Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Nat Chem Biol ; 17(1): 47-56, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32807969

RESUMEN

Identifying molecular and cellular processes that regulate reprogramming competence of transcription factors broadens our understanding of reprogramming mechanisms. In the present study, by a chemical screen targeting major epigenetic pathways in human reprogramming, we discovered that inhibiting specific epigenetic roadblocks including disruptor of telomeric silencing 1-like (DOT1L)-mediated H3K79/K27 methylation, but also other epigenetic pathways, catalyzed by lysine-specific histone demethylase 1A, DNA methyltransferases and histone deacetylases, allows induced pluripotent stem cell generation with almost all OCT factors. We found that simultaneous inhibition of these pathways not only dramatically enhances reprogramming competence of most OCT factors, but in fact enables dismantling of species-dependent reprogramming competence of OCT6, NR5A1, NR5A2, TET1 and GATA3. Harnessing these induced permissive epigenetic states, we performed an additional screen with 98 candidate genes. Thereby, we identified 25 transcriptional regulators (OTX2, SIX3, and so on) that can functionally replace OCT4 in inducing pluripotency. Our findings provide a conceptual framework for understanding how transcription factors elicit reprogramming in dependency of the donor cell epigenome that differs across species.


Asunto(s)
Reprogramación Celular , Epigénesis Genética , N-Metiltransferasa de Histona-Lisina/genética , Histonas/genética , Células Madre Embrionarias Humanas/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Animales , Línea Celular , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Células HEK293 , Células HeLa , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Células Madre Embrionarias Humanas/citología , Humanos , Células Madre Pluripotentes Inducidas/citología , Ratones , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Factores de Transcripción de Octámeros/genética , Factores de Transcripción de Octámeros/metabolismo , Factores de Transcripción Otx/genética , Factores de Transcripción Otx/metabolismo , Plásmidos/química , Plásmidos/metabolismo , Especificidad de la Especie , Transcripción Genética , Transfección , Proteína Homeobox SIX3
2.
J Craniofac Surg ; 34(1): 206-213, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36173963

RESUMEN

In this study, the authors report the experience of extended superficial musculoaponeurotic system (SMAS) face-lift with the vertical vector in Asian ethnicity and investigate the 3-dimensional change of facial contour. A total of 32 patients with Korean ethnicity underwent extended SMAS face-lift with vertical vector from 2015 to 2018. Patients with aging face were included for the study subjects, whereas those who were diagnosed with any craniofacial deformity were excluded. Using 3-dimensional photogrammetry, surface contour analysis was performed in the cheek region to calculate the mean, maximal, and minimal difference of surface projection and global root mean square error between the preoperative and 1-year postoperative period. The change of horizontal facial widths and jawline angle was evaluated. In contour analysis, the mean difference of surface contour was highest in anterior, followed by lateral cheek and lower face, sequentially. The maximal difference of surface contour was highest in anterior cheek, followed by lateral cheek and lower face, whereas the minimal difference of surface contour was lowest in lower face, followed by anterior cheek and lateral cheek, sequentially. No significant differences in the midfacial and lower facial distances were observed between the preoperative and postoperative periods. There was significant increase of jawline angle, from 20.78 to 23.14 degree of mean value ( P =0.001). In conclusion, the extended SMAS face-lift with vertical vector can be an optimal option for Asian subjects in terms of the midfacial volumetric shift, sharpening of jawline and avoidance of midfacial widening.


Asunto(s)
Ritidoplastia , Sistema Músculo-Aponeurótico Superficial , Humanos , Sistema Músculo-Aponeurótico Superficial/cirugía , Ritidoplastia/métodos , Mejilla/cirugía , Fotogrametría
3.
Genes Dev ; 29(5): 513-25, 2015 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-25737281

RESUMEN

The transcriptional activator MyoD serves as a master controller of myogenesis. Often in partnership with Mef2 (myocyte enhancer factor 2), MyoD binds to the promoters of hundreds of muscle genes in proliferating myoblasts yet activates these targets only upon receiving cues that launch differentiation. What regulates this off/on switch of MyoD function has been incompletely understood, although it is known to reflect the action of chromatin modifiers. Here, we identify KAP1 (KRAB [Krüppel-like associated box]-associated protein 1)/TRIM28 (tripartite motif protein 28) as a key regulator of MyoD function. In myoblasts, KAP1 is present with MyoD and Mef2 at many muscle genes, where it acts as a scaffold to recruit not only coactivators such as p300 and LSD1 but also corepressors such as G9a and HDAC1 (histone deacetylase 1), with promoter silencing as the net outcome. Upon differentiation, MSK1-mediated phosphorylation of KAP1 releases the corepressors from the scaffold, unleashing transcriptional activation by MyoD/Mef2 and their positive cofactors. Thus, our results reveal KAP1 as a previously unappreciated interpreter of cell signaling, which modulates the ability of MyoD to drive myogenesis.


Asunto(s)
Diferenciación Celular , Desarrollo de Músculos/fisiología , Músculo Esquelético/citología , Proteína MioD/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Represoras/metabolismo , Animales , Línea Celular , Regulación del Desarrollo de la Expresión Génica , Factores de Transcripción MEF2/metabolismo , Ratones , Proteína MioD/genética , Mioblastos/citología , Proteínas Nucleares/genética , Fosforilación , Proteínas Represoras/genética , Transducción de Señal , Proteína 28 que Contiene Motivos Tripartito
4.
Nucleic Acids Res ; 43(9): 4505-16, 2015 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-25813038

RESUMEN

Post-translational modifications of core histones affect various cellular processes, primarily through transcription. However, their relationship with the termination of transcription has remained largely unknown. In this study, we show that DNA damage-activated AKT phosphorylates threonine 45 of core histone H3 (H3-T45). By genome-wide chromatin immunoprecipitation sequencing (ChIP-seq) analysis, H3-T45 phosphorylation was distributed throughout DNA damage-responsive gene loci, particularly immediately after the transcription termination site. H3-T45 phosphorylation pattern showed close-resemblance to that of RNA polymerase II C-terminal domain (CTD) serine 2 phosphorylation, which establishes the transcription termination signal. AKT1 was more effective than AKT2 in phosphorylating H3-T45. Blocking H3-T45 phosphorylation by inhibiting AKT or through amino acid substitution limited RNA decay downstream of mRNA cleavage sites and decreased RNA polymerase II release from chromatin. Our findings suggest that AKT-mediated phosphorylation of H3-T45 regulates the processing of the 3' end of DNA damage-activated genes to facilitate transcriptional termination.


Asunto(s)
Daño del ADN , Histonas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Terminación de la Transcripción Genética , Línea Celular , Células HeLa , Histonas/química , Humanos , Células MCF-7 , Fosforilación , Treonina/metabolismo , Sitio de Iniciación de la Transcripción
5.
Stem Cells ; 33(9): 2699-711, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26059508

RESUMEN

Pluripotent stem cells (PSCs) have distinct metabolic properties that support their metabolic and energetic needs and affect their stemness. In particular, high glycolysis is critical for the generation and maintenance of PSCs. However, it is unknown how PSCs maintain and acquire this metabolic signature. In this study, we found that core pluripotency factors regulate glycolysis directly by controlling the expression of glycolytic enzymes. Specifically, Oct4 directly governs Hk2 and Pkm2, which are important glycolytic enzymes that determine the rate of glycolytic flux. The overexpression of Hk2 and Pkm2 sustains high levels of glycolysis during embryonic stem cell (ESC) differentiation. Moreover, the maintenance of high glycolysis levels by Hk2 and Pkm2 overexpression hampers differentiation and preserves the pluripotency of ESCs in the absence of leukemia inhibitory factor. Overall, our study identifies a direct molecular connection between core pluripotency factors and ESC metabolic signatures and demonstrates the significance of metabolism in cell fate determination.


Asunto(s)
Proteínas Portadoras/biosíntesis , Células Madre Embrionarias/metabolismo , Glucólisis/fisiología , Hexoquinasa/biosíntesis , Proteínas de la Membrana/biosíntesis , Factor 3 de Transcripción de Unión a Octámeros/biosíntesis , Células Madre Pluripotentes/metabolismo , Hormonas Tiroideas/biosíntesis , Animales , Diferenciación Celular/fisiología , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Proteínas de Unión a Hormona Tiroide
6.
Nucleic Acids Res ; 42(1): 224-34, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24078251

RESUMEN

Myocyte enhancer factor 2 (MEF2) is a family of transcription factors that regulates many processes, including muscle differentiation. Due to its many target genes, MEF2D requires tight regulation of transcription activity over time and by location. Epigenetic modifiers have been suggested to regulate MEF2-dependent transcription via modifications to histones and MEF2. However, the modulation of MEF2 activity by lysine methylation, an important posttranslational modification that alters the activities of transcription factors, has not been studied. We report the reversible lysine methylation of MEF2D by G9a and LSD1 as a regulatory mechanism of MEF2D activity and skeletal muscle differentiation. G9a methylates lysine-267 of MEF2D and represses its transcriptional activity, but LSD1 counteracts it. This residue is highly conserved between MEF2 members in mammals. During myogenic differentiation of C2C12 mouse skeletal muscle cells, the methylation of MEF2D by G9a decreased, on which MEF2D-dependent myogenic genes were upregulated. We have also identified lysine-267 as a methylation/demethylation site and demonstrate that the lysine methylation state of MEF2D regulates its transcriptional activity and skeletal muscle cell differentiation.


Asunto(s)
Diferenciación Celular/genética , Lisina/metabolismo , Factores de Transcripción MEF2/metabolismo , Mioblastos Esqueléticos/metabolismo , Animales , Línea Celular , Cromatina/metabolismo , Células HEK293 , Histona Demetilasas , N-Metiltransferasa de Histona-Lisina/antagonistas & inhibidores , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Factores de Transcripción MEF2/antagonistas & inhibidores , Factores de Transcripción MEF2/química , Metilación , Ratones , Mioblastos Esqueléticos/citología , Oxidorreductasas N-Desmetilantes/metabolismo , Procesamiento Proteico-Postraduccional , Transcripción Genética
7.
Exp Mol Med ; 56(2): 251-263, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38297159

RESUMEN

H3.3, the most common replacement variant for histone H3, has emerged as an important player in chromatin dynamics for controlling gene expression and genome integrity. While replicative variants H3.1 and H3.2 are primarily incorporated into nucleosomes during DNA synthesis, H3.3 is under the control of H3.3-specific histone chaperones for spatiotemporal incorporation throughout the cell cycle. Over the years, there has been progress in understanding the mechanisms by which H3.3 affects domain structure and function. Furthermore, H3.3 distribution and relative abundance profoundly impact cellular identity and plasticity during normal development and pathogenesis. Recurrent mutations in H3.3 and its chaperones have been identified in neoplastic transformation and developmental disorders, providing new insights into chromatin biology and disease. Here, we review recent findings emphasizing how two distinct histone chaperones, HIRA and DAXX, take part in the spatial and temporal distribution of H3.3 in different chromatin domains and ultimately achieve dynamic control of chromatin organization and function. Elucidating the H3.3 deposition pathways from the available histone pool will open new avenues for understanding the mechanisms by which H3.3 epigenetically regulates gene expression and its impact on cellular integrity and pathogenesis.


Asunto(s)
Proteínas de Ciclo Celular , Cromatina , Proteínas Co-Represoras , Histonas , Chaperonas Moleculares , Factores de Transcripción , Ciclo Celular , División Celular , Cromatina/genética , Chaperonas de Histonas/genética , Humanos , Chaperonas Moleculares/genética , Proteínas Co-Represoras/genética , Factores de Transcripción/genética , Proteínas de Ciclo Celular/genética
8.
BMB Rep ; 56(4): 252-257, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36789561

RESUMEN

The hypoxia-inducible factor-1α (HIF-1α) is a key regulator of hypoxic stress under physiological and pathological conditions. HIF-1α protein stability is tightly regulated by the ubiquitin-proteasome system (UPS) and autophagy in normoxia, hypoxia, and the tumor environment to mediate the hypoxic response. However, the mechanisms of how the UPS and autophagy interplay for HIF-1α proteostasis remain unclear. Here, we found a HIF-1α species propionylated at lysine (K) 709 by p300/CREB binding protein (CBP). HIF-1α stability and the choice of degradation pathway were affected by HIF-1α propionylation. K709-propionylation prevented HIF-1α from degradation through the UPS, while activated chaperon-mediated autophagy (CMA) induced the degradation of propionylated and nonpropionylated HIF-1α. CMA contributed to HIF-1α degradation in both normoxia and hypoxia. Furthermore, the pan-cancer analysis showed that CMA had a significant positive correlation with the hypoxic signatures, whereas SIRT1, responsible for K709-depropionylation correlated negatively with them. Altogether, our results revealed a novel mechanism of HIF-1α distribution into two different degradation pathways. [BMB Reports 2023; 56(4): 252-257].


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia , Neoplasias , Humanos , Neoplasias/patología , Complejo de la Endopetidasa Proteasomal , Hipoxia , Hipoxia de la Célula
9.
Elife ; 112022 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-35621159

RESUMEN

The transcription factor Oct4 is essential for the maintenance and induction of stem cell pluripotency, but its functional roles are not fully understood. Here, we investigate the functions of Oct4 by depleting and subsequently recovering it in mouse embryonic stem cells (ESCs) and conducting a time-resolved multiomics analysis. Oct4 depletion leads to an immediate loss of its binding to enhancers, accompanied by a decrease in mRNA synthesis from its target genes that are part of the transcriptional network that maintains pluripotency. Gradual decrease of Oct4 binding to enhancers does not immediately change the chromatin accessibility but reduces transcription of enhancers. Conversely, partial recovery of Oct4 expression results in a rapid increase in chromatin accessibility, whereas enhancer transcription does not fully recover. These results indicate different concentration-dependent activities of Oct4. Whereas normal ESC levels of Oct4 are required for transcription of pluripotency enhancers, low levels of Oct4 are sufficient to retain chromatin accessibility, likely together with other factors such as Sox2.


Asunto(s)
Cromatina , Células Madre Pluripotentes , Animales , Cromatina/metabolismo , Redes Reguladoras de Genes , Ratones , Células Madre Embrionarias de Ratones/metabolismo , Células Madre Pluripotentes/metabolismo , Transcripción Genética
10.
Mol Cells ; 45(4): 202-215, 2022 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-35014621

RESUMEN

The androgen receptor (AR) is an important therapeutic target for treating prostate cancer (PCa). Moreover, there is an increasing need for understanding the AR-independent progression of tumor cells such as neuroendocrine prostate cancer (NEPC). Menin, which is encoded by multiple endocrine neoplasia type 1 (MEN1), serves as a direct link between AR and the mixed-lineage leukemia (MLL) complex in PCa development by activating AR target genes through histone H3 lysine 4 methylation. Although menin is a critical component of AR signaling, its tumorigenic role in AR-independent PCa cells remains unknown. Here, we compared the role of menin in AR-positive and AR-negative PCa cells via RNAi-mediated or pharmacological inhibition of menin. We demonstrated that menin was involved in tumor cell growth and metastasis in PCa cells with low or deficient levels of AR. The inhibition of menin significantly diminished the growth of PCa cells and induced apoptosis, regardless of the presence of AR. Additionally, transcriptome analysis showed that the expression of many metastasis-associated genes was perturbed by menin inhibition in AR-negative DU145 cells. Furthermore, wound-healing assay results showed that menin promoted cell migration in AR-independent cellular contexts. Overall, these findings suggest a critical function of menin in tumorigenesis and provide a rationale for drug development against menin toward targeting high-risk metastatic PCa, especially those independent of AR.


Asunto(s)
Neoplasias de la Próstata , Receptores Androgénicos , Línea Celular Tumoral , Proliferación Celular , Humanos , Masculino , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Transducción de Señal , Factores de Transcripción
11.
Elife ; 102021 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-33770473

RESUMEN

Enhancer activity drives cell differentiation and cell fate determination, but it remains unclear how enhancers cooperate during these processes. Here we investigate enhancer cooperation during transdifferentiation of human leukemia B-cells to macrophages. Putative enhancers are established by binding of the pioneer factor C/EBPα followed by chromatin opening and enhancer RNA (eRNA) synthesis from H3K4-monomethylated regions. Using eRNA synthesis as a proxy for enhancer activity, we find that most putative enhancers cooperate in an additive way to regulate transcription of assigned target genes. However, transcription from 136 target genes depends exponentially on the summed activity of its putative paired enhancers, indicating that these enhancers cooperate synergistically. The target genes are cell type-specific, suggesting that enhancer synergy can contribute to cell fate determination. Enhancer synergy appears to depend on cell type-specific transcription factors, and such interacting enhancers are not predicted from occupancy or accessibility data that are used to detect superenhancers.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/genética , Diferenciación Celular/genética , Histonas/metabolismo , ARN/metabolismo , Transcripción Genética , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Cromatina/metabolismo , Elementos de Facilitación Genéticos , Humanos , Regiones Promotoras Genéticas , Células THP-1
12.
Cancers (Basel) ; 13(14)2021 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-34298635

RESUMEN

2-oxoglutarate and iron-dependent oxygenase domain-containing protein 1 (OGFOD1) expression is upregulated in a variety of cancers and has been related to poor prognosis. However, despite this significance to cancer progression, the precise oncogenic mechanism of OGFOD1 is not understood. We demonstrated that OGFOD1 plays a role in enhancing the transcriptional activity of RNA polymerase II in breast cancer cells. OGFOD1 directly binds to the C-terminal domain of RNA polymerase II to alter phosphorylation status. The elimination of OGFOD1 resulted in decreased tumor development. Additionally, cell cycle-dependent kinase 7 and cell cycle-dependent kinase 9, critical enzymes for activating RNA polymerase II, phosphorylated serine 256 of OGFOD1, whereas a non-phosphorylated mutant OGFOD1 failed to enhance transcriptional activation and tumor growth. Consequently, OGFOD1 helps promote tumor growth by enhancing RNA polymerase II, whereas simultaneous phosphorylation of OGFOD1 by CDK enzymes is essential in stimulating RNA polymerase II-mediated transcription both in vitro and in vivo, and expression of target genes.

13.
Biochem Biophys Res Commun ; 401(3): 327-32, 2010 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-20833138

RESUMEN

During myogenesis, transcriptional activities of two major myogenic factors, MyoD and myocyte enhancer factor 2 (Mef2) are regulated by histone modifications that switch on and off the target genes. However, the transition mechanism from repression to activation modes of histones has not been defined. Here we identify that lysine specific demethylase 1, (LSD1) is responsible for removing the repressive histone codes during C2C12 mouse myoblast differentiation. The potent role of LSD1 is suggested by the increment of its expression level during myogenic differentiation. Moreover, by performing co-immunoprecipitation and ChIP assay, physically interaction of LSD1 with MyoD and Mef2 on the target promoters was identified. Their interactions were resulted in upregulation of the transcription activities shown with increased luciferase activity. Interruption of demethylase activity of LSD1 using shRNA or chemical inhibitor, pargyline, treatment led to aberrant histone codes on myogenic promoters during skeletal muscle differentiation. We also demonstrate that inhibition of LSD1 impairs C2C12 mouse myoblast differentiation. Our results show for the first time the regulatory mechanism of myogenesis involving histone demethylase. Altogether, the present study suggests a de-repression model and expands the understanding on the dynamic regulation of chromatin during myogenesis.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Desarrollo de Músculos/genética , Músculo Esquelético/fisiología , Oxidorreductasas N-Desmetilantes/metabolismo , Regeneración/genética , Animales , Línea Celular , Histona Demetilasas , Factores de Transcripción MEF2 , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/enzimología , Proteína MioD/genética , Factores Reguladores Miogénicos/genética , Oxidorreductasas N-Desmetilantes/genética , Regiones Promotoras Genéticas
14.
Exp Mol Med ; 52(12): 2005-2019, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33311704

RESUMEN

Acetylation is the most studied histone acyl modification and has been recognized as a fundamental player in metabolic gene regulation, whereas other short-chain acyl modifications have only been recently identified, and little is known about their dynamics or molecular functions at the intersection of metabolism and epigenetic gene regulation. In this study, we aimed to understand the link between nonacetyl histone acyl modification, metabolic transcriptional regulation, and cellular adaptation. Using antibodies specific for butyrylated, propionylated, and crotonylated H3K23, we analyzed dynamic changes of H3K23 acylation upon various metabolic challenges. Here, we show that H3K23 modifications were highly responsive and reversibly regulated by nutrient availability. These modifications were commonly downregulated by the depletion of glucose and recovered based on glucose or fatty acid availability. Depletion of metabolic enzymes, namely, ATP citrate lyase, carnitine acetyltransferase, and acetyl-CoA synthetase, which are involved in Ac-CoA synthesis, resulted in global loss of H3K23 butyrylation, crotonylation, propionylation, and acetylation, with a profound impact on gene expression and cellular metabolic states. Our data indicate that Ac-CoA/CoA and central metabolic inputs are important for the maintenance of histone acylation. Additionally, genome-wide analysis revealed that acyl modifications are associated with gene activation. Our study shows that histone acylation acts as an immediate and reversible metabolic sensor enabling cellular adaptation to metabolic stress by reprogramming gene expression.


Asunto(s)
Adaptación Biológica , Metabolismo Energético , Histonas/metabolismo , Acetilcoenzima A/metabolismo , Acilación , Animales , Línea Celular , Cromatina/genética , Cromatina/metabolismo , Ensamble y Desensamble de Cromatina , Coenzima A/metabolismo , Epigénesis Genética , Cromatografía de Gases y Espectrometría de Masas , Regulación de la Expresión Génica , Glucosa/metabolismo , Humanos , Metaboloma , Metabolómica/métodos , Ratones , Estrés Fisiológico , Transcriptoma
15.
Nat Genet ; 52(7): 655-661, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32514124

RESUMEN

Three-dimensional organization of the genome is important for transcriptional regulation1-7. In mammals, CTCF and the cohesin complex create submegabase structures with elevated internal chromatin contact frequencies, called topologically associating domains (TADs)8-12. Although TADs can contribute to transcriptional regulation, ablation of TAD organization by disrupting CTCF or the cohesin complex causes modest gene expression changes13-16. In contrast, CTCF is required for cell cycle regulation17, embryonic development and formation of various adult cell types18. To uncouple the role of CTCF in cell-state transitions and cell proliferation, we studied the effect of CTCF depletion during the conversion of human leukemic B cells into macrophages with minimal cell division. CTCF depletion disrupts TAD organization but not cell transdifferentiation. In contrast, CTCF depletion in induced macrophages impairs the full-blown upregulation of inflammatory genes after exposure to endotoxin. Our results demonstrate that CTCF-dependent genome topology is not strictly required for a functional cell-fate conversion but facilitates a rapid and efficient response to an external stimulus.


Asunto(s)
Linfocitos B/fisiología , Factor de Unión a CCCTC/fisiología , Macrófagos/fisiología , Mielopoyesis/fisiología , Antígenos de Diferenciación/metabolismo , Factor de Unión a CCCTC/genética , Línea Celular Tumoral , Proliferación Celular/fisiología , Cromatina/fisiología , Regulación de la Expresión Génica , Humanos , Conformación Molecular , Mielopoyesis/genética , Conformación Proteica
16.
Exp Mol Med ; 49(10): e385, 2017 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-29026198

RESUMEN

The canonical Wnt pathway is critical for embryonic stem cell (ESC) pluripotency and aberrant control of ß-catenin leads to failure of exit from pluripotency and lineage commitments. Hence, maintaining the appropriate level of ß-catenin is important for the decision to commit to the appropriate lineage. However, how ß-catenin links to core transcription factors in ESCs remains elusive. C-terminal-binding protein (CtBP) in Drosophila is essential for Wnt-mediated target gene expression. In addition, Ctbp acts as an antagonist of ß-catenin/TCF activation in mammals. Recently, Ctbp2, a core Oct4-binding protein in ESCs, has been reported to play a key role in ESC pluripotency. However, the significance of the connection between Ctbp2 and ß-catenin with regard to ESC pluripotency remains elusive. Here, we demonstrate that C-terminal-binding protein 2 (Ctbp2) associates with major components of the ß-catenin destruction complex and limits the accessibility of ß-catenin to core transcription factors in undifferentiated ESCs. Ctbp2 knockdown leads to stabilization of ß-catenin, which then interacts with core pluripotency-maintaining factors that are occupied by Ctbp2, leading to incomplete exit from pluripotency. These findings suggest a suppressive function for Ctbp2 in reducing the protein level of ß-catenin, along with priming its position on core pluripotency genes to hinder ß-catenin deposition, which is central to commitment to the appropriate lineage.


Asunto(s)
Autorrenovación de las Células/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , beta Catenina/metabolismo , Oxidorreductasas de Alcohol , Animales , Sitios de Unión , Línea Celular , Proteínas Co-Represoras , Células Madre Embrionarias , Expresión Génica , Técnicas de Silenciamiento del Gen , Genes Reporteros , Ratones , Modelos Biológicos , Motivos de Nucleótidos , Unión Proteica , Estabilidad Proteica , ARN Interferente Pequeño/genética
17.
FEBS J ; 282(2): 361-71, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25367309

RESUMEN

ATP citrate lyase (ACLY) is a key enzyme that is involved in de novo lipogenesis by catalyzing conversion of cytosolic citrate into acetyl CoA and oxaloacetate. Up-regulation of ACLY in various types of tumors enhances fatty acid synthesis and supplies excess acetyl CoA for histone acetylation. However, there is evidence that its enzymatic activity alone is insufficient to explain ACLY silencing-mediated growth arrest in tumor cells. In this study, we found that ACLY knockdown in primary human cells triggers cellular senescence and activation of tumor suppressor p53. Provision of acetyl CoA to ACLY knockdown cells did not alleviate ACLY silencing-induced p53 activation, suggesting an independent role for ACLY activity. Instead, ACLY physically interacted with the catalytic subunit of AMP-activated protein kinase (AMPK) and inhibited AMPK activity. The activation of AMPK under ACLY knockdown conditions may lead to p53 activation, ultimately leading to cellular senescence. In cancer cells, ACLY silencing-induced p53 activation facilitated DNA damage-induced cell death. Taken together, our results suggest a novel function of ACLY in cellular senescence and tumorigenesis.


Asunto(s)
Proteínas Quinasas Activadas por AMP/genética , ATP Citrato (pro-S)-Liasa/genética , Senescencia Celular/genética , Neoplasias/genética , Proteína p53 Supresora de Tumor/biosíntesis , ATP Citrato (pro-S)-Liasa/metabolismo , Acetilcoenzima A/metabolismo , Animales , Carcinogénesis/genética , Citosol/metabolismo , Citosol/patología , Regulación Enzimológica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Neoplasias/patología , Ratas , Transducción de Señal/genética
18.
Biochem Biophys Res Commun ; 348(4): 1296-301, 2006 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-16920070

RESUMEN

G-rich is a Drosophila melanogaster selenoprotein, which is a homologue of human and mouse SelK. Subcellular localization analysis using GFP-tagged G-rich showed that G-rich was localized in the Golgi apparatus. The fusion protein was co-localized with the Golgi marker proteins but not with an endoplasmic reticulum (ER) marker protein in Drosophila SL2 cells. Bioinformatic analysis of G-rich suggests that this protein is either type II or type III transmembrane protein. To determine the type of transmembrane protein experimentally, GFP-G-rich in which GFP was tagged at the N-terminus of G-rich, or G-rich-GFP in which GFP was tagged at the C-terminus of G-rich, were expressed in SL2 cells. The tagged proteins were then digested with trypsin, and analyzed by Western blot analysis. The results showed that the C-terminus of the G-rich protein was exposed to the cytoplasm indicating it is a type III microsomal membrane protein. G-rich is the first selenoprotein identified in the Golgi apparatus.


Asunto(s)
Proteínas de Drosophila/análisis , Drosophila melanogaster/química , Aparato de Golgi/química , Proteínas de la Membrana/análisis , Selenoproteínas/análisis , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Línea Celular , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Drosophila melanogaster/citología , Drosophila melanogaster/genética , Membranas Intracelulares/química , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Datos de Secuencia Molecular , Selenoproteínas/química , Selenoproteínas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA