Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Cell Sci ; 136(23)2023 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-37997922

RESUMEN

The correct inheritance of chromatin structure is key for maintaining genome function and cell identity and preventing cellular transformation. DEK, a conserved non-histone chromatin protein, has recognized tumor-promoting properties, its overexpression being associated with poor prognosis in various cancer types. At the cellular level, DEK displays pleiotropic functions, influencing differentiation, apoptosis and stemness, but a characteristic oncogenic mechanism has remained elusive. Here, we report the identification of DEK bodies, focal assemblies of DEK that regularly occur at specific, yet unidentified, sites of heterochromatin replication exclusively in late S-phase. In these bodies, DEK localizes in direct proximity to active replisomes in agreement with a function in the early maturation of heterochromatin. A high-throughput siRNA screen, supported by mutational and biochemical analyses, identifies SUMO as one regulator of DEK body formation, linking DEK to the complex SUMO protein network that controls chromatin states and cell fate. This work combines and refines our previous data on DEK as a factor essential for heterochromatin integrity and facilitating replication under stress, and delineates an avenue of further study for unraveling the contribution of DEK to cancer development.


Asunto(s)
Heterocromatina , Neoplasias , Humanos , Proteínas de Unión a Poli-ADP-Ribosa/genética , Proteínas Cromosómicas no Histona/metabolismo , Proteínas Oncogénicas/genética , Proteínas Oncogénicas/metabolismo , Cromatina
2.
Genes Dev ; 25(7): 673-8, 2011 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-21460035

RESUMEN

Heterochromatin integrity is crucial for genome stability and regulation of gene expression, but the factors involved in mammalian heterochromatin biology are only incompletely understood. Here we identify the oncoprotein DEK, an abundant nuclear protein with a previously enigmatic in vivo function, as a Suppressor of Variegation [Su(var)] that is crucial to global heterochromatin integrity. We show that DEK interacts directly with Heterochromatin Protein 1 α (HP1α) and markedly enhances its binding to trimethylated H3K9 (H3K9me3), which is key for maintaining heterochromatic regions. Loss of Dek in Drosophila leads to a Su(var) phenotype and global reduction in heterochromatin. Thus, these findings show that DEK is a key factor in maintaining the balance between heterochromatin and euchromatin in vivo.


Asunto(s)
Proteínas Cromosómicas no Histona/metabolismo , Heterocromatina/genética , Heterocromatina/metabolismo , Proteínas Oncogénicas/metabolismo , Animales , Ciclo Celular/efectos de los fármacos , Homólogo de la Proteína Chromobox 5 , Proteínas Cromosómicas no Histona/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Inestabilidad Genómica/genética , Células HEK293 , Células HeLa , Inhibidores de Histona Desacetilasas/farmacología , Histonas/metabolismo , Humanos , Microscopía Electrónica de Transmisión , Proteínas Oncogénicas/genética , Proteínas de Unión a Poli-ADP-Ribosa , Unión Proteica , Receptores de la Familia Eph/genética , Receptores de la Familia Eph/metabolismo , Células Tumorales Cultivadas
3.
Proteins ; 86(1): 88-97, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29082557

RESUMEN

DEK is an oncoprotein that is overexpressed in many forms of cancer and participates in numerous cellular pathways. Of these different pathways, relevant interacting partners and functions of DEK are well described in regard to the regulation of chromatin structure, epigenetic marks, and transcription. Most of this understanding was derived by investigating DNA-binding and chromatin processing capabilities of the oncoprotein. To facilitate the generation of mechanism-driven hypotheses regarding DEK activities in underexplored areas, we have developed the first DEK interactome model using tandem-affinity purification and mass spectrometry. With this approach, we identify IMPDH2, DDX21, and RPL7a as novel DEK binding partners, hinting at new roles for the oncogene in de novo nucleotide biosynthesis and ribosome formation. Additionally, a hydroxyurea-specific interaction with replication protein A (RPA) was observed, suggesting that a DEK-RPA complex may form in response to DNA replication fork stalling. Taken together, these findings highlight diverse activities for DEK across cellular pathways and support a model wherein this molecule performs a plethora of functions.


Asunto(s)
Proteínas Cromosómicas no Histona/química , Proteínas Cromosómicas no Histona/metabolismo , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Proteínas Oncogénicas/química , Proteínas Oncogénicas/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa/química , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Sitios de Unión , Cromatina/química , Cromatina/metabolismo , Cromatografía Líquida de Alta Presión/métodos , ADN/química , Células HEK293 , Células HeLa , Humanos , Estructura Molecular , Unión Proteica , Conformación Proteica , Relación Estructura-Actividad , Espectrometría de Masas en Tándem/métodos
4.
Genome Res ; 23(9): 1505-13, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23657884

RESUMEN

Human endogenous retroviruses (HERVs) make up 8% of the human genome. The HERV-K (HML-2) family is the most recent group of these viruses to have inserted into the genome, and we have detected the activation of HERV-K (HML-2) proviruses in the blood of patients with HIV-1 infection. We report that HIV-1 infection activates expression of a novel HERV-K (HML-2) provirus, termed K111, present in multiple copies in the centromeres of chromosomes throughout the human genome yet not annotated in the most recent human genome assembly. Infection with HIV-1 or stimulation with the HIV-1 Tat protein leads to the activation of K111 proviruses. K111 is present as a single copy in the genome of the chimpanzee, yet K111 is not found in the genomes of other primates. Remarkably, K111 proviruses appear in the genomes of the extinct Neanderthal and Denisovan, while modern humans have at least 100 K111 proviruses spread across the centromeres of 15 chromosomes. Our studies suggest that the progenitor K111 integrated before the Homo-Pan divergence and expanded in copy number during the evolution of hominins, perhaps by recombination. The expansion of K111 provides sequence evidence suggesting that recombination between the centromeres of various chromosomes took place during the evolution of humans. K111 proviruses show significant sequence variations in each individual centromere, which may serve as markers in future efforts to annotate human centromere sequences. Further, this work is an example of the potential to discover previously unknown genomic sequences through the analysis of nucleic acids found in the blood of patients.


Asunto(s)
Retrovirus Endógenos/genética , Genoma Humano , Infecciones por VIH/genética , Provirus/genética , Integración Viral , Animales , Centrómero/genética , Centrómero/virología , Cromosomas Humanos/genética , Cromosomas Humanos/virología , Evolución Molecular , Hominidae/genética , Hominidae/virología , Humanos , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo
5.
Proc Natl Acad Sci U S A ; 110(17): 6847-52, 2013 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-23569252

RESUMEN

DEK is a biochemically distinct, conserved nonhistone protein that is vital to global heterochromatin integrity. In addition, DEK can be secreted and function as a chemotactic, proinflammatory factor. Here we show that exogenous DEK can penetrate cells, translocate to the nucleus, and there carry out its endogenous nuclear functions. Strikingly, adjacent cells can take up DEK secreted from synovial macrophages. DEK internalization is a heparan sulfate-dependent process, and cellular uptake of DEK into DEK knockdown cells corrects global heterochromatin depletion and DNA repair deficits, the phenotypic aberrations characteristic of these cells. These findings thus unify the extracellular and intracellular activities of DEK, and suggest that this paracrine loop involving DEK plays a role in chromatin biology.


Asunto(s)
Proteínas Cromosómicas no Histona/metabolismo , Reparación del ADN/fisiología , Heterocromatina/metabolismo , Histonas/metabolismo , Proteínas Oncogénicas/metabolismo , Comunicación Paracrina/fisiología , Fraccionamiento Celular , Células HeLa , Humanos , Procesamiento de Imagen Asistido por Computador , Immunoblotting , Inmunoprecipitación , Microscopía Fluorescente , Proteínas de Unión a Poli-ADP-Ribosa , Transporte de Proteínas/fisiología , ARN Interferente Pequeño/genética
6.
Blood Cells Mol Dis ; 54(1): 123-31, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25128083

RESUMEN

DEK is important in regulating cellular processes including proliferation, differentiation and maintenance of stem cell phenotype. The translocation t(6;9) in Acute Myeloid Leukemia (AML), which fuses DEK with NUP214, confers a poor prognosis and a higher risk of relapse. The over-expression of DEK in AML has been reported, but different studies have shown diminished levels in pediatric and promyelocytic leukemias. This study has characterized DEK expression, in silico, using a large multi-center cohort of leukemic and normal control cases. Overall, DEK was under-expressed in AML compared to normal bone marrow (NBM). Studying specific subtypes of AML confirmed either no significant change or a significant reduction in DEK expression compared to NBM. Importantly, the similarity of DEK expression between AML and NBM was confirmed using immunohistochemistry analysis of tissue mircorarrays. In addition, stratification of AML patients based on median DEK expression levels indicated that DEK showed no effect on the overall survival of patients. DEK expression during normal hematopoiesis did reveal a relationship with specific cell types implicating a distinct function during myeloid differentiation. Whilst DEK may play a potential role in hematopoiesis, it remains to be established whether it is important for leukemagenesis, except when involved in the t(6;9) translocation.


Asunto(s)
Proteínas Cromosómicas no Histona/biosíntesis , Proteínas de Unión al ADN/biosíntesis , Bases de Datos Genéticas , Regulación Leucémica de la Expresión Génica , Hematopoyesis , Leucemia Mieloide Aguda/metabolismo , Proteínas Oncogénicas/biosíntesis , Animales , Proteínas Cromosómicas no Histona/genética , Cromosomas Humanos Par 6/genética , Cromosomas Humanos Par 9/genética , Estudios de Cohortes , Proteínas de Unión al ADN/genética , Supervivencia sin Enfermedad , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Estudios Multicéntricos como Asunto , Proteínas Oncogénicas/genética , Proteínas de Unión a Poli-ADP-Ribosa , Tasa de Supervivencia , Translocación Genética
7.
Stem Cells ; 31(8): 1447-53, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23733396

RESUMEN

Understanding the factors that regulate hematopoiesis opens up the possibility of modifying these factors and their actions for clinical benefit. DEK, a non-histone nuclear phosphoprotein initially identified as a putative proto-oncogene, has recently been linked to regulate hematopoiesis. DEK has myelosuppressive activity in vitro on proliferation of human and mouse hematopoietic progenitor cells and enhancing activity on engraftment of long-term marrow repopulating mouse stem cells, has been linked in coordinate regulation with the transcription factor C/EBPα, for differentiation of myeloid cells, and apparently targets a long-term repopulating hematopoietic stem cell for leukemic transformation. This review covers the uniqueness of DEK, what is known about how it now functions as a nuclear protein and also as a secreted molecule that can act in paracrine fashion, and how it may be regulated in part by dipeptidylpeptidase 4, an enzyme known to truncate and modify a number of proteins involved in activities on hematopoietic cells. Examples are provided of possible future areas of investigation needed to better understand how DEK may be regulated and function as a regulator of hematopoiesis, information possibly translatable to other normal and diseased immature cell systems.


Asunto(s)
Proteínas Cromosómicas no Histona/fisiología , Proteínas de Unión al ADN/fisiología , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/fisiología , Proteínas Oncogénicas/fisiología , Animales , Humanos , Proteínas de Unión a Poli-ADP-Ribosa , Proto-Oncogenes Mas
8.
Arthritis Rheum ; 63(2): 556-67, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21280010

RESUMEN

OBJECTIVE: DEK is a nuclear phosphoprotein and autoantigen in a subset of children with juvenile idiopathic arthritis (JIA). Autoantibodies to DEK are also found in a broad spectrum of disorders associated with abnormal immune activation. We previously demonstrated that DEK is secreted by macrophages, is released by apoptotic T cells, and attracts leukocytes. Since DEK has been identified in the synovial fluid (SF) of patients with JIA, this study was undertaken to investigate how DEK protein and/or autoantibodies may contribute to the pathogenesis of JIA. METHODS: DEK autoantibodies, immune complexes (ICs), and synovial macrophages were purified from the SF of patients with JIA. DEK autoantibodies and ICs were purified by affinity-column chromatography and analyzed by 2-dimensional gel electrophoresis, immunoblotting, and enzyme-linked immunosorbent assay. DEK in supernatants and exosomes was purified by serial centrifugation and immunoprecipitation with magnetic beads, and posttranslational modifications of DEK were identified by nano-liquid chromatography tandem mass spectrometry (nano-LC-MS/MS). RESULTS: DEK autoantibodies and protein were found in the SF of patients with JIA. Secretion of DEK by synovial macrophages was observed both in a free form and via exosomes. DEK autoantibodies (IgG2) may activate the complement cascade, primarily recognize the C-terminal portion of DEK protein, and exhibit higher affinity for acetylated DEK. Consistent with these observations, DEK underwent acetylation on an unprecedented number of lysine residues, as demonstrated by nano-LC-MS/MS. CONCLUSION: These results indicate that DEK can contribute directly to joint inflammation in JIA by generating ICs through high-affinity interaction between DEK and DEK autoantibodies, a process enhanced by acetylation of DEK in the inflamed joint.


Asunto(s)
Artritis Juvenil/metabolismo , Autoantígenos/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Proteínas Oncogénicas/metabolismo , Procesamiento Proteico-Postraduccional , Membrana Sinovial/metabolismo , Acetilación , Complejo Antígeno-Anticuerpo/inmunología , Complejo Antígeno-Anticuerpo/metabolismo , Artritis Juvenil/inmunología , Artritis Juvenil/patología , Autoanticuerpos/sangre , Autoantígenos/inmunología , Niño , Proteínas Cromosómicas no Histona/inmunología , Humanos , Articulaciones/metabolismo , Articulaciones/patología , Macrófagos/metabolismo , Macrófagos/patología , Proteínas Oncogénicas/inmunología , Proteínas de Unión a Poli-ADP-Ribosa , Líquido Sinovial/química , Líquido Sinovial/metabolismo , Membrana Sinovial/patología
9.
J Cutan Pathol ; 39(8): 753-7, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22765016

RESUMEN

BACKGROUND: The chromatin architectural factor DEK maps to chromosome 6p and is frequently overexpressed in several neoplasms, including small cell lung carcinoma, where it is associated with poor prognosis, tumor initiation activity and chemoresistance. DEK expression has not been studied in cutaneous Merkel cell carcinoma. METHODS: We applied a DEK monoclonal antibody to 15 cases of Merkel cell carcinoma and 12 cases of small cell carcinoma. DEK nuclear immunoreactivity was scored based on percentage (0, negative; 1+, <25%; 2+, 25-50%; 3+, >50%) and intensity (weak, moderate or strong). RESULTS: All 15 Merkel cell carcinoma cases (100%) showed diffuse (3+) nuclear positivity (14 strong, 1 weak). Six of 12 small cell carcinoma cases (50%) showed diffuse (3+) and strong nuclear positivity, while one case exhibited focal (1+) weak nuclear positivity. The remaining five cases were negative for DEK expression. CONCLUSIONS: Our results suggest that DEK may be involved in the pathogenesis of Merkel cell carcinoma and therefore may provide therapeutic implications for Merkel cell carcinomas. In addition, the difference in DEK expression between Merkel cell carcinoma and small cell carcinoma suggests possible separate tumorigenesis pathways for the two tumors.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Carcinoma de Células de Merkel/metabolismo , Carcinoma de Células Pequeñas/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Neoplasias Pulmonares/metabolismo , Proteínas Oncogénicas/metabolismo , Neoplasias Cutáneas/metabolismo , Anciano , Anciano de 80 o más Años , Carcinoma de Células de Merkel/secundario , Carcinoma de Células de Merkel/cirugía , Carcinoma de Células Pequeñas/secundario , Carcinoma de Células Pequeñas/cirugía , Núcleo Celular/metabolismo , Núcleo Celular/patología , Femenino , Humanos , Neoplasias Pulmonares/patología , Ganglios Linfáticos/patología , Metástasis Linfática , Masculino , Persona de Mediana Edad , Proteínas de Unión a Poli-ADP-Ribosa , Estudios Retrospectivos , Neoplasias Cutáneas/patología , Neoplasias Cutáneas/cirugía , Análisis de Matrices Tisulares
10.
FEBS Lett ; 595(10): 1422-1437, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33704777

RESUMEN

In two proof-of-concept studies, we established and validated the Bacterial Growth Inhibition Screen (BGIS), which explores recombinant protein toxicity in Escherichia coli as a largely overlooked and alternative means for basic characterization of functional eukaryotic protein domains. By applying BGIS, we identified an unrecognized RNA-interacting domain in the DEK oncoprotein (this study) and successfully combined BGIS with random mutagenesis as a screening tool for loss-of-function mutants of the DNA modulating domain of DEK [1]. Collectively, our findings shed new light on the phenomenon of recombinant protein toxicity in E. coli. Given the easy and rapid implementation and wide applicability, BGIS will extend the repertoire of basic methods for the identification, analysis and unbiased manipulation of proteins.


Asunto(s)
Escherichia coli/efectos de los fármacos , Escherichia coli/crecimiento & desarrollo , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/toxicidad , Pruebas de Toxicidad/métodos , Animales , Sesgo , Biocatálisis , Proteínas Cromosómicas no Histona/química , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Proteínas Cromosómicas no Histona/toxicidad , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/toxicidad , Escherichia coli/genética , Humanos , Mutación con Pérdida de Función , Proteínas Oncogénicas/química , Proteínas Oncogénicas/genética , Proteínas Oncogénicas/metabolismo , Proteínas Oncogénicas/toxicidad , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/toxicidad , Proteínas de Unión a Poli-ADP-Ribosa/química , Proteínas de Unión a Poli-ADP-Ribosa/genética , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa/toxicidad , Dominios Proteicos/genética , ARN/metabolismo , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/toxicidad , Receptores de la Familia Eph/química , Receptores de la Familia Eph/genética , Receptores de la Familia Eph/metabolismo , Receptores de la Familia Eph/toxicidad , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Reproducibilidad de los Resultados , Factores de Tiempo , Pruebas de Toxicidad/normas
11.
FEBS Lett ; 595(10): 1438-1453, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33686684

RESUMEN

The DEK oncoprotein regulates cellular chromatin function via a number of protein-protein interactions. However, the biological relevance of its unique pseudo-SAP/SAP-box domain, which transmits DNA modulating activities in vitro, remains largely speculative. As hypothesis-driven mutations failed to yield DNA-binding null (DBN) mutants, we combined random mutagenesis with the Bacterial Growth Inhibition Screen (BGIS) to overcome this bottleneck. Re-expression of a DEK-DBN mutant in newly established human DEK knockout cells failed to reduce the increase in nuclear size as compared to wild type, indicating roles for DEK-DNA interactions in cellular chromatin organization. Our results extend the functional roles of DEK in metazoan chromatin and highlight the predictive ability of recombinant protein toxicity in E. coli for unbiased studies of eukaryotic DNA modulating protein domains.


Asunto(s)
Cromatina/metabolismo , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , ADN/metabolismo , Escherichia coli/efectos de los fármacos , Mutación con Pérdida de Función , Proteínas Oncogénicas/genética , Proteínas Oncogénicas/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa/genética , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Proteínas Recombinantes/toxicidad , Sesgo , Núcleo Celular/efectos de los fármacos , Tamaño de la Célula/efectos de los fármacos , Cromatina/química , Cromatina/genética , Proteínas Cromosómicas no Histona/química , Proteínas Cromosómicas no Histona/toxicidad , Escherichia coli/genética , Escherichia coli/crecimiento & desarrollo , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Genoma Bacteriano/efectos de los fármacos , Genoma Bacteriano/genética , Humanos , Mutagénesis , Nucleosomas/química , Nucleosomas/genética , Nucleosomas/metabolismo , Proteínas Oncogénicas/química , Proteínas Oncogénicas/toxicidad , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/toxicidad , Proteínas de Unión a Poli-ADP-Ribosa/química , Proteínas de Unión a Poli-ADP-Ribosa/toxicidad , Dominios Proteicos/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Pruebas de Toxicidad/métodos
12.
Biochemistry ; 49(33): 7119-30, 2010 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-20669926

RESUMEN

Poly(ADP-ribose) polymerase-1 (PARP-1) is a molecular DNA damage sensor that catalyzes the synthesis of the complex biopolymer poly(ADP-ribose) (PAR) under consumption of NAD(+). PAR engages in fundamental cellular processes such as DNA metabolism and transcription and interacts noncovalently with specific binding proteins involved in DNA repair and regulation of chromatin structure. A factor implicated in DNA repair and chromatin organization is the DEK oncoprotein, an abundant and conserved constituent of metazoan chromatin, and the only member of its protein class. We have recently demonstrated that DEK, under stress conditions, is covalently modified with PAR by PARP-1, leading to a partial release of DEK into the cytoplasm. Additionally, we have also observed a noncovalent interaction between DEK and PAR, which we detail here. Using sequence alignment, we identify three functional PAR-binding sites in the DEK primary sequence and confirm their functionality in PAR binding studies. Furthermore, we show that the noncovalent binding to DEK is dependent on PAR chain length as revealed by an overlay blot technique and a PAR electrophoretic mobility shift assay. Intriguingly, DEK promotes the formation of a defined complex with a 54mer PAR (K(D) = 6 x 10(-8) M), whereas no specific interaction is detected with a short PAR chain (18mer). In stark contrast to covalent poly(ADP-ribosyl)ation of DEK, the noncovalent interaction does not affect the overall ability of DEK to bind to DNA. Instead the noncovalent interaction interferes with subsequent DNA-dependent multimerization activities of DEK, as seen in South-Western, electrophoretic mobility shift, topology, and aggregation assays. In particular, noncovalent attachment of PAR to DEK promotes the formation of DEK-DEK complexes by competing with DNA binding. This was seen by the reduced affinity of PAR-bound DEK for DNA templates in solution. Taken together, our findings deepen the molecular understanding of the DEK-PAR interplay and support the existence of a cellular "PAR code" represented by PAR chain length.


Asunto(s)
Proteínas Cromosómicas no Histona/metabolismo , Proteínas Oncogénicas/metabolismo , Poli Adenosina Difosfato Ribosa/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Proteínas Cromosómicas no Histona/química , ADN/metabolismo , Humanos , Datos de Secuencia Molecular , Proteínas Oncogénicas/química , Poli Adenosina Difosfato Ribosa/química , Proteínas de Unión a Poli-ADP-Ribosa , Multimerización de Proteína , Estructura Terciaria de Proteína , Alineación de Secuencia
13.
Mol Cell Biol ; 26(24): 9484-96, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17030615

RESUMEN

The nuclear DNA-binding protein DEK is an autoantigen that has been implicated in the regulation of transcription, chromatin architecture, and mRNA processing. We demonstrate here that DEK is actively secreted by macrophages and is also found in synovial fluid samples from patients with juvenile arthritis. Secretion of DEK is modulated by casein kinase 2, stimulated by interleukin-8, and inhibited by dexamethasone and cyclosporine A, consistent with a role as a proinflammatory molecule. DEK is secreted in both a free form and in exosomes, vesicular structures in which transcription-modulating factors such as DEK have not previously been found. Furthermore, DEK functions as a chemotactic factor, attracting neutrophils, CD8+ T lymphocytes, and natural killer cells. Therefore, the DEK autoantigen, previously described as a strictly nuclear protein, is secreted and can act as an extracellular chemoattractant, suggesting a direct role for DEK in inflammation.


Asunto(s)
Artritis Juvenil/metabolismo , Artritis Juvenil/patología , Autoantígenos/fisiología , Factores Quimiotácticos/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Proteínas Oncogénicas/metabolismo , Artritis Juvenil/inmunología , Autoantígenos/metabolismo , Células Cultivadas , Factores Quimiotácticos/fisiología , Quimiotaxis de Leucocito , Niño , Proteínas Cromosómicas no Histona/fisiología , Líquido Extracelular/inmunología , Líquido Extracelular/metabolismo , Humanos , Macrófagos/inmunología , Macrófagos/metabolismo , Monocitos/citología , Monocitos/inmunología , Monocitos/metabolismo , Proteínas Oncogénicas/fisiología , Proteínas de Unión a Poli-ADP-Ribosa
14.
PLoS One ; 14(8): e0213130, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31408463

RESUMEN

DNA replication stress is a major source of genomic instability and is closely linked to tumor formation and progression. Poly(ADP-ribose)polymerases1/2 (PARP1/2) enzymes are activated in response to replication stress resulting in poly(ADP-ribose) (PAR) synthesis. PARylation plays an important role in the remodelling and repair of impaired replication forks, providing a rationale for targeting highly replicative cancer cells with PARP1/2 inhibitors. The human oncoprotein DEK is a unique, non-histone chromatin architectural protein whose deregulated expression is associated with the development of a wide variety of human cancers. Recently, we showed that DEK is a high-affinity target of PARylation and that it promotes the progression of impaired replication forks. Here, we investigated a potential functional link between PAR and DEK in the context of replication stress. Under conditions of mild replication stress induced either by topoisomerase1 inhibition with camptothecin or nucleotide depletion by hydroxyurea, we found that the effect of acute PARP1/2 inhibition on replication fork progression is dependent on DEK expression. Reducing DEK protein levels also overcomes the restart impairment of stalled forks provoked by blocking PARylation. Non-covalent DEK-PAR interaction via the central PAR-binding domain of DEK is crucial for counteracting PARP1/2 inhibition as shown for the formation of RPA positive foci in hydroxyurea treated cells. Finally, we show by iPOND and super resolved microscopy that DEK is not directly associated with the replisome since it binds to DNA at the stage of chromatin formation. Our report sheds new light on the still enigmatic molecular functions of DEK and suggests that DEK expression levels may influence the sensitivity of cancer cells to PARP1/2 inhibitors.


Asunto(s)
Neoplasias Óseas/patología , Proteínas Cromosómicas no Histona/metabolismo , Daño del ADN , Replicación del ADN , Proteínas Oncogénicas/metabolismo , Osteosarcoma/patología , Poli(ADP-Ribosa) Polimerasa-1/antagonistas & inhibidores , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Poli(ADP-Ribosa) Polimerasas/química , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/genética , Neoplasias Óseas/metabolismo , Proteínas Cromosómicas no Histona/genética , Inestabilidad Genómica , Humanos , Proteínas Oncogénicas/genética , Osteosarcoma/tratamiento farmacológico , Osteosarcoma/genética , Osteosarcoma/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa/genética , Células Tumorales Cultivadas
15.
J Clin Invest ; 129(6): 2555-2570, 2019 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-31107242

RESUMEN

The nuclear protein DEK is an endogenous DNA-binding chromatin factor regulating hematopoiesis. DEK is one of only 2 known secreted nuclear chromatin factors, but whether and how extracellular DEK regulates hematopoiesis is not known. We demonstrated that extracellular DEK greatly enhanced ex vivo expansion of cytokine-stimulated human and mouse hematopoietic stem cells (HSCs) and regulated HSC and hematopoietic progenitor cell (HPC) numbers in vivo and in vitro as determined both phenotypically (by flow cytometry) and functionally (through transplantation and colony formation assays). Recombinant DEK increased long-term HSC numbers and decreased HPC numbers through a mechanism mediated by the CXC chemokine receptor CXCR2 and heparan sulfate proteoglycans (HSPGs) (as determined utilizing Cxcr2-/- mice, blocking CXCR2 antibodies, and 3 different HSPG inhibitors) that was associated with enhanced phosphorylation of ERK1/2, AKT, and p38 MAPK. To determine whether extracellular DEK required nuclear function to regulate hematopoiesis, we utilized 2 mutant forms of DEK: one that lacked its nuclear translocation signal and one that lacked DNA-binding ability. Both altered HSC and HPC numbers in vivo or in vitro, suggesting the nuclear function of DEK is not required. Thus, DEK acts as a hematopoietic cytokine, with the potential for clinical applicability.


Asunto(s)
Proteínas Cromosómicas no Histona/metabolismo , Citocinas/metabolismo , Proteínas de Unión al ADN/metabolismo , Hematopoyesis , Células Madre Hematopoyéticas/metabolismo , Sistema de Señalización de MAP Quinasas , Proteínas Oncogénicas/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Animales , Proteínas Cromosómicas no Histona/genética , Citocinas/genética , Proteínas de Unión al ADN/genética , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Ratones , Ratones Noqueados , Mutación , Proteínas Oncogénicas/genética , Proteínas de Unión a Poli-ADP-Ribosa/genética , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas Sprague-Dawley , Receptores de Interleucina-8B
16.
Protein Sci ; 17(2): 205-15, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18227428

RESUMEN

The human DEK protein has a long-standing association with carcinogenesis since the DEK gene was originally identified in the t(6:9) chromosomal translocation in a subtype of patients with acute myelogenous leukemia (AML). Recent studies have partly unveiled DEK's cellular functions including apoptosis inhibition in primary cells as well as cancer cells, determination of 3' splice site of transcribed RNA, and suppression of transcription initiation by polymerase II. It has been previously shown that the N-terminal region of DEK, spanning residues 68-226, confers important in vitro and in vivo functions of DEK, which include double-stranded DNA (ds-DNA) binding, introduction of constrained positive supercoils into closed dsDNA, and apoptosis inhibition. In this paper, we describe the three-dimensional structure of the N-terminal domain of DEK (DEKntd) as determined using solution NMR. The C-terminal part of DEKntd, which contains a putative DNA-binding motif (SAF/SAP motif), folds into a helix-loop-helix structure. Interestingly, the N-terminal part of DEKntd shows a very similar structure to the C-terminal part, although the N-terminal and the C-terminal part differ distinctively in their amino acid sequences. As a consequence, the structure of DEKntd has a pseudo twofold plane symmetry. In addition, we tested dsDNA binding of DEKntd by monitoring changes of NMR chemical shifts upon addition of dsDNAs. We found that not only the C-terminal part containing the SAF/SAP motif but the N-terminal part is also involved in DEKntd's dsDNA binding. Our study illustrates a new structural variant and reveals novel dsDNA-binding properties for proteins containing the SAP/SAF motif.


Asunto(s)
Proteínas Cromosómicas no Histona/química , Proteínas Oncogénicas/química , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Proteínas Cromosómicas no Histona/metabolismo , ADN/metabolismo , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Humanos , Datos de Secuencia Molecular , Resonancia Magnética Nuclear Biomolecular , Proteínas Oncogénicas/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa , Conformación Proteica , Pliegue de Proteína , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína
17.
Biomed Pharmacother ; 106: 142-148, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29957464

RESUMEN

Breast and cervical cancer are the first and fourth cancer types with the highest prevalence in women, respectively. The developmental profiles of cancer in women can vary by genetic markers and cellular events. In turn, age and lifestyle influence in the cellular response and also on the cancer progression and relapse. The human DEK protein, a histone chaperone, belongs to a specific subclass of chromatin topology modulators, being involved in the regulation of DNA-dependent processes. These epigenetic mechanisms have dynamic and reversible nature, have been proposed as targets for different treatment approaches, especially in tumor therapy. The expression patterns of DEK vary between healthy and cancer cells. High expression of DEK is associated with poor prognosis in many cancer types, suggesting that DEK takes part in oncogenic activities via different molecular pathways, including inhibition of senescence and apoptosis. The focus of this review was to highlight the role of the DEK protein in these two female cancers.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Proteínas Oncogénicas/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Neoplasias del Cuello Uterino/metabolismo , Salud de la Mujer , Animales , Biomarcadores de Tumor/química , Biomarcadores de Tumor/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Neoplasias de la Mama/terapia , Proteínas Cromosómicas no Histona/química , Proteínas Cromosómicas no Histona/genética , Epigénesis Genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Proteínas Oncogénicas/química , Proteínas Oncogénicas/genética , Proteínas de Unión a Poli-ADP-Ribosa/química , Proteínas de Unión a Poli-ADP-Ribosa/genética , Pronóstico , Conformación Proteica , Relación Estructura-Actividad , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/patología , Neoplasias del Cuello Uterino/terapia
18.
Sci Rep ; 8(1): 6748, 2018 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-29712969

RESUMEN

Macrodomains are conserved protein folds associated with ADP-ribose binding and turnover. ADP-ribosylation is a posttranslational modification catalyzed primarily by ARTD (aka PARP) enzymes in cells. ARTDs transfer either single or multiple ADP-ribose units to substrates, resulting in mono- or poly-ADP-ribosylation. TARG1/C6orf130 is a macrodomain protein that hydrolyzes mono-ADP-ribosylation and interacts with poly-ADP-ribose chains. Interactome analyses revealed that TARG1 binds strongly to ribosomes and proteins associated with rRNA processing and ribosomal assembly factors. TARG1 localized to transcriptionally active nucleoli, which occurred independently of ADP-ribose binding. TARG1 shuttled continuously between nucleoli and nucleoplasm. In response to DNA damage, which activates ARTD1/2 (PARP1/2) and promotes synthesis of poly-ADP-ribose chains, TARG1 re-localized to the nucleoplasm. This was dependent on the ability of TARG1 to bind to poly-ADP-ribose. These findings are consistent with the observed ability of TARG1 to competitively interact with RNA and PAR chains. We propose a nucleolar role of TARG1 in ribosome assembly or quality control that is stalled when TARG1 is re-located to sites of DNA damage.


Asunto(s)
Núcleo Celular/genética , Daño del ADN/genética , Poli ADP Ribosilación/genética , Tioléster Hidrolasas/genética , Adenosina Difosfato Ribosa/genética , Adenosina Difosfato Ribosa/metabolismo , Núcleo Celular/metabolismo , Humanos , Proteínas del Complejo de Iniciación de Transcripción Pol1/genética , Poli(ADP-Ribosa) Polimerasa-1/genética , Poli Adenosina Difosfato Ribosa/genética , Poli(ADP-Ribosa) Polimerasas/genética , Procesamiento Proteico-Postraduccional/genética
19.
Mol Cell Biol ; 24(13): 6000-10, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15199153

RESUMEN

DEK was originally described as a proto-oncogene protein and is now known to be a major component of metazoan chromatin. DEK is able to modify the structure of DNA by introducing supercoils. In order to find interaction partners and functional domains of DEK, we performed yeast two-hybrid screens and mutational analyses. Two-hybrid screening yielded C-terminal fragments of DEK, suggesting that DEK is able to multimerize. We could localize the domain to amino acids 270 to 350 and show that multimerization is dependent on phosphorylation by CK2 kinase in vitro. We also found two DNA binding domains of DEK, one on a fragment including amino acids 87 to 187 and containing the SAF-box DNA binding motif, which is located between amino acids 149 and 187. This region is sufficient to introduce supercoils into DNA. The second DNA binding domain is located between amino acids 270 and 350 and thus overlaps the multimerization domain. We show that the two DNA-interacting domains differ in their binding properties and in their abilities to respond to CK2 phosphorylation.


Asunto(s)
Proteínas Cromosómicas no Histona/metabolismo , Proteínas Oncogénicas/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Quinasa de la Caseína II , Proteínas Cromosómicas no Histona/química , Proteínas Cromosómicas no Histona/fisiología , ADN Superhelicoidal/metabolismo , Dimerización , Células HeLa , Humanos , Proteínas Oncogénicas/química , Proteínas Oncogénicas/fisiología , Fosforilación , Proteínas de Unión a Poli-ADP-Ribosa , Proteínas Serina-Treonina Quinasas/metabolismo , Estructura Terciaria de Proteína/fisiología , Proto-Oncogenes Mas , Técnicas del Sistema de Dos Híbridos
20.
Mol Cell Biol ; 24(13): 6011-20, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15199154

RESUMEN

We have examined the posttranslational modification of the human chromatin protein DEK and found that DEK is phosphorylated by the protein kinase CK2 in vitro and in vivo. Phosphorylation sites were mapped by quadrupole ion trap mass spectrometry and found to be clustered in the C-terminal region of the DEK protein. Phosphorylation fluctuates during the cell cycle with a moderate peak during G(1) phase. Filter binding assays, as well as Southwestern analysis, demonstrate that phosphorylation weakens the binding of DEK to DNA. In vivo, however, phosphorylated DEK remains on chromatin. We present evidence that phosphorylated DEK is tethered to chromatin throughout the cell cycle by the un- or underphosphorylated form of DEK.


Asunto(s)
Proteínas Cromosómicas no Histona/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas Oncogénicas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Quinasa de la Caseína II , Ciclo Celular , Cromatina/metabolismo , ADN/metabolismo , Células HeLa , Humanos , Espectrometría de Masas , Fosforilación , Proteínas de Unión a Poli-ADP-Ribosa , Procesamiento Proteico-Postraduccional , Fase de Descanso del Ciclo Celular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA