Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Medicine (Baltimore) ; 102(24): e34036, 2023 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-37327301

RESUMEN

RATIONALE: Bone marrow failure (BMF) includes inherited and acquired BMFs. Acquired BMF can be secondary to various factors, such as autoimmune dysfunction, benzene, drugs, radiation, viral infection and so on. Fanconi anemia (FA) complementation group L (FANCL) is an E3 ubiquitin ligase that participates in the repair of DNA damage. Homozygous or compound heterozygous mutations of FANCL can lead to the onset of FA, which is one of the most common inherited BMFs. PATIENT CONCERNS AND DIAGNOSES: Here, we report a case of acquired BMF. This patient had a history of benzene exposure for half a year before the onset of the disease, and presented with progressive pancytopenia, especially the reduction of erythrocytes and megakaryocyte, without malformation. Interestingly, this patient and his brother/father had a heterozygous (non-homozygous/compound heterozygous) mutation (Exon9, c.745C > T, p.H249Y) in the FANCL gene. INTERVENTIONS AND OUTCOMES: The patient successfully underwent unrelated and fully compatible umbilical cord blood hematopoietic stem cell transplantation. LESSONS SUBSECTIONS: We report for the first time an acquired BMF case with FANCL gene heterozygous mutation, and the mutation site (Exon9, c.745C > T, p.H249Y) has never been reported. This case suggests that heterozygous mutations in FANCL gene may be associated with increased susceptibility to acquired BMF. Based on current reports and this case, we speculate that heterozygous mutations in the FA complementation gene may exist in a certain proportion of tumor and acquired BMF patients, but have not been detected. We recommend routine screening for FA complementation gene mutations in tumor and acquired BMF patients in clinical practice. If positive results are found, further screening can be conducted on their families.


Asunto(s)
Anemia de Fanconi , Pancitopenia , Humanos , Masculino , Benceno , Anemia de Fanconi/diagnóstico , Anemia de Fanconi/genética , Proteína del Grupo de Complementación L de la Anemia de Fanconi/genética , Heterocigoto , Mutación
2.
Mol Genet Genomic Med ; 9(7): e1693, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33960719

RESUMEN

BACKGROUND: Fanconi anemia (FA) is an inherited bone marrow failure syndrome associated with characteristic dysmorphology primarily caused by biallelic pathogenic germline variants in any of 22 different DNA repair genes. There are limited data on the specific molecular causes of FA in different ethnic groups. METHODS: We performed exome sequencing and copy number variant analyses on 19 patients with FA from 17 families undergoing hematopoietic cell transplantation evaluation in Pakistan. The scientific literature was reviewed, and we curated germline variants reported in patients with FA from South Asia and the Middle East. RESULTS: The genetic causes of FA were identified in 14 of the 17 families: seven FANCA, two FANCC, one FANCF, two FANCG, and two FANCL. Homozygous and compound heterozygous variants were present in 12 and two families, respectively. Nine families carried variants previously reported as pathogenic, including two families with the South Asian FANCL founder variant. We also identified five novel likely deleterious variants in FANCA, FANCF, and FANCG in affected patients. CONCLUSIONS: Our study supports the importance of determining the genomic landscape of FA in diverse populations, in order to improve understanding of FA etiology and assist in the counseling of families.


Asunto(s)
Anemia de Fanconi/genética , Frecuencia de los Genes , Adolescente , Asia , Niño , Preescolar , Variaciones en el Número de Copia de ADN , Exoma , Anemia de Fanconi/diagnóstico , Proteína del Grupo de Complementación F de la Anemia de Fanconi/genética , Proteína del Grupo de Complementación G de la Anemia de Fanconi/genética , Proteína del Grupo de Complementación L de la Anemia de Fanconi/genética , Femenino , Efecto Fundador , Humanos , Masculino , Medio Oriente , Mutación
3.
Mol Biol Rep ; 48(1): 585-593, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33394227

RESUMEN

Fanconi Anemia (FA) is a rare genetic disease with the incidence of 1 in 360,000 and is characterised by bone marrow failure, physical abnormalities, pancytopenia, and high frequency of chromosomal breakage and increased risk of evolving into malignancy. Telomere plays an important role in genomic stability, ageing process and cancers. Telomere shortening has been reported in FA. We studied telomere length in FA subjects and compared with complementation groups. Chromosomal breakage analysis from PHA stimulated, MMC induced peripheral blood culture was carried out in 37 clinically diagnosed FA. Molecular study of FANCA, G, and L was done through Sanger sequencing and next generation sequencing. Telomere length was estimated using real time quantitative polymerase chain reaction (qPCR) method. Student t-test was applied to test the significance. A high frequency of chromosomal breakage was observed in all the patients compared to healthy controls. We found significantly shorter telomere length in all the three complementation groups compare to age matched healthy controls. Among all complementation groups, FANCL showed severe telomere shortening (P value 0.0001). A negative correlation was observed between telomere length and chromosomal breakage frequency (R = -0.3116). Telomere shortening is not uncommon in FA subjects. However the telomere length shortening is different in complementation groups as FANCL showed severe telomere shortening in FA subjects. Though BM transplantation is essential for the management of the FA subjects, the telomere length can be considered as biological marker to understand the prognosis of the disease as FA subjects primarily treated with androgens.


Asunto(s)
Proteína del Grupo de Complementación A de la Anemia de Fanconi/genética , Proteína del Grupo de Complementación G de la Anemia de Fanconi/genética , Proteína del Grupo de Complementación L de la Anemia de Fanconi/genética , Anemia de Fanconi/genética , Acortamiento del Telómero/genética , Adolescente , Adulto , Niño , Preescolar , Rotura Cromosómica , Proteínas de Unión al ADN/genética , Anemia de Fanconi/patología , Femenino , Regulación de la Expresión Génica/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Telómero/genética , Adulto Joven
4.
PLoS One ; 15(8): e0237825, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32822435

RESUMEN

Cattle temperament is a complex and economically relevant trait. The objective of this study was to identify genomic regions and genes associated with cattle temperament. From a Brahman cattle population of 1,370 animals evaluated for temperament traits (Exit velocity-EV, Pen Score-PS, Temperament Score-TS), two groups of temperament-contrasting animals were identified based on their EV-average values ±1/2 standard deviation (SD). To be considered in the calm group, the EV of females ranged between 0.16-1.82 m/s (n = 50) and the EV of males ranged between 0.4-1.56 m/s (n = 48). Females were classified as temperamental if their EV ranged between 3.13-7.66 m/s (n = 46) and males were classified as temperamental if their EV ranged between 3.05-10.83 m/s (n = 45). Selected animals were genotyped using a total of 139,376 SNPs (GGP-HD-150K), evaluated for their association with EV. The Genome-Wide Association analysis (GWAS) identified fourteen SNPs: rs135340276, rs134895560, rs110190635, rs42949831, rs135982573, rs109393235, rs109531929, rs135087545, rs41839733, rs42486577, rs136661522, rs110882543, rs110864071, rs109722627, (P<8.1E-05), nine of them were located on intergenic regions, harboring seventeen genes, of which only ACER3, VRK2, FANCL and SLCO3A1 were considered candidate associated with bovine temperament due to their reported biological functions. Five SNPs were located at introns of the NRXN3, EXOC4, CACNG4 and SLC9A4 genes. The indicated candidate genes are implicated in a wide range of behavioural phenotypes and complex cognitive functions. The association of the fourteen SNPs on bovine temperament traits (EV, PS and TS) was evaluated; all these SNPs were significant for EV; only some were associated with PS and TS. Fourteen SNPs were associated with EV which allowed the identification of twenty-one candidate genes for Brahman temperament. From a functional point of view, the five intronic SNPs identified in this study, are candidates to address control of bovine temperament, further investigation will probe their role in expression of this trait.


Asunto(s)
Conducta Animal , Bovinos/genética , Bovinos/psicología , Emociones , Temperamento , Ceramidasa Alcalina/genética , Animales , Proteína del Grupo de Complementación L de la Anemia de Fanconi/genética , Femenino , Estudios de Asociación Genética , Estudio de Asociación del Genoma Completo , Genotipo , Masculino , Transportadores de Anión Orgánico/genética , Fenotipo , Polimorfismo de Nucleótido Simple , Proteínas Serina-Treonina Quinasas/genética , Intercambiadores de Sodio-Hidrógeno/genética , Proteínas de Transporte Vesicular/genética
7.
Biosci Rep ; 40(6)2020 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-32420600

RESUMEN

Fanconi Anemia (FA) is a rare genetic disorder characterized by developmental defects, bone marrow failure and high predisposition to cancer. The FA DNA repair pathway is required in humans to coordinate repair of DNA interstrand cross-links. The central event in the activation of the pathway is the monoubiquitination of FANCD2 and FANCI by the E2-E3 pair, Ube2T-FANCL, with the central UBC-RWD (URD) domain of FANCL recognizing the substrates. Whole genome sequencing studies of cancer cells from patients identified point mutations in the FANCL URD domain. We analysed 17 such variants of FANCL, including known substrate binding mutants (W212A, W214A and L248A, F252A, L254A, I265A), a FA mutation (R221C) and 14 cancer-associated mutations (F110S, I136V, L149V, L154S, A192G, E215Q, E217K, R221W, T224K, M247V, F252L, N270K, V287G, E289Q) through recombinant expression analysis, thermal shift assay, interaction with FANCD2, in vitro ubiquitination activity, and cellular sensitivity to an interstrand cross-linking agent. We find that the FANCL mutations I136V, L154S, W212A and L214A, R221W, R221C, and V287G are destabilizing, with N270K and E289Q destabilizing the C-terminal helices of the URD domain. The hydrophobic patch mutant (L248A, F252A, L254A, I265A), along with mutations E217K, T224K, and M247V, cause defects in the catalytic function of FANCL. This highlights the C-terminal lobe of the FANCL URD domain as important for the activity and function of FANCL. These mutations which affect the fold and activity of FANCL may contribute to tumorigenesis in these non-FA cancer patients, and this implicates FA genes in general cancer progression.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias Óseas/genética , Proteína del Grupo de Complementación L de la Anemia de Fanconi/genética , Anemia de Fanconi/genética , Mutación , Osteosarcoma/genética , Antineoplásicos Alquilantes/farmacología , Biomarcadores de Tumor/metabolismo , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/metabolismo , Neoplasias Óseas/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Análisis Mutacional de ADN , Anemia de Fanconi/diagnóstico , Anemia de Fanconi/metabolismo , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Proteína del Grupo de Complementación L de la Anemia de Fanconi/metabolismo , Predisposición Genética a la Enfermedad , Humanos , Mitomicina/farmacología , Osteosarcoma/tratamiento farmacológico , Osteosarcoma/metabolismo , Osteosarcoma/patología , Conformación Proteica en Hélice alfa , Pliegue de Proteína , Estabilidad Proteica , Proteolisis , Ubiquitinación
8.
Hum Mutat ; 41(5): 1033-1041, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32048394

RESUMEN

The Fanconi anemia (FA) pathway is mainly involved in DNA interstrand crosslinks (ICLs) repair in the genome. Several FA genes, including FANCD1/BRCA2, FANCM, and FANCU/XRCC2, have been identified as causative genes for premature ovary insufficiency (POI). Fanconi anemia group L protein (FANCL) cooperates with FANCT/UBE2T to ubiquitinate the FANCI-D2 dimer, which is a crucial event in the process of ICLs repair. Fancl-knockout mice phenocopy human POI, but the role of FANCL mutations in POI pathogenesis has not been confirmed. In the present work, potentially pathogenic mutations in the FANCL gene were screened in 200 Chinese patients with idiopathic POI and in 200 matched controls. Two novel heterozygous frameshift mutations, c.1048_1051delGTCT (p.Gln350Valfs*18) and c.739dupA (p.Met247Asnfs*4), were identified in the FANCL gene in POI patients but not in controls. Wild-type FANCL protein was predominantly localized in the nuclei, while both mutant FANCL proteins were retained in the cytoplasm. In addition, the FANCL variants exhibited impaired ubiquitin-ligase activity and compromised DNA repair ability after mitomycin C treatment. Furthermore, the FANCL variants were deleterious and might be associated with haploinsufficiency. Our results show that FANCL mutations are potentially causative for POI by disrupting DNA damage repair processes.


Asunto(s)
Proteína del Grupo de Complementación L de la Anemia de Fanconi/genética , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Mutación , Insuficiencia Ovárica Primaria/diagnóstico , Insuficiencia Ovárica Primaria/genética , Adulto , Biomarcadores , Línea Celular , Daño del ADN , Reparación del ADN , Proteína del Grupo de Complementación L de la Anemia de Fanconi/metabolismo , Femenino , Mutación del Sistema de Lectura , Técnicas de Silenciamiento del Gen , Estudios de Asociación Genética/métodos , Heterocigoto , Humanos , Insuficiencia Ovárica Primaria/metabolismo , Transporte de Proteínas , Interferencia de ARN , Factores de Riesgo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Adulto Joven
9.
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
10.
Endocrinology ; 159(11): 3699-3722, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30184072

RESUMEN

Sex determination and differentiation are complex processes. As a juvenile hermaphrodite or undifferentiated gonochorist, zebrafish undergo a special juvenile ovarian phase during sex differentiation, making it an excellent model for studying early oogenesis and folliculogenesis. We provide lines of evidence at morphological, molecular, and genetic levels for roles of factor in the germline α (Figla), an oocyte-specific transcription factor, in early zebrafish gonadogenesis. As in mammals, Figla/figla was also expressed in the gonads and its expression in the ovary was also restricted to early oocytes. Disruption of figla gene by CRISPR/Cas9 led to an all-male phenotype in the mutant. Detailed analysis of early gonadal development showed that the germ cells in the mutant were clustered in cysts and underwent meiosis, forming oocytes at prefollicular chromatin nucleolar (CN) stage (stage IA). However, the subsequent transition from cystic CN oocytes to individual follicular perinucleolar oocytes (stage IB) was blocked, resulting in an all-male phenotype in the mutant. The phenotype of figla mutant could not be rescued by estrogen treatment, in contrast to cyp19a1a mutant, and introduction of tp53 mutation also had no effect, unlike in fancd1 and fancl mutants. Transcriptome analysis revealed that many biological processes and pathways related to germ cell development, especially oogenesis, were upregulated in the presence of Figla and that the regulation of figla expression may involve heat shock proteins. Our results strongly suggest important roles for Figla in juvenile ovary development, especially the formation of individual follicles from cystic oocytes.


Asunto(s)
Oogénesis/genética , Organogénesis/genética , Folículo Ovárico/crecimiento & desarrollo , Ovario/crecimiento & desarrollo , Procesos de Determinación del Sexo/genética , Proteínas de Pez Cebra/genética , Animales , Aromatasa/genética , Proteína BRCA2/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Estradiol/farmacología , Estrógenos/farmacología , Proteína del Grupo de Complementación L de la Anemia de Fanconi/genética , Femenino , Perfilación de la Expresión Génica , Proteínas de Choque Térmico/metabolismo , Masculino , Meiosis , Mutación , Oogénesis/efectos de los fármacos , Organogénesis/efectos de los fármacos , Folículo Ovárico/efectos de los fármacos , Ovario/efectos de los fármacos , Fenotipo , Proteína p53 Supresora de Tumor/genética , Pez Cebra , Proteínas de Pez Cebra/metabolismo
11.
Cancer ; 123(20): 3943-3954, 2017 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-28678401

RESUMEN

BACKGROUND: Patients with Fanconi anemia (FA) have an increased risk for head and neck squamous cell carcinoma (HNSCC). The authors sought to determine the prevalence of undiagnosed FA and FA carriers among patients with HNSCC as well as an age cutoff for FA genetic screening. METHODS: Germline DNA samples from 417 patients with HNSCC aged <50 years were screened for sequence variants by targeted next-generation sequencing of the entire length of 16 FA genes. RESULTS: The sequence revealed 194 FA gene variants in 185 patients (44%). The variant spectrum was comprised of 183 nonsynonymous point mutations, 9 indels, 1 large deletion, and 1 synonymous variant that was predicted to effect splicing. One hundred eight patients (26%) had at least 1 rare variant that was predicted to be damaging, and 57 (14%) had at least 1 rare variant that was predicted to be damaging and had been previously reported. Fifteen patients carried 2 rare variants or an X-linked variant in an FA gene. Overall, an age cutoff for FA screening was not identified among young patients with HNSCC, because there were no significant differences in mutation rates when patients were stratified by age, tumor site, ethnicity, smoking status, or human papillomavirus status. However, an increased burden, or mutation load, of FA gene variants was observed in carriers of the genes FA complementation group D2 (FANCD2), FANCE, and FANCL in the HNSCC patient cohort relative to the 1000 Genomes population. CONCLUSIONS: FA germline functional variants offer a novel area of study in HNSCC tumorigenesis. FANCE and FANCL, which are components of the core complex, are known to be responsible for the recruitment and ubiquitination, respectively, of FANCD2, a critical step in the FA DNA repair pathway. In the current cohort, the increased mutation load of FANCD2, FANCE, and FANCL variants among younger patients with HNSCC indicates the importance of the FA pathway in HNSCC. Cancer 2017;123:3943-54. © 2017 American Cancer Society.


Asunto(s)
Carcinoma de Células Escamosas/genética , Anemia de Fanconi/genética , Neoplasias de Cabeza y Cuello/genética , Adulto , Edad de Inicio , Proteína BRCA2/genética , Análisis Mutacional de ADN , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/genética , Proteína del Grupo de Complementación E de la Anemia de Fanconi/genética , Proteína del Grupo de Complementación L de la Anemia de Fanconi/genética , Proteínas del Grupo de Complementación de la Anemia de Fanconi/genética , Femenino , Mutación de Línea Germinal , Heterocigoto , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple , Recombinasas/genética , Análisis de Secuencia de ADN , Carcinoma de Células Escamosas de Cabeza y Cuello
12.
Eur J Med Genet ; 60(7): 369-373, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28419882

RESUMEN

Fanconi Anemia (FA) is a rare genetically heterogeneous disorder with 17 known complement groups caused by mutations in different genes. FA complementation group L (FA-L, OMIM #608111) occurred in 0.2% of all FA and only eight mutant variants in the FANCL gene were documented. Phenotype and genotype correlation in FANCL associated FA is still obscure. Here we describe a Chinese girl with FA-L caused by a novel homozygous mutation c.822_823insCTTTCAGG (p.Asp275LeufsX13) in the FANCL gene. The patient's clinical course was typical for FA with progression to bone marrow failure, and death from acute myelomonocytic leukemia (AML-M4) at 9 years of age. Mutation analysis also detected a likely somatic c.2608G > A (p.Gly870Ser) in the SETBP1 gene. Consistent copy number losses of 7q and 18p and gains of 3q and 21q and accumulated non-clonal single cell chromosomal abnormalities were detected in blood leukocytes as her FA progressed. This is the first Chinese FA-L case caused by a novel FANCL mutation. The somatic gene mutation and copy number aberrations could be used to monitor disease progression and the clinical findings provide further information for genotype-phenotype correlation for FA-L.


Asunto(s)
Proteínas Portadoras/genética , Proteína del Grupo de Complementación L de la Anemia de Fanconi/genética , Anemia de Fanconi/genética , Proteínas Nucleares/genética , Adulto , Biomarcadores/sangre , Preescolar , Aberraciones Cromosómicas , Variaciones en el Número de Copia de ADN , Anemia de Fanconi/sangre , Anemia de Fanconi/diagnóstico , Femenino , Heterocigoto , Homocigoto , Humanos
15.
Oncogene ; 35(47): 6087-6095, 2016 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-27132514

RESUMEN

SALL4 is aberrantly expressed in human myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML). We have generated a SALL4 transgenic (SALL4B Tg) mouse model with pre-leukemic MDS-like symptoms that transform to AML over time. This makes our mouse model applicable for studying human MDS/AML diseases. Characterization of the leukemic initiation population in this model leads to the discovery that Fancl (Fanconi anemia, complementation group L) is downregulated in SALL4B Tg leukemic and pre-leukemic cells. Similar to the reported Fanconi anemia (FA) mouse model, chromosomal instability with radial changes can be detected in pre-leukemic SALL4B Tg bone marrow (BM) cells after DNA damage challenge. Results from additional studies using DNA damage repair reporter assays support a role of SALL4 in inhibiting the homologous recombination pathway. Intriguingly, unlike the FA mouse model, after DNA damage challenge, SALL4B Tg BM cells can survive and generate hematopoietic colonies. We further elucidated that the mechanism by which SALL4 promotes cell survival is through Bcl2 activation. Overall, our studies demonstrate for the first time that SALL4 has a negative impact in DNA damage repair, and support the model of dual functional properties of SALL4 in leukemogenesis through inhibiting DNA damage repair and promoting cell survival.


Asunto(s)
Daño del ADN , Reparación del ADN , Proteínas de Unión al ADN/metabolismo , Factores de Transcripción/metabolismo , Animales , Células de la Médula Ósea/metabolismo , Trasplante de Médula Ósea , Inestabilidad Cromosómica/efectos de los fármacos , Análisis por Conglomerados , Reparación del ADN por Unión de Extremidades , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/genética , Proteína del Grupo de Complementación L de la Anemia de Fanconi/genética , Proteína del Grupo de Complementación L de la Anemia de Fanconi/metabolismo , Femenino , Expresión Génica , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Genes bcl-2 , Células Madre Hematopoyéticas/metabolismo , Recombinación Homóloga , Humanos , Cariotipo , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Masculino , Ratones , Ratones Transgénicos , Mitomicina/farmacología , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/metabolismo , Factores de Transcripción/genética
16.
Oncotarget ; 7(22): 32351-61, 2016 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-27083049

RESUMEN

Mutations in the human RecQ helicase, BLM, causes Bloom Syndrome, which is a rare autosomal recessive disorder and characterized by genomic instability and an increased risk of cancer. Fanconi Anemia (FA), resulting from mutations in any of the 19 known FA genes and those yet to be known, is also characterized by chromosomal instability and a high incidence of cancer. BLM helicase and FA proteins, therefore, may work in a common tumor-suppressor signaling pathway. To date, it remains largely unclear as to how BLM and FA proteins work concurrently in the maintenance of genome stability. Here we report that BLM is involved in the early activation of FA group D2 protein (FANCD2). We found that FANCD2 activation is substantially delayed and attenuated in crosslinking agent-treated cells harboring deficient Blm compared to similarly treated control cells with sufficient BLM. We also identified that the domain VI of BLM plays an essential role in promoting FANCD2 activation in cells treated with DNA crosslinking agents, especially ultraviolet B. The similar biological effects performed by ΔVI-BLM and inactivated FANCD2 further confirm the relationship between BLM and FANCD2. Mutations within the domain VI of BLM detected in human cancer samples demonstrate the functional importance of this domain, suggesting human tumorigenicity resulting from mtBLM may be at least partly attributed to mitigated FANCD2 activation. Collectively, our data show a previously unknown regulatory liaison in advancing our understanding of how the cancer susceptibility gene products act in concert to maintain genome stability.


Asunto(s)
Síndrome de Bloom/enzimología , Proteína del Grupo de Complementación L de la Anemia de Fanconi/metabolismo , Anemia de Fanconi/enzimología , Neoplasias/enzimología , RecQ Helicasas/metabolismo , Transducción de Señal , Síndrome de Bloom/genética , Síndrome de Bloom/patología , Neoplasias Óseas/enzimología , Neoplasias Óseas/genética , Neoplasias Óseas/patología , Línea Celular Tumoral , Supervivencia Celular , Reactivos de Enlaces Cruzados/farmacología , Anemia de Fanconi/genética , Anemia de Fanconi/patología , Proteína del Grupo de Complementación L de la Anemia de Fanconi/química , Proteína del Grupo de Complementación L de la Anemia de Fanconi/genética , Femenino , Humanos , Mutación , Neoplasias/genética , Neoplasias/patología , Osteosarcoma/enzimología , Osteosarcoma/genética , Osteosarcoma/patología , Neoplasias Ováricas/enzimología , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Interferencia de ARN , RecQ Helicasas/química , RecQ Helicasas/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/efectos de la radiación , Transfección , Rayos Ultravioleta
17.
J Proteome Res ; 15(4): 1333-41, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26956768

RESUMEN

Metabolic profiling has great potential to help the diagnosis and prognosis of cancer patients. Fanconi Anemia (FA) tumor-suppressor signaling has been instrumental in understanding human tumorigenesis. However, this instrumental understanding has never been demonstrated at the metabolic level. Here, we show that impaired FA signaling can lead cells to exhibit metabolic signatures of tumorigenesis. This is consistent with our original studies of the roles of FA signaling in suppressing non-FA tumorigenesis at functional and genetic levels. Using ultraperformance liquid chromatography-mass spectroscopy and gas chromatography-mass spectrometry, we characterized metabolic alterations in bladder cancer cells carrying an intact or impaired FA pathway. The latter was obtained by ectopically expressing FAVL (FAVL-high), which we previously found to be capable of inactivating FA signaling. A total of 18 metabolites, end products of cell proliferation or apoptosis, were significantly different between FAVL-high and -low cells. Methionine, phenylalanine, and threonine, resulting from a tumorigenic process, were substantially increased in FAVL-high cells. With this study, we achieved genomic, functional, and metabolomic characterization of the roles of FA signaling in the development of human cancer. Furthermore, this study provides novel insights into how to translate FA basic research into strategies for producing effective biomarkers in human cancer diagnosis and prognosis.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Células Epiteliales/metabolismo , Proteína del Grupo de Complementación L de la Anemia de Fanconi/genética , Metaboloma , Transducción de Señal , Empalme Alternativo , Apoptosis , Línea Celular Tumoral , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Cromatografía Liquida , Células Epiteliales/patología , Proteína del Grupo de Complementación L de la Anemia de Fanconi/metabolismo , Expresión Génica , Humanos , Espectrometría de Masas , Metionina/metabolismo , Fenilalanina/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Treonina/metabolismo , Transgenes
18.
Clin Genet ; 90(4): 324-33, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-26822949

RESUMEN

Hereditary breast cancer comprises a minor but clinically meaningful breast cancer (BC) subgroup. Mutations in the major BC-susceptibility genes are important prognostic and predictive markers; however, their carriers represent only 25% of high-risk BC patients. To further characterize variants influencing BC risk, we performed SOLiD sequencing of 581 genes in 325 BC patients (negatively tested in previous BRCA1/BRCA2/PALB2 analyses). In 105 (32%) patients, we identified and confirmed 127 truncating variants (89 unique; nonsense, frameshift indels, and splice site), 19 patients harbored more than one truncation. Forty-six (36 unique) truncating variants in 25 DNA repair genes were found in 41 (12%) patients, including 16 variants in the Fanconi anemia (FA) genes. The most frequent variant in FA genes was c.1096_1099dupATTA in FANCL that also show a borderline association with increased BC risk in subsequent analysis of enlarged groups of BC patients and controls. Another 81 (53 unique) truncating variants were identified in 48 non-DNA repair genes in 74 patients (23%) including 16 patients carrying variants in genes coding proteins of estrogen metabolism/signaling. Our results highlight the importance of mutations in the FA genes' family, and indicate that estrogen metabolism genes may reveal a novel candidate genetic component for BC susceptibility.


Asunto(s)
Neoplasias de la Mama/genética , Reparación del ADN/genética , Mutación , Anciano , Anciano de 80 o más Años , Proteína BRCA1/genética , Proteína BRCA2/genética , Análisis Mutacional de ADN , Proteína del Grupo de Complementación L de la Anemia de Fanconi/genética , Proteína del Grupo de Complementación N de la Anemia de Fanconi , Femenino , Humanos , Persona de Mediana Edad , Proteínas Nucleares/genética , Proteínas Supresoras de Tumor/genética
19.
J Biomed Sci ; 22: 77, 2015 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-26385482

RESUMEN

BACKGROUND: Cisplatin is one of the most commonly used chemotherapy agent for lung cancer. The therapeutic efficacy of cisplatin is limited by the development of resistance. In this study, we test the effect of RNA interference (RNAi) targeting Fanconi anemia (FA)/BRCA pathway upstream genes on the sensitivity of cisplatin-sensitive (A549 and SK-MES-1) and -resistant (A549/DDP) lung cancer cells to cisplatin. RESULT: Using small interfering RNA (siRNA), knockdown of FANCF, FANCL, or FANCD2 inhibited function of the FA/BRCA pathway in A549, A549/DDP and SK-MES-1 cells, and potentiated sensitivity of the three cells to cisplatin. The extent of proliferation inhibition induced by cisplatin after knockdown of FANCF and/or FANCL in A549/DDP cells was significantly greater than in A549 and SK-MES-1 cells, suggesting that depletion of FANCF and/or FANCL can reverse resistance of cisplatin-resistant lung cancer cells to cisplatin. Furthermore, knockdown of FANCL resulted in higher cisplatin sensitivity and dramatically elevated apoptosis rates compared with knockdown of FANCF in A549/DDP cells, indicating that FANCL play an important role in the repair of cisplatin-induced DNA damage. CONCLUSION: Knockdown of FANCF, FANCL, or FANCD2 by RNAi could synergize the effect of cisplatin on suppressing cell proliferation in cisplatin-resistant lung cancer cells through inhibition of FA/BRCA pathway.


Asunto(s)
Proteína BRCA1 , Cisplatino/farmacología , Resistencia a Antineoplásicos , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi , Proteína del Grupo de Complementación F de la Anemia de Fanconi , Proteína del Grupo de Complementación L de la Anemia de Fanconi , Neoplasias Pulmonares , Interferencia de ARN , Transducción de Señal , Proteína BRCA1/antagonistas & inhibidores , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Línea Celular Tumoral , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/antagonistas & inhibidores , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/genética , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Proteína del Grupo de Complementación F de la Anemia de Fanconi/antagonistas & inhibidores , Proteína del Grupo de Complementación F de la Anemia de Fanconi/genética , Proteína del Grupo de Complementación F de la Anemia de Fanconi/metabolismo , Proteína del Grupo de Complementación L de la Anemia de Fanconi/antagonistas & inhibidores , Proteína del Grupo de Complementación L de la Anemia de Fanconi/genética , Proteína del Grupo de Complementación L de la Anemia de Fanconi/metabolismo , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
20.
J Biol Chem ; 290(34): 20995-21006, 2015 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-26149689

RESUMEN

The Fanconi Anemia (FA) DNA repair pathway is essential for the recognition and repair of DNA interstrand crosslinks (ICL). Inefficient repair of these ICL can lead to leukemia and bone marrow failure. A critical step in the pathway is the monoubiquitination of FANCD2 by the RING E3 ligase FANCL. FANCL comprises 3 domains, a RING domain that interacts with E2 conjugating enzymes, a central domain required for substrate interaction, and an N-terminal E2-like fold (ELF) domain. The ELF domain is found in all FANCL homologues, yet the function of the domain remains unknown. We report here that the ELF domain of FANCL is required to mediate a non-covalent interaction between FANCL and ubiquitin. The interaction involves the canonical Ile44 patch on ubiquitin, and a functionally conserved patch on FANCL. We show that the interaction is not necessary for the recognition of the core complex, it does not enhance the interaction between FANCL and Ube2T, and is not required for FANCD2 monoubiquitination in vitro. However, we demonstrate that the ELF domain is required to promote efficient DNA damage-induced FANCD2 monoubiquitination in vertebrate cells, suggesting an important function of ubiquitin binding by FANCL in vivo.


Asunto(s)
Reparación del ADN , Proteínas de Drosophila/química , Proteína del Grupo de Complementación L de la Anemia de Fanconi/química , Proteínas del Grupo de Complementación de la Anemia de Fanconi/química , Ubiquitina/química , Proteínas de Xenopus/química , Secuencia de Aminoácidos , Animales , Sitios de Unión , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Anemia de Fanconi/genética , Proteína del Grupo de Complementación L de la Anemia de Fanconi/genética , Proteína del Grupo de Complementación L de la 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 , Regulación de la Expresión Génica , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Unión Proteica , Pliegue de Proteína , Dominios y Motivos de Interacción de Proteínas , Estructura Secundaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Transducción de Señal , Ubiquitina/genética , Ubiquitina/metabolismo , Enzimas Ubiquitina-Conjugadoras/genética , Enzimas Ubiquitina-Conjugadoras/metabolismo , Ubiquitinación , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo , Xenopus laevis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...