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










Base de dados
Intervalo de ano de publicação
1.
Mol Ther Methods Clin Dev ; 29: 381-394, 2023 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-37251982

RESUMO

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.
Artigo em Inglês | MEDLINE | ID: mdl-35650213

RESUMO

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.


Assuntos
Doença de Parkinson , Células-Tronco Pluripotentes , Animais , Diferenciação Celular , Neurônios Dopaminérgicos , Humanos , Mesencéfalo , Doença de Parkinson/terapia , Ratos , Tretinoína/farmacologia
3.
J Parkinsons Dis ; 12(6): 1881-1896, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35466951

RESUMO

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.


Assuntos
Discinesia Induzida por Medicamentos , Células-Tronco Embrionárias Humanas , Doença de Parkinson , Anfetaminas/uso terapêutico , Animais , Antiparkinsonianos/uso terapêutico , Modelos Animais de Doenças , Dopamina , Discinesia Induzida por Medicamentos/tratamento farmacológico , Humanos , Levodopa/uso terapêutico , Oxidopamina/uso terapêutico , Oxidopamina/toxicidade , Doença de Parkinson/tratamento farmacológico , Ratos , Ratos Sprague-Dawley
4.
Stem Cell Reports ; 17(1): 159-172, 2022 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-34971563

RESUMO

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.


Assuntos
Diferenciação Celular , Neurônios Dopaminérgicos/citologia , Neurônios Dopaminérgicos/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Células-Tronco Embrionárias Humanas/citologia , Células-Tronco Embrionárias Humanas/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Sinapses/metabolismo , Biomarcadores , Rastreamento de Células , Expressão Gênica , Genes Reporter , Fatores de Troca do Nucleotídeo Guanina/genética , Humanos , Mesencéfalo/metabolismo , Fenótipo , Proteínas Serina-Treonina Quinases/genética , Transplante de Células-Tronco
5.
iScience ; 24(6): 102559, 2021 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-34142058

RESUMO

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.
Artigo em Inglês | MEDLINE | ID: mdl-32976765

RESUMO

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.


Assuntos
Reprogramação Celular , Neurônios Dopaminérgicos/citologia , Células-Tronco Fetais/citologia , Mesencéfalo/citologia , Células-Tronco Neurais/citologia , Neuroglia/citologia , Neurônios Dopaminérgicos/metabolismo , Células-Tronco Fetais/metabolismo , Fator 3-beta Nuclear de Hepatócito/metabolismo , Humanos , Modelos Biológicos , Células-Tronco Neurais/metabolismo , Neuroglia/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo
8.
Nat Commun ; 11(1): 2434, 2020 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-32415072

RESUMO

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.


Assuntos
Neurônios Dopaminérgicos/citologia , Doença de Parkinson/terapia , Transplante de Células-Tronco , Células-Tronco/citologia , Animais , Encéfalo/metabolismo , Diferenciação Celular , Corpo Estriado , Modelos Animais de Doenças , Dopamina/metabolismo , Células-Tronco Embrionárias/citologia , Feminino , Sobrevivência de Enxerto , Humanos , Família Multigênica , RNA-Seq , Ratos , Ratos Nus , Regeneração , Análise de Célula Única , Transcriptoma
9.
Heliyon ; 6(1): e03067, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31909251

RESUMO

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.
Artigo em Inglês | MEDLINE | ID: mdl-31818949

RESUMO

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.
Artigo em Inglês | MEDLINE | ID: mdl-31553914

RESUMO

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.


Assuntos
Gânglios da Base/fisiopatologia , Células-Tronco Embrionárias Humanas/metabolismo , Doença de Parkinson/genética , Animais , Modelos Animais de Doenças , Humanos , Camundongos Nus , Ratos
12.
Nat Commun ; 10(1): 581, 2019 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-30718509

RESUMO

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.


Assuntos
Encéfalo/citologia , Neurônios Dopaminérgicos/metabolismo , Análise de Sequência de RNA/métodos , Animais , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/metabolismo , Camundongos , Fatores de Transcrição/metabolismo
13.
J Comp Neurol ; 526(13): 2133-2146, 2018 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-30007046

RESUMO

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.


Assuntos
Neurônios Dopaminérgicos/fisiologia , Neurônios Dopaminérgicos/transplante , Mesencéfalo/cirurgia , Vias Neurais/fisiologia , Células-Tronco Neurais/fisiologia , Células-Tronco Neurais/transplante , Transtornos Parkinsonianos/cirurgia , Prosencéfalo/fisiologia , Sinapses/fisiologia , Anfetamina/farmacologia , Animais , Inibidores da Captação de Dopamina/farmacologia , Feminino , Humanos , Hidroxidopaminas , Camundongos , Fibras Nervosas/fisiologia , Transtornos Parkinsonianos/induzido quimicamente , Ratos Nus , Transplante de Células-Tronco , Comportamento Estereotipado/efeitos dos fármacos
14.
Nat Protoc ; 12(9): 1962-1979, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28858290

RESUMO

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.


Assuntos
Neurônios Dopaminérgicos , Células-Tronco Embrionárias/citologia , Mesencéfalo/cirurgia , Transplante de Células-Tronco , Animais , Diferenciação Celular , Engenharia Celular , Linhagem Celular , Neurônios Dopaminérgicos/química , Neurônios Dopaminérgicos/citologia , Neurônios Dopaminérgicos/transplante , Perfilação da Expressão Gênica , Humanos , Ratos
15.
Cell Stem Cell ; 20(1): 29-40, 2017 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-28094018

RESUMO

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.


Assuntos
Diferenciação Celular , Linhagem da Célula , Neurônios Dopaminérgicos/citologia , Análise de Célula Única/métodos , Núcleo Subtalâmico/citologia , Biomarcadores/metabolismo , Padronização Corporal/genética , Diferenciação Celular/genética , Linhagem da Célula/genética , Neurônios Dopaminérgicos/metabolismo , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Perfilação da Expressão Gênica , Redes Reguladoras de Genes , Humanos , Imuno-Histoquímica , Proteínas com Homeodomínio LIM/metabolismo , Neurogênese/genética , Análise de Sequência de RNA , Transdução de Sinais/genética , Fatores de Transcrição/metabolismo
16.
Cell Stem Cell ; 20(1): 135-148, 2017 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-28094017

RESUMO

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.


Assuntos
Biomarcadores/metabolismo , Diferenciação Celular , Células-Tronco Embrionárias Humanas/citologia , Células-Tronco Embrionárias Humanas/transplante , Doença de Parkinson/terapia , Transplante de Células-Tronco , Pesquisa Translacional Biomédica , Animais , Diferenciação Celular/efeitos dos fármacos , Linhagem da Célula/efeitos dos fármacos , Células Cultivadas , Dopamina/metabolismo , Neurônios Dopaminérgicos/citologia , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/metabolismo , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/metabolismo , Feminino , Fator 8 de Crescimento de Fibroblasto/metabolismo , Células-Tronco Embrionárias Humanas/efeitos dos fármacos , Humanos , Laminina/farmacologia , Mesencéfalo/metabolismo , Ratos Sprague-Dawley , Reprodutibilidade dos Testes , Análise de Sequência de RNA , Núcleo Subtalâmico/citologia , Núcleo Subtalâmico/metabolismo , Fatores de Tempo , Resultado do Tratamento
17.
Oncotarget ; 7(36): 58203-58217, 2016 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-27533460

RESUMO

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.


Assuntos
Regulação da Expressão Gênica , Genes Supressores de Tumor , Complexo Repressor Polycomb 1/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Receptor EphA7/genética , Animais , Linfócitos B , Técnicas de Cultura de Células/métodos , Núcleo Celular/metabolismo , Proliferação de Células/fisiologia , Células Cultivadas , Cerebelo/anatomia & histologia , Cerebelo/metabolismo , Metilação de DNA/fisiologia , Regulação para Baixo , Histonas/metabolismo , Imuno-Histoquímica , Antígeno Ki-67/metabolismo , Ventrículos Laterais/anatomia & histologia , Ventrículos Laterais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Análise em Microsséries , Células-Tronco Neurais , Complexo Repressor Polycomb 1/genética , Proteínas Proto-Oncogênicas/genética , Receptor EphA7/metabolismo , Baço/citologia , Transdução Genética , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...