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










Base de datos
Intervalo de año de publicación
1.
Science ; 382(6671): 725-731, 2023 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-37943933

RESUMEN

The global replacement of histones with protamines in sperm chromatin is widespread in animals, including insects, but its actual function remains enigmatic. We show that in the Drosophila paternal effect mutant paternal loss (pal), sperm chromatin retains germline histones H3 and H4 genome wide without impairing sperm viability. However, after fertilization, pal sperm chromosomes are targeted by the egg chromosomal passenger complex and engage into a catastrophic premature division in synchrony with female meiosis II. We show that pal encodes a rapidly evolving transition protein specifically required for the eviction of (H3-H4)2 tetramers from spermatid DNA after the removal of H2A-H2B dimers. Our study thus reveals an unsuspected role of histone eviction from insect sperm chromatin: safeguarding the integrity of the male pronucleus during female meiosis.


Asunto(s)
Amidina-Liasas , Cromatina , Proteínas de Drosophila , Drosophila melanogaster , Fertilización , Histonas , Herencia Paterna , Espermatozoides , Animales , Femenino , Masculino , Cromatina/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Histonas/metabolismo , Espermatozoides/metabolismo , Amidina-Liasas/genética , Amidina-Liasas/metabolismo , Empaquetamiento del ADN
2.
Nat Commun ; 14(1): 4187, 2023 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-37443316

RESUMEN

Spermiogenesis is a radical process of differentiation whereby sperm cells acquire a compact and specialized morphology to cope with the constraints of sexual reproduction while preserving their main cargo, an intact copy of the paternal genome. In animals, this often involves the replacement of most histones by sperm-specific nuclear basic proteins (SNBPs). Yet, how the SNBP-structured genome achieves compaction and accommodates shaping remain largely unknown. Here, we exploit confocal, electron and super-resolution microscopy, coupled with polymer modeling to identify the higher-order architecture of sperm chromatin in the needle-shaped nucleus of the emerging model cricket Gryllus bimaculatus. Accompanying spermatid differentiation, the SNBP-based genome is strikingly reorganized as ~25nm-thick fibers orderly coiled along the elongated nucleus axis. This chromatin spool is further found to achieve large-scale helical twisting in the final stages of spermiogenesis, favoring its ultracompaction. We reveal that these dramatic transitions may be recapitulated by a surprisingly simple biophysical principle based on a nucleated rigidification of chromatin linked to the histone-to-SNBP transition within a confined nuclear space. Our work highlights a unique, liquid crystal-like mode of higher-order genome organization in ultracompact cricket sperm, and establishes a multidisciplinary methodological framework to explore the diversity of non-canonical modes of DNA organization.


Asunto(s)
Gryllidae , Animales , Masculino , Gryllidae/genética , Semen/metabolismo , Cromatina/genética , Cromatina/metabolismo , Espermatogénesis/genética , Histonas/metabolismo , Espermatozoides/metabolismo
3.
PLoS Pathog ; 19(3): e1011211, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36928089

RESUMEN

Wolbachia are common bacteria among terrestrial arthropods. These endosymbionts transmitted through the female germline manipulate their host reproduction through several mechanisms whose most prevalent form called Cytoplasmic Incompatibility -CI- is a conditional sterility syndrome eventually favoring the infected progeny. Upon fertilization, the sperm derived from an infected male is only compatible with an egg harboring a compatible Wolbachia strain, this sperm leading otherwise to embryonic death. The Wolbachia Cif factors CidA and CidB responsible for CI and its neutralization function as a Toxin-Antitoxin system in the mosquito host Culex pipiens. However, the mechanism of CidB toxicity and its neutralization by the CidA antitoxin remain unexplored. Using transfected insect cell lines to perform a structure-function analysis of these effectors, we show that both CidA and CidB are chromatin interactors and CidA anchors CidB to the chromatin in a cell-cycle dependent-manner. In absence of CidA, the CidB toxin localizes to its own chromatin microenvironment and acts by preventing S-phase completion, independently of its deubiquitylase -DUB- domain. Experiments with transgenic Drosophila show that CidB DUB domain is required together with CidA during spermatogenesis to stabilize the CidA-CidB complex. Our study defines CidB functional regions and paves the way to elucidate the mechanism of its toxicity.


Asunto(s)
Proteínas de Drosophila , Wolbachia , Animales , Masculino , Cromatina/metabolismo , Wolbachia/fisiología , Semen/metabolismo , Animales Modificados Genéticamente , Drosophila/metabolismo , Citoplasma/metabolismo , Proteína A Centromérica/metabolismo , Proteínas de Drosophila/metabolismo
4.
Curr Biol ; 32(6): 1319-1331.e5, 2022 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-35134330

RESUMEN

Wolbachia are widespread endosymbiotic bacteria that manipulate the reproduction of arthropods through a diversity of cellular mechanisms. In cytoplasmic incompatibility (CI), a sterility syndrome originally discovered in the mosquito Culex pipiens, uninfected eggs fertilized by sperm from infected males are selectively killed during embryo development following the abortive segregation of paternal chromosomes in the zygote. Despite the recent discovery of Wolbachia CI factor (cif) genes, the mechanism by which they control the fate of paternal chromosomes at fertilization remains unknown. Here, we have analyzed the cytological distribution and cellular impact of CidA and CidB, a pair of Cif proteins from the Culex-infecting Wolbachia strain wPip. We show that expression of CidB in Drosophila S2R+ cells induces apoptosis unless CidA is co-expressed and associated with its partner. In transgenic Drosophila testes, both effectors colocalize in germ cells until the histone-to-protamine transition in which only CidB is retained in maturing spermatid nuclei. We further show that CidB is similarly targeted to maturing sperm of naturally infected Culex mosquitoes. At fertilization, CidB associates with paternal DNA regions exhibiting DNA replication stress, as a likely cause of incomplete replication of paternal chromosomes at the onset of the first mitosis. Importantly, we demonstrate that inactivation of the deubiquitylase activity of CidB does not abolish its cell toxicity or its ability to induce CI in Drosophila. Our study thus demonstrates that CI functions as a transgenerational toxin-antidote system and suggests that CidB acts by poisoning paternal DNA replication in incompatible crosses.


Asunto(s)
Culex , Wolbachia , Animales , Culex/genética , Citoplasma , Citosol , Drosophila , Masculino , Wolbachia/genética
5.
PLoS Genet ; 18(1): e1009615, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34982772

RESUMEN

The formation of a diploid zygote is a highly complex cellular process that is entirely controlled by maternal gene products stored in the egg cytoplasm. This highly specialized transcriptional program is tightly controlled at the chromatin level in the female germline. As an extreme case in point, the massive and specific ovarian expression of the essential thioredoxin Deadhead (DHD) is critically regulated in Drosophila by the histone demethylase Lid and its partner, the histone deacetylase complex Sin3A/Rpd3, via yet unknown mechanisms. Here, we identified Snr1 and Mod(mdg4) as essential for dhd expression and investigated how these epigenomic effectors act with Lid and Sin3A to hyperactivate dhd. Using Cut&Run chromatin profiling with a dedicated data analysis procedure, we found that dhd is intriguingly embedded in an H3K27me3/H3K9me3-enriched mini-domain flanked by DNA regulatory elements, including a dhd promoter-proximal element essential for its expression. Surprisingly, Lid, Sin3a, Snr1 and Mod(mdg4) impact H3K27me3 and this regulatory element in distinct manners. However, we show that these effectors activate dhd independently of H3K27me3/H3K9me3, and that dhd remains silent in the absence of these marks. Together, our study demonstrates an atypical and critical role for chromatin regulators Lid, Sin3A, Snr1 and Mod(mdg4) to trigger tissue-specific hyperactivation within a unique heterochromatin mini-domain.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/fisiología , Heterocromatina/genética , Histona Demetilasas/metabolismo , Proteínas de la Membrana/genética , Proteínas de Unión al ARN/metabolismo , Complejo Correpresor Histona Desacetilasa y Sin3/metabolismo , Tiorredoxinas/genética , Factores de Transcripción/metabolismo , Animales , Epigenómica , Femenino , Regulación de la Expresión Génica , Heterocromatina/química , Histonas/metabolismo , Masculino , Herencia Materna , Especificidad de Órganos , Ovario/química , Regiones Promotoras Genéticas , Elementos Reguladores de la Transcripción
6.
PLoS Genet ; 16(3): e1008543, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32134927

RESUMEN

Following fertilization of a mature oocyte, the formation of a diploid zygote involves a series of coordinated cellular events that ends with the first embryonic mitosis. In animals, this complex developmental transition is almost entirely controlled by maternal gene products. How such a crucial transcriptional program is established during oogenesis remains poorly understood. Here, we have performed an shRNA-based genetic screen in Drosophila to identify genes required to form a diploid zygote. We found that the Lid/KDM5 histone demethylase and its partner, the Sin3A-HDAC1 deacetylase complex, are necessary for sperm nuclear decompaction and karyogamy. Surprisingly, transcriptomic analyses revealed that these histone modifiers are required for the massive transcriptional activation of deadhead (dhd), which encodes a maternal thioredoxin involved in sperm chromatin remodeling. Unexpectedly, while lid knock-down tends to slightly favor the accumulation of its target, H3K4me3, on the genome, this mark was lost at the dhd locus. We propose that Lid/KDM5 and Sin3A cooperate to establish a local chromatin environment facilitating the unusually high expression of dhd, a key effector of the oocyte-to-zygote transition.


Asunto(s)
Proteínas de Drosophila/genética , Histona Demetilasas/genética , Oocitos/fisiología , Cigoto/fisiología , Animales , Núcleo Celular/genética , Cromatina/genética , Ensamble y Desensamble de Cromatina/genética , Drosophila/genética , Femenino , Regulación del Desarrollo de la Expresión Génica/genética , Histonas/genética , Masculino , Oogénesis/genética , Espermatozoides/fisiología , Transcripción Genética/genética
7.
Epigenetics Chromatin ; 11(1): 19, 2018 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-29751847

RESUMEN

BACKGROUND: Anti-Silencing Factor 1 (ASF1) is a conserved H3-H4 histone chaperone involved in both Replication-Coupled and Replication-Independent (RI) nucleosome assembly pathways. At DNA replication forks, ASF1 plays an important role in regulating the supply of H3.1/2 and H4 to the CAF-1 chromatin assembly complex. ASF1 also provides H3.3-H4 dimers to HIRA and DAXX chaperones for RI nucleosome assembly. The early Drosophila embryo is an attractive system to study chromatin assembly in a developmental context. The formation of a diploid zygote begins with the unique, genome-wide RI assembly of paternal chromatin following sperm protamine eviction. Then, within the same cytoplasm, syncytial embryonic nuclei undergo a series of rapid, synchronous S and M phases to form the blastoderm embryo. Here, we have investigated the implication of ASF1 in these two distinct assembly processes. RESULTS: We show that depletion of the maternal pool of ASF1 with a specific shRNA induces a fully penetrant, maternal effect embryo lethal phenotype. Unexpectedly, despite the depletion of ASF1 protein to undetectable levels, we show that asf1 knocked-down (KD) embryos can develop to various stages, thus demonstrating that ASF1 is not absolutely required for the amplification of cleavage nuclei. Remarkably, we found that ASF1 is required for the formation of the male pronucleus, although ASF1 protein does not reside in the decondensing sperm nucleus. In asf1 KD embryos, HIRA localizes to the male nucleus but is only capable of limited and insufficient chromatin assembly. Finally, we show that the conserved HIRA B domain, which is involved in ASF1-HIRA interaction, is dispensable for female fertility. CONCLUSIONS: We conclude that ASF1 is critically required to load H3.3-H4 dimers on the HIRA complex prior to histone deposition on paternal DNA. This separation of tasks could optimize the rapid assembly of paternal chromatin within the gigantic volume of the egg cell. In contrast, ASF1 is surprisingly dispensable for the amplification of cleavage nuclei, although chromatin integrity is likely compromised in KD embryos.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/embriología , Fertilización , Chaperonas de Histonas/metabolismo , Histonas/metabolismo , Factores de Transcripción/metabolismo , Animales , Sitios de Unión , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Núcleo Celular/genética , Núcleo Celular/metabolismo , Ensamble y Desensamble de Cromatina , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Femenino , Chaperonas de Histonas/química , Masculino , Protaminas/metabolismo , Dominios Proteicos , Espermatozoides/metabolismo , Factores de Transcripción/química
9.
Curr Biol ; 27(2): R53-R55, 2017 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-28118585

RESUMEN

Wolbachia are widespread endosymbiotic bacteria found in terrestrial arthropods and filarial nematodes [1]. In insects, Wolbachia generally rely on diverse strategies to manipulate their host's reproduction and favor their own vertical transmission through infected eggs [2]. One such mechanism is a sterility syndrome called 'cytoplasmic incompatibility'. Cytoplasmic incompatibility occurs at fertilization, when a spermatozoon from a Wolbachia-infected male fertilizes an uninfected egg. In this case, sperm-derived chromosomes fail to separate normally at the first zygotic division, thus preventing the development of a diploid embryo [3]. Moreover, the presence of Wolbachia in females rescues the integration of paternal chromosomes in the zygote and allows the development of a viable, infected individual. Although the molecular basis of cytoplasmic incompatibility is still unknown, a current model implies the existence of Wolbachia-induced reversible modifications on sperm DNA or chromatin that must be eliminated or neutralized shortly after fertilization by rescuing Wolbachia factors present in infected eggs [4]. In a recent Current Biology paper [5], Stéphanie Pontier and François Schweisguth recently challenged this model by proposing that Wolbachia perturbs a pheromone-based communication between male and female pupae in Drosophila melanogaster and Drosophila simulans, which controls the "compatibility range" of male and female gametes. However, we fail to detect any influence of pupal communication on cytoplasmic incompatibility in Drosophila simulans as well as in the parasitoid wasp Nasonia vitripennis. Our results thus question the robustness of their model.


Asunto(s)
Wolbachia , Animales , Citoplasma , Drosophila , Drosophila melanogaster , Femenino , Masculino , Pupa
10.
Nat Commun ; 7: 13539, 2016 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-27876811

RESUMEN

In most animals, the extreme compaction of sperm DNA is achieved after the massive replacement of histones with sperm nuclear basic proteins (SNBPs), such as protamines. In some species, the ultracompact sperm chromatin is stabilized by a network of disulfide bonds connecting cysteine residues present in SNBPs. Studies in mammals have established that the reduction of these disulfide crosslinks at fertilization is required for sperm nuclear decondensation and the formation of the male pronucleus. Here, we show that the Drosophila maternal thioredoxin Deadhead (DHD) is specifically required to unlock sperm chromatin at fertilization. In dhd mutant eggs, the sperm nucleus fails to decondense and the replacement of SNBPs with maternally-provided histones is severely delayed, thus preventing the participation of paternal chromosomes in embryo development. We demonstrate that DHD localizes to the sperm nucleus to reduce its disulfide targets and is then rapidly degraded after fertilization.


Asunto(s)
Cromatina/fisiología , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Fertilización/fisiología , Proteínas de la Membrana/metabolismo , Óvulo/fisiología , Espermatozoides/fisiología , Tiorredoxinas/metabolismo , Animales , Proteínas de Drosophila/genética , Drosophila melanogaster/embriología , Femenino , Regulación de la Expresión Génica/fisiología , Masculino , Proteínas de la Membrana/genética , Tiorredoxinas/genética
11.
Aging (Albany NY) ; 7(12): 1066-76, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26638179

RESUMEN

PURPOSE: Age induces a progressive decline in functional reserve and impacts cancer treatments. Telomere attrition leads to tissue senescence. We tested the hypothesis that telomere length (TL) could predict patient vulnerability and outcome with cancer treatment. PATIENTS AND METHODS: An ancillary study in the Elderly Women GINECO Trial 3 was performed to evaluate the impact of geriatric covariates on survival in elderly advanced ovarian cancer patients receiving six cycles of carboplatin. TL was estimated from peripheral blood at inclusion using standard procedures. RESULTS: TL (in base pairs) was estimated for 109/111 patients (median 6.1 kb; range [4.5-8.3 kb]). With a cut-off of 5.77 kb, TL discriminated two patient groups, long telomere (LT) and short telomeres (ST), with significantly different treatment completion rates of 0.80 (95% CI [0.71-0.89]) and 0.59 (95% CI [0.41-0.76]), respectively (odds ratio [OR]=2.8, p=0.02). ST patients were at higher risk of serious adverse events (SAE, OR=2.7; p=0.02) and had more unplanned hospital admissions (OR=2.1; p=0.08). After adjustment on FIGO stage, TL shorter than 6 kb was a risk factor of premature death (HR=1.57; p=0.06). CONCLUSION: This exploratory study identifies TL as predictive factor of decreased treatment completion, SAE risk, unplanned hospital admissions and OS after adjustment on FIGO stage.


Asunto(s)
Envejecimiento , Antineoplásicos/uso terapéutico , Carboplatino/uso terapéutico , Neoplasias Ováricas/metabolismo , Homeostasis del Telómero/fisiología , Anciano , Anciano de 80 o más Años , Biomarcadores , Femenino , Humanos , Estadificación de Neoplasias , Neoplasias Ováricas/tratamiento farmacológico , Embarazo , Factores de Tiempo
12.
Open Biol ; 5(8)2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26246493

RESUMEN

The union of haploid gametes at fertilization initiates the formation of the diploid zygote in sexually reproducing animals. This founding event of embryogenesis includes several fascinating cellular and nuclear processes, such as sperm-egg cellular interactions, sperm chromatin remodelling, centrosome formation or pronuclear migration. In comparison with other aspects of development, the exploration of animal fertilization at the functional level has remained so far relatively limited, even in classical model organisms. Here, we have reviewed our current knowledge of fertilization in Drosophila melanogaster, with a special emphasis on the genes involved in the complex transformation of the fertilizing sperm nucleus into a replicated set of paternal chromosomes.


Asunto(s)
Drosophila/genética , Fertilización/fisiología , Animales , División Celular , Núcleo Celular , Femenino , Masculino , Mutación , Interacciones Espermatozoide-Óvulo/fisiología , Espermatozoides/fisiología , Cigoto/metabolismo
13.
Chromosoma ; 124(2): 163-75, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25563491

RESUMEN

Drosophila development initiates with the formation of a diploid zygote followed by the rapid division of embryonic nuclei. This syncytial phase of development occurs almost entirely under maternal control and ends when the blastoderm embryo cellularizes and activates its zygotic genome. The biosynthesis and storage of histones in quantity sufficient for chromatin assembly of several thousands of genome copies represent a unique challenge for the developing embryo. In this article, we have reviewed our current understanding of the mechanisms involved in the production, storage, and deposition of histones in the fertilized egg and during the exponential amplification of cleavage nuclei.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila/embriología , Drosophila/genética , Histonas/genética , Animales , Blastodermo/embriología , Núcleo Celular/genética , Centrómero/genética , Centrómero/metabolismo , Ensamble y Desensamble de Cromatina/genética , Replicación del ADN , Proteínas de Drosophila/metabolismo , Desarrollo Embrionario/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Genómica , Histonas/biosíntesis , Masculino , Nucleosomas/genética , Nucleosomas/metabolismo , Cigoto/metabolismo
14.
Curr Biol ; 24(19): 2281-7, 2014 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-25242033

RESUMEN

The animal sperm nucleus is characterized by an extremely compacted organization of its DNA after the global replacement of histones with sperm-specific nuclear basic proteins, such as protamines. In the absence of DNA repair activity in the mature gamete, the integrity of the paternal genome is potentially challenged by the unique topological constraints exerted on sperm DNA. In addition, the maintenance of paternal DNA integrity during the rapid remodeling of sperm chromatin at fertilization has long been regarded as a maternal trait. However, little is known about the nature of the egg proteins involved in this essential aspect of zygote formation. We had previously characterized the unique phenotype of the classical Drosophila maternal effect mutant maternal haploid (mh), which specifically affects the integration of paternal chromosomes in the zygote. Here we show that MH is the fly ortholog of the recently identified human DVC1/Spartan protein, a conserved regulator of DNA damage tolerance. Like Spartan, MH protein is involved in the resistance to UV radiation and recruits the p97/TER94 segregase to stalled DNA replication forks in somatic cells. In the zygote, we found that the mh phenotype is consistent with perturbed or incomplete paternal DNA replication. Remarkably, however, the specific accumulation of MH in the male pronucleus before the first S phase suggests that this maternal protein is required to maintain paternal DNA integrity during nuclear decondensation or to set the paternal chromatin landscape in preparation of the first zygotic cycle.


Asunto(s)
Cromosomas/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Animales , Replicación del ADN , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/embriología , Drosophila melanogaster/metabolismo , Embrión no Mamífero/embriología , Embrión no Mamífero/metabolismo , Haploidia , Cigoto/metabolismo
15.
Nat Cell Biol ; 15(7): 818-28, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23792691

RESUMEN

Dysfunctional telomeres suppress tumour progression by activating cell-intrinsic programs that lead to growth arrest. Increased levels of TRF2, a key factor in telomere protection, are observed in various human malignancies and contribute to oncogenesis. We demonstrate here that a high level of TRF2 in tumour cells decreased their ability to recruit and activate natural killer (NK) cells. Conversely, a reduced dose of TRF2 enabled tumour cells to be more easily eliminated by NK cells. Consistent with these results, a progressive upregulation of TRF2 correlated with decreased NK cell density during the early development of human colon cancer. By screening for TRF2-bound genes, we found that HS3ST4--a gene encoding for the heparan sulphate (glucosamine) 3-O-sulphotransferase 4--was regulated by TRF2 and inhibited the recruitment of NK cells in an epistatic relationship with TRF2. Overall, these results reveal a TRF2-dependent pathway that is tumour-cell extrinsic and regulates NK cell immunity.


Asunto(s)
Neoplasias de la Mama/prevención & control , Neoplasias del Colon/prevención & control , Células Asesinas Naturales/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Melanoma Experimental/prevención & control , Sulfotransferasas/metabolismo , Proteína 2 de Unión a Repeticiones Teloméricas/metabolismo , Animales , Apoptosis , Western Blotting , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/metabolismo , Adhesión Celular , Proliferación Celular , Neoplasias del Colon/inmunología , Neoplasias del Colon/metabolismo , Cartilla de ADN/química , Receptor con Dominio Discoidina 1 , Femenino , Citometría de Flujo , Células HeLa , Humanos , Células Asesinas Naturales/metabolismo , Células Asesinas Naturales/patología , Linfocitos Infiltrantes de Tumor/patología , Melanoma Experimental/inmunología , Melanoma Experimental/metabolismo , Ratones , Ratones Desnudos , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sulfotransferasas/genética , Proteína 2 de Unión a Repeticiones Teloméricas/antagonistas & inhibidores , Proteína 2 de Unión a Repeticiones Teloméricas/genética , Células Tumorales Cultivadas
16.
Biochim Biophys Acta ; 1833(8): 1885-93, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23570868

RESUMEN

Telomere length maintenance is critical for organisms' long-term survival and cancer cell proliferation. Telomeres are kept within species-specific length ranges by the interplay between telomerase activity and telomeric chromatin organization. In this paper, we exploited telomerase immortalized human fibroblasts (cen3tel) that gradually underwent neoplastic transformation during culture propagation to study telomere composition and length regulation during the transformation process. Just after telomerase catalytic subunit (hTERT) expression, cen3tel telomeres shortened despite the presence of telomerase activity. At a later stage and concomitantly with transformation, cells started elongating telomeres, which reached a mean length greater than 100kb in about 900 population doublings. Super-telomeres were stable and compatible with cell growth and tumorigenesis. Telomere extension was associated with increasing levels of telomerase activity that were linked to the deregulation of endogenous telomerase RNA (hTERC) and exogenous telomerase reverse transcriptase (hTERT) expression. Notably, the increase in hTERC levels paralleled the increase in telomerase activity, suggesting that this subunit plays a role in regulating enzyme activity. Telomeres ranging in length between 10 and more than 100kb were maintained in an extendible state although TRF1 and TRF2 binding increased with telomere length. Super-telomeres neither influenced subtelomeric region global methylation nor the expression of the subtelomeric gene FRG1, attesting the lack of a clear-cut relationship between telomere length, subtelomeric DNA methylation and expression in human cells. The cellular levels of the telomeric proteins hTERT, TRF1, TRF2 and Hsp90 rose with transformation and were independent of telomere length, pointing to a role of these proteins in tumorigenesis.


Asunto(s)
Transformación Celular Neoplásica/genética , Fibroblastos/fisiología , Homeostasis del Telómero/genética , Telómero/genética , Telómero/metabolismo , Línea Celular Transformada , Transformación Celular Neoplásica/metabolismo , Metilación de ADN , Fibroblastos/metabolismo , Humanos , Proteínas de Microfilamentos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , ARN/genética , Proteínas de Unión al ARN , Telomerasa/genética , Proteína 1 de Unión a Repeticiones Teloméricas/genética , Proteína 1 de Unión a Repeticiones Teloméricas/metabolismo , Proteína 2 de Unión a Repeticiones Teloméricas/genética , Proteína 2 de Unión a Repeticiones Teloméricas/metabolismo
17.
PLoS Genet ; 9(2): e1003285, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23408912

RESUMEN

The differentiation of post-meiotic spermatids in animals is characterized by a unique reorganization of their nuclear architecture and chromatin composition. In many species, the formation of sperm nuclei involves the massive replacement of nucleosomes with protamines, followed by a phase of extreme nuclear compaction. At fertilization, the reconstitution of a nucleosome-based paternal chromatin after the removal of protamines requires the deposition of maternally provided histones before the first round of DNA replication. This process exclusively uses the histone H3 variant H3.3 and constitutes a unique case of genome-wide replication-independent (RI) de novo chromatin assembly. We had previously shown that the histone H3.3 chaperone HIRA plays a central role for paternal chromatin assembly in Drosophila. Although several conserved HIRA-interacting proteins have been identified from yeast to human, their conservation in Drosophila, as well as their actual implication in this highly peculiar RI nucleosome assembly process, is an open question. Here, we show that Yemanuclein (YEM), the Drosophila member of the Hpc2/Ubinuclein family, is essential for histone deposition in the male pronucleus. yem loss of function alleles affect male pronucleus formation in a way remarkably similar to Hira mutants and abolish RI paternal chromatin assembly. In addition, we demonstrate that HIRA and YEM proteins interact and are mutually dependent for their targeting to the decondensing male pronucleus. Finally, we show that the alternative ATRX/XNP-dependent H3.3 deposition pathway is not involved in paternal chromatin assembly, thus underlining the specific implication of the HIRA/YEM complex for this essential step of zygote formation.


Asunto(s)
Proteínas de Ciclo Celular , Proteínas de Unión al ADN , Proteínas de Drosophila , Chaperonas de Histonas , Proteínas Nucleares , Nucleosomas , Factores de Transcripción , Cigoto , Animales , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Núcleo Celular/genética , Núcleo Celular/metabolismo , Cromatina/metabolismo , Cromatina/ultraestructura , Ensamble y Desensamble de Cromatina , Replicación del ADN/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Fertilización/genética , Chaperonas de Histonas/genética , Chaperonas de Histonas/metabolismo , Histonas/genética , Histonas/metabolismo , Masculino , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Nucleosomas/genética , Nucleosomas/metabolismo , Espermatozoides/citología , Espermatozoides/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Cigoto/crecimiento & desarrollo , Cigoto/metabolismo
18.
PLoS One ; 7(9): e44253, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22962605

RESUMEN

Transposable elements (TEs) are a major source of genetic variability in genomes, creating genetic novelty and driving genome evolution. Analysis of sequenced genomes has revealed considerable diversity in TE families, copy number, and localization between different, closely related species. For instance, although the twin species Drosophila melanogaster and D. simulans share the same TE families, they display different amounts of TEs. Furthermore, previous analyses of wild type derived strains of D. simulans have revealed high polymorphism regarding TE copy number within this species. Several factors may influence the diversity and abundance of TEs in a genome, including molecular mechanisms such as epigenetic factors, which could be a source of variation in TE success. In this paper, we present the first analysis of the epigenetic status of four TE families (roo, tirant, 412 and F) in seven wild type strains of D. melanogaster and D. simulans. Our data shows intra- and inter-specific variations in the histone marks that adorn TE copies. Our results demonstrate that the chromatin state of common TEs varies among TE families, between closely related species and also between wild type strains.


Asunto(s)
Elementos Transponibles de ADN , Drosophila/genética , Histonas/genética , Procesamiento Proteico-Postraduccional , Animales , Evolución Biológica , Cromatina/metabolismo , Variaciones en el Número de Copia de ADN , Metilación de ADN , Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Epigénesis Genética , Expresión Génica , Variación Genética , Genoma de los Insectos , Genotipo , Histonas/metabolismo , Especificidad de la Especie
19.
Methods Mol Biol ; 859: 267-91, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22367878

RESUMEN

Hypomethylation of repetitive DNA elements is a common epigenetics event in cancer. Although it is believed that this hypomethylation impacts chromosomal and transcriptional stability of the genome, the extent of repetitive sequences contribution to the development and progression of human cancers remains to be clarified. Repetitive sequences have largely been ignored by genome-wide studies, and thus little is known about the DNA methylation profiles of different repetitive DNA elements types. As a step toward investigating epigenetic landscape of repetitive DNA, we have developed a repeat-specific DNA microarray called RepArray. The RepArray comprises 236 prototypic oligonucleotides that span the main repetitive elements families found in the human genome. Combined to a methylated DNA immunoprecipitation (MeDIP) approach, RepArray allows depicting simultaneously the global trends that affect multiple repeat classes through the analysis of a restricted number of targets. Here, we present the MeDIP-on-RepArray protocol as it was established in our laboratory to delineate DNA methylation changes after chemical or genetic disruption of DNA methyltransferase activity in cells. It might serve as a workflow guideline for screening DNA methylation changes on repetitive elements during development and aging, among tissues and in various types of stress or pathological situations.


Asunto(s)
Metilación de ADN , Elementos Transponibles de ADN/genética , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Análisis de Secuencia de ADN/métodos , ADN/aislamiento & purificación , División del ADN , Colorantes Fluorescentes/química , Genoma Humano , Humanos , Inmunoprecipitación/métodos , Reacción en Cadena de la Polimerasa/métodos , Secuencias Repetitivas de Ácidos Nucleicos/genética , Sonicación , Coloración y Etiquetado/métodos
20.
Blood ; 118(5): 1316-22, 2011 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-21355086

RESUMEN

Cells of B-cell chronic lymphocytic leukemia (B-CLL) are characterized by short telomeres despite a low proliferative index. Because telomere length has been reported to be a valuable prognosis criteria, there is a great interest in a deep understanding of the origin and consequences of telomere dysfunction in this pathology. Cases of chromosome fusion involving extremely short telomeres have been reported at advanced stage. In the present study, we address the question of the existence of early telomere dysfunction during the B-CLL time course. In a series restricted to 23 newly diagnosed Binet stage A CLL patients compared with 12 healthy donors, we found a significant increase in recruitment of DNA-damage factors to telomeres showing telomere dysfunction in the early stage of the disease. Remarkably, the presence of dysfunctional telomeres did not correlate with telomere shortening or chromatin marks deregulation but with a down-regulation of 2 shelterin genes: ACD (coding for TPP1; P = .0464) and TINF2 (coding for TIN2; P = .0177). We propose that telomeric deprotection in the early step of CLL is not merely the consequence of telomere shortening but also of shelterin alteration.


Asunto(s)
Daño del ADN/fisiología , Leucemia Linfocítica Crónica de Células B/genética , Proteínas de Unión a Telómeros/genética , Telómero/patología , Secuencia de Bases , Estudios de Cohortes , Progresión de la Enfermedad , Regulación Leucémica de la Expresión Génica , Humanos , Leucemia Linfocítica Crónica de Células B/patología , Modelos Biológicos , Datos de Secuencia Molecular , ARN Mensajero/análisis , ARN Mensajero/metabolismo , Complejo Shelterina , Telómero/genética , Proteínas de Unión a Telómeros/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...