Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Nat Rev Mol Cell Biol ; 17(5): 308-21, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26956196

RESUMEN

Mitochondrial dysfunction is a hallmark of ageing, and mitochondrial maintenance may lead to increased healthspan. Emerging evidence suggests a crucial role for signalling from the nucleus to mitochondria (NM signalling) in regulating mitochondrial function and ageing. An important initiator of NM signalling is nuclear DNA damage, which accumulates with age and may contribute to the development of age-associated diseases. DNA damage-dependent NM signalling constitutes a network that includes nuclear sirtuins and controls genomic stability and mitochondrial integrity. Pharmacological modulation of NM signalling is a promising novel approach for the prevention and treatment of age-associated diseases.


Asunto(s)
Envejecimiento , Daño del ADN , Mitocondrias/fisiología , Animales , Apoptosis , Núcleo Celular/genética , Reparación del ADN , Inestabilidad Genómica , Humanos , Mitofagia , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal
2.
Cell ; 148(3): 387-9, 2012 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-22304906

RESUMEN

Despite resveratrol's well-documented health benefits, its mechanism of action remains controversial. In particular, the direct molecular target of resveratrol has been elusive. Park et al. now show that resveratrol directly inhibits cAMP-dependent phosphodiesterases, triggering a cascade of events that converge on the important energy-sensing metabolic regulators AMPK, SIRT1, and PGC-1α.

3.
Nature ; 590(7846): 504-508, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33536620

RESUMEN

Amplification of chromosomal region 8p11-12 is a common genetic alteration that has been implicated in the aetiology of lung squamous cell carcinoma (LUSC)1-3. The FGFR1 gene is the main candidate driver of tumorigenesis within this region4. However, clinical trials evaluating FGFR1 inhibition as a targeted therapy have been unsuccessful5. Here we identify the histone H3 lysine 36 (H3K36) methyltransferase NSD3, the gene for which is located in the 8p11-12 amplicon, as a key regulator of LUSC tumorigenesis. In contrast to other 8p11-12 candidate LUSC drivers, increased expression of NSD3 correlated strongly with its gene amplification. Ablation of NSD3, but not of FGFR1, attenuated tumour growth and extended survival in a mouse model of LUSC. We identify an LUSC-associated variant NSD3(T1232A) that shows increased catalytic activity for dimethylation of H3K36 (H3K36me2) in vitro and in vivo. Structural dynamic analyses revealed that the T1232A substitution elicited localized mobility changes throughout the catalytic domain of NSD3 to relieve auto-inhibition and to increase accessibility of the H3 substrate. Expression of NSD3(T1232A) in vivo accelerated tumorigenesis and decreased overall survival in mouse models of LUSC. Pathological generation of H3K36me2 by NSD3(T1232A) reprograms the chromatin landscape to promote oncogenic gene expression signatures. Furthermore, NSD3, in a manner dependent on its catalytic activity, promoted transformation in human tracheobronchial cells and growth of xenografted human LUSC cell lines with amplification of 8p11-12. Depletion of NSD3 in patient-derived xenografts from primary LUSCs containing NSD3 amplification or the NSD3(T1232A)-encoding variant attenuated neoplastic growth in mice. Finally, NSD3-regulated LUSC-derived xenografts were hypersensitive to bromodomain inhibition. Thus, our work identifies NSD3 as a principal 8p11-12 amplicon-associated oncogenic driver in LUSC, and suggests that NSD3-dependency renders LUSC therapeutically vulnerable to bromodomain inhibition.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/química , Histonas/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Proteínas Nucleares/metabolismo , Animales , Biocatálisis , Carcinogénesis/genética , Carcinoma de Células Escamosas/genética , Femenino , N-Metiltransferasa de Histona-Lisina/deficiencia , N-Metiltransferasa de Histona-Lisina/genética , Humanos , Neoplasias Pulmonares/genética , Masculino , Metilación , Ratones , Modelos Moleculares , Mutación , Proteínas Nucleares/deficiencia , Proteínas Nucleares/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/deficiencia , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Cell ; 136(1): 62-74, 2009 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-19135889

RESUMEN

Members of the sirtuin (SIRT) family of NAD-dependent deacetylases promote longevity in multiple organisms. Deficiency of mammalian SIRT6 leads to shortened life span and an aging-like phenotype in mice, but the underlying molecular mechanisms are unclear. Here we show that SIRT6 functions at chromatin to attenuate NF-kappaB signaling. SIRT6 interacts with the NF-kappaB RELA subunit and deacetylates histone H3 lysine 9 (H3K9) at NF-kappaB target gene promoters. In SIRT6-deficient cells, hyperacetylation of H3K9 at these target promoters is associated with increased RELA promoter occupancy and enhanced NF-kappaB-dependent modulation of gene expression, apoptosis, and cellular senescence. Computational genomics analyses revealed increased activity of NF-kappaB-driven gene expression programs in multiple Sirt6-deficient tissues in vivo. Moreover, haploinsufficiency of RelA rescues the early lethality and degenerative syndrome of Sirt6-deficient mice. We propose that SIRT6 attenuates NF-kappaB signaling via H3K9 deacetylation at chromatin, and hyperactive NF-kappaB signaling may contribute to premature and normal aging.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , FN-kappa B/metabolismo , Sirtuinas/metabolismo , Factor de Transcripción ReIA/metabolismo , Acetilación , Animales , Línea Celular , Cromatina/metabolismo , Cruzamientos Genéticos , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Humanos , Longevidad/genética , Ratones , FN-kappa B/genética , Regiones Promotoras Genéticas , Sirtuinas/genética , Factor de Transcripción ReIA/genética
5.
Nat Immunol ; 12(1): 29-36, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21131967

RESUMEN

Signaling via the methylation of lysine residues in proteins has been linked to diverse biological and disease processes, yet the catalytic activity and substrate specificity of many human protein lysine methyltransferases (PKMTs) are unknown. We screened over 40 candidate PKMTs and identified SETD6 as a methyltransferase that monomethylated chromatin-associated transcription factor NF-κB subunit RelA at Lys310 (RelAK310me1). SETD6-mediated methylation rendered RelA inert and attenuated RelA-driven transcriptional programs, including inflammatory responses in primary immune cells. RelAK310me1 was recognized by the ankryin repeat of the histone methyltransferase GLP, which under basal conditions promoted a repressed chromatin state at RelA target genes through GLP-mediated methylation of histone H3 Lys9 (H3K9). NF-κB-activation-linked phosphorylation of RelA at Ser311 by protein kinase C-ζ (PKC-ζ) blocked the binding of GLP to RelAK310me1 and relieved repression of the target gene. Our findings establish a previously uncharacterized mechanism by which chromatin signaling regulates inflammation programs.


Asunto(s)
Artritis Reumatoide/inmunología , FN-kappa B/metabolismo , Proteína Metiltransferasas/metabolismo , Factor de Transcripción ReIA/metabolismo , Artritis Reumatoide/genética , Artritis Reumatoide/metabolismo , Ensamble y Desensamble de Cromatina/genética , Metilación de ADN , Células HEK293 , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Humanos , Inflamación , Lisina/metabolismo , FN-kappa B/genética , FN-kappa B/inmunología , Unión Proteica/genética , Proteína Metiltransferasas/genética , Proteína Metiltransferasas/inmunología , ARN Interferente Pequeño/genética , Transducción de Señal/genética , Transducción de Señal/inmunología , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/inmunología
6.
Mol Cell ; 50(3): 444-56, 2013 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-23583077

RESUMEN

Lysine methylation of histone proteins regulates chromatin dynamics and plays important roles in diverse physiological and pathological processes. However, beyond histone proteins, the proteome-wide extent of lysine methylation remains largely unknown. We have engineered the naturally occurring MBT domain repeats of L3MBTL1 to serve as a universal affinity reagent for detecting, enriching, and identifying proteins carrying a mono- or dimethylated lysine. The domain is broadly specific for methylated lysine ("pan-specific") and can be applied to any biological system. We have used our approach to demonstrate that SIRT1 is a substrate of the methyltransferase G9a both in vitro and in cells, to perform proteome-wide detection and enrichment of methylated proteins, and to identify candidate in-cell substrates of G9a and the related methyltransferase GLP. Together, our results demonstrate a powerful new approach for global and quantitative analysis of methylated lysine, and they represent the first systems biology understanding of lysine methylation.


Asunto(s)
Lisina/genética , Lisina/metabolismo , Proteoma/genética , Proteoma/metabolismo , Animales , Línea Celular , Células HEK293 , Humanos , Insectos , Metilación , Estructura Terciaria de Proteína , Proteómica/métodos , Células Sf9 , Sirtuina 1/genética , Sirtuina 1/metabolismo
7.
J Biol Chem ; 293(28): 11242-11250, 2018 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-29728458

RESUMEN

In the yeast Saccharomyces cerevisiae, genomic instability in rDNA repeat sequences is an underlying cause of cell aging and is suppressed by the chromatin-silencing factor Sir2. In humans, rDNA instability is observed in cancers and premature aging syndromes, but its underlying mechanisms and functional consequences remain unclear. Here, we uncovered a pivotal role of sirtuin 7 (SIRT7), a mammalian Sir2 homolog, in guarding against rDNA instability and show that this function of SIRT7 protects against senescence in primary human cells. We found that, mechanistically, SIRT7 is required for association of SNF2H (also called SMARCA5, SWI/SNF-related matrix-associated actin-dependent regulator of chromatin, subfamily A, member 5), a component of the nucleolar heterochromatin-silencing complex NoRC, with rDNA sequences. Defective rDNA-heterochromatin silencing in SIRT7-deficient cells unleashed rDNA instability, with excision and loss of rDNA gene copies, which in turn induced acute senescence. Mounting evidence indicates that accumulation of senescent cells significantly contributes to tissue dysfunction in aging-related pathologies. Our findings identify rDNA instability as a driver of mammalian cellular senescence and implicate SIRT7-dependent heterochromatin silencing in protecting against this process.


Asunto(s)
Neoplasias Óseas/patología , Senescencia Celular , ADN Ribosómico/genética , Epigénesis Genética , Inestabilidad Genómica , Osteosarcoma/patología , Sirtuinas/metabolismo , Neoplasias Óseas/genética , Neoplasias Óseas/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Osteosarcoma/genética , Osteosarcoma/metabolismo , Sirtuinas/genética , Transcripción Genética , Células Tumorales Cultivadas
8.
J Am Chem Soc ; 141(6): 2462-2473, 2019 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-30653310

RESUMEN

Using an engineered pyrrolysyl-tRNA synthetase mutant together with tRNACUAPyl, we have genetically encoded Nε-(7-azidoheptanoyl)-l-lysine (AzHeK) by amber codon in Escherichia coli for recombinant expression of a number of AzHeK-containing histone H3 proteins. We assembled in vitro acyl-nucleosomes from these recombinant acyl-H3 histones. All these acyl-nucleosomes contained an azide functionality that allowed quick click labeling with a strained alkyne dye for in-gel fluorescence analysis. Using these acyl-nucleosomes as substrates and click labeling as a detection method, we systematically investigated chromatin deacylation activities of SIRT7, a class III NAD+-dependent histone deacylase with roles in aging and cancer biology. Besides confirming the previously reported histone H3K18 deacylation activity, our results revealed that SIRT7 has an astonishingly high activity to catalyze deacylation of H3K36 and is also catalytically active to deacylate H3K37. We further demonstrated that this H3K36 deacylation activity is nucleosome dependent and can be significantly enhanced when appending the acyl-nucleosome substrate with a short double-stranded DNA that mimics the bridging DNA between nucleosomes in native chromatin. By overexpressing SIRT7 in human cells, we verified that SIRT7 natively removes acetylation from histone H3K36. Moreover, SIRT7-deficient cells exhibited H3K36 hyperacetylation in whole cell extracts, at rDNA sequences in nucleoli, and at select SIRT7 target loci, demonstrating the physiologic importance of SIRT7 in determining endogenous H3K36 acetylation levels. H3K36 acetylation has been detected at active gene promoters, but little is understood about its regulation and functions. Our findings establish H3K36 as a physiologic substrate of SIRT7 and implicate this modification in potential SIRT7 pathways in heterochromatin silencing and genomic stability.


Asunto(s)
Cromatina/metabolismo , Sirtuinas/metabolismo , Acilación , Biocatálisis , Dominio Catalítico , Química Clic , Histonas/química , Histonas/metabolismo , Lisina/metabolismo , Modelos Moleculares , Nucleosomas/metabolismo , Sirtuinas/química
9.
Nature ; 487(7405): 114-8, 2012 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-22722849

RESUMEN

Sirtuin proteins regulate diverse cellular pathways that influence genomic stability, metabolism and ageing. SIRT7 is a mammalian sirtuin whose biochemical activity, molecular targets and physiological functions have been unclear. Here we show that SIRT7 is an NAD(+)-dependent H3K18Ac (acetylated lysine 18 of histone H3) deacetylase that stabilizes the transformed state of cancer cells. Genome-wide binding studies reveal that SIRT7 binds to promoters of a specific set of gene targets, where it deacetylates H3K18Ac and promotes transcriptional repression. The spectrum of SIRT7 target genes is defined in part by its interaction with the cancer-associated E26 transformed specific (ETS) transcription factor ELK4, and comprises numerous genes with links to tumour suppression. Notably, selective hypoacetylation of H3K18Ac has been linked to oncogenic transformation, and in patients is associated with aggressive tumour phenotypes and poor prognosis. We find that deacetylation of H3K18Ac by SIRT7 is necessary for maintaining essential features of human cancer cells, including anchorage-independent growth and escape from contact inhibition. Moreover, SIRT7 is necessary for a global hypoacetylation of H3K18Ac associated with cellular transformation by the viral oncoprotein E1A. Finally, SIRT7 depletion markedly reduces the tumorigenicity of human cancer cell xenografts in mice. Together, our work establishes SIRT7 as a highly selective H3K18Ac deacetylase and demonstrates a pivotal role for SIRT7 in chromatin regulation, cellular transformation programs and tumour formation in vivo.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Histona Desacetilasas/metabolismo , Histonas/metabolismo , Lisina/metabolismo , Sirtuinas/metabolismo , Acetilación , Proteínas E1A de Adenovirus/genética , Proteínas E1A de Adenovirus/metabolismo , Animales , Secuencia de Bases , Sitios de Unión , Línea Celular Tumoral , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Cromatina/metabolismo , Inhibición de Contacto , Progresión de la Enfermedad , Humanos , Ratones , Trasplante de Neoplasias , Motivos de Nucleótidos , Fenotipo , Regiones Promotoras Genéticas , Proteínas Represoras/metabolismo , Sirtuinas/deficiencia , Sirtuinas/genética , Transcripción Genética , Trasplante Heterólogo , Proteína Elk-4 del Dominio ets/metabolismo
10.
Angew Chem Int Ed Engl ; 56(4): 1007-1011, 2017 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-27990725

RESUMEN

Sirtuins are protein deacylases regulating metabolism and stress responses, and are implicated in aging-related diseases. Small molecule activators for the human sirtuins Sirt1-7 are sought as chemical tools and potential therapeutics, such as for cancer. Activators are available for Sirt1 and exploit its unique N-terminus, whereas drug-like activators for Sirt2-7 are lacking. We synthesized and screened pyrrolo[1,2-a]quinoxaline derivatives, yielding the first synthetic Sirt6 activators. Biochemical assays show direct, substrate-independent compound binding to the Sirt6 catalytic core and potent activation of Sirt6-dependent deacetylation of peptide substrates and complete nucleosomes. Crystal structures of Sirt6/activator complexes reveal that the compounds bind to a Sirt6-specific acyl channel pocket and identify key interactions. Our results establish potent Sirt6 activation with small molecules and provide a structural basis for further development of Sirt6 activators as tools and therapeutics.


Asunto(s)
Pirroles/metabolismo , Quinoxalinas/metabolismo , Sirtuinas/metabolismo , Bibliotecas de Moléculas Pequeñas/metabolismo , Humanos , Modelos Moleculares , Estructura Molecular , Pirroles/química , Quinoxalinas/química , Sirtuinas/química , Bibliotecas de Moléculas Pequeñas/química
11.
Trends Biochem Sci ; 36(1): 39-46, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20729089

RESUMEN

Saccharomyces cerevisiae Sir2 is an NAD(+)-dependent histone deacetylase that links chromatin silencing to genomic stability, cellular metabolism and lifespan regulation. In mice, deficiency for the Sir2 family member SIRT6 leads to genomic instability, metabolic defects and degenerative pathologies associated with aging. Until recently, SIRT6 was an orphan enzyme whose catalytic activity and substrates were unclear. However, new mechanistic insights have come from the discovery that SIRT6 is a highly substrate-specific histone deacetylase that promotes proper chromatin function in several physiologic contexts, including telomere and genome stabilization, gene expression and DNA repair. By maintaining both the integrity and the expression of the mammalian genome, SIRT6 thus serves several roles that parallel Sir2 function. In this article, we review recent advances in understanding the mechanisms of SIRT6 action and their implications for human biology and disease.


Asunto(s)
Cromatina , Genoma , Sirtuinas/metabolismo , Acetilación , Animales , Senescencia Celular , Histonas/metabolismo , Humanos
12.
EMBO J ; 35(14): 1483-5, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-27302089
13.
J Biol Chem ; 288(25): 18439-47, 2013 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-23653361

RESUMEN

Aging is known to be the single most important risk factor for multiple diseases. Sirtuin 6, or SIRT6, has recently been identified as a critical regulator of transcription, genome stability, telomere integrity, DNA repair, and metabolic homeostasis. A knockout mouse model of SIRT6 has displayed dramatic phenotypes of accelerated aging. In keeping with its role in aging, we demonstrated that human dermal fibroblasts (HDFs) from older human subjects were more resistant to reprogramming by classic Yamanaka factors than those from younger human subjects, but the addition of SIRT6 during reprogramming improved such efficiency in older HDFs substantially. Despite the importance of SIRT6, little is known about the molecular mechanism of its regulation. We show, for the first, time posttranscriptional regulation of SIRT6 by miR-766 and inverse correlation in the expression of this microRNA in HDFs from different age groups. Our results suggest that SIRT6 regulates miR-766 transcription via a feedback regulatory loop, which has implications for the modulation of SIRT6 expression in reprogramming of aging cells.


Asunto(s)
Envejecimiento/fisiología , Reprogramación Celular/fisiología , Células Madre Pluripotentes Inducidas/fisiología , Sirtuinas/fisiología , Adolescente , Factores de Edad , Envejecimiento/genética , Células Cultivadas , Reprogramación Celular/genética , Análisis por Conglomerados , Dermis/citología , Cuerpos Embrioides/metabolismo , Cuerpos Embrioides/fisiología , Fibroblastos/citología , Fibroblastos/metabolismo , Fibroblastos/fisiología , Perfilación de la Expresión Génica , Humanos , Inmunohistoquímica , Células Madre Pluripotentes Inducidas/metabolismo , MicroARNs/genética , Persona de Mediana Edad , Modelos Genéticos , Células Madre Pluripotentes/metabolismo , Células Madre Pluripotentes/fisiología , Procesamiento Postranscripcional del ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sirtuinas/genética , Sirtuinas/metabolismo
14.
Nature ; 452(7186): 492-6, 2008 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-18337721

RESUMEN

The Sir2 deacetylase regulates chromatin silencing and lifespan in Saccharomyces cerevisiae. In mice, deficiency for the Sir2 family member SIRT6 leads to a shortened lifespan and a premature ageing-like phenotype. However, the molecular mechanisms of SIRT6 function are unclear. SIRT6 is a chromatin-associated protein, but no enzymatic activity of SIRT6 at chromatin has yet been detected, and the identity of physiological SIRT6 substrates is unknown. Here we show that the human SIRT6 protein is an NAD+-dependent, histone H3 lysine 9 (H3K9) deacetylase that modulates telomeric chromatin. SIRT6 associates specifically with telomeres, and SIRT6 depletion leads to telomere dysfunction with end-to-end chromosomal fusions and premature cellular senescence. Moreover, SIRT6-depleted cells exhibit abnormal telomere structures that resemble defects observed in Werner syndrome, a premature ageing disorder. At telomeric chromatin, SIRT6 deacetylates H3K9 and is required for the stable association of WRN, the factor that is mutated in Werner syndrome. We propose that SIRT6 contributes to the propagation of a specialized chromatin state at mammalian telomeres, which in turn is required for proper telomere metabolism and function. Our findings constitute the first identification of a physiological enzymatic activity of SIRT6, and link chromatin regulation by SIRT6 to telomere maintenance and a human premature ageing syndrome.


Asunto(s)
Cromatina/metabolismo , Histona Desacetilasas/metabolismo , Sirtuinas/metabolismo , Telómero/metabolismo , Acetilación , Línea Celular , Senescencia Celular/genética , Cromatina/genética , Replicación del ADN , Exodesoxirribonucleasas/metabolismo , Fibroblastos , Histona Desacetilasas/deficiencia , Histona Desacetilasas/genética , Histonas/química , Histonas/metabolismo , Humanos , Lisina/metabolismo , Fenotipo , Unión Proteica , RecQ Helicasas/metabolismo , Sirtuinas/deficiencia , Sirtuinas/genética , Telomerasa/genética , Telomerasa/metabolismo , Telómero/genética , Síndrome de Werner/genética , Helicasa del Síndrome de Werner
16.
Nature ; 442(7098): 96-9, 2006 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-16728974

RESUMEN

Dynamic regulation of diverse nuclear processes is intimately linked to covalent modifications of chromatin. Much attention has focused on methylation at lysine 4 of histone H3 (H3K4), owing to its association with euchromatic genomic regions. H3K4 can be mono-, di- or tri-methylated. Trimethylated H3K4 (H3K4me3) is preferentially detected at active genes, and is proposed to promote gene expression through recognition by transcription-activating effector molecules. Here we identify a novel class of methylated H3K4 effector domains--the PHD domains of the ING (for inhibitor of growth) family of tumour suppressor proteins. The ING PHD domains are specific and highly robust binding modules for H3K4me3 and H3K4me2. ING2, a native subunit of a repressive mSin3a-HDAC1 histone deacetylase complex, binds with high affinity to the trimethylated species. In response to DNA damage, recognition of H3K4me3 by the ING2 PHD domain stabilizes the mSin3a-HDAC1 complex at the promoters of proliferation genes. This pathway constitutes a new mechanism by which H3K4me3 functions in active gene repression. Furthermore, ING2 modulates cellular responses to genotoxic insults, and these functions are critically dependent on ING2 interaction with H3K4me3. Together, our findings establish a pivotal role for trimethylation of H3K4 in gene repression and, potentially, tumour suppressor mechanisms.


Asunto(s)
Silenciador del Gen , Histonas/química , Histonas/metabolismo , Proteínas de Homeodominio/química , Proteínas de Homeodominio/metabolismo , Lisina/metabolismo , Secuencias de Aminoácidos , Cromatina/metabolismo , Histona Desacetilasas/metabolismo , Proteínas de Homeodominio/genética , Metilación , Unión Proteica , Estructura Terciaria de Proteína , Especificidad por Sustrato , Proteínas Supresoras de Tumor/química , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
17.
Genes (Basel) ; 12(9)2021 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-34573442

RESUMEN

The modulation of dynamic histone acetylation states is key for organizing chromatin structure and modulating gene expression and is regulated by histone acetyltransferase (HAT) and histone deacetylase (HDAC) enzymes. The mammalian SIRT6 protein, a member of the Class III HDAC Sirtuin family of NAD+-dependent enzymes, plays pivotal roles in aging, metabolism, and cancer biology. Through its site-specific histone deacetylation activity, SIRT6 promotes chromatin silencing and transcriptional regulation of aging-associated, metabolic, and tumor suppressive gene expression programs. ATP citrate lyase (ACLY) is a nucleo-cytoplasmic enzyme that produces acetyl coenzyme A (acetyl-CoA), which is the required acetyl donor for lysine acetylation by HATs. In addition to playing a central role in generating cytosolic acetyl-CoA for de novo lipogenesis, a growing body of work indicates that ACLY also functions in the nucleus where it contributes to the nutrient-sensitive regulation of nuclear acetyl-CoA availability for histone acetylation in cancer cells. In this study, we have identified a novel function of SIRT6 in controlling nuclear levels of ACLY and ACLY-dependent tumor suppressive gene regulation. The inactivation of SIRT6 in cancer cells leads to the accumulation of nuclear ACLY protein and increases nuclear acetyl-CoA pools, which in turn drive locus-specific histone acetylation and the expression of cancer cell adhesion and migration genes that promote tumor invasiveness. Our findings uncover a novel mechanism of SIRT6 in suppressing invasive cancer cell phenotypes and identify acetyl-CoA responsive cell migration and adhesion genes as downstream targets of SIRT6.


Asunto(s)
ATP Citrato (pro-S)-Liasa/metabolismo , Histonas/metabolismo , Neoplasias/patología , Sirtuinas/metabolismo , ATP Citrato (pro-S)-Liasa/genética , Acetilcoenzima A/metabolismo , Acetilación , Adhesión Celular , Línea Celular Tumoral , Movimiento Celular , Regulación de la Expresión Génica , Células HEK293 , Humanos , Neoplasias/etiología , Neoplasias/metabolismo , Fenotipo , Sirtuinas/genética
18.
Cell Rep ; 34(3): 108638, 2021 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-33472068

RESUMEN

Histone acetylation levels are regulated by histone acetyltransferases (HATs) and histone deacetylases (HDACs) that antagonistically control the overall balance of this post-translational modification. HDAC inhibitors (HDACi) are potent agents that disrupt this balance and are used clinically to treat diseases including cancer. Despite their use, little is known about their effects on chromatin regulators, particularly those that signal through lysine acetylation. We apply quantitative genomic and proteomic approaches to demonstrate that HDACi robustly increases a low-abundance histone 4 polyacetylation state, which serves as a preferred binding substrate for several bromodomain-containing proteins, including BRD4. Increased H4 polyacetylation occurs in transcribed genes and correlates with the targeting of BRD4. Collectively, these results suggest that HDAC inhibition functions, at least in part, through expansion of a rare histone acetylation state, which then retargets lysine-acetyl readers associated with changes in gene expression, partially mimicking the effect of bromodomain inhibition.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Inhibidores de Histona Desacetilasas/uso terapéutico , Histonas/metabolismo , Factores de Transcripción/metabolismo , Acetilación , Inhibidores de Histona Desacetilasas/farmacología , Humanos
19.
Cell Metab ; 2(1): 67-76, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16054100

RESUMEN

The Saccharomyces cerevisiae chromatin silencing factor Sir2 suppresses genomic instability and extends replicative life span. In contrast, we find that mouse embryonic fibroblasts (MEFs) deficient for SIRT1, a mammalian Sir2 homolog, have dramatically increased resistance to replicative senescence. Extended replicative life span of SIRT1-deficient MEFs correlates with enhanced proliferative capacity under conditions of chronic, sublethal oxidative stress. In this context, SIRT1-deficient cells fail to normally upregulate either the p19(ARF) senescence regulator or its downstream target p53. However, upon acute DNA damage or oncogene expression, SIRT1-deficient cells show normal p19(ARF) induction and cell cycle arrest. Together, our findings demonstrate an unexpected SIRT1 function in promoting replicative senescence in response to chronic cellular stress and implicate p19(ARF) as a downstream effector in this pathway.


Asunto(s)
Senescencia Celular , Daño del ADN , Sirtuinas/metabolismo , Animales , Proliferación Celular/efectos de los fármacos , Senescencia Celular/efectos de los fármacos , Inhibidor p16 de la Quinasa Dependiente de Ciclina , Daño del ADN/efectos de los fármacos , Doxorrubicina/farmacología , Fibroblastos , Genes ras/genética , Peróxido de Hidrógeno/farmacología , Ratones , Ratones Noqueados , Células 3T3 NIH , Estrés Oxidativo/efectos de los fármacos , Fase S/efectos de los fármacos , Sirtuina 1 , Sirtuinas/deficiencia , Sirtuinas/genética , Proteína p14ARF Supresora de Tumor/metabolismo
20.
Sci Rep ; 10(1): 17425, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-33060621

RESUMEN

The tumor suppressor adenomatous polyposis coli (APC) is frequently mutated in colorectal cancers. APC and Axin are core components of a destruction complex that scaffolds GSK3ß and CK1 to earmark ß-catenin for proteosomal degradation. Disruption of APC results in pathologic stabilization of ß-catenin and oncogenesis. However, the molecular mechanism by which APC promotes ß-catenin degradation is unclear. Here, we find that the intrinsically disordered region (IDR) of APC, which contains multiple ß-catenin and Axin interacting sites, undergoes liquid-liquid phase separation (LLPS) in vitro. Expression of the APC IDR in colorectal cells promotes Axin puncta formation and ß-catenin degradation. Our results support the model that multivalent interactions between APC and Axin drives the ß-catenin destruction complex to form biomolecular condensates in cells, which concentrate key components to achieve high efficient degradation of ß-catenin.


Asunto(s)
Proteína Axina/metabolismo , Genes APC , beta Catenina/metabolismo , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Humanos , Proteolisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA