Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
1.
EMBO J ; 40(16): e102509, 2021 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-34155658

RESUMEN

The SAGA coactivator complex is essential for eukaryotic transcription and comprises four distinct modules, one of which contains the ubiquitin hydrolase USP22. In yeast, the USP22 ortholog deubiquitylates H2B, resulting in Pol II Ser2 phosphorylation and subsequent transcriptional elongation. In contrast to this H2B-associated role in transcription, we report here that human USP22 contributes to the early stages of stimulus-responsive transcription, where USP22 is required for pre-initiation complex (PIC) stability. Specifically, USP22 maintains long-range enhancer-promoter contacts and controls loading of Mediator tail and general transcription factors (GTFs) onto promoters, with Mediator core recruitment being USP22-independent. In addition, we identify Mediator tail subunits MED16 and MED24 and the Pol II subunit RBP1 as potential non-histone substrates of USP22. Overall, these findings define a role for human SAGA within the earliest steps of transcription.


Asunto(s)
Ubiquitina Tiolesterasa/genética , Apoptosis , Estrés del Retículo Endoplásmico/genética , Células HCT116 , Humanos , Complejo Mediador/genética , Regiones Promotoras Genéticas , ARN Polimerasa II , Transcripción Genética
2.
Mol Cell ; 66(2): 247-257.e5, 2017 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-28410996

RESUMEN

Recruitment of transcription factors (TFs) to repressed genes in euchromatin is essential to activate new transcriptional programs during cell differentiation. However, recruitment of all TFs, including pioneer factors, is impeded by condensed H3K27me3-containing chromatin. Single-cell and gene-specific analyses revealed that, during the first hours of induction of differentiation of mammalian embryonic stem cells (ESCs), accumulation of the repressive histone mark H3K27me3 is delayed after DNA replication, indicative of a decondensed chromatin structure in all regions of the replicating genome. This delay provides a critical "window of opportunity" for recruitment of lineage-specific TFs to DNA. Increasing the levels of post-replicative H3K27me3 or preventing S phase entry inhibited recruitment of new TFs to DNA and significantly blocked cell differentiation. These findings suggest that recruitment of lineage-specifying TFs occurs soon after replication and is facilitated by a decondensed chromatin structure. This insight may explain the developmental plasticity of stem cells and facilitate their exploitation for therapeutic purposes.


Asunto(s)
Diferenciación Celular , Linaje de la Célula , Ensamble y Desensamble de Cromatina , Cromatina/metabolismo , Replicación del ADN , ADN/biosíntesis , Células Madre Embrionarias/metabolismo , Histonas/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética , Animales , Sitios de Unión , Plasticidad de la Célula , Cromatina/química , ADN/química , ADN/genética , Metilación de ADN , Regulación del Desarrollo de la Expresión Génica , Histona Demetilasas/metabolismo , Histonas/química , Humanos , Metilación , Ratones , Proteínas Nucleares/metabolismo , Conformación de Ácido Nucleico , Unión Proteica , Relación Estructura-Actividad , Factores de Tiempo , Factores de Transcripción/genética
3.
Mol Cell ; 50(6): 781-2, 2013 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-23806333

RESUMEN

In this issue, Trinidad et al. (2013) show that CCT/TRiC is a chaperone required for p53 folding, thus providing another layer of regulation of p53 function, with implications for cancer therapeutics targeting the p53 pathway.


Asunto(s)
Chaperoninas del Grupo II/metabolismo , Pliegue de Proteína , Proteína p53 Supresora de Tumor/metabolismo , Humanos
4.
Proc Natl Acad Sci U S A ; 115(40): E9298-E9307, 2018 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-30224477

RESUMEN

Overexpression of the deubiquitylase ubiquitin-specific peptidase 22 (USP22) is a marker of aggressive cancer phenotypes like metastasis, therapy resistance, and poor survival. Functionally, this overexpression of USP22 actively contributes to tumorigenesis, as USP22 depletion blocks cancer cell cycle progression in vitro, and inhibits tumor progression in animal models of lung, breast, bladder, ovarian, and liver cancer, among others. Current models suggest that USP22 mediates these biological effects via its role in epigenetic regulation as a subunit of the Spt-Ada-Gcn5-acetyltransferase (SAGA) transcriptional cofactor complex. Challenging the dogma, we report here a nontranscriptional role for USP22 via a direct effect on the core cell cycle machinery: that is, the deubiquitylation of the G1 cyclin D1 (CCND1). Deubiquitylation by USP22 protects CCND1 from proteasome-mediated degradation and occurs separately from the canonical phosphorylation/ubiquitylation mechanism previously shown to regulate CCND1 stability. We demonstrate that control of CCND1 is a key mechanism by which USP22 mediates its known role in cell cycle progression. Finally, USP22 and CCND1 levels correlate in patient lung and colorectal cancer samples and our preclinical studies indicate that targeting USP22 in combination with CDK inhibitors may offer an approach for treating cancer patients whose tumors exhibit elevated CCND1.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Ciclina D1/metabolismo , Epigénesis Genética , Fase G1 , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares/metabolismo , Proteolisis , Tioléster Hidrolasas/metabolismo , Ubiquitinación , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Ciclina D1/genética , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Células MCF-7 , Estabilidad Proteica , Tioléster Hidrolasas/genética , Ubiquitina Tiolesterasa
5.
Carcinogenesis ; 41(1): 67-77, 2020 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-31067569

RESUMEN

Lung cancer is the leading cause of cancer-related deaths in the USA, and alterations in the tumor suppressor gene TP53 are the most frequent somatic mutation among all histologic subtypes of lung cancer. Mutations in TP53 frequently result in a protein that exhibits not only loss of tumor suppressor capability but also oncogenic gain-of-function (GOF). The canonical p53 hotspot mutants R175H and R273H, for example, confer upon tumors a metastatic phenotype in murine models of mutant p53. To the best of our knowledge, GOF phenotypes of the less often studied V157, R158 and A159 mutants-which occur with higher frequency in lung cancer compared with other solid tumors-have not been defined. In this study, we aimed to define whether the lung mutants are simply equivalent to full loss of the p53 locus, or whether they additionally acquire the ability to drive new downstream effector pathways. Using a publicly available human lung cancer dataset, we characterized patients with V157, R158 and A159 p53 mutations. In addition, we show here that cell lines with mutant p53-V157F, p53-R158L and p53-R158P exhibit a loss of expression of canonical wild-type p53 target genes. Furthermore, these lung-enriched p53 mutants regulate genes not previously linked to p53 function including PLAU. Paradoxically, mutant p53 represses genes associated with increased cell viability, migration and invasion. These findings collectively represent the first demonstration that lung-enriched p53 mutations at V157 and R158 regulate a novel transcriptome in human lung cancer cells and may confer de novo function.


Asunto(s)
Carcinogénesis/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares/genética , Transcriptoma/genética , Proteína p53 Supresora de Tumor/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Secuenciación de Inmunoprecipitación de Cromatina , Conjuntos de Datos como Asunto , Técnicas de Silenciamiento del Gen , Humanos , Proteínas de la Membrana/metabolismo , Mutación , Polimorfismo de Nucleótido Simple , ARN Interferente Pequeño/metabolismo , RNA-Seq
6.
EMBO J ; 35(2): 193-207, 2016 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-26658110

RESUMEN

Telomeres and tumor suppressor protein TP53 (p53) function in genome protection, but a direct role of p53 at telomeres has not yet been described. Here, we have identified non-canonical p53-binding sites within the human subtelomeres that suppress the accumulation of DNA damage at telomeric repeat DNA. These non-canonical subtelomeric p53-binding sites conferred transcription enhancer-like functions that include an increase in local histone H3K9 and H3K27 acetylation and stimulation of subtelomeric transcripts, including telomere repeat-containing RNA (TERRA). p53 suppressed formation of telomere-associated γH2AX and prevented telomere DNA degradation in response to DNA damage stress. Our findings indicate that p53 provides a direct chromatin-associated protection to human telomeres, as well as other fragile genomic sites. We propose that p53-associated chromatin modifications enhance local DNA repair or protection to provide a previously unrecognized tumor suppressor function of p53.


Asunto(s)
Proteínas Portadoras/metabolismo , Daño del ADN/genética , Telómero/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Portadoras/genética , Células HCT116 , Humanos , Unión Proteica , Telómero/genética , Proteína p53 Supresora de Tumor/genética
7.
BMC Cancer ; 19(1): 258, 2019 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-30902071

RESUMEN

BACKGROUND: The oncoprotein MYC has the dual capacity to drive cell cycle progression or induce apoptosis, depending on the cellular context. BAG1 was previously identified as a transcriptional target of MYC that functions as a critical determinant of this cell fate decision. The BAG1 protein is expressed as multiple isoforms, each having an array of distinct biochemical functions; however, the specific effector function of BAG1 that directs MYC-dependent cell survival has not been defined. METHODS: In our studies the human osteosarcoma line U2OS expressing a conditional MYC-ER allele was used to induce oncogenic levels of MYC. We interrogated MYC-driven survival processes by modifying BAG1 protein expression. The function of the separate BAG1 isoforms was investigated by depleting cells of endogenous BAG1 and reintroducing the distinct isoforms. Flow cytometry and immunoblot assays were performed to analyze the effect of specific BAG1 isoforms on MYC-dependent apoptosis. These experiments were repeated to determine the role of the HSP70 chaperone complex in BAG1 survival processes. Finally, a proteomic approach was used to identify a set of specific pro-survival proteins controlled by the HSP70/BAG1 complex. RESULTS: Loss of BAG1 resulted in robust MYC-induced apoptosis. Expression of the larger isoforms of BAG1, BAG1L and BAG1M, were insufficient to rescue survival in cells with oncogenic levels of MYC. Alternatively, reintroduction of BAG1S significantly reduced the level of apoptosis. Manipulation of the BAG1S interaction with HSP70 revealed that BAG1S provides its pro-survival function by serving as a cofactor for the HSP70 chaperone complex. Via a proteomic approach we identified and classified a set of pro-survival proteins controlled by this HSP70/BAG1 chaperone complex that contribute to the BAG1 anti-apoptotic phenotype. CONCLUSIONS: The small isoform of BAG1, BAG1S, in cooperation with the HSP70 chaperone complex, selectively mediates cell survival in MYC overexpressing tumor cells. We identified a set of specific pro-survival clients controlled by the HSP70/BAG1S chaperone complex. These clients define new nodes that could be therapeutically targeted to disrupt the survival of tumor cells driven by MYC activation. With MYC overexpression occurring in most human cancers, this introduces new strategies for cancer treatment.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Neoplasias Óseas/patología , Proteínas de Unión al ADN/metabolismo , Proteínas HSP70 de Choque Térmico/metabolismo , Osteosarcoma/patología , Factores de Transcripción/metabolismo , Apoptosis , Línea Celular Tumoral , Supervivencia Celular , Humanos , Isoformas de Proteínas/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo
8.
Mol Cell ; 42(5): 584-96, 2011 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-21658600

RESUMEN

Activation of p53 by DNA damage results in either cell-cycle arrest, allowing DNA repair and cell survival, or induction of apoptosis. As these opposite outcomes are both mediated by p53 stabilization, additional mechanisms to determine this decision must exist. Here, we show that glycogen synthase kinase-3 (GSK-3) is required for the p53-mediated induction of the proapoptotic BH3 only-protein PUMA, an essential mediator of p53-induced apoptosis. Inhibition of GSK-3 protected from cell death induced by DNA damage and promoted increased long-term cell survival. We demonstrate that GSK-3 phosphorylates serine 86 of the p53-acetyltransferase Tip60. A Tip60(S86A) mutant was less active to induce p53 K120 acetylation, histone 4 acetylation, and expression of PUMA. Our data suggest that GSK-3 mediated Tip60S86 phosphorylation provides a link between PI3K signaling and the choice for or against apoptosis induction by p53.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Apoptosis/fisiología , Glucógeno Sintasa Quinasa 3/fisiología , Histona Acetiltransferasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteína p53 Supresora de Tumor/fisiología , Acetilación , Línea Celular Tumoral , Daño del ADN , Glucógeno Sintasa Quinasa 3/metabolismo , Histona Acetiltransferasas/química , Histona Acetiltransferasas/fisiología , Humanos , Lisina Acetiltransferasa 5 , Fosforilación , Regiones Promotoras Genéticas
9.
FASEB J ; 31(3): 1165-1178, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27940549

RESUMEN

The human telomerase reverse transcriptase (hTERT) gene is repressed in most somatic cells, whereas the expression of the mouse mTert gene is widely detected. To understand the mechanisms of this human-specific repression, we constructed bacterial artificial chromosome (BAC) reporters using human and mouse genomic DNAs encompassing the TERT genes and neighboring loci. Upon chromosomal integration, the hTERT, but not the mTert, reporter was stringently repressed in telomerase-negative human cells in a histone deacetylase (HDAC)-dependent manner, replicating the expression of their respective endogenous genes. In chimeric BACs, the mTert promoter became strongly repressed in the human genomic context, but the hTERT promoter was highly active in the mouse genomic context. Furthermore, an unrelated herpes simplex virus-thymidine kinase (HSV-TK) promoter was strongly repressed in the human, but not in the mouse, genomic context. These results demonstrated that the repression of hTERT gene was dictated by distal elements and its chromatin environment. This repression depended on class I HDACs and involved multiple corepressor complexes, including HDAC1/2-containing Sin3B, nucleosome remodeling and histone deacetylase (NuRD), and corepressor of RE1 silencing transcription factor (CoREST) complexes. Together, our data indicate that the lack of telomerase expression in most human somatic cells results from its repressive genomic environment, providing new insight into the mechanism of long-recognized differential telomerase regulation in mammalian species.-Cheng, D., Zhao, Y., Wang, S., Zhang, F., Russo, M., McMahon, S. B., Zhu, J. Repression of telomerase gene promoter requires human-specific genomic context and is mediated by multiple HDAC1-containing corepressor complexes.


Asunto(s)
Genoma Humano , Histona Desacetilasa 1/metabolismo , Regiones Promotoras Genéticas , Telomerasa/genética , Animales , Ensamble y Desensamble de Cromatina , Cromosomas Artificiales Bacterianos/genética , Células HEK293 , Histona Desacetilasa 1/genética , Humanos , Células MCF-7 , Ratones , Especificidad de la Especie , Telomerasa/metabolismo
10.
Mol Cell ; 36(2): 174-5, 2009 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-19854127

RESUMEN

Dynamic lysine acetylation regulates proteins involved in diverse cellular processes, with individual enzymes often acetylating multiple substrates. Here, Li et al. (2009) show that the substrate specificity of hMOF/MYST1/KAT8 is controlled by differential interaction with two mutually exclusive partners.


Asunto(s)
Histona Acetiltransferasas/metabolismo , Acetilación , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Histonas/metabolismo , Humanos , Modelos Biológicos , Complejos Multiproteicos/química , Complejos Multiproteicos/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
11.
EMBO J ; 31(1): 58-70, 2012 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-22020126

RESUMEN

The MYST protein lysine acetyltransferases are evolutionarily conserved throughout eukaryotes and acetylate proteins to regulate diverse biological processes including gene regulation, DNA repair, cell-cycle regulation, stem cell homeostasis and development. Here, we demonstrate that MYST protein acetyltransferase activity requires active site lysine autoacetylation. The X-ray crystal structures of yeast Esa1 (yEsa1/KAT5) bound to a bisubstrate H4K16CoA inhibitor and human MOF (hMOF/KAT8/MYST1) reveal that they are autoacetylated at a strictly conserved lysine residue in MYST proteins (yEsa1-K262 and hMOF-K274) in the enzyme active site. The structure of hMOF also shows partial occupancy of K274 in the unacetylated form, revealing that the side chain reorients to a position that engages the catalytic glutamate residue and would block cognate protein substrate binding. Consistent with the structural findings, we present mass spectrometry data and biochemical experiments to demonstrate that this lysine autoacetylation on yEsa1, hMOF and its yeast orthologue, ySas2 (KAT8) occurs in solution and is required for acetylation and protein substrate binding in vitro. We also show that this autoacetylation occurs in vivo and is required for the cellular functions of these MYST proteins. These findings provide an avenue for the autoposttranslational regulation of MYST proteins that is distinct from other acetyltransferases but draws similarities to the phosphoregulation of protein kinases.


Asunto(s)
Histona Acetiltransferasas/metabolismo , Lisina/genética , Acetilación , Sitios de Unión , Dominio Catalítico , Cristalografía por Rayos X , Histona Acetiltransferasas/genética , Histonas/metabolismo , Humanos , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
12.
J Biol Chem ; 288(33): 24234-46, 2013 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-23760504

RESUMEN

Pluripotent embryonic stem cells (ESCs) undergo self-renewal until stimulated to differentiate along specific lineage pathways. Many of the transcriptional networks that drive reprogramming of a self-renewing ESC to a differentiating cell have been identified. However, fundamental questions remain unanswered about the epigenetic programs that control these changes in gene expression. Here we report that the histone ubiquitin hydrolase ubiquitin-specific protease 22 (USP22) is a critical epigenetic modifier that controls this transition from self-renewal to differentiation. USP22 is induced as ESCs differentiate and is necessary for differentiation into all three germ layers. We further report that USP22 is a transcriptional repressor of the locus encoding the core pluripotency factor sex-determining region Y-box 2 (SOX2) in ESCs, and this repression is required for efficient differentiation. USP22 occupies the Sox2 promoter and hydrolyzes monoubiquitin from ubiquitylated histone H2B and blocks transcription of the Sox2 locus. Our study reveals an epigenetic mechanism that represses the core pluripotency transcriptional network in ESCs, allowing ESCs to transition from a state of self-renewal into lineage-specific differentiation programs.


Asunto(s)
Diferenciación Celular/genética , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Endopeptidasas/metabolismo , Epigénesis Genética , Factores de Transcripción SOXB1/genética , Transcripción Genética , Proteasas Ubiquitina-Específicas/metabolismo , Animales , Línea Celular , Proliferación Celular , Endopeptidasas/genética , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Sitios Genéticos/genética , Histonas/metabolismo , Ratones , Fenotipo , Células Madre Pluripotentes/metabolismo , Unión Proteica/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores de Transcripción SOXB1/metabolismo , Sirtuina 1/metabolismo , Ubiquitina Tiolesterasa , Proteasas Ubiquitina-Específicas/genética , Ubiquitinación/genética
13.
Trends Biochem Sci ; 34(11): 571-8, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19819149

RESUMEN

Protein phosphorylation is regulated dynamically in eukaryotic cells via modulation of the enzymatic activity of kinases and phosphatases. Like phosphorylation, acetylation has emerged as a critical regulatory protein modification that is altered dynamically in response to diverse cellular cues. Moreover, acetyltransferases and deacetylases are tightly linked to cellular signaling pathways. Recent studies provide clues about the mechanisms utilized to regulate acetyltransferases and deacetylases. The therapeutic value of deacetylase inhibitors suggests that understanding acetylation pathways will directly impact our ability to rationally target these enzymes in patients. Recently discovered mechanisms that directly regulate the catalytic activity of acetyltransferases and deacetylases provide exciting new insights about these enzymes.


Asunto(s)
Acetiltransferasas/metabolismo , Histona Desacetilasas/metabolismo , Transducción de Señal , Acetilación , Animales , Catálisis , Histonas/metabolismo , Humanos , Modelos Biológicos , Proteína p53 Supresora de Tumor/metabolismo
14.
Biochim Biophys Acta ; 1819(9-10): 1075-9, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22766037

RESUMEN

Eukaryotes control nearly every cellular process in part by modulating the transcription of genes encoded by their nuclear genome. However, these cells are faced with the added complexity of possessing a second genome, within the mitochondria, which encodes critical components of several essential processes, including energy metabolism and macromolecule biosynthesis. As these cellular processes require gene products encoded by both genomes, cells have adopted strategies for linking mitochondrial gene expression to nuclear gene expression and other dynamic cellular events. Here we discuss examples of several mechanisms that have been identified, by which eukaryotic cells link extramitochondrial signals to dynamic alterations in mitochondrial transcription. This article is part of a Special Issue entitled: Mitochondrial Gene Expression.


Asunto(s)
Adaptación Biológica/genética , Mitocondrias , Proteínas Nucleares/metabolismo , Transcripción Genética , ADN Mitocondrial/genética , ADN Mitocondrial/metabolismo , ARN Polimerasas Dirigidas por ADN/genética , ARN Polimerasas Dirigidas por ADN/metabolismo , Regulación de la Expresión Génica , Humanos , Mitocondrias/genética , Mitocondrias/fisiología , Proteínas Nucleares/genética , Proteína Oncogénica p55(v-myc)/genética , Proteína Oncogénica p55(v-myc)/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Esteroides/metabolismo
15.
J Biol Chem ; 286(6): 4264-70, 2011 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-21148320

RESUMEN

In unstressed cells, the p53 tumor suppressor is highly unstable. DNA damage and other forms of cellular stress rapidly stabilize and activate p53. This process is regulated by a complex array of post-translational modifications that are dynamically deposited onto p53. Recent studies show that these modifications orchestrate p53-mediated processes such as cell cycle arrest and apoptosis. Cancer cells carry inherent genetic damage, but avoid arrest and apoptosis by inactivating p53. Defining the enzymatic machinery that regulates the stress-induced modification of p53 at single-residue resolution is critical to our understanding of the biochemical mechanisms that control this critical tumor suppressor. Specifically, acetylation of p53 at lysine 120, a DNA-binding domain residue mutated in human cancer, is essential for triggering apoptosis. Given the oncogenic properties of deacetylases and the success of deacetylase inhibitors as anticancer agents, we investigated the regulation of Lys(120) deacetylation using pharmacologic and genetic approaches. This analysis revealed that histone deacetylase 1 is predominantly responsible for the deacetylation of Lys(120). Furthermore, treatment with the clinical-grade histone deacetylase inhibitor entinostat enhances Lys(120) acetylation, an event that is mechanistically linked to its apoptotic effect. These data expand our understanding of the mechanisms controlling p53 function and suggest that regulation of p53 modification status at single-residue resolution by targeted therapeutics can selectively alter p53 pathway function. This knowledge may impact the rational application of deacetylase inhibitors in the treatment of human cancer.


Asunto(s)
Apoptosis , Proteína p53 Supresora de Tumor/metabolismo , Acetilación/efectos de los fármacos , Benzamidas/farmacología , Línea Celular Tumoral , Daño del ADN/efectos de los fármacos , Daño del ADN/genética , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Mutación Missense , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/metabolismo , Estructura Terciaria de Proteína , Piridinas/farmacología , Proteína p53 Supresora de Tumor/genética
16.
Methods ; 54(3): 339-47, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21513801

RESUMEN

While the post-translational modification of histones by the addition of ubiquitin was discovered decades ago, it has only recently been appreciated that the dynamic regulation of histone ubiquitylation patterns is an important mechanism for controlling a variety of biological processes. The processes include transcription, the recognition and repair of genomic damage and DNA replication, among others. Enzymes that catalyze the addition of ubiquitin to histones, such as the polycomb family, have been well-studied. In contrast, the enzymes that remove ubiquitin from histones are less well understood. The assay strategies described here provide a platform for the thorough in vitro and in vivo analysis of histone deubiquitylation. In some cases, these poorly characterized enzymes are likely to provide new opportunities for therapeutic targeting and a detailed understanding of their biochemical and biological activities is a prerequisite to these clinical advances.


Asunto(s)
Endopeptidasas/química , Pruebas de Enzimas/métodos , Histonas/química , Proteínas Recombinantes/química , Proteínas Ubiquitinadas/química , Animales , Baculoviridae/genética , Línea Celular , Cromatografía en Gel , Endopeptidasas/aislamiento & purificación , Precipitación Fraccionada , Vectores Genéticos , Humanos , Nucleosomas/química , Replegamiento Proteico , Proteínas Recombinantes/aislamiento & purificación , Levaduras
17.
Exp Cell Res ; 317(4): 488-95, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21111732

RESUMEN

MicroRNA145 (miR145), a tumor suppressor miR, has been reported to inhibit growth of human cancer cells, to induce differentiation and to cause apoptosis, all conditions that result in growth arrest. In order to clarify the functional effects of miR145, we have investigated its expression in diverse conditions and different cell lines. Our results show that miR145 levels definitely increase in differentiating cells and also in growth-arrested cells, even in the absence of differentiation. Increased expression during differentiation sometimes occurs as a late event, suggesting that miR145 could be required either early or late during the differentiation process.


Asunto(s)
Diferenciación Celular/genética , Inhibidores de Crecimiento/análisis , MicroARNs/análisis , Regulación hacia Arriba , Apoptosis , Butiratos/farmacología , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Ciclo Celular , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Inhibidores de Crecimiento/farmacología , Humanos , Cloruro de Litio/farmacología , MicroARNs/efectos de los fármacos , Proteínas Proto-Oncogénicas c-myc , Tretinoina/farmacología , Regulación hacia Arriba/efectos de los fármacos
18.
Cell Mol Gastroenterol Hepatol ; 13(4): 1276-1296, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34954189

RESUMEN

BACKGROUND & AIMS: Sporadic colorectal cancers arise from initiating mutations in APC, producing oncogenic ß-catenin/TCF-dependent transcriptional reprogramming. Similarly, the tumor suppressor axis regulated by the intestinal epithelial receptor GUCY2C is among the earliest pathways silenced in tumorigenesis. Retention of the receptor, but loss of its paracrine ligands, guanylin and uroguanylin, is an evolutionarily conserved feature of colorectal tumors, arising in the earliest dysplastic lesions. Here, we examined a mechanism of GUCY2C ligand transcriptional silencing by ß-catenin/TCF signaling. METHODS: We performed RNA sequencing analysis of 4 unique conditional human colon cancer cell models of ß-catenin/TCF signaling to map the core Wnt-transcriptional program. We then performed a comparative analysis of orthogonal approaches, including luciferase reporters, chromatin immunoprecipitation sequencing, CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats) knockout, and CRISPR epigenome editing, which were cross-validated with human tissue chromatin immunoprecipitation sequencing datasets, to identify functional gene enhancers mediating GUCY2C ligand loss. RESULTS: RNA sequencing analyses reveal the GUCY2C hormones as 2 of the most sensitive targets of ß-catenin/TCF signaling, reflecting transcriptional repression. The GUCY2C hormones share an insulated genomic locus containing a novel locus control region upstream of the guanylin promoter that mediates the coordinated silencing of both genes. Targeting this region with CRISPR epigenome editing reconstituted GUCY2C ligand expression, overcoming gene inactivation by mutant ß-catenin/TCF signaling. CONCLUSIONS: These studies reveal DNA elements regulating corepression of GUCY2C ligand transcription by ß-catenin/TCF signaling, reflecting a novel pathophysiological step in tumorigenesis. They offer unique genomic strategies that could reestablish hormone expression in the context of canonical oncogenic mutations to reconstitute the GUCY2C axis and oppose transformation.


Asunto(s)
Neoplasias Colorrectales , beta Catenina , Carcinogénesis/genética , Cateninas/genética , Cateninas/metabolismo , Neoplasias Colorrectales/patología , Humanos , Ligandos , Región de Control de Posición , Receptores de Enterotoxina/genética , Receptores de Enterotoxina/metabolismo , Factores de Transcripción TCF/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
19.
Proc Natl Acad Sci U S A ; 105(48): 18782-7, 2008 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-19033189

RESUMEN

Mammalian cells fuel their growth and proliferation through the catabolism of two main substrates: glucose and glutamine. Most of the remaining metabolites taken up by proliferating cells are not catabolized, but instead are used as building blocks during anabolic macromolecular synthesis. Investigations of phosphoinositol 3-kinase (PI3K) and its downstream effector AKT have confirmed that these oncogenes play a direct role in stimulating glucose uptake and metabolism, rendering the transformed cell addicted to glucose for the maintenance of survival. In contrast, less is known about the regulation of glutamine uptake and metabolism. Here, we report that the transcriptional regulatory properties of the oncogene Myc coordinate the expression of genes necessary for cells to engage in glutamine catabolism that exceeds the cellular requirement for protein and nucleotide biosynthesis. A consequence of this Myc-dependent glutaminolysis is the reprogramming of mitochondrial metabolism to depend on glutamine catabolism to sustain cellular viability and TCA cycle anapleurosis. The ability of Myc-expressing cells to engage in glutaminolysis does not depend on concomitant activation of PI3K or AKT. The stimulation of mitochondrial glutamine metabolism resulted in reduced glucose carbon entering the TCA cycle and a decreased contribution of glucose to the mitochondrial-dependent synthesis of phospholipids. These data suggest that oncogenic levels of Myc induce a transcriptional program that promotes glutaminolysis and triggers cellular addiction to glutamine as a bioenergetic substrate.


Asunto(s)
Metabolismo Energético/fisiología , Glutamina/metabolismo , Mitocondrias/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Transcripción Genética , Animales , Línea Celular , Fibroblastos/citología , Fibroblastos/metabolismo , Regulación de la Expresión Génica , Glucosa/metabolismo , Humanos , Ratones , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Transducción de Señal/fisiología
20.
Nat Commun ; 12(1): 484, 2021 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-33473123

RESUMEN

The tumor suppressor p53 integrates stress response pathways by selectively engaging one of several potential transcriptomes, thereby triggering cell fate decisions (e.g., cell cycle arrest, apoptosis). Foundational to this process is the binding of tetrameric p53 to 20-bp response elements (REs) in the genome (RRRCWWGYYYN0-13RRRCWWGYYY). In general, REs at cell cycle arrest targets (e.g. p21) are of higher affinity than those at apoptosis targets (e.g., BAX). However, the RE sequence code underlying selectivity remains undeciphered. Here, we identify molecular mechanisms mediating p53 binding to high- and low-affinity REs by showing that key determinants of the code are embedded in the DNA shape. We further demonstrate that differences in minor/major groove widths, encoded by G/C or A/T bp content at positions 3, 8, 13, and 18 in the RE, determine distinct p53 DNA-binding modes by inducing different Arg248 and Lys120 conformations and interactions. The predictive capacity of this code was confirmed in vivo using genome editing at the BAX RE to interconvert the DNA-binding modes, transcription pattern, and cell fate outcome.


Asunto(s)
Diferenciación Celular/genética , Diferenciación Celular/fisiología , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Apoptosis/genética , Ciclo Celular , Puntos de Control del Ciclo Celular , Línea Celular , ADN/química , Proteínas de Unión al ADN , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Modelos Moleculares , Conformación Molecular , Unión Proteica/genética , Elementos de Respuesta
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA