Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 81
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 186(2): 287-304.e26, 2023 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-36610399

RESUMEN

Whether and how certain transposable elements with viral origins, such as endogenous retroviruses (ERVs) dormant in our genomes, can become awakened and contribute to the aging process is largely unknown. In human senescent cells, we found that HERVK (HML-2), the most recently integrated human ERVs, are unlocked to transcribe viral genes and produce retrovirus-like particles (RVLPs). These HERVK RVLPs constitute a transmissible message to elicit senescence phenotypes in young cells, which can be blocked by neutralizing antibodies. The activation of ERVs was also observed in organs of aged primates and mice as well as in human tissues and serum from the elderly. Their repression alleviates cellular senescence and tissue degeneration and, to some extent, organismal aging. These findings indicate that the resurrection of ERVs is a hallmark and driving force of cellular senescence and tissue aging.


Asunto(s)
Envejecimiento , Retrovirus Endógenos , Anciano , Animales , Humanos , Ratones , Envejecimiento/genética , Envejecimiento/patología , Senescencia Celular , Retrovirus Endógenos/genética , Primates
2.
Cell ; 180(5): 984-1001.e22, 2020 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-32109414

RESUMEN

Aging causes a functional decline in tissues throughout the body that may be delayed by caloric restriction (CR). However, the cellular profiles and signatures of aging, as well as those ameliorated by CR, remain unclear. Here, we built comprehensive single-cell and single-nucleus transcriptomic atlases across various rat tissues undergoing aging and CR. CR attenuated aging-related changes in cell type composition, gene expression, and core transcriptional regulatory networks. Immune cells were increased during aging, and CR favorably reversed the aging-disturbed immune ecosystem. Computational prediction revealed that the abnormal cell-cell communication patterns observed during aging, including the excessive proinflammatory ligand-receptor interplay, were reversed by CR. Our work provides multi-tissue single-cell transcriptional landscapes associated with aging and CR in a mammal, enhances our understanding of the robustness of CR as a geroprotective intervention, and uncovers how metabolic intervention can act upon the immune system to modify the process of aging.


Asunto(s)
Envejecimiento/genética , Restricción Calórica , Sistema Inmunológico/metabolismo , Transcriptoma/genética , Envejecimiento/metabolismo , Envejecimiento/patología , Animales , Reprogramación Celular/genética , Regulación de la Expresión Génica/genética , Redes Reguladoras de Genes/genética , Humanos , Ratas , Análisis de la Célula Individual
3.
Nat Rev Mol Cell Biol ; 21(3): 137-150, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32020082

RESUMEN

Ageing is characterized by the functional decline of tissues and organs and the increased risk of ageing-associated disorders. Several 'rejuvenating' interventions have been proposed to delay ageing and the onset of age-associated decline and disease to extend healthspan and lifespan. These interventions include metabolic manipulation, partial reprogramming, heterochronic parabiosis, pharmaceutical administration and senescent cell ablation. As the ageing process is associated with altered epigenetic mechanisms of gene regulation, such as DNA methylation, histone modification and chromatin remodelling, and non-coding RNAs, the manipulation of these mechanisms is central to the effectiveness of age-delaying interventions. This Review discusses the epigenetic changes that occur during ageing and the rapidly increasing knowledge of how these epigenetic mechanisms have an effect on healthspan and lifespan extension, and outlines questions to guide future research on interventions to rejuvenate the epigenome and delay ageing processes.


Asunto(s)
Envejecimiento/genética , Epigénesis Genética/genética , Rejuvenecimiento/fisiología , Animales , Ensamble y Desensamble de Cromatina/genética , Metilación de ADN/genética , Epigenoma/genética , Epigenómica/métodos , Regulación de la Expresión Génica/genética , Código de Histonas/genética , Humanos , Longevidad/genética
4.
Cell ; 169(2): 243-257.e25, 2017 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-28388409

RESUMEN

Of all known cultured stem cell types, pluripotent stem cells (PSCs) sit atop the landscape of developmental potency and are characterized by their ability to generate all cell types of an adult organism. However, PSCs show limited contribution to the extraembryonic placental tissues in vivo. Here, we show that a chemical cocktail enables the derivation of stem cells with unique functional and molecular features from mice and humans, designated as extended pluripotent stem (EPS) cells, which are capable of chimerizing both embryonic and extraembryonic tissues. Notably, a single mouse EPS cell shows widespread chimeric contribution to both embryonic and extraembryonic lineages in vivo and permits generating single-EPS-cell-derived mice by tetraploid complementation. Furthermore, human EPS cells exhibit interspecies chimeric competency in mouse conceptuses. Our findings constitute a first step toward capturing pluripotent stem cells with extraembryonic developmental potentials in culture and open new avenues for basic and translational research. VIDEO ABSTRACT.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Células Madre Pluripotentes/citología , Animales , Blastocisto/citología , Línea Celular , Quimera/metabolismo , Dimetindeno/farmacología , Humanos , Indicadores y Reactivos/química , Ratones , Minociclina/química , Minociclina/farmacología , Células Madre Pluripotentes/efectos de los fármacos , Poli(ADP-Ribosa) Polimerasa-1/metabolismo
5.
Cell ; 165(1): 13-15, 2016 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-27015301

RESUMEN

Random or not, the mechanisms at play during the first cell-fate determination in mammalian embryos have been debated for years. Studies by Goolam et al. and White et al. shed new light on the molecular mechanisms underlying the intrinsic biases that lead to non-random lineage segregation in early mammalian development.


Asunto(s)
Diferenciación Celular , Embrión de Mamíferos , Animales , Tipificación del Cuerpo , Linaje de la Célula
6.
Cell ; 166(6): 1371-1385, 2016 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-27610564

RESUMEN

The discovery of induced pluripotent stem cells (iPSCs) a decade ago, which we are celebrating in this issue of Cell, represents a landmark discovery in biomedical research. Together with somatic cell nuclear transfer, iPSC generation reveals the remarkable plasticity associated with differentiated cells and provides an unprecedented means for modeling diseases using patient samples. In addition to transcriptional and epigenetic remodeling, cellular reprogramming to pluripotency is also accompanied by a rewiring of metabolic pathways, which ultimately leads to changes in cell identities.


Asunto(s)
Células Madre Pluripotentes Inducidas/metabolismo , Redes y Vías Metabólicas , Animales , Reprogramación Celular , Humanos , Células Madre Pluripotentes Inducidas/citología , Enfermedades Metabólicas/metabolismo , Modelos Biológicos
7.
Nature ; 624(7992): 611-620, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37907096

RESUMEN

Ageing is a critical factor in spinal-cord-associated disorders1, yet the ageing-specific mechanisms underlying this relationship remain poorly understood. Here, to address this knowledge gap, we combined single-nucleus RNA-sequencing analysis with behavioural and neurophysiological analysis in non-human primates (NHPs). We identified motor neuron senescence and neuroinflammation with microglial hyperactivation as intertwined hallmarks of spinal cord ageing. As an underlying mechanism, we identified a neurotoxic microglial state demarcated by elevated expression of CHIT1 (a secreted mammalian chitinase) specific to the aged spinal cords in NHP and human biopsies. In the aged spinal cord, CHIT1-positive microglia preferentially localize around motor neurons, and they have the ability to trigger senescence, partly by activating SMAD signalling. We further validated the driving role of secreted CHIT1 on MN senescence using multimodal experiments both in vivo, using the NHP spinal cord as a model, and in vitro, using a sophisticated system modelling the human motor-neuron-microenvironment interplay. Moreover, we demonstrated that ascorbic acid, a geroprotective compound, counteracted the pro-senescent effect of CHIT1 and mitigated motor neuron senescence in aged monkeys. Our findings provide the single-cell resolution cellular and molecular landscape of the aged primate spinal cord and identify a new biomarker and intervention target for spinal cord degeneration.


Asunto(s)
Senescencia Celular , Quitinasas , Microglía , Neuronas Motoras , Primates , Médula Espinal , Animales , Humanos , Biomarcadores/metabolismo , Quitinasas/metabolismo , Microglía/enzimología , Microglía/metabolismo , Microglía/patología , Neuronas Motoras/metabolismo , Enfermedades Neuroinflamatorias/metabolismo , Enfermedades Neuroinflamatorias/patología , Primates/metabolismo , Reproducibilidad de los Resultados , Análisis de Expresión Génica de una Sola Célula , Médula Espinal/metabolismo , Médula Espinal/patología
9.
Nucleic Acids Res ; 50(6): 3323-3347, 2022 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-35286396

RESUMEN

Aging in humans is intricately linked with alterations in circadian rhythms concomitant with physiological decline and stem cell exhaustion. However, whether the circadian machinery directly regulates stem cell aging, especially in primates, remains poorly understood. In this study, we found that deficiency of BMAL1, the only non-redundant circadian clock component, results in an accelerated aging phenotype in both human and cynomolgus monkey mesenchymal progenitor cells (MPCs). Unexpectedly, this phenotype was mainly attributed to a transcription-independent role of BMAL1 in stabilizing heterochromatin and thus preventing activation of the LINE1-cGAS-STING pathway. In senescent primate MPCs, we observed decreased capacity of BMAL1 to bind to LINE1 and synergistic activation of LINE1 expression. Likewise, in the skin and muscle tissues from the BMAL1-deficient cynomolgus monkey, we observed destabilized heterochromatin and aberrant LINE1 transcription. Altogether, these findings uncovered a noncanonical role of BMAL1 in stabilizing heterochromatin to inactivate LINE1 that drives aging in primate cells.


Asunto(s)
Factores de Transcripción ARNTL , Senescencia Celular , Relojes Circadianos , Macaca fascicularis/metabolismo , Factores de Transcripción ARNTL/genética , Factores de Transcripción ARNTL/metabolismo , Animales , Relojes Circadianos/genética , Ritmo Circadiano , Heterocromatina , Macaca fascicularis/genética
11.
Nat Rev Genet ; 18(3): 180-191, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28045100

RESUMEN

Pluripotency is a state that exists transiently in the early embryo and, remarkably, can be recapitulated in vitro by deriving embryonic stem cells or by reprogramming somatic cells to become induced pluripotent stem cells. The state of pluripotency, which is stabilized by an interconnected network of pluripotency-associated genes, integrates external signals and exerts control over the decision between self-renewal and differentiation at the transcriptional, post-transcriptional and epigenetic levels. Recent evidence of alternative pluripotency states indicates the regulatory flexibility of this network. Insights into the underlying principles of the pluripotency network may provide unprecedented opportunities for studying development and for regenerative medicine.


Asunto(s)
Diferenciación Celular/genética , Reprogramación Celular/genética , Células Madre Embrionarias/citología , Epigenómica , Redes Reguladoras de Genes , Células Madre Pluripotentes/citología , Animales , Células Madre Embrionarias/metabolismo , Humanos , Células Madre Pluripotentes/metabolismo , Medicina Regenerativa
12.
Nature ; 548(7668): 413-419, 2017 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-28783728

RESUMEN

Genome editing has potential for the targeted correction of germline mutations. Here we describe the correction of the heterozygous MYBPC3 mutation in human preimplantation embryos with precise CRISPR-Cas9-based targeting accuracy and high homology-directed repair efficiency by activating an endogenous, germline-specific DNA repair response. Induced double-strand breaks (DSBs) at the mutant paternal allele were predominantly repaired using the homologous wild-type maternal gene instead of a synthetic DNA template. By modulating the cell cycle stage at which the DSB was induced, we were able to avoid mosaicism in cleaving embryos and achieve a high yield of homozygous embryos carrying the wild-type MYBPC3 gene without evidence of off-target mutations. The efficiency, accuracy and safety of the approach presented suggest that it has potential to be used for the correction of heritable mutations in human embryos by complementing preimplantation genetic diagnosis. However, much remains to be considered before clinical applications, including the reproducibility of the technique with other heterozygous mutations.


Asunto(s)
Proteínas Portadoras/genética , Embrión de Mamíferos/metabolismo , Edición Génica/métodos , Mutación/genética , Adulto , Alelos , Blastocisto/metabolismo , Blastocisto/patología , División Celular , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Roturas del ADN de Doble Cadena , Embrión de Mamíferos/patología , Marcación de Gen , Prueba de Complementación Genética , Heterocigoto , Homocigoto , Humanos , Masculino , Mosaicismo , Reparación del ADN por Recombinación/genética , Fase S , Moldes Genéticos , Cigoto/metabolismo , Cigoto/patología
13.
Nature ; 540(7631): 51-59, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27905428

RESUMEN

Chimaeras are both monsters of the ancient imagination and a long-established research tool. Recent advances, particularly those dealing with the identification and generation of various kinds of stem cells, have broadened the repertoire and utility of mammalian interspecies chimaeras and carved out new paths towards understanding fundamental biology as well as potential clinical applications.


Asunto(s)
Quimera , Células Madre/citología , Animales , Evolución Biológica , Blastocisto/citología , Linaje de la Célula , Quimera/embriología , Evaluación Preclínica de Medicamentos , Humanos , Especificidad de la Especie , Investigación con Células Madre/ética , Investigación con Células Madre/legislación & jurisprudencia
14.
Nature ; 540(7632): 270-275, 2016 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-27919073

RESUMEN

Maternally inherited mitochondrial (mt)DNA mutations can cause fatal or severely debilitating syndromes in children, with disease severity dependent on the specific gene mutation and the ratio of mutant to wild-type mtDNA (heteroplasmy) in each cell and tissue. Pathogenic mtDNA mutations are relatively common, with an estimated 778 affected children born each year in the United States. Mitochondrial replacement therapies or techniques (MRT) circumventing mother-to-child mtDNA disease transmission involve replacement of oocyte maternal mtDNA. Here we report MRT outcomes in several families with common mtDNA syndromes. The mother's oocytes were of normal quality and mutation levels correlated with those in existing children. Efficient replacement of oocyte mutant mtDNA was performed by spindle transfer, resulting in embryos containing >99% donor mtDNA. Donor mtDNA was stably maintained in embryonic stem cells (ES cells) derived from most embryos. However, some ES cell lines demonstrated gradual loss of donor mtDNA and reversal to the maternal haplotype. In evaluating donor-to-maternal mtDNA interactions, it seems that compatibility relates to mtDNA replication efficiency rather than to mismatch or oxidative phosphorylation dysfunction. We identify a polymorphism within the conserved sequence box II region of the D-loop as a plausible cause of preferential replication of specific mtDNA haplotypes. In addition, some haplotypes confer proliferative and growth advantages to cells. Hence, we propose a matching paradigm for selecting compatible donor mtDNA for MRT.


Asunto(s)
ADN Mitocondrial/genética , ADN Mitocondrial/uso terapéutico , Herencia Materna/genética , Enfermedades Mitocondriales/genética , Enfermedades Mitocondriales/patología , Terapia de Reemplazo Mitocondrial/métodos , Mutación , Oocitos/metabolismo , Blastocisto/citología , Blastocisto/metabolismo , Línea Celular , Secuencia Conservada/genética , ADN Mitocondrial/biosíntesis , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Femenino , Haplotipos/genética , Humanos , Masculino , Meiosis , Enfermedades Mitocondriales/metabolismo , Enfermedades Mitocondriales/prevención & control , Donación de Oocito , Oocitos/citología , Oocitos/patología , Fosforilación Oxidativa , Linaje , Polimorfismo Genético
15.
Nature ; 540(7631): 144-149, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27851729

RESUMEN

Targeted genome editing via engineered nucleases is an exciting area of biomedical research and holds potential for clinical applications. Despite rapid advances in the field, in vivo targeted transgene integration is still infeasible because current tools are inefficient, especially for non-dividing cells, which compose most adult tissues. This poses a barrier for uncovering fundamental biological principles and developing treatments for a broad range of genetic disorders. Based on clustered regularly interspaced short palindromic repeat/Cas9 (CRISPR/Cas9) technology, here we devise a homology-independent targeted integration (HITI) strategy, which allows for robust DNA knock-in in both dividing and non-dividing cells in vitro and, more importantly, in vivo (for example, in neurons of postnatal mammals). As a proof of concept of its therapeutic potential, we demonstrate the efficacy of HITI in improving visual function using a rat model of the retinal degeneration condition retinitis pigmentosa. The HITI method presented here establishes new avenues for basic research and targeted gene therapies.


Asunto(s)
Sistemas CRISPR-Cas/genética , Edición Génica/métodos , Marcación de Gen/métodos , Genoma/genética , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/terapia , Animales , División Celular , Modelos Animales de Enfermedad , Técnicas de Sustitución del Gen , Terapia Genética/métodos , Neuronas/citología , Neuronas/metabolismo , Ratas , Homología de Secuencia
16.
Eur J Neurosci ; 49(4): 561-589, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30656775

RESUMEN

Recent advances in cell reprogramming have enabled assessment of disease-related cellular traits in patient-derived somatic cells, thus providing a versatile platform for disease modeling and drug development. Given the limited access to vital human brain cells, this technology is especially relevant for neurodegenerative disorders such as Parkinson's disease (PD) as a tool to decipher underlying pathomechanisms. Importantly, recent progress in genome-editing technologies has provided an ability to analyze isogenic induced pluripotent stem cell (iPSC) pairs that differ only in a single genetic change, thus allowing a thorough assessment of the molecular and cellular phenotypes that result from monogenetic risk factors. In this review, we summarize the current state of iPSC-based modeling of PD with a focus on leucine-rich repeat kinase 2 (LRRK2), one of the most prominent monogenetic risk factors for PD linked to both familial and idiopathic forms. The LRRK2 protein is a primarily cytosolic multi-domain protein contributing to regulation of several pathways including autophagy, mitochondrial function, vesicle transport, nuclear architecture and cell morphology. We summarize iPSC-based studies that contributed to improving our understanding of the function of LRRK2 and its variants in the context of PD etiopathology. These data, along with results obtained in our own studies, underscore the multifaceted role of LRRK2 in regulating cellular homeostasis on several levels, including proteostasis, mitochondrial dynamics and regulation of the cytoskeleton. Finally, we expound advantages and limitations of reprogramming technologies for disease modeling and drug development and provide an outlook on future challenges and expectations offered by this exciting technology.


Asunto(s)
Células Madre Pluripotentes Inducidas , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina , Mitofagia , Modelos Neurológicos , Enfermedad de Parkinson , Humanos , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/terapia
18.
Biochim Biophys Acta Mol Cell Res ; 1864(12): 2356-2368, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28888991

RESUMEN

Leucine-rich repeat kinase 2 (LRRK2), a multi-domain protein, is a key causative factor in Parkinson's disease (PD). Identification of novel substrates and the molecular mechanisms underlying the effects of LRRK2 are essential for understanding the pathogenesis of PD. In this study, we showed that LRRK2 played an important role in neuronal cell death by directly phosphorylating and activating apoptosis signal-regulating kinase 1 (ASK1). LRRK2 phosphorylated ASK1 at Thr832 that is adjacent to Thr845, which serves as an autophosphorylation site. Moreover, results of binding and kinase assays showed that LRRK2 acted as a scaffolding protein by interacting with each components of the ASK1-MKK3/6-p38 MAPK pathway through its specific domains and increasing the proximity to downstream targets. Furthermore, LRRK2-induced apoptosis was suppressed by ASK1 inhibition in neuronal stem cells derived from patients with PD. These results clearly indicate that LRRK2 acts as an upstream kinase in the ASK1 pathway and plays an important role in the pathogenesis of PD.


Asunto(s)
Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , MAP Quinasa Quinasa Quinasa 5/genética , Neuronas/metabolismo , Enfermedad de Parkinson/genética , Apoptosis/genética , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/metabolismo , MAP Quinasa Quinasa 3/genética , MAP Quinasa Quinasa 3/metabolismo , MAP Quinasa Quinasa Quinasa 5/metabolismo , Neuronas/patología , Enfermedad de Parkinson/patología , Fosforilación , Transducción de Señal/genética , Proteínas Quinasas p38 Activadas por Mitógenos/genética
19.
Circulation ; 131(14): 1278-1290, 2015 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-25739401

RESUMEN

BACKGROUND: Long noncoding RNAs (lncRNAs) have emerged as critical epigenetic regulators with important functions in development and disease. Here, we sought to identify and functionally characterize novel lncRNAs critical for vertebrate development. METHODS AND RESULTS: By relying on human pluripotent stem cell differentiation models, we investigated lncRNAs differentially regulated at key steps during human cardiovascular development with a special focus on vascular endothelial cells. RNA sequencing led to the generation of large data sets that serve as a gene expression roadmap highlighting gene expression changes during human pluripotent cell differentiation. Stage-specific analyses led to the identification of 3 previously uncharacterized lncRNAs, TERMINATOR, ALIEN, and PUNISHER, specifically expressed in undifferentiated pluripotent stem cells, cardiovascular progenitors, and differentiated endothelial cells, respectively. Functional characterization, including localization studies, dynamic expression analyses, epigenetic modification monitoring, and knockdown experiments in lower vertebrates, as well as murine embryos and human cells, confirmed a critical role for each lncRNA specific for each analyzed developmental stage. CONCLUSIONS: We have identified and functionally characterized 3 novel lncRNAs involved in vertebrate and human cardiovascular development, and we provide a comprehensive transcriptomic roadmap that sheds new light on the molecular mechanisms underlying human embryonic development, mesodermal commitment, and cardiovascular specification.


Asunto(s)
Sistema Cardiovascular/crecimiento & desarrollo , Células Endoteliales/química , Regulación del Desarrollo de la Expresión Génica/genética , Miocitos Cardíacos/química , Células Madre Pluripotentes/química , ARN Largo no Codificante/aislamiento & purificación , Vertebrados/genética , Animales , Sistema Cardiovascular/metabolismo , Diferenciación Celular , Linaje de la Célula , Mapeo Cromosómico , Desarrollo Embrionario/genética , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Corazón Fetal/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Datos de Secuencia Molecular , Morfolinos/farmacocinética , Miocitos Cardíacos/citología , ARN Largo no Codificante/fisiología , Análisis de Secuencia de ARN , Transcriptoma , Vertebrados/crecimiento & desarrollo , Pez Cebra/embriología
20.
Stem Cells ; 32(11): 2923-2938, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25175072

RESUMEN

Reprogramming technologies have emerged as a promising approach for future regenerative medicine. Here, we report on the establishment of a novel methodology allowing for the conversion of human fibroblasts into hematopoietic progenitor-like cells with macrophage differentiation potential. SOX2 overexpression in human fibroblasts, a gene found to be upregulated during hematopoietic reconstitution in mice, induced the rapid appearance of CD34+ cells with a concomitant upregulation of mesoderm-related markers. Profiling of cord blood hematopoietic progenitor cell populations identified miR-125b as a factor facilitating commitment of SOX2-generated CD34+ cells to immature hematopoietic-like progenitor cells with grafting potential. Further differentiation toward the monocytic lineage resulted in the appearance of CD14+ cells with functional phagocytic capacity. In vivo transplantation of SOX2/miR-125b-generated CD34+ cells facilitated the maturation of the engrafted cells toward CD45+ cells and ultimately the monocytic/macrophage lineage. Altogether, our results indicate that strategies combining lineage conversion and further lineage specification by in vivo or in vitro approaches could help to circumvent long-standing obstacles for the reprogramming of human cells into hematopoietic cells with clinical potential.


Asunto(s)
Diferenciación Celular/fisiología , Fibroblastos/citología , Monocitos/citología , Células Madre/citología , Animales , Antígenos CD34/metabolismo , Linaje de la Célula/fisiología , Células Cultivadas , Humanos , Antígenos Comunes de Leucocito/metabolismo , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA