Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
1.
Blood ; 143(26): 2749-2762, 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38498025

ABSTRACT

ABSTRACT: Identifying and targeting microenvironment-driven pathways that are active across acute myeloid leukemia (AML) genetic subtypes should allow the development of more broadly effective therapies. The proinflammatory cytokine interleukin-1ß (IL-1ß) is abundant in the AML microenvironment and promotes leukemic growth. Through RNA-sequencing analysis, we identify that IL-1ß-upregulated ASF1B (antisilencing function-1B), a histone chaperone, in AML progenitors compared with healthy progenitors. ASF1B, along with its paralogous protein ASF1A, recruits H3-H4 histones onto the replication fork during S-phase, a process regulated by Tousled-like kinase 1 and 2 (TLKs). Although ASF1s and TLKs are known to be overexpressed in multiple solid tumors and associated with poor prognosis, their functional roles in hematopoiesis and inflammation-driven leukemia remain unexplored. In this study, we identify that ASF1s and TLKs are overexpressed in multiple genetic subtypes of AML. We demonstrate that depletion of ASF1s significantly reduces leukemic cell growth in both in vitro and in vivo models using human cells. Using a murine model, we show that overexpression of ASF1B accelerates leukemia progression. Moreover, Asf1b or Tlk2 deletion delayed leukemia progression, whereas these proteins are dispensable for normal hematopoiesis. Through proteomics and phosphoproteomics analyses, we uncover that the TLK-ASF1 pathway promotes leukemogenesis by affecting the cell cycle and DNA damage pathways. Collectively, our findings identify the TLK1-ASF1 pathway as a novel mediator of inflammatory signaling and a promising therapeutic target for AML treatment across diverse genetic subtypes. Selective inhibition of this pathway offers potential opportunities to intervene effectively, address intratumoral heterogeneity, and ultimately improve clinical outcomes in AML.


Subject(s)
Cell Cycle Proteins , Disease Progression , Interleukin-1beta , Leukemia, Myeloid, Acute , Protein Serine-Threonine Kinases , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Leukemia, Myeloid, Acute/genetics , Humans , Animals , Mice , Interleukin-1beta/metabolism , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Signal Transduction , Molecular Chaperones/metabolism , Molecular Chaperones/genetics , Histone Chaperones/metabolism , Histone Chaperones/genetics , Histones/metabolism , Histones/genetics , Cell Line, Tumor , Serine-Arginine Splicing Factors/metabolism , Serine-Arginine Splicing Factors/genetics
2.
EMBO J ; 40(1): e106118, 2021 01 04.
Article in English | MEDLINE | ID: mdl-33226141

ABSTRACT

Mutations in centrosome genes deplete neural progenitor cells (NPCs) during brain development, causing microcephaly. While NPC attrition is linked to TP53-mediated cell death in several microcephaly models, how TP53 is activated remains unclear. In cultured cells, mitotic delays resulting from centrosome loss prevent the growth of unfit daughter cells by activating a pathway involving 53BP1, USP28, and TP53, termed the mitotic surveillance pathway. Whether this pathway is active in the developing brain is unknown. Here, we show that the depletion of centrosome proteins in NPCs prolongs mitosis and increases TP53-mediated apoptosis. Cell death after a delayed mitosis was rescued by inactivation of the mitotic surveillance pathway. Moreover, 53BP1 or USP28 deletion restored NPC proliferation and brain size without correcting the upstream centrosome defects or extended mitosis. By contrast, microcephaly caused by the loss of the non-centrosomal protein SMC5 is also TP53-dependent but is not rescued by loss of 53BP1 or USP28. Thus, we propose that mutations in centrosome genes cause microcephaly by delaying mitosis and pathologically activating the mitotic surveillance pathway in the developing brain.


Subject(s)
Centrosome/metabolism , Microcephaly/genetics , Microcephaly/metabolism , Mitosis/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor p53-Binding Protein 1/genetics , Ubiquitin Thiolesterase/genetics , Animals , Apoptosis/genetics , Brain/pathology , Cell Death/genetics , Cell Proliferation/genetics , Cells, Cultured , Mice , Mice, Knockout , Mutation/genetics , Signal Transduction/genetics
3.
Nucleic Acids Res ; 51(13): 6754-6769, 2023 07 21.
Article in English | MEDLINE | ID: mdl-37309898

ABSTRACT

The Sirtuin family of NAD+-dependent enzymes plays an important role in maintaining genome stability upon stress. Several mammalian Sirtuins have been linked directly or indirectly to the regulation of DNA damage during replication through Homologous recombination (HR). The role of one of them, SIRT1, is intriguing as it seems to have a general regulatory role in the DNA damage response (DDR) that has not yet been addressed. SIRT1-deficient cells show impaired DDR reflected in a decrease in repair capacity, increased genome instability and decreased levels of γH2AX. Here we unveil a close functional antagonism between SIRT1 and the PP4 phosphatase multiprotein complex in the regulation of the DDR. Upon DNA damage, SIRT1 interacts specifically with the catalytical subunit PP4c and promotes its inhibition by deacetylating the WH1 domain of the regulatory subunits PP4R3α/ß. This in turn regulates γH2AX and RPA2 phosphorylation, two key events in the signaling of DNA damage and repair by HR. We propose a mechanism whereby during stress, SIRT1 signaling ensures a global control of DNA damage signaling through PP4.


Subject(s)
DNA Damage , Sirtuin 1 , Animals , Humans , Mammals/metabolism , Phosphoric Monoester Hydrolases , Phosphorylation , Signal Transduction , Sirtuin 1/metabolism
4.
PLoS Biol ; 19(3): e3001176, 2021 03.
Article in English | MEDLINE | ID: mdl-33788831

ABSTRACT

Analysis of cancer mutagenic signatures provides information about the origin of mutations and can inform the use of clinical therapies, including immunotherapy. In particular, APOBEC3A (A3A) has emerged as a major driver of mutagenesis in cancer cells, and its expression results in DNA damage and susceptibility to treatment with inhibitors of the ATR and CHK1 checkpoint kinases. Here, we report the implementation of CRISPR/Cas-9 genetic screening to identify susceptibilities of multiple A3A-expressing lung adenocarcinoma (LUAD) cell lines. We identify HMCES, a protein recently linked to the protection of abasic sites, as a central protein for the tolerance of A3A expression. HMCES depletion results in synthetic lethality with A3A expression preferentially in a TP53-mutant background. Analysis of previous screening data reveals a strong association between A3A mutational signatures and sensitivity to HMCES loss and indicates that HMCES is specialized in protecting against a narrow spectrum of DNA damaging agents in addition to A3A. We experimentally show that both HMCES disruption and A3A expression increase susceptibility of cancer cells to ionizing radiation (IR), oxidative stress, and ATR inhibition, strategies that are often applied in tumor therapies. Overall, our results suggest that HMCES is an attractive target for selective treatment of A3A-expressing tumors.


Subject(s)
Adenocarcinoma of Lung/genetics , Cytidine Deaminase/genetics , DNA-Binding Proteins/genetics , Proteins/genetics , Adenocarcinoma of Lung/metabolism , Ataxia Telangiectasia Mutated Proteins/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Checkpoint Kinase 1/metabolism , Cytidine Deaminase/metabolism , Cytosine Deaminase/genetics , Cytosine Deaminase/metabolism , DNA/genetics , DNA/metabolism , DNA Damage/genetics , DNA Damage/physiology , DNA Replication/genetics , DNA Replication/physiology , DNA-Binding Proteins/metabolism , Humans , Proteins/metabolism
5.
Semin Cell Dev Biol ; 110: 51-60, 2021 02.
Article in English | MEDLINE | ID: mdl-32362381

ABSTRACT

Multiciliated cells (MCC) project dozens to hundreds of motile cilia from the cell surface to generate fluid flow across epithelial surfaces or turbulence to promote the transport of gametes. The MCC differentiation program is initiated by GEMC1 and MCIDAS, members of the geminin family, that activate key transcription factors, including p73 and FOXJ1, to control the multiciliogenesis program. To support the generation of multiple motile cilia, MCCs must undergo massive centriole amplification to generate a sufficient number of basal bodies (modified centrioles). This transcriptional program involves the generation of deuterosomes, unique structures that act as platforms to regulate centriole amplification, the reactivation of cell cycle programs to control centriole amplification and release, and extensive remodeling of the cytoskeleton. This review will focus on providing an overview of the transcriptional regulation of MCCs and its connection to key processes, in addition to highlighting exciting recent developments and open questions in the field.


Subject(s)
Cell Cycle Proteins/genetics , Centrioles/metabolism , Cilia/metabolism , Ciliopathies/genetics , Transcription Factors/genetics , Transcription, Genetic , Animals , Cell Cycle/genetics , Cell Cycle Proteins/metabolism , Cell Differentiation , Centrioles/ultrastructure , Cilia/ultrastructure , Ciliopathies/metabolism , Ciliopathies/pathology , Cytoskeleton/metabolism , Cytoskeleton/ultrastructure , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Humans , Signal Transduction , Transcription Factors/metabolism , Tumor Protein p73/genetics , Tumor Protein p73/metabolism
6.
Nat Rev Mol Cell Biol ; 12(2): 90-103, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21252998

ABSTRACT

The maintenance of genome stability depends on the DNA damage response (DDR), which is a functional network comprising signal transduction, cell cycle regulation and DNA repair. The metabolism of DNA double-strand breaks governed by the DDR is important for preventing genomic alterations and sporadic cancers, and hereditary defects in this response cause debilitating human pathologies, including developmental defects and cancer. The MRE11 complex, composed of the meiotic recombination 11 (MRE11), RAD50 and Nijmegen breakage syndrome 1 (NBS1; also known as nibrin) proteins is central to the DDR, and recent insights into its structure and function have been gained from in vitro structural analysis and studies of animal models in which the DDR response is deficient.


Subject(s)
DNA Repair , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Animals , DNA Breaks, Double-Stranded , DNA Repair Enzymes/chemistry , DNA Repair Enzymes/metabolism , Humans , Pyrococcus furiosus/chemistry , Pyrococcus furiosus/metabolism
7.
J Med Genet ; 59(2): 170-179, 2022 02.
Article in English | MEDLINE | ID: mdl-33323470

ABSTRACT

INTRODUCTION: The Tousled-like kinases 1 and 2 (TLK1 and TLK2) are involved in many fundamental processes, including DNA replication, cell cycle checkpoint recovery and chromatin remodelling. Mutations in TLK2 were recently associated with 'Mental Retardation Autosomal Dominant 57' (MRD57, MIM# 618050), a neurodevelopmental disorder characterised by a highly variable phenotype, including mild-to-moderate intellectual disability, behavioural abnormalities, facial dysmorphisms, microcephaly, epilepsy and skeletal anomalies. METHODS: We re-evaluate whole exome sequencing and array-CGH data from a large cohort of patients affected by neurodevelopmental disorders. Using spatial proteomics (BioID) and single-cell gel electrophoresis, we investigated the proximity interaction landscape of TLK2 and analysed the effects of p.(Asp551Gly) and a previously reported missense variant (c.1850C>T; p.(Ser617Leu)) on TLK2 interactions, localisation and activity. RESULTS: We identified three new unrelated MRD57 families. Two were sporadic and caused by a missense change (c.1652A>G; p.(Asp551Gly)) or a 39 kb deletion encompassing TLK2, and one was familial with three affected siblings who inherited a nonsense change from an affected mother (c.1423G>T; p.(Glu475Ter)). The clinical phenotypes were consistent with those of previously reported cases. The tested mutations strongly impaired TLK2 kinase activity. Proximal interactions between TLK2 and other factors implicated in neurological disorders, including CHD7, CHD8, BRD4 and NACC1, were identified. Finally, we demonstrated a more relaxed chromatin state in lymphoblastoid cells harbouring the p.(Asp551Gly) variant compared with control cells, conferring susceptibility to DNA damage. CONCLUSION: Our study identified novel TLK2 pathogenic variants, confirming and further expanding the MRD57-related phenotype. The molecular characterisation of missense variants increases our knowledge about TLK2 function and provides new insights into its role in neurodevelopmental disorders.


Subject(s)
Chromatin/metabolism , Neurodevelopmental Disorders/genetics , Protein Kinases/genetics , Adolescent , Adult , Child , Child, Preschool , Cohort Studies , DNA Mutational Analysis , Female , Humans , Male , Metabolome , Middle Aged , Mutation , Mutation, Missense , Neurodevelopmental Disorders/enzymology , Pedigree , Protein Interaction Mapping , Protein Kinases/metabolism , Exome Sequencing , Young Adult
8.
Development ; 146(8)2019 04 23.
Article in English | MEDLINE | ID: mdl-30936178

ABSTRACT

GEMC1 and MCIDAS are geminin family proteins that transcriptionally activate E2F4/5-target genes during multiciliogenesis, including Foxj1 and Ccno Male mice that lacked Gemc1, Mcidas or Ccno were found to be infertile, but the origin of this defect has remained unclear. Here, we show that all three genes are necessary for the generation of functional multiciliated cells in the efferent ducts that are required for spermatozoa to enter the epididymis. In mice that are mutant for Gemc1, Mcidas or Ccno, we observed a similar spectrum of phenotypes, including thinning of the seminiferous tubule epithelia, dilation of the rete testes, sperm agglutinations in the efferent ducts and lack of spermatozoa in the epididymis (azoospermia). These data suggest that defective efferent duct development is the dominant cause of male infertility in these mouse models, and this likely extends to individuals with the ciliopathy reduced generation of multiple motile cilia with mutations in MCIDAS and CCNO.


Subject(s)
Cell Cycle Proteins/deficiency , DNA Glycosylases/deficiency , Ejaculatory Ducts/metabolism , Ejaculatory Ducts/pathology , Infertility, Male/metabolism , Infertility, Male/pathology , Nuclear Proteins/deficiency , Animals , Cell Cycle Proteins/genetics , Cell Line , DNA Glycosylases/genetics , Epididymis/metabolism , Epididymis/pathology , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Infertility, Male/genetics , Male , Mice , Mice, Mutant Strains , Nuclear Proteins/genetics , Real-Time Polymerase Chain Reaction , Testis/metabolism , Testis/pathology
9.
Chromosoma ; 129(2): 115-120, 2020 06.
Article in English | MEDLINE | ID: mdl-32424716

ABSTRACT

The International University of Andalucía (UNIA) Current Trends in Biomedicine Workshop on Molecular Causes of Primary Microcephaly and Related Diseases took place in Baeza, Spain, November 18-20, 2019. This meeting brought together scientists from Europe, the USA and China to discuss recent advances in our molecular and genetic understanding of a group of rare neurodevelopmental diseases characterised by primary microcephaly, a condition in which head circumference is smaller than normal at birth. Microcephaly can be caused by inherited mutations that affect key cellular processes, or environmental exposure to radiation or other toxins. It can also result from viral infection, as exemplified by the recent Zika virus outbreak in South America. Here we summarise a number of the scientific advances presented and topics discussed at the meeting.


Subject(s)
Disease Susceptibility , Microcephaly/etiology , Neurodevelopmental Disorders/etiology , Centrosome , Cilia , Congresses as Topic , DNA Damage , DNA Replication , Genetic Association Studies , Genetic Predisposition to Disease , Genetic Testing , Humans , Microcephaly/diagnosis , Neurodevelopmental Disorders/diagnosis , Neurogenesis/genetics , Phenotype , Spindle Apparatus/genetics , Spindle Apparatus/metabolism
10.
Mol Cell ; 52(4): 566-73, 2013 Nov 21.
Article in English | MEDLINE | ID: mdl-24267451

ABSTRACT

DNA damage can stall the DNA replication machinery, leading to genomic instability. Thus, numerous mechanisms exist to complete genome duplication in the absence of a pristine DNA template, but identification of the enzymes involved remains incomplete. Here, we establish that Primase-Polymerase (PrimPol; CCDC111), an archaeal-eukaryotic primase (AEP) in eukaryotic cells, is involved in chromosomal DNA replication. PrimPol is required for replication fork progression on ultraviolet (UV) light-damaged DNA templates, possibly mediated by its ability to catalyze translesion synthesis (TLS) of these lesions. This PrimPol UV lesion bypass pathway is not epistatic with the Pol η-dependent pathway and, as a consequence, protects xeroderma pigmentosum variant (XP-V) patient cells from UV-induced cytotoxicity. In addition, we establish that PrimPol is also required for efficient replication fork progression during an unperturbed S phase. These and other findings indicate that PrimPol is an important player in replication fork progression in eukaryotic cells.


Subject(s)
Chromosomes, Human/genetics , DNA Adducts/genetics , DNA Primase/physiology , DNA Replication , DNA-Directed DNA Polymerase/physiology , Multifunctional Enzymes/physiology , Amino Acid Sequence , Animals , Cell Proliferation , Cell Survival , Chickens , DNA Adducts/chemistry , DNA Adducts/metabolism , DNA Damage , DNA Primase/chemistry , DNA, Single-Stranded/chemistry , DNA-Directed DNA Polymerase/chemistry , G2 Phase Cell Cycle Checkpoints , Gene Knockdown Techniques , HEK293 Cells , Humans , Mice , Mice, Knockout , Molecular Sequence Data , Multifunctional Enzymes/chemistry , Ultraviolet Rays , Xenopus
11.
EMBO J ; 35(9): 942-60, 2016 05 02.
Article in English | MEDLINE | ID: mdl-26933123

ABSTRACT

The generation of multiciliated cells (MCCs) is required for the proper function of many tissues, including the respiratory tract, brain, and germline. Defects in MCC development have been demonstrated to cause a subclass of mucociliary clearance disorders termed reduced generation of multiple motile cilia (RGMC). To date, only two genes, Multicilin (MCIDAS) and cyclin O (CCNO) have been identified in this disorder in humans. Here, we describe mice lacking GEMC1 (GMNC), a protein with a similar domain organization as Multicilin that has been implicated in DNA replication control. We have found that GEMC1-deficient mice are growth impaired, develop hydrocephaly with a high penetrance, and are infertile, due to defects in the formation of MCCs in the brain, respiratory tract, and germline. Our data demonstrate that GEMC1 is a critical regulator of MCC differentiation and a candidate gene for human RGMC or related disorders.


Subject(s)
Carrier Proteins/metabolism , Cell Differentiation , Cilia/genetics , Cilia/physiology , Growth Disorders/genetics , Growth Disorders/pathology , Animals , Carrier Proteins/genetics , Cell Cycle Proteins , Mice , Mice, Knockout
12.
Cell Mol Life Sci ; 76(19): 3827-3841, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31302748

ABSTRACT

The Tousled-like kinases (TLKs) are an evolutionarily conserved family of serine-threonine kinases that have been implicated in DNA replication, DNA repair, transcription, chromatin structure, viral latency, cell cycle checkpoint control and chromosomal stability in various organisms. The functions of the TLKs appear to depend largely on their ability to regulate the H3/H4 histone chaperone ASF1, although numerous TLK substrates have been proposed. Over the last few years, a clearer picture of TLK function has emerged through the identification of new partners, the definition of specific roles in development and the elucidation of their structural and biochemical properties. In addition, the TLKs have been clearly linked to human disease; both TLK1 and TLK2 are frequently amplified in human cancers and TLK2 mutations have been identified in patients with neurodevelopmental disorders characterized by intellectual disability (ID), autism spectrum disorder (ASD) and microcephaly. A better understanding of the substrates, regulation and diverse roles of the TLKs is needed to understand their functions in neurodevelopment and determine if they are viable targets for cancer therapy. In this review, we will summarize current knowledge of TLK biology and its potential implications in development and disease.


Subject(s)
Genomic Instability , Protein Serine-Threonine Kinases/metabolism , Animals , Epigenesis, Genetic , Humans , Mice , Neoplasms/drug therapy , Neoplasms/genetics , Neurodevelopmental Disorders/genetics , Protein Domains , Protein Kinases/chemistry , Protein Kinases/genetics , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/physiology
13.
Blood ; 126(22): 2502-10, 2015 Nov 26.
Article in English | MEDLINE | ID: mdl-26324700

ABSTRACT

Nijmegen breakage syndrome 1 (NBS1) is a component of the MRE11 complex, which is a sensor of DNA double-strand breaks and plays a crucial role in the DNA damage response. Because activated macrophages produce large amounts of reactive oxygen species (ROS) that can cause DNA lesions, we examined the role of NBS1 in macrophage functional activity. Proliferative and proinflammatory (interferon gamma [IFN-γ] and lipopolysaccharide [LPS]) stimuli led to increased NBS1 levels in macrophages. In mice expressing a hypomorphic allele of Nbs1, Nbs1(∆B/∆B), macrophage activation-induced ROS caused increased levels of DNA damage that were associated with defects in proliferation, delayed differentiation, and increased senescence. Furthermore, upon stimulation, Nbs1(∆B/∆B) macrophages exhibited increased expression of proinflammatory cytokines. In the in vivo 2,4-dinitrofluorobenzene model of inflammation, Nbs1(∆B/∆B) animals showed increased weight and ear thickness. By using the sterile inflammation by zymosan injection, we found that macrophage proliferation was drastically decreased in the peritoneal cavity of Nbs1(∆B/∆B) mice. Our findings show that NBS1 is crucial for macrophage function during normal aging. These results have implications for understanding the immune defects observed in patients with NBS and related disorders.


Subject(s)
Aging/immunology , Cell Cycle Proteins/immunology , Homeostasis/immunology , Macrophage Activation/immunology , Macrophages/immunology , Nuclear Proteins/immunology , Aging/pathology , Animals , DNA Repair Enzymes/immunology , DNA-Binding Proteins/immunology , Homeostasis/drug effects , Inflammation/chemically induced , Inflammation/immunology , Inflammation/pathology , Interferon-gamma/immunology , Lipopolysaccharides/toxicity , MRE11 Homologue Protein , Macrophage Activation/drug effects , Macrophages/pathology , Mice , Zymosan/toxicity
14.
Mol Cell ; 34(1): 13-25, 2009 Apr 10.
Article in English | MEDLINE | ID: mdl-19362533

ABSTRACT

Recent work has highlighted the importance of alternative, error-prone mechanisms for joining DNA double-strand breaks (DSBs) in mammalian cells. These noncanonical, nonhomologous end-joining (NHEJ) pathways threaten genomic stability but remain poorly characterized. The RAG postcleavage complex normally prevents V(D)J recombination-associated DSBs from accessing alternative NHEJ. Because the MRE11/RAD50/NBS1 complex localizes to RAG-mediated DSBs and possesses DNA end tethering, processing, and joining activities, we asked whether it plays a role in the mechanism of alternative NHEJ or participates in regulating access of DSBs to alternative repair pathways. We find that NBS1 is required for alternative NHEJ of hairpin coding ends, suppresses alternative NHEJ of signal ends, and promotes proper resolution of inversional recombination intermediates. These data demonstrate that the MRE11 complex functions at two distinct levels, regulating repair pathway choice (likely through enhancing the stability of DNA end complexes) and participating in alternative NHEJ of coding ends.


Subject(s)
Cell Cycle Proteins/physiology , DNA Breaks, Double-Stranded , Nuclear Proteins/physiology , Recombination, Genetic , ATP-Binding Cassette Transporters/metabolism , ATP-Binding Cassette Transporters/physiology , Acid Anhydride Hydrolases , Animals , Cell Cycle Proteins/genetics , Cells, Cultured , DNA Repair/physiology , DNA Repair Enzymes/metabolism , DNA Repair Enzymes/physiology , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/physiology , Endonucleases , MRE11 Homologue Protein , Mice , Mutation , Nuclear Proteins/genetics , Protein Kinase C/genetics , VDJ Recombinases/metabolism
15.
Nucleic Acids Res ; 43(15): 7371-87, 2015 Sep 03.
Article in English | MEDLINE | ID: mdl-26160886

ABSTRACT

The maintenance of genome stability is critical for the suppression of diverse human pathologies that include developmental disorders, premature aging, infertility and predisposition to cancer. The DNA damage response (DDR) orchestrates the appropriate cellular responses following the detection of lesions to prevent genomic instability. The MRE11 complex is a sensor of DNA double strand breaks (DSBs) and plays key roles in multiple aspects of the DDR, including DNA end resection that is critical for signaling and DNA repair. The MRE11 complex has been shown to function both upstream and in concert with the 5'-3' exonuclease EXO1 in DNA resection, but it remains unclear to what extent EXO1 influences DSB responses independently of the MRE11 complex. Here we examine the genetic relationship of the MRE11 complex and EXO1 during mammalian development and in response to DNA damage. Deletion of Exo1 in mice expressing a hypomorphic allele of Nbs1 leads to severe developmental impairment, embryonic death and chromosomal instability. While EXO1 plays a minimal role in normal cells, its loss strongly influences DNA replication, DNA repair, checkpoint signaling and damage sensitivity in NBS1 hypomorphic cells. Collectively, our results establish a key role for EXO1 in modulating the severity of hypomorphic MRE11 complex mutations.


Subject(s)
Cell Cycle Proteins/genetics , DNA Repair Enzymes/physiology , DNA Repair , Embryonic Development , Exodeoxyribonucleases/physiology , Nuclear Proteins/genetics , Alleles , Animals , Ataxia Telangiectasia Mutated Proteins/metabolism , Camptothecin/toxicity , Cells, Cultured , Chromosomal Instability , DNA Breaks, Double-Stranded , DNA Repair Enzymes/genetics , DNA Replication , DNA-Binding Proteins , Embryonic Development/genetics , Exodeoxyribonucleases/genetics , G2 Phase Cell Cycle Checkpoints , Gene Deletion , Genes, Lethal , Mice , Mutation
16.
Genes Dev ; 23(2): 171-80, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-19171781

ABSTRACT

The MRN complex (Mre11/RAD50/NBS1) and ATM (ataxia telangiectasia, mutated) are critical for the cellular response to DNA damage. ATM disruption causes ataxia telangiectasia (A-T), while MRN dysfunction can lead to A-T-like disease (ATLD) or Nijmegen breakage syndrome (NBS). Neuropathology is a hallmark of these diseases, whereby neurodegeneration occurs in A-T and ATLD while microcephaly characterizes NBS. To understand the contrasting neuropathology resulting from Mre11 or Nbs1 hypomorphic mutations, we analyzed neural tissue from Mre11(ATLD1/ATLD1) and Nbs1(DeltaB/DeltaB) mice after genotoxic stress. We found a pronounced resistance to DNA damage-induced apoptosis after ionizing radiation or DNA ligase IV (Lig4) loss in the Mre11(ATLD1/ATLD1) nervous system that was associated with defective Atm activation and phosphorylation of its substrates Chk2 and p53. Conversely, DNA damage-induced Atm phosphorylation was defective in Nbs1(DeltaB/DeltaB) neural tissue, although apoptosis occurred normally. We also conditionally disrupted Lig4 throughout the nervous system using Nestin-cre (Lig4(Nes-Cre)), and while viable, these mice showed pronounced microcephaly and a prominent age-related accumulation of DNA damage throughout the brain. Either Atm-/- or Mre11(ATLD1/ATLD1) genetic backgrounds, but not Nbs1(DeltaB/DeltaB), rescued Lig4(Nes-Cre) microcephaly. Thus, DNA damage signaling in the nervous system is different between ATLD and NBS and likely explains their respective neuropathology.


Subject(s)
Apoptosis , Ataxia Telangiectasia/physiopathology , DNA Damage/physiology , Neurons/physiology , Nijmegen Breakage Syndrome/physiopathology , Signal Transduction/genetics , Animals , Apoptosis/radiation effects , Ataxia Telangiectasia/genetics , Ataxia Telangiectasia Mutated Proteins , Brain/pathology , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , DNA Damage/genetics , DNA Ligase ATP , DNA Ligases/metabolism , DNA Repair Enzymes/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Enzyme Activation/physiology , Female , MRE11 Homologue Protein , Male , Mice , Mice, Transgenic , Microcephaly/pathology , Mutation , Neurons/cytology , Neurons/radiation effects , Nijmegen Breakage Syndrome/genetics , Nuclear Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Radiation, Ionizing , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
17.
Angew Chem Int Ed Engl ; 56(13): 3531-3535, 2017 03 20.
Article in English | MEDLINE | ID: mdl-28220994

ABSTRACT

A novel metabolomics approach for NMR-based stable isotope tracer studies called PEPA is presented, and its performance validated using human cancer cells. PEPA detects the position of carbon label in isotopically enriched metabolites and quantifies fractional enrichment by indirect determination of 13 C-satellite peaks using 1D-1 H-NMR spectra. In comparison with 13 C-NMR, TOCSY and HSQC, PEPA improves sensitivity, accelerates the elucidation of 13 C positions in labeled metabolites and the quantification of the percentage of stable isotope enrichment. Altogether, PEPA provides a novel framework for extending the high-throughput of 1 H-NMR metabolic profiling to stable isotope tracing in metabolomics, facilitating and complementing the information derived from 2D-NMR experiments and expanding the range of isotopically enriched metabolites detected in cellular extracts.


Subject(s)
Metabolomics/methods , Proton Magnetic Resonance Spectroscopy/methods , Carbon Isotopes/analysis , Cell Line, Tumor , High-Throughput Screening Assays/methods , Humans , Metabolome , Protons
18.
Mol Cell ; 31(1): 21-32, 2008 Jul 11.
Article in English | MEDLINE | ID: mdl-18614044

ABSTRACT

The Mre11 complex (Mre11, Rad50, and Nbs1) and Chk2 have been implicated in the DNA-damage response, an inducible process required for the suppression of malignancy. The Mre11 complex is predominantly required for repair and checkpoint activation in S phase, whereas Chk2 governs apoptosis. We examined the relationship between the Mre11 complex and Chk2 in the DNA-damage response via the establishment of Nbs1(DeltaB/DeltaB) Chk2(-/-) and Mre11(ATLD1/ATLD1) Chk2(-/-) mice. Chk2 deficiency did not modify the checkpoint defects or chromosomal instability of Mre11 complex mutants; however, the double-mutant mice exhibited synergistic defects in DNA-damage-induced p53 regulation and apoptosis. Nbs1(DeltaB/DeltaB) Chk2(-/-) and Mre11(ATLD1/ATLD1) Chk2(-/-) mice were also predisposed to tumors. In contrast, DNA-PKcs-deficient mice, in which G1-specific chromosome breaks are present, did not exhibit synergy with Chk2(-/-) mutants. These data suggest that Chk2 suppresses the oncogenic potential of DNA damage arising during S and G2 phases of the cell cycle.


Subject(s)
DNA Damage , DNA Replication , Precancerous Conditions/enzymology , Protein Serine-Threonine Kinases/metabolism , Alleles , Animals , Apoptosis , Cell Cycle , Cell Cycle Proteins/metabolism , Checkpoint Kinase 2 , Chromosomal Instability , Cyclin-Dependent Kinase Inhibitor p16/metabolism , DNA Repair , DNA Repair Enzymes/metabolism , DNA, Complementary/genetics , DNA-Activated Protein Kinase/metabolism , DNA-Binding Proteins/metabolism , Exons/genetics , Genome/genetics , MRE11 Homologue Protein , Mice , Mutation/genetics , Nuclear Proteins/metabolism , Precancerous Conditions/pathology , Protein Serine-Threonine Kinases/deficiency , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism
20.
J Biol Chem ; 289(50): 34838-50, 2014 Dec 12.
Article in English | MEDLINE | ID: mdl-25359778

ABSTRACT

USP28 (ubiquitin-specific protease 28) is a deubiquitinating enzyme that has been implicated in the DNA damage response, the regulation of Myc signaling, and cancer progression. The half-life stability of major regulators of critical cellular pathways depends on the activities of specific ubiquitin E3 ligases that target them for proteosomal degradation and deubiquitinating enzymes that promote their stabilization. One function of the post-translational small ubiquitin modifier (SUMO) is the regulation of enzymatic activity of protein targets. In this work, we demonstrate that the SUMO modification of the N-terminal domain of USP28 negatively regulates its deubiquitinating activity, revealing a role for the N-terminal region as a regulatory module in the control of USP28 activity. Despite the presence of ubiquitin-binding domains in the N-terminal domain, its truncation does not impair deubiquitinating activity on diubiquitin or polyubiquitin chain substrates. In contrast to other characterized USP deubiquitinases, our results indicate that USP28 has a chain preference activity for Lys(11), Lys(48), and Lys(63) diubiquitin linkages.


Subject(s)
SUMO-1 Protein/metabolism , Sumoylation , Ubiquitin Thiolesterase/metabolism , Amino Acid Sequence , Catalytic Domain , Humans , Models, Molecular , Molecular Sequence Data , Protein Processing, Post-Translational , Substrate Specificity , Ubiquitin Thiolesterase/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL