Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Neuron ; 110(20): 3217-3219, 2022 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-36265437

RESUMEN

Abnormal activity of LINE-1 transposable elements has been associated with neurological disease. In this issue of Neuron, Takahashi et al. (2022) show that L1 hyperactivity occurs in the neurodegenerative syndrome ataxia telangiectasia and causes ataxia and cerebellar degeneration in mice.


Asunto(s)
Ataxia Telangiectasia , Enfermedades Neurodegenerativas , Animales , Ratones , Elementos Transponibles de ADN/genética , Ataxia Telangiectasia/genética , Neuronas , Enfermedades Neurodegenerativas/genética , Ataxia/genética
2.
Sci Adv ; 7(51): eabg6363, 2021 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-34910524

RESUMEN

The pathogenesis of inherited genome instability neurodegenerative syndromes remains largely unknown. Here, we report new disease-relevant murine models of genome instability­driven neurodegeneration involving disabled ATM and APTX that develop debilitating ataxia. We show that neurodegeneration and ataxia result from transcriptional interference in the cerebellum via aberrant messenger RNA splicing. Unexpectedly, these splicing defects were restricted to only Purkinje cells, disrupting the expression of critical homeostatic regulators including ITPR1, GRID2, and CA8. Abundant genotoxic R loops were also found at these Purkinje cell gene loci, further exacerbating DNA damage and transcriptional disruption. Using ATAC-seq to profile global chromatin accessibility in the cerebellum, we found a notably unique chromatin conformation specifically in Purkinje chromatin at the affected gene loci, thereby promoting susceptibility to DNA damage. These data reveal the pathogenic basis of DNA damage in the nervous system and suggest chromatin conformation as a feature in directing genome instability­associated neuropathology.

3.
Mol Cell ; 81(12): 2611-2624.e10, 2021 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-33857404

RESUMEN

The Shieldin complex shields double-strand DNA breaks (DSBs) from nucleolytic resection. Curiously, the penultimate Shieldin component, SHLD1, is one of the least abundant mammalian proteins. Here, we report that the transcription factors THAP1, YY1, and HCF1 bind directly to the SHLD1 promoter, where they cooperatively maintain the low basal expression of SHLD1, thereby ensuring a proper balance between end protection and resection during DSB repair. The loss of THAP1-dependent SHLD1 expression confers cross-resistance to poly (ADP-ribose) polymerase (PARP) inhibitor and cisplatin in BRCA1-deficient cells and shorter progression-free survival in ovarian cancer patients. Moreover, the embryonic lethality and PARPi sensitivity of BRCA1-deficient mice is rescued by ablation of SHLD1. Our study uncovers a transcriptional network that directly controls DSB repair choice and suggests a potential link between DNA damage and pathogenic THAP1 mutations, found in patients with the neurodevelopmental movement disorder adult-onset torsion dystonia type 6.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Animales , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Proteínas de Ciclo Celular/genética , ADN/metabolismo , Roturas del ADN de Doble Cadena/efectos de los fármacos , Reparación del ADN por Unión de Extremidades/efectos de los fármacos , Reparación del ADN/genética , Distonía/genética , Femenino , Factor C1 de la Célula Huésped/metabolismo , Proteínas Mad2/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Reparación del ADN por Recombinación/efectos de los fármacos , Proteínas de Unión a Telómeros/metabolismo , Proteína 1 de Unión al Supresor Tumoral P53/metabolismo , Factor de Transcripción YY1/metabolismo
4.
Mol Cell ; 77(1): 26-38.e7, 2020 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-31653568

RESUMEN

53BP1 activity drives genome instability and lethality in BRCA1-deficient mice by inhibiting homologous recombination (HR). The anti-recombinogenic functions of 53BP1 require phosphorylation-dependent interactions with PTIP and RIF1/shieldin effector complexes. While RIF1/shieldin blocks 5'-3' nucleolytic processing of DNA ends, it remains unclear how PTIP antagonizes HR. Here, we show that mutation of the PTIP interaction site in 53BP1 (S25A) allows sufficient DNA2-dependent end resection to rescue the lethality of BRCA1Δ11 mice, despite increasing RIF1 "end-blocking" at DNA damage sites. However, double-mutant cells fail to complete HR, as excessive shieldin activity also inhibits RNF168-mediated loading of PALB2/RAD51. As a result, BRCA1Δ1153BP1S25A mice exhibit hallmark features of HR insufficiency, including premature aging and hypersensitivity to PARPi. Disruption of shieldin or forced targeting of PALB2 to ssDNA in BRCA1D1153BP1S25A cells restores RNF168 recruitment, RAD51 nucleofilament formation, and PARPi resistance. Our study therefore reveals a critical function of shieldin post-resection that limits the loading of RAD51.


Asunto(s)
Recombinación Homóloga/genética , Proteína 1 de Unión al Supresor Tumoral P53/genética , Envejecimiento/efectos de los fármacos , Envejecimiento/genética , Animales , Proteína BRCA1/genética , Roturas del ADN de Doble Cadena/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Daño del ADN/genética , Inestabilidad Genómica/efectos de los fármacos , Inestabilidad Genómica/genética , Recombinación Homóloga/efectos de los fármacos , Ratones , Mutación/efectos de los fármacos , Mutación/genética , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Recombinasa Rad51/genética , Ubiquitina-Proteína Ligasas/genética
5.
Proc Natl Acad Sci U S A ; 115(52): E12285-E12294, 2018 12 26.
Artículo en Inglés | MEDLINE | ID: mdl-30538199

RESUMEN

Frequent oxidative modification of the neural genome is a by-product of the high oxygen consumption of the nervous system. Rapid correction of oxidative DNA lesions is essential, as genome stability is a paramount determinant of neural homeostasis. Apurinic/apyrimidinic endonuclease 1 (APE1; also known as "APEX1" or "REF1") is a key enzyme for the repair of oxidative DNA damage, although the specific role(s) for this enzyme in the development and maintenance of the nervous system is largely unknown. Here, using conditional inactivation of murine Ape1, we identify critical roles for this protein in the brain selectively after birth, coinciding with tissue oxygenation shifting from a placental supply to respiration. While mice lacking APE1 throughout neurogenesis were viable with little discernible phenotype at birth, rapid and pronounced brain-wide degenerative changes associated with DNA damage were observed immediately after birth leading to early death. Unexpectedly, Ape1Nes-cre mice appeared hypothermic with persistent shivering associated with the loss of thermoregulatory serotonergic neurons. We found that APE1 is critical for the selective regulation of Fos1-induced hippocampal immediate early gene expression. Finally, loss of APE1 in combination with p53 inactivation resulted in a profound susceptibility to brain tumors, including medulloblastoma and glioblastoma, implicating oxidative DNA lesions as an etiologic agent in these diseases. Our study reveals APE1 as a major suppressor of deleterious oxidative DNA damage and uncovers specific and broad pathogenic consequences of respiratory oxygenation in the postnatal nervous system.


Asunto(s)
Regulación de la Temperatura Corporal , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/fisiopatología , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Homeostasis , Animales , Daño del ADN , ADN-(Sitio Apurínico o Apirimidínico) Liasa/genética , Femenino , Genoma , Hipocampo/metabolismo , Humanos , Masculino , Ratones , Ratones Noqueados , Neurogénesis , Estrés Oxidativo , Neuronas Serotoninérgicas/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
6.
J Neurosci ; 37(4): 893-905, 2017 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-28123024

RESUMEN

The DNA damage response (DDR) orchestrates a network of cellular processes that integrates cell-cycle control and DNA repair or apoptosis, which serves to maintain genome stability. DNA-PKcs (the catalytic subunit of the DNA-dependent kinase, encoded by PRKDC), ATM (ataxia telangiectasia, mutated), and ATR (ATM and Rad3-related) are related PI3K-like protein kinases and central regulators of the DDR. Defects in these kinases have been linked to neurodegenerative or neurodevelopmental syndromes. In all cases, the key neuroprotective function of these kinases is uncertain. It also remains unclear how interactions between the three DNA damage-responsive kinases coordinate genome stability, particularly in a physiological context. Here, we used a genetic approach to identify the neural function of DNA-PKcs and the interplay between ATM and ATR during neurogenesis. We found that DNA-PKcs loss in the mouse sensitized neuronal progenitors to apoptosis after ionizing radiation because of excessive DNA damage. DNA-PKcs was also required to prevent endogenous DNA damage accumulation throughout the adult brain. In contrast, ATR coordinated the DDR during neurogenesis to direct apoptosis in cycling neural progenitors, whereas ATM regulated apoptosis in both proliferative and noncycling cells. We also found that ATR controls a DNA damage-induced G2/M checkpoint in cortical progenitors, independent of ATM and DNA-PKcs. These nonoverlapping roles were further confirmed via sustained murine embryonic or cortical development after all three kinases were simultaneously inactivated. Thus, our results illustrate how DNA-PKcs, ATM, and ATR have unique and essential roles during the DDR, collectively ensuring comprehensive genome maintenance in the nervous system. SIGNIFICANCE STATEMENT: The DNA damage response (DDR) is essential for prevention of a broad spectrum of different human neurologic diseases. However, a detailed understanding of the DDR at a physiological level is lacking. In contrast to many in vitro cellular studies, here we demonstrate independent biological roles for the DDR kinases DNA-PKcs, ATM, and ATR during neurogenesis. We show that DNA-PKcs is central to DNA repair in nonproliferating cells, and restricts DNA damage accumulation, whereas ATR controls damage-induced G2 checkpoint control and apoptosis in proliferating cells. Conversely, ATM is critical for controlling apoptosis in immature noncycling neural cells after DNA damage. These data demonstrate functionally distinct, but cooperative, roles for each kinase in preserving genome stability in the nervous system.


Asunto(s)
Daño del ADN/fisiología , Proteína Quinasa Activada por ADN/metabolismo , Proteínas de Unión al ADN/metabolismo , Neurogénesis/fisiología , Neuronas/metabolismo , Proteínas Nucleares/metabolismo , Animales , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Corteza Cerebral/citología , Corteza Cerebral/embriología , Corteza Cerebral/metabolismo , Proteína Quinasa Activada por ADN/genética , Proteínas de Unión al ADN/genética , Femenino , Genoma/fisiología , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Proteínas Nucleares/genética
7.
Mech Ageing Dev ; 161(Pt A): 121-129, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27125728

RESUMEN

A variety of human neurologic diseases are caused by inherited defects in DNA repair. In many cases, these syndromes almost exclusively impact the nervous system, underscoring the critical requirement for genome stability in this tissue. A striking example of this is defective enzymatic activity of polynucleotide kinase-phosphatase (PNKP), leading to microcephaly or neurodegeneration. Notably, the broad neural impact of mutations in PNKP can result in markedly different disease entities, even when the inherited mutation is the same. For example microcephaly with seizures (MCSZ) results from various hypomorphic PNKP mutations, as does ataxia with oculomotor apraxia 4 (AOA4). Thus, other contributing factors influence the neural phenotype when PNKP is disabled. Here we consider the role for PNKP in maintaining brain function and how perturbation in its activity can account for the varied pathology of neurodegeneration or microcephaly present in MCSZ and AOA4 respectively.


Asunto(s)
Enzimas Reparadoras del ADN/genética , Microcefalia/genética , Mutación , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Convulsiones/genética , Ataxias Espinocerebelosas/congénito , Animales , Enzimas Reparadoras del ADN/metabolismo , Humanos , Microcefalia/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Convulsiones/metabolismo , Ataxias Espinocerebelosas/genética , Ataxias Espinocerebelosas/metabolismo
8.
EMBO J ; 34(19): 2465-80, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26290337

RESUMEN

Polynucleotide kinase-phosphatase (PNKP) is a DNA repair factor possessing both 5'-kinase and 3'-phosphatase activities to modify ends of a DNA break prior to ligation. Recently, decreased PNKP levels were identified as the cause of severe neuropathology present in the human microcephaly with seizures (MCSZ) syndrome. Utilizing novel murine Pnkp alleles that attenuate expression and a T424GfsX48 frame-shift allele identified in MCSZ individuals, we determined how PNKP inactivation impacts neurogenesis. Mice with PNKP inactivation in neural progenitors manifest neurodevelopmental abnormalities and postnatal death. This severe phenotype involved defective base excision repair and non-homologous end-joining, pathways required for repair of both DNA single- and double-strand breaks. Although mice homozygous for the T424GfsX48 allele were lethal embryonically, attenuated PNKP levels (akin to MCSZ) showed general neurodevelopmental defects, including microcephaly, indicating a critical developmental PNKP threshold. Directed postnatal neural inactivation of PNKP affected specific subpopulations including oligodendrocytes, indicating a broad requirement for genome maintenance, both during and after neurogenesis. These data illuminate the basis for selective neural vulnerability in DNA repair deficiency disease.


Asunto(s)
Reparación del ADN , Mutación del Sistema de Lectura , Inestabilidad Genómica , Células-Madre Neurales/enzimología , Oligodendroglía/enzimología , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Animales , Humanos , Ratones , Ratones Mutantes , Microcefalia/enzimología , Microcefalia/genética , Microcefalia/patología , Células-Madre Neurales/patología , Oligodendroglía/patología , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética
9.
Nature ; 501(7468): 569-72, 2013 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-24013173

RESUMEN

Replication fork maintenance pathways preserve chromosomes, but their faulty application at nonallelic repeats could generate rearrangements causing cancer, genomic disorders and speciation. Potential causal mechanisms are homologous recombination and error-free postreplication repair (EF-PRR). Homologous recombination repairs damage-induced DNA double-strand breaks (DSBs) and single-ended DSBs within replication. To facilitate homologous recombination, the recombinase RAD51 and mediator BRCA2 form a filament on the 3' DNA strand at a break to enable annealing to the complementary sister chromatid while the RecQ helicase, BLM (Bloom syndrome mutated) suppresses crossing over to prevent recombination. Homologous recombination also stabilizes and restarts replication forks without a DSB. EF-PRR bypasses DNA incongruities that impede replication by ubiquitinating PCNA (proliferating cell nuclear antigen) using the RAD6-RAD18 and UBC13-MMS2-RAD5 ubiquitin ligase complexes. Some components are common to both homologous recombination and EF-PRR such as RAD51 and RAD18. Here we delineate two pathways that spontaneously fuse inverted repeats to generate unstable chromosomal rearrangements in wild-type mouse embryonic stem (ES) cells. Gamma-radiation induced a BLM-regulated pathway that selectively fused identical, but not mismatched, repeats. By contrast, ultraviolet light induced a RAD18-dependent pathway that efficiently fused mismatched repeats. Furthermore, TREX2 (a 3'→5' exonuclease) suppressed identical repeat fusion but enhanced mismatched repeat fusion, clearly separating these pathways. TREX2 associated with UBC13 and enhanced PCNA ubiquitination in response to ultraviolet light, consistent with it being a novel member of EF-PRR. RAD18 and TREX2 also suppressed replication fork stalling in response to nucleotide depletion. Interestingly, replication fork stalling induced fusion for identical and mismatched repeats, implicating faulty replication as a causal mechanism for both pathways.


Asunto(s)
Inestabilidad Cromosómica/genética , Cromosomas de los Mamíferos/genética , Reparación del ADN/genética , Replicación del ADN/genética , Recombinación Homóloga/genética , Secuencias Invertidas Repetidas/genética , Animales , Secuencia de Bases , Rotura Cromosómica , Roturas del ADN de Doble Cadena , Proteínas de Unión al ADN/metabolismo , Células Madre Embrionarias/metabolismo , Exodesoxirribonucleasas/metabolismo , Hidroxiurea/farmacología , Ratones , Nucleótidos/deficiencia , Nucleótidos/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Recombinasa Rad51/metabolismo , RecQ Helicasas/metabolismo , Enzimas Ubiquitina-Conjugadoras/metabolismo , Ubiquitinación/efectos de la radiación , Rayos Ultravioleta
10.
Genetics ; 188(4): 787-97, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21546543

RESUMEN

Trex2 is a 3' → 5' exonuclease that removes 3'-mismatched sequences in a biochemical assay; however, its biological function remains unclear. To address biology we previously generated trex2(null) mouse embryonic stem (ES) cells and expressed in these cells wild-type human TREX2 cDNA (Trex2(hTX2)) or cDNA with a single-amino-acid change in the catalytic domain (Trex2(H188A)) or in the DNA-binding domain (Trex2(R167A)). We found the trex2(null) and Trex2(H188A) cells exhibited spontaneous broken chromosomes and trex2(null) cells exhibited spontaneous chromosomal rearrangements. We also found ectopically expressed human TREX2 was active at the 3' ends of I-SceI-induced chromosomal double-strand breaks (DSBs). Therefore, we hypothesized Trex2 participates in DNA DSB repair by modifying 3' ends. This may be especially important for ends with damaged nucleotides. Here we present data that are unexpected and prompt a new model. We found Trex2-altered cells (null, H188A, and R167A) were not hypersensitive to camptothecin, a type-1 topoisomerase inhibitor that induces DSBs at replication forks. In addition, Trex2-altered cells were not hypersensitive to γ-radiation, an agent that causes DSBs throughout the cell cycle. This observation held true even in cells compromised for one of the two major DSB repair pathways: homology-directed repair (HDR) or nonhomologous end joining (NHEJ). Trex2 deletion also enhanced repair of an I-SceI-induced DSB by both HDR and NHEJ without affecting pathway choice. Interestingly, however, trex2(null) cells exhibited reduced spontaneous sister chromatid exchanges (SCEs) but this was not due to a defect in HDR-mediated crossing over. Therefore, reduced spontaneous SCE could be a manifestation of the same defect that caused spontaneous broken chromosomes and spontaneous chromosomal rearrangements. These unexpected data suggest Trex2 does not enable DSB repair and prompt a new model that posits Trex2 suppresses the formation of broken chromosomes.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN/genética , Exodesoxirribonucleasas/metabolismo , Fosfoproteínas/metabolismo , Intercambio de Cromátides Hermanas/genética , Animales , Camptotecina/farmacología , Línea Celular , Roturas del ADN de Doble Cadena/efectos de los fármacos , Roturas del ADN de Doble Cadena/efectos de la radiación , Desoxirribonucleasas de Localización Especificada Tipo II/metabolismo , Exodesoxirribonucleasas/genética , Rayos gamma/efectos adversos , Técnicas de Inactivación de Genes , Marcación de Gen , Humanos , Ratones , Ratones de la Cepa 129 , Fosfoproteínas/genética , RecQ Helicasas/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Inhibidores de Topoisomerasa I/farmacología
11.
Mutat Res ; 662(1-2): 84-7, 2009 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-19094998

RESUMEN

TREX2 is a 3'-->5' exonuclease that binds to DNA and removes 3' mismatched nucleotides. By an in vitro structure function analysis, we found a single amino acid change (H188A) completely ablates exonuclease activity and impairs DNA binding by about 60% while another change (R167A) impairs DNA binding by about 85% without impacting exonuclease activity. For a biological analysis, we generated trex2null cells by deleting the entire Trex2 coding sequences in mouse embryonic stem (ES) cells. We found Trex2 deletion caused high levels of Robertsonian translocations (RbTs) showing Trex2 is important for chromosomal maintenance. Here we evaluate the exonuclease and DNA binding domains by expressing in trex2(null) cells coding sequences for wild type human TREX2 (Trex2hTX2) or human TREX2 with the H188A change (Trex2H188A) or the R167A change (Trex2R167A). These cDNAs are positioned adjacent to the mouse Trex2 promoter by Cre-mediated knock-in. By observing metaphase spreads, we found Trex2H188A cells exhibited high levels of double-strand breaks (DSBs) and chromosomal fragments. Therefore, TREX2 may suppress spontaneous DSBs or exonuclease defective TREX2 may induce them in a dominate-negative manner. We also found Trex2hTX2, hTrex2H188A and hTrex2R167A cells did not exhibit RbTs. Thus, neither the exonuclease nor DNA binding domains suppress RbTs suggesting TREX2 possesses additional biochemical activities.


Asunto(s)
Aberraciones Cromosómicas , Roturas del ADN de Doble Cadena , Exodesoxirribonucleasas/deficiencia , Fosfoproteínas/deficiencia , Translocación Genética , Animales , ADN Complementario/genética , Exodesoxirribonucleasas/metabolismo , Técnicas de Sustitución del Gen , Humanos , Ratones , Fosfoproteínas/metabolismo
12.
Cancer Res ; 67(19): 9077-83, 2007 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-17909011

RESUMEN

Cisplatin, an anticancer drug, forms DNA interstrand cross-links (ICL) that interfere with replication, whereas TREX2 is a 3'-->5' exonuclease that removes 3' mismatched nucleotides and promotes cellular proliferation. Here, we show that TREX2 is depleted in human cells derived from cancer after exposure to cisplatin but not other genotoxins including another cross-linking agent, mitomycin C (MMC), indicating a potential role for TREX2 depletion in cisplatin-induced cytotoxicity. To better understand TREX2 cellular function, we deleted TREX2 in mouse embryonic stem (ES) cells by gene targeting and find these cells exhibit reduced proliferation and gross chromosomal rearrangements including Robertsonian translocations (RbT). Quite interestingly, ES cells exposed to cisplatin also exhibit RbTs. By contrast, RbTs are not observed for ES cells exposed to MMC, indicating that RbTs are not caused by ICLs but instead TREX2 depletion by either cisplatin exposure or mutation. Taken together, our results show that cisplatin depletes TREX2 and causes genomic instability that is similarly observed in TREX2-mutant cells. Thus, cisplatin has two potential cytotoxic activities: (a) the generation of ICLs and (b) the depletion of TREX2.


Asunto(s)
Antineoplásicos/farmacología , Cisplatino/farmacología , Daño del ADN , Exodesoxirribonucleasas/deficiencia , Fosfoproteínas/deficiencia , Translocación Genética/efectos de los fármacos , Procesos de Crecimiento Celular/efectos de los fármacos , Procesos de Crecimiento Celular/fisiología , Núcleo Celular/efectos de los fármacos , Núcleo Celular/enzimología , Relación Dosis-Respuesta a Droga , Exodesoxirribonucleasas/metabolismo , Células HeLa , Humanos , Mitomicina/farmacología , Fosfoproteínas/metabolismo
13.
Nucleic Acids Res ; 35(8): 2682-94, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17426129

RESUMEN

TREX2 is an autonomous nonprocessive 3' --> 5' exonuclease, suggesting that it maintains genome integrity. To investigate TREX2's biochemical and cellular properties, we show that endogenous TREX2 is expressed widely in mouse tissues and human cell lines. Unexpectedly, endogenous human TREX2 is predominantly expressed as a 30-kDa protein (not 26 kDa, as previously believed), which is likely encoded by longer isoforms (TREX2(L1) and/or TREX2(L2)) that possess similar capacity for self-association, DNA binding and catalytic activity. Site-directed mutagenesis analysis shows that the three functional activities of TREX2 are distinct, yet integrated. Mutation of amino acids putatively important for homodimerization significantly impairs both DNA binding and exonuclease activity, while mutation of amino acids (except R163) in the DNA binding and exonuclease domains affects their corresponding activities. Interestingly, however, DNA-binding domain mutations do not impact catalytic activity, while exonuclease domain mutations diminish DNA binding. To understand TREX2 cellular properties, we find endogenous TREX2 is down regulated during G2/M and nuclear TREX2 displays a punctate staining pattern. Furthermore, TREX2 knockdown reduces cell proliferation. Taken together, our results suggest that TREX2 plays an important function during DNA metabolism and cellular proliferation.


Asunto(s)
Exodesoxirribonucleasas/química , Exodesoxirribonucleasas/metabolismo , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Empalme Alternativo , Secuencia de Aminoácidos , Animales , Ciclo Celular , Línea Celular , Proliferación Celular , Exodesoxirribonucleasas/genética , Células HeLa , Humanos , Ratones , Datos de Secuencia Molecular , Fosfoproteínas/genética , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estructura Terciaria de Proteína , Relación Estructura-Actividad
14.
Genesis ; 45(5): 275-81, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17457929

RESUMEN

The HPRT minigene is a selection cassette used for gene targeting in mouse embryonic stem (ES) cells and, it is unique since selection may be applied for its presence and absence. This minigene has two exon clusters separated by a small intron and splicing sequences. We find these exon clusters splice into exons from the target gene forming two different classes of chimeric transcripts. The first class is expressed by the endogenous promoter and includes upstream target gene exons spliced into minigene exons 3-8. The second class is expressed by the minigene's PGK promoter and includes minigene exons 1-2 spliced into downstream target gene exons. These chimeric transcripts may produce chimeric proteins that could influence phenotype. Therefore, we have designed two floxed HPRT minigenes that permit removal of either the 5' half of the minigene or the entire minigene via Cre-mediated recombination.


Asunto(s)
Quimera/genética , Marcación de Gen/métodos , Hipoxantina Fosforribosiltransferasa/genética , Empalme Alternativo , Animales , Secuencia de Bases , Línea Celular , Cartilla de ADN/genética , Células Madre Embrionarias/metabolismo , Exones , Vectores Genéticos , Ratones , Mutagénesis Insercional , Regiones Promotoras Genéticas , Recombinación Genética , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...