Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
2.
Blood ; 135(6): 411-428, 2020 02 06.
Article in English | MEDLINE | ID: mdl-31794600

ABSTRACT

Spontaneous regression is a recognized phenomenon in chronic lymphocytic leukemia (CLL) but its biological basis remains unknown. We undertook a detailed investigation of the biological and clinical features of 20 spontaneous CLL regression cases incorporating phenotypic, functional, transcriptomic, and genomic studies at sequential time points. All spontaneously regressed tumors were IGHV-mutated with no restricted IGHV usage or B-cell receptor (BCR) stereotypy. They exhibited shortened telomeres similar to nonregressing CLL, indicating prior proliferation. They also displayed low Ki-67, CD49d, cell-surface immunoglobulin M (IgM) expression and IgM-signaling response but high CXCR4 expression, indicating low proliferative activity associated with poor migration to proliferation centers, with these features becoming increasingly marked during regression. Spontaneously regressed CLL displayed a transcriptome profile characterized by downregulation of metabolic processes as well as MYC and its downstream targets compared with nonregressing CLL. Moreover, spontaneous regression was associated with reversal of T-cell exhaustion features including reduced programmed cell death 1 expression and increased T-cell proliferation. Interestingly, archetypal CLL genomic aberrations including HIST1H1B and TP53 mutations and del(13q14) were found in some spontaneously regressing tumors, but genetic composition remained stable during regression. Conversely, a single case of CLL relapse following spontaneous regression was associated with increased BCR signaling, CLL proliferation, and clonal evolution. These observations indicate that spontaneously regressing CLL appear to undergo a period of proliferation before entering a more quiescent state, and that a complex interaction between genomic alterations and the microenvironment determines disease course. Together, the findings provide novel insight into the biological processes underpinning spontaneous CLL regression, with implications for CLL treatment.


Subject(s)
Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Adult , Aged , Aged, 80 and over , Cell Proliferation , Female , Gene Expression Regulation, Leukemic , Humans , Immunoglobulin Heavy Chains/genetics , Immunoglobulin M/genetics , Ki-67 Antigen/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Male , Middle Aged , Mutation , Polymorphism, Single Nucleotide , Receptors, CXCR4/genetics , Tumor Microenvironment
3.
Nat Rev Dis Primers ; 5(1): 64, 2019 09 19.
Article in English | MEDLINE | ID: mdl-31537806

ABSTRACT

Fanconi anaemia (FA), ataxia telangiectasia (A-T), Nijmegen breakage syndrome (NBS) and Bloom syndrome (BS) are clinically distinct, chromosome instability (or breakage) disorders. Each disorder has its own pattern of chromosomal damage, with cells from these patients being hypersensitive to particular genotoxic drugs, indicating that the underlying defect in each case is likely to be different. In addition, each syndrome shows a predisposition to cancer. Study of the molecular and genetic basis of these disorders has revealed mechanisms of recognition and repair of DNA double-strand breaks, DNA interstrand crosslinks and DNA damage during DNA replication. Specialist clinics for each disorder have provided the concentration of expertise needed to tackle their characteristic clinical problems and improve outcomes. Although some treatments of the consequences of a disorder may be possible, for example, haematopoietic stem cell transplantation in FA and NBS, future early intervention to prevent complications of disease will depend on a greater understanding of the roles of the affected DNA repair pathways in development. An important realization has been the predisposition to cancer in carriers of some of these gene mutations.


Subject(s)
DNA Repair-Deficiency Disorders/diagnosis , DNA Repair-Deficiency Disorders/genetics , Ataxia Telangiectasia/diagnosis , Ataxia Telangiectasia/genetics , Ataxia Telangiectasia/physiopathology , Bloom Syndrome/diagnosis , Bloom Syndrome/genetics , Bloom Syndrome/physiopathology , DNA Damage/genetics , DNA Repair-Deficiency Disorders/physiopathology , Fanconi Anemia/diagnosis , Fanconi Anemia/genetics , Fanconi Anemia/physiopathology , Humans , Nijmegen Breakage Syndrome/diagnosis , Nijmegen Breakage Syndrome/genetics , Nijmegen Breakage Syndrome/physiopathology
4.
Blood ; 130(2): 156-166, 2017 07 13.
Article in English | MEDLINE | ID: mdl-28495793

ABSTRACT

The role of deubiquitylase ubiquitin-specific protease 7 (USP7) in the regulation of the p53-dependent DNA damage response (DDR) pathway is well established. Whereas previous studies have mostly focused on the mechanisms underlying how USP7 directly controls p53 stability, we recently showed that USP7 modulates the stability of the DNA damage responsive E3 ubiquitin ligase RAD18. This suggests that targeting USP7 may have therapeutic potential even in tumors with defective p53 or ibrutinib resistance. To test this hypothesis, we studied the effect of USP7 inhibition in chronic lymphocytic leukemia (CLL) where the ataxia telangiectasia mutated (ATM)-p53 pathway is inactivated with relatively high frequency, leading to treatment resistance and poor clinical outcome. We demonstrate that USP7 is upregulated in CLL cells, and its loss or inhibition disrupts homologous recombination repair (HRR). Consequently, USP7 inhibition induces significant tumor-cell killing independently of ATM and p53 through the accumulation of genotoxic levels of DNA damage. Moreover, USP7 inhibition sensitized p53-defective, chemotherapy-resistant CLL cells to clinically achievable doses of HRR-inducing chemotherapeutic agents in vitro and in vivo in a murine xenograft model. Together, these results identify USP7 as a promising therapeutic target for the treatment of hematological malignancies with DDR defects, where ATM/p53-dependent apoptosis is compromised.


Subject(s)
Gene Expression Regulation, Neoplastic , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Recombinational DNA Repair/drug effects , Tumor Suppressor Protein p53/genetics , Ubiquitin-Specific Proteases/genetics , Adenine/analogs & derivatives , Animals , Antineoplastic Agents/pharmacology , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Line, Tumor , DNA Damage , Drug Resistance, Neoplasm/genetics , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Mice , Mice, Inbred NOD , Piperidines , Primary Cell Culture , Pyrazoles/pharmacology , Pyrimidines/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Specific Peptidase 7 , Ubiquitin-Specific Proteases/antagonists & inhibitors , Ubiquitin-Specific Proteases/metabolism , Xenograft Model Antitumor Assays
5.
PLoS Genet ; 12(3): e1005945, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26990772

ABSTRACT

Patients with biallelic truncating mutations in PALB2 have a severe form of Fanconi anaemia (FA-N), with a predisposition for developing embryonal-type tumours in infancy. Here we describe two unusual patients from a single family, carrying biallelic PALB2 mutations, one truncating, c.1676_1677delAAinsG;(p.Gln559ArgfsTer2), and the second, c.2586+1G>A; p.Thr839_Lys862del resulting in an in frame skip of exon 6 (24 amino acids). Strikingly, the affected individuals did not exhibit the severe developmental defects typical of FA-N patients and initially presented with B cell non-Hodgkin lymphoma. The expressed p.Thr839_Lys862del mutant PALB2 protein retained the ability to interact with BRCA2, previously unreported in FA-N patients. There was also a large increased chromosomal radiosensitivity following irradiation in G2 and increased sensitivity to mitomycin C. Although patient cells were unable to form Rad51 foci following exposure to either DNA damaging agent, U2OS cells, in which the mutant PALB2 with in frame skip of exon 6 was induced, did show recruitment of Rad51 to foci following damage. We conclude that a very mild form of FA-N exists arising from a hypomorphic PALB2 allele.


Subject(s)
Fanconi Anemia/genetics , Lymphoma, Non-Hodgkin/genetics , Nuclear Proteins/genetics , Rad51 Recombinase/genetics , Tumor Suppressor Proteins/genetics , Alleles , BRCA2 Protein/genetics , BRCA2 Protein/metabolism , Chromosomes/genetics , DNA Damage/genetics , Fanconi Anemia/pathology , Fanconi Anemia Complementation Group N Protein , Humans , Lymphocytes/metabolism , Lymphocytes/pathology , Lymphoma, Non-Hodgkin/pathology , Mutation
6.
Dis Model Mech ; 8(11): 1401-12, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26398941

ABSTRACT

Chronic lymphocytic leukaemia (CLL) cells require microenvironmental support for their proliferation. This can be recapitulated in highly immunocompromised hosts in the presence of T cells and other supporting cells. Current primary CLL xenograft models suffer from limited duration of tumour cell engraftment coupled with gradual T-cell outgrowth. Thus, a greater understanding of the interaction between CLL and T cells could improve their utility. In this study, using two distinct mouse xenograft models, we investigated whether xenografts recapitulate CLL biology, including natural environmental interactions with B-cell receptors and T cells, and whether manipulation of autologous T cells can expand the duration of CLL engraftment. We observed that primary CLL xenografts recapitulated both the tumour phenotype and T-cell repertoire observed in patients and that engraftment was significantly shorter for progressive tumours. A reduction in the number of patient T cells that were injected into the mice to 2-5% of the initial number or specific depletion of CD8(+) cells extended the limited xenograft duration of progressive cases to that characteristic of indolent disease. We conclude that manipulation of T cells can enhance current CLL xenograft models and thus expand their utility for investigation of tumour biology and pre-clinical drug assessment.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunocompromised Host , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Lymphocytes, Tumor-Infiltrating/immunology , T-Lymphocyte Subsets/immunology , Tumor Microenvironment , Animals , CD8-Positive T-Lymphocytes/pathology , Cell Proliferation , Cell Survival , Cells, Cultured , Coculture Techniques , Cytotoxicity, Immunologic , Graft Survival , Heterografts , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Lymphocyte Activation , Lymphocyte Depletion , Lymphocytes, Tumor-Infiltrating/pathology , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Phenotype , Spleen/immunology , T-Lymphocyte Subsets/pathology , Time Factors
7.
Haematologica ; 100(8): 1076-85, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25840602

ABSTRACT

Inactivation of the Ataxia Telangiectasia Mutated gene in chronic lymphocytic leukemia results in resistance to p53-dependent apoptosis and inferior responses to treatment with DNA damaging agents. Hence, p53-independent strategies are required to target Ataxia Telangiectasia Mutated-deficient chronic lymphocytic leukemia. As Ataxia Telangiectasia Mutated has been implicated in redox homeostasis, we investigated the effect of the Ataxia Telangiectasia Mutated-null chronic lymphocytic leukemia genotype on cellular responses to oxidative stress with a view to therapeutic targeting. We found that in comparison to Ataxia Telangiectasia Mutated-wild type chronic lymphocytic leukemia, pro-oxidant treatment of Ataxia Telangiectasia Mutated-null cells led to reduced binding of NF-E2 p45-related factor-2 to antioxidant response elements and thus decreased expression of target genes. Furthermore, Ataxia Telangiectasia Mutated-null chronic lymphocytic leukemia cells contained lower levels of antioxidants and elevated mitochondrial reactive oxygen species. Consequently, Ataxia Telangiectasia Mutated-null chronic lymphocytic leukemia, but not tumors with 11q deletion or TP53 mutations, exhibited differentially increased sensitivity to pro-oxidants both in vitro and in vivo. We found that cell death was mediated by a p53- and caspase-independent mechanism associated with apoptosis inducing factor activity. Together, these data suggest that defective redox-homeostasis represents an attractive therapeutic target for Ataxia Telangiectasia Mutated-null chronic lymphocytic leukemia.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/genetics , Homozygote , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Mutation , Oxidants/metabolism , Phenotype , Animals , Antioxidants/metabolism , Apoptosis , Caspases/metabolism , Disease Models, Animal , Gene Expression Regulation, Leukemic , Humans , Mitochondria/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Protein Binding , Reactive Oxygen Species/metabolism , Response Elements , Superoxides/metabolism , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays
8.
J Clin Invest ; 124(7): 3137-46, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24911150

ABSTRACT

Numerous human disorders, including Cockayne syndrome, UV-sensitive syndrome, xeroderma pigmentosum, and trichothiodystrophy, result from the mutation of genes encoding molecules important for nucleotide excision repair. Here, we describe a syndrome in which the cardinal clinical features include short stature, hearing loss, premature aging, telangiectasia, neurodegeneration, and photosensitivity, resulting from a homozygous missense (p.Ser228Ile) sequence alteration of the proliferating cell nuclear antigen (PCNA). PCNA is a highly conserved sliding clamp protein essential for DNA replication and repair. Due to this fundamental role, mutations in PCNA that profoundly impair protein function would be incompatible with life. Interestingly, while the p.Ser228Ile alteration appeared to have no effect on protein levels or DNA replication, patient cells exhibited marked abnormalities in response to UV irradiation, displaying substantial reductions in both UV survival and RNA synthesis recovery. The p.Ser228Ile change also profoundly altered PCNA's interaction with Flap endonuclease 1 and DNA Ligase 1, DNA metabolism enzymes. Together, our findings detail a mutation of PCNA in humans associated with a neurodegenerative phenotype, displaying clinical and molecular features common to other DNA repair disorders, which we showed to be attributable to a hypomorphic amino acid alteration.


Subject(s)
DNA Repair-Deficiency Disorders/genetics , Mutant Proteins/genetics , Mutation, Missense , Proliferating Cell Nuclear Antigen/genetics , Adolescent , Adult , Aging, Premature/genetics , Amino Acid Substitution , Child , Chromosomes, Human, Pair 20/genetics , DNA Mutational Analysis , DNA Repair-Deficiency Disorders/pathology , DNA Repair-Deficiency Disorders/physiopathology , Dwarfism/genetics , Female , Hearing Loss/genetics , Homozygote , Humans , Male , Models, Molecular , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Nerve Degeneration/genetics , Pedigree , Phenotype , Photosensitivity Disorders/genetics , Proliferating Cell Nuclear Antigen/chemistry , Proliferating Cell Nuclear Antigen/metabolism , Protein Structure, Quaternary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Syndrome , Telangiectasis/genetics
9.
Neuromolecular Med ; 15(3): 447-57, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23632773

ABSTRACT

Variant ataxia telangiectasia (A-T) may be an underdiagnosed entity. We correlate data from radiosensitivity and kinase assays with clinical and molecular data from a patient with variant A-T and relatives. The coding region of ATM was sequenced. To evaluate the functional effect of the mutations, we performed kinase assays and developed a novel S-G2 micronucleus test. Our patient presented with mild dystonia, moderately dysarthric speech, increased serum α-fetoprotein but no ataxia nor telangiectasias, no nystagmus or oculomotor dyspraxia. She has a severe IgA deficiency, but does not have recurrent infections. She is compound heterozygote for ATM c.8122G>A (p.Asp2708Asn) and c.8851-1G>T, leading to in frame loss of 63 nucleotides at the cDNA level. A trace amount of ATM protein is translated from both alleles. Residual kinase activity is derived only from the p.Asp2708Asn allele. The conventional G0 micronucleus test, based on irradiation of resting lymphocytes, revealed a radiosensitive phenotype for the patient, but not for the heterozygous relatives. As ATM is involved in homologous recombination and G2/M cell cycle checkpoint, we optimized an S-G2 micronucleus assay, allowing to evaluate micronuclei in lymphocytes irradiated in the S and G2 phases. This test showed increased radiosensitivity for both the patient and the heterozygous carriers. Intriguingly, heterozygous carriers of c.8851-1G>T (mutation associated with absence of kinase activity) showed a stronger radiosensitive phenotype with this assay than heterozygous carriers of p.Asp2708Asn (mutation associated with residual kinase activity). The modified S-G2 micronucleus assay provided phenotypic insight into complement the diagnosis of this atypical A-T patient.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia/genetics , Adult , Amino Acid Substitution , Ataxia Telangiectasia Mutated Proteins/chemistry , Ataxia Telangiectasia Mutated Proteins/physiology , Breast Neoplasms/genetics , Caffeine/pharmacology , Child , Exons/genetics , Female , G2 Phase , Heterozygote , Humans , Lymphocytes/drug effects , Lymphocytes/radiation effects , Male , Micronucleus Tests , Mutation, Missense , Neoplastic Syndromes, Hereditary/genetics , Neurologic Examination , Pedigree , Phenotype , RNA Splice Sites/genetics , Radiation Tolerance/genetics , Recombinational DNA Repair/genetics , Rhabdomyosarcoma, Embryonal/genetics , S Phase , Sequence Analysis, DNA
10.
Mov Disord ; 28(4): 524-8, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23143971

ABSTRACT

BACKGROUND: The major clinical feature of ataxia telangiectasia (A-T) is severe progressive neurodegeneration with onset in infancy. This classical A-T phenotype is caused by biallelic null mutations in the ATM gene, leading to the absence of ATM protein and increased cellular radiosensitivity. We report an unusual case of A-T in a 41-year-old mother, A-T210, who had very mild neurological symptoms despite complete loss of ATM protein. METHODS: A neurological examination was performed, cellular radiosensitivity was assessed, and the ATM gene was sequenced. Skin fibroblasts and a lymphoblastoid cell line (LCL) were assayed for ATM protein expression and kinase activity. RESULTS: Patient A-T210 showed mild chorea, dystonia, and gait ataxia, walked independently, and drove a car. LCL and skin fibroblasts were radiosensitive and did not express ATM protein. Two ATM-null mutations were identified. CONCLUSIONS: The severe neurodegeneration resulting from loss of ATM can be mitigated in some circumstances.


Subject(s)
Ataxia Telangiectasia/genetics , Mutation/genetics , Adult , Ataxia Telangiectasia/diagnosis , Ataxia Telangiectasia/metabolism , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Line , Female , Genotype , Humans , Phenotype , Radiation Tolerance
11.
PLoS Genet ; 8(11): e1002945, 2012.
Article in English | MEDLINE | ID: mdl-23144622

ABSTRACT

A homozygous mutational change in the Ataxia-Telangiectasia and RAD3 related (ATR) gene was previously reported in two related families displaying Seckel Syndrome (SS). Here, we provide the first identification of a Seckel Syndrome patient with mutations in ATRIP, the gene encoding ATR-Interacting Protein (ATRIP), the partner protein of ATR required for ATR stability and recruitment to the site of DNA damage. The patient has compound heterozygous mutations in ATRIP resulting in reduced ATRIP and ATR expression. A nonsense mutational change in one ATRIP allele results in a C-terminal truncated protein, which impairs ATR-ATRIP interaction; the other allele is abnormally spliced. We additionally describe two further unrelated patients native to the UK with the same novel, heterozygous mutations in ATR, which cause dramatically reduced ATR expression. All patient-derived cells showed defective DNA damage responses that can be attributed to impaired ATR-ATRIP function. Seckel Syndrome is characterised by microcephaly and growth delay, features also displayed by several related disorders including Majewski (microcephalic) osteodysplastic primordial dwarfism (MOPD) type II and Meier-Gorlin Syndrome (MGS). The identification of an ATRIP-deficient patient provides a novel genetic defect for Seckel Syndrome. Coupled with the identification of further ATR-deficient patients, our findings allow a spectrum of clinical features that can be ascribed to the ATR-ATRIP deficient sub-class of Seckel Syndrome. ATR-ATRIP patients are characterised by extremely severe microcephaly and growth delay, microtia (small ears), micrognathia (small and receding chin), and dental crowding. While aberrant bone development was mild in the original ATR-SS patient, some of the patients described here display skeletal abnormalities including, in one patient, small patellae, a feature characteristically observed in Meier-Gorlin Syndrome. Collectively, our analysis exposes an overlapping clinical manifestation between the disorders but allows an expanded spectrum of clinical features for ATR-ATRIP Seckel Syndrome to be defined.


Subject(s)
Adaptor Proteins, Signal Transducing , Cell Cycle Proteins , DNA-Binding Proteins , Dwarfism/genetics , Growth Disorders , Micrognathism , Protein Serine-Threonine Kinases , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Codon, Nonsense , Congenital Microtia , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dwarfism/pathology , Ear/abnormalities , Ear/pathology , Female , Fetal Growth Retardation/genetics , Fetal Growth Retardation/pathology , Gene Expression Regulation , Growth Disorders/genetics , Growth Disorders/pathology , Heterozygote , Humans , Male , Microcephaly/genetics , Microcephaly/pathology , Micrognathism/genetics , Micrognathism/pathology , Osteochondrodysplasias/genetics , Osteochondrodysplasias/pathology , Patella/abnormalities , Patella/pathology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA Splicing , Signal Transduction/genetics
12.
J Clin Oncol ; 30(36): 4524-32, 2012 Dec 20.
Article in English | MEDLINE | ID: mdl-23091097

ABSTRACT

PURPOSE: The prognostic significance of ATM mutations in chronic lymphocytic leukemia (CLL) is unclear. We assessed their impact in the context of a prospective randomized trial. PATIENTS AND METHODS: We analyzed the ATM gene in 224 patients treated on the Leukemia Research Fund Chronic Lymphocytic Leukemia 4 (LRF-CLL4) trial with chlorambucil or fludarabine with and without cyclophosphamide. ATM status was analyzed by denaturing high-performance liquid chromatography and was related to treatment response, survival, and the impact of TP53 alterations for the same patient cohort. RESULTS: We identified 36 ATM mutations in 33 tumors, 16 with and 17 without 11q deletion. Mutations were associated with advanced disease stage and involvement of multiple lymphoid sites. Patients with both ATM mutation and 11q deletion showed significantly reduced progression-free survival (median, 7.4 months) compared with those with ATM wild type (28.6 months), 11q deletion alone (17.1 months), or ATM mutation alone (30.8 months), but survival was similar to that in patients with monoallelic (6.7 months) or biallelic (3.4 months) TP53 alterations. This effect was independent of treatment, immunoglobulin heavy chain variable gene (IGHV) status, age, sex, or disease stage. Overall survival for patients with biallelic ATM alterations was also significantly reduced compared with those with ATM wild type or ATM mutation alone (median, 42.2 v 85.5 v 77.6 months, respectively). CONCLUSION: The combination of 11q deletion and ATM mutation in CLL is associated with significantly shorter progression-free and overall survival following first-line treatment with alkylating agents and purine analogs. Assessment of ATM mutation status in patients with 11q deletion may influence the choice of subsequent therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Cycle Proteins/genetics , Chlorambucil/therapeutic use , DNA-Binding Proteins/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Mutation , Protein Serine-Threonine Kinases/genetics , Tumor Suppressor Proteins/genetics , Vidarabine/analogs & derivatives , Aged , Alleles , Ataxia Telangiectasia Mutated Proteins , Cyclophosphamide/administration & dosage , Disease Progression , Disease-Free Survival , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Leukemic , Gene Silencing , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/enzymology , Male , Middle Aged , Prognosis , Prospective Studies , Survival Analysis , United Kingdom , Vidarabine/administration & dosage , Vidarabine/therapeutic use
13.
Mol Cell ; 45(4): 505-16, 2012 Feb 24.
Article in English | MEDLINE | ID: mdl-22365830

ABSTRACT

DNA double-strand break (DSB) signaling and repair are critical for cell viability, and rely on highly coordinated pathways whose molecular organization is still incompletely understood. Here, we show that heterogeneous nuclear ribonucleoprotein U-like (hnRNPUL) proteins 1 and 2 play key roles in cellular responses to DSBs. We identify human hnRNPUL1 and -2 as binding partners for the DSB sensor complex MRE11-RAD50-NBS1 (MRN) and demonstrate that hnRNPUL1 and -2 are recruited to DNA damage in an interdependent manner that requires MRN. Moreover, we show that hnRNPUL1 and -2 stimulate DNA-end resection and promote ATR-dependent signaling and DSB repair by homologous recombination, thereby contributing to cell survival upon exposure to DSB-inducing agents. Finally, we establish that hnRNPUL1 and -2 function downstream of MRN and CtBP-interacting protein (CtIP) to promote recruitment of the BLM helicase to DNA breaks. Collectively, these results provide insights into how mammalian cells respond to DSBs.


Subject(s)
DNA Breaks, Double-Stranded , DNA End-Joining Repair , Heterogeneous-Nuclear Ribonucleoproteins/physiology , Nuclear Proteins/physiology , Transcription Factors/physiology , Acid Anhydride Hydrolases , Carrier Proteins/genetics , Carrier Proteins/metabolism , Carrier Proteins/physiology , Cell Cycle Proteins/metabolism , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/metabolism , Endodeoxyribonucleases , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Humans , MRE11 Homologue Protein , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism
14.
Nucleic Acids Res ; 40(6): 2518-26, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22139912

ABSTRACT

The loss of telomere function can result in the fusion of telomeres with other telomeric loci, or non-telomeric double-stranded DNA breaks. Sequence analysis of fusion events between short dysfunctional telomeres in human cells has revealed that fusion is characterized by a distinct molecular signature consisting of extensive deletions and micro-homology at the fusion points. This signature is consistent with alternative error-prone end-joining processes. We have examined the role that Mre11 may play in the fusion of short telomeres in human cells; to do this, we have analysed telomere fusion events in cells derived from ataxia-telangiectasia-like disorder (ATLD) patients that exhibit hypomorphic mutations in MRE11. The telomere dynamics of ATLD fibroblasts were indistinguishable from wild-type fibroblasts and they were proficient in the fusion of short telomeres. However, we observed a high frequency of insertion of DNA sequences at the fusion points that created localized sequence duplications. These data indicate that Mre11 plays a role in the fusion of short dysfunctional telomeres in human cells and are consistent with the hypothesis that as part of the MRN complex it serves to stabilize the joining complex, thereby controlling the fidelity of the fusion reaction.


Subject(s)
DNA-Binding Proteins/physiology , Telomere/chemistry , Ataxia Telangiectasia/genetics , Cell Line , DNA-Binding Proteins/genetics , Humans , MRE11 Homologue Protein , Mutation , Repetitive Sequences, Nucleic Acid , Sequence Deletion , Telomere/metabolism
16.
Clin Immunol ; 140(1): 26-36, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21459046

ABSTRACT

ATM kinase modulates pathways implicated in premature ageing and ATM genotype predicts survival, yet immunodeficiency in ataxia telangiectasia is regarded as mild and unrelated to age. We address this paradox in a molecularly characterised sequential adult cohort with classical and mild variant ataxia telangiectasia. Immunodeficiency has the characteristics of premature ageing across multiple cellular and molecular immune parameters. This immune ageing occurs without previous CMV infection. Age predicts immunodeficiency in genetically homogeneous ataxia telangiectasia, and in comparison with controls, calendar age is exceeded by immunological age defined by thymic naïve CD4+ T cell levels. Applying ataxia telangiectasia as a model of immune ageing, pneumococcal vaccine responses, characteristically deficient in physiological ageing, are predicted by thymic naïve CD4+ T cell levels. These data suggest inherited defects of DNA repair may provide valuable insight into physiological ageing. Thymic naïve CD4+ T cells may provide a biomarker for vaccine responsiveness in elderly cohorts.


Subject(s)
Aging/immunology , Ataxia Telangiectasia/immunology , CD4-Positive T-Lymphocytes/immunology , Adult , Ataxia Telangiectasia/genetics , Ataxia Telangiectasia Mutated Proteins , Cell Count , Cell Cycle Proteins/genetics , Cell Separation , DNA-Binding Proteins/genetics , Female , Flow Cytometry , Humans , Male , Protein Serine-Threonine Kinases/genetics , Tumor Suppressor Proteins/genetics
17.
Genome Biol ; 11(11): R110, 2010.
Article in English | MEDLINE | ID: mdl-21078160

ABSTRACT

BACKGROUND: Immunolabeling of metaphase chromosome spreads can map components of the human epigenome at the single cell level. Previously, there has been no systematic attempt to explore the potential of this approach for epigenomic mapping and thereby to complement approaches based on chromatin immunoprecipitation (ChIP) and sequencing technologies. RESULTS: By immunostaining and immunofluorescence microscopy, we have defined the distribution of selected histone modifications across metaphase chromosomes from normal human lymphoblastoid cells and constructed immunostained karyotypes. Histone modifications H3K9ac, H3K27ac and H3K4me3 are all located in the same set of sharply defined immunofluorescent bands, corresponding to 10- to 50-Mb genomic segments. Primary fibroblasts gave broadly the same banding pattern. Bands co-localize with regions relatively rich in genes and CpG islands. Staining intensity usually correlates with gene/CpG island content, but occasional exceptions suggest that other factors, such as transcription or SINE density, also contribute. H3K27me3, a mark associated with gene silencing, defines a set of bands that only occasionally overlap with gene-rich regions. Comparison of metaphase bands with histone modification levels across the interphase genome (ENCODE, ChIP-seq) shows a close correspondence for H3K4me3 and H3K27ac, but major differences for H3K27me3. CONCLUSIONS: At metaphase the human genome is packaged as chromatin in which combinations of histone modifications distinguish distinct regions along the euchromatic chromosome arms. These regions reflect the high-level interphase distributions of some histone modifications, and may be involved in heritability of epigenetic states, but we also find evidence for extensive remodeling of the epigenome at mitosis.


Subject(s)
Chromatin Immunoprecipitation/methods , Epigenomics , Genome, Human , Histones/chemistry , Metaphase/genetics , Cell Line , Chromatin/chemistry , CpG Islands , Epigenesis, Genetic , Female , Gene Silencing , Humans , Karyotyping , Male , Microarray Analysis , Mitosis , Protein Processing, Post-Translational
18.
Blood ; 116(22): 4578-87, 2010 Nov 25.
Article in English | MEDLINE | ID: mdl-20739657

ABSTRACT

The Ataxia Telangiectasia Mutated (ATM) gene is frequently inactivated in lymphoid malignancies such as chronic lymphocytic leukemia (CLL), T-prolymphocytic leukemia (T-PLL), and mantle cell lymphoma (MCL) and is associated with defective apoptosis in response to alkylating agents and purine analogues. ATM mutant cells exhibit impaired DNA double strand break repair. Poly (ADP-ribose) polymerase (PARP) inhibition that imposes the requirement for DNA double strand break repair should selectively sensitize ATM-deficient tumor cells to killing. We investigated in vitro sensitivity to the poly (ADP-ribose) polymerase inhibitor olaparib (AZD2281) of 5 ATM mutant lymphoblastoid cell lines (LCL), an ATM mutant MCL cell line, an ATM knockdown PGA CLL cell line, and 9 ATM-deficient primary CLLs induced to cycle and observed differential killing compared with ATM wildtype counterparts. Pharmacologic inhibition of ATM and ATM knockdown confirmed the effect was ATM-dependent and mediated through mitotic catastrophe independently of apoptosis. A nonobese diabetic/severe combined immunodeficient (NOD/SCID) murine xenograft model of an ATM mutant MCL cell line demonstrated significantly reduced tumor load and an increased survival of animals after olaparib treatment in vivo. Addition of olaparib sensitized ATM null tumor cells to DNA-damaging agents. We suggest that olaparib would be an appropriate agent for treating refractory ATM mutant lymphoid tumors.


Subject(s)
Antineoplastic Agents/therapeutic use , Cell Cycle Proteins/genetics , DNA-Binding Proteins/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Lymphoma, Mantle-Cell/drug therapy , Phthalazines/therapeutic use , Piperazines/therapeutic use , Poly(ADP-ribose) Polymerase Inhibitors , Protein Serine-Threonine Kinases/genetics , Tumor Suppressor Proteins/genetics , Animals , Antineoplastic Agents/pharmacology , Ataxia Telangiectasia Mutated Proteins , Cell Line, Tumor , Cell Proliferation/drug effects , Cells, Cultured , DNA Damage/drug effects , Gene Knockdown Techniques , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Lymphoma, Mantle-Cell/genetics , Mice , Mice, SCID , Mutation , Phthalazines/pharmacology , Piperazines/pharmacology
19.
Proc Natl Acad Sci U S A ; 107(27): 12251-6, 2010 Jul 06.
Article in English | MEDLINE | ID: mdl-20566845

ABSTRACT

Activation of the cellular DNA damage response is detrimental to adenovirus (Ad) infection. Ad has therefore evolved a number of strategies to inhibit ATM- and ATR-dependent signaling pathways during infection. Recent work suggests that the Ad5 E4orf3 protein prevents ATR activation through its ability to mislocalize the MRN complex. Here we provide evidence to indicate that Ad12 has evolved a different strategy from Ad5 to inhibit ATR. We show that Ad12 utilizes a CUL2/RBX1/elongin C-containing ubiquitin ligase to promote the proteasomal degradation of the ATR activator protein topoisomerase-IIbeta-binding protein 1 (TOPBP1). Ad12 also uses this complex to degrade p53 during infection, in contrast to Ad5, which requires a CUL5-based ubiquitin ligase. Although Ad12-mediated degradation of p53 is dependent upon both E1B-55K and E4orf6, Ad12-mediated degradation of TOPBP1 is solely dependent on E4orf6. We propose that Ad12 E4orf6 has two principal activities: to recruit the CUL2-based ubiquitin ligase and to act as substrate receptor for TOPBP1. In support of the idea that Ad12 E4orf6 specifically prevents ATR activation during infection by targeting TOPBP1 for degradation, we demonstrate that Ad12 E4orf6 can inhibit the ATR-dependent phosphorylation of CHK1 in response to replication stress. Taken together, these data provide insights into how Ad modulates ATR signaling pathways during infection.


Subject(s)
Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Viral Proteins/metabolism , Adenoviridae/genetics , Adenoviridae/metabolism , Adenoviridae/physiology , Adenovirus E1B Proteins/genetics , Adenovirus E1B Proteins/metabolism , Ataxia Telangiectasia Mutated Proteins , Blotting, Western , Carrier Proteins/genetics , Cell Cycle Proteins/genetics , Cell Line, Tumor , Checkpoint Kinase 1 , Cullin Proteins/genetics , Cullin Proteins/metabolism , DNA-Binding Proteins/genetics , Elongin , Fluorescent Antibody Technique , HeLa Cells , Host-Pathogen Interactions , Humans , Microscopy, Confocal , Mutation , Nuclear Proteins/genetics , Phosphorylation , Proteasome Endopeptidase Complex/metabolism , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , RNA Interference , Transcription Factors/genetics , Transcription Factors/metabolism , Transfection , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Viral Proteins/genetics
20.
Hum Mutat ; 30(8): 1222-30, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19431188

ABSTRACT

Ataxia-telangiectasia mutated (ATM) is the gene mutated in the cancer-predisposing disorder ataxia-telangiectasia (A-T). We modeled ATM sequence variants identified in UK A-T patients to determine the stability and kinase activity of the resulting proteins as well as the distribution of these mutations across the coding region. Of 20 missense changes modeled, 10 proteins showed ATM kinase activity and 10 showed none. In the majority of cases the mutant ATM protein was unstable, although this was variable. Reduction in ATM kinase activity can result either from the presence of low levels of unstable mutant protein with relatively normal specific kinase activity or from stable mutant protein with deficient ATM kinase activation. Indeed, ATM mutant proteins without kinase activity toward downstream targets were still able to autophosphorylate on serine 1981, although in a much less efficient manner, suggesting that this was not sufficient for ATM activation. In terms of function, green fluorescent protein (GFP)-tagged kinase inactive ATM proteins could form ionizing radiation (IR)-induced foci (IRIF), at least temporarily, which colocalized with the DNA double-strand break (DSB) marker gammaH2AX. Consistent with this, both kinase active and inactive mutant ATM proteins were able to interfere with phosphorylation of targets by endogenous ATM. Since the majority of missense mutations occurred C-terminal to aa1966, including all 10 mutations with absence of kinase activity, the implication was that mutations N-terminal to this, with exceptions, are less likely to result in loss of kinase activity and therefore, are less likely to be identified in A-T patients.


Subject(s)
Cell Cycle Proteins/genetics , DNA-Binding Proteins/genetics , Mutation, Missense , Protein Serine-Threonine Kinases/genetics , Tumor Suppressor Proteins/genetics , Ataxia Telangiectasia Mutated Proteins , Blotting, Western , Cell Cycle Proteins/metabolism , Cell Division , DNA Damage , DNA Repair , DNA-Binding Proteins/metabolism , Enzyme Activation , G2 Phase , Humans , Infrared Rays , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...