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










Base de datos
Intervalo de año de publicación
1.
Cell Stem Cell ; 30(10): 1299-1314.e9, 2023 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-37802036

RESUMEN

Cell replacement therapies for Parkinson's disease (PD) based on transplantation of pluripotent stem cell-derived dopaminergic neurons are now entering clinical trials. Here, we present quality, safety, and efficacy data supporting the first-in-human STEM-PD phase I/IIa clinical trial along with the trial design. The STEM-PD product was manufactured under GMP and quality tested in vitro and in vivo to meet regulatory requirements. Importantly, no adverse effects were observed upon testing of the product in a 39-week rat GLP safety study for toxicity, tumorigenicity, and biodistribution, and a non-GLP efficacy study confirmed that the transplanted cells mediated full functional recovery in a pre-clinical rat model of PD. We further observed highly comparable efficacy results between two different GMP batches, verifying that the product can be serially manufactured. A fully in vivo-tested batch of STEM-PD is now being used in a clinical trial of 8 patients with moderate PD, initiated in 2022.


Asunto(s)
Células Madre Embrionarias Humanas , Enfermedad de Parkinson , Humanos , Ratas , Animales , Enfermedad de Parkinson/terapia , Distribución Tisular , Diferenciación Celular/fisiología , Trasplante de Células Madre/métodos , Neuronas Dopaminérgicas/fisiología
2.
Nat Commun ; 13(1): 3046, 2022 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-35650213

RESUMEN

Stem cell therapies for Parkinson's disease (PD) have entered first-in-human clinical trials using a set of technically related methods to produce mesencephalic dopamine (mDA) neurons from human pluripotent stem cells (hPSCs). Here, we outline an approach for high-yield derivation of mDA neurons that principally differs from alternative technologies by utilizing retinoic acid (RA) signaling, instead of WNT and FGF8 signaling, to specify mesencephalic fate. Unlike most morphogen signals, where precise concentration determines cell fate, it is the duration of RA exposure that is the key-parameter for mesencephalic specification. This concentration-insensitive patterning approach provides robustness and reduces the need for protocol-adjustments between hPSC-lines. RA-specified progenitors promptly differentiate into functional mDA neurons in vitro, and successfully engraft and relieve motor deficits after transplantation in a rat PD model. Our study provides a potential alternative route for cell therapy and disease modelling that due to its robustness could be particularly expedient when use of autologous- or immunologically matched cells is considered.


Asunto(s)
Enfermedad de Parkinson , Células Madre Pluripotentes , Animales , Diferenciación Celular , Neuronas Dopaminérgicas , Humanos , Mesencéfalo , Enfermedad de Parkinson/terapia , Ratas , Tretinoina/farmacología
3.
Stem Cell Reports ; 17(1): 159-172, 2022 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-34971563

RESUMEN

Transplantation in Parkinson's disease using human embryonic stem cell (hESC)-derived dopaminergic (DA) neurons is a promising future treatment option. However, many of the mechanisms that govern their differentiation, maturation, and integration into the host circuitry remain elusive. Here, we engrafted hESCs differentiated toward a ventral midbrain DA phenotype into the midbrain of a preclinical rodent model of Parkinson's disease. We then injected a novel DA-neurotropic retrograde MNM008 adeno-associated virus vector capsid, into specific DA target regions to generate starter cells based on their axonal projections. Using monosynaptic rabies-based tracing, we demonstrated for the first time that grafted hESC-derived DA neurons receive distinctly different afferent inputs depending on their projections. The similarities to the host DA system suggest a previously unknown directed circuit integration. By evaluating the differential host-to-graft connectivity based on projection patterns, this novel approach offers a tool to answer outstanding questions regarding the integration of grafted hESC-derived DA neurons.


Asunto(s)
Diferenciación Celular , Neuronas Dopaminérgicas/citología , Neuronas Dopaminérgicas/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Células Madre Embrionarias Humanas/citología , Células Madre Embrionarias Humanas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Sinapsis/metabolismo , Biomarcadores , Rastreo Celular , Expresión Génica , Genes Reporteros , Factores de Intercambio de Guanina Nucleótido/genética , Humanos , Mesencéfalo/metabolismo , Fenotipo , Proteínas Serina-Treonina Quinasas/genética , Trasplante de Células Madre
5.
Nat Commun ; 11(1): 2434, 2020 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-32415072

RESUMEN

Cell replacement is a long-standing and realistic goal for the treatment of Parkinson's disease (PD). Cells for transplantation can be obtained from fetal brain tissue or from stem cells. However, after transplantation, dopamine (DA) neurons are seen to be a minor component of grafts, and it has remained difficult to determine the identity of other cell types. Here, we report analysis by single-cell RNA sequencing (scRNA-seq) combined with comprehensive histological analyses to characterize intracerebral grafts from human embryonic stem cells (hESCs) and fetal tissue after functional maturation in a pre-clinical rat PD model. We show that neurons and astrocytes are major components in both fetal and stem cell-derived grafts. Additionally, we identify a cell type closely resembling a class of recently identified perivascular-like cells in stem cell-derived grafts. Thus, this study uncovers previously unknown cellular diversity in a clinically relevant cell replacement PD model.


Asunto(s)
Neuronas Dopaminérgicas/citología , Enfermedad de Parkinson/terapia , Trasplante de Células Madre , Células Madre/citología , Animales , Encéfalo/metabolismo , Diferenciación Celular , Cuerpo Estriado , Modelos Animales de Enfermedad , Dopamina/metabolismo , Células Madre Embrionarias/citología , Femenino , Supervivencia de Injerto , Humanos , Familia de Multigenes , RNA-Seq , Ratas , Ratas Desnudas , Regeneración , Análisis de la Célula Individual , Transcriptoma
6.
Stem Cells ; 38(6): 716-726, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32101353

RESUMEN

Fetal neural progenitor grafts have been evaluated in preclinical animal models of spinal cord injury and Parkinson's disease for decades, but the initial reliance on primary tissue as a cell source limited the scale of their clinical translatability. With the development of robust methods to differentiate human pluripotent stem cells to specific neural subtypes, cell replacement therapy holds renewed promise to treat a variety of neurodegenerative diseases and injuries at scale. As these cell sources are evaluated in preclinical models, new transsynaptic tracing methods are making it possible to study the connectivity between host and graft neurons with greater speed and detail than was previously possible. To date, these studies have revealed that widespread, long-lasting, and anatomically appropriate synaptic contacts are established between host and graft neurons, as well as new aspects of host-graft connectivity which may be relevant to clinical cell replacement therapy. It is not yet clear, however, whether the synaptic connectivity between graft and host neurons is as cell-type specific as it is in the endogenous nervous system, or whether that connectivity is responsible for the functional efficacy of cell replacement therapy. Here, we review evidence suggesting that the new contacts established between host and graft neurons may indeed be cell-type specific, and how transsynaptic tracing can be used in the future to further elucidate the mechanisms of graft-mediated functional recovery in spinal cord injury and Parkinson's disease.


Asunto(s)
Células-Madre Neurales/trasplante , Enfermedad de Parkinson/terapia , Traumatismos de la Médula Espinal/terapia , Humanos
7.
Cell Rep ; 28(13): 3462-3473.e5, 2019 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-31553914

RESUMEN

Cell replacement is currently being explored as a therapeutic approach for neurodegenerative disease. Using stem cells as a source, transplantable progenitors can now be generated under conditions compliant with clinical application in patients. In this study, we elucidate factors controlling target-appropriate innervation and circuitry integration of human embryonic stem cell (hESC)-derived grafts after transplantation to the adult brain. We show that cell-intrinsic factors determine graft-derived axonal innervation, whereas synaptic inputs from host neurons primarily reflect the graft location. Furthermore, we provide evidence that hESC-derived dopaminergic grafts transplanted in a long-term preclinical rat model of Parkinson's disease (PD) receive synaptic input from subtypes of host cortical, striatal, and pallidal neurons that are known to regulate the function of endogenous nigral dopamine neurons. This refined understanding of how graft neurons integrate with host circuitry will be important for the design of clinical stem-cell-based replacement therapies for PD, as well as for other neurodegenerative diseases.


Asunto(s)
Ganglios Basales/fisiopatología , Células Madre Embrionarias Humanas/metabolismo , Enfermedad de Parkinson/genética , Animales , Modelos Animales de Enfermedad , Humanos , Ratones Desnudos , Ratas
8.
MedEdPORTAL ; 15: 10839, 2019 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-31976362

RESUMEN

Introduction: Cardiac auscultation skills are essential to the development of a competent physician. We created a hypothesis-driven cardiac auscultation laboratory session utilizing a high-fidelity simulator to teach these skills to second-year medical students at our institution. This program was grounded in deliberate practice opportunities to aid in the acquisition of cardiac auscultation skills. Methods: This session aimed to help students identify and discriminate between normal and pathologic heart sounds in the context of a clinical vignette. Faculty facilitators guided students through unknown patient cases and utilized the auscultation manikin to simulate corresponding heart sounds. Time was also allotted for students to auscultate the manikins and practice their cardiac physical examination skills. Results: This program has been in place at our institution since 2016 and has been well received by students and facilitators. Since its initial introduction in 2016, 183 second-year medical students have completed the cardiac auscultation lab session each year, for a total of 549 students. Evaluations of the session have improved as faculty have become more familiar with the mechanics of operating the auscultation manikin. Discussion: The cardiac exam and heart sounds lab can be adapted to any simulator model that is capable of producing heart sounds and can be done in a large- or small-group format. Enough time should be allotted to adequately work through all components of the laboratory. Student and faculty feedback has helped us further refine the session since its initial introduction to the curriculum.


Asunto(s)
Competencia Clínica/normas , Auscultación Cardíaca/normas , Cardiopatías/diagnóstico , Ruidos Cardíacos/fisiología , Maniquíes , Simulación de Paciente , Estudiantes de Medicina , Curriculum , Evaluación Educacional , Cardiopatías/fisiopatología , Humanos
9.
Nat Methods ; 15(9): 723-731, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30082899

RESUMEN

Spinal cord neural stem cells (NSCs) have great potential to reconstitute damaged spinal neural circuitry, but they have yet to be generated in vitro. We now report the derivation of spinal cord NSCs from human pluripotent stem cells (hPSCs). Our observations show that these spinal cord NSCs differentiate into a diverse population of spinal cord neurons occupying multiple positions along the dorso-ventral axis, and can be maintained for prolonged time periods. Grafts into injured spinal cords were rich with excitatory neurons, extended large numbers of axons over long distances, innervated their target structures, and enabled robust corticospinal regeneration. The grafts synaptically integrated into multiple host intraspinal and supraspinal systems, including the corticospinal projection, and improved functional outcomes after injury. hPSC-derived spinal cord NSCs could enable a broad range of biomedical applications for in vitro disease modeling and constitute an improved clinically translatable cell source for 'replacement' strategies in several spinal cord disorders.


Asunto(s)
Células-Madre Neurales/patología , Células Madre Pluripotentes/patología , Traumatismos de la Médula Espinal/patología , Médula Espinal/patología , Linaje de la Célula , Humanos
10.
J Comp Neurol ; 526(13): 2133-2146, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-30007046

RESUMEN

Dopamine (DA) neurons derived from human embryonic stem cells (hESCs) are a promising unlimited source of cells for cell replacement therapy in Parkinson's disease (PD). A number of studies have demonstrated functionality of DA neurons originating from hESCs when grafted to the striatum of rodent and non-human primate models of PD. However, several questions remain in regard to their axonal outgrowth potential and capacity to integrate into host circuitry. Here, ventral midbrain (VM) patterned hESC-derived progenitors were grafted into the midbrain of 6-hydroxydopamine-lesioned rats, and analyzed at 6, 18, and 24 weeks for a time-course evaluation of specificity and extent of graft-derived fiber outgrowth as well as potential for functional recovery. To investigate synaptic integration of the transplanted cells, we used rabies-based monosynaptic tracing to reveal the origin and extent of host presynaptic inputs to grafts at 6 weeks. The results reveal the capacity of grafted neurons to extend axonal projections toward appropriate forebrain target structures progressively over 24 weeks. The timing and extent of graft-derived dopaminergic fibers innervating the dorsolateral striatum matched reduction in amphetamine-induced rotational asymmetry in the animals where recovery could be observed. Monosynaptic tracing demonstrated that grafted cells integrate with host circuitry 6 weeks after transplantation, in a manner that is comparable with endogenous midbrain connectivity. Thus, we demonstrate that VM patterned hESC-derived progenitors grafted to midbrain have the capacity to extensively innervate appropriate forebrain targets, integrate into the host circuitry and that functional recovery can be achieved when grafting fetal or hESC-derived DA neurons to the midbrain.


Asunto(s)
Neuronas Dopaminérgicas/fisiología , Neuronas Dopaminérgicas/trasplante , Mesencéfalo/cirugía , Vías Nerviosas/fisiología , Células-Madre Neurales/fisiología , Células-Madre Neurales/trasplante , Trastornos Parkinsonianos/cirugía , Prosencéfalo/fisiología , Sinapsis/fisiología , Anfetamina/farmacología , Animales , Inhibidores de Captación de Dopamina/farmacología , Femenino , Humanos , Hidroxidopaminas , Ratones , Fibras Nerviosas/fisiología , Trastornos Parkinsonianos/inducido químicamente , Ratas Desnudas , Trasplante de Células Madre , Conducta Estereotipada/efectos de los fármacos
11.
Nat Commun ; 9(1): 84, 2018 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-29311559

RESUMEN

Neural progenitor cell (NPC) transplantation has high therapeutic potential in neurological disorders. Functional restoration may depend on the formation of reciprocal connections between host and graft. While it has been reported that axons extending out of neural grafts in the brain form contacts onto phenotypically appropriate host target regions, it is not known whether adult, injured host axons regenerating into NPC grafts also form appropriate connections. We report that spinal cord NPCs grafted into the injured adult rat spinal cord self-assemble organotypic, dorsal horn-like domains. These clusters are extensively innervated by regenerating adult host sensory axons and are avoided by corticospinal axons. Moreover, host axon regeneration into grafts increases significantly after enrichment with appropriate neuronal targets. Together, these findings demonstrate that injured adult axons retain the ability to recognize appropriate targets and avoid inappropriate targets within neural progenitor grafts, suggesting that restoration of complex circuitry after SCI may be achievable.


Asunto(s)
Axones/fisiología , Neuronas Motoras/fisiología , Regeneración Nerviosa/fisiología , Células-Madre Neurales/trasplante , Células Receptoras Sensoriales/fisiología , Asta Dorsal de la Médula Espinal/fisiología , Traumatismos de la Médula Espinal/terapia , Animales , Femenino , Masculino , Células-Madre Neurales/fisiología , Ratas , Médula Espinal/citología , Trasplante de Células Madre
12.
Stem Cell Reports ; 8(6): 1525-1533, 2017 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-28479302

RESUMEN

Neural progenitor cells grafted to sites of spinal cord injury have supported electrophysiological and functional recovery in several studies. Mechanisms associated with graft-related improvements in outcome appear dependent on functional synaptic integration of graft and host systems, although the extent and diversity of synaptic integration of grafts with hosts are unknown. Using transgenic mouse spinal neural progenitor cell grafts expressing the TVA and G-protein components of the modified rabies virus system, we initiated monosynaptic tracing strictly from graft neurons placed in sites of cervical spinal cord injury. We find that graft neurons receive synaptic inputs from virtually every known host system that normally innervates the spinal cord, including numerous cortical, brainstem, spinal cord, and dorsal root ganglia inputs. Thus, implanted neural progenitor cells receive an extensive range of host neural inputs to the injury site, potentially enabling functional restoration across multiple systems.


Asunto(s)
Virus de la Rabia/metabolismo , Traumatismos de la Médula Espinal/patología , Animales , Proteínas Aviares/metabolismo , Diferenciación Celular , Células Cultivadas , Ganglios Espinales/metabolismo , Glicoproteínas/genética , Glicoproteínas/metabolismo , Ratones , Ratones Desnudos , Ratones Transgénicos , Microscopía Fluorescente , Células-Madre Neurales/citología , Células-Madre Neurales/trasplante , Células-Madre Neurales/virología , Neuronas/metabolismo , Virus de la Rabia/genética , Receptores Virales/metabolismo , Recuperación de la Función , Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/terapia , Proteínas Virales/genética , Proteínas Virales/metabolismo
13.
Sci Rep ; 7: 40285, 2017 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-28071742

RESUMEN

Transdifferentiation has been described as a novel method for converting human fibroblasts into induced cardiomyocyte-like cells. Such an approach can produce differentiated cells to study physiology or pathophysiology, examine drug interactions or toxicities, and engineer cardiac tissues. Here we describe the transdifferentiation of human dermal fibroblasts towards the cardiac cell lineage via the induced expression of transcription factors GATA4, TBX5, MEF2C, MYOCD, NKX2-5, and delivery of microRNAs miR-1 and miR-133a. Cells undergoing transdifferentiation expressed ACTN2 and TNNT2 and partially organized their cytoskeleton in a cross-striated manner. The conversion process was associated with significant upregulation of a cohort of cardiac-specific genes, activation of pathways associated with muscle contraction and physiology, and downregulation of fibroblastic markers. We used a genetically encoded calcium indicator and readily detected active calcium transients although no spontaneous contractions were observed in transdifferentiated cells. Finally, we determined that inhibition of Janus kinase 1, inhibition of Glycogen synthase kinase 3, or addition of NRG1 significantly enhanced the efficiency of transdifferentiation. Overall, we describe a method for achieving transdifferentiation of human dermal fibroblasts into induced cardiomyocyte-like cells via transcription factor overexpression, microRNA delivery, and molecular pathway manipulation.


Asunto(s)
Linaje de la Célula/genética , Transdiferenciación Celular/genética , Fibroblastos/citología , Miocitos Cardíacos/citología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Transdiferenciación Celular/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Humanos , MicroARNs/genética , Proteínas Nucleares/genética , Bibliotecas de Moléculas Pequeñas/farmacología , Transactivadores/genética , Factores de Transcripción/genética , Activación Transcripcional/efectos de los fármacos , Activación Transcripcional/genética
14.
Cell Stem Cell ; 19(3): 406-14, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27524438

RESUMEN

Overexpression of exogenous fate-specifying transcription factors can directly reprogram differentiated somatic cells to target cell types. Here, we show that similar reprogramming can also be achieved through the direct activation of endogenous genes using engineered CRISPR/Cas9-based transcriptional activators. We use this approach to induce activation of the endogenous Brn2, Ascl1, and Myt1l genes (BAM factors) to convert mouse embryonic fibroblasts to induced neuronal cells. This direct activation of endogenous genes rapidly remodeled the epigenetic state of the target loci and induced sustained endogenous gene expression during reprogramming. Thus, transcriptional activation and epigenetic remodeling of endogenous master transcription factors are sufficient for conversion between cell types. The rapid and sustained activation of endogenous genes in their native chromatin context by this approach may facilitate reprogramming with transient methods that avoid genomic integration and provides a new strategy for overcoming epigenetic barriers to cell fate specification.


Asunto(s)
Sistemas CRISPR-Cas/genética , Epigénesis Genética , Fibroblastos/citología , Sitios Genéticos , Neuronas/citología , Transactivadores/metabolismo , Animales , Células Cultivadas , Embrión de Mamíferos/citología , Fibroblastos/metabolismo , Marcadores Genéticos , Vectores Genéticos/metabolismo , Células HEK293 , Humanos , Lentivirus/genética , Ratones Endogámicos C57BL , Neuronas/metabolismo , ARN Guía de Kinetoplastida/metabolismo , Factores de Transcripción/metabolismo , Activación Transcripcional
15.
Nanomedicine ; 12(2): 399-409, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26711960

RESUMEN

Safety concerns and/or the stochastic nature of current transduction approaches have hampered nuclear reprogramming's clinical translation. We report a novel non-viral nanotechnology-based platform permitting deterministic large-scale transfection with single-cell resolution. The superior capabilities of our technology are demonstrated by modification of the well-established direct neuronal reprogramming paradigm using overexpression of the transcription factors Brn2, Ascl1, and Myt1l (BAM). Reprogramming efficiencies were comparable to viral methodologies (up to ~9-12%) without the constraints of capsid size and with the ability to control plasmid dosage, in addition to showing superior performance relative to existing non-viral methods. Furthermore, increased neuronal complexity could be tailored by varying BAM ratio and by including additional proneural genes to the BAM cocktail. Furthermore, high-throughput NEP allowed easy interrogation of the reprogramming process. We discovered that BAM-mediated reprogramming is regulated by AsclI dosage, the S-phase cyclin CCNA2, and that some induced neurons passed through a nestin-positive cell stage. FROM THE CLINICAL EDITOR: In the field of regenerative medicine, the ability to direct cell fate by nuclear reprogramming is an important facet in terms of clinical application. In this article, the authors described their novel technique of cell reprogramming through overexpression of the transcription factors Brn2, Ascl1, and Myt1l (BAM) by in situ electroporation through nanochannels. This new technique could provide a platform for further future designs.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Reprogramación Celular , Proteínas de Unión al ADN/genética , ADN/administración & dosificación , Proteínas del Tejido Nervioso/genética , Neuronas/citología , Factores del Dominio POU/genética , Factores de Transcripción/genética , Transfección/métodos , Animales , Línea Celular , ADN/genética , Electroporación/métodos , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Ratones , Neuronas/metabolismo , Plásmidos/administración & dosificación , Plásmidos/genética , Regulación hacia Arriba
16.
Biomaterials ; 35(20): 5327-5336, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24709523

RESUMEN

Cellular reprogramming holds tremendous potential for cell therapy and regenerative medicine. Recently, fibroblasts have been directly converted into induced neurons (iNs) by overexpression of the neuronal transcription factors Ascl1, Brn2 and Myt1L. Hypothesizing that cell-topography interactions could influence the fibroblast-to-neuron reprogramming process, we investigated the effects of various topographies on iNs produced by direct reprogramming. Final iN purity and conversion efficiency were increased on micrograting substrates. Neurite branching was increased on microposts and decreased on microgratings, with a simplified dendritic arbor characterized by the reduction of MAP2(+) neurites. Neurite outgrowth increased significantly on various topographies. DNA microarray analysis detected 20 differentially expressed genes in iNs reprogrammed on smooth versus microgratings, and quantitative PCR (qPCR) confirmed the upregulation of Vip and downregulation of Thy1 and Bmp5 on microgratings. Electrophysiology and calcium imaging verified the functionality of these iNs. This study demonstrates the potential of applying topographical cues to optimize cellular reprogramming.


Asunto(s)
Reprogramación Celular , Fibroblastos/citología , Neuronas/citología , Animales , Materiales Biocompatibles/química , Técnicas de Cultivo de Célula , Células Cultivadas , Expresión Génica , Inmunohistoquímica , Ratones , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuritas , Neurogénesis/efectos de los fármacos , Medicina Regenerativa , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
17.
Nat Methods ; 10(10): 973-6, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23892895

RESUMEN

Technologies for engineering synthetic transcription factors have enabled many advances in medical and scientific research. In contrast to existing methods based on engineering of DNA-binding proteins, we created a Cas9-based transactivator that is targeted to DNA sequences by guide RNA molecules. Coexpression of this transactivator and combinations of guide RNAs in human cells induced specific expression of endogenous target genes, demonstrating a simple and versatile approach for RNA-guided gene activation.


Asunto(s)
Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Ingeniería de Proteínas/métodos , Edición de ARN , Factores de Transcripción/genética , Activación Transcripcional , Células HEK293 , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Proteína Antagonista del Receptor de Interleucina 1/genética , Ribonucleasas/genética , ARN Pequeño no Traducido
18.
PLoS One ; 8(5): e63577, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23704920

RESUMEN

Transient overexpression of defined combinations of master regulator genes can effectively induce cellular reprogramming: the acquisition of an alternative predicted phenotype from a differentiated cell lineage. This can be of particular importance in cardiac regenerative medicine wherein the heart lacks the capacity to heal itself, but simultaneously contains a large pool of fibroblasts. In this study we determined the cardio-inducing capacity of ten transcription factors to actuate cellular reprogramming of mouse embryonic fibroblasts into cardiomyocyte-like cells. Overexpression of transcription factors MYOCD and SRF alone or in conjunction with Mesp1 and SMARCD3 enhanced the basal but necessary cardio-inducing effect of the previously reported GATA4, TBX5, and MEF2C. In particular, combinations of five or seven transcription factors enhanced the activation of cardiac reporter vectors, and induced an upregulation of cardiac-specific genes. Global gene expression analysis also demonstrated a significantly greater cardio-inducing effect when the transcription factors MYOCD and SRF were used. Detection of cross-striated cells was highly dependent on the cell culture conditions and was enhanced by the addition of valproic acid and JAK inhibitor. Although we detected Ca(2+) transient oscillations in the reprogrammed cells, we did not detect significant changes in resting membrane potential or spontaneously contracting cells. This study further elucidates the cardio-inducing effect of the transcriptional networks involved in cardiac cellular reprogramming, contributing to the ongoing rational design of a robust protocol required for cardiac regenerative therapies.


Asunto(s)
Reprogramación Celular , Proteínas Cromosómicas no Histona/metabolismo , Factor de Transcripción GATA4/metabolismo , Factores de Transcripción MEF2/metabolismo , Proteínas Musculares/metabolismo , Miocitos Cardíacos/metabolismo , Proteínas de Dominio T Box/metabolismo , Factores de Transcripción/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Citoesqueleto/metabolismo , Fenómenos Electrofisiológicos , Embrión de Mamíferos/citología , Fibroblastos/metabolismo , Regulación de la Expresión Génica , Redes Reguladoras de Genes/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Miocitos Cardíacos/citología , Células 3T3 NIH , Proteínas Nucleares/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Especificidad de Órganos , Factor de Respuesta Sérica/metabolismo , Sus scrofa , Transactivadores/metabolismo , Transcripción Genética , Transcriptoma/genética
19.
Mol Ther Nucleic Acids ; 1: e32, 2012 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-23344148

RESUMEN

Transdifferentiation, where differentiated cells are reprogrammed into another lineage without going through an intermediate proliferative stem cell-like stage, is the next frontier of regenerative medicine. Wernig et al. first described the direct conversion of fibroblasts into functional induced neuronal cells (iNs). Subsequent reports of transdifferentiation into clinically relevant neuronal subtypes have further endorsed the prospect of autologous cell therapy for neurodegenerative disorders. So far, all published neuronal transdifferentiation protocols rely on lentiviruses, which likely precludes their clinical translation. Instead, we delivered plasmids encoding neuronal transcription factors (Brn2, Ascl1, Myt1l) to primary mouse embryonic fibroblasts with a bioreducible linear poly(amido amine). The low toxicity and high transfection efficiency of this gene carrier allowed repeated dosing to sustain high transgene expression levels. Serial 0.5 µg cm(-2) doses of reprogramming factors delivered at 48-hour intervals produced up to 7.6% Tuj1(+) (neuron-specific class III ß-tubulin) cells, a subset of which expressed MAP2 (microtubule-associated protein 2), tau, and synaptophysin. A synapsin-red fluorescent protein (RFP) reporter helped to identify more mature, electrophysiologically active cells, with 24/26 patch-clamped RFP(+) cells firing action potentials. Some non-virally induced neuronal cells (NiNs) were observed firing multiple and spontaneous action potentials. This study demonstrates the feasibility of nonviral neuronal transdifferentiation, and may be amenable to other transdifferentiation processes.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA