dissociated ROs using a label-free ghost cytometry (LF-GC)-based sorting system, where a machine-based classifier was trained in advance with another RPC reporter line. The sorted cells reproducibly formed retinal spheroids large enough for transplantation and developed mature photoreceptors in the retinal degeneration rats. This method of enriching early RPCs with no specific surface antigens and without any reporters or chemical labeling is promising for robust preparation of graft tissues during cell-based therapy.


Asunto(s)
Células Madre Pluripotentes , Degeneración Retiniana , Retinitis Pigmentosa , Humanos , Animales , Ratas , Especies Reactivas de Oxígeno , Retina , Células Madre Pluripotentes/trasplante , Degeneración Retiniana/terapia , Retinitis Pigmentosa/terapia , Trasplante de Células Madre/métodos
3.
Artículo en Inglés | MEDLINE | ID: mdl-36617642

RESUMEN

Retinal degeneration is an increasing global burden without cure for the majority of patients. Once retinal cells have degenerated, vision is permanently lost. Different strategies have been developed in recent years to prevent retinal degeneration or to restore sight (e.g., gene therapy, cell therapy, and electronic implants). Herein, we present current treatment strategies with a focus on cell therapy for photoreceptor replacement using human pluripotent stem cells. We will describe the state of the art and discuss obstacles and limitations observed in preclinical animal models as well as future directions to improve graft integration and functionality.


Asunto(s)
Células Madre Pluripotentes , Degeneración Retiniana , Animales , Humanos , Degeneración Retiniana/terapia , Células Madre Pluripotentes/trasplante , Células Fotorreceptoras , Trasplante de Células Madre
4.
Diabet Med ; 39(12): e14992, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36302085

RESUMEN

Islet transplantation from organ donors can considerably improve glucose homeostasis and well-being in individuals with type 1 diabetes, where the beta cells are destroyed by the autoimmune attack, but there are insufficient donor islets to make this a widespread therapy. Strategies are therefore being developed to generate unlimited amounts of insulin-producing beta cells from pluripotent stem cells, with the aim that they will be transplanted to treat diabetes. Whilst much progress has been made in recent years in the directed differentiation of pluripotent stem cells to beta-like cells, essential gaps still exist in generating stem cell-derived beta cells that are fully functional in vitro. This short review provides details of recent multi-'omics' studies of the human fetal pancreas, which are revealing granular information on the various cell types in the developing pancreas. It is anticipated that this fine mapping of the pancreatic cells at single-cell resolution will provide additional insights that can be utilised to reproducibly produce human beta cells in vitro that have the functional characteristics of beta cells within native human islets.


Asunto(s)
Células Secretoras de Insulina , Trasplante de Islotes Pancreáticos , Células Madre Pluripotentes , Humanos , Páncreas/metabolismo , Diferenciación Celular , Células Madre Pluripotentes/metabolismo , Células Madre Pluripotentes/trasplante , Células Secretoras de Insulina/metabolismo
6.
Cells ; 10(12)2021 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-34943842

RESUMEN

Spinal cord injury (SCI) is a devastating condition of the central nervous system that strongly reduces the patient's quality of life and has large financial costs for the healthcare system. Cell therapy has shown considerable therapeutic potential for SCI treatment in different animal models. Although many different cell types have been investigated with the goal of promoting repair and recovery from injury, stem cells appear to be the most promising. Here, we review the experimental approaches that have been carried out with pluripotent stem cells, a cell type that, due to its inherent plasticity, self-renewal, and differentiation potential, represents an attractive source for the development of new cell therapies for SCI. We will focus on several key observations that illustrate the potential of cell therapy for SCI, and we will attempt to draw some conclusions from the studies performed to date.


Asunto(s)
Células Madre Pluripotentes/trasplante , Traumatismos de la Médula Espinal/terapia , Regeneración de la Medula Espinal , Animales , Ensayos Clínicos como Asunto , Células Madre Embrionarias/trasplante , Humanos , Células Madre Pluripotentes Inducidas/trasplante
7.
Cells ; 10(11)2021 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-34831333

RESUMEN

Personalized regenerative medicine and biomedical research have been galvanized and revolutionized by human pluripotent stem cells in combination with recent advances in genomics, artificial intelligence, and genome engineering. More recently, we have witnessed the unprecedented breakthrough life-saving translation of mRNA-based vaccines for COVID-19 to contain the global pandemic and the investment in billions of US dollars in space exploration projects and the blooming space-tourism industry fueled by the latest reusable space vessels. Now, it is time to examine where the translation of pluripotent stem cell research stands currently, which has been touted for more than the last two decades to cure and treat millions of patients with severe debilitating degenerative diseases and tissue injuries. This review attempts to highlight the accomplishments of pluripotent stem cell research together with cutting-edge genomics and genome editing tools and, also, the promises that have still not been transformed into clinical applications, with cardiovascular research as a case example. This review also brings to our attention the scientific and socioeconomic challenges that need to be effectively addressed to see the full potential of pluripotent stem cells at the clinical bedside.


Asunto(s)
Enfermedades Cardiovasculares/terapia , Genómica , Células Madre Pluripotentes/trasplante , Inteligencia Artificial , Enfermedades Cardiovasculares/genética , Enfermedades Cardiovasculares/patología , Sistema Cardiovascular/citología , Sistema Cardiovascular/crecimiento & desarrollo , Diferenciación Celular , Descubrimiento de Drogas , Edición Génica , Humanos , Modelos Biológicos , Células Madre Pluripotentes/citología , Medicina de Precisión , Medicina Regenerativa , Seguridad , Investigación Biomédica Traslacional
8.
Int J Mol Sci ; 22(22)2021 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-34830410

RESUMEN

Human pluripotent stem cell-derived neural progenitor cells (NPCs) have the potential to recover from nerve injury. We previously reported that human placenta-derived mesenchymal stem cells (PSCs) have neuroprotective effects. To evaluate the potential benefit of NPCs, we compared them to PSCs using R28 cells under hypoxic conditions and a rat model of optic nerve injury. NPCs and PSCs (2 × 106 cells) were injected into the subtenon space. After 1, 2, and 4 weeks, we examined changes in target proteins in the retina and optic nerve. NPCs significantly induced vascular endothelial growth factor (Vegf) compared to age-matched shams and PSC groups at 2 weeks; they also induced neurofilaments in the retina compared to the sham group at 4 weeks. In addition, the expression of brain-derived neurotrophic factor (Bdnf) was high in the retina in the NPC group at 2 weeks, while expression in the optic nerve was high in both the NPC and PSC groups. The low expression of ionized calcium-binding adapter molecule 1 (Iba1) in the retina had recovered at 2 weeks after NPC injection and at 4 weeks after PSC injection. The expression of the inflammatory protein NLR family, pyrin domain containing 3 (Nlrp3) was significantly reduced at 1 week, and that of tumor necrosis factor-α (Tnf-α) in the optic nerves of the NPC group was lower at 2 weeks. Regarding retinal ganglion cells, the expressions of Brn3a and Tuj1 in the retina were enhanced in the NPC group compared to sham controls at 4 weeks. NPC injections increased Gap43 expression from 2 weeks and reduced Iba1 expression in the optic nerves during the recovery period. In addition, R28 cells exposed to hypoxic conditions showed increased cell survival when cocultured with NPCs compared to PSCs. Both Wnt/ß-catenin signaling and increased Nf-ĸb could contribute to the rescue of damaged retinal ganglion cells via upregulation of neuroprotective factors, microglial engagement, and anti-inflammatory regulation by NPCs. This study suggests that NPCs could be useful for the cellular treatment of various optic neuropathies, together with cell therapy using mesenchymal stem cells.


Asunto(s)
Células-Madre Neurales/trasplante , Enfermedades del Nervio Óptico/terapia , Traumatismos del Nervio Óptico/terapia , Nervio Óptico/crecimiento & desarrollo , Células Madre Pluripotentes/trasplante , Animales , Axones/metabolismo , Axones/fisiología , Supervivencia Celular/genética , Tratamiento Basado en Trasplante de Células y Tejidos , Modelos Animales de Enfermedad , Femenino , Humanos , Regeneración Nerviosa/genética , Nervio Óptico/patología , Nervio Óptico/trasplante , Enfermedades del Nervio Óptico/patología , Embarazo , Ratas , Células Ganglionares de la Retina/trasplante
9.
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
10.
In Vitro Cell Dev Biol Anim ; 57(6): 587-597, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-34212340

RESUMEN

Conventional methods for obtaining pancreatic ß cells are based on simulating the embryonic development phase of endocrine cells via hierarchical differentiation of pluripotent stem cells (PSCs). Accordingly, we attempted to modify the protocols for obtaining insulin-secreting cells (ISCs) by sequential differentiation of a human embryonic stem cell (hESC), using the HS181 cell line. Furthermore, we hypothesize that actual pancreatic endocrine cells may arise from trans-differentiation of mature ductal cells after the embryonic developmental stage and throughout the rest of life. According to the hypothesis, ductal cells are trans-differentiated into endocrine and exocrine cells, undergoing a partial epithelial to mesenchymal transition (EMT). To address this issue, we developed two new protocols based on hESC differentiation to obtain ductal cells and then induce EMT in cells to obtain hormone-secreting islet-like cells (HSCs). The ductal (pre-EMT exocrine) cells were then induced to undergo partial EMT by treating with Wnt3a and activin A, in hypoxia. The cell derived from the latter method significantly expressed the main endocrine cell-specific markers and also ß cells, in particular. These experiments not only support our hypothetical model but also offer a promising approach to develop new methods to compensate ß cell depletion in patients with type 1 diabetes mellitus (T1DM). Although this protocol of generating islet-like cells from ductal cells has a potential to treat T1DM, this strategy may be exploited to optimize the function of these cells in an animal model and future clinical applications.


Asunto(s)
Transdiferenciación Celular/genética , Diabetes Mellitus Tipo 1/terapia , Células Madre Embrionarias Humanas/citología , Células Madre Pluripotentes/citología , Proteína Wnt3A/genética , Técnicas de Cultivo de Célula , Diferenciación Celular/genética , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patología , Células Endocrinas/citología , Transición Epitelial-Mesenquimal/genética , Células Madre Embrionarias Humanas/trasplante , Humanos , Insulina/genética , Insulina/metabolismo , Secreción de Insulina/genética , Células Secretoras de Insulina/patología , Células Secretoras de Insulina/trasplante , Páncreas/crecimiento & desarrollo , Páncreas/patología , Células Madre Pluripotentes/trasplante
11.
Science ; 373(6554): 516-522, 2021 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-34326233

RESUMEN

Technological advancements in blood glucose monitoring and therapeutic insulin administration have improved the quality of life for people with type 1 diabetes. However, these efforts fall short of replicating the exquisite metabolic control provided by native islets. We examine the integrated advancements in islet cell replacement and immunomodulatory therapies that are coalescing to enable the restoration of endogenous glucose regulation. We highlight advances in stem cell biology and graft site design, which offer innovative sources of cellular material and improved engraftment. We also cover cutting-edge approaches for preventing allograft rejection and recurrent autoimmunity. These insights reflect a growing understanding of type 1 diabetes etiology, ß cell biology, and biomaterial design, together highlighting therapeutic opportunities to durably replace the ß cells destroyed in type 1 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 1/terapia , Inmunomodulación , Células Secretoras de Insulina/trasplante , Trasplante de Islotes Pancreáticos , Animales , Autoinmunidad , Glucemia/metabolismo , Diferenciación Celular , Ingeniería Celular , Microambiente Celular , Diabetes Mellitus Tipo 1/metabolismo , Rechazo de Injerto/prevención & control , Supervivencia de Injerto , Humanos , Tolerancia Inmunológica , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/fisiología , Islotes Pancreáticos/fisiología , Células Madre Pluripotentes/trasplante , Trasplante de Células Madre
12.
Cell ; 184(8): 2020-2032.e14, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33861963

RESUMEN

Interspecies chimera formation with human pluripotent stem cells (hPSCs) represents a necessary alternative to evaluate hPSC pluripotency in vivo and might constitute a promising strategy for various regenerative medicine applications, including the generation of organs and tissues for transplantation. Studies using mouse and pig embryos suggest that hPSCs do not robustly contribute to chimera formation in species evolutionarily distant to humans. We studied the chimeric competency of human extended pluripotent stem cells (hEPSCs) in cynomolgus monkey (Macaca fascicularis) embryos cultured ex vivo. We demonstrate that hEPSCs survived, proliferated, and generated several peri- and early post-implantation cell lineages inside monkey embryos. We also uncovered signaling events underlying interspecific crosstalk that may help shape the unique developmental trajectories of human and monkey cells within chimeric embryos. These results may help to better understand early human development and primate evolution and develop strategies to improve human chimerism in evolutionarily distant species.


Asunto(s)
Quimerismo , Embrión de Mamíferos/citología , Células Madre Pluripotentes/citología , Animales , Blastocisto/citología , Blastocisto/metabolismo , Diferenciación Celular , Linaje de la Célula , Células Cultivadas , Embrión de Mamíferos/metabolismo , Femenino , Humanos , Macaca fascicularis , Células Madre Pluripotentes/metabolismo , Células Madre Pluripotentes/trasplante , RNA-Seq , Análisis de la Célula Individual , Transcriptoma
13.
Sci Rep ; 11(1): 7177, 2021 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-33785778

RESUMEN

Cellular therapies based on human pluripotent stem cells (hPSCs) offer considerable promise for treating numerous diseases including diabetes and end stage liver failure. Stem cell spheroids may be cultured in stirred bioreactors to scale up cell production to cell numbers relevant for use in humans. Despite significant progress in bioreactor culture of stem cells, areas for improvement remain. In this study, we demonstrate that microfluidic encapsulation of hPSCs and formation of spheroids. A co-axial droplet microfluidic device was used to fabricate 400 µm diameter capsules with a poly(ethylene glycol) hydrogel shell and an aqueous core. Spheroid formation was demonstrated for three hPSC lines to highlight broad utility of this encapsulation technology. In-capsule differentiation of stem cell spheroids into pancreatic ß-cells in suspension culture was also demonstrated.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Células Madre Pluripotentes/fisiología , Esferoides Celulares/fisiología , Reactores Biológicos , Cápsulas/química , Técnicas de Cultivo de Célula/instrumentación , Diferenciación Celular , Línea Celular , Supervivencia Celular , Trasplante de Células/métodos , Diabetes Mellitus/terapia , Enfermedad Hepática en Estado Terminal/terapia , Humanos , Hidrogeles/química , Células Secretoras de Insulina/fisiología , Técnicas Analíticas Microfluídicas/instrumentación , Células Madre Pluripotentes/trasplante , Polietilenglicoles/química
14.
Cell Rep ; 34(12): 108889, 2021 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-33761348

RESUMEN

Spinal cord injury (SCI) often results in spasticity. There is currently no effective therapy for spasticity. Here, we describe a method to efficiently differentiate human pluripotent stem cells from spinal GABA neurons. After transplantation into the injured rat spinal cord, the DREADD (designer receptors exclusively activated by designer drug)-expressing spinal progenitors differentiate into GABA neurons, mitigating spasticity-like response of the rat hindlimbs and locomotion deficits in 3 months. Administering clozapine-N-oxide, which activates the grafted GABA neurons, further alleviates spasticity-like response, suggesting an integration of grafted GABA neurons into the local neural circuit. These results highlight the therapeutic potential of the spinal GABA neurons for SCI.


Asunto(s)
Neuronas GABAérgicas/patología , Espasticidad Muscular/patología , Espasticidad Muscular/fisiopatología , Traumatismos de la Médula Espinal/patología , Traumatismos de la Médula Espinal/fisiopatología , Médula Espinal/patología , Potenciales de Acción/fisiología , Animales , Diferenciación Celular , Supervivencia Celular , Humanos , Locomoción , Vértebras Lumbares/patología , Vértebras Lumbares/fisiopatología , Masculino , Neuronas Motoras/patología , Neuronas Motoras/ultraestructura , Espasticidad Muscular/complicaciones , Inhibición Neural , Células Madre Pluripotentes/metabolismo , Células Madre Pluripotentes/trasplante , Ratas Sprague-Dawley , Médula Espinal/fisiopatología , Traumatismos de la Médula Espinal/complicaciones , Traumatismos de la Médula Espinal/terapia , Sinapsis/metabolismo , Sinapsis/ultraestructura
15.
Stem Cells ; 39(7): 882-896, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33657251

RESUMEN

Retinal dystrophies often lead to blindness. Developing therapeutic interventions to restore vision is therefore of paramount importance. Here we demonstrate the ability of pluripotent stem cell-derived cone precursors to engraft and restore light responses in the Pde6brd1 mouse, an end-stage photoreceptor degeneration model. Our data show that up to 1.5% of precursors integrate into the host retina, differentiate into cones, and engraft in close apposition to the host bipolar cells. Half of the transplanted mice exhibited visual behavior and of these 33% showed binocular light sensitivity. The majority of retinal ganglion cells exhibited contrast-sensitive ON, OFF or ON-OFF light responses and even motion sensitivity; however, quite a few exhibited unusual responses (eg, light-induced suppression), presumably reflecting remodeling of the neural retina. Our data indicate that despite relatively low engraftment yield, pluripotent stem cell-derived cone precursors can elicit light responsiveness even at advanced degeneration stages. Further work is needed to improve engraftment yield and counteract retinal remodeling to achieve useful clinical applications.


Asunto(s)
Células Madre Pluripotentes , Células Fotorreceptoras Retinianas Conos , Degeneración Retiniana , Trasplante de Células Madre , Animales , Ratones , Células Madre Pluripotentes/trasplante , Degeneración Retiniana/terapia , Células Ganglionares de la Retina/patología
16.
Diabetologia ; 64(5): 1030-1036, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33454830

RESUMEN

Present-day treatments for people that are insulin dependent require multiple insulin injections, sometimes with an insulin pump, coupled with regular blood glucose monitoring. The availability of modified insulins, each with peaks of activity at varying times, has improved diabetes management. On the other hand, there have been impressive results leading to insulin independence by transplantation of cadaveric islets coupled with immune suppression. This review focuses on the possibility of treating diabetes with cellular transplants, specifically with the use of pluripotent stem cells, to produce a virtually unlimited and uniform supply of human islet-like clusters by directed differentiation. Prospects for improving the in vitro differentiation of human endocrine cells for the study of endocrine function and their possible clinical uses are also discussed.


Asunto(s)
Trasplante de Islotes Pancreáticos , Células Madre Pluripotentes/fisiología , Animales , Diferenciación Celular , Diabetes Mellitus/terapia , Humanos , Islotes Pancreáticos/citología , Islotes Pancreáticos/fisiología , Trasplante de Islotes Pancreáticos/métodos , Trasplante de Islotes Pancreáticos/tendencias , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/trasplante
17.
Stem Cells ; 39(5): 522-535, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33497522

RESUMEN

Strategies to mitigate the pathologies from diabetes range from simply administering insulin to prescribing complex drug/biologic regimens combined with lifestyle changes. There is a substantial effort to better understand ß-cell physiology during diabetes pathogenesis as a means to develop improved therapies. The convergence of multiple fields ranging from developmental biology to microfluidic engineering has led to the development of new experimental systems to better study complex aspects of diabetes and ß-cell biology. Here we discuss the available insulin-secreting cell types used in research, ranging from primary human ß-cells, to cell lines, to pluripotent stem cell-derived ß-like cells. Each of these sources possess inherent strengths and weaknesses pertinent to specific applications, especially in the context of engineered platforms. We then outline how insulin-expressing cells have been used in engineered platforms and how recent advances allow for better mimicry of in vivo conditions. Chief among these conditions are ß-cell interactions with other endocrine organs. This facet is beginning to be thoroughly addressed by the organ-on-a-chip community, but holds enormous potential in the development of novel diabetes therapeutics. Furthermore, high throughput strategies focused on studying ß-cell biology, improving ß-cell differentiation, or proliferation have led to enormous contributions in the field and will no doubt be instrumental in bringing new diabetes therapeutics to the clinic.


Asunto(s)
Diabetes Mellitus/terapia , Células Secretoras de Insulina/metabolismo , Insulina/biosíntesis , Células Madre Pluripotentes/metabolismo , Comunicación Celular/genética , Diferenciación Celular/genética , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patología , Humanos , Insulina/genética , Células Secretoras de Insulina/trasplante , Dispositivos Laboratorio en un Chip , Células Madre Pluripotentes/trasplante
18.
Stem Cell Reports ; 16(1): 39-55, 2021 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-33357408

RESUMEN

Human pluripotent stem cells (hPSCs) are commonly kept in a primed state but also able to acquire a more immature naive state under specific conditions in vitro. Acquisition of naive state changes several properties of hPSCs and might affect their contribution to embryonic development in vivo. However, the lack of an appropriate animal test system has made it difficult to assess potential differences for chimera formation between naive and primed hPSCs. Here, we report that the developing chicken embryo is a permissive host for hPSCs, allowing analysis of the pluripotency potential of hPSCs. Transplantation of naive-like and primed hPSCs at matched developmental stages resulted in robust chimerism. Importantly, the ability of naive-like but not of primed hPSCs to form chimera was substantially reduced when injected at non-matched developmental stages. We propose that contribution to chick embryogenesis is an informative and versatile test to identify different pluripotent states of hPSCs.


Asunto(s)
Embrión de Pollo/metabolismo , Quimerismo/veterinaria , Células Madre Pluripotentes/trasplante , Animales , Diferenciación Celular , Linaje de la Célula , Embrión de Pollo/citología , Pollos , Desarrollo Embrionario , Edición Génica , Humanos , Proteínas con Homeodominio LIM/genética , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Factores de Transcripción/genética , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo
19.
Trends Cardiovasc Med ; 31(2): 85-90, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-31983535

RESUMEN

Today, cell replacement therapy using pluripotent stem cell-derived cardiomyocytes (PSC-CMs) remains a research endeavor, with several hurdles that must be overcome before delivery of PSC-CMs can become a therapeutic reality. In this review, we highlight major findings to date from pre-clinical studies involving delivery of PSC-CMs and consider remaining challenges that must be addressed for successful clinical translation. Our goal is to provide an overview of the current status of cardiomyocyte replacement therapy and what challenges must be addressed before successful clinical translation of such therapies will be possible.


Asunto(s)
Cardiomiopatías/cirugía , Miocardio/patología , Miocitos Cardíacos/trasplante , Células Madre Pluripotentes/trasplante , Regeneración , Medicina Regenerativa/tendencias , Trasplante de Células Madre/tendencias , Animales , Cardiomiopatías/patología , Cardiomiopatías/fisiopatología , Diferenciación Celular , Difusión de Innovaciones , Predicción , Humanos , Recuperación de la Función , Trasplante de Células Madre/efectos adversos
20.
Stem Cell Reports ; 16(1): 10-19, 2021 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-33275879

RESUMEN

Inducible expression of PAX7 in differentiating pluripotent stem cells (PSCs) allows massively scalable generation of human myogenic progenitors, which upon transplantation into dystrophic muscles give rise to donor-derived myofibers and satellite cells. Therefore, PSC-derived PAX7+ myogenic progenitors represent an attractive therapeutic approach to promote muscle regeneration. Work to date has used lentiviral vectors (LVs) that randomly integrate inducible PAX7 transgenes. Here, we investigated whether equivalent induction of the myogenic program could be achieved by targeting the PAX7 transgene into genomic safe harbor (GSH) sites. Across multiple PSC lines, we find that this approach consistently generates expandable myogenic progenitors in vitro, although scalability of expansion is moderately reduced compared with the LV approach. Importantly, transplantation of GSH-targeted myogenic progenitors produces robust engraftment, comparable with LV counterparts. These findings provide proof of concept for the use of GSH targeting as a potential alternative approach to generate therapeutic PSC-derived myogenic progenitors for clinical applications.


Asunto(s)
Factor de Transcripción PAX7/genética , Células Madre/metabolismo , Animales , Diferenciación Celular , Línea Celular , Modelos Animales de Enfermedad , Distrofina/genética , Cuerpos Embrioides/citología , Cuerpos Embrioides/metabolismo , Sitios Genéticos , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Humanos , Lentivirus/genética , Ratones , Desarrollo de Músculos , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/patología , Distrofia Muscular de Duchenne/terapia , Factor de Transcripción PAX7/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Células Madre Pluripotentes/trasplante , Trasplante de Células Madre , Células Madre/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...