Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Am J Hum Genet ; 106(2): 202-214, 2020 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-32004449

RESUMEN

Cell-free DNA (cf.DNA) is a powerful noninvasive biomarker for cancer and prenatal testing, and it circulates in plasma as short fragments. To elucidate the biology of cf.DNA fragmentation, we explored the roles of deoxyribonuclease 1 (DNASE1), deoxyribonuclease 1 like 3 (DNASE1L3), and DNA fragmentation factor subunit beta (DFFB) with mice deficient in each of these nucleases. By analyzing the ends of cf.DNA fragments in each type of nuclease-deficient mice with those in wild-type mice, we show that each nuclease has a specific cutting preference that reveals the stepwise process of cf.DNA fragmentation. Essentially, we demonstrate that cf.DNA is generated first intracellularly with DFFB, intracellular DNASE1L3, and other nucleases. Then, cf.DNA fragmentation continues extracellularly with circulating DNASE1L3 and DNASE1. With the use of heparin to disrupt the nucleosomal structure, we also show that the 10 bp periodicity originates from the cutting of DNA within an intact nucleosomal structure. Altogether, this work establishes a model of cf.DNA fragmentation.


Asunto(s)
Ácidos Nucleicos Libres de Células/metabolismo , Cromatina/metabolismo , Fragmentación del ADN , Desoxirribonucleasa I/fisiología , Desoxirribonucleasas/fisiología , Endodesoxirribonucleasas/fisiología , Nucleosomas/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa/fisiología , Animales , Ácidos Nucleicos Libres de Células/genética , Cromatina/genética , Femenino , Masculino , Ratones , Ratones Noqueados , Nucleosomas/genética
2.
Mol Cell ; 55(6): 829-842, 2014 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-25201414

RESUMEN

Breakpoint junctions of the chromosomal translocations that occur in human cancers display hallmarks of nonhomologous end-joining (NHEJ). In mouse cells, translocations are suppressed by canonical NHEJ (c-NHEJ) components, which include DNA ligase IV (LIG4), and instead arise from alternative NHEJ (alt-NHEJ). Here we used designer nucleases (ZFNs, TALENs, and CRISPR/Cas9) to introduce DSBs on two chromosomes to study translocation joining mechanisms in human cells. Remarkably, translocations were altered in cells deficient for LIG4 or its interacting protein XRCC4. Translocation junctions had significantly longer deletions and more microhomology, indicative of alt-NHEJ. Thus, unlike mouse cells, translocations in human cells are generated by c-NHEJ. Human cancer translocations induced by paired Cas9 nicks also showed a dependence on c-NHEJ, despite having distinct joining characteristics. These results demonstrate an unexpected and striking species-specific difference for common genomic rearrangements associated with tumorigenesis.


Asunto(s)
Reparación del ADN por Unión de Extremidades , ADN Ligasas/genética , Proteínas de Unión al ADN/genética , Desoxirribonucleasas/fisiología , Translocación Genética/genética , Animales , Cromosomas Humanos , ADN Ligasa (ATP) , Humanos , Ratones , Eliminación de Secuencia , Especificidad de la Especie , Células Tumorales Cultivadas
3.
PLoS One ; 9(4): e92593, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24705134

RESUMEN

Many viruses target cytoplasmic polyA binding protein (PABPC) to effect widespread inhibition of host gene expression, a process termed viral host-shutoff (vhs). During lytic replication of Epstein Barr Virus (EBV) we observed that PABPC was efficiently translocated from the cytoplasm to the nucleus. Translocated PABPC was diffusely distributed but was excluded from viral replication compartments. Vhs during EBV infection is regulated by the viral alkaline nuclease, BGLF5. Transfection of BGLF5 alone into BGLF5-KO cells or uninfected 293 cells promoted translocation of PAPBC that was distributed in clumps in the nucleus. ZEBRA, a viral bZIP protein, performs essential functions in the lytic program of EBV, including activation or repression of downstream viral genes. ZEBRA is also an essential replication protein that binds to viral oriLyt and interacts with other viral replication proteins. We report that ZEBRA also functions as a regulator of vhs. ZEBRA translocated PABPC to the nucleus, controlled the intranuclear distribution of PABPC, and caused global shutoff of host gene expression. Transfection of ZEBRA alone into 293 cells caused nuclear translocation of PABPC in the majority of cells in which ZEBRA was expressed. Co-transfection of ZEBRA with BGLF5 into BGLF5-KO cells or uninfected 293 cells rescued the diffuse intranuclear pattern of PABPC seen during lytic replication. ZEBRA mutants defective for DNA-binding were capable of regulating the intranuclear distribution of PABPC, and caused PABPC to co-localize with ZEBRA. One ZEBRA mutant, Z(S186E), was deficient in translocation yet was capable of altering the intranuclear distribution of PABPC. Therefore ZEBRA-mediated nuclear translocation of PABPC and regulation of intranuclear PABPC distribution are distinct events. Using a click chemistry-based assay for new protein synthesis, we show that ZEBRA and BGLF5 each function as viral host shutoff factors.


Asunto(s)
Núcleo Celular/metabolismo , Desoxirribonucleasas/fisiología , Herpesvirus Humano 4 , Proteína I de Unión a Poli(A)/metabolismo , Transactivadores/fisiología , Proteínas Virales/fisiología , Transporte Activo de Núcleo Celular , Núcleo Celular/virología , Células Cultivadas , Infecciones por Virus de Epstein-Barr/virología , Células HEK293 , Herpesvirus Humano 4/patogenicidad , Herpesvirus Humano 4/fisiología , Interacciones Huésped-Patógeno/genética , Humanos , Distribución Tisular , Replicación Viral/genética
4.
Mol Ther ; 21(10): 1889-97, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23883864

RESUMEN

Chronic hepatitis B virus (HBV) infection remains an important global health problem. Stability of the episomal covalently closed circular HBV DNA (cccDNA) is largely responsible for the modest curative efficacy of available therapy. Since licensed anti-HBV drugs have a post-transcriptional mechanism of action, disabling cccDNA is potentially of therapeutic benefit. To develop this approach, we engineered mutagenic transcription activator-like effector nucleases (TALENs) that target four HBV-specific sites within the viral genome. TALENs with cognate sequences in the S or C open-reading frames (ORFs) efficiently disrupted sequences at the intended sites and suppressed markers of viral replication. Following triple transfection of cultured HepG2.2.15 cells under mildly hypothermic conditions, the S TALEN caused targeted mutation in ~35% of cccDNA molecules. Markers of viral replication were also inhibited in vivo in a murine hydrodynamic injection model of HBV replication. HBV target sites within S and C ORFs of the injected HBV DNA were mutated without evidence of toxicity. These findings are the first to demonstrate a targeted nuclease-mediated disruption of HBV cccDNA. Efficacy in vivo also indicates that these engineered nucleases have potential for use in treatment of chronic HBV infection.


Asunto(s)
ADN Circular/genética , ADN Viral/genética , Desoxirribonucleasas/genética , Desoxirribonucleasas/fisiología , Virus de la Hepatitis B/genética , Virus de la Hepatitis B/fisiología , Replicación Viral , Animales , Secuencia de Bases , Línea Celular , Replicación del ADN , Modelos Animales de Enfermedad , Terapia Genética , Vectores Genéticos , Células Hep G2 , Hepatitis B/patología , Hepatitis B/terapia , Humanos , Ratones , Datos de Secuencia Molecular , Mutagénesis , Ingeniería de Proteínas , Transfección
5.
Gastroenterology ; 142(7): 1483-92.e6, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22387394

RESUMEN

BACKGROUND & AIMS: Polymorphisms that reduce the function of nucleotide-binding oligomerization domain (NOD)2, a bacterial sensor, have been associated with Crohn's disease (CD). No proteins that regulate NOD2 activity have been identified as selective pharmacologic targets. We sought to discover regulators of NOD2 that might be pharmacologic targets for CD therapies. METHODS: Carbamoyl phosphate synthetase/aspartate transcarbamylase/dihydroorotase (CAD) is an enzyme required for de novo pyrimidine nucleotide synthesis; it was identified as a NOD2-interacting protein by immunoprecipitation-coupled mass spectrometry. CAD expression was assessed in colon tissues from individuals with and without inflammatory bowel disease by immunohistochemistry. The interaction between CAD and NOD2 was assessed in human HCT116 intestinal epithelial cells by immunoprecipitation, immunoblot, reporter gene, and gentamicin protection assays. We also analyzed human cell lines that express variants of NOD2 and the effects of RNA interference, overexpression and CAD inhibitors. RESULTS: CAD was identified as a NOD2-interacting protein expressed at increased levels in the intestinal epithelium of patients with CD compared with controls. Overexpression of CAD inhibited NOD2-dependent activation of nuclear factor κB and p38 mitogen-activated protein kinase, as well as intracellular killing of Salmonella. Reduction of CAD expression or administration of CAD inhibitors increased NOD2-dependent signaling and antibacterial functions of NOD2 variants that are and are not associated with CD. CONCLUSIONS: The nucleotide synthesis enzyme CAD is a negative regulator of NOD2. The antibacterial function of NOD2 variants that have been associated with CD increased in response to pharmacologic inhibition of CAD. CAD is a potential therapeutic target for CD.


Asunto(s)
Aspartato Carbamoiltransferasa/fisiología , Carbamoil-Fosfato Sintasa (Glutamina-Hidrolizante)/fisiología , Enfermedad de Crohn/inmunología , Desoxirribonucleasas/fisiología , Dihidroorotasa/fisiología , Mucosa Intestinal/microbiología , Proteína Adaptadora de Señalización NOD2/inmunología , Aspartato Carbamoiltransferasa/antagonistas & inhibidores , Aspartato Carbamoiltransferasa/uso terapéutico , Carbamoil-Fosfato Sintasa (Glutamina-Hidrolizante)/antagonistas & inhibidores , Carbamoil-Fosfato Sintasa (Glutamina-Hidrolizante)/uso terapéutico , Línea Celular , Células Cultivadas , Enfermedad de Crohn/tratamiento farmacológico , Enfermedad de Crohn/microbiología , Dihidroorotasa/antagonistas & inhibidores , Dihidroorotasa/uso terapéutico , Inhibidores Enzimáticos/farmacología , Humanos , Inmunohistoquímica , Inmunoprecipitación , Mucosa Intestinal/inmunología , Espectrometría de Masas , FN-kappa B/fisiología , Proteína Adaptadora de Señalización NOD2/fisiología , Salmonella/crecimiento & desarrollo , Salmonella/inmunología , Transducción de Señal
6.
Chromosoma ; 121(1): 21-36, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22057367

RESUMEN

Chromosomal aberrations are often associated with incomplete genome duplication, for instance at common fragile sites, or as a consequence of chemical alterations in the DNA template that block replication forks. Studies of the cancer-prone disease Fanconi anaemia (FA) have provided important insights into the resolution of replication problems. The repair of interstrand DNA crosslinks induced by chemotherapy drugs is coupled with DNA replication and controlled by FA proteins. We discuss here the recent discovery of new FA-associated proteins and the development of new tractable repair systems that have dramatically improved our understanding of crosslink repair. We focus also on how FA proteins protect against replication failure in the context of fragile sites and on the identification of reactive metabolites that account for the development of Fanconi anaemia symptoms.


Asunto(s)
Replicación del ADN/genética , Proteínas del Grupo de Complementación de la Anemia de Fanconi/fisiología , Animales , Daño del ADN/genética , Daño del ADN/fisiología , Reparación del ADN/genética , Reparación del ADN/fisiología , Replicación del ADN/fisiología , Desoxirribonucleasas/metabolismo , Desoxirribonucleasas/fisiología , Anemia de Fanconi/genética , Anemia de Fanconi/metabolismo , Proteínas del Grupo de Complementación de la Anemia de Fanconi/genética , Proteínas del Grupo de Complementación de la Anemia de Fanconi/metabolismo , Humanos , Modelos Biológicos , Transducción de Señal/genética
7.
J Immunol ; 186(3): 1694-702, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21191071

RESUMEN

Viruses use a wide range of strategies to modulate the host immune response. The human gammaherpesvirus EBV, causative agent of infectious mononucleosis and several malignant tumors, encodes proteins that subvert immune responses, notably those mediated by T cells. Less is known about EBV interference with innate immunity, more specifically at the level of TLR-mediated pathogen recognition. The viral dsDNA sensor TLR9 is expressed on B cells, a natural target of EBV infection. Here, we show that EBV particles trigger innate immune signaling pathways through TLR9. Furthermore, using an in vitro system for productive EBV infection, it has now been possible to compare the expression of TLRs by EBV(-) and EBV(+) human B cells during the latent and lytic phases of infection. Several TLRs were found to be differentially expressed either in latently EBV-infected cells or after induction of the lytic cycle. In particular, TLR9 expression was profoundly decreased at both the RNA and protein levels during productive EBV infection. We identified the EBV lytic-phase protein BGLF5 as a protein that contributes to downregulating TLR9 levels through RNA degradation. Reducing the levels of a pattern-recognition receptor capable of sensing the presence of EBV provides a mechanism by which the virus could obstruct host innate antiviral responses.


Asunto(s)
Desoxirribonucleasas/fisiología , Regulación hacia Abajo/inmunología , Infecciones por Virus de Epstein-Barr/inmunología , Infecciones por Virus de Epstein-Barr/virología , Herpesvirus Humano 4/inmunología , Receptor Toll-Like 9/antagonistas & inhibidores , Receptor Toll-Like 9/biosíntesis , Proteínas Virales/fisiología , Latencia del Virus/inmunología , Subgrupos de Linfocitos B/inmunología , Subgrupos de Linfocitos B/patología , Subgrupos de Linfocitos B/virología , Linfoma de Burkitt/inmunología , Linfoma de Burkitt/patología , Linfoma de Burkitt/virología , Línea Celular Tumoral , Células Cultivadas , Regulación hacia Abajo/genética , Infecciones por Virus de Epstein-Barr/metabolismo , Regulación Viral de la Expresión Génica/inmunología , Células HEK293 , Herpesvirus Humano 4/patogenicidad , Humanos , ARN Viral/antagonistas & inhibidores , ARN Viral/metabolismo , Receptor Toll-Like 9/genética , Virión/inmunología , Activación Viral/inmunología
9.
J Virol ; 83(18): 9554-66, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19587049

RESUMEN

Lytic infection with the two human gammaherpesviruses, Kaposi's sarcoma-associated herpesvirus (KSHV) and Epstein-Barr virus (EBV), leads to significant depletion of the cellular transcriptome. This host shutoff phenotype is driven by the conserved herpesviral alkaline exonuclease, termed SOX in KSHV and BGLF5 in EBV, which in gammaherpesviruses has evolved the genetically separable ability to target cellular mRNA. We now show that host shutoff is also a prominent consequence of murine gammaherpesvirus 68 (MHV68) infection, which is widely used as a model system to study pathogenesis of these viruses in vivo. The effector of MHV68-induced host shutoff is its SOX homolog, here termed muSOX. There is remarkable functional conservation of muSOX host shutoff activities with those of KSHV SOX, including the recently described ability of SOX to induce mRNA hyperadenylation in the nucleus as well as cause nuclear relocalization of the poly(A) binding protein. SOX and muSOX localize to both the nucleus and cytoplasm of infected cells. Using spatially restricted variants of these proteins, we go on to demonstrate that all known host shutoff-related activities of SOX and muSOX are orchestrated exclusively from the cytoplasm. These results have important mechanistic implications for how SOX and muSOX target nascent cellular transcripts in the nucleus. Furthermore, our findings establish MHV68 as a new, genetically tractable model to study host shutoff.


Asunto(s)
Citoplasma/virología , Gammaherpesvirinae/patogenicidad , Infecciones por Herpesviridae/virología , Interacciones Huésped-Patógeno , ARN Mensajero/metabolismo , Animales , Desoxirribonucleasas/fisiología , Gammaherpesvirinae/enzimología , Herpesvirus Humano 4 , Herpesvirus Humano 8 , Humanos , Ratones , Rhadinovirus/patogenicidad , Infecciones Tumorales por Virus , Proteínas Virales/fisiología
10.
Chromosome Res ; 17(2): 265-75, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19308706

RESUMEN

Bacteria and archaea possess several different SMC-like proteins, which perform essential functions in a variety of chromosome dynamics, such as chromosome compaction, segregation, and DNA repair. SMC-like proteins localize to distinct sites within the cells at different time points in the cell cycle, or are recruited to sites of DNA breaks and damage. The bacterial SMC (MukB) complex appears to perform a condensin-like function, while SbcC and RecN act early during DNA repair, but apparently at different sites within the cells. Thus, bacterial SMC-like proteins have dynamic functions in chromosome segregation and maintenance of genetic stability.


Asunto(s)
Proteínas Bacterianas/fisiología , Proteínas de Ciclo Celular/fisiología , Reparación del ADN/fisiología , Complejos Multiproteicos/fisiología , Adenosina Trifosfato/fisiología , Proteínas Arqueales/química , Proteínas Arqueales/fisiología , Bacillus subtilis/citología , Bacillus subtilis/genética , Proteínas Bacterianas/química , Proteínas de Ciclo Celular/química , Proteínas Cromosómicas no Histona/química , Proteínas Cromosómicas no Histona/fisiología , Segregación Cromosómica/fisiología , Cromosomas Bacterianos/fisiología , Cromosomas Bacterianos/ultraestructura , Daño del ADN , Enzimas de Restricción del ADN/química , Enzimas de Restricción del ADN/fisiología , ADN Bacteriano/genética , Desoxirribonucleasas/química , Desoxirribonucleasas/fisiología , Escherichia coli/citología , Escherichia coli/genética , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/fisiología , Modelos Genéticos , Modelos Moleculares , Estructura Terciaria de Proteína , Relación Estructura-Actividad
11.
Int J Biochem Cell Biol ; 41(6): 1249-53, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19150506

RESUMEN

Defining the factors that lead to genomic instability is one of the most important fields in cancer biology. DNA damage can arise from exogenous sources or as a result of normal cellular metabolism. Regardless of the cause, when damaged DNA is not properly repaired the genome acquires mutation(s). Under normal circumstances, to prevent such chromosome instability the cell activates the checkpoint response, which inhibits cell cycle progression until DNA repair is complete. The Mre11 complex is formed by three components: Mre11, Rad50, and Nbs1/Xrs2 and is involved in the signaling pathways that lead to both checkpoint activation and DNA repair. In response to DNA damage two functions of the complex will be discussed, one involves its role in initiating kinase activation and the second involves its ability to tether and link DNA strands. This review will highlight the functions of the Mre11 complex during the process of DNA double strand break recognition and repair, and during the process of replication. Understanding how the Mre11 complex is working at the molecular level is important for understanding why disruptions in components of the complex lead to genomic instability and cancer predisposition syndromes in humans.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN , Replicación del ADN , Proteínas de Unión al ADN/fisiología , Desoxirribonucleasas/fisiología , Animales , Humanos , Proteína Homóloga de MRE11
12.
Cell Res ; 18(1): 125-33, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18087292

RESUMEN

Nonhomologous DNA end joining (NHEJ) is the primary pathway for repair of double-strand DNA breaks in human cells and in multicellular eukaryotes. The causes of double-strand breaks often fragment the DNA at the site of damage, resulting in the loss of information there. NHEJ does not restore the lost information and may resect additional nucleotides during the repair process. The ability to repair a wide range of overhang and damage configurations reflects the flexibility of the nuclease, polymerases, and ligase of NHEJ. The flexibility of the individual components also explains the large number of ways in which NHEJ can repair any given pair of DNA ends. The loss of information locally at sites of NHEJ repair may contribute to cancer and aging, but the action by NHEJ ensures that entire segments of chromosomes are not lost.


Asunto(s)
Adaptación Biológica/genética , Envejecimiento/genética , ADN Ligasas/fisiología , Reparación del ADN/genética , ADN Polimerasa Dirigida por ADN/fisiología , Desoxirribonucleasas/fisiología , Sistema Inmunológico/enzimología , Neoplasias/enzimología , Envejecimiento/fisiología , Animales , ADN Ligasa (ATP) , ADN Ligasas/genética , ADN Ligasas/metabolismo , Reparación del ADN/fisiología , Enzimas Reparadoras del ADN/metabolismo , Enzimas Reparadoras del ADN/fisiología , Proteína Quinasa Activada por ADN/metabolismo , Proteína Quinasa Activada por ADN/fisiología , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/fisiología , ADN Polimerasa Dirigida por ADN/genética , Desoxirribonucleasas/genética , Endonucleasas , Humanos , Sistema Inmunológico/metabolismo , Cambio de Clase de Inmunoglobulina/genética , Modelos Biológicos , Neoplasias/genética , Proteínas Nucleares/metabolismo , Proteínas Nucleares/fisiología , Recombinación Genética , Exones VDJ/genética , Vertebrados/genética
13.
J Invest Dermatol ; 127(1): 24-30, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16902420

RESUMEN

The removal of keratinocyte (KC) nuclear DNA by deoxyribonucleases (DNases) is an important step in the formation of normal stratum corneum (SC). However, the molecular identity of the DNA-degrading enzymes has so far remained elusive. Here we show that the endonuclease DNase1-like 2 (DNase1L2) is preferentially expressed in the epidermis and that its expression correlates with terminal differentiation of KC in vitro and in vivo. In biopsies of normal skin, DNase1L2 mRNA was regularly found in suprabasal KC and DNase1L2 protein was highly abundant in the stratum granulosum. In contrast to normal skin, DNase1L2 expression was downregulated in parakeratotic epidermis such as in psoriatic lesions. When DNase1L2 gene expression was knocked down by small interfering RNA in a human skin equivalent model, nuclei were maintained through all layers of the SC. Taken together, our data demonstrate that DNase1L2 plays an essential role in DNA degradation during terminal differentiation of epidermal KC.


Asunto(s)
ADN/metabolismo , Desoxirribonucleasas/fisiología , Células Epidérmicas , Queratinocitos/citología , Enfermedad de Bowen/metabolismo , Diferenciación Celular , Células Cultivadas , Desoxirribonucleasa I , Desoxirribonucleasas/genética , Endodesoxirribonucleasas , Regulación de la Expresión Génica , Humanos , Queratinocitos/metabolismo , Psoriasis/metabolismo , ARN Mensajero/análisis
14.
Br J Cancer ; 95(12): 1696-700, 2006 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-17146478

RESUMEN

Previously it was shown that horizontal DNA transfer between mammalian cells can occur through the uptake of apoptotic bodies, where genes from the apoptotic cells were transferred to neighbouring cells phagocytosing the apoptotic bodies. The regulation of this process is poorly understood. It was shown that the ability of cells as recipient of horizontally transferred DNA was enhanced by deficiency of p53 or p21. However, little is known with regard to the regulation of DNA from donor apoptotic cells. Here we report that the DNA fragmentation factor/caspase-activated DNase (DFF/CAD), which is the endonuclease responsible for DNA fragmentation during apoptosis, plays a significant role in regulation of horizontal DNA transfer. Cells with inhibited DFF/CAD function are poor donors for horizontal gene transfer (HGT) while their ability of being recipients of HGT is not affected.


Asunto(s)
Apoptosis/fisiología , Fragmentación del ADN , Transferencia de Gen Horizontal/genética , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas Reguladoras de la Apoptosis/farmacología , Caspasas/metabolismo , Células Cultivadas , Desoxirribonucleasas/antagonistas & inhibidores , Desoxirribonucleasas/genética , Desoxirribonucleasas/fisiología , Fibroblastos/metabolismo , Ratones , Ratones Noqueados/embriología , Fagocitosis , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/fisiología
15.
World J Gastroenterol ; 12(27): 4377-82, 2006 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-16865781

RESUMEN

AIM: To investigate loss of heterozygosity (LOH) and microsatellite instability (MSI) on the chromosomal region 1p36-pter in cholangiocarcinoma (CCA) patients and determine the association between microsatellite alterations and clinicopathological parameters. METHODS: Ten polymorphic microsatellite markers were determined for LOH and MSI using GS-3000 gel scan fragment autoanalyzer. RESULTS: Sixty-eight out of 90 cases (75.6%) showed LOH in one or more loci. LOH was found most frequently at D1S199 (40.0%), D1S507 (34.6%), D1S2845 (30.5%), and D1S2734 (30.1%). MSI was found in 34 of 90 cases (37.8%) at one or more loci. Fine mapping at 1p36 showed two distinctive regions of common loss, which were D1S2845 and the 25.5-cM region between D1S507 and D1S2734, indicating the existence of putative tumor suppressor genes that is likely to play important roles in the development of CCA. Patients with LOH at D1S234 showed less lymphatic invasion (P = 0.017), whereas patients with LOH at D1S2676 exhibited more lymphatic invasion than those without (P = 0.031). LOH at D1S2845 showed a significant correlation with nerve invasion (P = 0.029). Moreover, patients who demonstrated MSI at D1S228 showed a poor prognosis (P = 0.0026). CONCLUSION: Allelic loss plays a major role in microsatellite alterations at chromosome 1p36, which may contribute to carcinogenesis and pathogenesis of liver fluke related CCA and these alterations can be used as molecular prognostic indicators for CCA patients.


Asunto(s)
Neoplasias de los Conductos Biliares/genética , Conductos Biliares Intrahepáticos/patología , Colangiocarcinoma/genética , Cromosomas Humanos Par 1/genética , ADN de Neoplasias/genética , Repeticiones de Microsatélite/genética , Alelos , Animales , Neoplasias de los Conductos Biliares/diagnóstico , Neoplasias de los Conductos Biliares/etiología , Neoplasias de los Conductos Biliares/patología , Conductos Biliares Intrahepáticos/parasitología , Colangiocarcinoma/diagnóstico , Colangiocarcinoma/etiología , Colangiocarcinoma/patología , Inestabilidad Cromosómica/genética , Mapeo Cromosómico , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Desoxirribonucleasas/genética , Desoxirribonucleasas/fisiología , Progresión de la Enfermedad , Fasciola hepatica/patogenicidad , Fascioloidiasis/complicaciones , Fascioloidiasis/diagnóstico , Fascioloidiasis/patología , Genes Supresores de Tumor/fisiología , N-Metiltransferasa de Histona-Lisina , Humanos , Pérdida de Heterocigocidad/genética , Invasividad Neoplásica/genética , Proteínas Nucleares/genética , Proteínas Nucleares/fisiología , Proteínas de Unión a Poli-ADP-Ribosa , Pronóstico , Factores de Transcripción/genética , Factores de Transcripción/fisiología , Proteína de Unión al GTP cdc42/genética , Proteína de Unión al GTP cdc42/fisiología
16.
Proc Natl Acad Sci U S A ; 103(5): 1504-9, 2006 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-16432220

RESUMEN

DNA fragmentation is a hallmark of apoptosis (programmed cell death). However, the biological function of apoptotic DNA fragmentation remains unclear. Here, we show that DNA fragmentation factor plays an important role for maintaining genomic stability. Inhibition or loss of the DNA fragmentation factor (DFF)/caspase-activated DNase (CAD), whose nuclease activity is responsible for digesting genomic DNA during apoptosis, led to significant increases in spontaneous or induced gene mutations, gene amplifications, and chromosomal instability in primary mouse cells and transformed human cell lines. The mechanism underlying genetic instability in DFF/CAD-deficient cells, at least in part, involves a small but significant elevation in the survival of cells exposed to ionizing radiation, suggesting that apoptotic DNA fragmentation factor contributes to genomic stability by ensuring the removal of cells that have suffered DNA damage. In support of this hypothesis are the observations of increased cellular transformation of mouse embryonic cells from the DFF/CAD-null mice and significantly enhanced susceptibility to radiation-induced carcinogenesis in these mice. These data, in combination with published reports on the existence of tumor-specific gene mutations/deletions in the DFF/CAD genes in human cancer samples, suggest that apoptotic DNA fragmentation factor is required for the maintenance of genetic stability and may play a role in tumor suppression.


Asunto(s)
Cromosomas/efectos de la radiación , Fragmentación del ADN , Desoxirribonucleasas/fisiología , Genoma , Neoplasias Inducidas por Radiación/patología , Animales , Apoptosis , Proteínas Reguladoras de la Apoptosis , Línea Celular , Línea Celular Tumoral , Transformación Celular Neoplásica , Células Cultivadas , Cromosomas/ultraestructura , Relación Dosis-Respuesta en la Radiación , Fibroblastos/metabolismo , Regulación de la Expresión Génica , Humanos , Ratones , Ratones Transgénicos , Mutación , Neoplasias/metabolismo , Plásmidos/metabolismo , Factores de Tiempo , Transgenes
17.
Curr Dir Autoimmun ; 9: 162-72, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16394660

RESUMEN

Apoptotic cells are swiftly engulfed by macrophages and immature dendritic cells. Inefficient clearance of apoptotic cells has been implicated as a cause of inflammation and autoimmune diseases. Milk fat globule-EGF factor 8 (MFG-E8) and developmental endothelial locus-1 (Del-1) are glycoproteins secreted from macrophages that pass apoptotic cells to phagocytes. MFG-E8, but not Del-1, is expressed in the tingible-body macrophages at the germinal centers of the second lymphoid tissues. MFG-E8-deficient mice carry many unengulfed apoptotic cells in the germinal centers of the spleen, and develop a lupus-like autoimmune disease. In this review, we discuss the importance of the MFG-E8-mediated clearance of apoptotic cells in the prevention of autoimmune diseases.


Asunto(s)
Antígenos de Superficie/fisiología , Apoptosis , Enfermedades Autoinmunes/etiología , Fagocitosis , Animales , Proteínas de Unión al Calcio , Proteínas Portadoras/fisiología , Moléculas de Adhesión Celular , Desoxirribonucleasas/fisiología , Centro Germinal/fisiología , Humanos , Macrófagos/fisiología , Proteínas de la Leche
18.
J Biol Chem ; 280(42): 35670-83, 2005 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-16049016

RESUMEN

We have assessed the contribution of apoptosis-inducing factor (AIF) and inhibitor of caspase-activated DNase (ICAD) to the nuclear morphology and DNA degradation pattern in staurosporine-induced apoptosis. Expression of D117E ICAD, a mutant that is resistant to caspase cleavage at residue 117, prevented low molecular weight (LMW) DNA fragmentation, stage II nuclear morphology, and detection of terminal deoxynucleotidyl transferase staining. However, high molecular weight (HMW) DNA fragmentation and stage I nuclear morphology remained unaffected. On the other hand, expression of either D224E or wild type ICAD had no effect on DNA fragmentation or nuclear morphology. In addition, both HMW and LMW DNA degradation required functional executor caspases. Interestingly, silencing of endogenous AIF abolished type I nuclear morphology without any effect on HMW or LMW DNA fragmentation. Together, these results demonstrate that AIF is responsible for stage I nuclear morphology and suggest that HMW DNA degradation is a caspase-activated DNase and AIF-independent process.


Asunto(s)
Factor Inductor de la Apoptosis/fisiología , Núcleo Celular/metabolismo , ADN/química , Desoxirribonucleasas/antagonistas & inhibidores , Desoxirribonucleasas/fisiología , Apoptosis , Western Blotting , Línea Celular Tumoral , Fragmentación del ADN , Desoxirribonucleasas/química , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Humanos , Etiquetado Corte-Fin in Situ , Microscopía Electrónica , Microscopía Fluorescente , Peso Molecular , Mutagénesis , Fenotipo , Plásmidos/metabolismo , Estaurosporina/farmacología , Transfección
19.
Chromosoma ; 113(4): 157-66, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15309560

RESUMEN

The MRE11/RAD50/NBS1 complex (Mre11 complex) is a central player in most aspects of the cellular response to DNA double-strand breaks, including homologous recombination, non-homologous end joining, telomere maintenance and DNA damage checkpoint activation. Several of these findings are explained by the unusual enzymatic activities and macromolecular structure of the Mre11 complex. The Mre11 complex possesses an ATP-stimulated nuclease to process heterogeneous DNA ends and long coiled-coil tails to link DNA ends and/or sister chromatids. However, the mechanistic role of the Mre11 complex in checkpoint activation has been unclear until recently. New data suggest that the Mre11 complex can directly activate the ATM checkpoint kinase at DNA breaks. These findings, together with newly determined functional interactions, identify the Mre11 complex as an architectural and mechanistic keystone of cellular response events emerging from DNA breaks.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Rotura Cromosómica , Enzimas Reparadoras del ADN/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas Nucleares/metabolismo , Ácido Anhídrido Hidrolasas , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/fisiología , ADN Helicasas/fisiología , Enzimas Reparadoras del ADN/química , Enzimas Reparadoras del ADN/genética , Replicación del ADN , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Desoxirribonucleasas/fisiología , Humanos , Proteína Homóloga de MRE11 , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas Supresoras de Tumor/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA