Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Stem Cells ; 42(4): 385-401, 2024 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-38206366

RESUMO

Pancreatic ductal progenitor cells have been proposed to contribute to adult tissue maintenance and regeneration after injury, but the identity of such ductal cells remains elusive. Here, from adult mice, we identify a near homogenous population of ductal progenitor-like clusters, with an average of 8 cells per cluster. They are a rare subpopulation, about 0.1% of the total pancreatic cells, and can be sorted using a fluorescence-activated cell sorter with the CD133highCD71lowFSCmid-high phenotype. They exhibit properties in self-renewal and tri-lineage differentiation (including endocrine-like cells) in a unique 3-dimensional colony assay system. An in vitro lineage tracing experiment, using a novel HprtDsRed/+ mouse model, demonstrates that a single cell from a cluster clonally gives rise to a colony. Droplet RNAseq analysis demonstrates that these ductal clusters express embryonic multipotent progenitor cell markers Sox9, Pdx1, and Nkx6-1, and genes involved in actin cytoskeleton regulation, inflammation responses, organ development, and cancer. Surprisingly, these ductal clusters resist prolonged trypsin digestion in vitro, preferentially survive in vivo after a severe acinar cell injury and become proliferative within 14 days post-injury. Thus, the ductal clusters are the fundamental units of progenitor-like cells in the adult murine pancreas with implications in diabetes treatment and tumorigenicity.


Assuntos
Células Acinares , Ductos Pancreáticos , Camundongos , Animais , Pâncreas , Células-Tronco , Diferenciação Celular
2.
Nucleic Acids Res ; 50(8): 4500-4514, 2022 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-35451487

RESUMO

Histone H3.3 is an H3 variant which differs from the canonical H3.1/2 at four residues, including a serine residue at position 31 which is evolutionarily conserved. The H3.3 S31 residue is phosphorylated (H3.3 S31Ph) at heterochromatin regions including telomeres and pericentric repeats. However, the role of H3.3 S31Ph in these regions remains unknown. In this study, we find that H3.3 S31Ph regulates heterochromatin accessibility at telomeres during replication through regulation of H3K9/K36 histone demethylase KDM4B. In mouse embryonic stem (ES) cells, substitution of S31 with an alanine residue (H3.3 A31 -phosphorylation null mutant) results in increased KDM4B activity that removes H3K9me3 from telomeres. In contrast, substitution with a glutamic acid (H3.3 E31, mimics S31 phosphorylation) inhibits KDM4B, leading to increased H3K9me3 and DNA damage at telomeres. H3.3 E31 expression also increases damage at other heterochromatin regions including the pericentric heterochromatin and Y chromosome-specific satellite DNA repeats. We propose that H3.3 S31Ph regulation of KDM4B is required to control heterochromatin accessibility of repetitive DNA and preserve chromatin integrity.


Assuntos
Heterocromatina , Histonas , Animais , Camundongos , Histonas/genética , Histonas/metabolismo , Heterocromatina/genética , Histona Desmetilases/metabolismo , Fosforilação , Montagem e Desmontagem da Cromatina
3.
Mol Hum Reprod ; 27(11)2021 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-34590701

RESUMO

PIWI-interacting small RNAs (piRNAs) maintain genome stability in animal germ cells, with a predominant role in silencing transposable elements. Mutations in the piRNA pathway in the mouse uniformly lead to failed spermatogenesis and male sterility. By contrast, mutant females are fertile. In keeping with this paradigm, we previously reported male sterility and female fertility associated with loss of the enzyme HENMT1, which is responsible for stabilising piRNAs through the catalysation of 3'-terminal 2'-O-methylation. However, the Henmt1 mutant females were poor breeders, suggesting they could be subfertile. Therefore, we investigated oogenesis and female fertility in these mice in greater detail. Here, we show that mutant females indeed have a 3- to 4-fold reduction in follicle number and reduced litter sizes. In addition, meiosis-II mutant oocytes display various spindle abnormalities and have a dramatically altered transcriptome which includes a down-regulation of transcripts required for microtubule function. This down-regulation could explain the spindle defects observed with consequent reductions in litter size. We suggest these various effects on oogenesis could be exacerbated by asynapsis, an apparently universal feature of piRNA mutants of both sexes. Our findings reveal that loss of the piRNA pathway in females has significant functional consequences.


Assuntos
Fertilidade , Infertilidade Feminina/enzimologia , Meiose , Metiltransferases/metabolismo , Oócitos/enzimologia , Oogênese , RNA Interferente Pequeno/metabolismo , Animais , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Infertilidade Feminina/genética , Infertilidade Feminina/fisiopatologia , Metiltransferases/genética , Camundongos , RNA Interferente Pequeno/genética , Transcriptoma
4.
Proc Natl Acad Sci U S A ; 115(18): 4737-4742, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29669917

RESUMO

ATRX (alpha thalassemia/mental retardation X-linked) complexes with DAXX to deposit histone variant H3.3 into repetitive heterochromatin. Recent genome sequencing studies in cancers have revealed mutations in ATRX and their association with ALT (alternative lengthening of telomeres) activation. Here we report depletion of ATRX in mouse ES cells leads to selective loss in ribosomal RNA gene (rDNA) copy number. Supporting this, ATRX-mutated human ALT-positive tumors also show a substantially lower rDNA copy than ALT-negative tumors. Further investigation shows that the rDNA copy loss and repeat instability are caused by a disruption in H3.3 deposition and thus a failure in heterochromatin formation at rDNA repeats in the absence of ATRX. We also find that ATRX-depleted cells are reduced in ribosomal RNA transcription output and show increased sensitivity to RNA polymerase I (Pol I) transcription inhibitor CX5461. In addition, human ALT-positive cancer cell lines are also more sensitive to CX5461 treatment. Our study provides insights into the contribution of ATRX loss of function to tumorigenesis through the loss of rDNA stability and suggests the therapeutic potential of targeting Pol I transcription in ALT cancers.


Assuntos
DNA de Neoplasias/metabolismo , DNA Ribossômico/metabolismo , Dosagem de Genes , Mutação , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Proteína Nuclear Ligada ao X/metabolismo , Benzotiazóis/farmacologia , Linhagem Celular Tumoral , DNA de Neoplasias/genética , DNA Ribossômico/genética , Instabilidade Genômica , Humanos , Naftiridinas/farmacologia , Proteínas de Neoplasias/genética , Neoplasias/genética , Neoplasias/patologia , RNA Polimerase I/antagonistas & inibidores , RNA Polimerase I/genética , RNA Polimerase I/metabolismo , Transcrição Gênica/efeitos dos fármacos , Transcrição Gênica/genética , Proteína Nuclear Ligada ao X/genética
5.
PLoS Genet ; 11(2): e1004964, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25675407

RESUMO

Histones package DNA and regulate epigenetic states. For the latter, probably the most important histone is H3. Mammals have three near-identical H3 isoforms: canonical H3.1 and H3.2, and the replication-independent variant H3.3. This variant can accumulate in slowly dividing somatic cells, replacing canonical H3. Some replication-independent histones, through their ability to incorporate outside S-phase, are functionally important in the very slowly dividing mammalian germ line. Much remains to be learned of H3.3 functions in germ cell development. Histone H3.3 presents a unique genetic paradigm in that two conventional intron-containing genes encode the identical protein. Here, we present a comprehensive analysis of the developmental effects of null mutations in each of these genes. H3f3a mutants were viable to adulthood. Females were fertile, while males were subfertile with dysmorphic spermatozoa. H3f3b mutants were growth-deficient, dying at birth. H3f3b heterozygotes were also growth-deficient, with males being sterile because of arrest of round spermatids. This sterility was not accompanied by abnormalities in sex chromosome inactivation in meiosis I. Conditional ablation of H3f3b at the beginning of folliculogenesis resulted in zygote cleavage failure, establishing H3f3b as a maternal-effect gene, and revealing a requirement for H3.3 in the first mitosis. Simultaneous ablation of H3f3a and H3f3b in folliculogenesis resulted in early primary oocyte death, demonstrating a crucial role for H3.3 in oogenesis. These findings reveal a heavy reliance on H3.3 for growth, gametogenesis, and fertilization, identifying developmental processes that are particularly susceptible to H3.3 deficiency. They also reveal partial redundancy in function of H3f3a and H3f3b, with the latter gene being generally the most important.


Assuntos
Sobrevivência Celular/genética , Cromatina/genética , Fertilidade/genética , Histonas/genética , Oogênese , Animais , Replicação do DNA/genética , Feminino , Feto , Masculino , Meiose/genética , Camundongos , Oócitos/crescimento & desenvolvimento , Espermatócitos/crescimento & desenvolvimento , Espermatócitos/patologia , Espermatozoides/crescimento & desenvolvimento , Espermatozoides/patologia , Zigoto
6.
Nucleic Acids Res ; 43(5): 2603-14, 2015 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-25690891

RESUMO

Human ALT cancers show high mutation rates in ATRX and DAXX. Although it is well known that the absence of ATRX/DAXX disrupts H3.3 deposition at heterochromatin, its impact on H3.3 deposition and post-translational modification in the global genome remains unclear. Here, we explore the dynamics of phosphorylated H3.3 serine 31 (H3.3S31ph) in human ALT cancer cells. While H3.3S31ph is found only at pericentric satellite DNA repeats during mitosis in most somatic human cells, a high level of H3.3S31ph is detected on the entire chromosome in ALT cells, attributable to an elevated CHK1 activity in these cells. Drug inhibition of CHK1 activity during mitosis and expression of mutant H3.3S31A in these ALT cells result in a decrease in H3.3S31ph levels accompanied with increased levels of phosphorylated H2AX serine 139 on chromosome arms and at the telomeres. Furthermore, the inhibition of CHK1 activity in these cells also reduces cell viability. Our findings suggest a novel role of CHK1 as an H3.3S31 kinase, and that CHK1-mediated H3.3S31ph plays an important role in the maintenance of chromatin integrity and cell survival in ALT cancer cells.


Assuntos
Montagem e Desmontagem da Cromatina , Cromatina/metabolismo , Histonas/metabolismo , Proteínas Quinases/metabolismo , Western Blotting , Linhagem Celular Transformada , Linhagem Celular Tumoral , Sobrevivência Celular/genética , Quinase 1 do Ponto de Checagem , Cromatina/genética , DNA Helicases/genética , DNA Helicases/metabolismo , Células HT29 , Células HeLa , Histonas/genética , Humanos , Microscopia de Fluorescência , Mutação , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosforilação , Proteínas Quinases/genética , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Serina/genética , Serina/metabolismo , Telômero/genética , Telômero/metabolismo , Proteína Nuclear Ligada ao X
7.
Nucleic Acids Res ; 43(21): 10227-37, 2015 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-26304540

RESUMO

In addition to being a hallmark at active genes, histone variant H3.3 is deposited by ATRX at repressive chromatin regions, including the telomeres. It is unclear how H3.3 promotes heterochromatin assembly. We show that H3.3 is targeted for K9 trimethylation to establish a heterochromatic state enriched in trimethylated H3.3K9 at telomeres. In H3f3a(-/-) and H3f3b(-/-) mouse embryonic stem cells (ESCs), H3.3 deficiency results in reduced levels of H3K9me3, H4K20me3 and ATRX at telomeres. The H3f3b(-/-) cells show increased levels of telomeric damage and sister chromatid exchange (t-SCE) activity when telomeres are compromised by treatment with a G-quadruplex (G4) DNA binding ligand or by ASF1 depletion. Overexpression of wild-type H3.3 (but not a H3.3K9 mutant) in H3f3b(-/-) cells increases H3K9 trimethylation level at telomeres and represses t-SCE activity induced by a G4 ligand. This study demonstrates the importance of H3.3K9 trimethylation in heterochromatin formation at telomeres. It provides insights into H3.3 function in maintaining integrity of mammalian constitutive heterochromatin, adding to its role in mediating transcription memory in the genome.


Assuntos
Heterocromatina/metabolismo , Código das Histonas , Histonas/metabolismo , Lisina/metabolismo , Telômero/metabolismo , Animais , Células Cultivadas , Dano ao DNA , Deleção de Genes , Histonas/química , Histonas/genética , Metilação , Camundongos , Troca de Cromátide Irmã , Transcrição Gênica
8.
Genomics ; 107(2-3): 88-99, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26802803

RESUMO

Natural antisense transcripts (NATs) are involved in cellular development and regulatory processes. Multiple NATs at the Sox4 gene locus are spatiotemporally regulated throughout murine cerebral corticogenesis. In the study, we evaluated the potential functional role of Sox4 NATs at Sox4 gene locus. We demonstrated Sox4 sense and NATs formed dsRNA aggregates in the cytoplasm of brain cells. Over expression of Sox4 NATs in NIH/3T3 cells generally did not alter the level of Sox4 mRNA expression or protein translation. Upregulation of a Sox4 NAT known as Sox4ot1 led to the production of a novel small RNA, Sox4_sir3. Its biogenesis is Dicer1-dependent and has characteristics resemble piRNA. Expression of Sox4_sir3 was observed in the marginal and germinative zones of the developing and postnatal brains suggesting a potential role in regulating neurogenesis. We proposed that Sox4 sense-NATs serve as Dicer1-dependent templates to produce a novel endo-siRNA- or piRNA-like Sox4_sir3.


Assuntos
Encéfalo/crescimento & desenvolvimento , RNA Antissenso/genética , RNA de Cadeia Dupla/metabolismo , RNA Interferente Pequeno/metabolismo , Fatores de Transcrição SOXC/genética , Animais , Encéfalo/metabolismo , Citoplasma/metabolismo , RNA Helicases DEAD-box/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Células NIH 3T3 , Neurogênese , RNA Antissenso/metabolismo , RNA de Cadeia Dupla/genética , Ribonuclease III/metabolismo , Fatores de Transcrição SOXC/metabolismo
9.
Chromosoma ; 123(6): 587-95, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25007861

RESUMO

Histone variants can incorporate into the nucleosome outside of S-phase. Some are known to play important roles in mammalian germ cell development, this cell lineage being characterized by long phases of quiescence, a protracted meiotic phase, and genome-wide epigenetic reformatting events. The best known example of such an event is the global-scale erasure of DNA methylation in sexually indifferent primordial germ cells, then its re-establishment in fetal prospermatogonia and growing oocytes. Histone H3 and its post-translationally modified forms provide important waypoints in the establishment of epigenetic states. Using mass spectrometry and immunoblotting, we show that the H3.3 replacement variant is present at an unusually high amount in mouse prospermatogonia at the peak stage of global DNA methylation re-establishment. We speculate that H3.3 facilitates this process through achieving a greater level of accessibility of chromatin modifiers to DNA.


Assuntos
Metilação de DNA , Epigênese Genética , Histonas/fisiologia , Espermatogônias/metabolismo , Animais , Western Blotting , Montagem e Desmontagem da Cromatina , Feminino , Cromatografia Gasosa-Espectrometria de Massas , Expressão Gênica , Histonas/genética , Histonas/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Nucleossomos/genética , Nucleossomos/metabolismo
10.
Cell Mol Life Sci ; 71(7): 1117-22, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24445814

RESUMO

The field of epigenetics is expanding rapidly, yet there is persistent uncertainty in the definition of the term. The word was coined in the mid-twentieth century as a descriptor of how intrinsic, yet largely unknown, forces act with genes to channel progenitor cells along pathways of differentiation. Near the end of the twentieth century, epigenetics was defined more specifically as the study of changes in gene activity states. In some definitions, only those activity states that are inherited across cell division were considered. Other definitions were broader, also including activity states that are transient, or occurring in non-dividing cells. The greatest point of disagreement in these current definitions, is if the term should concern only inherited activity states. To alleviate this disparity, an alternative term, 'memigenetics', could be used in place of epigenetics to describe inherited chromatin activity states. The advantage of this term is that it is self-defining, and would serve to emphasize the important concept of cell memory. It would also free the term epigenetics to be used in a broader sense in accord with the meaning of the prefix 'epi', that is, as a descriptor of what is 'over' DNA at any point in time.


Assuntos
Epigenômica , Regulação da Expressão Gênica , Cromatina/metabolismo , Cromatina/fisiologia , Epigênese Genética , Genótipo , Fenótipo , Terminologia como Assunto
11.
Nucleic Acids Res ; 41(8): 4447-58, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23444137

RESUMO

We have previously shown that α-thalassemia mental retardation X-linked (ATRX) and histone H3.3 are key regulators of telomeric chromatin in mouse embryonic stem cells. The function of ATRX and H3.3 in the maintenance of telomere chromatin integrity is further demonstrated by recent studies that show the strong association of ATRX/H3.3 mutations with alternative lengthening of telomeres in telomerase-negative human cancer cells. Here, we demonstrate that ATRX and H3.3 co-localize with the telomeric DNA and associated proteins within the promyelocytic leukemia (PML) bodies in mouse ES cells. The assembly of these telomere-associated PML bodies is most prominent at S phase. RNA interference (RNAi)-mediated knockdown of PML expression induces the disassembly of these nuclear bodies and a telomere dysfunction phenotype in mouse ES cells. Loss of function of PML bodies in mouse ES cells also disrupts binding of ATRX/H3.3 and proper establishment of histone methylation pattern at the telomere. Our study demonstrates that PML bodies act as epigenetic regulators by serving as platforms for the assembly of the telomeric chromatin to ensure a faithful inheritance of epigenetic information at the telomere.


Assuntos
Estruturas do Núcleo Celular/metabolismo , Cromatina/metabolismo , Células-Tronco Embrionárias/metabolismo , Telômero/metabolismo , Animais , Linhagem Celular Tumoral , Estruturas do Núcleo Celular/química , DNA Helicases/análise , Reparo do DNA , Epigênese Genética , Histonas/análise , Humanos , Camundongos , Células NIH 3T3 , Proteínas Nucleares/análise , Proteínas Nucleares/fisiologia , Fenótipo , Fase S , Proteína Nuclear Ligada ao X
12.
PLoS Genet ; 8(9): e1002919, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22969435

RESUMO

Reduced DNA methylation has been reported in DICER1-deficient mouse ES cells. Reductions seen at pericentric satellite repeats have suggested that siRNAs are required for the proper assembly of heterochromatin. More recent studies have postulated that the reduced methylation is an indirect effect: the loss of Mir290 cluster miRNAs leads to upregulation of the transcriptional repressor RBL2 that targets the downregulation of DNA methyltransferase (Dnmt) genes. However, the observations have been inconsistent. We surmised that the inconsistency could be related to cell line "age," given that DNA methylation is lost progressively with passage in DNMT-deficient ES cells. We therefore subjected Dicer1(-/-) ES cells to two experimental regimes to rigorously test the level of functional DNMT activity. First, we cultured them for a prolonged period. If DNMT activity was reduced, further losses of methylation would occur. Second, we measured their DNMT activity in a rebound DNA methylation assay: DNA methylation was stripped from Cre/loxP conditionally mutant Dicer1 ES cells using a shRNA targeting Dnmt1 mRNA. Cre expression then converted these cells to Dicer1(-/-), allowing for DNMT1 recovery and forcing the cells to remethylate in the absence of RNAi. In both cases, we found functional DNMT activity to be normal. Finally, we also show that the level of RBL2 protein is not at excess levels in Dicer1(-/-) ES cells as has been assumed. These studies reveal that reduced functional DNMT activity is not a salient feature of DICER1-deficient ES cells. We suggest that the reduced DNA methylation sometimes observed in these cells could be due to stochastic alterations in DNA methylation patterns that could offer growth or survival advantages in culture, or to the dysregulation of pathways acting in opposition to the DNMT pathway.


Assuntos
RNA Helicases DEAD-box/genética , Metilação de DNA , Células-Tronco Embrionárias/metabolismo , Ribonuclease III/genética , Animais , RNA Helicases DEAD-box/metabolismo , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/metabolismo , Camundongos , MicroRNAs/metabolismo , Proteína p130 Retinoblastoma-Like/metabolismo , Ribonuclease III/metabolismo
13.
Genesis ; 51(2): 142-6, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23315948

RESUMO

Post-translational modifications to residues in core histones convey epigenetic information. Their function can be evaluated in amino acid substitution mutants, although to date this method has not been used in mice. To this end, we have evaluated gene targeting vectors designed for Cre recombinase-mediated conditional allelic replacement at the two unlinked genes encoding the histone variant H3.3. The conditional alleles consist of an uninterrupted wild-type H3.3 coding sequence upstream of a desired alternative or proxy coding sequence. The arrangement of two loxP sites allows Cre-mediated replacement of the wild-type coding sequence with the proxy. To demonstrate proof of principle, at each locus we replaced the wild-type coding sequence with a fluorescent reporter. This produced null alleles that will be useful to analyse the effects of H3.3 deficiency in development. Each targeting vector can readily be retrofitted with a proxy coding sequence encoding a modified H3.3 protein. Such vectors will allow for the conditional substitution of specific residues in order to dissect the roles of H3.3 post-translational modifications in development and disease.


Assuntos
Alelos , Histonas/genética , Regiões 3' não Traduzidas , Animais , Regulação da Expressão Gênica no Desenvolvimento , Marcação de Genes , Integrases/genética , Camundongos , Mutação , Processamento de Proteína Pós-Traducional
14.
Genome Res ; 20(3): 351-60, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20110566

RESUMO

ATRX (alpha thalassemia/mental retardation syndrome X-linked) belongs to the SWI2/SNF2 family of chromatin remodeling proteins. Besides the ATPase/helicase domain at its C terminus, it contains a PHD-like zinc finger at the N terminus. Mutations in the ATRX gene are associated with X-linked mental retardation (XLMR) often accompanied by alpha thalassemia (ATRX syndrome). Although ATRX has been postulated to be a transcriptional regulator, its precise roles remain undefined. We demonstrate ATRX localization at the telomeres in interphase mouse embryonic stem (ES) cells in synchrony with the incorporation of H3.3 during telomere replication at S phase. Moreover, we found that chromobox homolog 5 (CBX5) (also known as heterochromatin protein 1 alpha, or HP1 alpha) is also present at the telomeres in ES cells. We show by coimmunoprecipitation that this localization is dependent on the association of ATRX with histone H3.3, and that mutating the K4 residue of H3.3 significantly diminishes ATRX and H3.3 interaction. RNAi-knockdown of ATRX induces a telomere-dysfunction phenotype and significantly reduces CBX5 enrichment at the telomeres. These findings suggest a novel function of ATRX, working in conjunction with H3.3 and CBX5, as a key regulator of ES-cell telomere chromatin.


Assuntos
DNA Helicases/metabolismo , Histonas/metabolismo , Proteínas Nucleares/metabolismo , Células-Tronco Pluripotentes/metabolismo , Telômero/metabolismo , Adenosina Trifosfatases/química , Animais , Cromatina/metabolismo , Cromatina/fisiologia , Montagem e Desmontagem da Cromatina , Homólogo 5 da Proteína Cromobox , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/metabolismo , Cromossomos/metabolismo , DNA Helicases/genética , Replicação do DNA/genética , Replicação do DNA/fisiologia , Células-Tronco Embrionárias/metabolismo , Genes , Histonas/genética , Humanos , Deficiência Intelectual/genética , Interfase/genética , Deficiência Intelectual Ligada ao Cromossomo X/genética , Camundongos , Mutação , Proteínas Nucleares/genética , Proteína Nuclear Ligada ao X , Talassemia alfa/genética
15.
PLoS Genet ; 6(1): e1000803, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20062522

RESUMO

The misexpressed imprinted genes causing developmental failure of mouse parthenogenones are poorly defined. To obtain further insight, we investigated misexpressions that could cause the pronounced growth deficiency and death of fetuses with maternal duplication of distal chromosome (Chr) 7 (MatDup.dist7). Their small size could involve inactivity of Igf2, encoding a growth factor, with some contribution by over-expression of Cdkn1c, encoding a negative growth regulator. Mice lacking Igf2 expression are usually viable, and MatDup.dist7 death has been attributed to the misexpression of Cdkn1c or other imprinted genes. To examine the role of misexpressions determined by two maternal copies of the Igf2/H19 imprinting control region (ICR)-a chromatin insulator, we introduced a mutant ICR (ICR(Delta)) into MatDup.dist7 fetuses. This activated Igf2, with correction of H19 expression and other imprinted transcripts expected. Substantial growth enhancement and full postnatal viability was obtained, demonstrating that the aberrant MatDup.dist7 phenotype is highly dependent on the presence of two unmethylated maternal Igf2/H19 ICRs. Activation of Igf2 is likely the predominant correction that rescued growth and viability. Further experiments involved the introduction of a null allele of Cdkn1c to alleviate its over-expression. Results were not consistent with the possibility that this misexpression alone, or in combination with Igf2 inactivity, mediates MatDup.dist7 death. Rather, a network of misexpressions derived from dist7 is probably involved. Our results are consistent with the idea that reduced expression of IGF2 plays a role in the aetiology of the human imprinting-related growth-deficit disorder, Silver-Russell syndrome.


Assuntos
Cromossomos de Mamíferos/genética , Impressão Genômica , Elementos Isolantes , Fator de Crescimento Insulin-Like II/genética , RNA não Traduzido/genética , Síndrome de Silver-Russell/mortalidade , Animais , Cromossomos de Mamíferos/metabolismo , Inibidor de Quinase Dependente de Ciclina p57/genética , Inibidor de Quinase Dependente de Ciclina p57/metabolismo , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Fator de Crescimento Insulin-Like II/metabolismo , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , RNA Longo não Codificante , RNA não Traduzido/metabolismo , Síndrome de Silver-Russell/embriologia , Síndrome de Silver-Russell/genética , Síndrome de Silver-Russell/metabolismo , Sobrevida
16.
Genome Biol ; 24(1): 284, 2023 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-38066546

RESUMO

BACKGROUND: Point mutations in histone variant H3.3 (H3.3K27M, H3.3G34R) and the H3.3-specific ATRX/DAXX chaperone complex are frequent events in pediatric gliomas. These H3.3 point mutations affect many chromatin modifications but the exact oncogenic mechanisms are currently unclear. Histone H3.3 is known to localize to nuclear compartments known as promyelocytic leukemia (PML) nuclear bodies, which are frequently mutated and confirmed as oncogenic drivers in acute promyelocytic leukemia. RESULTS: We find that the pediatric glioma-associated H3.3 point mutations disrupt the formation of PML nuclear bodies and this prevents differentiation down glial lineages. Similar to leukemias driven by PML mutations, H3.3-mutated glioma cells are sensitive to drugs that target PML bodies. We also find that point mutations in IDH1/2-which are common events in adult gliomas and myeloid leukemias-also disrupt the formation of PML bodies. CONCLUSIONS: We identify PML as a contributor to oncogenesis in a subset of gliomas and show that targeting PML bodies is effective in treating these H3.3-mutated pediatric gliomas.


Assuntos
Neoplasias Encefálicas , Glioma , Histonas , Adulto , Criança , Humanos , Neoplasias Encefálicas/genética , Glioma/genética , Histonas/genética , Mutação , Corpos Nucleares da Leucemia Promielocítica/genética , Corpos Nucleares da Leucemia Promielocítica/patologia
17.
Biochem Cell Biol ; 90(1): 70-7, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22003849

RESUMO

RNAi and Dicer-dependent siRNAs are required for constitutive heterochromatin formation in fission yeast and for establishing DNA methylation at repetitive elements in plants. In the mammalian male germ line, DICER1-independent piRNAs are required for the full establishment of DNA methylation of dispersed repetitive transposable elements. However, in other mammalian cell types, no clear picture has yet emerged of the role of RNAi in establishing heterochromatin and DNA methylation. In mouse embryonic stem cells, which remain viable on loss of DICER1 and ablation of RNAi, while no firm evidence has been obtained for defective heterochromatin formation, there are indications of defective DNA methylation. The latter has been attributed to an indirect effect of reduced DNA methyltransferase (DNMT) activity due to a loss of miRNA-mediated gene regulation. However, it is unclear whether the reductions in DNMT activity were sufficient to affect DNA methylation. We consider it equally likely that the defects in DNA methylation that can be observed in DICER1-deficient embryonic stem cells are the result of nonspecific effects related to RNAi loss aside from reduced DNMT activity.


Assuntos
Metilação de DNA , Mamíferos/genética , Interferência de RNA , Animais , Humanos , Metiltransferases/metabolismo
18.
Sci Adv ; 8(24): eabl8070, 2022 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-35704569

RESUMO

Eggs contain about 200,000 mitochondria that generate adenosine triphosphate and metabolites essential for oocyte development. Mitochondria also integrate metabolism and transcription via metabolites that regulate epigenetic modifiers, but there is no direct evidence linking oocyte mitochondrial function to the maternal epigenome and subsequent embryo development. Here, we have disrupted oocyte mitochondrial function via deletion of the mitochondrial fission factor Drp1. Fission-deficient oocytes exhibit a high frequency of failure in peri- and postimplantation development. This is associated with altered mitochondrial function, changes in the oocyte transcriptome and proteome, altered subcortical maternal complex, and a decrease in oocyte DNA methylation and H3K27me3. Transplanting pronuclei of fertilized Drp1 knockout oocytes to normal ooplasm fails to rescue embryonic lethality. We conclude that mitochondrial function plays a role in establishing the maternal epigenome, with serious consequences for embryo development.


Assuntos
Desenvolvimento Embrionário , Oócitos , Citoplasma/metabolismo , Dinaminas/genética , Dinaminas/metabolismo , Desenvolvimento Embrionário/genética , Feminino , Humanos , Mitocôndrias/metabolismo , Oócitos/metabolismo , Gravidez
19.
BMC Genomics ; 12: 176, 2011 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-21466694

RESUMO

BACKGROUND: MicroRNAs (miRNAs) are small non-coding RNAs that can exert multilevel inhibition/repression at a post-transcriptional or protein synthesis level during disease or development. Characterisation of miRNAs in adult mammalian brains by deep sequencing has been reported previously. However, to date, no small RNA profiling of the developing brain has been undertaken using this method. We have performed deep sequencing and small RNA analysis of a developing (E15.5) mouse brain. RESULTS: We identified the expression of 294 known miRNAs in the E15.5 developing mouse brain, which were mostly represented by let-7 family and other brain-specific miRNAs such as miR-9 and miR-124. We also discovered 4 putative 22-23 nt miRNAs: mm_br_e15_1181, mm_br_e15_279920, mm_br_e15_96719 and mm_br_e15_294354 each with a 70-76 nt predicted pre-miRNA. We validated the 4 putative miRNAs and further characterised one of them, mm_br_e15_1181, throughout embryogenesis. Mm_br_e15_1181 biogenesis was Dicer1-dependent and was expressed in E3.5 blastocysts and E7 whole embryos. Embryo-wide expression patterns were observed at E9.5 and E11.5 followed by a near complete loss of expression by E13.5, with expression restricted to a specialised layer of cells within the developing and early postnatal brain. Mm_br_e15_1181 was upregulated during neurodifferentiation of P19 teratocarcinoma cells. This novel miRNA has been identified as miR-3099. CONCLUSIONS: We have generated and analysed the first deep sequencing dataset of small RNA sequences of the developing mouse brain. The analysis revealed a novel miRNA, miR-3099, with potential regulatory effects on early embryogenesis, and involvement in neuronal cell differentiation/function in the brain during late embryonic and early neonatal development.


Assuntos
Encéfalo/metabolismo , MicroRNAs/genética , Animais , Encéfalo/crescimento & desenvolvimento , Embrião de Mamíferos/metabolismo , Desenvolvimento Embrionário/genética , Perfilação da Expressão Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Camundongos , MicroRNAs/metabolismo
20.
Cell Rep ; 34(6): 108729, 2021 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-33567274

RESUMO

Beckwith-Wiedemann syndrome (BWS) and Silver-Russell syndrome (SRS) are imprinting disorders manifesting as aberrant fetal growth and severe postnatal-growth-related complications. Based on the insulator model, one-third of BWS cases and two-thirds of SRS cases are consistent with misexpression of insulin-like growth factor 2 (IGF2), an important facilitator of fetal growth. We propose that the IGF2-dependent BWS and SRS cases can be identified by prenatal diagnosis and can be prevented by prenatal intervention targeting IGF2. We test this hypothesis using our mouse models of IGF2-dependent BWS and SRS. We find that genetically normalizing IGF2 levels in a double rescue experiment corrects the fetal overgrowth phenotype in the BWS model and the growth retardation in the SRS model. In addition, we pharmacologically rescue the BWS growth phenotype by reducing IGF2 signaling during late gestation. This animal study encourages clinical investigations to target IGF2 for prenatal diagnosis and prenatal prevention in human BWS and SRS.


Assuntos
Síndrome de Beckwith-Wiedemann , Marcação de Genes , Fator de Crescimento Insulin-Like II , Diagnóstico Pré-Natal , Síndrome de Silver-Russell , Animais , Síndrome de Beckwith-Wiedemann/diagnóstico , Síndrome de Beckwith-Wiedemann/embriologia , Síndrome de Beckwith-Wiedemann/genética , Síndrome de Beckwith-Wiedemann/terapia , Modelos Animais de Doenças , Feminino , Humanos , Fator de Crescimento Insulin-Like II/genética , Fator de Crescimento Insulin-Like II/metabolismo , Camundongos , Gravidez , Síndrome de Silver-Russell/diagnóstico , Síndrome de Silver-Russell/embriologia , Síndrome de Silver-Russell/genética , Síndrome de Silver-Russell/terapia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA