14 research outputs found

    Dynamics and dispensability of variant-specific histone H1 Lys-26/Ser-27 and Thr-165 post-translational modifications

    Get PDF
    Jean-Michel Terme et al.In mammals, the linker histone H1, involved in DNA packaging into chromatin, is represented by a family of variants. H1 tails undergo post-translational modifications (PTMs) that can be detected by mass spectrometry. We developed antibodies to analyze several of these as yet unexplored PTMs including the combination of H1.4 K26 acetylation or trimethylation and S27 phosphorylation. H1.2-T165 phosphorylation was detected at S and G2/M phases of the cell cycle and was dispensable for chromatin binding and cell proliferation; while the H1.4-K26 residue was essential for proper cell cycle progression. We conclude that histone H1 PTMs are dynamic over the cell cycle and that the recognition of modified lysines may be affected by phosphorylation of adjacent residues. © 2014 Federation of European Biochemical Societies. Published by Elsevier B.V. All rights reserved.This work was supported by the Spanish Ministry of Science and Innovation (MICINN) and European Regional Development Fund (Grant BFU2011-23057 to A.J., and Grant BFU2008-00460 to P.S.), and by the Regional Government of Catalonia (Generalitat de Catalunya; Grant 2009-SGR-1222 to A.J.). J.-M.T. received a JAE-Doc contract from the Spanish National Research Council (CSIC)-MICINN; R.M. a TA contract from CSIC-MICINN; and L.M.-A. an FPU predoctoral fellowship from MICINNPeer Reviewe

    Histone H1 depletion triggers an interferon response in cancer cells via activation of heterochromatic repeats

    Get PDF
    Histone H1 has seven variants in human somatic cells and contributes to chromatin compaction and transcriptional regulation. Knock-down (KD) of each H1 variant in breast cancer cells results in altered gene expression and proliferation differently in a variant specific manner with H1.2 and H1.4 KDs being most deleterious. Here we show combined depletion of H1.2 and H1.4 has a strong deleterious effect resulting in a strong interferon (IFN) response, as evidenced by an up-regulation of many IFN-stimulated genes (ISGs) not seen in individual nor in other combinations of H1 variant KDs. Although H1 participates to repress ISG promoters, IFN activation upon H1.2 and H1.4 KD is mainly generated through the activation of the IFN response by cytosolic nucleic acid receptors and IFN synthesis, and without changes in histone modifications at induced ISG promoters. H1.2 and H1.4 co-KD also promotes the appearance of accessibility sites genome wide and, particularly, at satellites and other repeats. The IFN response may be triggered by the expression of noncoding RNA generated from heterochromatic repeats or endogenous retroviruses upon H1 KD. In conclusion, redundant H1-mediated silencing of heterochromatin is important to maintain cell homeostasis and to avoid an unspecific IFN response

    Genomic distribution and functional specificity of human histone H1 subtypes

    Get PDF
    Seven linker histone H1 variants exist in human somatic cells with distinct prevalence among cell types and during differentiation. Despite being key chromatin structural components, it remains elusive how they participate in the regulation of nuclear processes. Moreover, it is not well understood whether the different variants have specific roles or are differentially distributed along the genome. By taking advantage of specific antibodies for H1 variants and HA-tagged recombinant H1s expressed in breast cancer cells, the distribution of somatic variants H1.2 to H1.5, H1.0 and H1X has been investigated by combining ChIP-qPCR, ChIP-chip, and ChIP-seq analysis. All H1 variants bind gene promoters and are depleted from the TSS in active genes, and also from regulatory sites. The extension of H1 depletion at promoters is dependent on the transcriptional status of the gene and differs between variants. Analyses show that histone H1 is not uniformly distributed along the genome and differences among variants exist, being H1.2 the variant showing a more specific pattern and a strongest correlation with gene repression in breast cancer cells. Results suggest that different variants may be present at different chromatin types, and this may depend on the cell type, differentiation state, and whether cells are originated from a neoplastic process. In a second part of the thesis, it is shown that a previously reported H1.4 knock-down cell line presents and off-target effect against lamin B2. Therefore, it has been developed a new inducible knock-down cell line specifically inhibiting H1.4, which resembles previously characterized H1.2 knock-down. Finally, combined depletion of H1.4/lamin B2 and H1.2/H1.4 causes similar effects in T47D breast cancer cell line.Fins a set variants de la histona H1 s han identificat en mamífers, les quals mostres una prevalença diferent entre tipus cel lulars i durant el procés de diferenciació. Tot i que la histona H1 juga un paper clau en l estructuració de la cromatina, no s acaba d entendre encara com participa exactament en els diferent processos cel lulars. A més a més, encara no està clar si les diferents variants tenen funcions específiques ni si es distribueixen igual al llarg del genoma. Mitjançant anticossos específics per algunes variants d H1 i de línies cel lulars de càncer de mama que expressen H1s recombinant fusionades a un pèptid HA, s ha estudiat la distribució genòmica de H1.2 a H1.5, H1.0 i H1X, combinant ChIP-qPCR, ChIP-chip i ChIP-seq. Totes les H1s es troben a promotors gènics i empobrides a l inici de transcripció dels gens actius, i també a les regions reguladores. El grau de disminució d H1 al promotor depèn de l estat transcripcional del gen i presenta diferències entre variants. Els anàlisis mostren que la histona H1 no es distribueix uniformement al genoma i que hi ha diferències entre variants, essent H1.2 la variant que presenta un patró més específic i una correlació més forta amb repressió gènica a cèl lules de càncer de mama. Aquests resultats suggereixen que variants d H1 diferents es troben presents als diversos tipus de cromatina, i aquest fet podria dependre de la línia cel lular, l estat de diferenciació, o de si les cèl lules s han originat durant un procés neoplàsic. En una segona part de la tesi, es mostra que una línia cel lular anteriorment descrita que inhibeix H1.4 presenta un efecte inespecífic contra lamina B2. Així, s ha desenvolupat una altra línia que inhibeix H1.4 específicament, la qual s assembla a un mutant anteriorment caracteritzat que inhibeix H1.2. Finalment, la inhibició combinada de H1.4/lamina B2 i H1.2/H1.4 provoca efectes fenotípics semblants a cèl lules de càncer de mama T47D

    Progesterone Induction of the 11β-Hydroxysteroid Dehydrogenase Type 2 Promoter in Breast Cancer Cells Involves Coordinated Recruitment of STAT5A and Progesterone Receptor to a Distal Enhancer and Polymerase Tracking▿ †

    Get PDF
    Steroid hormone receptors regulate gene expression, interacting with target DNA sequences but also activating cytoplasmic signaling pathways. Using the human 11β-hydroxysteroid dehydrogenase type 2 (11β-HSD2) gene as a model, we have investigated the contributions of both effects on a human progesterone-responsive promoter in breast cancer cells. Chromatin immunoprecipitation has identified two different mechanisms of hormone-induced progesterone receptor (PR) recruitment to the 11β-HSD2 promoter: (i) direct PR binding to DNA at the proximal promoter, abrogated when PR contains a mutated DNA binding domain (DBD), and (ii) STAT5A (signal transducer and activator of transcription 5A)-mediated recruitment of PR to an upstream distal region, impaired by AG490, a JAK/STAT pathway inhibitor. The JAK/STAT inhibitor, as well as expression of dominant-negative STAT5A, impairs hormone induction of 11β-HSD2. On the other hand, the DBD-mutated PR fully supports 11β-HSD2 expression. These results, along with data from a deletion analysis, indicate that the distal region is crucial for hormone regulation of 11β-HSD2. We show active RNA polymerase II tracking from the distal region upon PR and STAT5A binding, concomitant with synthesis of noncoding, hormone-dependent RNAs, suggesting that this region works as a hormone-dependent transcriptional enhancer. In conclusion, coordination of PR transcriptional effects and cytoplasmic signaling activation, in particular the JAK/STAT pathway, are critical in regulating progestin-induced endogenous 11β-HSD2 gene expression in breast cancer cells. This is not unique to this promoter, as AG490 also alters the expression of other progesterone-regulated genes

    Mapping of six somatic linker histone H1 variants in human breast cancer cells uncovers specific features of H1.2

    Get PDF
    Seven linker histone H1 variants are present in human somatic cells with distinct prevalence across cell types. Despite being key structural components of chromatin, it is not known whether the different variants have specific roles in the regulation of nuclear processes or are differentially distributed throughout the genome. Using variant-specific antibodies to H1 and hemagglutinin (HA)-tagged recombinant H1 variants expressed in breast cancer cells, we have investigated the distribution of six H1 variants in promoters and genome-wide. H1 is depleted at promoters depending on its transcriptional status and differs between variants. Notably, H1.2 is less abundant than other variants at the transcription start sites of inactive genes, and promoters enriched in H1.2 are different from those enriched in other variants and tend to be repressed. Additionally, H1.2 is enriched at chromosomal domains characterized by low guanine–cytosine (GC) content and is associated with lamina-associated domains. Meanwhile, other variants are associated with higher GC content, CpG islands and gene-rich domains. For instance, H1.0 and H1X are enriched at gene-rich chromosomes, whereas H1.2 is depleted. In short, histone H1 is not uniformly distributed along the genome and there are differences between variants, H1.2 being the one showing the most specific pattern and strongest correlation with low gene expression.Ministerio de Ciencia e Innovació n of Spain (MICINN) and FEDER [BFU2011-23057 to A.J.]; Ministerio de Economía y Competitividad [SAF2012-36199 to N.L.-B.]; Generalitat de Catalunya [2009-SGR-1222 to A.J.]; JAE-Doc contract from CSIC-MICINN (to J.-M.T.); TA contract from CSIC-MICINN (to R.M.); FPU predoctoral fellowship from MICINN (to L.M.-A.). Funding for open access charge: Ministerio de Ciencia e Innovación of Spain (MICINN) and FEDER [BFU2011-23057 to A.J.

    Mapping of six somatic linker histone H1 variants in human breast cancer cells uncovers specific features of H1.2

    No full text
    Seven linker histone H1 variants are present in human somatic cells with distinct prevalence across cell types. Despite being key structural components of chromatin, it is not known whether the different variants have specific roles in the regulation of nuclear processes or are differentially distributed throughout the genome. Using variant-specific antibodies to H1 and hemagglutinin (HA)-tagged recombinant H1 variants expressed in breast cancer cells, we have investigated the distribution of six H1 variants in promoters and genome-wide. H1 is depleted at promoters depending on its transcriptional status and differs between variants. Notably, H1.2 is less abundant than other variants at the transcription start sites of inactive genes, and promoters enriched in H1.2 are different from those enriched in other variants and tend to be repressed. Additionally, H1.2 is enriched at chromosomal domains characterized by low guanine–cytosine (GC) content and is associated with lamina-associated domains. Meanwhile, other variants are associated with higher GC content, CpG islands and gene-rich domains. For instance, H1.0 and H1X are enriched at gene-rich chromosomes, whereas H1.2 is depleted. In short, histone H1 is not uniformly distributed along the genome and there are differences between variants, H1.2 being the one showing the most specific pattern and strongest correlation with low gene expression.Ministerio de Ciencia e Innovació n of Spain (MICINN) and FEDER [BFU2011-23057 to A.J.]; Ministerio de Economía y Competitividad [SAF2012-36199 to N.L.-B.]; Generalitat de Catalunya [2009-SGR-1222 to A.J.]; JAE-Doc contract from CSIC-MICINN (to J.-M.T.); TA contract from CSIC-MICINN (to R.M.); FPU predoctoral fellowship from MICINN (to L.M.-A.). Funding for open access charge: Ministerio de Ciencia e Innovación of Spain (MICINN) and FEDER [BFU2011-23057 to A.J.

    Histone H1 depletion triggers an interferon response in cancer cells via activation of heterochromatic repeats

    No full text
    Histone H1 has seven variants in human somatic cells and contributes to chromatin compaction and transcriptional regulation. Knock-down (KD) of each H1 variant in breast cancer cells results in altered gene expression and proliferation differently in a variant specific manner with H1.2 and H1.4 KDs being most deleterious. Here we show combined depletion of H1.2 and H1.4 has a strong deleterious effect resulting in a strong interferon (IFN) response, as evidenced by an up-regulation of many IFN-stimulated genes (ISGs) not seen in individual nor in other combinations of H1 variant KDs. Although H1 participates to repress ISG promoters, IFN activation upon H1.2 and H1.4 KD is mainly generated through the activation of the IFN response by cytosolic nucleic acid receptors and IFN synthesis, and without changes in histone modifications at induced ISG promoters. H1.2 and H1.4 co-KD also promotes the appearance of accessibility sites genome wide and, particularly, at satellites and other repeats. The IFN response may be triggered by the expression of noncoding RNA generated from heterochromatic repeats or endogenous retroviruses upon H1 KD. In conclusion, redundant H1-mediated silencing of heterochromatin is important to maintain cell homeostasis and to avoid an unspecific IFN response

    Histone H1 depletion triggers an interferon response in cancer cells via activation of heterochromatic repeats

    No full text
    Histone H1 has seven variants in human somatic cells and contributes to chromatin compaction and transcriptional regulation. Knock-down (KD) of each H1 variant in breast cancer cells results in altered gene expression and proliferation differently in a variant specific manner with H1.2 and H1.4 KDs being most deleterious. Here we show combined depletion of H1.2 and H1.4 has a strong deleterious effect resulting in a strong interferon (IFN) response, as evidenced by an up-regulation of many IFN-stimulated genes (ISGs) not seen in individual nor in other combinations of H1 variant KDs. Although H1 participates to repress ISG promoters, IFN activation upon H1.2 and H1.4 KD is mainly generated through the activation of the IFN response by cytosolic nucleic acid receptors and IFN synthesis, and without changes in histone modifications at induced ISG promoters. H1.2 and H1.4 co-KD also promotes the appearance of accessibility sites genome wide and, particularly, at satellites and other repeats. The IFN response may be triggered by the expression of noncoding RNA generated from heterochromatic repeats or endogenous retroviruses upon H1 KD. In conclusion, redundant H1-mediated silencing of heterochromatin is important to maintain cell homeostasis and to avoid an unspecific IFN response

    Genome distribution of replication-independent histone H1 variants shows H1.0 associated with nucleolar domains and H1X associated with RNA polymerase II-enriched regions

    No full text
    © 2015 by The American Society for Biochemistry and Molecular Biology, Inc. Unlike core histones, the linker histone H1 family is more evolutionarily diverse, and many organisms have multiple H1 variants or subtypes. In mammals, the H1 family includes seven somatic H1 variants; H1.1 to H1.5 are expressed in a replication-dependent manner, whereas H1.0 and H1X are replication-independent. Using ChIP-sequencing data and cell fractionation, we have compared the genomic distribution of H1.0 and H1X in human breast cancer cells, in which we previously observed differential distribution of H1.2 compared with the other subtypes. We have found H1.0 to be enriched at nucleolus-associated DNA repeats and chromatin domains, whereas H1X is associated with coding regions, RNA polymerase II-enriched regions, and hypomethylated CpG islands. Further, H1X accumulates within constitutive or included exons and retained introns and toward the 3′ end of expressed genes. Inducible H1X knockdown does not affect cell proliferation but dysregulates a subset of genes related to cell movement and transport. In H1X-depleted cells, the promoters of up-regulated genes are not occupied specifically by this variant, have a lower than average H1 content, and, unexpectedly, do not form an H1 valley upon induction. We conclude that H1 variants are not distributed evenly across the genome and may participate with some specificity in chromatin domain organization or gene regulation.This work was supported by funding from the Spanish Ministry of Science and Innovation (MICINN), European Regional Development Fund Grant BFU2011-23057, and Generalitat de Catalunya Grant 2009-SGR-1222Peer Reviewe
    corecore