Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 171
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 80(6): 996-1012.e9, 2020 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-33147438

RESUMEN

Reactive aldehydes arise as by-products of metabolism and are normally cleared by multiple families of enzymes. We find that mice lacking two aldehyde detoxifying enzymes, mitochondrial ALDH2 and cytoplasmic ADH5, have greatly shortened lifespans and develop leukemia. Hematopoiesis is disrupted profoundly, with a reduction of hematopoietic stem cells and common lymphoid progenitors causing a severely depleted acquired immune system. We show that formaldehyde is a common substrate of ALDH2 and ADH5 and establish methods to quantify elevated blood formaldehyde and formaldehyde-DNA adducts in tissues. Bone-marrow-derived progenitors actively engage DNA repair but also imprint a formaldehyde-driven mutation signature similar to aging-associated human cancer mutation signatures. Furthermore, we identify analogous genetic defects in children causing a previously uncharacterized inherited bone marrow failure and pre-leukemic syndrome. Endogenous formaldehyde clearance alone is therefore critical for hematopoiesis and in limiting mutagenesis in somatic tissues.


Asunto(s)
Alcohol Deshidrogenasa/genética , Aldehído Deshidrogenasa Mitocondrial/genética , Formaldehído/sangre , Leucemia/genética , Adolescente , Aldehídos/sangre , Animales , Niño , Preescolar , Aductos de ADN/genética , Daño del ADN/efectos de los fármacos , Reparación del ADN/efectos de los fármacos , Femenino , Formaldehído/toxicidad , Hematopoyesis/genética , Células Madre Hematopoyéticas/metabolismo , Humanos , Lactante , Leucemia/sangre , Leucemia/patología , Masculino , Ratones , Mutación/genética , Especificidad por Sustrato
2.
Mol Cell ; 71(1): 25-41.e6, 2018 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-29937342

RESUMEN

Components of the Fanconi anemia and homologous recombination pathways play a vital role in protecting newly replicated DNA from uncontrolled nucleolytic degradation, safeguarding genome stability. Here we report that histone methylation by the lysine methyltransferase SETD1A is crucial for protecting stalled replication forks from deleterious resection. Depletion of SETD1A sensitizes cells to replication stress and leads to uncontrolled DNA2-dependent resection of damaged replication forks. The ability of SETD1A to prevent degradation of these structures is mediated by its ability to catalyze methylation on Lys4 of histone H3 (H3K4) at replication forks, which enhances FANCD2-dependent histone chaperone activity. Suppressing H3K4 methylation or expression of a chaperone-defective FANCD2 mutant leads to loss of RAD51 nucleofilament stability and severe nucleolytic degradation of replication forks. Our work identifies epigenetic modification and histone mobility as critical regulatory mechanisms in maintaining genome stability by restraining nucleases from irreparably damaging stalled replication forks.


Asunto(s)
ADN/biosíntesis , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Chaperonas Moleculares/metabolismo , Nucleosomas/metabolismo , Células A549 , ADN/genética , Replicación del ADN/fisiología , Epigénesis Genética/fisiología , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/genética , Células HeLa , N-Metiltransferasa de Histona-Lisina/genética , Histonas/genética , Humanos , Metilación , Chaperonas Moleculares/genética , Nucleosomas/genética , Recombinasa Rad51/genética , Recombinasa Rad51/metabolismo
3.
Mol Cell ; 66(5): 622-634.e8, 2017 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-28575658

RESUMEN

RFWD3 is a recently identified Fanconi anemia protein FANCW whose E3 ligase activity toward RPA is essential in homologous recombination (HR) repair. However, how RPA ubiquitination promotes HR remained unknown. Here, we identified RAD51, the central HR protein, as another target of RFWD3. We show that RFWD3 polyubiquitinates both RPA and RAD51 in vitro and in vivo. Phosphorylation by ATR and ATM kinases is required for this activity in vivo. RFWD3 inhibits persistent mitomycin C (MMC)-induced RAD51 and RPA foci by promoting VCP/p97-mediated protein dynamics and subsequent degradation. Furthermore, MMC-induced chromatin loading of MCM8 and RAD54 is defective in cells with inactivated RFWD3 or expressing a ubiquitination-deficient mutant RAD51. Collectively, our data reveal a mechanism that facilitates timely removal of RPA and RAD51 from DNA damage sites, which is crucial for progression to the late-phase HR and suppression of the FA phenotype.


Asunto(s)
Cromatina/enzimología , Daño del ADN , ADN/metabolismo , Anemia de Fanconi/enzimología , Recombinasa Rad51/metabolismo , Reparación del ADN por Recombinación , Proteína de Replicación A/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/metabolismo , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Sitios de Unión , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Cromatina/efectos de los fármacos , Cromatina/genética , Cromatina/efectos de la radiación , ADN/genética , Anemia de Fanconi/genética , Humanos , Proteínas de Mantenimiento de Minicromosoma/metabolismo , Mitomicina/farmacología , Mutación , Fosforilación , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , Proteolisis , Interferencia de ARN , Recombinasa Rad51/genética , Reparación del ADN por Recombinación/efectos de los fármacos , Reparación del ADN por Recombinación/efectos de la radiación , Proteína de Replicación A/genética , Transfección , Ubiquitina-Proteína Ligasas/genética , Proteína que Contiene Valosina
4.
Genes Cells ; 28(9): 642-645, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37341149

RESUMEN

Ataxia-telangiectasia (A-T) is a rare devastating hereditary condition, which affects multiple organ systems including cerebellar motor function as well as DNA repair, resulting in a higher incidence of cancer and immunodeficiency. The genetic defect in A-T lies in ATM kinase, which is activated by DNA damage and regulates a plethora of substrates including the p53 tumor suppressor. We have organized an international meeting "The 19th Ataxia-Telangiectasia Workshop 2023 (ATW2023)" with support from the Molecular Biology Society of Japan (MBSJ) and other funders. Here, we report that ATW2023 was successfully held in Kyoto from March 2nd to 5th, 2023 with more than 150 participants traveling from all over the world, despite the still smoldering COVID-19 pandemic. In this meeting report, we will briefly describe the highlights of the meeting and would like to express our gratitude to the MBSJ for the financial support.


Asunto(s)
Ataxia Telangiectasia , COVID-19 , Humanos , Ataxia Telangiectasia/genética , Ataxia Telangiectasia/patología , Proteínas de la Ataxia Telangiectasia Mutada , Pandemias , Daño del ADN , Reparación del ADN , Proteínas de Ciclo Celular/metabolismo
5.
Genes Cells ; 28(9): 663-673, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37469008

RESUMEN

The SLFN11 gene participates in cell fate decision following cancer chemotherapy and encodes the N-terminal ribonuclease (RNase) domain and the C-terminal helicase/ATPase domain. How these domains contribute to the chemotherapeutic response remains controversial. Here, we expressed SLFN11 containing mutations in two critical residues required for RNase activity in SLFN11-/- cells. We found that this mutant was still able to suppress DNA damage tolerance, destabilized the stalled replication forks, and perturbed recruitment of the fork protector RAD51. In contrast, we confirmed that the helicase domain was essential to accelerate fork degradation. The fork degradation by the RNase mutant was dependent on both DNA2 and MRE11 nuclease, but not on MRE11's novel interactor FXR1. Collectively, these results supported the view that the RNase domain function is dispensable for SLFN11 to mediate cell fate decision during replication stress response.


Asunto(s)
Replicación del ADN , Ribonucleasas , Ribonucleasas/genética , ADN Helicasas/genética , ADN Helicasas/metabolismo , Mutación , Daño del ADN
6.
Blood ; 137(3): 336-348, 2021 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-32735670

RESUMEN

Fanconi anemia (FA) is a hereditary disorder caused by mutations in any 1 of 22 FA genes. The disease is characterized by hypersensitivity to interstrand crosslink (ICL) inducers such as mitomycin C (MMC). In addition to promoting ICL repair, FA proteins such as RAD51, BRCA2, or FANCD2 protect stalled replication forks from nucleolytic degradation during replication stress, which may have a profound impact on FA pathophysiology. Recent studies showed that expression of the putative DNA/RNA helicase SLFN11 in cancer cells correlates with cell death on chemotherapeutic treatment. However, the underlying mechanisms of SLFN11-mediated DNA damage sensitivity remain unclear. Because SLFN11 expression is high in hematopoietic stem cells, we hypothesized that SLFN11 depletion might ameliorate the phenotypes of FA cells. Here we report that SLFN11 knockdown in the FA patient-derived FANCD2-deficient PD20 cell line improved cell survival on treatment with ICL inducers. FANCD2-/-SLFN11-/- HAP1 cells also displayed phenotypic rescue, including reduced levels of MMC-induced chromosome breakage compared with FANCD2-/- cells. Importantly, we found that SLFN11 promotes extensive fork degradation in FANCD2-/- cells. The degradation process is mediated by the nucleases MRE11 or DNA2 and depends on the SLFN11 ATPase activity. This observation was accompanied by an increased RAD51 binding at stalled forks, consistent with the role of RAD51 antagonizing nuclease recruitment and subsequent fork degradation. Suppression of SLFN11 protects nascent DNA tracts even in wild-type cells. We conclude that SLFN11 destabilizes stalled replication forks, and this function may contribute to the attrition of hematopoietic stem cells in FA.


Asunto(s)
Replicación del ADN , Anemia de Fanconi/patología , Proteínas Nucleares/metabolismo , Puntos de Control del Ciclo Celular , Línea Celular , Rotura Cromosómica , Reactivos de Enlaces Cruzados/farmacología , ADN Helicasas/metabolismo , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/genética , Técnicas de Silenciamiento del Gen , Humanos , Proteína Homóloga de MRE11/metabolismo , Modelos Biológicos , Mutación/genética , Fenotipo , ARN Interferente Pequeño/metabolismo , Recombinasa Rad51/metabolismo
7.
Blood ; 137(15): 2021-2032, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33512438

RESUMEN

We have recently discovered Japanese children with a novel Fanconi anemia-like inherited bone marrow failure syndrome (IBMFS). This disorder is likely caused by the loss of a catabolic system directed toward endogenous formaldehyde due to biallelic variants in ADH5 combined with a heterozygous ALDH2*2 dominant-negative allele (rs671), which is associated with alcohol-induced Asian flushing. Phytohemagglutinin-stimulated lymphocytes from these patients displayed highly increased numbers of spontaneous sister chromatid exchanges (SCEs), reflecting homologous recombination repair of formaldehyde damage. Here, we report that, in contrast, patient-derived fibroblasts showed normal levels of SCEs, suggesting that different cell types or conditions generate various amounts of formaldehyde. To obtain insights about endogenous formaldehyde production and how defects in ADH5/ALDH2 affect human hematopoiesis, we constructed disease model cell lines, including induced pluripotent stem cells (iPSCs). We found that ADH5 is the primary defense against formaldehyde, and ALDH2 provides a backup. DNA repair capacity in the ADH5/ALDH2-deficient cell lines can be overwhelmed by exogenous low-dose formaldehyde, as indicated by higher levels of DNA damage than in FANCD2-deficient cells. Although ADH5/ALDH2-deficient cell lines were healthy and showed stable growth, disease model iPSCs displayed drastically defective cell expansion when stimulated into hematopoietic differentiation in vitro, displaying increased levels of DNA damage. The expansion defect was partially reversed by treatment with a new small molecule termed C1, which is an agonist of ALDH2, thus identifying a potential therapeutic strategy for the patients. We propose that hematopoiesis or lymphocyte blastogenesis may entail formaldehyde generation that necessitates elimination by ADH5/ALDH2 enzymes.


Asunto(s)
Aldehído Deshidrogenasa Mitocondrial/genética , Síndromes Congénitos de Insuficiencia de la Médula Ósea/genética , Anemia de Fanconi/genética , Células Madre Pluripotentes Inducidas/patología , Sistemas CRISPR-Cas , Línea Celular , Células Cultivadas , Síndromes Congénitos de Insuficiencia de la Médula Ósea/diagnóstico , Síndromes Congénitos de Insuficiencia de la Médula Ósea/patología , Daño del ADN , Anemia de Fanconi/diagnóstico , Anemia de Fanconi/patología , Eliminación de Gen , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Mutación
8.
Mol Biol Rep ; 50(10): 8385-8395, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37615925

RESUMEN

BACKGROUND: Fanconi anemia (FA) is a devastating hereditary disorder for which we desperately need a novel therapeutic strategy. It is caused by mutations in one of at least 22 genes in the FA pathway and is characterized by developmental abnormalities, bone marrow failure, and cancer predisposition. The FA pathway is required for the efficient repair of damaged DNA, including interstrand cross-links (ICL). Recent studies indicate formaldehyde as an ultimate endogenous cause of DNA damage in FA pathophysiology. Formaldehyde can form DNA adducts as well as ICLs by inducing covalent linkages between opposite strands of double-stranded DNA. METHODS AND RESULTS: In this study, we generated a disease model of FA in zebrafish by disrupting the ube2t or fancd2 gene, which resulted in a striking phenotype of female-to-male sex reversal. Since formaldehyde is detoxified from the body by alcohol dehydrogenase 5 (ADH5), we generated fancd2-/-/adh5-/- zebrafish. We observed a body size reduction and a lower number of mature spermatozoa than wild-type or single knockout zebrafish. To evaluate if increased activity in ADH5 can affect the FA phenotype, we overexpressed human ADH5 in fancd2-/- zebrafish. The progress of spermatogenesis seemed to be partially recovered due to ADH5 overexpression. CONCLUSIONS: Our results suggest potential utility of an ADH5 enzyme activator as a therapeutic measure for the clearance of formaldehyde and treatment of FA.


Asunto(s)
Anemia de Fanconi , Pez Cebra , Animales , Masculino , Humanos , Femenino , Pez Cebra/genética , Anemia de Fanconi/genética , Daño del ADN , Reparación del ADN , Fenotipo , Formaldehído
10.
Rinsho Ketsueki ; 64(7): 639-645, 2023.
Artículo en Japonés | MEDLINE | ID: mdl-37544724

RESUMEN

Fanconi anemia (FA), a hereditary bone marrow failure syndrome, has been suggested to be caused by a defect in DNA repair that removes endogenous DNA damage due to aldehydes. In seven Japanese children with aplastic anemia who clinically resembled FA, we identified biallelic variants of the ADH5 gene, encoding formaldehyde degrading enzyme, and a heterozygous ALDH2 variant (rs671). We conclude that the combined defects in ADH5/ALDH2 caused a new disorder now termed Aldehyde Degradation Deficiency Syndrome (ADDS). We suggest that this disease is caused by defective removal of formaldehyde produced by histone demethylation during hematopoietic cell differentiation. Therapeutic targeting of formaldehyde may reduce the hematopoietic deficits of FA as well as ADDS.


Asunto(s)
Anemia de Fanconi , Niño , Humanos , Anemia de Fanconi/genética , Aldehído Deshidrogenasa Mitocondrial/genética , Aldehídos/metabolismo , Formaldehído , Hematopoyesis/genética , Mecanismos de Defensa
11.
EMBO Rep ; 21(2): e48222, 2020 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-31867888

RESUMEN

SMC5/6 function in genome integrity remains elusive. Here, we show that SMC5 dysfunction in avian DT40 B cells causes mitotic delay and hypersensitivity toward DNA intra- and inter-strand crosslinkers (ICLs), with smc5 mutants being epistatic to FANCC and FANCM mutations affecting the Fanconi anemia (FA) pathway. Mutations in the checkpoint clamp loader RAD17 and the DNA helicase DDX11, acting in an FA-like pathway, do not aggravate the damage sensitivity caused by SMC5 dysfunction in DT40 cells. SMC5/6 knockdown in HeLa cells causes MMC sensitivity, increases nuclear bridges, micronuclei, and mitotic catastrophes in a manner similar and non-additive to FANCD2 knockdown. In both DT40 and HeLa systems, SMC5/6 deficiency does not affect FANCD2 ubiquitylation and, unlike FANCD2 depletion, RAD51 focus formation. SMC5/6 components further physically interact with FANCD2-I in human cells. Altogether, our data suggest that SMC5/6 functions jointly with the FA pathway to support genome integrity and DNA repair and may be implicated in FA or FA-related human disorders.


Asunto(s)
Proteínas de Ciclo Celular/genética , Proteínas Cromosómicas no Histona/genética , Anemia de Fanconi , ARN Helicasas DEAD-box , Daño del ADN/genética , ADN Helicasas/genética , Reparación del ADN/genética , Anemia de Fanconi/genética , Inestabilidad Genómica , Células HeLa , Humanos
12.
Proc Natl Acad Sci U S A ; 115(33): 8412-8417, 2018 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-30061412

RESUMEN

Warsaw breakage syndrome, a developmental disorder caused by mutations in the DDX11/ChlR1 helicase, shows cellular features of genome instability similar to Fanconi anemia (FA). Here we report that DDX11-deficient avian DT40 cells exhibit interstrand crosslink (ICL)-induced chromatid breakage, with DDX11 functioning as backup for the FA pathway in regard to ICL repair. Importantly, we establish that DDX11 acts jointly with the 9-1-1 checkpoint clamp and its loader, RAD17, primarily in a postreplicative fashion, to promote homologous recombination repair of bulky lesions, but is not required for intra-S checkpoint activation or efficient fork progression. Notably, we find that DDX11 also promotes diversification of the chicken Ig-variable gene, a process triggered by programmed abasic sites, by facilitating both hypermutation and homeologous recombination-mediated gene conversion. Altogether, our results uncover that DDX11 orchestrates jointly with 9-1-1 and its loader, RAD17, DNA damage tolerance at sites of bulky lesions, and endogenous abasic sites. These functions may explain the essential roles of DDX11 and its similarity with 9-1-1 during development.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , ARN Helicasas DEAD-box/fisiología , ADN Helicasas/fisiología , Reparación del ADN , Replicación del ADN , Animales , Pollos , Anemia de Fanconi/genética , Inestabilidad Genómica , Recombinación Homóloga , Humanos , Hipermutación Somática de Inmunoglobulina
13.
Rinsho Ketsueki ; 62(6): 547-553, 2021.
Artículo en Japonés | MEDLINE | ID: mdl-34219079

RESUMEN

We have recently described the identification of a novel inherited bone marrow failure syndrome. The first set of patients was diagnosed through the exome analysis of cells from Japanese patients with hypoplastic anemia, which have been deposited to the JCRB cell bank for quite some time previously. Originally, these cases were diagnosed with a novel disorder based on increased levels of sister chromatid exchanges in lymphocytes; however, causative genes were clarified only after applying the recently developed next-generation sequencing technology. Aldehyde degradation deficiency syndrome (ADDS) is caused by combined defects in two genes, ADH5 and ALDH2, which are both critical for degrading endogenously generated formaldehyde. Formaldehyde is highly reactive and toxic to biological molecules including DNA, and its endogenous generation in the absence of the degradation system results in DNA damage that overwhelms the DNA repair capacity, leading to the development of BMF with loss of hematopoietic stem cells and progression to MDS/leukemia. In this short review, we would like to summarize what is known today about ADDS for a wide readership of hematology clinicians in Japan.


Asunto(s)
Anemia de Fanconi , Proteínas Adaptadoras Transductoras de Señales , Alcohol Deshidrogenasa , Aldehído Deshidrogenasa Mitocondrial/metabolismo , Aldehídos , Síndromes Congénitos de Insuficiencia de la Médula Ósea , Humanos , Japón
14.
Hum Mutat ; 41(1): 122-128, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31513304

RESUMEN

Fanconi anemia (FA) is a rare genetic disorder characterized by bone marrow failure, predisposition to cancer, and congenital abnormalities. FA is caused by pathogenic variants in any of 22 genes involved in the DNA repair pathway responsible for removing interstrand crosslinks. FANCL, an E3 ubiquitin ligase, is an integral component of the pathway, but patients affected by disease-causing FANCL variants are rare, with only nine cases reported worldwide. We report here a FANCL founder variant, anticipated to be synonymous, c.1092G>A;p.K364=, but demonstrated to induce aberrant splicing, c.1021_1092del;p.W341_K364del, that accounts for the onset of FA in 13 cases from South Asia, 12 from India and one from Pakistan. We comprehensively illustrate the pathogenic nature of the variant, provide evidence for a founder effect, and propose including this variant in genetic screening of suspected FA patients in India and Pakistan, as well as those with ancestry from these regions of South Asia.


Asunto(s)
Proteína del Grupo de Complementación L de la Anemia de Fanconi/genética , Anemia de Fanconi/epidemiología , Anemia de Fanconi/genética , Efecto Fundador , Variación Genética , Alelos , Asia/epidemiología , Aberraciones Cromosómicas , Consanguinidad , Femenino , Genotipo , Humanos , India/epidemiología , Masculino , Mutación , Prevalencia
15.
Mol Cell ; 47(4): 511-22, 2012 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-22771115

RESUMEN

DNA interstrand crosslinks (ICLs) are highly toxic lesions that stall the replication fork to initiate the repair process during the S phase of vertebrates. Proteins involved in Fanconi anemia (FA), nucleotide excision repair (NER), and translesion synthesis (TS) collaboratively lead to homologous recombination (HR) repair. However, it is not understood how ICL-induced HR repair is carried out and completed. Here, we showed that the replicative helicase-related Mcm family of proteins, Mcm8 and Mcm9, forms a complex required for HR repair induced by ICLs. Chicken DT40 cells lacking MCM8 or MCM9 are viable but highly sensitive to ICL-inducing agents, and exhibit more chromosome aberrations in the presence of mitomycin C compared with wild-type cells. During ICL repair, Mcm8 and Mcm9 form nuclear foci that partly colocalize with Rad51. Mcm8-9 works downstream of the FA and BRCA2/Rad51 pathways, and is required for HR that promotes sister chromatid exchanges, probably as a hexameric ATPase/helicase.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Reactivos de Enlaces Cruzados/farmacología , Proteínas de Unión al ADN/metabolismo , Reparación del ADN por Recombinación/efectos de los fármacos , Reparación del ADN por Recombinación/genética , Adenosina Trifosfatasas/metabolismo , Animales , Proteína BRCA2/genética , Proteína BRCA2/metabolismo , Proteínas de Ciclo Celular/genética , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Células Cultivadas , Pollos , Aberraciones Cromosómicas/efectos de los fármacos , ADN/genética , ADN/metabolismo , ADN Helicasas/genética , ADN Helicasas/metabolismo , Replicación del ADN/efectos de los fármacos , Replicación del ADN/genética , Proteínas de Unión al ADN/genética , Anemia de Fanconi/genética , Anemia de Fanconi/metabolismo , Recombinación Homóloga/efectos de los fármacos , Recombinación Homóloga/genética , Mitomicina/farmacología , Datos de Secuencia Molecular , Intercambio de Cromátides Hermanas/efectos de los fármacos , Intercambio de Cromátides Hermanas/genética
16.
Mol Cell ; 47(1): 61-75, 2012 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-22705371

RESUMEN

The Fanconi anemia (FA) protein network is necessary for repair of DNA interstrand crosslinks (ICLs), but its control mechanism remains unclear. Here we show that the network is regulated by a ubiquitin signaling cascade initiated by RNF8 and its partner, UBC13, and mediated by FAAP20, a component of the FA core complex. FAAP20 preferentially binds the ubiquitin product of RNF8-UBC13, and this ubiquitin-binding activity and RNF8-UBC13 are both required for recruitment of FAAP20 to ICLs. Both RNF8 and FAAP20 are required for recruitment of FA core complex and FANCD2 to ICLs, whereas RNF168 can modulate efficiency of the recruitment. RNF8 and FAAP20 are needed for efficient FANCD2 monoubiquitination, a key step of the FA network; RNF8 and the FA core complex work in the same pathway to promote cellular resistance to ICLs. Thus, the RNF8-FAAP20 ubiquitin cascade is critical for recruiting FA core complex to ICLs and for normal function of the FA network.


Asunto(s)
Reparación del ADN , Proteínas de Unión al ADN/metabolismo , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Proteínas del Grupo de Complementación de la Anemia de Fanconi/metabolismo , Ubiquitinación , Secuencia de Aminoácidos , Animales , Línea Celular Tumoral , Proteínas de Unión al ADN/genética , Anemia de Fanconi/genética , Anemia de Fanconi/metabolismo , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/genética , Proteínas del Grupo de Complementación de la Anemia de Fanconi/química , Proteínas del Grupo de Complementación de la Anemia de Fanconi/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Células HeLa , Humanos , Immunoblotting , Lisina/química , Lisina/genética , Lisina/metabolismo , Microscopía Fluorescente , Datos de Secuencia Molecular , Mutación , Unión Proteica , Estructura Terciaria de Proteína , Interferencia de ARN , Homología de Secuencia de Aminoácido , Transducción de Señal , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
17.
Nucleic Acids Res ; 46(6): 2932-2944, 2018 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-29394375

RESUMEN

During mild replication stress provoked by low dose aphidicolin (APH) treatment, the key Fanconi anemia protein FANCD2 accumulates on common fragile sites, observed as sister foci, and protects genome stability. To gain further insights into FANCD2 function and its regulatory mechanisms, we examined the genome-wide chromatin localization of FANCD2 in this setting by ChIP-seq analysis. We found that FANCD2 mostly accumulates in the central regions of a set of large transcribed genes that were extensively overlapped with known CFS. Consistent with previous studies, we found that this FANCD2 retention is R-loop-dependent. However, FANCD2 monoubiquitination and RPA foci formation were still induced in cells depleted of R-loops. Interestingly, we detected increased Proximal Ligation Assay dots between FANCD2 and R-loops following APH treatment, which was suppressed by transcriptional inhibition. Collectively, our data suggested that R-loops are required to retain FANCD2 in chromatin at the middle intronic region of large genes, while the replication stress-induced upstream events leading to the FA pathway activation are not triggered by R-loops.


Asunto(s)
Cromatina/genética , Sitios Frágiles del Cromosoma/genética , Replicación del ADN/genética , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/genética , Inestabilidad Genómica/genética , Afidicolina/farmacología , Línea Celular Tumoral , Cromatina/metabolismo , ADN/química , ADN/genética , ADN/metabolismo , Daño del ADN , Reparación del ADN , Replicación del ADN/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Conformación de Ácido Nucleico , Transducción de Señal/genética , Ubiquitinación/efectos de los fármacos
18.
Int J Mol Sci ; 21(2)2020 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-31936623

RESUMEN

Melanoma is one of the most lethal and malignant cancers and its incidence is increasing worldwide, and Japan is not an exception. Although there are numerous therapeutic options for melanoma, the prognosis is still poor once it has metastasized. The main concern after removal of a primary melanoma is whether it has metastasized, and early detection of metastatic melanoma would be effective in improving the prognosis of patients. Thus, it is very important to identify reliable methods to detect metastases as early as possible. Although many prognostic biomarkers (mainly for metastases) of melanoma have been reported, there are very few effective for an early diagnosis. Serum and urinary biomarkers for melanoma diagnosis have especially received great interest because of the relative ease of sample collection and handling. Several serum and urinary biomarkers appear to have significant potential both as prognostic indicators and as targets for future therapeutic methods, but still there are no efficient serum and urinary biomarkers for early detection, accurate diagnosis and prognosis, efficient monitoring of the disease and reliable prediction of survival and recurrence. Levels of 5-S-cysteinyldopa (5SCD) in the serum or urine as biomarkers of melanoma have been found to be significantly elevated earlier and to reflect melanoma progression better than physical examinations, laboratory tests and imaging techniques, such as scintigraphy and echography. With recent developments in the treatment of melanoma, studies reporting combinations of 5SCD levels and new applications for the treatment of melanoma are gradually increasing. This review summarizes the usefulness of 5SCD, the most widely used and well-known melanoma marker in the serum and urine, compares 5SCD and other useful markers, and finally its application to other fields.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Cisteinildopa/metabolismo , Melanoma/metabolismo , Neoplasias Cutáneas/metabolismo , Cisteinildopa/sangre , Monitoreo de Drogas , Humanos , Melanoma/sangre , Melanoma/patología , Metaboloma , Neoplasias Cutáneas/sangre , Neoplasias Cutáneas/patología
19.
Haematologica ; 104(10): 1962-1973, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-30792206

RESUMEN

Fanconi anemia is a rare recessive disease characterized by multiple congenital abnormalities, progressive bone marrow failure, and a predisposition to malignancies. It results from mutations in one of the 22 known FANC genes. The number of Japanese Fanconi anemia patients with a defined genetic diagnosis was relatively limited. In this study, we reveal the genetic subtyping and the characteristics of mutated FANC genes in Japan and clarify the genotype-phenotype correlations. We studied 117 Japanese patients and successfully subtyped 97% of the cases. FANCA and FANCG pathogenic variants accounted for the disease in 58% and 25% of Fanconi anemia patients, respectively. We identified one FANCA and two FANCG hot spot mutations, which are found at low percentages (0.04-0.1%) in the whole-genome reference panel of 3,554 Japanese individuals (Tohoku Medical Megabank). FANCB was the third most common complementation group and only one FANCC case was identified in our series. Based on the data from the Tohoku Medical Megabank, we estimate that approximately 2.6% of Japanese are carriers of disease-causing FANC gene variants, excluding missense mutations. This is the largest series of subtyped Japanese Fanconi anemia patients to date and the results will be useful for future clinical management.


Asunto(s)
Proteínas del Grupo de Complementación de la Anemia de Fanconi/genética , Anemia de Fanconi/genética , Mutación , Anemia de Fanconi/epidemiología , Femenino , Estudio de Asociación del Genoma Completo , Humanos , Japón/epidemiología , Masculino
20.
Ann Hematol ; 98(2): 271-280, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30368588

RESUMEN

Fanconi anemia (FA) is a genetically and clinically heterogeneous disorder that predisposes patients to bone marrow failure (BMF), myelodysplastic syndromes (MDS), and acute myeloid leukemia (AML). To study which genetic and phenotypic factors predict clinical outcomes for Japanese FA patients, we examined the FA genes, bone marrow karyotype, and aldehyde dehydrogenase-2 (ALDH2) genotype; variants of which are associated with accelerated progression of BMF in FA. In 88 patients, we found morphologic MDS/AML in 33 patients, including refractory cytopenia in 16, refractory anemia with excess blasts (RAEB) in 7, and AML in 10. The major mutated FA genes observed in this study were FANCA (n = 52) and FANCG (n = 23). The distribution of the ALDH2 variant alleles did not differ significantly between patients with mutations in FANCA and FANCG. However, patients with FANCG mutations had inferior BMF-free survival and received hematopoietic stem cell transplantation (HSCT) at a younger age than those with FANCA mutations. In FANCA, patients with the c.2546delC mutation (n = 24) related to poorer MDS/AML-free survival and a younger age at HSCT than those without this mutation. All patients with RAEB/AML had an abnormal karyotype and poorer prognosis after HSCT; specifically, the presence of a structurally complex karyotype with a monosomy (n = 6) was associated with dismal prognosis. In conclusion, the best practice for a clinician may be to integrate the morphological, cytogenetic, and genetic data to optimize HSCT timing in Japanese FA patients.


Asunto(s)
Aldehído Deshidrogenasa Mitocondrial/genética , Secuencia de Bases , Anemia de Fanconi/genética , Anemia de Fanconi/mortalidad , Genotipo , Eliminación de Secuencia , Factores de Edad , Aldehído Deshidrogenasa Mitocondrial/metabolismo , Alelos , Aloinjertos , Pueblo Asiatico , Supervivencia sin Enfermedad , Anemia de Fanconi/enzimología , Anemia de Fanconi/terapia , Proteína del Grupo de Complementación A de la Anemia de Fanconi/genética , Proteína del Grupo de Complementación A de la Anemia de Fanconi/metabolismo , Proteína del Grupo de Complementación G de la Anemia de Fanconi/genética , Proteína del Grupo de Complementación G de la Anemia de Fanconi/metabolismo , Femenino , Frecuencia de los Genes , Trasplante de Células Madre Hematopoyéticas , Humanos , Japón , Masculino , Tasa de Supervivencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA