Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 150
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 81(14): 2989-3006.e9, 2021 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-34197737

RESUMEN

Stalled DNA replication fork restart after stress as orchestrated by ATR kinase, BLM helicase, and structure-specific nucleases enables replication, cell survival, and genome stability. Here we unveil human exonuclease V (EXO5) as an ATR-regulated DNA structure-specific nuclease and BLM partner for replication fork restart. We find that elevated EXO5 in tumors correlates with increased mutation loads and poor patient survival, suggesting that EXO5 upregulation has oncogenic potential. Structural, mechanistic, and mutational analyses of EXO5 and EXO5-DNA complexes reveal a single-stranded DNA binding channel with an adjacent ATR phosphorylation motif (T88Q89) that regulates EXO5 nuclease activity and BLM binding identified by mass spectrometric analysis. EXO5 phospho-mimetic mutant rescues the restart defect from EXO5 depletion that decreases fork progression, DNA damage repair, and cell survival. EXO5 depletion furthermore rescues survival of FANCA-deficient cells and indicates EXO5 functions epistatically with SMARCAL1 and BLM. Thus, an EXO5 axis connects ATR and BLM in directing replication fork restart.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/genética , Replicación del ADN/genética , ADN/genética , Exonucleasas/genética , Inestabilidad Genómica/genética , RecQ Helicasas/genética , Línea Celular , Línea Celular Tumoral , Daño del ADN/genética , ADN Helicasas/genética , Análisis Mutacional de ADN/métodos , Reparación del ADN/genética , Proteínas de Unión al ADN/genética , Células HEK293 , Células HeLa , Humanos , Mutación/genética , Oncogenes/genética , Fosforilación/genética , Regulación hacia Arriba/genética
3.
Mol Cell ; 62(3): 409-421, 2016 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-27153538

RESUMEN

The NuA4/TIP60 acetyltransferase complex is a key regulator of genome expression and stability. Here we identified MBTD1 as a stable subunit of the complex, and we reveal that, via a histone reader domain for H4K20me1/2, MBTD1 allows TIP60 to associate with specific gene promoters and to promote the repair of DNA double-strand breaks by homologous recombination. It was previously suggested that TIP60-dependent acetylation of H4 regulates binding of the non-homologous end joining factor 53BP1, which engages chromatin through simultaneous binding of H4K20me2 and H2AK15ub. We find that the TIP60 complex regulates association of 53BP1 partly by competing for H4K20me2 and by regulating H2AK15ub. Ubiquitylation of H2AK15 by RNF168 inhibits chromatin acetylation by TIP60, while this residue can be acetylated by TIP60 in vivo, blocking its ubiquitylation. Altogether, these results uncover an intricate mechanism orchestrated by the TIP60 complex to regulate 53BP1-dependent repair through competitive bivalent binding and modification of chromatin.


Asunto(s)
Ensamble y Desensamble de Cromatina , Proteínas Cromosómicas no Histona/metabolismo , Histona Acetiltransferasas/metabolismo , Histonas/metabolismo , Procesamiento Proteico-Postraduccional , Proteína 1 de Unión al Supresor Tumoral P53/metabolismo , Acetilación , Sitios de Unión , Unión Competitiva , Sistemas CRISPR-Cas , Proteínas Cromosómicas no Histona/genética , Roturas del ADN de Doble Cadena , Reparación del ADN , Histona Acetiltransferasas/genética , Histonas/genética , Humanos , Células K562 , Lisina Acetiltransferasa 5 , Regiones Promotoras Genéticas , Unión Proteica , Interferencia de ARN , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transducción de Señal , Factores de Tiempo , Transcripción Genética , Transfección , Proteína 1 de Unión al Supresor Tumoral P53/genética , Ubiquitinación
4.
J Transl Med ; 21(1): 286, 2023 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-37118828

RESUMEN

BACKGROUND: Osteosarcoma is a type of bone cancer that predominantly affects young individuals, including children and adolescents. The disease progresses through heterogeneous genetic alterations, and patients often develop pulmonary metastases even after the primary tumors have been surgically removed. Ubiquitin-specific peptidases (USPs) regulate several critical cellular processes, such as cell cycle progression, transcriptional activation, and signal transduction. Various studies have revealed the significance of USP37 in the regulation of replication stress and oncogenesis. METHODS: In this study, the Cancer Genome Atlas (TCGA) database was analyzed to investigate USP37 expression. RNA sequencing was utilized to assess the impact of USP37 overexpression and depletion on gene expression in osteosarcoma cells. Various molecular assays, including colony formation, immunofluorescence, immunoprecipitation, and DNA replication restart, were employed to examine the physical interaction between USP37 and PCNA, as well as its physiological effects in osteosarcoma cells. Additionally, molecular docking studies were conducted to gain insight into the nature of the interaction between USP37 and PCNA. Furthermore, immunohistochemistry was performed on archived tissue blocks from osteosarcoma patients to establish a correlation between USP37 and PCNA expression. RESULTS: Analysis of the TCGA database revealed that increased expression of USP37 was linked to decreased progression-free survival (PFS) in osteosarcoma patients. Next-generation sequencing analysis of osteosarcoma cells demonstrated that overexpression or knockdown of USP37 led to the expression of different sets of genes. USP37 overexpression provided a survival advantage, while its depletion heightened sensitivity to replication stress in osteosarcoma cells. USP37 was found to physically interact with PCNA, and molecular docking studies indicated that the interaction occurs through unique residues. In response to genotoxic stress, cells that overexpressed USP37 resolved DNA damage foci more quickly than control cells or cells in which USP37 was depleted. The expression of USP37 varied in archived osteosarcoma tissues, with intermediate expression seen in 52% of cases in the cohort examined. CONCLUSION: The results of this investigation propose that USP37 plays a vital role in promoting replication stress tolerance in osteosarcoma cells. The interaction between USP37 and PCNA is involved in the regulation of replication stress, and disrupting it could potentially trigger synthetic lethality in osteosarcoma. This study has expanded our knowledge of the mechanism through which USP37 regulates replication stress, and its potential as a therapeutic target in osteosarcoma merits additional exploration.


Asunto(s)
Neoplasias Óseas , Osteosarcoma , Niño , Humanos , Adolescente , Antígeno Nuclear de Célula en Proliferación , Endopeptidasas/genética , Endopeptidasas/metabolismo , Simulación del Acoplamiento Molecular , Proteasas Ubiquitina-Específicas , Osteosarcoma/genética , Neoplasias Óseas/genética
5.
Subcell Biochem ; 100: 115-141, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36301493

RESUMEN

The accurate repair of genomic damage mediated by ionizing radiation (IR), chemo- or radiomimetic drugs, or other exogenous agents, is necessary for maintenance of genome integrity, preservation of cellular viability and prevention of oncogenic transformation. Eukaryotes have conserved mechanisms designed to perceive and repair the damaged DNA quite efficiently. Among the different types of DNA damage, double strand breaks (DSB) are the most detrimental. The cellular DNA DSB response is a hierarchical signaling network that integrates damage sensing and repair with chromatin structural changes that involve a range of pre-existing and induced covalent modifications. Recent studies have revealed that pre-existing histone modifications are important contributors within this signaling/repair network. This chapter discusses the role of a critical histone acetyl transferase (HAT) known as MOF (males absent on the first) and the histone deacetylases (HDACs) Sirtuins on histone H4K16 acetylation (H4K16ac) and DNA damage repair. We also discuss the role of this important histone modification in light of metabolic rewiring and its role in regulating human pathophysiologic states.


Asunto(s)
Envejecimiento , Histona Acetiltransferasas , Neoplasias , Sirtuinas , Humanos , Acetilación , Cromatina , ADN/metabolismo , Daño del ADN , Reparación del ADN , Histona Acetiltransferasas/genética , Histona Acetiltransferasas/metabolismo , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Histonas/metabolismo , Neoplasias/genética , Sirtuinas/genética , Sirtuinas/metabolismo
6.
Int J Mol Sci ; 24(3)2023 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-36769195

RESUMEN

Cellular senescence-the irreversible cell cycle arrest driven by a variety of mechanisms and, more specifically, the senescence-associated secretory phenotype (SASP)-is an important area of research in the context of different age-related diseases, such as cardiovascular disease and cancer. SASP factors play both beneficial and detrimental roles in age-related disease progression depending on the source of the SASPs, the target cells, and the microenvironment. The impact of senescence and the SASP on different cell types, the immune system, and the vascular system has been widely discussed. However, the impact of replicative or stress-induced senescence on lymphatic biology and pathological lymphangiogenesis remains underexplored. The lymphatic system plays a crucial role in the maintenance of body fluid homeostasis and immune surveillance. The perturbation of lymphatic function can hamper normal physiological function. Natural aging or stress-induced premature aging influences the lymphatic vessel structure and function, which significantly affect the role of lymphatics in tumor dissemination and metastasis. In this review, we focus on the role of senescence on lymphatic pathobiology, its impact on cancer, and potential therapeutic interventions to manipulate the aged or senescent lymphatic system for disease management.


Asunto(s)
Senescencia Celular , Microambiente Tumoral , Humanos , Metástasis Linfática , Senescencia Celular/genética , Puntos de Control del Ciclo Celular
7.
Int J Mol Sci ; 23(14)2022 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-35887150

RESUMEN

Most living organisms have in their genome a sizable proportion of DNA sequences capable of mobilization; these sequences are commonly referred to as transposons, transposable elements (TEs), or jumping genes. Although long thought to have no biological significance, advances in DNA sequencing and analytical technologies have enabled precise characterization of TEs and confirmed their ubiquitous presence across all forms of life. These findings have ignited intense debates over their biological significance. The available evidence now supports the notion that TEs exert major influence over many biological aspects of organismal life. Transposable elements contribute significantly to the evolution of the genome by giving rise to genetic variations in both active and passive modes. Due to their intrinsic nature of mobility within the genome, TEs primarily cause gene disruption and large-scale genomic alterations including inversions, deletions, and duplications. Besides genomic instability, growing evidence also points to many physiologically important functions of TEs, such as gene regulation through cis-acting control elements and modulation of the transcriptome through epigenetic control. In this review, we discuss the latest evidence demonstrating the impact of TEs on genome stability and the underling mechanisms, including those developed to mitigate the deleterious impact of TEs on genomic stability and human health. We have also highlighted the potential therapeutic application of TEs.


Asunto(s)
Elementos Transponibles de ADN , Inestabilidad Genómica , Elementos Transponibles de ADN/genética , Evolución Molecular , Genómica , Humanos , Secuencias Reguladoras de Ácidos Nucleicos , Transcriptoma
8.
Chromosoma ; 129(3-4): 215-226, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32651609

RESUMEN

Heterochromatin protein 1ß (HP1ß), encoded by the Cbx1 gene, has been functionally linked to chromatin condensation, transcriptional regulation, and DNA damage repair. Here we report that testis-specific Cbx1 conditional knockout (Cbx1 cKO) impairs male germ cell development in mice. Depletion of HP1ß negatively affected sperm maturation and increased seminiferous tubule degeneration in Cbx1 cKO mice. In addition, the spermatogonia have elevated γ-H2AX foci levels as do Cbx1 deficient mouse embryonic fibroblasts (MEFs) as compared to wild-type (WT) control MEFs. The increase in γ-H2AX foci in proliferating Cbx1 cKO cells indicates defective replication-dependent DNA damage repair. Depletion or loss of HP1ß from human cells and MEFs increased DNA replication fork stalling and firing of new origins of replication, indicating defective DNA synthesis. Taken together, these results suggest that loss of HP1ß in proliferating cells leads to DNA replication defects with associated DNA damage that impact spermatogenesis.


Asunto(s)
Proteínas Cromosómicas no Histona/genética , Replicación del ADN , Regulación del Desarrollo de la Expresión Génica , Espermatogénesis/genética , Alelos , Animales , Apoptosis/genética , Línea Celular , Homólogo de la Proteína Chromobox 5 , Proteínas Cromosómicas no Histona/metabolismo , Replicación del ADN/efectos de los fármacos , Técnica del Anticuerpo Fluorescente , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Marcación de Gen , Sitios Genéticos , Histonas/metabolismo , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones Noqueados , Fenotipo , Maduración del Esperma/genética , Espermatogénesis/efectos de los fármacos , Espermatogonias/citología , Espermatogonias/metabolismo
9.
Haematologica ; 106(2): 495-512, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-32029507

RESUMEN

Ataxia telangiectasia mutated (ATM), a critical DNA damage sensor with protein kinase activity,is frequently altered in human cancers including mantle cell lymphoma (MCL). Loss of ATM protein is linked to accumulation of nonfunctional mitochondria and defective mitophagy, in both murine thymocytes and in A-T cells. However, the mechanistic role of ATM kinase in cancer cell mitophagy is unknown. Here, we provide evidence that FCCP-induced mitophagy in MCL and other cancer cell lines is dependent on ATM but independent of its kinase function. While Granta-519 MCL cells possess single copy and kinase dead ATM and are resistant to FCCP-induced mitophagy, both Jeko-1 and Mino cells are ATM proficient and induce mitophagy. Stable knockdown of ATM in Jeko-1 and Mino cells conferred resistance to mitophagy and was associated with reduced ATP production, oxygen consumption, and increased mROS. ATM interacts with the E3 ubiquitin ligase Parkin in a kinase-independent manner. Knockdown of ATM in HeLa cells resulted in proteasomal degradation of GFP-Parkin which was rescued by the proteasome inhibitor, MG132 suggesting that ATM-Parkin interaction is important for Parkin stability. Neither loss of ATM kinase activity in primary B cell lymphomas nor inhibition of ATM kinase in MCL, A-T and HeLa cell lines mitigated FCCP or CCCP-induced mitophagy suggesting that ATM kinase activity is dispensable for mitophagy. Malignant B-cell lymphomas without detectable ATM, Parkin, Pink1, and Parkin-Ub ser65 phosphorylation were resistant to mitophagy, providing the first molecular evidence of ATM's role in mitophagy in MCL and other B-cell lymphomas.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada , Ataxia Telangiectasia , Linfoma de Células del Manto , Adulto , Animales , Células HeLa , Humanos , Linfoma de Células del Manto/genética , Ratones , Mitofagia/genética , Fosforilación , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
10.
PLoS Genet ; 14(5): e1007300, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29795555

RESUMEN

Three waves of H2AX phosphorylation (γH2AX) have been observed in male meiotic prophase I: the first is ATM-dependent and occurs at leptonema, while the second and third are ATR-dependent, occuring at zygonema and pachynema, respectively. The third wave of H2AX phosphorylation marks and silences unsynapsed chromosomes. Little is known about H2AX phosphorylation expands to chromatin-wide regions in spermatocytes. Here, we report that histone acetyltransferase (HAT) MOF is involved in all three waves of H2AX phosphorylation expansion. Germ cell-specific deletion of Mof in spermatocytes by Stra8-Cre (Mof cKO) caused global loss of H4K16ac. In leptotene and zygotene spermatocytes of cKO mice, the γH2AX signals were observed only along the chromosomal axes, and chromatin-wide H2AX phosphorylation was lost. In almost 40% of early-mid pachytene spermatocytes from Mof cKO mice, γH2AX and MDC1 were detected along the unsynapsed axes of the sex chromosomes, but failed to expand, which consequently caused meiotic sex chromosome inactivation (MSCI) failure. Furthermore, though RAD51 was proficiently recruited to double-strand break (DSB) sites, defects in DSB repair and crossover formation were observed in Mof cKO spermatocytes, indicating that MOF facilitates meiotic DSB repair after RAD51 recruitment. We propose that MOF regulates male meiosis and is involved in the expansion of all three waves of H2AX phosphorylation from the leptotene to pachytene stages, initiated by ATM and ATR, respectively.


Asunto(s)
Histona Acetiltransferasas/metabolismo , Histonas/metabolismo , Espermatogénesis/fisiología , Animales , Roturas del ADN de Doble Cadena , Reparación del ADN , Femenino , Histona Acetiltransferasas/genética , Masculino , Meiosis , Ratones Noqueados , Ratones Transgénicos , Fase Paquiteno , Fosforilación , Recombinasa Rad51/genética , Recombinasa Rad51/metabolismo , Espermatocitos/citología , Testículo/fisiología
11.
J Biol Chem ; 294(37): 13619-13628, 2019 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-31320473

RESUMEN

Prion diseases are a group of incurable neurodegenerative disorders that affect humans and animals via infection with proteinaceous particles called prions. Prions are composed of PrPSc, a misfolded version of the cellular prion protein (PrPC). During disease progression, PrPSc replicates by interacting with PrPC and inducing its conversion to PrPSc As PrPSc accumulates, cellular stress mechanisms are activated to maintain cellular proteostasis, including increased protein chaperone levels. However, the exact roles of several of these chaperones remain unclear. Here, using various methodologies to monitor prion replication (i.e. protein misfolding cyclic amplification and cellular and animal infectivity bioassays), we studied the potential role of the molecular chaperone heat shock protein 70 (HSP70) in prion replication in vitro and in vivo Our results indicated that pharmacological induction of the heat shock response in cells chronically infected with prions significantly decreased PrPSc accumulation. We also found that HSP70 alters prion replication in vitro More importantly, prion infection of mice lacking the genes encoding stress-induced HSP70 exhibited accelerated prion disease progression compared with WT mice. In parallel with HSP70 being known to respond to endogenous and exogenous stressors such as heat, infection, toxicants, and ischemia, our results indicate that HSP70 may also play an important role in suppressing or delaying prion disease progression, opening opportunities for therapeutic intervention.


Asunto(s)
Proteínas HSP70 de Choque Térmico/metabolismo , Enfermedades por Prión/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/patología , Progresión de la Enfermedad , Retículo Endoplásmico/fisiología , Estrés del Retículo Endoplásmico/fisiología , Femenino , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP70 de Choque Térmico/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Priónicas/metabolismo , Priones/metabolismo , Pliegue de Proteína
12.
Development ; 144(12): 2165-2174, 2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-28506985

RESUMEN

Proper oocyte development is crucial for female fertility and requires timely and accurate control of gene expression. K (lysine) acetyltransferase 8 (KAT8), an important component of the X chromosome dosage compensation system in Drosophila, regulates gene activity by acetylating histone H4 preferentially at lysine 16. To explore the function of KAT8 during mouse oocyte development, we crossed Kat8flox/flox mice with Gdf9-Cre mice to specifically delete Kat8 in oocytes. Oocyte Kat8 deletion resulted in female infertility, with follicle development failure in the secondary and preantral follicle stages. RNA-seq analysis revealed that Kat8 deficiency in oocytes results in significant downregulation of antioxidant genes, with a consequent increase in reactive oxygen species. Intraperitoneal injection of the antioxidant N-acetylcysteine rescued defective follicle and oocyte development resulting from Kat8 deficiency. Chromatin immunoprecipitation assays indicated that KAT8 regulates antioxidant gene expression by direct binding to promoter regions. Taken together, our findings demonstrate that KAT8 is essential for female fertility by regulating antioxidant gene expression and identify KAT8 as the first histone acetyltransferase with an essential function in oogenesis.


Asunto(s)
Histona Acetiltransferasas/metabolismo , Oogénesis/fisiología , Especies Reactivas de Oxígeno/metabolismo , Animales , Antioxidantes/metabolismo , Apoptosis , Femenino , Fertilidad/genética , Fertilidad/fisiología , Regulación del Desarrollo de la Expresión Génica , Células de la Granulosa/metabolismo , Heterocromatina/genética , Heterocromatina/metabolismo , Histona Acetiltransferasas/deficiencia , Histona Acetiltransferasas/genética , Infertilidad Femenina/genética , Infertilidad Femenina/metabolismo , Infertilidad Femenina/patología , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Oocitos/citología , Oocitos/metabolismo , Oogénesis/genética , Folículo Ovárico/citología , Folículo Ovárico/metabolismo , Embarazo
13.
Hepatology ; 69(6): 2608-2622, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30693543

RESUMEN

Replication fork stability during DNA replication is vital for maintenance of genomic stability and suppression of cancer development in mammals. ATR (ataxia-telangiectasia mutated [ATM] and RAD3-related) is a master regulatory kinase that activates the replication stress response to overcome replication barriers. Although many downstream effectors of ATR have been established, the upstream regulators of ATR and the effect of such regulation on liver cancer remain unclear. The ubiquitin conjugase BRUCE (BIR Repeat containing Ubiquitin-Conjugating Enzyme) is a guardian of chromosome integrity and activator of ATM signaling, which promotes DNA double-strand break repair through homologous recombination. Here we demonstrate the functions for BRUCE in ATR activation in vitro and liver tumor suppression in vivo. BRUCE is recruited to induced DNA damage sites. Depletion of BRUCE inhibited multiple ATR-dependent signaling events during replication stress, including activation of ATR itself, phosphorylation of its downstream targets CHK1 and RPA, and the mono-ubiquitination of FANCD2. Consequently, BRUCE deficiency resulted in stalled DNA replication forks and increased firing of new replication origins. The in vivo impact of BRUCE loss on liver tumorigenesis was determined using the hepatocellular carcinoma model induced by genotoxin diethylnitrosamine. Liver-specific knockout of murine Bruce impaired ATR activation and exacerbated inflammation, fibrosis and hepatocellular carcinoma, which exhibited a trabecular architecture, closely resembling human hepatocellular carcinoma (HCC). In humans, the clinical relevance of BRUCE down-regulation in liver disease was found in hepatitis, cirrhosis, and HCC specimens, and deleterious somatic mutations of the Bruce gene was found in human hepatocellular carcinoma in the Cancer Genome Atlas database. Conclusion: These findings establish a BRUCE-ATR signaling axis in accurate DNA replication and suppression of liver cancer in mice and humans and provides a clinically relevant HCC mouse model.


Asunto(s)
Carcinoma Hepatocelular/genética , Replicación del ADN/genética , Proteínas Inhibidoras de la Apoptosis/genética , Neoplasias Hepáticas/genética , Transducción de Señal/genética , Animales , Proteínas de la Ataxia Telangiectasia Mutada/genética , Carcinogénesis , Carcinoma Hepatocelular/patología , Reparación del ADN/genética , Modelos Animales de Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Inestabilidad Genómica , Humanos , Neoplasias Hepáticas/patología , Masculino , Ratones , Ratones Noqueados , Distribución Aleatoria , Sensibilidad y Especificidad , Proteínas Supresoras de Tumor/genética
14.
Genet Med ; 21(1): 62-70, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-29895858

RESUMEN

PURPOSE: Fanconi anemia (FA) genes play important roles in spermatogenesis. In mice, disruption of Fancm impairs male fertility and testicular integrity, but whether FANCM pathogenic variants (PV) similarly affect fertility in men is unknown. Here we characterize a Pakistani family having three infertile brothers, two manifesting oligoasthenospermia and one exhibiting azoospermia, born to first-cousin parents. A homozygous PV in FANCM (c.1946_1958del, p.P648Lfs*16) was found cosegregating with male infertility. Our objective is to validate that FANCM p.P648Lfs*16 is the PV causing infertility in this family. METHODS: Exome and Sanger sequencing were used for PV screening. DNA interstrand crosslink (ICL) sensitivity was assessed in lymphocytes from patients. A mouse model carrying a PV nearly equivalent to that in the patients (FancmΔC/ΔC) was generated, followed by functional analysis in spermatogenesis. RESULTS: The loss-of-function FANCM PV increased ICL sensitivity in lymphocytes of patients and FancmΔC/ΔC spermatogonia. Adult FancmΔC/ΔC mice showed spermatogenic failure, with germ cell loss in 50.2% of testicular tubules and round-spermatid maturation arrest in 43.5% of tubules. In addition, neither bone marrow failure nor cancer/tumor was detected in all the patients or adult FancmΔC/ΔC mice. CONCLUSION: These findings revealed male infertility to be a novel phenotype of human patients with a biallelic FANCM PV.


Asunto(s)
ADN Helicasas/genética , Predisposición Genética a la Enfermedad , Infertilidad Masculina/genética , Espermatogénesis/genética , Adulto , Animales , Mutación del Sistema de Lectura , Homocigoto , Humanos , Infertilidad Masculina/patología , Mutación con Pérdida de Función/genética , Masculino , Ratones , Oligospermia/genética , Oligospermia/patología , Linaje , Fenotipo , Testículo/patología
15.
Genet Med ; 21(1): 266, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30158692

RESUMEN

Hao Win, Hui Ma and Sajjad Hussain were incorrectly affiliated to 'Department of Radiation Oncology, The Houston Methodist Research Institute, Houston, TX 77030 USA'. These authors should only have been affiliated to 'Hefei National Laboratory for Physical Sciences at Microscale, The First Affiliated Hospital of USTC, USTC-SJH Joint Center for Human Reproduction and Genetics, The CAS Key Laboratory of Innate Immunity and Chronic Diseases, School of Life Sciences, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center of Genetics and Development, University of Science and Technology of China, Hefei 230027, China'. They were also not noted as contributing equally to the paper. Both these errors have now been corrected in the PDF and HTML versions of the paper.

16.
Blood ; 129(1): 48-59, 2017 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-27827827

RESUMEN

K(lysine) acetyltransferase 8 (KAT8, also known as MOF) mediates the acetylation of histone H4 at lysine 16 (H4K16ac) and is crucial for murine embryogenesis. Lysine acetyltransferases have been shown to regulate various stages of normal hematopoiesis. However, the function of MOF in hematopoietic stem cell (HSC) development has not yet been elucidated. We set out to study the role of MOF in general hematopoiesis by using a Vav1-cre-induced conditional murine Mof knockout system and found that MOF is critical for hematopoietic cell maintenance and HSC engraftment capacity in adult hematopoiesis. Rescue experiments with a MOF histone acetyltransferase domain mutant illustrated the requirement for MOF acetyltransferase activity in the clonogenic capacity of HSCs and progenitors. In stark contrast, fetal steady-state hematopoiesis at embryonic day (E) 14.5 was not affected by homozygous Mof deletion despite dramatic loss of global H4K16ac. Hematopoietic defects start manifesting in late gestation at E17.5. The discovery that MOF and its H4K16ac activity are required for adult but not early and midgestational hematopoiesis supports the notion that multiple chromatin regulators may be crucial for hematopoiesis at varying stages of development. MOF is therefore a developmental-stage-specific chromatin regulator found to be essential for adult but not early fetal hematopoiesis.


Asunto(s)
Hematopoyesis/fisiología , Histona Acetiltransferasas/metabolismo , Animales , Feto , Células Madre Hematopoyéticas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
17.
Blood ; 129(18): 2479-2492, 2017 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-28270450

RESUMEN

Hematopoietic stem and progenitor cells (HSPCs) are vulnerable to endogenous damage and defects in DNA repair can limit their function. The 2 single-stranded DNA (ssDNA) binding proteins SSB1 and SSB2 are crucial regulators of the DNA damage response; however, their overlapping roles during normal physiology are incompletely understood. We generated mice in which both Ssb1 and Ssb2 were constitutively or conditionally deleted. Constitutive Ssb1/Ssb2 double knockout (DKO) caused early embryonic lethality, whereas conditional Ssb1/Ssb2 double knockout (cDKO) in adult mice resulted in acute lethality due to bone marrow failure and intestinal atrophy featuring stem and progenitor cell depletion, a phenotype unexpected from the previously reported single knockout models of Ssb1 or Ssb2 Mechanistically, cDKO HSPCs showed altered replication fork dynamics, massive accumulation of DNA damage, genome-wide double-strand breaks enriched at Ssb-binding regions and CpG islands, together with the accumulation of R-loops and cytosolic ssDNA. Transcriptional profiling of cDKO HSPCs revealed the activation of p53 and interferon (IFN) pathways, which enforced cell cycling in quiescent HSPCs, resulting in their apoptotic death. The rapid cell death phenotype was reproducible in in vitro cultured cDKO-hematopoietic stem cells, which were significantly rescued by nucleotide supplementation or after depletion of p53. Collectively, Ssb1 and Ssb2 control crucial aspects of HSPC function, including proliferation and survival in vivo by resolving replicative stress to maintain genomic stability.


Asunto(s)
Proliferación Celular/fisiología , Roturas del ADN de Doble Cadena , Inestabilidad Genómica/fisiología , Células Madre Hematopoyéticas/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Animales , Supervivencia Celular/fisiología , Islas de CpG/fisiología , Células Madre Hematopoyéticas/citología , Ratones , Ratones Noqueados , Proteínas Supresoras de la Señalización de Citocinas/genética , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
18.
Proc Natl Acad Sci U S A ; 112(11): E1210-9, 2015 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-25733871

RESUMEN

The DNA damage response (DDR) is crucial for genomic integrity. BRIT1 (breast cancer susceptibility gene C terminus-repeat inhibitor of human telomerase repeat transcriptase expression), a tumor suppressor and early DDR factor, is recruited to DNA double-strand breaks (DSBs) by phosphorylated H2A histone family, member X (γ-H2AX), where it promotes chromatin relaxation by recruiting the switch/sucrose nonfermentable (SWI-SNF) chromatin remodeler to facilitate DDR. However, regulation of BRIT1 recruitment is not fully understood. The baculovirus IAP repeat (BIR)-containing ubiquitin-conjugating enzyme (BRUCE) is an inhibitor of apoptosis protein (IAP). Here, we report a non-IAP function of BRUCE in the regulation of the BRIT1-SWI-SNF DSB-response pathway and genomic stability. We demonstrate that BRIT1 is K63 ubiquitinated in unstimulated cells and that deubiquitination of BRIT1 is a prerequisite for its recruitment to DSB sites by γ-H2AX. We show mechanistically that BRUCE acts as a scaffold, bridging the ubiquitin-specific peptidase 8 (USP8) and BRIT1 in a complex to coordinate USP8-catalyzed deubiquitination of BRIT1. Loss of BRUCE or USP8 impairs BRIT1 deubiquitination, BRIT1 binding with γ-H2AX, the formation of BRIT1 DNA damage foci, and chromatin relaxation. Moreover, BRUCE-depleted cells display reduced homologous recombination repair, and BRUCE-mutant mice exhibit repair defects and genomic instability. These findings identify BRUCE and USP8 as two hitherto uncharacterized critical DDR regulators and uncover a deubiquitination regulation of BRIT1 assembly at damaged chromatin for efficient DDR and genomic stability.


Asunto(s)
Roturas del ADN de Doble Cadena , Endopeptidasas/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Proteínas Inhibidoras de la Apoptosis/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Ubiquitinación , Animales , Proteínas de Ciclo Celular , Núcleo Celular/metabolismo , Cromatina/metabolismo , Cromosomas de los Mamíferos/metabolismo , Proteínas del Citoesqueleto , Reparación del ADN , Embrión de Mamíferos/citología , Fibroblastos/metabolismo , Inestabilidad Genómica , Células HEK293 , Histonas/metabolismo , Recombinación Homóloga/genética , Humanos , Lisina/metabolismo , Ratones , Complejos Multiproteicos/metabolismo , Poliubiquitina/metabolismo , Transporte de Proteínas
19.
PLoS Genet ; 11(1): e1004749, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25633985

RESUMEN

DNA strand-breaks (SBs) with non-ligatable ends are generated by ionizing radiation, oxidative stress, various chemotherapeutic agents, and also as base excision repair (BER) intermediates. Several neurological diseases have already been identified as being due to a deficiency in DNA end-processing activities. Two common dirty ends, 3'-P and 5'-OH, are processed by mammalian polynucleotide kinase 3'-phosphatase (PNKP), a bifunctional enzyme with 3'-phosphatase and 5'-kinase activities. We have made the unexpected observation that PNKP stably associates with Ataxin-3 (ATXN3), a polyglutamine repeat-containing protein mutated in spinocerebellar ataxia type 3 (SCA3), also known as Machado-Joseph Disease (MJD). This disease is one of the most common dominantly inherited ataxias worldwide; the defect in SCA3 is due to CAG repeat expansion (from the normal 14-41 to 55-82 repeats) in the ATXN3 coding region. However, how the expanded form gains its toxic function is still not clearly understood. Here we report that purified wild-type (WT) ATXN3 stimulates, and by contrast the mutant form specifically inhibits, PNKP's 3' phosphatase activity in vitro. ATXN3-deficient cells also show decreased PNKP activity. Furthermore, transgenic mice conditionally expressing the pathological form of human ATXN3 also showed decreased 3'-phosphatase activity of PNKP, mostly in the deep cerebellar nuclei, one of the most affected regions in MJD patients' brain. Finally, long amplicon quantitative PCR analysis of human MJD patients' brain samples showed a significant accumulation of DNA strand breaks. Our results thus indicate that the accumulation of DNA strand breaks due to functional deficiency of PNKP is etiologically linked to the pathogenesis of SCA3/MJD.


Asunto(s)
Enzimas Reparadoras del ADN/genética , Enfermedad de Machado-Joseph/genética , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Proteínas Represoras/genética , Expansión de Repetición de Trinucleótido/genética , Animales , Ataxina-3 , Línea Celular , Daño del ADN/genética , Reparación del ADN/genética , Enzimas Reparadoras del ADN/metabolismo , Humanos , Enfermedad de Machado-Joseph/enzimología , Enfermedad de Machado-Joseph/fisiopatología , Mamíferos , Ratones , Ratones Transgénicos , Mutación , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Estrés Oxidativo/genética , Fosforilación , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Proteínas Represoras/metabolismo
20.
J Biol Chem ; 290(41): 24636-48, 2015 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-26245904

RESUMEN

Why mammalian cells possess multiple DNA glycosylases (DGs) with overlapping substrate ranges for repairing oxidatively damaged bases via the base excision repair (BER) pathway is a long-standing question. To determine the biological role of these DGs, null animal models have been generated. Here, we report the generation and characterization of mice lacking Neil2 (Nei-like 2). As in mice deficient in each of the other four oxidized base-specific DGs (OGG1, NTH1, NEIL1, and NEIL3), Neil2-null mice show no overt phenotype. However, middle-aged to old Neil2-null mice show the accumulation of oxidative genomic damage, mostly in the transcribed regions. Immuno-pulldown analysis from wild-type (WT) mouse tissue showed the association of NEIL2 with RNA polymerase II, along with Cockayne syndrome group B protein, TFIIH, and other BER proteins. Chromatin immunoprecipitation analysis from mouse tissue showed co-occupancy of NEIL2 and RNA polymerase II only on the transcribed genes, consistent with our earlier in vitro findings on NEIL2's role in transcription-coupled BER. This study provides the first in vivo evidence of genomic region-specific repair in mammals. Furthermore, telomere loss and genomic instability were observed at a higher frequency in embryonic fibroblasts from Neil2-null mice than from the WT. Moreover, Neil2-null mice are much more responsive to inflammatory agents than WT mice. Taken together, our results underscore the importance of NEIL2 in protecting mammals from the development of various pathologies that are linked to genomic instability and/or inflammation. NEIL2 is thus likely to play an important role in long term genomic maintenance, particularly in long-lived mammals such as humans.


Asunto(s)
ADN Glicosilasas/deficiencia , ADN Glicosilasas/genética , ADN/metabolismo , Genoma/genética , Transcripción Genética , Envejecimiento/genética , Envejecimiento/metabolismo , Animales , Línea Celular , ADN/genética , Daño del ADN , Técnicas de Inactivación de Genes , Inestabilidad Genómica , Homeostasis , Humanos , Inflamación/genética , Inflamación/metabolismo , Ratones , Oxidación-Reducción , ARN Polimerasa II/metabolismo , Telómero/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA