Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Cell ; 157(2): 472-485, 2014 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-24725412

RESUMEN

Mutations in leucine-rich repeat kinase 2 (LRRK2) are a common cause of familial and sporadic Parkinson's disease (PD). Elevated LRRK2 kinase activity and neurodegeneration are linked, but the phosphosubstrate that connects LRRK2 kinase activity to neurodegeneration is not known. Here, we show that ribosomal protein s15 is a key pathogenic LRRK2 substrate in Drosophila and human neuron PD models. Phosphodeficient s15 carrying a threonine 136 to alanine substitution rescues dopamine neuron degeneration and age-related locomotor deficits in G2019S LRRK2 transgenic Drosophila and substantially reduces G2019S LRRK2-mediated neurite loss and cell death in human dopamine and cortical neurons. Remarkably, pathogenic LRRK2 stimulates both cap-dependent and cap-independent mRNA translation and induces a bulk increase in protein synthesis in Drosophila, which can be prevented by phosphodeficient T136A s15. These results reveal a novel mechanism of PD pathogenesis linked to elevated LRRK2 kinase activity and aberrant protein synthesis in vivo.


Asunto(s)
Neuronas/metabolismo , Enfermedad de Parkinson/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Ribosómicas/metabolismo , Secuencia de Aminoácidos , Animales , Drosophila melanogaster , Humanos , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina , Datos de Secuencia Molecular , Neuronas/patología , Enfermedad de Parkinson/patología , Proteínas Ribosómicas/química
2.
J Cell Sci ; 127(Pt 3): 641-52, 2014 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-24338367

RESUMEN

Abnormal generation of inhibitory neurons that synthesize γ-aminobutyric acid (GABAergic) is characteristic of neuropsychological disorders. We provide evidence that the extracellular matrix molecule tenascin-R (TNR) - which is predominantly expressed by a subpopulation of interneurons - plays a role in the generation of GABAergic and granule neurons in the murine dentate gyrus by regulating fate determination of neural stem or progenitor cells (NSCs). During development, absence of TNR in constitutively TNR-deficient (TNR(-/-)) mice results in increased numbers of dentate gyrus GABAergic neurons, decreased expression of its receptor ß1 integrin, increased activation of p38 MAPK and increased expression of the GABAergic specification gene Ascl1. Postnatally, increased GABAergic input to adult hippocampal NSCs in TNR(-/-) mice is associated not only with increased numbers of GABAergic and, particularly, parvalbumin-immunoreactive neurons, as seen during development, but also with increased numbers of granule neurons, thus contributing to the increased differentiation of NSCs into granule cells. These findings indicate the importance of TNR in the regulation of hippocampal neurogenesis and suggest that TNR acts through distinct direct and indirect mechanisms during development and in the adult.


Asunto(s)
Proliferación Celular , Giro Dentado/crecimiento & desarrollo , Neurogénesis/genética , Tenascina/genética , Animales , Diferenciación Celular , Embrión de Mamíferos , Desarrollo Embrionario/genética , Matriz Extracelular/genética , Regulación del Desarrollo de la Expresión Génica , Hipocampo/metabolismo , Ratones , Neuronas/metabolismo , Células Madre/metabolismo , Tenascina/metabolismo
3.
Neurobiol Dis ; 56: 104-15, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23639788

RESUMEN

Alzheimer's disease (AD) is a devastating neurodegenerative disorder and the most common cause of elderly dementia. In an effort to contribute to the potential of molecular approaches to reduce degenerative processes we have tested the possibility that the neural adhesion molecule L1 ameliorates some characteristic cellular and molecular parameters associated with the disease in a mouse model of AD. Three-month-old mice overexpressing mutated forms of amyloid precursor protein and presenilin-1 under the control of a neuron-specific promoter received an injection of adeno-associated virus encoding the neuronal isoform of full-length L1 (AAV-L1) or, as negative control, green fluorescent protein (AAV-GFP) into the hippocampus and occipital cortex. Four months after virus injection, the mice were analyzed for histological and biochemical parameters of AD. AAV-L1 injection decreased the Aß plaque load, levels of Aß42, Aß42/40 ratio and astrogliosis compared with AAV-GFP controls. AAV-L1 injected mice also had increased densities of inhibitory synaptic terminals on pyramidal cells in the hippocampus when compared with AAV-GFP controls. Numbers of microglial cells/macrophages were similar in both groups, but numbers of microglial cells/macrophages per plaque were increased in AAV-L1 injected mice. To probe for a molecular mechanism that may underlie these effects, we analyzed whether L1 would directly and specifically interact with Aß. In a label-free binding assay, concentration dependent binding of the extracellular domain of L1, but not of the close homolog of L1 to Aß40 and Aß42 was seen, with the fibronectin type III homologous repeats 1-3 of L1 mediating this effect. Aggregation of Aß42 in vitro was reduced in the presence of the extracellular domain of L1. The combined observations indicate that L1, when overexpressed in neurons and glia, reduces several histopathological hallmarks of AD in mice, possibly by reduction of Aß aggregation. L1 thus appears to be a candidate molecule to ameliorate the pathology of AD, when applied in therapeutically viable treatment schemes.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Molécula L1 de Adhesión de Célula Nerviosa/metabolismo , Molécula L1 de Adhesión de Célula Nerviosa/uso terapéutico , Enfermedad de Alzheimer/patología , Animales , Western Blotting , Encéfalo/patología , Dependovirus/genética , Ensayo de Inmunoadsorción Enzimática , Gliosis/patología , Proteínas Fluorescentes Verdes , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones Transgénicos , Microglía/efectos de los fármacos , Lóbulo Occipital/metabolismo , Lóbulo Occipital/patología , Placa Amiloide/patología , Unión Proteica , Células Piramidales/efectos de los fármacos , Receptores CCR2/metabolismo , Fijación del Tejido
5.
Brain ; 133(Pt 1): 189-204, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19995872

RESUMEN

Parkinson's disease is the second most common neurodegenerative disease, after Alzheimer's disease, and the most common movement disorder. Drug treatment and deep brain stimulation can ameliorate symptoms, but the progressive degeneration of dopaminergic neurons in the substantia nigra eventually leads to severe motor dysfunction. The transplantation of stem cells has emerged as a promising approach to replace lost neurons in order to restore dopamine levels in the striatum and reactivate functional circuits. We have generated substrate-adherent embryonic stem cell-derived neural aggregates overexpressing the neural cell adhesion molecule L1, because it has shown beneficial functions after central nervous system injury. L1 enhances neurite outgrowth and neuronal migration, differentiation and survival as well as myelination. In a previous study, L1 was shown to enhance functional recovery in a mouse model of Huntington's disease. In another study, a new differentiation protocol for murine embryonic stem cells was established allowing the transplantation of stem cell-derived neural aggregates consisting of differentiated neurons and radial glial cells into the lesioned brain. In the present study, this embryonic stem cell line was engineered to overexpress L1 constitutively at all stages of differentiation and used to generate stem cell-derived neural aggregates. These were monitored in their effects on stem cell survival and differentiation, rescue of endogenous dopaminergic neurons and ability to influence functional recovery after transplantation in an animal model of Parkinson's disease. Female C57BL/6J mice (2 months old) were treated with the mitochondrial toxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine intraperitoneally to deplete dopaminergic neurons selectively, followed by unilateral transplantation of stem cell-derived neural aggregates into the striatum. Mice grafted with L1 overexpressing stem cell-derived neural aggregates showed better functional recovery when compared to mice transplanted with wild-type stem cell-derived neural aggregates and vehicle-injected mice. Morphological analysis revealed increased numbers and migration of surviving transplanted cells, as well as increased numbers of dopaminergic neurons, leading to enhanced levels of dopamine in the striatum ipsilateral to the grafted side in L1 overexpressing stem cell-derived neural aggregates, when compared to wild-type stem cell-derived neural aggregates. The striatal levels of gamma-aminobutyric acid were not affected by L1 overexpressing stem cell-derived neural aggregates. Furthermore, L1 overexpressing, but not wild-type stem cell-derived neural aggregates, enhanced survival of endogenous host dopaminergic neurons after transplantation adjacent to the substantia nigra pars compacta. Thus, L1 overexpressing stem cell-derived neural aggregates enhance survival and migration of transplanted cells, differentiation into dopaminergic neurons, survival of endogenous dopaminergic neurons, and functional recovery after syngeneic transplantation in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine mouse model of Parkinson's disease.


Asunto(s)
Células Madre Embrionarias/trasplante , Molécula L1 de Adhesión de Célula Nerviosa/biosíntesis , Neuronas/metabolismo , Trastornos Parkinsonianos/metabolismo , Recuperación de la Función/fisiología , Trasplante de Células Madre , Animales , Agregación Celular/fisiología , Diferenciación Celular/fisiología , Células Cultivadas , Pollos , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Femenino , Ratones , Ratones Endogámicos C57BL , Neuronas/citología , Trastornos Parkinsonianos/patología , Trastornos Parkinsonianos/cirugía , Sustancia Negra/citología , Sustancia Negra/metabolismo , Sustancia Negra/cirugía
6.
J Neurosci ; 29(47): 14752-63, 2009 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-19940170

RESUMEN

The dopaminergic system plays an important role in the etiology of schizophrenia, and most antipsychotic drugs exert their functions by blocking dopamine D(2) receptors (D(2)Rs). Since the signaling strength mediated by D(2)Rs is regulated by internalization and degradation processes, it is crucial to identify molecules that modulate D(2)R localization at the cell surface. Here, we show that the neural cell adhesion molecule (NCAM) promotes D(2)R internalization/desensitization and subsequent degradation via direct interaction with a short peptide in the third intracellular loop of the D(2)R. NCAM deficiency in mice leads to increased numbers of D(2)Rs at the cell surface and augmented D(2)R signaling as a result of impaired D(2)R internalization. Furthermore, NCAM-deficient mice show higher sensitivity to the psychostimulant apomorphine and exaggerated activity of dopamine-related locomotor behavior. These results demonstrate that, in addition to its classical function in cell adhesion, NCAM is involved in regulating the trafficking of the neurotransmitter receptor D(2)R as well as receptor-mediated signaling and behavior, thus implicating NCAM as modulator of the dopaminergic system and a potential pharmacological target for dopamine-related neurological and psychiatric disorders.


Asunto(s)
Encéfalo/metabolismo , Dopamina/metabolismo , Endocitosis/fisiología , Moléculas de Adhesión de Célula Nerviosa/metabolismo , Receptores de Dopamina D2/metabolismo , Transmisión Sináptica/fisiología , Animales , Apomorfina/farmacología , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Encéfalo/efectos de los fármacos , Línea Celular , Células Cultivadas , Agonistas de Dopamina/farmacología , Endocitosis/efectos de los fármacos , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Moléculas de Adhesión de Célula Nerviosa/genética , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Estructura Terciaria de Proteína/fisiología , Receptores de Dopamina D2/química , Receptores de Dopamina D2/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Transmisión Sináptica/efectos de los fármacos
7.
eNeuro ; 6(4)2019.
Artículo en Inglés | MEDLINE | ID: mdl-31413152

RESUMEN

The human cerebral cortex is a complex structure with tightly interconnected excitatory and inhibitory neuronal networks. In order to study human cortical function, we recently developed a method to generate cortical neurons from human induced pluripotent stem cells (hiPSCs) that form both excitatory and inhibitory neuronal networks resembling the composition of the human cortex. These cultures and organoids recapitulate neuronal populations representative of the six cortical layers and a balanced excitatory and inhibitory network that is functional and homeostatically stable. To determine whether hiPSC-derived neurons can integrate and retain physiologic activities in vivo, we labeled hiPSCs with red fluorescent protein (RFP) and introduced hiPSC-derived neural progenitors to rat brains. Efficient neural induction, followed by differentiation resulted in a RFP+ neural population with traits of forebrain identity and a balanced synaptic activity composed of both excitatory neurons and inhibitory interneurons. Ten weeks after transplantation, grafted cells structurally integrated into the rat forebrain. Remarkably, these hiPSC-derived neurons were able to fire, exhibiting both excitatory and inhibitory postsynaptic currents, which culminates in the establishment of neuronal connectivity with the host circuitry. This study demonstrates that neural progenitors derived from hiPSCs can differentiate into functional cortical neurons and can participate in neural network activity through functional synaptic integration in vivo, thereby contributing to information processing.


Asunto(s)
Potenciales Postsinápticos Excitadores , Células Madre Pluripotentes Inducidas/fisiología , Células Madre Pluripotentes Inducidas/trasplante , Potenciales Postsinápticos Inhibidores , Neuronas/fisiología , Prosencéfalo/fisiología , Animales , Animales Recién Nacidos , Línea Celular , Femenino , Humanos , Interneuronas/fisiología , Masculino , Ratas Desnudas
8.
Cell Discov ; 3: 17038, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29387450

RESUMEN

Imprinted genes are vulnerable to environmental influences during early embryonic development, thereby contributing to the onset of disease in adulthood. Monoallelic methylation at several germline imprints has been reported as DNMT1-dependent. However, which of these two epigenetic attributes, DNMT1-dependence or allelic methylation, renders imprinted genes susceptible to environmental stressors has not been determined. Herein, we developed a new approach, referred to as NORED, to identify 2468 DNMT1-dependent DNA methylation patterns in the mouse genome. We further developed an algorithm based on a genetic variation-independent approach (referred to as MethylMosaic) to detect 2487 regions with bimodal methylation patterns. Two approaches identified 207 regions, including known imprinted germline allele-specific methylation patterns (ASMs), that were both NORED and MethylMosaic regions. Examination of methylation in four independent mouse embryonic stem cell lines shows that two regions identified by both NORED and MethylMosaic (Hcn2 and Park7) did not display parent-of-origin-dependent allelic methylation. In these four F1 hybrid cell lines, genetic variation in Cast allele at Hcn2 locus introduces a transcription factor binding site for MTF-1 that may predispose Cast allelic hypomethylation in a reciprocal cross with either C57 or 129 strains. In contrast, each allele of Hcn2 ASM in J1 inbred cell line and Park7 ASM in four F1 hybrid cell lines seems to exhibit similar propensity to be either hypo- or hypermethylated, suggesting a 'random, switchable' ASM. Together with published results, our data on ASMs prompted us to propose a hypothesis of regional 'autosomal chromosome inactivation (ACI)' that may control a subset of autosomal genes. Therefore, our results open a new avenue to understand monoallelic methylation and provide a rich resource of candidate genes to examine in environmental and nutritional exposure models.

9.
Sci Transl Med ; 8(333): 333ra48, 2016 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-27053772

RESUMEN

Translating neuroprotective treatments from discovery in cell and animal models to the clinic has proven challenging. To reduce the gap between basic studies of neurotoxicity and neuroprotection and clinically relevant therapies, we developed a human cortical neuron culture system from human embryonic stem cells or human inducible pluripotent stem cells that generated both excitatory and inhibitory neuronal networks resembling the composition of the human cortex. This methodology used timed administration of retinoic acid to FOXG1(+) neural precursor cells leading to differentiation of neuronal populations representative of the six cortical layers with both excitatory and inhibitory neuronal networks that were functional and homeostatically stable. In human cortical neuronal cultures, excitotoxicity or ischemia due to oxygen and glucose deprivation led to cell death that was dependent on N-methyl-D-aspartate (NMDA) receptors, nitric oxide (NO), and poly(ADP-ribose) polymerase (PARP) (a cell death pathway called parthanatos that is distinct from apoptosis, necroptosis, and other forms of cell death). Neuronal cell death was attenuated by PARP inhibitors that are currently in clinical trials for cancer treatment. This culture system provides a new platform for the study of human cortical neurotoxicity and suggests that PARP inhibitors may be useful for ameliorating excitotoxic and ischemic cell death in human neurons.


Asunto(s)
Corteza Cerebral/citología , Interneuronas/citología , Inhibición Neural/efectos de los fármacos , Neurotoxinas/toxicidad , Muerte Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Línea Celular , Separación Celular , Células Cultivadas , Células Madre Embrionarias/citología , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/metabolismo , Factores de Transcripción Forkhead/metabolismo , Glucosa/deficiencia , Proteínas Hedgehog/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Interneuronas/efectos de los fármacos , Interneuronas/metabolismo , Modelos Biológicos , N-Metilaspartato/farmacología , Red Nerviosa/efectos de los fármacos , Proteínas del Tejido Nervioso/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/metabolismo , Óxido Nítrico/metabolismo , Oxígeno , Transducción de Señal/efectos de los fármacos , Tretinoina/farmacología
10.
Mol Cell Ther ; 2(18)2014 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-25400937

RESUMEN

BACKGROUND: Small non-coding microRNA RNA molecules can regulate stem cell function. The role of microRNAs in neural stem/progenitor cells (NS/PCs) differentiation is not entirely clear. METHODS: MiRNA profiling, loss and gain of function studies coupled with dendritic tree development morphometric analysis and calcium influx imaging were utilized to investigate the role of micoRNA-223 in differentiating NS/PCs. RESULTS: MiRNA profiling in human NS/PCs before and after differentiation in vitro reveals modulation of miRNAs following differentiation of NS/PCs. MiR-223, a microRNA well characterized as a hematopoietic-specific miRNA was identified. Cell-autonomous inhibition of miR-223 in the adult mouse dentate gyrus NS/PCs led to a significant increase in immature neurons soma size, dendritic tree total length, branch number per neuron and complexity, while neuronal migration in the dentate gyrus remained unaffected. Overexpression of miR-223 decreased dendritic tree total length, branch number and complexity in neurons differentiated from human embryonic stem cells (hESCs). Inhibition of miR-223 enhanced N-methyl-D-aspartate (NMDA) induced calcium influx in human neurons differentiated from NS/PCs. CONCLUSIONS: Taken together, these findings indicate that miR-223 regulates the differentiation of neurons derived from NS/PCs.

11.
J Neurotrauma ; 28(9): 1921-37, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21671795

RESUMEN

A major obstacle for the transplantation of neural stem cells (NSCs) into the lesioned spinal cord is their predominant astrocytic differentiation after transplantation. We took advantage of this predominant astrocytic differentiation of NSCs and expressed the paradigmatic beneficial neural cell adhesion molecule L1 in radial glial cells and reactive and nonreactive astrocytes as novel cellular vehicles to express L1 under the control of the promoter for the human glial fibrillary acidic protein (GFAP-L1 NSCs). Behavioral analysis and electrophysiological H-reflex recordings revealed that mice transplanted with GFAP-L1 NSCs showed enhanced locomotor recovery in comparison to mice injected with wild type (WT) NSCs or control mice injected with phosphate-buffered saline (PBS). This functional recovery was further accelerated in mice transplanted with L1-expressing radial glial cells that had been immunoisolated from GFAP-L1 NSCs (GFAP-L1-i cells). Morphological analysis revealed that mice grafted with GFAP-L1 NSCs exhibited increased neuronal differentiation and migration of transplanted cells, as well as increased soma size and cholinergic synaptic coverage of host motoneurons and increased numbers of endogenous catecholaminergic nerve fibers caudal to the lesion site. These findings show that L1-expressing astrocytes and radial glial cells isolated from GFAP-L1 NSC cultures represent a novel strategy for improving functional recovery after spinal cord injury, encouraging the use of the human GFAP promoter to target beneficial transgene expression in transplanted stem cells.


Asunto(s)
Molécula L1 de Adhesión de Célula Nerviosa/metabolismo , Células-Madre Neurales/trasplante , Neuroglía/trasplante , Recuperación de la Función/fisiología , Traumatismos de la Médula Espinal/terapia , Animales , Movimiento Celular , Proliferación Celular , Femenino , Gliosis/metabolismo , Ratones , Actividad Motora/fisiología , Células-Madre Neurales/metabolismo , Neurogénesis/fisiología , Neuroglía/metabolismo , Traumatismos de la Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/fisiopatología
12.
PLoS One ; 6(3): e17126, 2011 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-21445247

RESUMEN

An obstacle to early stem cell transplantation into the acutely injured spinal cord is poor survival of transplanted cells. Transplantation of embryonic stem cells as substrate adherent embryonic stem cell-derived neural aggregates (SENAs) consisting mainly of neurons and radial glial cells has been shown to enhance survival of grafted cells in the injured mouse brain. In the attempt to promote the beneficial function of these SENAs, murine embryonic stem cells constitutively overexpressing the neural cell adhesion molecule L1 which favors axonal growth and survival of grafted and imperiled cells in the inhibitory environment of the adult mammalian central nervous system were differentiated into SENAs and transplanted into the spinal cord three days after compression lesion. Mice transplanted with L1 overexpressing SENAs showed improved locomotor function when compared to mice injected with wild-type SENAs. L1 overexpressing SENAs showed an increased number of surviving cells, enhanced neuronal differentiation and reduced glial differentiation after transplantation when compared to SENAs not engineered to overexpress L1. Furthermore, L1 overexpressing SENAs rescued imperiled host motoneurons and parvalbumin-positive interneurons and increased numbers of catecholaminergic nerve fibers distal to the lesion. In addition to encouraging the use of embryonic stem cells for early therapy after spinal cord injury L1 overexpression in the microenvironment of the lesioned spinal cord is a novel finding in its functions that would make it more attractive for pre-clinical studies in spinal cord regeneration and most likely other diseases of the nervous system.


Asunto(s)
Células Madre Embrionarias/metabolismo , Interneuronas/citología , Molécula L1 de Adhesión de Célula Nerviosa/metabolismo , Traumatismos de la Médula Espinal/rehabilitación , Animales , Astrocitos/citología , Diferenciación Celular , Ratones , Neuroglía/citología , Traumatismos de la Médula Espinal/metabolismo , Trasplante de Células Madre
13.
Neuroreport ; 19(17): 1679-83, 2008 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-18815584

RESUMEN

We had reported that neural stem cells from human fetal striatum (hsNSCs) expressed neural stem cell markers, and were capable of differentiation into neurons, astrocytes, and oligodendrocytes in vitro. To examine multipotency of hsNSCs, some experiments of transgerm layer differentiation in vitro were carried out. Our data indicated that hsNSCs could also generate osteocytes, adipocytes, and hepatocyte-like cells in vitro. Meanwhile, we injected hsNSCs into murine blastocysts at embryonic day 3.5 of gestation. Microinjection of hsNSCs led to the generation of chimeric embryos. Embryos at embryonic day 3.5 of gestation were shown to contribute to the hsNSC-derived cells by PCR-southern blot of 17alphamod, a special method to discover human cells from animals. Analysis of the donor distribution in different tissues showed that donor-derived cells seeded to various tissues. The cellular nature of the human donor cells in chimeric tissues is, however, currently unknown, and further work will be done to identify what differentiated phenotypes have developed from the human cells.


Asunto(s)
Diferenciación Celular , Células Madre Multipotentes/citología , Neuronas/citología , Adipocitos/citología , Animales , Astrocitos/citología , Blastocisto/citología , Blastocisto/metabolismo , Southern Blotting , Células Cultivadas , Cuerpo Estriado , Femenino , Feto , Células Madre Hematopoyéticas/citología , Humanos , Masculino , Ratones , Ratones Endogámicos ICR , Microinyecciones , Células Madre Multipotentes/metabolismo , Células Madre Multipotentes/trasplante , Miogenina/genética , Neuronas/metabolismo , Neuronas/trasplante , Oligodendroglía/citología , Osteocitos/citología , Reacción en Cadena de la Polimerasa , Embarazo , Trasplante de Células Madre/métodos , Trasplante Heterólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA