Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Stem Cells ; 31(4): 717-28, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23307555

RESUMEN

The pluripotency of mouse embryonic stem cells (mESCs) is controlled by a network of transcription factors, mi-RNAs, and signaling pathways. Here, we present a new regulatory circuit that connects miR-335, Oct4, and the Retinoblastoma pathway to control mESC self-renewal and differentiation. Oct4 drives the expression of Nipp1 and Ccnf that inhibit the activity of the protein phosphatase 1 (PP1) complex to establish hyperphosphorylation of the retinoblastoma protein 1 (pRb) as a hallmark feature of self-renewing mESCs. The Oct4-Nipp1/Ccnf-PP1-pRb axis promoting mESC self-renewal is under control of miR-335 that regulates Oct4 and Rb expression. During mESC differentiation, miR-335 upregulation co-operates with the transcriptional repression of Oct4 to facilitate the collapse of the Oct4-Nipp1/Ccnf-PP1-pRb axis, pRb dephosphorylation, the exit from self-renewal, and the establishment of a pRb-regulated cell cycle program. Our results introduce Oct4-dependent control of the Rb pathway as novel regulatory circuit controlling mESC self-renewal and differentiation.


Asunto(s)
Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , MicroARNs/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Proteína de Retinoblastoma/metabolismo , Animales , Western Blotting , Puntos de Control del Ciclo Celular/genética , Puntos de Control del Ciclo Celular/fisiología , Células Cultivadas , Inmunoprecipitación de Cromatina , Citometría de Flujo , Inmunoprecipitación , Ratones , MicroARNs/genética , Factor 3 de Transcripción de Unión a Octámeros/genética , Unión Proteica , Proteína de Retinoblastoma/genética
2.
EMBO J ; 28(16): 2323-36, 2009 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-19629032

RESUMEN

Protection of chromosome ends from DNA repair and degradation activities is mediated by specialized protein complexes bound to telomere repeats. Recently, it has become apparent that epigenetic regulation of the telomeric chromatin template critically impacts on telomere function and telomere-length homeostasis from yeast to man. Across all species, telomeric repeats as well as the adjacent subtelomeric regions carry features of repressive chromatin. Disruption of this silent chromatin environment results in loss of telomere-length control and increased telomere recombination. In turn, progressive telomere loss reduces chromatin compaction at telomeric and subtelomeric domains. The recent discoveries of telomere chromatin regulation during early mammalian development, as well as during nuclear reprogramming, further highlights a central role of telomere chromatin changes in ontogenesis. In addition, telomeres were recently shown to generate long, non-coding RNAs that remain associated to telomeric chromatin and will provide new insights into the regulation of telomere length and telomere chromatin. In this review, we will discuss the epigenetic regulation of telomeres across species, with special emphasis on mammalian telomeres. We will also discuss the links between epigenetic alterations at mammalian telomeres and telomere-associated diseases.


Asunto(s)
Cromatina/genética , Epigénesis Genética , ARN/genética , Telomerasa/genética , Telómero/genética , Telómero/metabolismo , Animales , Cromatina/metabolismo , Cromatina/patología , Drosophila/genética , Drosophila/metabolismo , Humanos , ARN/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Schizosaccharomyces/genética , Schizosaccharomyces/metabolismo , Telomerasa/metabolismo , Telómero/patología
3.
Biochimie ; 214(Pt A): 141-155, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37619810

RESUMEN

R-loops are atypical, three-stranded nucleic acid structures that contain a stretch of RNA:DNA hybrids and an unpaired, single stranded DNA loop. R-loops are physiological relevant and can act as regulators of gene expression, chromatin structure, DNA damage repair and DNA replication. However, unscheduled and persistent R-loops are mutagenic and can mediate replication-transcription conflicts, leading to DNA damage and genome instability if left unchecked. Detailed transcriptome analysis unveiled that 85% of the human genome, including repetitive regions, hold transcriptional activity. This anticipates that R-loops management plays a central role for the regulation and integrity of genomes. This function is expected to have a particular relevance for repetitive sequences that make up to 75% of the human genome. Here, we review the impact of R-loops on the function and stability of repetitive regions such as centromeres, telomeres, rDNA arrays, transposable elements and triplet repeat expansions and discuss their relevance for associated pathological conditions.


Asunto(s)
ADN , Estructuras R-Loop , Humanos , ADN/química , Secuencias Repetitivas de Ácidos Nucleicos , ARN/química , ADN de Cadena Simple , Replicación del ADN , Inestabilidad Genómica
4.
EMBO Mol Med ; 15(1): e16033, 2023 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-36426578

RESUMEN

The telomeric repeat-binding factor 2 (TRF2) is a telomere-capping protein that plays a key role in the maintenance of telomere structure and function. It is highly expressed in different cancer types, and it contributes to cancer progression. To date, anti-cancer strategies to target TRF2 remain a challenge. Here, we developed a miRNA-based approach to reduce TRF2 expression. By performing a high-throughput luciferase screening of 54 candidate miRNAs, we identified miR-182-3p as a specific and efficient post-transcriptional regulator of TRF2. Ectopic expression of miR-182-3p drastically reduced TRF2 protein levels in a panel of telomerase- or alternative lengthening of telomeres (ALT)-positive cancer cell lines. Moreover, miR-182-3p induced DNA damage at telomeric and pericentromeric sites, eventually leading to strong apoptosis activation. We also observed that treatment with lipid nanoparticles (LNPs) containing miR-182-3p impaired tumor growth in triple-negative breast cancer (TNBC) models, including patient-derived tumor xenografts (PDTXs), without affecting mouse survival or tissue function. Finally, LNPs-miR-182-3p were able to cross the blood-brain barrier and reduce intracranial tumors representing a possible therapeutic option for metastatic brain lesions.


Asunto(s)
MicroARNs , Neoplasias de la Mama Triple Negativas , Animales , Humanos , Ratones , Apoptosis , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , MicroARNs/metabolismo , Telómero/metabolismo , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología
5.
Semin Cell Dev Biol ; 21(2): 186-93, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19815087

RESUMEN

In eukaryotes, terminal chromosome repeats are bound by a specialized nucleoprotein complex that controls telomere length and protects chromosome ends from DNA repair and degradation. In mammals the "shelterin" complex mediates these central functions at telomeres. In the recent years it has become evident that also the heterochromatic structure of mammalian telomeres is implicated in telomere length regulation. Impaired telomeric chromatin compaction results in a loss of telomere length control. Progressive telomere shortening affects chromatin compaction at telomeric and subtelomeric repeats and activates alternative telomere maintenance mechanisms. Dynamics of chromatin structure of telomeres during early mammalian development and nuclear reprogramming further indicates a central role of telomeric heterochromatin in organismal development. In addition, the recent discovery that telomeres are transcribed, giving rise to UUAGGG-repeat containing TelRNAs/TERRA, opens a new level of chromatin regulation at telomeres. Understanding the links between the epigenetic status of telomeres, TERRA/TelRNA and telomere homeostasis will open new avenues for our understanding of organismal development, cancer and ageing.


Asunto(s)
Cromatina/genética , Mamíferos/genética , ARN/genética , Animales , Cromatina/metabolismo , Cromosomas/metabolismo , Epigénesis Genética , Humanos , Mamíferos/metabolismo , ARN/metabolismo , Telómero/genética , Telómero/metabolismo , Proteínas de Unión a Telómeros/genética , Proteínas de Unión a Telómeros/metabolismo
6.
Proc Natl Acad Sci U S A ; 106(46): 19393-8, 2009 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-19887628

RESUMEN

Telomeres are heterochromatic structures at chromosome ends essential for chromosomal stability. Telomere shortening and the accumulation of dysfunctional telomeres are associated with organismal aging. Using telomerase-deficient TRF2-overexpressing mice (K5TRF2/Terc(-/-)) as a model for accelerated aging, we show that telomere shortening is paralleled by a gradual deregulation of the mammalian transcriptome leading to cumulative changes in a defined set of genes, including up-regulation of the mTOR and Akt survival pathways and down-regulation of cell cycle and DNA repair pathways. Increased DNA damage from dysfunctional telomeres leads to reduced deposition of H3K27me3 onto the inactive X chromosome (Xi), impaired association of the Xi with telomeric transcript accumulations (Tacs), and reactivation of an X chromosome-linked K5TRF2 transgene that is subjected to X-chromosome inactivation in female mice with sufficiently long telomeres. Exogenously induced DNA damage also disrupts Xi-Tacs, suggesting DNA damage at the origin of these alterations. Collectively, these findings suggest that critically short telomeres activate a persistent DNA damage response that alters gene expression programs in a nonstochastic manner toward cell cycle arrest and activation of survival pathways, as well as impacts the maintenance of epigenetic memory and nuclear organization, thereby contributing to organismal aging.


Asunto(s)
Envejecimiento Prematuro/genética , Daño del ADN/genética , Piel/metabolismo , Telómero/metabolismo , Inactivación del Cromosoma X , Envejecimiento Prematuro/metabolismo , Envejecimiento Prematuro/patología , Animales , Ciclo Celular/genética , Femenino , Perfilación de la Expresión Génica , Queratina-15 , Queratina-5/genética , Masculino , Ratones , Ratones Transgénicos , Piel/patología , Telomerasa/genética , Telomerasa/metabolismo , Proteína 2 de Unión a Repeticiones Teloméricas/genética , Proteína 2 de Unión a Repeticiones Teloméricas/metabolismo , Transcripción Genética
7.
FEBS Lett ; 596(1): 42-52, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34817067

RESUMEN

Mutations in many genes that control the expression, the function, or the stability of telomerase cause telomere biology disorders (TBDs), such as dyskeratosis congenita, pulmonary fibrosis, and aplastic anemia. Mutations in a subset of the genes associated with TBDs cause reductions of the telomerase RNA moiety hTR, thus limiting telomerase activity. We have recently found that loss of the trimethylguanosine synthase TGS1 increases both hTR abundance and telomerase activity and leads to telomere elongation. Here, we show that treatment with the S-adenosylmethionine analog sinefungin inhibits TGS1 activity, increases the hTR levels, and promotes telomere lengthening in different cell types. Our results hold promise for restoring telomere length in stem and progenitor cells from TBD patients with reduced hTR levels.


Asunto(s)
Metiltransferasas
8.
Nat Commun ; 13(1): 2302, 2022 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-35484160

RESUMEN

Pathways that direct the selection of the telomerase-dependent or recombination-based, alternative lengthening of telomere (ALT) maintenance pathway in cancer cells are poorly understood. Using human lung cancer cells and tumor organoids we show that formation of the 2,2,7-trimethylguanosine (TMG) cap structure at the human telomerase RNA 5' end by the Trimethylguanosine Synthase 1 (TGS1) is central for recruiting telomerase to telomeres and engaging Cajal bodies in telomere maintenance. TGS1 depletion or inhibition by the natural nucleoside sinefungin impairs telomerase recruitment to telomeres leading to Exonuclease 1 mediated generation of telomere 3' end protrusions that engage in RAD51-dependent, homology directed recombination and the activation of key features of the ALT pathway. This indicates a critical role for 2,2,7-TMG capping of the RNA component of human telomerase (hTR) in enforcing telomerase-dependent telomere maintenance to restrict the formation of telomeric substrates conductive to ALT. Our work introduces a targetable pathway of telomere maintenance that holds relevance for telomere-related diseases such as cancer and aging.


Asunto(s)
Telomerasa , Guanosina , Humanos , ARN/genética , Telomerasa/genética , Telomerasa/metabolismo , Telómero/genética , Telómero/metabolismo
9.
Methods Mol Biol ; 2324: 203-217, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34165717

RESUMEN

There is accumulating evidence that pseudogenes can produce functionally relevant lncRNAs in a tightly controlled manner. This class of transcripts has been demonstrated to play an important role in development and disease, by controlling parental gene expression. Classically, pseudogene derived lncRNAs compete with parental transcripts for miRNAs or factors that control parental mRNA metabolisms. Recently, pseudogene lncRNAs were demonstrated to take over the control of classic chromatin modifying enzymes and alter parental gene promoter activity or genome wide gene expression. Here, we discuss a new mechanism of parental gene expression controlled by the mOct4P4 lncRNA, a sense transcript derived from the murine Oct4 pseudogene 4. mOct4P4 lncRNA specifically interacts with the RNA binding protein FUS and the Histone Methyltransferase SUV39H1 to target heterochromatin formation at the parental Oct4 promoter in trans. In addition, we will address key issues for the functional dissection of epigenetic control of parental gene promoters by pseudogene lncRNAs.


Asunto(s)
Cromatina/metabolismo , Epigénesis Genética/genética , Regiones Promotoras Genéticas/genética , Seudogenes/genética , ARN Largo no Codificante/genética , Alelos , Animales , Sistemas CRISPR-Cas , Proteínas de la Cápside/metabolismo , Cromatina/genética , Cromatina/ultraestructura , Inmunoprecipitación de Cromatina , Células Madre Embrionarias , Regulación de la Expresión Génica , Humanos , Levivirus/genética , Metiltransferasas/metabolismo , Ratones , Factor 3 de Transcripción de Unión a Octámeros/genética , ARN Mensajero/metabolismo , ARN Viral/metabolismo , Proteína FUS de Unión a ARN/metabolismo , Proteínas Represoras/metabolismo
10.
Commun Biol ; 3(1): 632, 2020 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-33128015

RESUMEN

The resurrection of pseudogenes during evolution produced lncRNAs with new biological function. Here we show that pseudogene-evolution created an Oct4 pseudogene lncRNA that is able to direct epigenetic silencing of the parental Oct4 gene via a 2-step, lncRNA dependent mechanism. The murine Oct4 pseudogene 4 (mOct4P4) lncRNA recruits the RNA binding protein FUS to allow the binding of the SUV39H1 HMTase to a defined mOct4P4 lncRNA sequence element. The mOct4P4-FUS-SUV39H1 silencing complex holds target site specificity for the parental Oct4 promoter and interference with individual components results in loss of Oct4 silencing. SUV39H1 and FUS do not bind parental Oct4 mRNA, confirming the acquisition of a new biological function by the mOct4P4 lncRNA. Importantly, all features of mOct4P4 function are recapitulated by the human hOCT4P3 pseudogene lncRNA, indicating evolutionary conservation. Our data highlight the biological relevance of rapidly evolving lncRNAs that infiltrate into central epigenetic regulatory circuits in vertebrate cells.


Asunto(s)
Metiltransferasas/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/genética , Seudogenes , ARN Largo no Codificante/genética , Proteína FUS de Unión a ARN/genética , Proteínas Represoras/metabolismo , Animales , Línea Celular Tumoral , Epigénesis Genética , Femenino , Silenciador del Gen , Humanos , Metiltransferasas/genética , Ratones , Complejos Multiproteicos/genética , Neoplasias Ováricas/genética , Regiones Promotoras Genéticas , Proteína FUS de Unión a ARN/metabolismo , Proteínas Represoras/genética
11.
Cancers (Basel) ; 12(12)2020 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-33371412

RESUMEN

Tumor organoids are tridimensional cell culture systems that are generated in vitro from surgically resected patients' tumors. They can be propagated in culture maintaining several features of the tumor of origin, including cellular and genetic heterogeneity, thus representing a promising tool for precision cancer medicine. Here, we established patient-derived tumor organoids (PDOs) from different breast cancer subtypes (luminal A, luminal B, human epidermal growth factor receptor 2 (HER2)-enriched, and triple negative). The established model systems showed histological and genomic concordance with parental tumors. However, in PDOs, the ratio of diverse cell populations was frequently different from that originally observed in parental tumors. We showed that tumor organoids represent a valuable system to test the efficacy of standard therapeutic treatments and to identify drug resistant populations within tumors. We also report that inhibitors of mechanosignaling and of Yes-associated protein 1 (YAP) activation can restore chemosensitivity in drug resistant tumor organoids.

12.
Cell Rep ; 30(5): 1358-1372.e5, 2020 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-32023455

RESUMEN

Biogenesis of the human telomerase RNA (hTR) involves a complex series of posttranscriptional modifications, including hypermethylation of the 5' mono-methylguanosine cap to a tri-methylguanosine cap (TMG). How the TMG cap affects hTR maturation is unknown. Here, we show that depletion of trimethylguanosine synthase 1 (TGS1), the enzyme responsible for cap hypermethylation, increases levels of hTR and telomerase. Diminished trimethylation increases hTR association with the cap-binding complex (CBC) and with Sm chaperone proteins. Loss of TGS1 causes an increase in accumulation of mature hTR in both the nucleus and the cytoplasm compared with controls. In TGS1 mutant cells, increased hTR assembles with telomerase reverse transcriptase (TERT) protein to yield elevated active telomerase complexes and increased telomerase activity, resulting in telomere elongation in cultured human cells. Our results show that TGS1-mediated hypermethylation of the hTR cap inhibits hTR accumulation, restrains levels of assembled telomerase, and limits telomere elongation.


Asunto(s)
Metiltransferasas/deficiencia , ARN/metabolismo , Telomerasa/metabolismo , Telómero/metabolismo , Biocatálisis , Cuerpos Enrollados/metabolismo , Guanosina/metabolismo , Células HEK293 , Células HeLa , Humanos , Metilación , Metiltransferasas/genética , Modelos Biológicos , Mutación/genética , Poliadenilación , Caperuzas de ARN/metabolismo , Fracciones Subcelulares/metabolismo
13.
Nat Commun ; 10(1): 1001, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30824709

RESUMEN

In vertebrates, the telomere repeat containing long, non-coding RNA TERRA is prone to form RNA:DNA hybrids at telomeres. This results in the formation of R-loop structures, replication stress and telomere instability, but also contributes to alternative lengthening of telomeres (ALT). Here, we identify the TERRA binding proteins NONO and SFPQ as novel regulators of RNA:DNA hybrid related telomere instability. NONO and SFPQ locate at telomeres and have a common role in suppressing RNA:DNA hybrids and replication defects at telomeres. NONO and SFPQ act as heterodimers to suppress fragility and homologous recombination at telomeres, respectively. Combining increased telomere fragility with unleashing telomere recombination upon NONO/SFPQ loss of function causes massive recombination events, involving 35% of telomeres in ALT cells. Our data identify the RNA binding proteins SFPQ and NONO as novel regulators at telomeres that collaborate to ensure telomere integrity by suppressing telomere fragility and homologous recombination triggered by RNA:DNA hybrids.


Asunto(s)
ADN/metabolismo , Proteínas Asociadas a Matriz Nuclear/metabolismo , Hibridación de Ácido Nucleico , Factores de Transcripción de Octámeros/metabolismo , Factor de Empalme Asociado a PTB/metabolismo , Proteínas de Unión al ARN/metabolismo , ARN/metabolismo , Telómero/metabolismo , Animales , Línea Celular Tumoral , Replicación del ADN , Proteínas de Unión al ADN , Recombinación Homóloga , Humanos , Ratones , ARN no Traducido , Homeostasis del Telómero , Proteínas de Unión a Telómeros/metabolismo
14.
Elife ; 82019 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-31426913

RESUMEN

The mechanisms that regulate pluripotency are still largely unknown. Here, we show that Telomere Repeat Binding Factor 1 (TRF1), a component of the shelterin complex, regulates the genome-wide binding of polycomb and polycomb H3K27me3 repressive marks to pluripotency genes, thereby exerting vast epigenetic changes that contribute to the maintenance of mouse ES cells in a naïve state. We further show that TRF1 mediates these effects by regulating TERRA, the lncRNAs transcribed from telomeres. We find that TERRAs are enriched at polycomb and stem cell genes in pluripotent cells and that TRF1 abrogation results in increased TERRA levels and in higher TERRA binding to those genes, coincidental with the induction of cell-fate programs and the loss of the naïve state. These results are consistent with a model in which TRF1-dependent changes in TERRA levels modulate polycomb recruitment to pluripotency and differentiation genes. These unprecedented findings explain why TRF1 is essential for the induction and maintenance of pluripotency.


Asunto(s)
Epigénesis Genética , Células Madre Pluripotentes Inducidas/metabolismo , Complejo Represivo Polycomb 2/metabolismo , ARN Largo no Codificante/metabolismo , Proteína 1 de Unión a Repeticiones Teloméricas/metabolismo , Transcripción Genética , Animales , Diferenciación Celular , Células Cultivadas , Ratones
15.
Oncotarget ; 8(56): 95674-95691, 2017 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-29221158

RESUMEN

The catalytic subunit of the telomerase complex, hTERT, ensures unlimited proliferative potential of cancer cells by maintaining telomere function and protecting from apoptosis. Using a miRNA screening approach we identified miR-296-5p and miR-512-5p as miRNAs that target hTERT in breast cancer cells. Ectopic miR-296-5p and miR-512-5p reduce telomerase activity, drive telomere shortening and cause proliferation defects by enhancing senescence and apoptosis in breast cancer cells. In line with the relevance of hTERT expression for human cancer we found that miR-296-5p and miR-512-5p expression is reduced in human breast cancer. Accordingly, high expression of miR-296-5p and miR-512-5p target genes including hTERT is linked with significantly reduced distant metastasis free survival and relapse free survival of basal type breast cancer patients. This suggests relevance of the identified miRNAs in basal type breast cancer. Epigenetic silencing of miR-296 and miR-512 encoding genes is responsible for low levels of miR-296-5p and miR-512-5p expression in basal type breast cancer cells. Disrupting gene silencing results in a dramatic upregulation of miR-296-5p and miR-512-5p levels leading to reduced hTERT expression and increased sensitivity to the induction of apoptosis. Altogether, our data suggest that epigenetic regulatory circuits in basal type breast cancer may contribute to high hTERT levels by silencing miR-296-5p and miR-512-5p expression, thereby contributing to the aggressiveness of basal type breast cancer.

16.
Cell Cycle ; 14(9): 1438-46, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25789788

RESUMEN

The Suv39h1 and Suv39h2 H3K9 histone methyltransferases (HMTs) have a conserved role in the formation of constitutive heterochromatin and gene silencing. Using a transgenic mouse model system we demonstrate that elevated expression of Suv39h1 increases global H3K9me3 levels in vivo. More specifically, Suv39h1 overexpression enhances the imposition of H3K9me3 levels at constitutive heterochromatin at telomeric and major satellite repeats in primary mouse embryonic fibroblasts. Chromatin compaction is paralleled by telomere shortening, indicating that telomere length is controlled by H3K9me3 density at telomeres. We further show that increased Suv39h1 levels result in an impaired clonogenic potential of transgenic epidermal stem cells and Ras/E1A transduced transgenic primary mouse embryonic fibroblasts. Importantly, Suv39h1 overexpression in mice confers resistance to a DMBA/TPA induced skin carcinogenesis protocol that is characterized by the accumulation of activating H-ras mutations. Our results provide genetic evidence that Suv39h1 controls telomere homeostasis and mediates resistance to oncogenic stress in vivo. This identifies Suv39h1 as an interesting target to improve oncogene induced senescence in premalignant lesions.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Epidermis/enzimología , Metiltransferasas/metabolismo , Proteínas Represoras/metabolismo , Neoplasias Cutáneas/prevención & control , Homeostasis del Telómero , Telómero/metabolismo , 9,10-Dimetil-1,2-benzantraceno , Animales , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Ensamble y Desensamble de Cromatina , Proteínas Cromosómicas no Histona/metabolismo , Epidermis/patología , Femenino , Histonas/metabolismo , Humanos , Masculino , Metilación , Metiltransferasas/genética , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Transgénicos , Mutación , Proteínas Represoras/genética , Neoplasias Cutáneas/inducido químicamente , Neoplasias Cutáneas/enzimología , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Acortamiento del Telómero , Proteínas ras/genética , Proteínas ras/metabolismo
17.
Nat Commun ; 6: 7631, 2015 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-26158551

RESUMEN

Pseudogene-derived, long non-coding RNAs (lncRNAs) act as epigenetic regulators of gene expression. Here we present a panel of new mouse Oct4 pseudogenes and demonstrate that the X-linked Oct4 pseudogene Oct4P4 critically impacts mouse embryonic stem cells (mESCs) self-renewal. Sense Oct4P4 transcription produces a spliced, nuclear-restricted lncRNA that is efficiently upregulated during mESC differentiation. Oct4P4 lncRNA forms a complex with the SUV39H1 HMTase to direct the imposition of H3K9me3 and HP1α to the promoter of the ancestral Oct4 gene, located on chromosome 17, leading to gene silencing and reduced mESC self-renewal. Targeting Oct4P4 expression in primary mouse embryonic fibroblasts causes the re-acquisition of self-renewing features of mESC. We demonstrate that Oct4P4 lncRNA plays an important role in inducing and maintaining silencing of the ancestral Oct4 gene in differentiating mESCs. Our data introduces a sense pseudogene-lncRNA-based mechanism of epigenetic gene regulation that controls the cross-talk between pseudogenes and their ancestral genes.


Asunto(s)
Epigénesis Genética/genética , Regulación del Desarrollo de la Expresión Génica , Metiltransferasas/metabolismo , Células Madre Embrionarias de Ratones/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/genética , Seudogenes/genética , ARN Largo no Codificante/genética , Proteínas Represoras/metabolismo , Animales , Línea Celular , Línea Celular Tumoral , Autorrenovación de las Células/genética , Inmunoprecipitación de Cromatina , Homólogo de la Proteína Chromobox 5 , Proteínas Cromosómicas no Histona/metabolismo , Histonas/metabolismo , Inmunoprecipitación , Ratones , Células Madre Embrionarias de Ratones/citología , Células 3T3 NIH
18.
Cell Rep ; 8(2): 487-500, 2014 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-25043184

RESUMEN

Sox4 expression is restricted in mammals to embryonic structures and some adult tissues, such as lymphoid organs, pancreas, intestine, and skin. During embryogenesis, Sox4 regulates mesenchymal and neural progenitor survival, as well as lymphocyte and myeloid differentiation, and contributes to pancreas, bone, and heart development. Aberrant Sox4 expression is linked to malignant transformation and metastasis in several types of cancer. To understand the role of Sox4 in the adult organism, we first generated mice with reduced whole-body Sox4 expression. These mice display accelerated aging and reduced cancer incidence. To specifically address a role for Sox4 in adult stem cells, we conditionally deleted Sox4 (Sox4(cKO)) in stratified epithelia. Sox4(cKO) mice show increased skin stem cell quiescence and resistance to chemical carcinogenesis concomitantly with downregulation of cell cycle, DNA repair, and activated hair follicle stem cell pathways. Altogether, these findings highlight the importance of Sox4 in regulating adult tissue homeostasis and cancer.


Asunto(s)
Células Madre Adultas/metabolismo , Envejecimiento/genética , Carcinogénesis/genética , Folículo Piloso/metabolismo , Factores de Transcripción SOXC/metabolismo , Células Madre Adultas/citología , Animales , Carcinogénesis/metabolismo , Ciclo Celular , Reparación del ADN , Folículo Piloso/citología , Queratinocitos/citología , Queratinocitos/metabolismo , Ratones , Factores de Transcripción SOXC/genética , Vía de Señalización Wnt
19.
Cancer Res ; 74(15): 4145-56, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24876105

RESUMEN

Telomeres consist of DNA tandem repeats that recruit the multiprotein complex shelterin to build a chromatin structure that protects chromosome ends. Although cancer formation is linked to alterations in telomere homeostasis, there is little understanding of how shelterin function is limited in cancer cells. Using a small-scale screening approach, we identified miR-155 as a key regulator in breast cancer cell expression of the shelterin component TERF1 (TRF1). miR-155 targeted a conserved sequence motif in the 3'UTR of TRF1, resulting in its translational repression. miR-155 was upregulated commonly in breast cancer specimens, as associated with reduced TRF1 protein expression, metastasis-free survival, and relapse-free survival in estrogen receptor-positive cases. Modulating miR-155 expression in cells altered TRF1 levels and TRF1 abundance at telomeres. Compromising TRF1 expression by elevating miR-155 increased telomere fragility and altered the structure of metaphase chromosomes. In contrast, reducing miR-155 levels improved telomere function and genomic stability. These results implied that miR-155 upregulation antagonizes telomere integrity in breast cancer cells, increasing genomic instability linked to poor clinical outcome in estrogen receptor-positive disease. Our work argued that miRNA-dependent regulation of shelterin function has a clinically significant impact on telomere function, suggesting the existence of "telo-miRNAs" that have an impact on cancer and aging.


Asunto(s)
Neoplasias de la Mama/genética , MicroARNs/genética , Telómero/genética , Proteína 1 de Unión a Repeticiones Teloméricas/genética , Regiones no Traducidas 3' , Animales , Secuencia de Bases , Neoplasias de la Mama/metabolismo , Técnicas de Cultivo de Célula , Femenino , Células HCT116 , Células HeLa , Humanos , Células MCF-7 , MicroARNs/metabolismo , Datos de Secuencia Molecular , Homología de Secuencia de Ácido Nucleico , Telómero/metabolismo , Proteína 1 de Unión a Repeticiones Teloméricas/metabolismo , Transfección
20.
Cancer Res ; 70(17): 6925-33, 2010 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-20713524

RESUMEN

Loss-of-function mutations of retinoblastoma family (Rb) proteins drive tumorigenesis by overcoming barriers to cellular proliferation. Consequently, factors modulating Rb function are of great clinical import. Here, we show that miR-335 is differentially expressed in human cancer cells and that it tightly regulates the expression of Rb1 (pRb/p105) by specifically targeting a conserved sequence motif in its 3' untranslated region. We found that by altering Rb1 (pRb/p105) levels, miR-335 activates the p53 tumor suppressor pathway to limit cell proliferation and neoplastic cell transformation. DNA damage elicited an increase in miR-335 expression in a p53-dependent manner. miR-335 and p53 cooperated in a positive feedback loop to drive cell cycle arrest. Together, these results indicate that miR-335 helps control proliferation by balancing the activities of the Rb and p53 tumor suppressor pathways. Further, they establish that miR-335 activation plays an important role in the induction of p53-dependent cell cycle arrest after DNA damage.


Asunto(s)
MicroARNs/genética , Neoplasias/genética , Proteína de Retinoblastoma/genética , Proteína p53 Supresora de Tumor/metabolismo , Animales , Ciclo Celular , Procesos de Crecimiento Celular/genética , Línea Celular Tumoral , Daño del ADN , Retroalimentación , Humanos , Ratones , MicroARNs/biosíntesis , Células 3T3 NIH , Neoplasias/metabolismo , Neoplasias/patología , ARN Interferente Pequeño/genética , Proteína de Retinoblastoma/biosíntesis , Proteína de Retinoblastoma/metabolismo , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA