Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Genes Dev ; 38(7-8): 308-321, 2024 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-38719541

RESUMO

The transcription factor Oct4/Pou5f1 is a component of the regulatory circuitry governing pluripotency and is widely used to induce pluripotency from somatic cells. Here we used domain swapping and mutagenesis to study Oct4's reprogramming ability, identifying a redox-sensitive DNA binding domain, cysteine residue (Cys48), as a key determinant of reprogramming and differentiation. Oct4 Cys48 sensitizes the protein to oxidative inhibition of DNA binding activity and promotes oxidation-mediated protein ubiquitylation. Pou5f1 C48S point mutation has little effect on undifferentiated embryonic stem cells (ESCs) but upon retinoic acid (RA) treatment causes retention of Oct4 expression, deregulated gene expression, and aberrant differentiation. Pou5f1 C48S ESCs also form less differentiated teratomas and contribute poorly to adult somatic tissues. Finally, we describe Pou5f1 C48S (Janky) mice, which in the homozygous condition are severely developmentally restricted after E4.5. Rare animals bypassing this restriction appear normal at birth but are sterile. Collectively, these findings uncover a novel Oct4 redox mechanism involved in both entry into and exit from pluripotency.


Assuntos
Diferenciação Celular , Reprogramação Celular , Fator 3 de Transcrição de Octâmero , Oxirredução , Fator 3 de Transcrição de Octâmero/metabolismo , Fator 3 de Transcrição de Octâmero/genética , Animais , Camundongos , Diferenciação Celular/genética , Reprogramação Celular/genética , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Tretinoína/farmacologia , Tretinoína/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Humanos
2.
J Biol Chem ; 289(52): 36070-88, 2014 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-25406310

RESUMO

Usher syndrome (USH) is the leading genetic cause of combined hearing and vision loss. Among the three USH clinical types, type 2 (USH2) occurs most commonly. USH2A, GPR98, and WHRN are three known causative genes of USH2, whereas PDZD7 is a modifier gene found in USH2 patients. The proteins encoded by these four USH genes have been proposed to form a multiprotein complex, the USH2 complex, due to interactions found among some of these proteins in vitro, their colocalization in vivo, and mutual dependence of some of these proteins for their normal in vivo localizations. However, evidence showing the formation of the USH2 complex is missing, and details on how this complex is formed remain elusive. Here, we systematically investigated interactions among the intracellular regions of the four USH proteins using colocalization, yeast two-hybrid, and pull-down assays. We show that multiple domains of the four USH proteins interact among one another. Importantly, both WHRN and PDZD7 are required for the complex formation with USH2A and GPR98. In this USH2 quaternary complex, WHRN prefers to bind to USH2A, whereas PDZD7 prefers to bind to GPR98. Interaction between WHRN and PDZD7 is the bridge between USH2A and GPR98. Additionally, the USH2 quaternary complex has a variable stoichiometry. These findings suggest that a non-obligate, short term, and dynamic USH2 quaternary protein complex may exist in vivo. Our work provides valuable insight into the physiological role of the USH2 complex in vivo and informs possible reconstruction of the USH2 complex for future therapy.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas de Membrana/metabolismo , Síndromes de Usher/metabolismo , Sequência de Aminoácidos , Animais , Células COS , Proteínas de Transporte/química , Chlorocebus aethiops , Proteínas da Matriz Extracelular/química , Proteínas da Matriz Extracelular/metabolismo , Células HEK293 , Humanos , Proteínas de Membrana/química , Camundongos , Domínios PDZ , Ligação Proteica , Multimerização Proteica , Estrutura Quaternária de Proteína , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/metabolismo , Técnicas do Sistema de Duplo-Híbrido , Síndromes de Usher/genética
3.
Hum Mol Genet ; 21(3): 692-710, 2012 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-22048959

RESUMO

Whirlin mutations cause retinal degeneration and hearing loss in Usher syndrome type II (USH2) and non-syndromic deafness, DFNB31. Its protein recruits other USH2 causative proteins to form a complex at the periciliary membrane complex in photoreceptors and the ankle link of the stereocilia in hair cells. However, the biological function of this USH2 protein complex is largely unknown. Using a yeast two-hybrid screen, we identified espin, an actin-binding/bundling protein involved in human deafness when defective, as a whirlin-interacting protein. The interaction between these two proteins was confirmed by their coimmunoprecipitation and colocalization in cultured cells. This interaction involves multiple domains of both proteins and only occurs when espin does not bind to actin. Espin was partially colocalized with whirlin in the retina and the inner ear. In whirlin knockout mice, espin expression changed significantly in these two tissues. Further studies found that whirlin increased the mobility of espin and actin at the actin bundles cross-linked by espin and, eventually, affected the dimension of these actin bundles. In whirlin knockout mice, the stereocilia were thickened in inner hair cells. We conclude that the interaction between whirlin and espin and the balance between their expressions are required to maintain the actin bundle network in photoreceptors and hair cells. Disruption of this actin bundle network contributes to the pathogenic mechanism of hearing loss and retinal degeneration caused by whirlin and espin mutations. Espin is a component of the USH2 protein complex and could be a candidate gene for Usher syndrome.


Assuntos
Proteínas de Membrana/metabolismo , Proteínas dos Microfilamentos/metabolismo , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/ultraestrutura , Animais , Encéfalo/metabolismo , Células COS , Chlorocebus aethiops , Orelha Interna/metabolismo , Células HEK293 , Células Ciliadas Auditivas/química , Células Ciliadas Auditivas/ultraestrutura , Humanos , Proteínas de Membrana/química , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Proteínas dos Microfilamentos/análise , Proteínas dos Microfilamentos/química , Células Fotorreceptoras de Vertebrados/química , Domínios e Motivos de Interação entre Proteínas , Pseudópodes/metabolismo , Retina/metabolismo , Estereocílios/ultraestrutura , Síndromes de Usher/etiologia
4.
FASEB J ; 27(7): 2807-17, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23580612

RESUMO

The Oct1 transcription factor is a potent regulator of stress responses, metabolism, and tumorigenicity. Although Oct1 is regulated by phosphorylation and ubiquitination, the presence and importance of other modifications is unknown. Here we show that Oct1 is modified by O-linked ß-N-acetylglucosamine (O-GlcNAc) moieties. We map two sites of O-GlcNAcylation at positions T255 and S728 within human Oct1. Under anchorage-independent overgrowth conditions, Oct1 associates 3-fold more strongly with the Gadd45a promoter and mediates transcriptional repression. Increased binding correlates with quantitative reductions in Oct1 nuclear periphery-associated puncta, and a reduced association with lamin B1. The O-GlcNAc modification sites are important for both Gadd45a repression and anchorage-independent survival. In contrast to chronic overgrowth conditions, following acute nutrient starvation Oct1 mediates Gadd45a activation. The O-GlcNAc sites are also important for Gadd45a activation under these conditions. We also, for the first time, identify specific Oct1 ubiquitination sites. The findings suggest that Oct1 integrates metabolic and stress signals via O-GlcNAc modification to regulate target gene activity.


Assuntos
Acetilglucosamina/metabolismo , Fator de Transcrição Brn-3A/metabolismo , Ativação Transcricional , Células 3T3 , Sequência de Aminoácidos , Animais , Sítios de Ligação/genética , Proteínas de Ciclo Celular/genética , Embrião de Mamíferos/citologia , Fibroblastos/metabolismo , Regulação da Expressão Gênica , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HeLa , Humanos , Immunoblotting , Camundongos , Camundongos Knockout , Microscopia de Fluorescência , Dados de Sequência Molecular , Mutação , Proteínas Nucleares/genética , Regiões Promotoras Genéticas/genética , Ligação Proteica , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição Brn-3A/genética
5.
bioRxiv ; 2024 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-36865286

RESUMO

The transcription factor Oct4/Pou5f1 is a component of the regulatory circuitry governing pluripotency and is widely used to induce pluripotency from somatic cells. Here we use domain swapping and mutagenesis to study Oct4s reprogramming ability, identifying a redox-sensitive DNA binding domain cysteine residue (Cys48) as a key determinant of reprogramming and differentiation. Oct4 Cys48 sensitizes the protein to oxidative inhibition of DNA binding activity and promotes oxidation-mediated protein ubiquitylation. Pou5f1C48S point mutation has little effect on undifferentiated embryonic stem cells (ESCs), but upon retinoic acid (RA) treatment causes retention of Oct4 expression, deregulated gene expression and aberrant differentiation. Pou5f1C48S ESCs also form less differentiated teratomas and contribute poorly to adult somatic tissues. Finally, we describe Pou5f1C48S (Janky) mice, which in the homozygous condition are severely developmentally restricted after E4.5. Rare animals bypassing this restriction appear normal at birth but are sterile. Collectively, these findings uncover a novel Oct4 redox mechanism involved in both entry into and exit from pluripotency.

6.
Sci Signal ; 16(781): eadd5750, 2023 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-37071732

RESUMO

The transition between pluripotent and tissue-specific states is a key aspect of development. Understanding the pathways driving these transitions will facilitate the engineering of properly differentiated cells for experimental and therapeutic uses. Here, we showed that during mesoderm differentiation, the transcription factor Oct1 activated developmental lineage-appropriate genes that were silent in pluripotent cells. Using mouse embryonic stem cells (ESCs) with an inducible knockout of Oct1, we showed that Oct1 deficiency resulted in poor induction of mesoderm-specific genes, leading to impaired mesodermal and terminal muscle differentiation. Oct1-deficient cells exhibited poor temporal coordination of the induction of lineage-specific genes and showed inappropriate developmental lineage branching, resulting in poorly differentiated cell states retaining epithelial characteristics. In ESCs, Oct1 localized with the pluripotency factor Oct4 at mesoderm-associated genes and remained bound to those loci during differentiation after the dissociation of Oct4. Binding events for Oct1 overlapped with those for the histone lysine demethylase Utx, and an interaction between Oct1 and Utx suggested that these two proteins cooperate to activate gene expression. The specificity of the ubiquitous Oct1 for the induction of mesodermal genes could be partially explained by the frequent coexistence of Smad and Oct binding sites at mesoderm-specific genes and the cooperative stimulation of mesodermal gene transcription by Oct1 and Smad3. Together, these results identify Oct1 as a key mediator of mesoderm lineage-specific gene induction.


Assuntos
Células-Tronco Embrionárias , Fatores de Transcrição , Animais , Camundongos , Fatores de Transcrição/metabolismo , Diferenciação Celular , Sítios de Ligação , Mesoderma/metabolismo , Linhagem da Célula
7.
Sci Adv ; 8(24): eabm4982, 2022 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-35704571

RESUMO

In response to various types of infection, naïve CD4+ T cells differentiate into diverse helper T cell subsets; however, the epigenetic programs that regulate differentiation in response to viral infection remain poorly understood. Demethylation of CpG dinucleotides by Tet methylcytosine dioxygenases is a key component of epigenetic programing that promotes specific gene expression, cellular differentiation, and function. We report that following viral infection, Tet2-deficient CD4+ T cells preferentially differentiate into highly functional germinal center T follicular helper (TFH) cells that provide enhanced help for B cells. Using genome-wide DNA methylation and transcription factor binding analyses, we find that Tet2 coordinates with multiple transcription factors, including Foxo1 and Runx1, to mediate the demethylation and expression of target genes, including genes encoding repressors of TFH differentiation. Our findings establish Tet2 as an important regulator of TFH cell differentiation and reveal pathways that could be targeted to enhance immune responses against infectious disease.


Assuntos
Centro Germinativo , Células T Auxiliares Foliculares , Diferenciação Celular/genética , Ativação Linfocitária , Linfócitos T Auxiliares-Indutores
8.
J Exp Med ; 218(3)2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33295943

RESUMO

The transcriptional coregulator OCA-B promotes expression of T cell target genes in cases of repeated antigen exposure, a necessary feature of autoimmunity. We hypothesized that T cell-specific OCA-B deletion and pharmacologic OCA-B inhibition would protect mice from autoimmune diabetes. We developed an Ocab conditional allele and backcrossed it onto a diabetes-prone NOD/ShiLtJ strain background. T cell-specific OCA-B loss protected mice from spontaneous disease. Protection was associated with large reductions in islet CD8+ T cell receptor specificities associated with diabetes pathogenesis. CD4+ clones associated with diabetes were present but associated with anergic phenotypes. The protective effect of OCA-B loss was recapitulated using autoantigen-specific NY8.3 mice but diminished in monoclonal models specific to artificial or neoantigens. Rationally designed membrane-penetrating OCA-B peptide inhibitors normalized glucose levels and reduced T cell infiltration and proinflammatory cytokine expression in newly diabetic NOD mice. Together, the results indicate that OCA-B is a potent autoimmune regulator and a promising target for pharmacologic inhibition.


Assuntos
Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/imunologia , Pâncreas/patologia , Linfócitos T/imunologia , Transativadores/metabolismo , Transcrição Gênica , Alelos , Sequência de Aminoácidos , Animais , Autoantígenos/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Cruzamentos Genéticos , Citocinas/metabolismo , Diabetes Mellitus Tipo 1/prevenção & controle , Modelos Animais de Doenças , Feminino , Deleção de Genes , Células Germinativas/metabolismo , Humanos , Mediadores da Inflamação/metabolismo , Linfonodos/metabolismo , Ativação Linfocitária , Masculino , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Ovalbumina , Pâncreas/metabolismo , Peptídeos/farmacologia , Receptores de Antígenos de Linfócitos T/metabolismo , Baço/patologia , Transativadores/deficiência
9.
Stem Cells Dev ; 27(24): 1693-1701, 2018 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-30319048

RESUMO

The histone chaperone facilitates chromatin transactions (FACT) is associated with nuclear processes, including DNA transcription, replication, and repair. We previously showed that FACT is transiently recruited to pluripotency-associated target genes by newly bound Oct4. In this study, we tested the effects of FACT depletion by knockout or chemical inhibition on the induction and maintenance of pluripotency. Clustered regularly interspaced short palindromic repeat (CRISPR)-mediated deletion of the FACT subunit Spt16 did not affect the viability or proliferation of fibroblasts but blocked their ability to form induced pluripotent stem cells. Similarly, a small molecule inhibitor of FACT blocked the induction of pluripotency at an early step in reprogramming, without affecting the viability, proliferation, undifferentiated state, or the expression of core pluripotency genes. Notably, trypsinization and passage of pluripotent cells transiently reintroduced a requirement for FACT. Although FACT has been considered to be an essential transcription elongation factor, these results contribute to the emerging view that it instead promotes transitions between stable chromatin states, including during reprogramming to pluripotency.


Assuntos
Reprogramação Celular , Células-Tronco Pluripotentes/citologia , Fatores de Transcrição/metabolismo , Células 3T3 , Animais , Células Cultivadas , Cromatina/metabolismo , Camundongos , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/genética
10.
Elife ; 62017 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-28537559

RESUMO

Embryonic stem cells co-express Oct4 and Oct1, a related protein with similar DNA-binding specificity. To study the role of Oct1 in ESC pluripotency and transcriptional control, we constructed germline and inducible-conditional Oct1-deficient ESC lines. ESCs lacking Oct1 show normal appearance, self-renewal and growth but manifest defects upon differentiation. They fail to form beating cardiomyocytes, generate neurons poorly, form small, poorly differentiated teratomas, and cannot generate chimeric mice. Upon RA-mediated differentiation, Oct1-deficient cells induce lineage-appropriate developmentally poised genes poorly while lineage-inappropriate genes, including extra-embryonic genes, are aberrantly expressed. In ESCs, Oct1 co-occupies a specific set of targets with Oct4, but does not occupy differentially expressed developmental targets. Instead, Oct1 occupies these targets as cells differentiate and Oct4 declines. These results identify a dynamic interplay between Oct1 and Oct4, in particular during the critical window immediately after loss of pluripotency when cells make the earliest developmental fate decisions.


Assuntos
Diferenciação Celular , Regulação da Expressão Gênica no Desenvolvimento , Células-Tronco Embrionárias Murinas/fisiologia , Fator 1 de Transcrição de Octâmero/metabolismo , Transcrição Gênica , Animais , Camundongos , Fator 3 de Transcrição de Octâmero/metabolismo
11.
Elife ; 52016 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-27661449

RESUMO

PAS domain containing protein kinase (Pask) is an evolutionarily conserved protein kinase implicated in energy homeostasis and metabolic regulation across eukaryotic species. We now describe an unexpected role of Pask in promoting the differentiation of myogenic progenitor cells, embryonic stem cells and adipogenic progenitor cells. This function of Pask is dependent upon its ability to phosphorylate Wdr5, a member of several protein complexes including those that catalyze histone H3 Lysine 4 trimethylation (H3K4me3) during transcriptional activation. Our findings suggest that, during myoblast differentiation, Pask stimulates the conversion of repressive H3K4me1 to activating H3K4me3 marks on the promoter of the differentiation gene myogenin (Myog) via Wdr5 phosphorylation. This enhances accessibility of the MyoD transcription factor and enables transcriptional activation of the Myog promoter to initiate muscle differentiation. Thus, as an upstream kinase of Wdr5, Pask integrates signaling cues with the transcriptional network to regulate the differentiation of progenitor cells.


Assuntos
Código das Histonas , Histona-Lisina N-Metiltransferase/metabolismo , Desenvolvimento Muscular/fisiologia , Músculos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Animais , Diferenciação Celular , Linhagem Celular , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Células HEK293 , Histona-Lisina N-Metiltransferase/genética , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Células-Tronco Embrionárias Murinas/metabolismo , Células Musculares/fisiologia , Músculo Esquelético , Músculos/lesões , Proteína MyoD/metabolismo , Mioblastos/patologia , Miogenina/genética , Miogenina/metabolismo , Fosforilação , Regiões Promotoras Genéticas , Proteínas Serina-Treonina Quinases/genética , Células-Tronco , Ativação Transcricional
12.
Invest Ophthalmol Vis Sci ; 52(5): 2343-51, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21212183

RESUMO

PURPOSE: Whirlin is the causative gene for Usher syndrome type IID (USH2D), a condition manifested as both retinitis pigmentosa and congenital deafness. Mutations in this gene cause disruption of the USH2 protein complex composed of USH2A and VLGR1 at the periciliary membrane complex (PMC) in photoreceptors. In this study, the adeno-associated virus (AAV)-mediated whirlin replacement was evaluated as a treatment option. METHODS: Murine whirlin cDNA driven by the human rhodopsin kinase promoter (hRK) was packaged as an AAV2/5 vector and delivered into the whirlin knockout retina through subretinal injection. The efficiency, efficacy, and safety of this treatment were examined using immunofluorescent staining, confocal imaging, immunoelectron microscopy, Western blot analysis, histologic analysis, and electroretinogram. RESULTS: The AAV-mediated whirlin expression started at two weeks, reached its maximum level at 10 weeks, and lasted up to six months post injection. The transgenic whirlin product had a molecular size and an expression level comparable to the wild-type. It was distributed at the PMC in both rod and cone photoreceptors from the central to peripheral retina. Importantly, the transgenic whirlin restored the cellular localization and expression level of both USH2A and VLGR1 and did not cause defects in the retinal histology and function in the whirlin knockout mouse. CONCLUSIONS: Whirlin transgene recruits USH2A and VLGR1 to the PMC and is sufficient for the formation of the USH2 protein complex in photoreceptors. The combined hRK and AAV gene delivery system could be an effective gene therapy approach to treat retinal degeneration in USH2D patients.


Assuntos
Proteínas da Matriz Extracelular/genética , Regulação da Expressão Gênica/fisiologia , Vetores Genéticos , Proteínas de Membrana/genética , Células Fotorreceptoras de Vertebrados/metabolismo , Animais , Animais Recém-Nascidos , Western Blotting , Dependovirus/genética , Eletrorretinografia , Proteínas da Matriz Extracelular/metabolismo , Técnica Indireta de Fluorescência para Anticorpo , Receptor Quinase 1 Acoplada a Proteína G/genética , Deleção de Genes , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Confocal , Microscopia Imunoeletrônica , Plasmídeos , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Transgenes , Síndromes de Usher/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA