Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Nucleic Acids Res ; 50(5): 2681-2699, 2022 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-35189637

RESUMEN

Cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS) is activated in cells with defective DNA damage repair and signaling (DDR) factors, but a direct role for DDR factors in regulating cGAS activation in response to micronuclear DNA is still poorly understood. Here, we provide novel evidence that Nijmegen breakage syndrome 1 (NBS1) protein, a well-studied DNA double-strand break (DSB) sensor-in coordination with Ataxia Telangiectasia Mutated (ATM), a protein kinase, and Carboxy-terminal binding protein 1 interacting protein (CtIP), a DNA end resection factor-functions as an upstream regulator that prevents cGAS from binding micronuclear DNA. When NBS1 binds to micronuclear DNA via its fork-head-associated domain, it recruits CtIP and ATM via its N- and C-terminal domains, respectively. Subsequently, ATM stabilizes NBS1's interaction with micronuclear DNA, and CtIP converts DSB ends into single-strand DNA ends; these two key events prevent cGAS from binding micronuclear DNA. Additionally, by using a cGAS tripartite system, we show that cells lacking NBS1 not only recruit cGAS to a major fraction of micronuclear DNA but also activate cGAS in response to these micronuclear DNA. Collectively, our results underscore how NBS1 and its binding partners prevent cGAS from binding micronuclear DNA, in addition to their classical functions in DDR signaling.


Asunto(s)
Proteínas de Ciclo Celular , Proteínas Supresoras de Tumor , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Proteínas de Ciclo Celular/metabolismo , ADN/genética , Daño del ADN , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteínas Nucleares/metabolismo , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/metabolismo , Proteínas Serina-Treonina Quinasas , Proteínas Supresoras de Tumor/genética
2.
J Biol Chem ; 298(7): 102102, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35667440

RESUMEN

Oxidative DNA damage contributes to aging and the pathogenesis of numerous human diseases including cancer. 8-hydroxyguanine (8-oxoG) is the major product of oxidative DNA lesions. Although OGG1-mediated base excision repair is the primary mechanism for 8-oxoG removal, DNA mismatch repair has also been implicated in processing oxidative DNA damage. However, the mechanism of the latter is not fully understood. Here, we treated human cells defective in various 8-oxoG repair factors with H2O2 and performed biochemical, live cell imaging, and chromatin immunoprecipitation sequencing analyses to determine their response to the treatment. We show that the mismatch repair processing of oxidative DNA damage involves cohesive interactions between mismatch recognition protein MutSα, histone mark H3K36me3, and H3K36 trimethyltransferase SETD2, which activates the ATM DNA damage signaling pathway. We found that cells depleted of MutSα or SETD2 accumulate 8-oxoG adducts and fail to trigger H2O2-induced ATM activation. Furthermore, we show that SETD2 physically interacts with both MutSα and ATM, which suggests a role for SETD2 in transducing DNA damage signals from lesion-bound MutSα to ATM. Consistently, MutSα and SETD2 are highly coenriched at oxidative damage sites. The data presented here support a model wherein MutSα, SETD2, ATM, and H3K36me3 constitute a positive feedback loop to help cells cope with oxidative DNA damage.


Asunto(s)
Reparación de la Incompatibilidad de ADN , N-Metiltransferasa de Histona-Lisina , Proteínas MutS , Estrés Oxidativo , Daño del ADN , Código de Histonas , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/genética , Humanos , Peróxido de Hidrógeno/farmacología , Proteínas MutS/genética , Proteínas MutS/metabolismo
3.
Proc Natl Acad Sci U S A ; 117(38): 23597-23605, 2020 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-32900932

RESUMEN

Trinucleotide repeat (TNR) expansions cause nearly 20 severe human neurological diseases which are currently untreatable. For some of these diseases, ongoing somatic expansions accelerate disease progression and may influence age of onset. This new knowledge emphasizes the importance of understanding the protein factors that drive expansions. Recent genetic evidence indicates that the mismatch repair factor MutSß (Msh2-Msh3 complex) and the histone deacetylase HDAC3 function in the same pathway to drive triplet repeat expansions. Here we tested the hypothesis that HDAC3 deacetylates MutSß and thereby activates it to drive expansions. The HDAC3-selective inhibitor RGFP966 was used to examine its biological and biochemical consequences in human tissue culture cells. HDAC3 inhibition efficiently suppresses repeat expansion without impeding canonical mismatch repair activity. Five key lysine residues in Msh3 are direct targets of HDAC3 deacetylation. In cells expressing Msh3 in which these lysine residues are mutated to arginine, the inhibitory effect of RGFP966 on expansions is largely bypassed, consistent with the direct deacetylation hypothesis. RGFP966 treatment does not alter MutSß subunit abundance or complex formation but does partially control its subcellular localization. Deacetylation sites in Msh3 overlap a nuclear localization signal, and we show that localization of MutSß is partially dependent on HDAC3 activity. Together, these results indicate that MutSß is a key target of HDAC3 deacetylation and provide insights into an innovative regulatory mechanism for triplet repeat expansions. The results suggest expansion activity may be druggable and support HDAC3-selective inhibition as an attractive therapy in some triplet repeat expansion diseases.


Asunto(s)
Reparación de la Incompatibilidad de ADN/genética , Histona Desacetilasas , Expansión de Repetición de Trinucleótido/genética , Acetilación/efectos de los fármacos , Acrilamidas/farmacología , Línea Celular , Células Cultivadas , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Humanos , Fenilendiaminas/farmacología
4.
J Biol Chem ; 297(4): 101144, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34473992

RESUMEN

Huntington's disease (HD), a neurodegenerative disease characterized by progressive dementia, psychiatric problems, and chorea, is known to be caused by CAG repeat expansions in the HD gene HTT. However, the mechanism of this pathology is not fully understood. The translesion DNA polymerase θ (Polθ) carries a large insertion sequence in its catalytic domain, which has been shown to allow DNA loop-outs in the primer strand. As a result of high levels of oxidative DNA damage in neural cells and Polθ's subsequent involvement in base excision repair of oxidative DNA damage, we hypothesized that Polθ contributes to CAG repeat expansion while repairing oxidative damage within HTT. Here, we performed Polθ-catalyzed in vitro DNA synthesis using various CAG•CTG repeat DNA substrates that are similar to base excision repair intermediates. We show that Polθ efficiently extends (CAG)n•(CTG)n hairpin primers, resulting in hairpin retention and repeat expansion. Polθ also triggers repeat expansions to pass the threshold for HD when the DNA template contains 35 repeats upward. Strikingly, Polθ depleted of the catalytic insertion fails to induce repeat expansions regardless of primers and templates used, indicating that the insertion sequence is responsible for Polθ's error-causing activity. In addition, the level of chromatin-bound Polθ in HD cells is significantly higher than in non-HD cells and exactly correlates with the degree of CAG repeat expansion, implying Polθ's involvement in triplet repeat instability. Therefore, we have identified Polθ as a potent factor that promotes CAG•CTG repeat expansions in HD and other neurodegenerative disorders.


Asunto(s)
Reparación del ADN , ADN Polimerasa Dirigida por ADN/química , Enfermedad de Huntington/enzimología , Expansión de Repetición de Trinucleótido , Dominio Catalítico , Daño del ADN , ADN Polimerasa Dirigida por ADN/genética , ADN Polimerasa Dirigida por ADN/metabolismo , Células HeLa , Humanos , Enfermedad de Huntington/genética , ADN Polimerasa theta
5.
J Biol Chem ; 294(17): 7037-7045, 2019 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-30858175

RESUMEN

Proliferating cell nuclear antigen (PCNA) and its posttranslational modifications regulate DNA metabolic reactions, including DNA replication and repair, at replication forks. PCNA phosphorylation at Tyr-211 (PCNA-Y211p) inhibits DNA mismatch repair and induces misincorporation during DNA synthesis. Here, we describe an unexpected role of PCNA-Y211p in cancer promotion and development. Cells expressing phosphorylation-mimicking PCNA, PCNA-Y211D, show elevated hallmarks specific to the epithelial-mesenchymal transition (EMT), including the up-regulation of the EMT-promoting factor Snail and the down-regulation of EMT-inhibitory factors E-cadherin and GSK3ß. The PCNA-Y211D-expressing cells also exhibited active cell migration and underwent G2/M arrest. Interestingly, all of these EMT-associated activities required the activation of ATM and Akt kinases, as inactivating these protein kinases by gene knockdown or inhibitors blocked EMT-associated signaling and cell migration. We concluded that PCNA phosphorylation promotes cancer progression via the ATM/Akt/GSK3ß/Snail signaling pathway. In conclusion, this study identifies a novel PCNA function and reveals the molecular basis of phosphorylated PCNA-mediated cancer development and progression.


Asunto(s)
Neoplasias/patología , Antígeno Nuclear de Célula en Proliferación/metabolismo , Transducción de Señal , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Movimiento Celular , Progresión de la Enfermedad , Transición Epitelial-Mesenquimal , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Células HeLa , Humanos , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factores de Transcripción de la Familia Snail/metabolismo
6.
EMBO J ; 34(13): 1829-43, 2015 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-25921062

RESUMEN

During nuclear DNA replication, proofreading-deficient DNA polymerase α (Pol α) initiates Okazaki fragment synthesis with lower fidelity than bulk replication by proofreading-proficient Pol δ or Pol ε. Here, we provide evidence that the exonuclease activity of mammalian flap endonuclease (FEN1) excises Pol α replication errors in a MutSα-dependent, MutLα-independent mismatch repair process we call Pol α-segment error editing (AEE). We show that MSH2 interacts with FEN1 and facilitates its nuclease activity to remove mismatches near the 5' ends of DNA substrates. Mouse cells and mice encoding FEN1 mutations display AEE deficiency, a strong mutator phenotype, enhanced cellular transformation, and increased cancer susceptibility. The results identify a novel role for FEN1 in a specialized mismatch repair pathway and a new cancer etiological mechanism.


Asunto(s)
Reparación de la Incompatibilidad de ADN , ADN Polimerasa I/metabolismo , ADN/metabolismo , Endonucleasas de ADN Solapado/metabolismo , Proteína MutS de Unión a los Apareamientos Incorrectos del ADN/metabolismo , Animales , Células Cultivadas , Reparación de la Incompatibilidad de ADN/genética , Replicación del ADN/genética , Embrión de Mamíferos , Femenino , Endonucleasas de ADN Solapado/clasificación , Endonucleasas de ADN Solapado/genética , Células HEK293 , Células HeLa , Humanos , Masculino , Ratones , Ratones Transgénicos , Proteína MutS de Unión a los Apareamientos Incorrectos del ADN/genética , Saccharomyces cerevisiae
7.
RNA ; 22(11): 1710-1718, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27672132

RESUMEN

RNA nanotechnology is rapidly emerging. Due to advantageous pharmacokinetics and favorable in vivo biodistribution, RNA nanoparticles have shown promise in targeted delivery of therapeutics. RNA nanotechnology applies bottom-up assembly, thus elucidation of the mechanism of interaction between multiple components is of fundamental importance. The tendency of diminishing concern about RNA instability has accelerated by the finding of the novel thermostable three-way junction (3WJ) motif of the phi29 DNA-packaging motor. The kinetics of these three components, each averaging 18 nucleotides (nt), was investigated to elucidate the mechanism for producing the stable 3WJ. The three fragments coassembled into the 3WJ with extraordinary speed and affinity via a two-step reaction mechanism, 3WJb + 3WJc ↔ 3WJbc + 3WJa ↔ 3WJabc The first step of reaction between 3WJb and 3WJc is highly dynamic since these two fragments only contain 8 nt for complementation. In the second step, the 3WJa, which contains 17 nt complementary to the 3WJbc complex, locks the unstable 3WJbc complex into a highly stable 3WJ. The resulting pRNA-3WJ is more stable than any of the dimer species as shown in the much more rapid association rates and slowest dissociation rate constant. The second step occurs at a very high association rate that is difficult to quantify, resulting in a rapid formation of a stable 3WJ. Elucidation of the mechanism of three-component collision in producing the ultrastable 3WJ proves a promising platform for bottom-up assembly of RNA nanoparticles as a new class of anion polymers for material science, electronic elements, or therapeutic reagents.


Asunto(s)
Bacteriófagos/genética , Empaquetamiento del ADN , ADN Viral/genética , Estabilidad del ARN , ADN Viral/química , Dimerización , Cinética , Resonancia por Plasmón de Superficie
8.
Proc Natl Acad Sci U S A ; 112(18): 5667-72, 2015 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-25825764

RESUMEN

Proliferating cell nuclear antigen (PCNA) plays essential roles in eukaryotic cells during DNA replication, DNA mismatch repair (MMR), and other events at the replication fork. Earlier studies show that PCNA is regulated by posttranslational modifications, including phosphorylation of tyrosine 211 (Y211) by the epidermal growth factor receptor (EGFR). However, the functional significance of Y211-phosphorylated PCNA remains unknown. Here, we show that PCNA phosphorylation by EGFR alters its interaction with mismatch-recognition proteins MutSα and MutSß and interferes with PCNA-dependent activation of MutLα endonuclease, thereby inhibiting MMR at the initiation step. Evidence is also provided that Y211-phosphorylated PCNA induces nucleotide misincorporation during DNA synthesis. These findings reveal a novel mechanism by which Y211-phosphorylated PCNA promotes cancer development and progression via facilitating error-prone DNA replication and suppressing the MMR function.


Asunto(s)
Reparación de la Incompatibilidad de ADN , ADN/biosíntesis , Receptores ErbB/metabolismo , Neoplasias/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Línea Celular Tumoral , Proliferación Celular , Neoplasias Colorrectales/metabolismo , Progresión de la Enfermedad , Genoma , Genoma Humano , Células HeLa , Humanos , Microscopía Fluorescente , Modelos Moleculares , Mutación , Neoplasias/genética , Fosforilación , Unión Proteica , Análisis de Secuencia de ADN , Tirosina/química
10.
J Biol Chem ; 290(23): 14536-41, 2015 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-25907674

RESUMEN

Both genotoxic and non-genotoxic chemicals can act as carcinogens. However, while genotoxic compounds lead directly to mutations that promote unregulated cell growth, the mechanism by which non-genotoxic carcinogens lead to cellular transformation is poorly understood. Using a model non-genotoxic carcinogen, arsenic, we show here that exposure to arsenic inhibits mismatch repair (MMR) in human cells, possibly through its ability to stimulate epidermal growth factor receptor (EGFR)-dependent tyrosine phosphorylation of proliferating cellular nuclear antigen (PCNA). HeLa cells exposed to exogenous arsenic demonstrate a dose- and time-dependent increase in the levels of EGFR and tyrosine 211-phosphorylated PCNA. Cell extracts derived from arsenic-treated HeLa cells are defective in MMR, and unphosphorylated recombinant PCNA restores normal MMR activity to these extracts. These results suggest a model in which arsenic induces expression of EGFR, which in turn phosphorylates PCNA, and phosphorylated PCNA then inhibits MMR, leading to increased susceptibility to carcinogenesis. This study suggests a putative novel mechanism of action for arsenic and other non-genotoxic carcinogens.


Asunto(s)
Arsénico/toxicidad , Carcinogénesis/inducido químicamente , Carcinógenos/toxicidad , Reparación de la Incompatibilidad de ADN/efectos de los fármacos , Receptores ErbB/genética , Antígeno Nuclear de Célula en Proliferación/metabolismo , Carcinogénesis/genética , Carcinogénesis/metabolismo , Receptores ErbB/análisis , Receptores ErbB/metabolismo , Células HeLa , Humanos , Fosforilación/efectos de los fármacos , Antígeno Nuclear de Célula en Proliferación/análisis , Regulación hacia Arriba/efectos de los fármacos
11.
Cell Res ; 31(5): 542-553, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33510387

RESUMEN

DNA mismatch repair (MMR) relies on MutS and MutL ATPases for mismatch recognition and strand-specific nuclease recruitment to remove mispaired bases in daughter strands. However, whether the MutS-MutL complex coordinates MMR by ATP-dependent sliding on DNA or protein-protein interactions between the mismatch and strand discrimination signal is ambiguous. Using functional MMR assays and systems preventing proteins from sliding, we show that sliding of human MutSα is required not for MMR initiation, but for final mismatch removal. MutSα recruits MutLα to form a mismatch-bound complex, which initiates MMR by nicking the daughter strand 5' to the mismatch. Exonuclease 1 (Exo1) is then recruited to the nick and conducts 5' → 3' excision. ATP-dependent MutSα dissociation from the mismatch is necessary for Exo1 to remove the mispaired base when the excision reaches the mismatch. Therefore, our study has resolved a long-standing puzzle, and provided new insights into the mechanism of MMR initiation and mispair removal.


Asunto(s)
Disparidad de Par Base , Reparación de la Incompatibilidad de ADN , Adenosina Trifosfatasas , ADN , Reparación del ADN , Proteínas de Unión al ADN/metabolismo , Humanos , Proteína MutS de Unión a los Apareamientos Incorrectos del ADN/genética
12.
Nat Commun ; 12(1): 2954, 2021 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-34012010

RESUMEN

How cancer cells cope with high levels of replication stress during rapid proliferation is currently unclear. Here, we show that macrophage migration inhibitory factor (MIF) is a 3' flap nuclease that translocates to the nucleus in S phase. Poly(ADP-ribose) polymerase 1 co-localizes with MIF to the DNA replication fork, where MIF nuclease activity is required to resolve replication stress and facilitates tumor growth. MIF loss in cancer cells leads to mutation frequency increases, cell cycle delays and DNA synthesis and cell growth inhibition, which can be rescued by restoring MIF, but not nuclease-deficient MIF mutant. MIF is significantly upregulated in breast tumors and correlates with poor overall survival in patients. We propose that MIF is a unique 3' nuclease, excises flaps at the immediate 3' end during DNA synthesis and favors cancer cells evading replication stress-induced threat for their growth.


Asunto(s)
Neoplasias de la Mama/metabolismo , Replicación del ADN/fisiología , Endonucleasas de ADN Solapado/metabolismo , Oxidorreductasas Intramoleculares/metabolismo , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular , ADN/química , ADN/metabolismo , Daño del ADN , ADN Polimerasa III/genética , ADN Polimerasa III/metabolismo , Replicación del ADN/genética , Femenino , Endonucleasas de ADN Solapado/deficiencia , Endonucleasas de ADN Solapado/genética , Técnicas de Inactivación de Genes , Inestabilidad Genómica , Células HCT116 , Humanos , Oxidorreductasas Intramoleculares/deficiencia , Oxidorreductasas Intramoleculares/genética , Factores Inhibidores de la Migración de Macrófagos/deficiencia , Factores Inhibidores de la Migración de Macrófagos/genética , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Conformación de Ácido Nucleico , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Fase S , Especificidad por Sustrato
13.
Cancer Cell ; 39(1): 109-121.e5, 2021 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-33338427

RESUMEN

Tumors with defective mismatch repair (dMMR) are responsive to immunotherapy because of dMMR-induced neoantigens and activation of the cGAS-STING pathway. While neoantigens result from the hypermutable nature of dMMR, it is unknown how dMMR activates the cGAS-STING pathway. We show here that loss of the MutLα subunit MLH1, whose defect is responsible for ~50% of dMMR cancers, results in loss of MutLα-specific regulation of exonuclease 1 (Exo1) during DNA repair. This leads to unrestrained DNA excision by Exo1, which causes increased single-strand DNA formation, RPA exhaustion, DNA breaks, and aberrant DNA repair intermediates. Ultimately, this generates chromosomal abnormalities and the release of nuclear DNA into the cytoplasm, activating the cGAS-STING pathway. In this study, we discovered a hitherto unknown MMR mechanism that modulates genome stability and has implications for cancer therapy.


Asunto(s)
Aberraciones Cromosómicas , Enzimas Reparadoras del ADN/metabolismo , Exodesoxirribonucleasas/metabolismo , Homólogo 1 de la Proteína MutL/deficiencia , Neoplasias/genética , Transducción de Señal , Animales , Línea Celular Tumoral , Roturas del ADN de Cadena Simple , Reparación de la Incompatibilidad de ADN , Reparación del ADN , ADN de Cadena Simple/metabolismo , Células HeLa , Humanos , Proteínas de la Membrana/metabolismo , Ratones , Homólogo 1 de la Proteína MutL/metabolismo , Neoplasias/metabolismo , Nucleotidiltransferasas/metabolismo , Proteína de Replicación A/metabolismo
14.
Nat Chem ; 10(8): 873-880, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29915346

RESUMEN

The human DNA repair enzyme MUTYH excises mispaired adenine residues in oxidized DNA. Homozygous MUTYH mutations underlie the autosomal, recessive cancer syndrome MUTYH-associated polyposis. We report a MUTYH variant, p.C306W (c.918C>G), with a tryptophan residue in place of native cysteine, that ligates the [4Fe4S] cluster in a patient with colonic polyposis and family history of early age colon cancer. In bacterial MutY, the [4Fe4S] cluster is redox active, allowing rapid localization to target lesions by long-range, DNA-mediated signalling. In the current study, using DNA electrochemistry, we determine that wild-type MUTYH is similarly redox-active, but MUTYH C306W undergoes rapid oxidative degradation of its cluster to [3Fe4S]+, with loss of redox signalling. In MUTYH C306W, oxidative cluster degradation leads to decreased DNA binding and enzyme function. This study confirms redox activity in eukaryotic DNA repair proteins and establishes MUTYH C306W as a pathogenic variant, highlighting the essential role of redox signalling by the [4Fe4S] cluster.


Asunto(s)
Poliposis Adenomatosa del Colon/metabolismo , Neoplasias del Colon/metabolismo , ADN Glicosilasas/metabolismo , Proteínas Hierro-Azufre/metabolismo , ADN Glicosilasas/genética , Variación Genética/genética , Humanos , Mutación , Oxidación-Reducción
15.
Nat Commun ; 8: 15102, 2017 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-28440315

RESUMEN

TET2 is a dioxygenase that catalyses multiple steps of 5-methylcytosine oxidation. Although TET2 mutations frequently occur in various types of haematological malignancies, the mechanism by which they increase risk for these cancers remains poorly understood. Here we show that Tet2-/- mice develop spontaneous myeloid, T- and B-cell malignancies after long latencies. Exome sequencing of Tet2-/- tumours reveals accumulation of numerous mutations, including Apc, Nf1, Flt3, Cbl, Notch1 and Mll2, which are recurrently deleted/mutated in human haematological malignancies. Single-cell-targeted sequencing of wild-type and premalignant Tet2-/- Lin-c-Kit+ cells shows higher mutation frequencies in Tet2-/- cells. We further show that the increased mutational burden is particularly high at genomic sites that gained 5-hydroxymethylcytosine, where TET2 normally binds. Furthermore, TET2-mutated myeloid malignancy patients have significantly more mutational events than patients with wild-type TET2. Thus, Tet2 loss leads to hypermutagenicity in haematopoietic stem/progenitor cells, suggesting a novel TET2 loss-mediated mechanism of haematological malignancy pathogenesis.


Asunto(s)
5-Metilcitosina/análogos & derivados , Proteínas de Unión al ADN/genética , Neoplasias Hematológicas/genética , Células Madre Hematopoyéticas/metabolismo , Tasa de Mutación , Proteínas Proto-Oncogénicas/genética , 5-Metilcitosina/metabolismo , 5-Metilcitosina/toxicidad , Anciano , Animales , Línea Celular Tumoral , Desmetilación del ADN , Análisis Mutacional de ADN/métodos , Proteínas de Unión al ADN/metabolismo , Dioxigenasas , Epigénesis Genética/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias Hematológicas/mortalidad , Neoplasias Hematológicas/patología , Humanos , Ratones , Ratones Noqueados , Mutagénesis/genética , Proteínas Proto-Oncogénicas/metabolismo , ARN Interferente Pequeño/metabolismo , Análisis de la Célula Individual/métodos , Secuenciación del Exoma/métodos
16.
DNA Repair (Amst) ; 38: 68-74, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26719139

RESUMEN

DNA mismatch repair (MMR) protects genome integrity by correcting DNA replication-associated mispairs, modulating DNA damage-induced cell cycle checkpoints and regulating homeologous recombination. Loss of MMR function leads to cancer development. This review describes progress in understanding how MMR is carried out in the context of chromatin and how chromatin organization/compaction, epigenetic mechanisms and posttranslational modifications of MMR proteins influence and regulate MMR in eukaryotic cells.


Asunto(s)
Reparación de la Incompatibilidad de ADN , Células Eucariotas/metabolismo , Código de Histonas , Procesamiento Proteico-Postraduccional , Animales , Ensamble y Desensamble de Cromatina , Humanos , Modelos Biológicos
18.
Cell Res ; 22(6): 973-85, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22290424

RESUMEN

MicroRNAs (miRNAs) are critical post-transcriptional regulators and are derived from hairpin-shaped primary transcripts via a series of processing steps. However, how the production of individual miRNAs is regulated remains largely unknown. Similarly, loss or overexpression of the key mismatch repair protein MutLα (MLH1-PMS2 heterodimer) leads to genome instability and tumorigenesis, but the mechanisms controlling MutLα expression are unknown. Here we demonstrate in vitro and in vivo that MLH1 and miR-422a participate in a feedback loop that regulates the level of both molecules. Using a defined in-vitro miRNA processing system, we show that MutLα stimulates the conversion of pri-miR-422a to pre-miR-422a, as well as the processing of other miRNAs tested, implicating MutLα as a general stimulating factor for miRNA biogenesis. This newly identified MutLα function requires its ATPase and pri-miRNA binding activities. In contrast, miR-422a downregulates MutLα levels by suppressing MLH1 expression through base pairing with the MLH1 3'-untranslated region. A model depicting this feedback mechanism is discussed.


Asunto(s)
Reparación de la Incompatibilidad de ADN , Enzimas Reparadoras del ADN/metabolismo , MicroARNs/metabolismo , Regiones no Traducidas 3' , Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/metabolismo , Enzimas Reparadoras del ADN/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Dimerización , Células HCT116 , Células HEK293 , Células HeLa , Humanos , Endonucleasa PMS2 de Reparación del Emparejamiento Incorrecto , Homólogo 1 de la Proteína MutL , Proteínas MutL , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA