Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
EMBO J ; 42(12): e111272, 2023 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-37143403

RESUMEN

Patients with chronic obstructive pulmonary disease (COPD) are still waiting for curative treatments. Considering its environmental cause, we hypothesized that COPD will be associated with altered epigenetic signaling in lung cells. We generated genome-wide DNA methylation maps at single CpG resolution of primary human lung fibroblasts (HLFs) across COPD stages. We show that the epigenetic landscape is changed early in COPD, with DNA methylation changes occurring predominantly in regulatory regions. RNA sequencing of matched fibroblasts demonstrated dysregulation of genes involved in proliferation, DNA repair, and extracellular matrix organization. Data integration identified 110 candidate regulators of disease phenotypes that were linked to fibroblast repair processes using phenotypic screens. Our study provides high-resolution multi-omic maps of HLFs across COPD stages. We reveal novel transcriptomic and epigenetic signatures associated with COPD onset and progression and identify new candidate regulators involved in the pathogenesis of chronic lung diseases. The presence of various epigenetic factors among the candidates demonstrates that epigenetic regulation in COPD is an exciting research field that holds promise for novel therapeutic avenues for patients.


Asunto(s)
Enfermedad Pulmonar Obstructiva Crónica , Transcriptoma , Humanos , Epigénesis Genética , Enfermedad Pulmonar Obstructiva Crónica/genética , Enfermedad Pulmonar Obstructiva Crónica/patología , Pulmón/patología , Perfilación de la Expresión Génica , Metilación de ADN
2.
Int J Cancer ; 146(12): 3423-3434, 2020 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-31745977

RESUMEN

Protein kinase D3 (PKD3) is upregulated in triple-negative breast cancer (TNBC) and associated with cell proliferation and metastasis development but its precise pro-oncogenic function is unknown. Here we show that PKD3 is required for the maintenance of the TNBC stem cell population. The depletion of PKD3 in MDA-MB-231 cells reduced the cancer stem cell frequency in vitro and tumor initiation potential in vivo. We further provide evidence that the RhoGEF GEF-H1 is upstream of PKD3 activation in TNBC stem cells. Most importantly, pharmacological PKD inhibition in combination with paclitaxel synergistically decreased oncosphere and colony formation efficiency in vitro and tumor recurrence in vivo. Based on our results we propose that targeting the GEF-H1/PKD3 signaling pathway in combination with chemotherapy might provide an effective therapeutic option for TNBC.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Células Madre Neoplásicas/patología , Proteína Quinasa C/metabolismo , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Neoplasias de la Mama Triple Negativas/patología , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Apoptosis/efectos de los fármacos , Apoptosis/genética , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Proliferación Celular/efectos de los fármacos , Supervivencia Celular , Sinergismo Farmacológico , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Células Madre Neoplásicas/efectos de los fármacos , Paclitaxel/farmacología , Paclitaxel/uso terapéutico , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/genética , Pirimidinas/farmacología , Pirimidinas/uso terapéutico , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Genome Res ; 24(11): 1842-53, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25301795

RESUMEN

Post-translational modifications (PTMs) of histones constitute a major chromatin indexing mechanism, and their proper characterization is of highest biological importance. So far, PTM-specific antibodies have been the standard reagent for studying histone PTMs despite caveats such as lot-to-lot variability of specificity and binding affinity. Herein, we successfully employed naturally occurring and engineered histone modification interacting domains for detection and identification of histone PTMs and ChIP-like enrichment of different types of chromatin. Our results demonstrate that histone interacting domains are robust and highly specific reagents that can replace or complement histone modification antibodies. These domains can be produced recombinantly in Escherichia coli at low cost and constant quality. Protein design of reading domains allows for generation of novel specificities, addition of affinity tags, and preparation of PTM binding pocket variants as matching negative controls, which is not possible with antibodies.


Asunto(s)
Anticuerpos/metabolismo , Histonas/metabolismo , Mapeo de Interacción de Proteínas/métodos , Procesamiento Proteico-Postraduccional , Secuencia de Aminoácidos , Anticuerpos/inmunología , Sitios de Unión/genética , Western Blotting , Inmunoprecipitación de Cromatina , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , ADN Metiltransferasa 3A , Células HEK293 , Histonas/inmunología , Humanos , Lisina/metabolismo , Metilación , Péptidos/metabolismo , Complejo Represivo Polycomb 1/genética , Complejo Represivo Polycomb 1/metabolismo , Análisis por Matrices de Proteínas/métodos , Unión Proteica , Reproducibilidad de los Resultados
4.
Hum Mol Genet ; 20(11): 2195-203, 2011 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-21421568

RESUMEN

Mutations in the ATRX protein are associated with the alpha-thalassemia and mental retardation X-linked syndrome (ATR-X). Almost half of the disease-causing mutations occur in its ATRX-Dnmt3-Dnmt3L (ADD) domain. By employing peptide arrays, chromatin pull-down and peptide binding assays, we show specific binding of the ADD domain to H3 histone tail peptides containing H3K9me3. Peptide binding was disrupted by the presence of the H3K4me3 and H3K4me2 modification marks indicating that the ATRX-ADD domain has a combined readout of these two important marks (absence of H3K4me2 and H3K4me3 and presence of H3K9me3). Disease-causing mutations reduced ATRX-ADD binding to H3 tail peptides. ATRX variants, which fail in the H3K9me3 interaction, show a loss of heterochromatic localization in cells, which indicates the chromatin targeting function of the ADD domain of ATRX. Disruption of H3K9me3 binding may be a general pathogenicity pathway of ATRX mutations in the ADD domain which may explain the clustering of disease mutations in this part of the ATRX protein.


Asunto(s)
ADN Helicasas/metabolismo , Histonas/metabolismo , Lisina/metabolismo , Proteínas Nucleares/metabolismo , Péptidos/metabolismo , Células 3T3 , Secuencia de Aminoácidos , Animales , Cromatina/metabolismo , Clonación Molecular , ADN Helicasas/genética , Expresión Génica , Células HEK293 , Humanos , Lisina/genética , Discapacidad Intelectual Ligada al Cromosoma X/genética , Discapacidad Intelectual Ligada al Cromosoma X/patología , Metilación , Ratones , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Mutación , Proteínas Nucleares/genética , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Estructura Cuaternaria de Proteína , Proteína Nuclear Ligada al Cromosoma X , Talasemia alfa/genética , Talasemia alfa/patología
5.
J Biol Chem ; 285(34): 26114-20, 2010 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-20547484

RESUMEN

The Dnmt3a DNA methyltransferase contains in its N-terminal part a PWWP domain that is involved in chromatin targeting. Here, we have investigated the interaction of the PWWP domain with modified histone tails using peptide arrays and show that it specifically recognizes the histone 3 lysine 36 trimethylation mark. H3K36me3 is known to be a repressive modification correlated with DNA methylation in mammals and heterochromatin in Schizosaccharomyces pombe. These results were confirmed by equilibrium peptide binding studies and pulldown experiments with native histones and purified native nucleosomes. The PWWP-H3K36me3 interaction is important for the subnuclear localization of enhanced yellow fluorescent protein-fused Dnmt3a. Furthermore, the PWWP-H3K36me3 interaction increases the activity of Dnmt3a for methylation of nucleosomal DNA as observed using native nucleosomes isolated from human cells after demethylation of the DNA with 5-aza-2'-deoxycytidine as substrate for methylation with Dnmt3a. These data suggest that the interaction of the PWWP domain with H3K36me3 is involved in targeting of Dnmt3a to chromatin carrying that mark, a model that is in agreement with several studies on the genome-wide distribution of DNA methylation and H3K36me3.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/fisiología , Metilación de ADN , Histonas/metabolismo , Animales , Línea Celular , ADN (Citosina-5-)-Metiltransferasas/genética , ADN Metiltransferasa 3A , Humanos , Lisina/metabolismo , Metilación , Ratones , Análisis por Matrices de Proteínas , Unión Proteica , Estructura Terciaria de Proteína , Transfección
6.
BMC Biochem ; 12: 48, 2011 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-21884582

RESUMEN

BACKGROUND: Epigenetic reading domains are involved in the regulation of gene expression and chromatin state by interacting with histones in a post-translational modification specific manner. A detailed knowledge of the target modifications of reading domains, including enhancing and inhibiting secondary modifications, will lead to a better understanding of the biological signaling processes mediated by reading domains. RESULTS: We describe the application of Celluspots peptide arrays which contain 384 histone peptides carrying 59 post translational modifications in different combinations as an inexpensive, reliable and fast method for initial screening for specific interactions of reading domains with modified histone peptides. To validate the method, we tested the binding specificities of seven known epigenetic reading domains on Celluspots peptide arrays, viz. the HP1ß and MPP8 Chromo domains, JMJD2A and 53BP1 Tudor domains, Dnmt3a PWWP domain, Rag2 PHD domain and BRD2 Bromo domain. In general, the binding results agreed with literature data with respect to the primary specificity of the reading domains, but in almost all cases we obtained additional new information concerning the influence of secondary modifications surrounding the target modification. CONCLUSIONS: We conclude that Celluspots peptide arrays are powerful screening tools for studying the specificity of putative reading domains binding to modified histone peptides.


Asunto(s)
Epigénesis Genética , Histonas/química , Histonas/metabolismo , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Análisis por Matrices de Proteínas/métodos , Proteínas Cromosómicas no Histona/química , Proteínas Cromosómicas no Histona/metabolismo , ADN (Citosina-5-)-Metiltransferasas/química , ADN (Citosina-5-)-Metiltransferasas/metabolismo , ADN Metiltransferasa 3A , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/química , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Histona Demetilasas con Dominio de Jumonji/química , Histona Demetilasas con Dominio de Jumonji/metabolismo , Modelos Moleculares , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Unión Proteica , Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/metabolismo , Estructura Terciaria de Proteína , Especificidad por Sustrato , Factores de Transcripción , Proteína 1 de Unión al Supresor Tumoral P53
7.
JCI Insight ; 6(6)2021 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-33630765

RESUMEN

Complexity of lung microenvironment and changes in cellular composition during disease make it exceptionally hard to understand molecular mechanisms driving development of chronic lung diseases. Although recent advances in cell type-resolved approaches hold great promise for studying complex diseases, their implementation relies on local access to fresh tissue, as traditional tissue storage methods do not allow viable cell isolation. To overcome these hurdles, we developed a versatile workflow that allows storage of lung tissue with high viability, permits thorough sample quality check before cell isolation, and befits sequencing-based profiling. We demonstrate that cryopreservation enables isolation of multiple cell types from both healthy and diseased lungs. Basal cells from cryopreserved airways retain their differentiation ability, indicating that cellular identity is not altered by cryopreservation. Importantly, using RNA sequencing and EPIC Array, we show that gene expression and DNA methylation signatures are preserved upon cryopreservation, emphasizing the suitability of our workflow for omics profiling of lung cells. Moreover, we obtained high-quality single-cell RNA-sequencing data of cells from cryopreserved human lungs, demonstrating that cryopreservation empowers single-cell approaches. Overall, thanks to its simplicity, our workflow is well suited for prospective tissue collection by academic collaborators and biobanks, opening worldwide access to viable human tissue.


Asunto(s)
Criopreservación , Epigénesis Genética , Pulmón/metabolismo , Transcripción Genética , Metilación de ADN , Expresión Génica , Humanos , Pulmón/citología , Análisis de Secuencia de ARN/métodos , Flujo de Trabajo
8.
Nat Chem Biol ; 4(6): 344-6, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18438403

RESUMEN

By methylation of peptide arrays, we determined the specificity profile of the protein methyltransferase G9a. We show that it mostly recognizes an Arg-Lys sequence and that its activity is inhibited by methylation of the arginine residue. Using the specificity profile, we identified new non-histone protein targets of G9a, including CDYL1, WIZ, ACINUS and G9a (automethylation), as well as peptides derived from CSB. We demonstrate potential downstream signaling pathways for methylation of non-histone proteins.


Asunto(s)
N-Metiltransferasa de Histona-Lisina/química , Histona Metiltransferasas , N-Metiltransferasa de Histona-Lisina/metabolismo , Metilación , Análisis por Matrices de Proteínas , Proteína Metiltransferasas , Transducción de Señal , Especificidad por Sustrato
9.
Histochem Cell Biol ; 131(1): 103-14, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18648844

RESUMEN

The fibroblast-like synoviocyte is a CD13-positive cell-type containing numerous caveolae, both single and interconnected clusters. In unstimulated cells, all single caveolae at the cell surface and the majority of those localized deeper into the cytoplasm were freely accessible from the medium, as judged from electron microscopy of synoviocytes exposed to the membrane impermeable marker Ruthenium Red. Caveolar internalization could be induced by a CD13 antibody or by cholera toxin B subunit (CTB). Thus, in experiments using sequential labeling with Alexa 488- and 594-conjugated CTB, about 50% of CTB-positive caveolae were internalized by 5 min of chase, and these remained inaccessible from the cell surface for periods up to 24 h. No colocalization with an endosomal marker, EEA1, or Lysotracker was observed, indicating that internalized caveolae clusters represent a static compartment. Vimentin was identified as the most abundant protein in detergent resistant membranes (DRM's), and by immunogold electron microscopy caveolae were seen in intimate contact with intermediate-size filaments. These observations indicate that vimentin-based filaments are responsible for the spatio-temporal fixation of caveolae clusters. RECK, a glycosylphosphatidylinositol-anchored protein acting as a negative regulator of cell surface metalloproteinases, was also localized to the caveolae clusters. We propose that these clusters function as static reservoirs of specialized lipid raft domains where proteins involved in cell-cell interactions, such as CD13, can be sequestered by binding to RECK in a regulatory manner.


Asunto(s)
Caveolas/metabolismo , Fibroblastos/citología , Membrana Sinovial/citología , Vimentina/metabolismo , Antígenos CD13/metabolismo , Caveolas/ultraestructura , Fibroblastos/metabolismo , Humanos , Microscopía Fluorescente , Membrana Sinovial/metabolismo
10.
Epigenetics Chromatin ; 11(1): 4, 2018 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-29370823

RESUMEN

BACKGROUND: Protein posttranslational modifications (PTMs) occur broadly in the human proteome, and their biological outcome is often mediated indirectly by reader proteins that specifically bind to modified proteins and trigger downstream effects. Particularly, many lysine methylation sites among histone and nonhistone proteins have been characterized; however, the list of readers associated with them is incomplete. RESULTS: This study introduces a modified yeast three-hybrid system (Y3H) to screen for methyllysine readers. A lysine methyltransferase is expressed together with its target protein or protein domain functioning as bait, and a human cDNA library serves as prey. Proof of principle was established using H3K9me3 as a bait and known H3K9me3 readers like the chromodomains of CBX1 or MPP8 as prey. We next conducted an unbiased screen using a library composed of human-specific open reading frames. It led to the identification of already known lysine methylation-dependent readers and of novel methyllysine reader candidates, which were further confirmed by co-localization with H3K9me3 in human cell nuclei. CONCLUSIONS: Our approach introduces a cost-effective method for screening reading domains binding to histone and nonhistone proteins which is not limited by expression levels of the candidate reading proteins. Identification of already known and novel H3K9me3 readers proofs the power of the Y3H assay which will allow for proteome-wide screens of PTM readers.


Asunto(s)
Proteínas Cromosómicas no Histona/química , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Lisina/química , Fosfoproteínas/química , Animales , Línea Celular , Homólogo de la Proteína Chromobox 5 , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Clonación Molecular , Epigénesis Genética , Biblioteca de Genes , N-Metiltransferasa de Histona-Lisina/genética , Humanos , Metilación , Ratones , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Procesamiento Proteico-Postraduccional , Técnicas del Sistema de Dos Híbridos
11.
Nat Commun ; 8(1): 2057, 2017 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-29234025

RESUMEN

SETDB1 is an essential H3K9 methyltransferase involved in silencing of retroviruses and gene regulation. We show here that its triple Tudor domain (3TD) specifically binds to doubly modified histone H3 containing K14 acetylation and K9 methylation. Crystal structures of 3TD in complex with H3K14ac/K9me peptides reveal that peptide binding and K14ac recognition occurs at the interface between Tudor domains (TD) TD2 and TD3. Structural and biochemical data demonstrate a pocket switch mechanism in histone code reading, because K9me1 or K9me2 is preferentially recognized by the aromatic cage of TD3, while K9me3 selectively binds to TD2. Mutations in the K14ac/K9me binding sites change the sub-nuclear localization of 3TD. ChIP-seq analyses show that SETDB1 is enriched at H3K9me3 regions and K9me3/K14ac is enriched at SETDB1 binding sites overlapping with LINE elements, suggesting that recruitment of the SETDB1 complex to K14ac/K9me regions has a role in silencing of active genomic regions.


Asunto(s)
Histonas/metabolismo , Elementos de Nucleótido Esparcido Largo/fisiología , Proteína Metiltransferasas/fisiología , Procesamiento Proteico-Postraduccional/fisiología , Dominio Tudor/fisiología , Acetilación , Animales , Sitios de Unión/fisiología , Cristalografía por Rayos X , Células HEK293 , N-Metiltransferasa de Histona-Lisina , Histonas/genética , Humanos , Metilación , Ratones , Células Madre Embrionarias de Ratones , Unión Proteica/fisiología , Proteína Metiltransferasas/química , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
13.
J Mol Biol ; 426(8): 1651-60, 2014 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-23954330

RESUMEN

PHF1 associates with the Polycomb repressive complex 2 and it was demonstrated to stimulate its H3K27-trimethylation activity. We studied the interaction of the PHF1 Tudor domain with modified histone peptides and found that it recognizes H3K36me3 and H3tK27me3 (on the histone variant H3t) and that it uses the same trimethyllysine binding pocket for the interaction with both peptides. Since both peptide sequences are very different, this result indicates that reading domains can have dual specificities. Sub-nuclear localization studies of full-length PHF1 in human HEK293 cells revealed that it co-localizes with K27me3, but not with K36me3, and that this co-localization depends on the trimethyllysine binding pocket indicating that K27me3 is an in vivo target for the PHF1 Tudor domain. Our data suggest that PHF1 binds to H3tK27me3 in human chromatin, and H3t has a more general role in Polycomb regulation.


Asunto(s)
Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Histonas/química , Histonas/metabolismo , Factores de Transcripción/química , Factores de Transcripción/metabolismo , Secuencia de Aminoácidos , Sitios de Unión/genética , Proteínas de Unión al ADN/genética , Variación Genética , Células HEK293 , Histonas/genética , Humanos , Lisina/análogos & derivados , Lisina/química , Metilación , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Complejo Represivo Polycomb 2/metabolismo , Proteínas del Grupo Polycomb , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homología de Secuencia de Aminoácido , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA