Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Nat Commun ; 15(1): 2960, 2024 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-38580649

RESUMEN

DNA methylation is an essential epigenetic chromatin modification, and its maintenance in mammals requires the protein UHRF1. It is yet unclear if UHRF1 functions solely by stimulating DNA methylation maintenance by DNMT1, or if it has important additional functions. Using degron alleles, we show that UHRF1 depletion causes a much greater loss of DNA methylation than DNMT1 depletion. This is not caused by passive demethylation as UHRF1-depleted cells proliferate more slowly than DNMT1-depleted cells. Instead, bioinformatics, proteomics and genetics experiments establish that UHRF1, besides activating DNMT1, interacts with DNMT3A and DNMT3B and promotes their activity. In addition, we show that UHRF1 antagonizes active DNA demethylation by TET2. Therefore, UHRF1 has non-canonical roles that contribute importantly to DNA methylation homeostasis; these findings have practical implications for epigenetics in health and disease.


Asunto(s)
Metilación de ADN , Neoplasias , Humanos , Proteínas Potenciadoras de Unión a CCAAT/genética , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Cromatina , ADN (Citosina-5-)-Metiltransferasa 1/genética , ADN (Citosina-5-)-Metiltransferasa 1/metabolismo , Neoplasias/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
2.
Life Sci Alliance ; 6(9)2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37468166

RESUMEN

Fun30 is the prototype of the Fun30-SMARCAD1-ETL subfamily of nucleosome remodelers involved in DNA repair and gene silencing. These proteins appear to act as single-subunit nucleosome remodelers, but their molecular mechanisms are, at this point, poorly understood. Using multiple sequence alignment and structure prediction, we identify an evolutionarily conserved domain that is modeled to contain a SAM-like fold with one long, protruding helix, which we term SAM-key. Deletion of the SAM-key within budding yeast Fun30 leads to a defect in DNA repair and gene silencing similar to that of the fun30Δ mutant. In vitro, Fun30 protein lacking the SAM-key is able to bind nucleosomes but is deficient in DNA-stimulated ATPase activity and nucleosome sliding and eviction. A structural model based on AlphaFold2 prediction and verified by crosslinking-MS indicates an interaction of the long SAM-key helix with protrusion I, a subdomain located between the two ATPase lobes that is critical for control of enzymatic activity. Mutation of the interaction interface phenocopies the domain deletion with a lack of DNA-stimulated ATPase activation and a nucleosome-remodeling defect, thereby confirming a role of the SAM-key helix in regulating ATPase activity. Our data thereby demonstrate a central role of the SAM-key domain in mediating the activation of Fun30 catalytic activity, thus highlighting the importance of allosteric activation for this class of enzymes.


Asunto(s)
Nucleosomas , Proteínas de Saccharomyces cerevisiae , Nucleosomas/genética , Nucleosomas/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , ADN/metabolismo , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/metabolismo
3.
Nucleic Acids Res ; 50(15): 8491-8511, 2022 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-35904814

RESUMEN

DNA methylation (5-methylcytosine (5mC)) is critical for genome stability and transcriptional regulation in mammals. The discovery that ten-eleven translocation (TET) proteins catalyze the oxidation of 5mC to 5-hydroxymethylcytosine (5hmC), 5-formylcytosine (5fC), and 5-carboxylcytosine (5caC) revolutionized our perspective on the complexity and regulation of DNA modifications. However, to what extent the regulatory functions of TET1 can be attributed to its catalytic activity remains unclear. Here, we use genome engineering and quantitative multi-omics approaches to dissect the precise catalytic vs. non-catalytic functions of TET1 in murine embryonic stem cells (mESCs). Our study identifies TET1 as an essential interaction hub for multiple chromatin modifying complexes and a global regulator of histone modifications. Strikingly, we find that the majority of transcriptional regulation depends on non-catalytic functions of TET1. In particular, we show that TET1 is critical for the establishment of H3K9me3 and H4K20me3 at endogenous retroviral elements (ERVs) and their silencing that is independent of its canonical role in DNA demethylation. Furthermore, we provide evidence that this repression of ERVs depends on the interaction between TET1 and SIN3A. In summary, we demonstrate that the non-catalytic functions of TET1 are critical for regulation of gene expression and the silencing of endogenous retroviruses in mESCs.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Retrovirus Endógenos , Proteínas Proto-Oncogénicas/metabolismo , 5-Metilcitosina/metabolismo , Animales , Citosina/metabolismo , Desmetilación del ADN , Metilación de ADN , Proteínas de Unión al ADN/genética , Retrovirus Endógenos/genética , Retrovirus Endógenos/metabolismo , Expresión Génica , Mamíferos/genética , Ratones , Proteínas Proto-Oncogénicas/genética
4.
Nucleic Acids Res ; 49(6): 3020-3032, 2021 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-33300031

RESUMEN

DNA methylation is essential to development and cellular physiology in mammals. Faulty DNA methylation is frequently observed in human diseases like cancer and neurological disorders. Molecularly, this epigenetic mark is linked to other chromatin modifications and it regulates key genomic processes, including transcription and splicing. Each round of DNA replication generates two hemi-methylated copies of the genome. These must be converted back to symmetrically methylated DNA before the next S-phase, or the mark will fade away; therefore the maintenance of DNA methylation is essential. Mechanistically, the maintenance of this epigenetic modification takes place during and after DNA replication, and occurs within the very dynamic context of chromatin re-assembly. Here, we review recent discoveries and unresolved questions regarding the mechanisms, dynamics and fidelity of DNA methylation maintenance in mammals. We also discuss how it could be regulated in normal development and misregulated in disease.


Asunto(s)
Metilación de ADN , Mamíferos/genética , Animales , Ensamble y Desensamble de Cromatina , ADN (Citosina-5-)-Metiltransferasa 1/metabolismo , Replicación del ADN , Epigénesis Genética , Humanos , Neoplasias/genética , Enfermedades del Sistema Nervioso/genética , Ubiquitina-Proteína Ligasas/metabolismo
5.
Nat Struct Mol Biol ; 26(10): 899-909, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31548724

RESUMEN

Polycomb repressive complex 2 (PRC2) maintains repression of cell-type-specific genes but also associates with genes ectopically in cancer. While it is currently unknown how PRC2 is removed from genes, such knowledge would be useful for the targeted reversal of deleterious PRC2 recruitment events. Here, we show that G-tract RNA specifically removes PRC2 from genes in human and mouse cells. PRC2 preferentially binds G tracts within nascent precursor mRNA (pre-mRNA), especially within predicted G-quadruplex structures. G-quadruplex RNA evicts the PRC2 catalytic core from the substrate nucleosome. In cells, PRC2 transfers from chromatin to pre-mRNA upon gene activation, and chromatin-associated G-tract RNA removes PRC2, leading to H3K27me3 depletion from genes. Targeting G-tract RNA to the tumor suppressor gene CDKN2A in malignant rhabdoid tumor cells reactivates the gene and induces senescence. These data support a model in which pre-mRNA evicts PRC2 during gene activation and provides the means to selectively remove PRC2 from specific genes.


Asunto(s)
Complejo Represivo Polycomb 2/metabolismo , Precursores del ARN/metabolismo , Animales , Línea Celular , Cromatina/metabolismo , G-Cuádruplex , Histonas/metabolismo , Humanos , Ratones , Nucleosomas/metabolismo , Unión Proteica , Precursores del ARN/química , Activación Transcripcional
6.
Mol Cell Oncol ; 6(4): 1605820, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31211233

RESUMEN

Mutations caused by DNA damage are a main driver of cancer. We discovered that recognition of newly synthesised histone H4 directs breast cancer type 1 susceptibility protein (BRCA1) to post-replicative chromatin. The switch from mutagenic to error-free DNA double strand break repair by homologous recombination is therefore controlled by chromatin.

7.
Nat Cell Biol ; 21(3): 311-318, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30804502

RESUMEN

Genotoxic DNA double-strand breaks (DSBs) can be repaired by error-free homologous recombination (HR) or mutagenic non-homologous end-joining1. HR supresses tumorigenesis1, but is restricted to the S and G2 phases of the cell cycle when a sister chromatid is present2. Breast cancer type 1 susceptibility protein (BRCA1) promotes HR by antagonizing the anti-resection factor TP53-binding protein 1(53BP1) (refs. 2-5), but it remains unknown how BRCA1 function is limited to the S and G2 phases. We show that BRCA1 recruitment requires recognition of histone H4 unmethylated at lysine 20 (H4K20me0), linking DSB repair pathway choice directly to sister chromatid availability. We identify the ankyrin repeat domain of BRCA1-associated RING domain protein 1 (BARD1)-the obligate BRCA1 binding partner3-as a reader of H4K20me0 present on new histones in post-replicative chromatin6. BARD1 ankyrin repeat domain mutations disabling H4K20me0 recognition abrogate accumulation of BRCA1 at DSBs, causing aberrant build-up of 53BP1, and allowing anti-resection activity to prevail in S and G2. Consequently, BARD1 recognition of H4K20me0 is required for HR and resistance to poly (ADP-ribose) polymerase inhibitors. Collectively, this reveals that BRCA1-BARD1 monitors the replicative state of the genome to oppose 53BP1 function, routing only DSBs within sister chromatids to HR.


Asunto(s)
Proteína BRCA1/metabolismo , Cromátides/metabolismo , Histonas/metabolismo , Recombinación Homóloga , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Secuencia de Aminoácidos , Proteína BRCA1/genética , Línea Celular Tumoral , Cromátides/genética , Roturas del ADN de Doble Cadena , Reparación del ADN , Fase G2/genética , Células HCT116 , Células HeLa , Humanos , Lisina/metabolismo , Metilación , Fase S/genética , Homología de Secuencia de Aminoácido , Proteínas Supresoras de Tumor/genética , Ubiquitina-Proteína Ligasas/genética
8.
PLoS Genet ; 13(8): e1006959, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28806732

RESUMEN

KDM2A is a histone demethylase associated with transcriptional silencing, however very little is known about its in vivo role in development and disease. Here we demonstrate that loss of the orthologue kdm2aa in zebrafish causes widespread transcriptional disruption and leads to spontaneous melanomas at a high frequency. Fish homozygous for two independent premature stop codon alleles show reduced growth and survival, a strong male sex bias, and homozygous females exhibit a progressive oogenesis defect. kdm2aa mutant fish also develop melanomas from early adulthood onwards which are independent from mutations in braf and other common oncogenes and tumour suppressors as revealed by deep whole exome sequencing. In addition to effects on translation and DNA replication gene expression, high-replicate RNA-seq in morphologically normal individuals demonstrates a stable regulatory response of epigenetic modifiers and the specific de-repression of a group of zinc finger genes residing in constitutive heterochromatin. Together our data reveal a complex role for Kdm2aa in regulating normal mRNA levels and carcinogenesis. These findings establish kdm2aa mutants as the first single gene knockout model of melanoma biology.


Asunto(s)
Histona Demetilasas con Dominio de Jumonji/genética , Melanoma/genética , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas de Pez Cebra/genética , Pez Cebra/genética , Animales , Replicación del ADN , Modelos Animales de Enfermedad , Epigénesis Genética , Exoma , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Técnicas de Inactivación de Genes , Masculino , Mutación , ARN Mensajero/genética , ARN Mensajero/metabolismo , Análisis de Secuencia de ARN , Pez Cebra/embriología
9.
Nucleic Acids Res ; 45(3): 1114-1129, 2017 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-28180290

RESUMEN

Functional genomic elements are marked by characteristic DNA and histone modification signatures. How combinatorial chromatin modification states are recognized by epigenetic reader proteins and how this is linked to their biological function is largely unknown. Here we provide a detailed molecular analysis of chromatin recognition by the lysine demethylase KDM2A. Using biochemical approaches we identify a nucleosome interaction module within KDM2A consisting of a CXXC type zinc finger, a PHD domain and a newly identified Heterochromatin Protein 1 (HP1) interaction motif that mediates direct binding between KDM2A and HP1. This nucleosome interaction module enables KDM2A to decode nucleosomal H3K9me3 modification in addition to CpG methylation signals. The multivalent engagement with DNA and HP1 results in a nucleosome binding circuit in which KDM2A can be recruited to H3K9me3-modified chromatin through HP1, and HP1 can be recruited to unmodified chromatin by KDM2A. A KDM2A mutant deficient in HP1-binding is inactive in an in vivo overexpression assay in zebrafish embryos demonstrating that the HP1 interaction is essential for KDM2A function. Our results reveal a complex regulation of chromatin binding for both KDM2A and HP1 that is modulated by DNA- and H3K9-methylation, and suggest a direct role for KDM2A in chromatin silencing.


Asunto(s)
Proteínas Cromosómicas no Histona/química , Proteínas Cromosómicas no Histona/metabolismo , Proteínas F-Box/química , Proteínas F-Box/metabolismo , Histona Demetilasas con Dominio de Jumonji/química , Histona Demetilasas con Dominio de Jumonji/metabolismo , Animales , Animales Modificados Genéticamente , Línea Celular , Homólogo de la Proteína Chromobox 5 , Proteínas Cromosómicas no Histona/genética , Islas de CpG , Cricetinae , Metilación de ADN , Proteínas F-Box/genética , Células HEK293 , Células HeLa , Histonas/metabolismo , Humanos , Histona Demetilasas con Dominio de Jumonji/genética , Modelos Genéticos , Mutación , Neoplasias/genética , Neoplasias/metabolismo , Nucleosomas/genética , Nucleosomas/metabolismo , Dominios y Motivos de Interacción de Proteínas , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Técnicas del Sistema de Dos Híbridos , Pez Cebra , Dedos de Zinc
10.
Nature ; 534(7609): 714-718, 2016 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-27338793

RESUMEN

After DNA replication, chromosomal processes including DNA repair and transcription take place in the context of sister chromatids. While cell cycle regulation can guide these processes globally, mechanisms to distinguish pre- and post-replicative states locally remain unknown. Here we reveal that new histones incorporated during DNA replication provide a signature of post-replicative chromatin, read by the human TONSL­MMS22L homologous recombination complex. We identify the TONSL ankyrin repeat domain (ARD) as a reader of histone H4 tails unmethylated at K20 (H4K20me0), which are specific to new histones incorporated during DNA replication and mark post-replicative chromatin until the G2/M phase of the cell cycle. Accordingly, TONSL­MMS22L binds new histones H3­H4 both before and after incorporation into nucleosomes, remaining on replicated chromatin until late G2/M. H4K20me0 recognition is required for TONSL­MMS22L binding to chromatin and accumulation at challenged replication forks and DNA lesions. Consequently, TONSL ARD mutants are toxic, compromising genome stability, cell viability and resistance to replication stress. Together, these data reveal a histone-reader-based mechanism for recognizing the post-replicative state, offering a new angle to understand DNA repair with the potential for targeted cancer therapy.


Asunto(s)
Cromatina/química , Cromatina/metabolismo , Reparación del ADN , Replicación del ADN , Proteínas de Unión al ADN/metabolismo , Histonas/metabolismo , FN-kappa B/metabolismo , Proteínas Nucleares/metabolismo , Cromatina/genética , Inestabilidad Genómica , Histonas/química , Recombinación Homóloga , Humanos , Lisina/metabolismo , Metilación , Modelos Moleculares , Chaperonas Moleculares/metabolismo , Unión Proteica , Estructura Terciaria de Proteína
11.
Chromosoma ; 125(1): 75-93, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26188466

RESUMEN

The regulation of chromatin by epigenetic mechanisms plays a central role in gene expression and is essential for development and maintenance of cell identity and function. Aberrant chromatin regulation is observed in many diseases where it leads to defects in epigenetic gene regulation resulting in pathological gene expression programmes. These defects are caused by inherited or acquired mutations in genes encoding enzymes that deposit or remove DNA and histone modifications and that shape chromatin architecture. Chromatin deregulation often results in neurodevelopmental disorders and intellectual disabilities, frequently linked to physical and developmental abnormalities, but can also cause neurodegenerative diseases, immunodeficiency, or muscle wasting syndromes. Epigenetic diseases can either be of monogenic origin or manifest themselves as complex multifactorial diseases such as in congenital heart disease, autism spectrum disorders, or cancer in which mutations in chromatin regulators are contributing factors. The environment directly influences the epigenome and can induce changes that cause or predispose to diseases through risk factors such as stress, malnutrition or exposure to harmful chemicals. The plasticity of chromatin regulation makes targeting the enzymatic machinery an attractive strategy for therapeutic intervention and an increasing number of small molecule inhibitors against a variety of epigenetic regulators are in clinical use or under development. In this review, we will give an overview of the molecular lesions that underlie epigenetic diseases, and we will discuss the impact of the environment and prospects for epigenetic therapies.


Asunto(s)
Ensamble y Desensamble de Cromatina , Cromatina/enzimología , Epigénesis Genética , Animales , Trastorno del Espectro Autista/genética , Cromatina/efectos de los fármacos , Cromatina/genética , ADN/metabolismo , Contaminantes Ambientales/farmacología , Femenino , Predisposición Genética a la Enfermedad , Cardiopatías/genética , Histonas/metabolismo , Humanos , Masculino , Mutación , Neoplasias/genética , Enfermedades Neurodegenerativas/genética
12.
Brief Funct Genomics ; 12(3): 205-18, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23401080

RESUMEN

The involvement of epigenetic processes in the origin and progression of cancer is now widely appreciated. Consequently, targeting the enzymatic machinery that controls the epigenetic regulation of the genome has emerged as an attractive new strategy for therapeutic intervention. The development of epigenetic drugs requires a detailed knowledge of the processes that govern chromatin regulation. Over the recent years, mass spectrometry (MS) has become an indispensable tool in epigenetics research. In this review, we will give an overview of the applications of MS-based proteomics in studying various aspects of chromatin biology. We will focus on the use of MS in the discovery and mapping of histone modifications and how novel proteomic approaches are being utilized to identify and study chromatin-associated proteins and multi-subunit complexes. Finally, we will discuss the application of proteomic methods in the diagnosis and prognosis of cancer based on epigenetic biomarkers and comment on their future impact on cancer epigenetics.


Asunto(s)
Epigenómica/métodos , Neoplasias/genética , Neoplasias/metabolismo , Proteómica/métodos , Animales , Humanos , Espectrometría de Masas
13.
J Mol Biol ; 422(1): 3-17, 2012 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-22575888

RESUMEN

The NuRD (nucleosome remodeling and deacetylase) complex serves as a crucial epigenetic regulator of cell differentiation, proliferation, and hematopoietic development by coupling the deacetylation and demethylation of histones, nucleosome mobilization, and the recruitment of transcription factors. The core nucleosome remodeling function of the mammalian NuRD complex is executed by the helicase-domain-containing ATPase CHD4 (Mi-2ß) subunit, which also contains N-terminal plant homeodomain (PHD) and chromo domains. The mode of regulation of chromatin remodeling by CHD4 is not well understood, nor is the role of its PHD and chromo domains. Here, we use small-angle X-ray scattering, nucleosome binding ATPase and remodeling assays, limited proteolysis, cross-linking, and tandem mass spectrometry to propose a three-dimensional structural model describing the overall shape and domain interactions of CHD4 and discuss the relevance of these for regulating the remodeling of chromatin by the NuRD complex.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Autoantígenos/química , Autoantígenos/metabolismo , Ensamble y Desensamble de Cromatina , Cromatina/metabolismo , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/química , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/metabolismo , Sitios de Unión , Ensayo de Cambio de Movilidad Electroforética , Humanos , Modelos Biológicos , Nucleosomas/metabolismo , Estructura Terciaria de Proteína , Proteolisis
14.
Cell ; 143(3): 470-84, 2010 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-21029866

RESUMEN

Modifications on histones or on DNA recruit proteins that regulate chromatin function. Here, we use nucleosomes methylated on DNA and on histone H3 in an affinity assay, in conjunction with a SILAC-based proteomic analysis, to identify "crosstalk" between these two distinct classes of modification. Our analysis reveals proteins whose binding to nucleosomes is regulated by methylation of CpGs, H3K4, H3K9, and H3K27 or a combination thereof. We identify the origin recognition complex (ORC), including LRWD1 as a subunit, to be a methylation-sensitive nucleosome interactor that is recruited cooperatively by DNA and histone methylation. Other interactors, such as the lysine demethylase Fbxl11/KDM2A, recognize nucleosomes methylated on histones, but their recruitment is disrupted by DNA methylation. These data establish SILAC nucleosome affinity purifications (SNAP) as a tool for studying the dynamics between different chromatin modifications and provide a modification binding "profile" for proteins regulated by DNA and histone methylation.


Asunto(s)
Ensamble y Desensamble de Cromatina , Metilación de ADN , Proteínas de Unión al ADN/metabolismo , Histonas/metabolismo , Línea Celular Tumoral , Islas de CpG , Proteínas F-Box/metabolismo , Células HeLa , Humanos , Histona Demetilasas con Dominio de Jumonji/metabolismo , Metilación , Nucleosomas/metabolismo , Complejo de Reconocimiento del Origen/metabolismo , Oxidorreductasas N-Desmetilantes/metabolismo , Proteómica/métodos , Coloración y Etiquetado/métodos
15.
Nature ; 461(7265): 819-22, 2009 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-19783980

RESUMEN

Activation of Janus kinase 2 (JAK2) by chromosomal translocations or point mutations is a frequent event in haematological malignancies. JAK2 is a non-receptor tyrosine kinase that regulates several cellular processes by inducing cytoplasmic signalling cascades. Here we show that human JAK2 is present in the nucleus of haematopoietic cells and directly phosphorylates Tyr 41 (Y41) on histone H3. Heterochromatin protein 1alpha (HP1alpha), but not HP1beta, specifically binds to this region of H3 through its chromo-shadow domain. Phosphorylation of H3Y41 by JAK2 prevents this binding. Inhibition of JAK2 activity in human leukaemic cells decreases both the expression of the haematopoietic oncogene lmo2 and the phosphorylation of H3Y41 at its promoter, while simultaneously increasing the binding of HP1alpha at the same site. Tauhese results identify a previously unrecognized nuclear role for JAK2 in the phosphorylation of H3Y41 and reveal a direct mechanistic link between two genes, jak2 and lmo2, involved in normal haematopoiesis and leukaemia.


Asunto(s)
Cromatina/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Histonas/metabolismo , Janus Quinasa 2/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Sitios de Unión , Línea Celular , Núcleo Celular/enzimología , Cromatina/química , Homólogo de la Proteína Chromobox 5 , Proteínas de Unión al ADN/genética , Regulación Neoplásica de la Expresión Génica , Hematopoyesis/genética , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/enzimología , Histonas/química , Histonas/genética , Humanos , Janus Quinasa 2/antagonistas & inhibidores , Proteínas con Dominio LIM , Leucemia/enzimología , Leucemia/genética , Leucemia/metabolismo , Leucemia/patología , Metaloproteínas/genética , Ratones , Oncogenes/genética , Fosforilación , Regiones Promotoras Genéticas/genética , Unión Proteica , Proteínas Proto-Oncogénicas , Transducción de Señal , Tirosina/metabolismo
16.
Mol Biol Cell ; 17(8): 3356-68, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16707568

RESUMEN

Protein misfolding is linked to different neurodegenerative disorders like Alzheimer's disease, polyglutamine, and prion diseases. We investigated the cytotoxic effects of aberrant conformers of the prion protein (PrP) and show that toxicity is specifically linked to misfolding of PrP in the cytosolic compartment and involves binding of PrP to the anti-apoptotic protein Bcl-2. PrP targeted to different cellular compartments, including the cytosol, nucleus, and mitochondria, adopted a misfolded and partially proteinase K-resistant conformation. However, only in the cytosol did the accumulation of misfolded PrP induce apoptosis. Apoptotic cell death was also induced by two pathogenic mutants of PrP, which are partially localized in the cytosol. A mechanistic analysis revealed that the toxic potential is linked to an internal domain of PrP (amino acids 115-156) and involves coaggregation of cytosolic PrP with Bcl-2. Increased expression of the chaperones Hsp70 and Hsp40 prevented the formation of PrP/Bcl-2 coaggregates and interfered with PrP-induced apoptosis. Our study reveals a compartment-specific toxicity of PrP misfolding that involves coaggregation of Bcl-2 and indicates a protective role of molecular chaperones.


Asunto(s)
Citosol/metabolismo , Proteínas PrPC/química , Proteínas PrPC/metabolismo , Pliegue de Proteína , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Animales , Apoptosis , Compartimento Celular , Células Cultivadas , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Ratones , Ratones Endogámicos C57BL , Chaperonas Moleculares/metabolismo , Mutación/genética , Enfermedades por Prión/metabolismo , Unión Proteica , Estructura Secundaria de Proteína , Transporte de Proteínas
17.
Mol Cell ; 14(6): 801-11, 2004 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-15200957

RESUMEN

Apoptotic cell death and survival is controlled by pro- and antiapoptotic proteins. Because these proteins act on each other, cell fate is dictated by the relative activity of pro- versus antiapoptotic proteins. Here we report that BRUCE, a conserved 528 kDa peripheral membrane protein of the trans-Golgi network, protects cells against apoptosis and functions as an inhibitor of apoptosis (IAP). By using wild-type and mutant forms we show that BRUCE inhibits caspase activity and apoptosis depending on its BIR domain. Upon apoptosis induction, BRUCE is antagonized by three mechanisms: first, through binding to Smac; second, by the protease HtrA2; and third, by caspase-mediated cleavage. In addition to its IAP activity BRUCE has the distinctive property of functioning as a chimeric E2/E3 ubiquitin ligase with Smac being a substrate. Our work suggests that, owing to its two activities and its localization, BRUCE may function as a specialized regulator of cell death pathways.


Asunto(s)
Apoptosis , Proteínas de Neoplasias/fisiología , Enzimas Ubiquitina-Conjugadoras/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Caspasas/metabolismo , Caspasas/farmacología , Línea Celular Tumoral , ADN Complementario/metabolismo , Células HeLa , Humanos , Proteínas Inhibidoras de la Apoptosis , Ratones , Mutación , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal , Enzimas Ubiquitina-Conjugadoras/análisis , Ubiquitina-Proteína Ligasas/análisis , Red trans-Golgi/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA