Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Mol Ther Methods Clin Dev ; 29: 381-394, 2023 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-37251982

RESUMEN

Cell therapy for Parkinson's disease has experienced substantial growth in the past decades with several ongoing clinical trials. Despite increasing refinement of differentiation protocols and standardization of the transplanted neural precursors, the transcriptomic analysis of cells in the transplant after its full maturation in vivo has not been thoroughly investigated. Here, we present spatial transcriptomics analysis of fully differentiated grafts in their host tissue. Unlike earlier transcriptomics analyses using single-cell technologies, we observe that cells derived from human embryonic stem cells (hESCs) in the grafts adopt mature dopaminergic signatures. We show that the presence of phenotypic dopaminergic genes, which were found to be differentially expressed in the transplants, is concentrated toward the edges of the grafts, in agreement with the immunohistochemical analyses. Deconvolution shows dopamine neurons being the dominating cell type in many features beneath the graft area. These findings further support the preferred environmental niche of TH-positive cells and confirm their dopaminergic phenotype through the presence of multiple dopaminergic markers.

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.
J Parkinsons Dis ; 12(6): 1881-1896, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35466951

RESUMEN

BACKGROUND: First-in-human studies to test the efficacy and safety of human embryonic stem cells (hESC)-derived dopaminergic cells in the treatment of Parkinson's disease (PD) are imminent. Pre-clinical studies using hESC-derived dopamine neuron transplants in rat models have indicated that the benefits parallel those shown with fetal tissue but have thus far failed to consider how ongoing L-DOPA administration might impact on the graft. OBJECTIVE: To determine whether L-DOPA impacts on survival and functional recovery following grafting of hESC-derived dopaminergic neurons. METHODS: Unilateral 6-OHDA lesioned rats were administered with either saline or L-DOPA prior to, and for 18 weeks following surgical implantation of dopaminergic neural progenitors derived from RC17 hESCs according to two distinct protocols in independent laboratories. RESULTS: Grafts from both protocols elicited reduction in amphetamine-induced rotations. Reduced L-DOPA-induced dyskinesia preceded the improvement in amphetamine-induced rotations. Furthermore, L-DOPA had no effect on overall survival (HuNu) or dopaminergic neuron content of the graft (TH positive cells) but did lead to an increase in the number of GIRK2 positive neurons. CONCLUSION: Critically, we found that L-DOPA was not detrimental to graft function, potentially enhancing graft maturation and promoting an A9 phenotype. Early improvement of L-DOPA-induced dyskinesia suggests that grafts may support the handling of exogenously supplied dopamine earlier than improvements in amphetamine-induced behaviours indicate. Given that one of the protocols will be employed in the production of cells for the European STEM-PD clinical trial, this is vital information for the management of patients and achieving optimal outcomes following transplantation of hESC-derived grafts for PD.


Asunto(s)
Discinesia Inducida por Medicamentos , Células Madre Embrionarias Humanas , Enfermedad de Parkinson , Anfetaminas/uso terapéutico , Animales , Antiparkinsonianos/uso terapéutico , Modelos Animales de Enfermedad , Dopamina , Discinesia Inducida por Medicamentos/tratamiento farmacológico , Humanos , Levodopa/uso terapéutico , Oxidopamina/uso terapéutico , Oxidopamina/toxicidad , Enfermedad de Parkinson/tratamiento farmacológico , Ratas , Ratas Sprague-Dawley
4.
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.
iScience ; 24(6): 102559, 2021 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-34142058

RESUMEN

Rostrocaudal patterning of the neural tube is a defining event in vertebrate brain development. This process is driven by morphogen gradients which specify the fate of neural progenitor cells, leading to the partitioning of the tube. Although this is extensively studied experimentally, an integrated view of the genetic circuitry is lacking. Here, we present a minimal gene regulatory model for rostrocaudal patterning, whose tristable topology was determined in a data-driven way. Using this model, we identified the repression of hindbrain fate as promising strategy for the improvement of current protocols for the generation of dopaminergic neurons. Furthermore, we combined our model with an established minimal model for dorsoventral patterning on a realistic 3D neural tube and found that key features of neural tube patterning could be recapitulated. Doing so, we demonstrate how data and models from different sources can be combined to simulate complex in vivo processes.

6.
Stem Cell Reports ; 15(4): 869-882, 2020 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-32976765

RESUMEN

Human glial progenitor cells (hGPCs) are promising cellular substrates to explore for the in situ production of new neurons for brain repair. Proof of concept for direct neuronal reprogramming of glial progenitors has been obtained in mouse models in vivo, but conversion using human cells has not yet been demonstrated. Such studies have been difficult to perform since hGPCs are born late during human fetal development, with limited accessibility for in vitro culture. In this study, we show proof of concept of hGPC conversion using fetal cells and also establish a renewable and reproducible stem cell-based hGPC system for direct neural conversion in vitro. Using this system, we have identified optimal combinations of fate determinants for the efficient dopaminergic (DA) conversion of hGPCs, thereby yielding a therapeutically relevant cell type that selectively degenerates in Parkinson's disease. The induced DA neurons show a progressive, subtype-specific phenotypic maturation and acquire functional electrophysiological properties indicative of DA phenotype.


Asunto(s)
Reprogramación Celular , Neuronas Dopaminérgicas/citología , Células Madre Fetales/citología , Mesencéfalo/citología , Células-Madre Neurales/citología , Neuroglía/citología , Neuronas Dopaminérgicas/metabolismo , Células Madre Fetales/metabolismo , Factor Nuclear 3-beta del Hepatocito/metabolismo , Humanos , Modelos Biológicos , Células-Madre Neurales/metabolismo , Neuroglía/metabolismo , Neuronas/citología , Neuronas/metabolismo , Tirosina 3-Monooxigenasa/metabolismo
8.
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
9.
Heliyon ; 6(1): e03067, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31909251

RESUMEN

Human embryonic stem cells (hESCs) and induced pluripotent stem cells (iPSCs) can be differentiated into many different cell types of the central nervous system. One challenge when using pluripotent stem cells is to develop robust and efficient differentiation protocols that result in homogenous cultures of the desired cell type. Here, we have utilized the SMAD-inhibitors SB431542 and Noggin in a fully defined monolayer culture model to differentiate human pluripotent cells into homogenous forebrain neural progenitors. Temporal fate analysis revealed that this protocol results in forebrain-patterned neural progenitor cells that start to express early neuronal markers after two weeks of differentiation, allowing for the analysis of gene expression changes during neurogenesis. Using this system, we were able to identify many previously uncharacterized long intergenic non-coding RNAs that display dynamic expression during human forebrain neurogenesis.

10.
Proc Natl Acad Sci U S A ; 116(52): 27053-27062, 2019 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-31818949

RESUMEN

Adeno-associated virus (AAV) capsid modification enables the generation of recombinant vectors with tailored properties and tropism. Most approaches to date depend on random screening, enrichment, and serendipity. The approach explored here, called BRAVE (barcoded rational AAV vector evolution), enables efficient selection of engineered capsid structures on a large scale using only a single screening round in vivo. The approach stands in contrast to previous methods that require multiple generations of enrichment. With the BRAVE approach, each virus particle displays a peptide, derived from a protein, of known function on the AAV capsid surface, and a unique molecular barcode in the packaged genome. The sequencing of RNA-expressed barcodes from a single-generation in vivo screen allows the mapping of putative binding sequences from hundreds of proteins simultaneously. Using the BRAVE approach and hidden Markov model-based clustering, we present 25 synthetic capsid variants with refined properties, such as retrograde axonal transport in specific subtypes of neurons, as shown for both rodent and human dopaminergic neurons.

11.
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
12.
Nat Commun ; 10(1): 581, 2019 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-30718509

RESUMEN

Midbrain dopamine (mDA) neurons constitute a heterogenous group of cells that have been intensely studied, not least because their degeneration causes major symptoms in Parkinson's disease. Understanding the diversity of mDA neurons - previously well characterized anatomically - requires a systematic molecular classification at the genome-wide gene expression level. Here, we use single cell RNA sequencing of isolated mouse neurons expressing the transcription factor Pitx3, a marker for mDA neurons. Analyses include cells isolated during development up until adulthood and the results are validated by histological characterization of newly identified markers. This identifies seven neuron subgroups divided in two major branches of developing Pitx3-expressing neurons. Five of them express dopaminergic markers, while two express glutamatergic and GABAergic markers, respectively. Analysis also indicate evolutionary conservation of diversity in humans. This comprehensive molecular characterization will provide a valuable resource for elucidating mDA neuron subgroup development and function in the mammalian brain.


Asunto(s)
Encéfalo/citología , Neuronas Dopaminérgicas/metabolismo , Análisis de Secuencia de ARN/métodos , Animales , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Ratones , Factores de Transcripción/metabolismo
13.
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
14.
Nat Protoc ; 12(9): 1962-1979, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28858290

RESUMEN

Generation of precisely patterned neural cells from human pluripotent stem cells (hPSCs) is instrumental in developing disease models and stem cell therapies. Here, we provide a detailed 16-d protocol for obtaining high-purity ventral midbrain (VM) dopamine (DA) progenitors for intracerebral transplantation into animal models and for in vitro maturation into neurons. We have successfully transplanted such cells into the rat; however, in principle, the cells can be used for transplantation into any animal model, and the protocol is designed to also be compatible with clinical transplantation into humans. We show how to precisely set the balance of patterning factors to obtain specifically the caudal VM progenitors that give rise to DA-rich grafts. By specifying how to perform quality control (QC), troubleshooting and adaptation of the procedure, this protocol will facilitate implementation in different laboratories and with a variety of hPSC lines. To facilitate reproducibility of experiments and enable shipping of cells between centers, we present a method for cryopreservation of the progenitors for subsequent direct transplantation or terminal differentiation into DA neurons. This protocol is free of xeno-derived products and can be performed under good manufacturing practice (GMP) conditions.


Asunto(s)
Neuronas Dopaminérgicas , Células Madre Embrionarias/citología , Mesencéfalo/cirugía , Trasplante de Células Madre , Animales , Diferenciación Celular , Ingeniería Celular , Línea Celular , Neuronas Dopaminérgicas/química , Neuronas Dopaminérgicas/citología , Neuronas Dopaminérgicas/trasplante , Perfilación de la Expresión Génica , Humanos , Ratas
15.
Cell Stem Cell ; 20(1): 29-40, 2017 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-28094018

RESUMEN

Stem cell engineering and grafting of mesencephalic dopamine (mesDA) neurons is a promising strategy for brain repair in Parkinson's disease (PD). Refinement of differentiation protocols to optimize this approach will require deeper understanding of mesDA neuron development. Here, we studied this process using transcriptome-wide single-cell RNA sequencing of mouse neural progenitors expressing the mesDA neuron determinant Lmx1a. This approach resolved the differentiation of mesDA and neighboring neuronal lineages and revealed a remarkably close relationship between developing mesDA and subthalamic nucleus (STN) neurons, while also highlighting a distinct transcription factor set that can distinguish between them. While previous hESC mesDA differentiation protocols have relied on markers that are shared between the two lineages, we found that application of these highlighted markers can help to refine current stem cell engineering protocols, increasing the proportion of appropriately patterned mesDA progenitors. Our results, therefore, have important implications for cell replacement therapy in PD.


Asunto(s)
Diferenciación Celular , Linaje de la Célula , Neuronas Dopaminérgicas/citología , Análisis de la Célula Individual/métodos , Núcleo Subtalámico/citología , Biomarcadores/metabolismo , Tipificación del Cuerpo/genética , Diferenciación Celular/genética , Linaje de la Célula/genética , Neuronas Dopaminérgicas/metabolismo , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Humanos , Inmunohistoquímica , Proteínas con Homeodominio LIM/metabolismo , Neurogénesis/genética , Análisis de Secuencia de ARN , Transducción de Señal/genética , Factores de Transcripción/metabolismo
16.
Cell Stem Cell ; 20(1): 135-148, 2017 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-28094017

RESUMEN

Stem cell treatments for neurodegenerative diseases are expected to reach clinical trials soon. Most of the approaches currently under development involve transplantation of immature progenitors that subsequently undergo phenotypic and functional maturation in vivo, and predicting the long-term graft outcome already at the progenitor stage remains a challenge. Here, we took an unbiased approach to identify predictive markers expressed in dopamine neuron progenitors that correlate with graft outcome in an animal model of Parkinson's disease through gene expression analysis of >30 batches of grafted human embryonic stem cell (hESC)-derived progenitors. We found that many of the commonly used markers did not accurately predict in vivo subtype-specific maturation. Instead, we identified a specific set of markers associated with the caudal midbrain that correlate with high dopaminergic yield after transplantation in vivo. Using these markers, we developed a good manufacturing practice (GMP) differentiation protocol for highly efficient and reproducible production of transplantable dopamine progenitors from hESCs.


Asunto(s)
Biomarcadores/metabolismo , Diferenciación Celular , Células Madre Embrionarias Humanas/citología , Células Madre Embrionarias Humanas/trasplante , Enfermedad de Parkinson/terapia , Trasplante de Células Madre , Investigación Biomédica Traslacional , Animales , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula/efectos de los fármacos , Células Cultivadas , Dopamina/metabolismo , Neuronas Dopaminérgicas/citología , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Femenino , Factor 8 de Crecimiento de Fibroblastos/metabolismo , Células Madre Embrionarias Humanas/efectos de los fármacos , Humanos , Laminina/farmacología , Mesencéfalo/metabolismo , Ratas Sprague-Dawley , Reproducibilidad de los Resultados , Análisis de Secuencia de ARN , Núcleo Subtalámico/citología , Núcleo Subtalámico/metabolismo , Factores de Tiempo , Resultado del Tratamiento
17.
Oncotarget ; 7(36): 58203-58217, 2016 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-27533460

RESUMEN

Bmi1 was originally identified as a gene that contributes to the development of mouse lymphoma by inhibiting MYC-induced apoptosis through repression of Ink4a and Arf. It codes for the Polycomb group protein BMI-1 and acts primarily as a transcriptional repressor via chromatin modifications. Although it binds to a large number of genomic regions, the direct BMI-1 target genes described so far do not explain the full spectrum of BMI-1-mediated effects. Here we identify the putative tumor suppressor gene EphA7 as a novel direct BMI-1 target in neural cells and lymphocytes. EphA7 silencing has been reported in several different human tumor types including lymphomas, and our data suggest BMI1 overexpression as a novel mechanism leading to EphA7 inactivation via H3K27 trimethylation and DNA methylation.


Asunto(s)
Regulación de la Expresión Génica , Genes Supresores de Tumor , Complejo Represivo Polycomb 1/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Receptor EphA7/genética , Animales , Linfocitos B , Técnicas de Cultivo de Célula/métodos , Núcleo Celular/metabolismo , Proliferación Celular/fisiología , Células Cultivadas , Cerebelo/anatomía & histología , Cerebelo/metabolismo , Metilación de ADN/fisiología , Regulación hacia Abajo , Histonas/metabolismo , Inmunohistoquímica , Antígeno Ki-67/metabolismo , Ventrículos Laterales/anatomía & histología , Ventrículos Laterales/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis por Micromatrices , Células-Madre Neurales , Complejo Represivo Polycomb 1/genética , Proteínas Proto-Oncogénicas/genética , Receptor EphA7/metabolismo , Bazo/citología , Transducción Genética , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...