Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
PLoS Biol ; 21(12): e3002419, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38048364

RESUMO

Circadian regulation of gene expression is prevalent and plays critical roles in cell differentiation. However, its roles in the reprogramming of differentiated cells remain largely unknown. Here, we found that one of the master circadian regulators PER1 promoted virus-mediated reprogramming of mouse embryonic fibroblasts (MEFs) to induced neurons (iNs) and induced pluripotent stem cells (iPSCs). Unexpectedly, PER1 achieved this by repressing inflammatory activation of contaminating macrophages in the MEF culture, rather than by directly modulating the reprogrammability of MEFs. More specifically, we found that transduced viruses activated inflammatory genes in macrophages, such as Tnf encoding TNFα, one of the central inflammatory regulators and an autocrine activator of macrophages. TNFα inhibited iN reprogramming, whereas a TNFα inhibitor promoted iN reprogramming, connecting the inflammatory responses to iN reprogramming. In addition, macrophages were induced to proliferate and mature by non-macrophage cells serving as feeders, which also supported up-regulation of TNFα in macrophages without virus transduction. Furthermore, the 2 inflammatory responses were repressed by the circadian regulator PER1 in macrophages, making reprogrammability dependent on time-of-day of virus transduction. Similar results were obtained with iPSC reprogramming, suggesting a wide occurrence of macrophage-mediated inhibition of cell reprogramming. This study uncovers mechanistic links between cell reprogramming, bystander inflammatory macrophages, and circadian rhythms, which are particularly relevant to in vivo reprogramming and organoid formation incorporating immune cells.


Assuntos
Células-Tronco Pluripotentes Induzidas , Fator de Necrose Tumoral alfa , Animais , Camundongos , Diferenciação Celular , Reprogramação Celular , Fibroblastos/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
2.
Stem Cells ; 29(9): 1349-61, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21732495

RESUMO

Induced pluripotent stem cells (iPSCs) can be created by reprogramming differentiated cells through introduction of defined genes, most commonly Oct4, Sox2, Klf4, and c-Myc (OSKM). However, this process is slow and extremely inefficient. Here, we demonstrate radical acceleration of iPSC creation with a fusion gene between Oct4 and the powerful transactivation domain (TAD) of MyoD (M(3)O). Transduction of M(3) O as well as Sox2, Klf4, and c-Myc into fibroblasts effectively remodeled patterns of DNA methylation, chromatin accessibility, histone modifications, and protein binding at pluripotency genes, raising the efficiency of making mouse and human iPSCs more than 50-fold in comparison to OSKM. These results identified that one of the most critical barriers to iPSC creation is poor chromatin accessibility and protein recruitment to pluripotency genes. The MyoD TAD has a capability of overcoming this problem. Our approach of fusing TADs to unrelated transcription factors has far-reaching implications as a powerful tool for transcriptional reprogramming beyond application to iPSC technology.


Assuntos
Reprogramação Celular , Montagem e Desmontagem da Cromatina , Células-Tronco Pluripotentes Induzidas/fisiologia , Proteína MyoD/genética , Animais , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Fator 4 Semelhante a Kruppel , Camundongos , Fator 3 de Transcrição de Octâmero/genética , Estrutura Terciária de Proteína , Ativação Transcricional
3.
Nat Cell Biol ; 5(3): 205-10, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12589397

RESUMO

Egg cytoplasm has the capability to reprogramme differentiated somatic nuclei, as shown by nuclear transplantation in animal cloning. The nucleoli of donor nuclei are rapidly disassembled on injection into interphase eggs and are correctly reassembled when donor transcription initiates in the early embryos of frogs and mammals, recapitulating the physiological nucleolar dynamics of early embryogenesis. This is one of the most remarkable structural reorganizations of somatic nuclei in nuclear cloning. Despite the long history of nuclear cloning, almost nothing is known about the molecular mechanism of nucleolar disassembly in egg cytoplasm. Here we show that the Xenopus germ cell proteins FRGY2a and FRGY2b reversibly disassemble somatic nucleoli in egg cytoplasm, independently of continuing ribosomal RNA transcription. The carboxy-terminal domain of FRGY2a, which localizes to the nucleoli, is sufficient for nucleolar disassembly in transfected cells. Our results show that a single protein fragment can trigger reversible disassembly of the complex nucleolar structure.


Assuntos
Nucléolo Celular/ultraestrutura , Células Germinativas/metabolismo , Isoformas de Proteínas/fisiologia , Proteínas de Ligação a RNA/fisiologia , Fatores de Transcrição/fisiologia , Proteínas de Xenopus/fisiologia , Animais , Nucléolo Celular/metabolismo , Microscopia Eletrônica , Microscopia de Fluorescência , Dados de Sequência Molecular , Isoformas de Proteínas/metabolismo , Proteínas de Ligação a RNA/metabolismo , Proteínas Recombinantes/metabolismo , Fatores de Transcrição/metabolismo , Xenopus , Proteínas de Xenopus/metabolismo
4.
J Cell Biol ; 220(7)2021 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-34009269

RESUMO

Circadian rhythms regulate cell proliferation and differentiation, but circadian control of tissue regeneration remains elusive at the molecular level. Here, we show that proper myoblast differentiation and muscle regeneration are regulated by the circadian master regulators Per1 and Per2. Depletion of Per1 or Per2 suppressed myoblast differentiation in vitro and muscle regeneration in vivo, demonstrating their nonredundant functions. Both Per1 and Per2 were required for the activation of Igf2, an autocrine promoter of myoblast differentiation, accompanied by Per-dependent recruitment of RNA polymerase II, dynamic histone modifications at the Igf2 promoter and enhancer, and the promoter-enhancer interaction. This circadian epigenetic priming created a preferred time window for initiating myoblast differentiation. Consistently, muscle regeneration was faster if initiated at night, when Per1, Per2, and Igf2 were highly expressed compared with morning. This study reveals the circadian timing as a significant factor for effective muscle cell differentiation and regeneration.


Assuntos
Ritmo Circadiano/genética , Fator de Crescimento Insulin-Like II/genética , Proteínas Circadianas Period/genética , Regeneração/genética , Animais , Diferenciação Celular/genética , Proliferação de Células/genética , Elementos Facilitadores Genéticos/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Humanos , Camundongos , Camundongos Knockout , Músculo Esquelético/crescimento & desenvolvimento , Mioblastos/metabolismo , Regiões Promotoras Genéticas/genética , RNA Mensageiro/genética
5.
J Biol Chem ; 284(39): 26685-94, 2009 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-19648109

RESUMO

Nucleostemin (NS) is a nucleolar protein involved in the regulation of cell proliferation. Both overexpression and knockdown of NS increase the activity of the tumor suppressor protein p53, resulting in cell cycle arrest. In addition, NS regulates processing of pre-rRNA and consequently the level of total protein synthesis. Here, we describe a previously uncharacterized function of NS in the maintenance of the tripartite nucleolar structure as well as the integrity of small nucleolar ribonucleoproteins (snoRNPs). NS is also necessary to maintain the telomerase complex which shares common protein subunits with the H/ACA box snoRNPs. First, immunofluorescence microscopy and electron microscopy demonstrated that knockdown of NS disorganized the nucleolar architecture, in particular, the dense fibrillar component where snoRNPs are localized. Second, gel filtration chromatography and immunoprecipitation indicated that NS depletion leads to dissociation of the components of snoRNPs and the telomerase complex. Third, NS depletion reduced both telomerase activity and the cellular level of pseudouridine, an H/ACA snoRNP-mediated modification of rRNA and other RNAs that are important for their folding and stability. These morphological, biochemical and functional studies demonstrate that NS plays an important role to maintain nucleolar structure and function on a more fundamental level than previously thought.


Assuntos
Proteínas de Transporte/fisiologia , Nucléolo Celular/metabolismo , Proteínas Nucleares/fisiologia , Ribonucleoproteínas Nucleolares Pequenas/metabolismo , Telomerase/metabolismo , Western Blotting , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Nucléolo Celular/ultraestrutura , Cromatografia em Camada Fina , Proteínas de Ligação ao GTP , Células HeLa , Humanos , Imunoprecipitação , Microscopia Eletrônica , Microscopia de Fluorescência , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Pseudouridina/metabolismo , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa
6.
Mol Cell Biol ; 26(4): 1259-71, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16449640

RESUMO

Somatic cell nuclear cloning has repeatedly demonstrated striking reversibility of epigenetic regulation of cell differentiation. Upon injection into eggs, the donor nuclei exhibit global chromatin decondensation, which might contribute to reprogramming the nuclei by derepressing dormant genes. Decondensation of sperm chromatin in eggs is explained by the replacement of sperm-specific histone variants with egg-type histones by the egg protein nucleoplasmin (Npm). However, little is known about the mechanisms of chromatin decondensation in somatic nuclei that do not contain condensation-specific histone variants. Here we found that Npm could widely decondense chromatin in undifferentiated mouse cells without overt histone exchanges but with specific epigenetic modifications that are relevant to open chromatin structure. These modifications included nucleus-wide multiple histone H3 phosphorylation, acetylation of Lys 14 in histone H3, and release of heterochromatin proteins HP1beta and TIF1beta from the nuclei. The protein kinase inhibitor staurosporine inhibited chromatin decondensation and these epigenetic modifications with the exception of H3 acetylation, potentially linking these chromatin events. At the functional level, Npm pretreatment of mouse nuclei facilitated activation of four oocyte-specific genes from the nuclei injected into Xenopus laevis oocytes. Future molecular elucidation of chromatin decondensation by Npm will significantly contribute to our understanding of the plasticity of cell differentiation.


Assuntos
Cromatina/metabolismo , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Proteínas de Xenopus/metabolismo , Animais , Sequência de Bases , Diferenciação Celular/genética , Núcleo Celular/metabolismo , Cromatina/genética , Montagem e Desmontagem da Cromatina , Clonagem de Organismos , DNA/genética , Epigênese Genética , Feminino , Heterocromatina/genética , Heterocromatina/metabolismo , Histonas/metabolismo , Técnicas In Vitro , Masculino , Camundongos , Modelos Biológicos , Técnicas de Transferência Nuclear , Nucleoplasminas , Oócitos/metabolismo , Espermatozoides/metabolismo , Transplante Heterólogo , Xenopus laevis
7.
Cell Rep ; 25(9): 2537-2551.e8, 2018 11 27.
Artigo em Inglês | MEDLINE | ID: mdl-30485818

RESUMO

Fkbp5 is a widely expressed peptidyl prolyl isomerase that serves as a molecular chaperone through conformational changes of binding partners. Although it regulates diverse protein functions, little is known about its roles in myogenesis. We found here that Fkbp5 plays critical roles in myoblast differentiation through two mechanisms. First, it sequesters Cdk4 within the Hsp90 storage complex and prevents the formation of the cyclin D1-Cdk4 complex, which is a major inhibitor of differentiation. Second, Fkbp5 promotes cis-trans isomerization of the Thr172-Pro173 peptide bond in Cdk4 and inhibits phosphorylation of Thr172, an essential step for Cdk4 activation. Consistent with these in vitro findings, muscle regeneration is delayed in Fkbp5-/- mice. The related protein Fkbp4 also sequesters Cdk4 within the Hsp90 complex but does not isomerize Cdk4 or induce Thr173 phosphorylation despite its highly similar sequence. This study demonstrates protein isomerization as a critical regulatory mechanism of myogenesis by targeting Cdk4.


Assuntos
Diferenciação Celular , Quinase 4 Dependente de Ciclina/metabolismo , Mioblastos/citologia , Mioblastos/metabolismo , Proteínas de Ligação a Tacrolimo/metabolismo , Animais , Linhagem Celular , Proliferação de Células , Proteínas de Choque Térmico HSP90/metabolismo , Isomerismo , Masculino , Camundongos Knockout , Músculos/fisiologia , Peptídeos/metabolismo , Prolina/metabolismo , Ligação Proteica , Regeneração , Proteínas de Ligação a Tacrolimo/deficiência
8.
Cell Rep ; 22(8): 2118-2132, 2018 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-29466738

RESUMO

Circadian rhythms regulate cell proliferation and differentiation; however, little is known about their roles in myogenic differentiation. Our synchronized differentiation studies demonstrate that myoblast proliferation and subsequent myotube formation by cell fusion occur in circadian manners. We found that one of the core regulators of circadian rhythms, Cry2, but not Cry1, is critical for the circadian patterns of these two critical steps in myogenic differentiation. This is achieved through the specific interaction between Cry2 and Bclaf1, which stabilizes mRNAs encoding cyclin D1, a G1/S phase transition regulator, and Tmem176b, a transmembrane regulator for myogenic cell fusion. Myoblasts lacking Cry2 display premature cell cycle exit and form short myotubes because of inefficient cell fusion. Consistently, muscle regeneration is impaired in Cry2-/- mice. Bclaf1 knockdown recapitulated the phenotypes of Cry2 knockdown: early cell cycle exit and inefficient cell fusion. This study uncovers a post-transcriptional regulation of myogenic differentiation by circadian rhythms.


Assuntos
Diferenciação Celular , Ritmo Circadiano , Criptocromos/metabolismo , Ciclina D1/genética , Proteínas de Membrana/metabolismo , Desenvolvimento Muscular , Estabilidade de RNA/genética , Proteínas Repressoras/metabolismo , Animais , Ciclo Celular/genética , Fusão Celular , Linhagem Celular , Ciclina D1/metabolismo , Regulação da Expressão Gênica , Camundongos Knockout , Músculos/metabolismo , Mioblastos/citologia , Mioblastos/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Regeneração
9.
Cardiovasc Res ; 100(1): 105-13, 2013 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-23794713

RESUMO

AIMS: Fibroblasts can be directly reprogrammed to cardiomyocyte-like cells by introducing defined genes. However, the reprogramming efficiency remains low, delaying the clinical application of this strategy to regenerative cardiology. We previously showed that fusion of the MyoD transactivation domain to the pluripotency transcription factor Oct4 facilitated the transcriptional activity of Oct4, resulting in highly efficient production of induced pluripotent stem cells. We examined whether the same approach can be applied to cardiac transcription factors to facilitate cardiac reprogramming. METHODS AND RESULTS: We fused the MyoD domain to Mef2c, Gata4, Hand2, and Tbx5 and transduced these genes in various combinations into mouse non-cardiac fibroblasts. Transduction of the chimeric Mef2c with the wild-types of the other three genes produced much larger beating clusters of cardiomyocyte-like cells faster than the combination of the four wild-type genes, with an efficiency of 3.5%, >15-fold greater than the wild-type genes. CONCLUSION: Fusion of a powerful transactivation domain to heterologous factors can increase the efficiency of direct reprogramming of fibroblasts to cardiomyocytes.


Assuntos
Diferenciação Celular , Fibroblastos/citologia , Proteína MyoD/fisiologia , Miócitos Cardíacos/citologia , Ativação Transcricional , Animais , Imunofluorescência , Células-Tronco Pluripotentes Induzidas , Fatores de Transcrição MEF2/fisiologia , Camundongos , Proteína MyoD/química , Fator 3 de Transcrição de Octâmero/fisiologia , Estrutura Terciária de Proteína
10.
PLoS One ; 7(3): e34149, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22479546

RESUMO

A major difficulty of producing induced pluripotent stem cells (iPSCs) has been the low efficiency of reprogramming differentiated cells into pluripotent cells. We previously showed that 5% of mouse embryonic fibroblasts (MEFs) were reprogrammed into iPSCs when they were transduced with a fusion gene composed of Oct4 and the transactivation domain of MyoD (called M(3)O), along with Sox2, Klf4 and c-Myc (SKM). In addition, M(3)O facilitated chromatin remodeling of pluripotency genes in the majority of transduced MEFs, including cells that did not become iPSCs. These observations suggested the possibility that more than 5% of cells had acquired the ability to become iPSCs given more favorable culture conditions. Here, we raised the efficiency of making mouse iPSCs with M(3)O-SKM to 26% by culturing transduced cells at low density in serum-free culture medium. In contrast, the efficiency increased from 0.1% to only 2% with the combination of wild-type Oct4 and SKM (OSKM) under the same culture condition. For human iPSCs, M(3)O-SKM achieved 7% efficiency under a similar serum-free culture condition, in comparison to 1% efficiency with OSKM. This study highlights the power of combining the transactivation domain of MyoD with a favorable culture environment.


Assuntos
Técnicas de Cultura de Células , Células-Tronco Pluripotentes Induzidas/citologia , Proteína MyoD/metabolismo , Fosfatase Alcalina/metabolismo , Animais , Montagem e Desmontagem da Cromatina , Técnicas de Cocultura/métodos , Meios de Cultura Livres de Soro/farmacologia , Fibroblastos/citologia , Humanos , Fator 4 Semelhante a Kruppel , Camundongos , Camundongos Transgênicos , Microscopia de Fluorescência/métodos , Mioblastos/citologia , Fator 3 de Transcrição de Octâmero/metabolismo , Estrutura Terciária de Proteína , Teratoma/metabolismo , Ativação Transcricional
11.
J Biol Chem ; 284(8): 4968-77, 2009 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-19106111

RESUMO

Nucleostemin is a nucleolar protein widely expressed in proliferating cells. Nucleostemin is involved in the regulation of cell proliferation, and both depletion and overexpression of nucleostemin induce cell cycle arrest through the p53 signaling pathway. Although the presence of p53-independent functions of nucleostemin has been previously suggested, the identities of these additional functions remained to be investigated. Here, we show that nucleostemin has a novel role as an integrated component of ribosome biogenesis, particularly pre-rRNA processing. Nucleostemin forms a large protein complex (>700 kDa) that co-fractionates with the pre-60 S ribosomal subunit in a sucrose gradient. This complex contains proteins related to pre-rRNA processing, such as Pes1, DDX21, and EBP2, in addition to several ribosomal proteins. We show that the nucleolar retention of DDX21 and EBP2 is dependent on the presence of nucleostemin in the nucleolus. Furthermore, the knockdown of nucleostemin delays the processing of 32 S pre-rRNA into 28 S rRNA. This is accompanied by a substantial decrease of protein synthesis as well as the levels of rRNAs and some mRNAs. In addition, overexpressed nucleostemin significantly promotes the processing of 32 S pre-rRNA. Collectively, these biochemical and functional studies demonstrate a novel role of nucleostemin in ribosome biogenesis. This is a key aspect of the role of nucleostemin in regulating cell proliferation.


Assuntos
Proteínas de Transporte/metabolismo , Ciclo Celular/fisiologia , Proteínas Nucleares/metabolismo , Precursores de RNA/metabolismo , Processamento Pós-Transcricional do RNA/fisiologia , RNA Ribossômico 28S/metabolismo , Ribossomos/metabolismo , Proteínas de Transporte/genética , RNA Helicases DEAD-box/genética , RNA Helicases DEAD-box/metabolismo , Proteínas de Ligação ao GTP , Células HeLa , Humanos , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Proteínas Nucleares/genética , Proteínas/genética , Proteínas/metabolismo , Precursores de RNA/genética , RNA Ribossômico 28S/genética , Proteínas de Ligação a RNA , Ribossomos/genética , Transdução de Sinais/fisiologia , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
12.
Biochem Biophys Res Commun ; 361(3): 611-4, 2007 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-17669361

RESUMO

The transcription factors Oct4 and Nanog are essential for the maintenance of an undifferentiated and pluripotent state in early embryonic cells, embryonic stem cells and embryonal carcinoma cells in humans and mice. These factors are co-localized to promoters of more than 300 genes, and synergistically regulate their activities. Currently, the molecular interaction between these two factors has not been well-characterized. During attempts to co-immunoprecipitate Oct4 and Nanog we found that cross-linking with dithiobis[succinimidylpropionate] was necessary to maintain their interaction. This result was supported by gel filtration analysis. Surprisingly, formaldehyde, a cross-linker commonly used during chromatin immunoprecipitation of Oct4 and Nanog, did not preserve the complex. Our findings demonstrate the effectiveness of using DSP to mitigate the instability of the interaction between these two particular proteins. Additionally, this solution may potentially allow us to identify novel members of the Oct4-Nanog complex, leading to better understanding of the regulatory mechanisms behind pluripotency.


Assuntos
Reagentes de Ligações Cruzadas/farmacologia , Proteínas de Ligação a DNA/isolamento & purificação , Proteínas de Homeodomínio/isolamento & purificação , Imunoprecipitação , Fator 3 de Transcrição de Octâmero/isolamento & purificação , Succinimidas/farmacologia , Animais , Carcinoma Embrionário/metabolismo , Linhagem Celular Tumoral , Permeabilidade da Membrana Celular , Cromatografia em Gel , Proteínas de Ligação a DNA/metabolismo , Proteínas de Homeodomínio/metabolismo , Masculino , Mercaptoetanol/metabolismo , Camundongos , Proteína Homeobox Nanog , Fator 3 de Transcrição de Octâmero/metabolismo , Dodecilsulfato de Sódio/metabolismo , Neoplasias Testiculares/metabolismo
13.
J Biol Chem ; 281(12): 8153-60, 2006 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-16415342

RESUMO

In Xenopus somatic cell nuclear cloning, the nucleoli of donor nuclei rapidly and almost completely disappear in egg cytoplasm. We previously showed that the germ cell-specific proteins FRGY2a and FRGY2b were responsible for this unusually drastic nucleolar disassembly. The nucleolar disassembly occurs without inhibition of pre-rRNA transcription, a well known trigger for nucleolar segregation, and the mechanism for the nucleolar disassembly by FRGY2a and FRGY2b remains largely unknown. In this study, we searched for FRGY2a-interacting proteins and investigated the functional consequences of their interactions through a series of experiments. We showed that during the nucleolar disassembly, FRGY2a localized to the nucleoli of isolated nuclei and was capable of disassembling purified nucleoli, suggesting a direct interaction between FRGY2a and nucleolar components. Using a His tag pulldown approach, we identified the abundant and multifunctional nucleolar protein B23 as a potential target of FRGY2a and its related human protein YB1. A specific interaction between FRGY2a/YB1 and B23 was confirmed by co-immunoprecipitation. Finally, B23 knockdown using short interfering RNA and a subsequent add-back experiment confirmed that B23 was necessary for nucleolar disassembly by YB1. We propose that FRGY2a and YB1 disassemble nucleoli by sequestering B23, which is associated with pre-ribosomes and other structurally important nucleolar components.


Assuntos
Nucléolo Celular/metabolismo , Proteínas Nucleares/fisiologia , Proteínas de Ligação a RNA/fisiologia , Fatores de Transcrição/fisiologia , Proteínas de Xenopus/fisiologia , Animais , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Clonagem Molecular , DNA Complementar/metabolismo , Células HeLa , Histidina/química , Humanos , Imunoprecipitação , Espectrometria de Massas , Microscopia Eletrônica , Microscopia de Fluorescência , Proteínas Nucleares/metabolismo , Nucleofosmina , Ligação Proteica , Estrutura Terciária de Proteína , RNA Interferente Pequeno/metabolismo , Proteínas de Ligação a RNA/metabolismo , Proteínas Recombinantes/química , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição/metabolismo , Transfecção , Xenopus , Proteínas de Xenopus/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA