Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Database (Oxford) ; 20242024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38805752

RESUMEN

Mechanical aspects of tissues and cells critically influence a myriad of biological processes and can substantially alter the course of diverse diseases. The emergence of diverse methodologies adapted from physical science now permits the precise quantification of the cellular forces and the mechanical properties of tissues and cells. Despite the rising interest in tissue and cellular mechanics across fields like biology, bioengineering and medicine, there remains a noticeable absence of a comprehensive and readily accessible repository of this pertinent information. To fill this gap, we present MechanoBase, a comprehensive tissue and cellular mechanics database, curating 57 480 records from 5634 PubMed articles. The records archived in MechanoBase encompass a range of mechanical properties and forces, such as modulus and tractions, which have been measured utilizing various technical approaches. These measurements span hundreds of biosamples across more than 400 species studied under diverse conditions. Aiming for broad applicability, we design MechanoBase with user-friendly search, browsing and data download features, making it a versatile tool for exploring biomechanical attributes in various biological contexts. Moreover, we add complementary resources, including the principles of popular techniques, the concepts of mechanobiology terms and the cellular and tissue-level expression of related genes, offering scientists unprecedented access to a wealth of knowledge in this field of research. Database URL: https://zhanglab-web.tongji.edu.cn/mechanobase/ and https://compbio-zhanglab.org/mechanobase/.


Asunto(s)
Bases de Datos Factuales , Humanos , Fenómenos Biomecánicos , Animales
2.
DNA Repair (Amst) ; 134: 103627, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38219597

RESUMEN

DNA double-strand breaks (DSBs) are harmful to mammalian cells and a few of them can cause cell death. Accumulating DSBs in these cells to analyze their genomic distribution and their potential impact on chromatin structure is difficult. In this study, we used CRISPR to generate Ku80-/- human cells and arrested the cells in G1 phase to accumulate DSBs before conducting END-seq and Nanopore analysis. Our analysis revealed that DNA with high methylation level accumulates DSB hotspots in Ku80-/- human cells. Furthermore, we identified chromosome structural variants (SVs) using Nanopore sequencing and observed a higher number of SVs in Ku80-/- human cells. Based on our findings, we suggest that the high efficiency of Ku80 knockout in human HCT116 cells makes it a promising model for characterizing SVs in the context of 3D chromatin structure and studying the alternative-end joining (Alt-EJ) DSB repair pathway.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN , Autoantígeno Ku , Animales , Humanos , Cromatina , ADN , Reparación del ADN por Unión de Extremidades , Reparación del ADN/genética , Células HCT116 , Autoantígeno Ku/genética , Autoantígeno Ku/metabolismo , Mamíferos/metabolismo
3.
Nat Struct Mol Biol ; 28(6): 487-500, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34117478

RESUMEN

Fanconi anemia (FA) is a devastating hereditary disease characterized by bone marrow failure (BMF) and acute myeloid leukemia (AML). As FA-deficient cells are hypersensitive to DNA interstrand crosslinks (ICLs), ICLs are widely assumed to be the lesions responsible for FA symptoms. Here, we show that FA-mutated cells are hypersensitive to persistent replication stress and that FA proteins play a role in the break-induced-replication (BIR)-like pathway for fork restart. Both the BIR-like pathway and ICL repair share almost identical molecular mechanisms of 53BP1-BRCA1-controlled signaling response, SLX4- and FAN1-mediated fork cleavage and POLD3-dependent DNA synthesis, suggesting that the FA pathway is intrinsically one of the BIR-like pathways. Replication stress not only triggers BMF in FA-deficient mice, but also specifically induces monosomy 7, which is associated with progression to AML in patients with FA, in FA-deficient cells.


Asunto(s)
Replicación del ADN , Proteínas del Grupo de Complementación de la Anemia de Fanconi/fisiología , Anemia de Fanconi/genética , Aneuploidia , Animales , Trastornos de Fallo de la Médula Ósea/etiología , Línea Celular Transformada , Pollos , Rotura Cromosómica , Deleción Cromosómica , Cromosomas Humanos Par 7/genética , ADN Polimerasa III/fisiología , Replicación del ADN/genética , Progresión de la Enfermedad , Anemia de Fanconi/metabolismo , Proteínas del Grupo de Complementación de la Anemia de Fanconi/deficiencia , Proteínas del Grupo de Complementación de la Anemia de Fanconi/genética , Femenino , Células HCT116 , Células HEK293 , Humanos , Hidroxiurea/farmacología , Leucemia Mieloide Aguda/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Genéticos , Especificidad de la Especie , Proteína 1 de Unión al Supresor Tumoral P53/fisiología , Ubiquitina-Proteína Ligasas/fisiología
4.
Biomolecules ; 9(12)2019 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-31795137

RESUMEN

Classical non-homologous end joining (NHEJ) is a molecular pathway that detects, processes, and ligates DNA double-strand breaks (DSBs) throughout the cell cycle. Mutations in several NHEJ genes result in neurological abnormalities and immunodeficiency both in humans and mice. The NHEJ pathway is required for V(D)J recombination in developing B and T lymphocytes, and for class switch recombination in mature B cells. The Ku heterodimer formed by Ku70 and Ku80 recognizes DSBs and facilitates the recruitment of accessory factors (e.g., DNA-PKcs, Artemis, Paxx and Mri/Cyren) and downstream core factor subunits X-ray repair cross-complementing group 4 (XRCC4), XRCC4-like factor (XLF), and DNA ligase 4 (Lig4). Accessory factors might be dispensable for the process, depending on the genetic background and DNA lesion type. To determine the physiological role of Mri in DNA repair and development, we introduced a frame-shift mutation in the Mri gene in mice. We then analyzed the development of Mri-deficient mice as well as wild type and immunodeficient controls. Mice lacking Mri possessed reduced levels of class switch recombination in B lymphocytes and slow proliferation of neuronal progenitors when compared to wild type littermates. Human cell lines lacking Mri were as sensitive to DSBs as the wild type controls. Overall, we concluded that Mri/Cyren is largely dispensable for DNA repair and mouse development.


Asunto(s)
Reparación del ADN por Unión de Extremidades/genética , Ratones Noqueados , Animales , Linfocitos B/inmunología , Línea Celular , Proliferación Celular , Enzimas Reparadoras del ADN/genética , Proteínas de Unión al ADN/genética , Humanos , Cambio de Clase de Inmunoglobulina , Inmunoglobulina G/inmunología , Modelos Animales , Células Madre , Linfocitos T/inmunología
5.
FEBS Open Bio ; 9(7): 1315-1326, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31141305

RESUMEN

DNA double-strand breaks (DSBs) are highly cytotoxic lesions, and unrepaired or misrepaired DSBs can lead to various human diseases, including immunodeficiency, neurological abnormalities, growth retardation, and cancer. Nonhomologous end joining (NHEJ) is the major DSB repair pathway in mammals. Ku70 and Ku80 are DSB sensors that facilitate the recruitment of downstream factors, including protein kinase DNA-dependent protein kinase, catalytic subunit (DNA-PKcs), structural components [X-ray repair cross-complementing protein 4 (XRCC4), XRCC4-like factor (XLF), and paralogue of XRCC4 and XLF (PAXX)], and DNA ligase IV (LIG4), which complete DNA repair. DSBs also trigger the activation of the DNA damage response pathway, in which protein kinase ataxia-telangiectasia mutated (ATM) phosphorylates multiple substrates, including histone H2AX. Traditionally, research on NHEJ factors was performed using in vivo mouse models and murine cells. However, the current knowledge of the genetic interactions between NHEJ factors in human cells is incomplete. Here, we obtained genetically modified human HAP1 cell lines, which lacked one or two NHEJ factors, including LIG4, XRCC4, XLF, PAXX, DNA-PKcs, DNA-PKcs/XRCC4, and DNA-PKcs/PAXX. We examined the genomic instability of HAP1 cells, as well as their sensitivity to DSB-inducing agents. In addition, we determined the genetic interaction between XRCC4 paralogues (XRCC4, XLF, and PAXX) and DNA-PKcs. We found that in human cells, XLF, but not PAXX or XRCC4, genetically interacts with DNA-PKcs. Moreover, ATM possesses overlapping functions with DNA-PKcs, XLF, and XRCC4, but not with PAXX in response to DSBs. Finally, NHEJ-deficient HAP1 cells show increased chromosomal and chromatid breaks, when compared to the WT parental control. Overall, we found that HAP1 is a suitable model to study the genetic interactions in human cells.


Asunto(s)
Enzimas Reparadoras del ADN/genética , Enzimas Reparadoras del ADN/metabolismo , Proteína Quinasa Activada por ADN/genética , Proteínas de la Ataxia Telangiectasia Mutada/genética , Dominio Catalítico/genética , Línea Celular , ADN/genética , Roturas del ADN de Doble Cadena , Reparación del ADN por Unión de Extremidades/genética , Metilación de ADN/genética , Reparación del ADN/genética , Proteína Quinasa Activada por ADN/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Epistasis Genética/genética , Humanos , Autoantígeno Ku/genética , Proteínas del Tejido Nervioso/genética , Fosforilación
6.
Biomolecules ; 10(1)2019 12 30.
Artículo en Inglés | MEDLINE | ID: mdl-31905950

RESUMEN

DNA double-strand breaks (DSBs) trigger the Ataxia telangiectasia mutated (ATM)-dependent DNA damage response (DDR), which consists of histone H2AX, MDC1, RNF168, 53BP1, PTIP, RIF1, Rev7, and Shieldin. Early stages of B and T lymphocyte development are dependent on recombination activating gene (RAG)-induced DSBs that form the basis for further V(D)J recombination. Non-homologous end joining (NHEJ) pathway factors recognize, process, and ligate DSBs. Based on numerous loss-of-function studies, DDR factors were thought to be dispensable for the V(D)J recombination. In particular, mice lacking Mediator of DNA Damage Checkpoint Protein 1 (MDC1) possessed nearly wild-type levels of mature B and T lymphocytes in the spleen, thymus, and bone marrow. NHEJ factor XRCC4-like factor (XLF)/Cernunnos is functionally redundant with ATM, histone H2AX, and p53-binding protein 1 (53BP1) during the lymphocyte development in mice. Here, we genetically inactivated MDC1, XLF, or both MDC1 and XLF in murine vAbl pro-B cell lines and, using chromosomally integrated substrates, demonstrated that MDC1 stimulates the V(D)J recombination in cells lacking XLF. Moreover, combined inactivation of MDC1 and XLF in mice resulted in synthetic lethality. Together, these findings suggest that MDC1 and XLF are functionally redundant during the mouse development, in general, and the V(D)J recombination, in particular.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas de Unión al ADN/deficiencia , Recombinación V(D)J , Animales , Línea Celular , Proliferación Celular , Daño del ADN , Reparación del ADN , Proteínas de Unión al ADN/genética , Ratones , Ratones Noqueados , Ratones Transgénicos
7.
DNA Repair (Amst) ; 73: 164-169, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30579708

RESUMEN

Non-homologous end joining (NHEJ) is a DNA repair pathway that senses, processes and ligates DNA double-strand breaks (DSBs) throughout the cell cycle. During NHEJ, core Ku70 and Ku80 subunits bind DSBs as a heterodimer and promote further recruitment of accessory factors (e.g., PAXX, Mri, DNA-PKcs, Artemis) and downstream core subunits XRCC4 and DNA ligase 4 (Lig4). Inactivation of Ku70 or Ku80 genes in mice results in immunodeficiency and high levels of genomic instability; deletion of individual Dna-pkcs, Xlf, Paxx or Mri genes results in viable mice with no or modest DNA repair defects. However, combined inactivation of either Xlf and Dna-pkcs, or Xlf and Paxx, or Xlf and Mri, leads to synthetic lethality in mice, which correlates with increased levels of apoptosis in the central nervous system. Here, we demonstrated that inactivation of pro-apoptotic factor Trp53 rescues embryonic lethality of Xlf-/-Paxx-/- and Xlf-/-Dna-pkcs-/- double knockout mice. Moreover, combined inactivation of Paxx and Dna-pkcs results in live-born fertile Paxx-/-Dna-pkcs-/- mice indistinguishable from Dna-pkcs-/- knockout controls.


Asunto(s)
Reparación del ADN , Proteínas de Unión al ADN/genética , Silenciador del Gen , Mutaciones Letales Sintéticas , Proteína p53 Supresora de Tumor/deficiencia , Proteína p53 Supresora de Tumor/genética , Animales , Línea Celular , Enzimas Reparadoras del ADN/genética , Proteína Quinasa Activada por ADN/deficiencia , Proteína Quinasa Activada por ADN/genética , Proteínas de Unión al ADN/deficiencia , Técnicas de Inactivación de Genes , Humanos , Ratones , Proteínas Nucleares/deficiencia , Proteínas Nucleares/genética
8.
FEBS Open Bio ; 8(3): 426-434, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29511619

RESUMEN

DNA repair consists of several cellular pathways which recognize and repair damaged DNA. The classical nonhomologous DNA end-joining (NHEJ) pathway repairs double-strand breaks in DNA. It is required for maturation of both B and T lymphocytes by supporting V(D)J recombination as well as B-cell differentiation during class switch recombination (CSR). Inactivation of NHEJ factors Ku70, Ku80, XRCC4, DNA ligase 4, DNA-PKcs, and Artemis impairs V(D)J recombination and blocks lymphocyte development. Paralogue of XRCC4 and XLF (PAXX) is an accessory NHEJ factor that has a significant impact on the repair of DNA lesions induced by ionizing radiation in human, murine, and chicken cells. However, the role of PAXX during development is poorly understood. To determine the physiological role of PAXX, we deleted part of the Paxx promoter and the first two exons in mice. Further, we compared Paxx-knockout mice with wild-type (WT) and NHEJ-deficient controls including Ku80- and Dna-pkcs-null and severe combined immunodeficiency mice. Surprisingly, Paxx-deficient mice were not distinguishable from the WT littermates; they were the same weight and size, fertility status, had normal spleen, thymus, and bone marrow. Paxx-deficient mice had the same number of chromosomal and chromatid breaks as WT mice. Moreover, Paxx-deficient primary B lymphocytes had the same level of CSR as lymphocytes isolated from WT mice. We concluded that PAXX is dispensable for normal mouse development.

9.
FEBS Open Bio ; 8(3): 442-448, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29511621

RESUMEN

To ensure genome stability, mammalian cells employ several DNA repair pathways. Nonhomologous DNA end joining (NHEJ) is the DNA repair process that fixes double-strand breaks throughout the cell cycle. NHEJ is involved in the development of B and T lymphocytes through its function in V(D)J recombination and class switch recombination (CSR). NHEJ consists of several core and accessory factors, including Ku70, Ku80, XRCC4, DNA ligase 4, DNA-PKcs, Artemis, and XLF. Paralog of XRCC4 and XLF (PAXX) is the recently described accessory NHEJ factor that structurally resembles XRCC4 and XLF and interacts with Ku70/Ku80. To determine the physiological role of PAXX in mammalian cells, we purchased and characterized a set of custom-generated and commercially available NHEJ-deficient human haploid HAP1 cells, PAXXΔ, XRCC4Δ , and XLFΔ . In our studies, HAP1 PAXXΔ cells demonstrated modest sensitivity to DNA damage, which was comparable to wild-type controls. By contrast, XRCC4Δ and XLFΔ HAP1 cells possessed significant DNA repair defects measured as sensitivity to double-strand break inducing agents and chromosomal breaks. To investigate the role of PAXX in CSR, we generated and characterized Paxx-/- and Aid-/- murine lymphoid CH12F3 cells. CSR to IgA was nearly at wild-type levels in the Paxx-/- cells and completely ablated in the absence of activation-induced cytidine deaminase (AID). In addition, Paxx-/- CH12F3 cells were hypersensitive to zeocin when compared to wild-type controls. We concluded that Paxx-deficient mammalian cells maintain robust NHEJ and CSR.

10.
Elife ; 62017 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-29106372

RESUMEN

The cellular pathways that restart stalled replication forks are essential for genome stability and tumor prevention. However, how many of these pathways exist in cells and how these pathways are selectively activated remain unclear. Here, we describe two major fork restart pathways, and demonstrate that their selection is governed by 53BP1 and BRCA1, which are known to control the pathway choice to repair double-strand DNA breaks (DSBs). Specifically, 53BP1 promotes a fork cleavage-free pathway, whereas BRCA1 facilitates a break-induced replication (BIR) pathway coupled with SLX-MUS complex-mediated fork cleavage. The defect in the first pathway, but not DSB repair, in a 53BP1 mutant is largely corrected by disrupting BRCA1, and vice versa. Moreover, PLK1 temporally regulates the switch of these two pathways through enhancing the assembly of the SLX-MUS complex. Our results reveal two distinct fork restart pathways, which are antagonistically controlled by 53BP1 and BRCA1 in a DSB repair-independent manner.


Asunto(s)
Proteína BRCA1/metabolismo , Replicación del ADN , Proteína 1 de Unión al Supresor Tumoral P53/metabolismo , Línea Celular , Roturas del ADN de Doble Cadena , Reparación del ADN , Humanos
11.
DNA Repair (Amst) ; 57: 133-138, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28759779

RESUMEN

DNA double-strand breaks (DSBs) are recognized and repaired by the Classical Non-Homologous End-Joining (C-NHEJ) and Homologous Recombination pathways. C-NHEJ includes the core Ku70 and Ku80 (or Ku86) heterodimer that binds DSBs and thus promotes recruitment of accessory downstream NHEJ factors XLF, PAXX, DNA-PKcs, Artemis and other core subunits, XRCC4 and DNA Ligase 4 (Lig4). In the absence of core C-NHEJ factors, DNA repair can be performed by Alternative End-Joining, which likely depends on DNA Ligase 1 and DNA Ligase 3. Genetic inactivation of C-NHEJ factors, such as Ku70, Ku80, XLF, PAXX and DNA-PKcs results in viable mice showing increased levels of genomic instability and sensitivity to DSBs. Knockouts of XRCC4 or Lig4, on the other hand, as well as combined inactivation of XLF and DNA-PKcs, or XLF and PAXX, result in late embryonic lethality in mice, which in most cases correlate with severe apoptosis in the central nervous system. Here, we demonstrate that inactivation of the Ku70 gene rescues the synthetic lethality between XLF and DNA-PKcs, resulting in triple knockout mice that are indistinguishable from Ku70-deficient littermates by size or levels of genomic instability. Moreover, we find that combined inactivation of Ku70 and XLF results in viable mice. Together, these findings suggest that Ku70 is epistatic with XLF and DNA-PKcs and support a model in which inactivation of Ku70 allows DNA lesions to become accessible to alternative DNA repair pathways.


Asunto(s)
Reparación del ADN por Unión de Extremidades , Proteína Quinasa Activada por ADN/genética , Proteínas de Unión al ADN/genética , Epistasis Genética , Autoantígeno Ku/genética , Proteínas Nucleares/genética , Mutaciones Letales Sintéticas , Animales , ADN/metabolismo , Roturas del ADN de Doble Cadena , Proteína Quinasa Activada por ADN/metabolismo , Proteínas de Unión al ADN/metabolismo , Autoantígeno Ku/metabolismo , Ratones , Ratones Noqueados , Proteínas Nucleares/metabolismo
12.
Nat Commun ; 6: 6233, 2015 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-25670504

RESUMEN

Non-homologous end joining (NHEJ) is a major pathway to repair DNA double-strand breaks (DSBs), which can display different types of broken ends. However, it is unclear how NHEJ factors organize to repair diverse types of DNA breaks. Here, through systematic analysis of the human NHEJ factor interactome, we identify PAXX as a direct interactor of Ku. The crystal structure of PAXX is similar to those of XRCC4 and XLF. Importantly, PAXX-deficient cells are sensitive to DSB-causing agents. Moreover, epistasis analysis demonstrates that PAXX functions together with XLF in response to ionizing radiation-induced complex DSBs, whereas they function redundantly in response to Topo2 inhibitor-induced simple DSBs. Consistently, PAXX and XLF coordinately promote the ligation of complex but not simple DNA ends in vitro. Altogether, our data identify PAXX as a new NHEJ factor and provide insight regarding the organization of NHEJ factors responding to diverse types of DSB ends.


Asunto(s)
Reparación del ADN por Unión de Extremidades , Proteínas de Unión al ADN/metabolismo , Mapeo de Interacción de Proteínas , Homología de Secuencia de Aminoácido , Animales , Antígenos Nucleares/metabolismo , Línea Celular , Pollos , Cristalografía por Rayos X , Roturas del ADN de Doble Cadena , ADN Ligasa (ATP) , ADN Ligasas/metabolismo , Enzimas Reparadoras del ADN/metabolismo , Proteínas de Unión al ADN/química , Células HEK293 , Humanos , Autoantígeno Ku , Espectrometría de Masas , Modelos Biológicos , Modelos Moleculares , Unión Proteica , Estructura Terciaria de Proteína , Homología Estructural de Proteína
13.
Mol Cell ; 49(5): 872-83, 2013 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-23333306

RESUMEN

DNA double-strand break (DSB) repair pathway choice is governed by the opposing activities of 53BP1 and BRCA1. 53BP1 stimulates nonhomologous end joining (NHEJ), whereas BRCA1 promotes end resection and homologous recombination (HR). Here we show that 53BP1 is an inhibitor of BRCA1 accumulation at DSB sites, specifically in the G1 phase of the cell cycle. ATM-dependent phosphorylation of 53BP1 physically recruits RIF1 to DSB sites, and we identify RIF1 as the critical effector of 53BP1 during DSB repair. Remarkably, RIF1 accumulation at DSB sites is strongly antagonized by BRCA1 and its interacting partner CtIP. Lastly, we show that depletion of RIF1 is able to restore end resection and RAD51 loading in BRCA1-depleted cells. This work therefore identifies a cell cycle-regulated circuit, underpinned by RIF1 and BRCA1, that governs DSB repair pathway choice to ensure that NHEJ dominates in G1 and HR is favored from S phase onward.


Asunto(s)
Proteína BRCA1/genética , Proteínas Portadoras/genética , Ciclo Celular/genética , Reparación del ADN , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas Nucleares/genética , Proteínas de Unión a Telómeros/genética , Proteína BRCA1/metabolismo , Sitios de Unión , Proteínas Portadoras/metabolismo , Reparación del ADN por Unión de Extremidades/genética , Endodesoxirribonucleasas , Células HEK293 , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Nucleares/metabolismo , Fase S , Proteínas de Unión a Telómeros/metabolismo , Proteína 1 de Unión al Supresor Tumoral P53
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...