Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
1.
Development ; 149(22)2022 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-36383700

RESUMEN

'Age reprogramming' refers to the process by which the molecular and cellular pathways of a cell that are subject to age-related decline are rejuvenated without passage through an embryonic stage. This process differs from the rejuvenation observed in differentiated derivatives of induced pluripotent stem cells, which involves passage through an embryonic stage and loss of cellular identity. Accordingly, the study of age reprogramming can provide an understanding of how ageing can be reversed while retaining cellular identity and the specialised function(s) of a cell, which will be of benefit to regenerative medicine. Here, we highlight recent work that has provided a more nuanced understanding of age reprogramming and point to some open questions in the field that might be explored in the future.


Asunto(s)
Células Madre Pluripotentes Inducidas , Rejuvenecimiento , Reprogramación Celular/genética , Epigénesis Genética , Medicina Regenerativa
2.
Chromosoma ; 132(4): 317-328, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37700063

RESUMEN

Using a new method for bulk preparation of early stage embryos, we have investigated the role played by putative Planococcus citri H3K9 and H4K20 histone methyl transferases (HMTases) in regulating heterochromatinization of the imprinted paternal chromosomal set in male embryos. We found that H3K9 and H420 HMTases are required for heterochromatinization of the paternal chromosomes. We present evidence that both HMTases maintain the paternal "imprint" during the cleavage divisions when both parental chromosome sets are euchromatic. A testable model that accommodates our findings is proposed.


Asunto(s)
Heterocromatina , Metiltransferasas , Masculino , Humanos , Metiltransferasas/genética , Heterocromatina/genética , Histonas/genética
3.
Chromosoma ; 128(2): 69-80, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30719566

RESUMEN

Chromosomal imprinting requires an epigenetic system that "imprints" one of the two parental chromosomes such that it results in a heritable (cell-to-cell) change in behavior of the "imprinted" chromosome. Imprinting takes place when the parental genomes are separate, which occurs during gamete formation in the respective germ-lines and post-fertilization during the period when the parental pro-nuclei lie separately within the ooplasm of the zygote. In the mouse, chromosomal imprinting is regulated by germ-line specific DNA methylation. But the methylation machinery in the respective germ-lines does not discriminate between imprinted and non-imprinted regions. As a consequence, the mouse oocyte nucleus contains over a thousand oocyte-specific germ-line differentially methylated regions (gDMRs). Upon fertilization, the sperm provides a few hundred sperm-specific gDMRs of its own. Combined, there are around 1600 imprinted and non-imprinted gDMRs in the pro-nuclei of the newly fertilized zygote. It is a remarkable fact that beginning in the maternal ooplasm, there are mechanisms that manage to preserve DNA methylation at ~ 26 known imprinted gDMRs in the face of the ongoing genome-wide DNA de-methylation that characterizes pre-implantation development. Specificity is achieved through the binding of KRAB-zinc finger proteins to their cognate recognition sequences within the gDMRs of imprinted genes. This in turn nucleates the assembly of localized heterochromatin-like complexes that preserve methylation at imprinted gDMRs through recruitment of the maintenance methyl transferase Dnmt1. These studies have shown that a germ-line imprint may cause parent-of-origin-specific behavior only if "licensed" by mechanisms that operate post-fertilization. Study of the germ-line and post-fertilization contributions to the imprinting of chromosomes in classical insect systems (Coccidae and Sciaridae) show that the ooplasm is the likely site where imprinting takes place. By comparing molecular and genetic studies across these three species, we suggest that mechanisms which operate post-fertilization play a key role in chromosomal imprinting phenomena in animals and conserved components of heterochromatin are shared by these mechanisms.


Asunto(s)
Impresión Genómica , Herencia Materna , Cromosoma X/genética , Animales , Metilación de ADN , Epigénesis Genética , Células Germinativas/metabolismo , Ratones , Cromosoma X/metabolismo
4.
Exp Cell Res ; 338(1): 70-81, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-26364738

RESUMEN

Diverting a protein from its intracellular location is a unique property of intrabodies. To interfere with the intracellular traffic of heterochromatin protein 1ß (HP1ß) in living cells, we have generated a cytoplasmic targeted anti-HP1ß intrabody, specifically directed against the C-terminal portion of the molecule. HP1ß is a conserved component of mouse and human constitutive heterochromatin involved in diverse nuclear functions including gene silencing, DNA repair and nuclear membrane assembly. We found that the anti-HP1ß intrabody sequesters HP1ß into cytoplasmic aggregates, inhibiting its traffic to the nucleus. Lamin B receptor (LBR) and a subset of core histones (H3/H4) are also specifically co-sequestered in the cytoplasm of anti-HP1ß intrabody-expressing cells. Methylated histone H3 at K9 (Me9H3), a marker of constitutive heterochromatin, is not affected by the anti-HP1ß intrabody expression. Hyper-acetylating conditions completely dislodge H3 from HP1ß:LBR containing aggregates. The expression of anti-HP1ß scFv fragments induces apoptosis, associated with an alteration of nuclear morphology. Both these phenotypes are specifically rescued either by overexpression of recombinant full length HP1ß or by HP1ß mutant containing the chromoshadow domain, but not by recombinant LBR protein. The HP1ß-chromodomain mutant, on the other hand, does not rescue the phenotypes, but does compete with LBR for binding to HP1ß. These findings provide new insights into the mode of action of cytoplasmic-targeted intrabodies and the interaction between HP1ß and its binding partners involved in peripheral heterochromatin organisation.


Asunto(s)
Proteínas Cromosómicas no Histona/metabolismo , Citoplasma/metabolismo , Histonas/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Anticuerpos de Cadena Única/metabolismo , Secuencia de Aminoácidos , Animales , Forma del Núcleo Celular , Homólogo de la Proteína Chromobox 5 , Proteínas Cromosómicas no Histona/química , Células HeLa , Humanos , Ratones , Datos de Secuencia Molecular , Células 3T3 NIH , Agregado de Proteínas , Dominios y Motivos de Interacción de Proteínas , Transporte de Proteínas , Receptores Citoplasmáticos y Nucleares/química , Receptor de Lamina B
5.
Trends Biochem Sci ; 35(2): 115-23, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19836960

RESUMEN

A large body of work in various organisms has shown that the presence of HP1 structural proteins and methylated lysine 9 of histone H3 (H3K9me) represent the characteristic hallmarks of heterochromatin. We propose that a more critical assessment of the physiological importance of the H3K9me-HP1 interaction is warranted in light of recent studies on the mammalian HP1 beta protein. Based on this new research, we conclude that the essential function of HP1 beta (and perhaps that of its orthologues in other species) lies outside the canonical heterochromatic H3K9me-HP1 interaction. We suggest instead that binding of a small fraction of HP1 beta to the H3 histone fold performs a critical role in heterochromatin function and organismal survival.


Asunto(s)
Proteínas Cromosómicas no Histona/metabolismo , Animales , Homólogo de la Proteína Chromobox 5 , Proteínas Cromosómicas no Histona/química , Histonas/química , Histonas/metabolismo , Humanos
6.
J Biol Chem ; 288(35): 25285-25296, 2013 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-23836914

RESUMEN

Pericentric regions form epigenetically organized silent heterochromatin structures that accumulate histone H3 lysine 9 trimethylation (H3K9me3) and HP1. At pericentric regions, Suv39h is the major enzyme that generates H3K9me3. Suv39h also interacts directly with HP1, a methylated H3K9-binding protein. However, it is not well characterized how HP1 interaction is important for Suv39h accumulation and Suv39h-mediated H3K9me3 formation at the pericentromere. To address this, we introduced the HP1 binding-defective N-terminally truncated mouse Suv39h1 (ΔN) into Suv39h-deficient embryonic stem cells. Interestingly, pericentric accumulation of ΔN and ΔN-mediated H3K9me3 was observed to recover, but HP1 accumulation was only marginally restored. ΔN also rescued DNA methyltransferase Dnmt3a and -3b accumulation and DNA methylation of the pericentromere. In contrast, other pericentric heterochromatin features, such as ATRX protein association and H4K20me3, were not recovered. Finally, derepressed major satellite repeats were partially silenced by ΔN expression. These findings clearly showed that the Suv39h-HP1 binding is dispensable for pericentric H3K9me3 and DNA methylation, but this interaction and HP1 recruitment/accumulation seem to be crucial for complete formation of heterochromatin.


Asunto(s)
Proteínas Cromosómicas no Histona/metabolismo , Metilación de ADN/fisiología , Heterocromatina/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Metiltransferasas/metabolismo , Proteínas Represoras/metabolismo , Animales , Homólogo de la Proteína Chromobox 5 , Proteínas Cromosómicas no Histona/genética , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , ADN Helicasas/genética , ADN Helicasas/metabolismo , ADN Metiltransferasa 3A , Células Madre Embrionarias , Células HEK293 , Heterocromatina/genética , N-Metiltransferasa de Histona-Lisina/genética , Histonas/genética , Humanos , Metiltransferasas/genética , Ratones , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Represoras/genética , Proteína Nuclear Ligada al Cromosoma X , ADN Metiltransferasa 3B
8.
Genes Cells ; 17(5): 337-43, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22487104

RESUMEN

Induced pluripotent stem (iPS) cells have provided a rational means of obtaining histo-compatible tissues for 'patient-specific' regenerative therapies (Hanna et al. 2010; Yamanaka & Blau 2010). Despite the obvious potential of iPS cell-based therapies, there are certain problems that must be overcome before these therapies can become safe and routine (Ohi et al. 2011; Pera 2011). As an alternative, we have recently explored the possibility of using 'epigenetic rejuvenation', where the specialized functions of an old cell are rejuvenated in the absence of any change in its differentiated state (Singh & Zacouto 2010). The mechanism(s) that underpin 'epigenetic rejuvenation' are unknown and here we discuss model systems, using key epigenetic modifiers, which might shed light on the processes involved. Epigenetic rejuvenation has advantages over iPS cell techniques that are currently being pursued. First, the genetic and epigenetic abnormalities that arise through the cycle of dedifferentiation of somatic cells to iPS cells followed by redifferentiation of iPS cells into the desired cell type are avoided (Gore et al. 2011; Hussein et al. 2011; Pera 2011): epigenetic rejuvenation does not require passage through the de-/redifferentiation cycle. Second, because the aim of epigenetic rejuvenation is to ensure that the differentiated cell type retains its specialized function it makes redundant the question of transcriptional memory that is inimical to iPS cell-based therapies (Ohi et al. 2011). Third, to produce unrelated cell types using the iPS technology takes a long time, around three weeks, whereas epigenetic rejuvenation of old cells will take only a matter of days. Epigenetic rejuvenation provides the most safe, rapid and cheap route to successful regenerative medicine.


Asunto(s)
Epigenómica , Animales , Diferenciación Celular , Senescencia Celular , Heterocromatina/metabolismo , Humanos , Rejuvenecimiento
9.
Nat Genet ; 34(1): 75-9, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12679815

RESUMEN

Besides serving to package nuclear DNA, histones carry information in the form of a diverse array of post-translational modifications. Methylation of histones H3 and H4 has been implicated in long-term epigenetic 'memory'. Dimethylation or trimethylation of Lys4 of histone H3 (H3 Lys4) has been found in expressible euchromatin of yeasts and mammals. In contrast, methylation of Lys9 of histone H3 (H3 Lys9) has been implicated in establishing and maintaining the largely quiescent heterochromatin of mammals, yeasts, Drosophila melanogaster and plants. We have previously shown that a DNA methylation mutant of Neurospora crassa, dim-5 (defective in methylation), has a nonsense mutation in the SET domain of an H3-specific histone methyltransferase and that substitutions of H3 Lys9 cause gross hypomethylation of DNA. Similarly, the KRYPTONITE histone methyltransferase is required for full DNA methylation in Arabidopsis thaliana. We used biochemical, genetic and immunological methods to investigate the specific mark for DNA methylation in N. crassa. Here we show that trimethylated H3 Lys9, but not dimethylated H3 Lys9, marks chromatin regions for cytosine methylation and that DIM-5 specifically creates this mark.


Asunto(s)
Metilación de ADN , ADN de Hongos/química , ADN de Hongos/metabolismo , Histonas/química , Histonas/metabolismo , Neurospora crassa/metabolismo , ADN de Hongos/genética , Proteínas Fúngicas/química , Proteínas Fúngicas/metabolismo , Genes Fúngicos , Histona Metiltransferasas , N-Metiltransferasa de Histona-Lisina/genética , Lisina/química , Metilación , Metiltransferasas/genética , Mutación , Neurospora crassa/genética , Proteína Metiltransferasas , Procesamiento Proteico-Postraduccional
10.
Front Cell Dev Biol ; 11: 1293122, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38020886

RESUMEN

Pericentric heterochromatin (PCH) plays an essential role in the maintenance of genome integrity and alterations in PCH have been linked to cancer and aging. HP1 α, ß, and γ, are hallmarks of constitutive heterochromatin that are thought to promote PCH structure through binding to heterochromatin-specific histone modifications and interaction with a wide range of factors. Among the less understood components of PCH is the histone H2A variant H2A.Z, whose role in the organization and maintenance of PCH is poorly defined. Here we show that there is a complex interplay between H2A.Z and HP1 isoforms in PCH. While the loss of HP1α results in the accumulation of H2A.Z.1 in PCH, which is associated with a significant decrease in its mobile fraction, H2A.Z.1 binds preferentially to HP1ß in these regions. Of note, H2A.Z.1 downregulation results in increased heterochromatinization and instability of PCH, reflected by accumulation of the major epigenetic hallmarks of heterochromatin in these regions and increased frequency of chromosome aberrations related to centromeric/pericentromeric defects. Our studies support a role for H2A.Z in genome stability and unveil a key role of H2A.Z in the regulation of heterochromatin-specific epigenetic modifications through a complex interplay with the HP1 isoforms.

12.
J Cell Biol ; 174(2): 185-94, 2006 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-16831888

RESUMEN

Mouse chromocenters are clusters of late-replicating pericentric heterochromatin containing HP1 bound to trimethylated lysine 9 of histone H3 (Me3K9H3). Using a cell-free system to initiate replication within G1-phase nuclei, we demonstrate that chromocenters acquire the property of late replication coincident with their reorganization after mitosis and the establishment of a global replication timing program. HP1 dissociated during mitosis but rebound before the establishment of late replication, and removing HP1 from chromocenters by competition with Me3K9H3 peptides did not result in early replication, demonstrating that this interaction is neither necessary nor sufficient for late replication. However, in cells lacking the Suv39h1,2 methyltransferases responsible for K9H3 trimethylation and HP1 binding at chromocenters, replication of chromocenter DNA was advanced by 10-15% of the length of S phase. Reintroduction of Suv39h1 activity restored the later replication time. We conclude that Suv39 activity is required for the fine-tuning of pericentric heterochromatin replication relative to other late-replicating domains, whereas separate factors establish a global replication timing program during early G1 phase.


Asunto(s)
Centrosoma/metabolismo , Momento de Replicación del ADN/fisiología , Heterocromatina/metabolismo , Animales , Células Cultivadas , Homólogo de la Proteína Chromobox 5 , Proteínas Cromosómicas no Histona/metabolismo , ADN Satélite/genética , Fibroblastos/citología , Fase G1 , Histonas/metabolismo , Lisina/metabolismo , Metilación , Metiltransferasas/metabolismo , Ratones , Unión Proteica , Proteínas Represoras/metabolismo , Xenopus
13.
Front Immunol ; 12: 738958, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34721405

RESUMEN

Immune checkpoint blockade (ICB) relieves CD8+ T-cell exhaustion in most mutated tumors, and TCF-1 is implicated in converting progenitor exhausted cells to functional effector cells. However, identifying mechanisms that can prevent functional senescence and potentiate CD8+ T-cell persistence for ICB non-responsive and resistant tumors remains elusive. We demonstrate that targeting Cbx3/HP1γ in CD8+ T cells augments transcription initiation and chromatin remodeling leading to increased transcriptional activity at Lef1 and Il21r. LEF-1 and IL-21R are necessary for Cbx3/HP1γ-deficient CD8+ effector T cells to persist and control ovarian cancer, melanoma, and neuroblastoma in preclinical models. The enhanced persistence of Cbx3/HP1γ-deficient CD8+ T cells facilitates remodeling of the tumor chemokine/receptor landscape ensuring their optimal invasion at the expense of CD4+ Tregs. Thus, CD8+ T cells heightened effector function consequent to Cbx3/HP1γ deficiency may be distinct from functional reactivation by ICB, implicating Cbx3/HP1γ as a viable cancer T-cell-based therapy target for ICB resistant, non-responsive solid tumors.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Homólogo de la Proteína Chromobox 5/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Linfocitos Infiltrantes de Tumor/metabolismo , Factor de Unión 1 al Potenciador Linfoide/metabolismo , Melanoma Experimental/metabolismo , Neuroblastoma/metabolismo , Neoplasias Ováricas/metabolismo , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/trasplante , Diferenciación Celular , Línea Celular Tumoral , Homólogo de la Proteína Chromobox 5/genética , Proteínas Cromosómicas no Histona/genética , Técnicas de Cocultivo , Femenino , Regulación Neoplásica de la Expresión Génica , Inmunoterapia Adoptiva , Subunidad alfa del Receptor de Interleucina-21/genética , Subunidad alfa del Receptor de Interleucina-21/metabolismo , Activación de Linfocitos , Linfocitos Infiltrantes de Tumor/inmunología , Factor de Unión 1 al Potenciador Linfoide/genética , Melanoma Experimental/genética , Melanoma Experimental/inmunología , Melanoma Experimental/terapia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Neuroblastoma/genética , Neuroblastoma/inmunología , Neuroblastoma/terapia , Neoplasias Ováricas/genética , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/terapia , Transducción de Señal , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Carga Tumoral , Microambiente Tumoral
14.
J Biol Chem ; 284(47): 32635-41, 2009 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-19801677

RESUMEN

Mast cells (MCs) play a critical role in innate and adaptive immunity through the release of cytokines, chemokines, lipid mediators, biogenic amines, and proteases. We recently showed that the activities of MC proteases are transcriptionally regulated by intracellularly retained interleukin-15 (IL-15), and we provided evidence that this cytokine acts as a specific regulator of mouse mast cell protease-2 (mMCP-2). Here, we show that in wild-type bone marrow-derived mast cells (BMMCs) IL-15 inhibits mMCP-2 transcription indirectly by inducing differential expression and mMCP-2 promoter binding of the bifunctional transcription factors C/EBPbeta and YY1. In wild-type BMMCs, C/EBPbeta expression predominates over YY1 expression, and thus C/EBPbeta preferentially binds to the mMCP-2 promoter. In IL-15-deficient BMMCs, the opposite is found: YY1 expression predominates and binds to the mMCP-2 promoter at the expense of C/EBPbeta. Hypertranscription of the mMCP-2 gene in IL-15-deficient BMMCs is associated with histone acetylation and, intriguingly, with methylation of non-CpG dinucleotides within the MCP-2 promoter. This suggests a novel model of cytokine-controlled protease transcription: non-CpG methylation maintains a chromosomal domain in an "open" configuration that is permissive for gene expression.


Asunto(s)
Quimasas/biosíntesis , Interleucina-15/fisiología , Transcripción Genética , Animales , Secuencia de Bases , Células de la Médula Ósea/citología , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Células COS , Chlorocebus aethiops , Epigénesis Genética , Histonas/química , Humanos , Mastocitos/citología , Ratones , Modelos Biológicos , Datos de Secuencia Molecular
15.
Chromosoma ; 118(4): 501-12, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19458957

RESUMEN

The establishment of sex-specific epigenetic marks during gametogenesis is one of the key feature of genomic imprinting. By immunocytological analysis, we thoroughly characterized the chromatin remodeling events that take place during gametogenesis in the mealybug Planococcus citri, in which an entire haploid set of (imprinted) chromosomes undergoes facultative heterochromatinization in male embryos. Building on our previous work, we have investigated the interplay of several epigenetic marks in the regulation of this genome-wide phenomenon. We characterized the germline patterns of histone modifications, Me(3)K9H3, Me(2)K9H3, and Me(3)K20H4, and of heterochromatic proteins, PCHET2 (HP1-like) and HP2-like during male and female gametogenesis. We found that at all stages in oogenesis chromatin is devoid of any detectable epigenetic marks. On the other hand, spermatogenesis is accompanied by a complex pattern of redistribution of epigenetic marks from euchromatin to heterochromatin, and vice versa. At the end of spermatogenesis, sperm heads are decorated by all the molecules we tested, except for PCHET2. However, only Me(3)K9H3 and Me(2)K9H3 are still detectable in the male pronucleus. We suggest that the histone H3 lysine 9 methylation is the signal used to establish the male-specific imprinting on the paternal genome, thus allowing it to be distinguished from the maternal genome in the developing embryo.


Asunto(s)
Epigénesis Genética , Impresión Genómica , Hemípteros/genética , Espermatogénesis/genética , Acetilación , Animales , Núcleo Celular/metabolismo , Ensamble y Desensamble de Cromatina , Femenino , Hemípteros/metabolismo , Hemípteros/fisiología , Heterocromatina/genética , Heterocromatina/metabolismo , Histonas/metabolismo , Inmunohistoquímica , Proteínas de Insectos/metabolismo , Lisina/metabolismo , Masculino , Metilación , Microscopía Fluorescente , Proteínas Nucleares/metabolismo , Espermatogénesis/fisiología
16.
Cytokine Growth Factor Rev ; 18(3-4): 223-32, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17509926

RESUMEN

Interleukin-21 is the most recently discovered member of the type-I cytokine family. Structurally, IL-21 shows homology to IL-2, IL-4, and IL-15 proteins. IL-21 shares the common gamma-chain with the other three cytokines but, in addition, binds to a unique IL-21Ralpha chain, and activates the JAK/STAT pathway. IL-21 is mainly produced by activated T-cells but targets a broad range of lymphoid and myeloid cells of the immune system and therefore is able to regulate innate and acquired immune responses. This review intends to give the reader an overview of the recent findings concerning the biology of IL-21 and its physiological role in immunity, infection, and cancer.


Asunto(s)
Regulación de la Expresión Génica , Infecciones/genética , Infecciones/metabolismo , Interleucinas/metabolismo , Interleucinas/fisiología , Neoplasias/genética , Neoplasias/metabolismo , Receptores de Interleucina-21/fisiología , Animales , Enfermedades Autoinmunes/metabolismo , Linfocitos B/metabolismo , Células Dendríticas/metabolismo , Humanos , Sistema Inmunológico , Células Asesinas Naturales/metabolismo , Ratones , Ratones Transgénicos , Receptores de Interleucina-21/metabolismo , Transducción de Señal
17.
R Soc Open Sci ; 7(2): 191976, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32257349

RESUMEN

The relationship between compartmentalization of the genome and epigenetics is long and hoary. In 1928, Heitz defined heterochromatin as the largest differentiated chromatin compartment in eukaryotic nuclei. Müller's discovery of position-effect variegation in 1930 went on to show that heterochromatin is a cytologically visible state of heritable (epigenetic) gene repression. Current insights into compartmentalization have come from a high-throughput top-down approach where contact frequency (Hi-C) maps revealed the presence of compartmental domains that segregate the genome into heterochromatin and euchromatin. It has been argued that the compartmentalization seen in Hi-C maps is owing to the physiochemical process of phase separation. Oddly, the insights provided by these experimental and conceptual advances have remained largely silent on how Hi-C maps and phase separation relate to epigenetics. Addressing this issue directly in mammals, we have made use of a bottom-up approach starting with the hallmarks of constitutive heterochromatin, heterochromatin protein 1 (HP1) and its binding partner the H3K9me2/3 determinant of the histone code. They are key epigenetic regulators in eukaryotes. Both hallmarks are also found outside mammalian constitutive heterochromatin as constituents of larger (0.1-5 Mb) heterochromatin-like domains and smaller (less than 100 kb) complexes. The well-documented ability of HP1 proteins to function as bridges between H3K9me2/3-marked nucleosomes contributes to polymer-polymer phase separation that packages epigenetically heritable chromatin states during interphase. Contacts mediated by HP1 'bridging' are likely to have been detected in Hi-C maps, as evidenced by the B4 heterochromatic subcompartment that emerges from contacts between large KRAB-ZNF heterochromatin-like domains. Further, mutational analyses have revealed a finer, innate, compartmentalization in Hi-C experiments that probably reflect contacts involving smaller domains/complexes. Proteins that bridge (modified) DNA and histones in nucleosomal fibres-where the HP1-H3K9me2/3 interaction represents the most evolutionarily conserved paradigm-could drive and generate the fundamental compartmentalization of the interphase nucleus. This has implications for the mechanism(s) that maintains cellular identity, be it a terminally differentiated fibroblast or a pluripotent embryonic stem cell.

18.
Cells ; 9(8)2020 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-32796726

RESUMEN

The hallmarks of constitutive heterochromatin, HP1 and H3K9me2/3, assemble heterochromatin-like domains/complexes outside canonical constitutively heterochromatic territories where they regulate chromatin template-dependent processes. Domains are more than 100 kb in size; complexes less than 100 kb. They are present in the genomes of organisms ranging from fission yeast to human, with an expansion in size and number in mammals. Some of the likely functions of domains/complexes include silencing of the donor mating type region in fission yeast, preservation of DNA methylation at imprinted germline differentially methylated regions (gDMRs) and regulation of the phylotypic progression during vertebrate development. Far cis- and trans-contacts between micro-phase separated domains/complexes in mammalian nuclei contribute to the emergence of epigenetic compartmental domains (ECDs) detected in Hi-C maps. A thermodynamic description of micro-phase separation of heterochromatin-like domains/complexes may require a gestalt shift away from the monomer as the "unit of incompatibility" that determines the sign and magnitude of the Flory-Huggins parameter, χ. Instead, a more dynamic structure, the oligo-nucleosomal "clutch", consisting of between 2 and 10 nucleosomes is both the long sought-after secondary structure of chromatin and its unit of incompatibility. Based on this assumption we present a simple theoretical framework that enables an estimation of χ for domains/complexes flanked by euchromatin and thereby an indication of their tendency to phase separate. The degree of phase separation is specified by χN, where N is the number of "clutches" in a domain/complex. Our approach could provide an additional tool for understanding the biophysics of the 3D genome.


Asunto(s)
Heterocromatina/metabolismo , Animales , Metilación de ADN/genética , Metilación de ADN/fisiología , Epigénesis Genética/genética , Epigénesis Genética/fisiología , Genoma Humano/genética , Heterocromatina/genética , Humanos
19.
J Biosci ; 44(4)2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31502583

RESUMEN

It has been proposed that age reprogramming enables old cells to be rejuvenated without passage through an embryonic stage (Singh and Zacouto in J. Biosci. 35 315-319, 2010). As such, age reprogramming stands apart from the induced pluripotent stem (iPS) and nuclear transfer-embryonic stem (NT-ES) cell therapies where histo-compatible cells are produced only after passage through an embryonic stage. It avoids many of the disadvantages associated with iPS and NT-ES cell therapies. Experimental evidence in support of age reprogramming is burgeoning. Here, we discuss possible new approaches to enhance age reprogramming, which will have considerable benefits for regenerative therapies.


Asunto(s)
Envejecimiento/genética , Reprogramación Celular/genética , Epigénesis Genética/genética , Células Madre Embrionarias/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Medicina Regenerativa
20.
Mol Biol Cell ; 16(6): 2872-81, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15788566

RESUMEN

Mono-, di-, and trimethylation of specific histone residues adds an additional level of complexity to the range of histone modifications that may contribute to a histone code. However, it has not been clear whether different methylated states reside stably at different chromatin sites or whether they represent dynamic intermediates at the same chromatin sites. Here, we have used recently developed antibodies that are highly specific for mono-, di-, and trimethylated lysine 9 of histone H3 (MeK9H3) to examine the subnuclear localization and replication timing of chromatin containing these epigenetic marks in mammalian cells. Me1K9H3 was largely restricted to early replicating, small punctate domains in the nuclear interior. Me2K9H3 was the predominant MeK9 epitope at the nuclear and nucleolar periphery and colocalized with sites of DNA synthesis primarily in mid-S phase. Me3K9H3 decorated late-replicating pericentric heterochromatin in mouse cells and sites of DAPI-dense intranuclear heterochromatin in human and hamster cells that replicated throughout S phase. Disruption of the Suv39h1,2 or G9a methyltransferases in murine embryonic stem cells resulted in a redistribution of methyl epitopes, but did not alter the overall spatiotemporal replication program. These results demonstrate that mono-, di-, and trimethylated states of K9H3 largely occupy distinct chromosome domains.


Asunto(s)
Núcleo Celular/metabolismo , ADN/biosíntesis , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Lisina/metabolismo , Animales , Anticuerpos Monoclonales/metabolismo , Células CHO , Línea Celular Tumoral , Cricetinae , Cricetulus , Epigénesis Genética , Epítopos , Colorantes Fluorescentes , Heterocromatina/metabolismo , Humanos , Indoles , Metilación , Ratones , Microscopía Confocal , Fase S
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA