Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 171
Filter
1.
Oncogene ; 35(8): 965-76, 2016 Feb 25.
Article in English | MEDLINE | ID: mdl-25961918

ABSTRACT

Rad18 functions at the cross-roads of three different DNA damage response (DDR) pathways involved in protecting stressed replication forks: homologous recombination repair, DNA inter-strand cross-link repair and DNA damage tolerance. Although Rad18 serves to facilitate replication of damaged genomes by promoting translesion synthesis (TLS), this comes at a cost of potentially error-prone lesion bypass. In contrast, loss of Rad18-dependent TLS potentiates the collapse of stalled forks and leads to incomplete genome replication. Given the pivotal nature with which Rad18 governs the fine balance between replication fidelity and genome stability, Rad18 levels and activity have a major impact on genomic integrity. Here, we identify the de-ubiquitylating enzyme USP7 as a critical regulator of Rad18 protein levels. Loss of USP7 destabilizes Rad18 and compromises UV-induced PCNA mono-ubiquitylation and Pol η recruitment to stalled replication forks. USP7-depleted cells also fail to elongate nascent daughter strand DNA following UV irradiation and show reduced DNA damage tolerance. We demonstrate that USP7 associates with Rad18 directly via a consensus USP7-binding motif and can disassemble Rad18-dependent poly-ubiquitin chains both in vitro and in vivo. Taken together, these observations identify USP7 as a novel component of the cellular DDR involved in preserving the genome stability.


Subject(s)
DNA Damage , DNA Repair , DNA-Binding Proteins/metabolism , Ubiquitin Thiolesterase/metabolism , Ubiquitin-Protein Ligases/metabolism , Amino Acid Motifs , Cell Line , HeLa Cells , Humans , Protein Binding , Protein Stability , Ubiquitin/metabolism , Ubiquitin-Specific Peptidase 7
2.
Br J Dermatol ; 169(6): 1279-87, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23889214

ABSTRACT

BACKGROUND: Xeroderma pigmentosum (XP) is a rare autosomal recessive disorder of DNA repair. It is divided into eight complementation groups: XP-A to XP-G (classical XP) and XP variant (XP-V). Severe and prolonged sunburn reactions on minimal sun exposure have been considered a cardinal feature of classical XP. However, it has recently become clear that not all patients have abnormal sunburn reactions. OBJECTIVES: To examine sunburn reactions in a cohort of patients with XP and correlate this to the complementation group. METHODS: Sixty patients with XP attending the U.K. National XP Service from 2010 to 2012 were studied. Their history of burning after minimal sun exposure was assessed using a newly developed sunburn severity score. The age at which the first skin cancer was histologically diagnosed in each patient, and the presence of any neurological abnormality, was also recorded. RESULTS: Sunburn severity scores were abnormally high in patients with XP-A, XP-D, XP-F and XP-G compared with non-XP controls. There was no significant difference in sunburn score of patients with XP-C, XP-E and XP-V compared with controls (P > 0·05). Patients with XP-C, XP-E and XP-V were more likely to have skin cancer diagnosed at an earlier age than those with severe sunburn on minimal sun exposure. In addition, patients with XP with severe sunburn had an increased frequency of neurological abnormalities. CONCLUSIONS: Not all patients with XP have a history of severe and prolonged sunburn on minimal sun exposure. The normal sunburn response of patients with XP-C, XP-E and XP-V may relate to the preservation of transcription-coupled DNA repair in these groups. Those with a history of severe sunburn on minimal sun exposure developed their first skin cancer at an older age compared with patients with XP-C, XP-E and XP-V, but they had an increased frequency of neurological abnormalities. Physicians need to be aware that about half of all patients with XP will present without a history of abnormal sunburn.


Subject(s)
Sunburn/pathology , Xeroderma Pigmentosum/pathology , Adolescent , Adult , Age of Onset , Case-Control Studies , Female , Humans , Kaplan-Meier Estimate , Male , Melanoma/ethnology , Melanoma/mortality , Melanoma/pathology , Middle Aged , Nervous System Diseases/ethnology , Nervous System Diseases/mortality , Nervous System Diseases/pathology , Skin Neoplasms/ethnology , Skin Neoplasms/pathology , Sunburn/ethnology , Sunburn/mortality , Xeroderma Pigmentosum/ethnology , Xeroderma Pigmentosum/mortality , Young Adult
3.
Hum Mutat ; 31(2): 113-26, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19894250

ABSTRACT

Cockayne syndrome is an autosomal recessive multisystem disorder characterized principally by neurological and sensory impairment, cachectic dwarfism, and photosensitivity. This rare disease is linked to mutations in the CSB/ERCC6 and CSA/ERCC8 genes encoding proteins involved in the transcription-coupled DNA repair pathway. The clinical spectrum of Cockayne syndrome encompasses a wide range of severity from severe prenatal forms to mild and late-onset presentations. We have reviewed the 45 published mutations in CSA and CSB to date and we report 43 new mutations in these genes together with the corresponding clinical data. Among the 84 reported kindreds, 52 (62%) have mutations in the CSB gene. Many types of mutations are scattered along the whole coding sequence of both genes, but clusters of missense mutations can be recognized and highlight the role of particular motifs in the proteins. Genotype-phenotype correlation hypotheses are considered with regard to these new molecular and clinical data. Additional cases of molecular prenatal diagnosis are reported and the strategy for prenatal testing is discussed. Two web-based locus-specific databases have been created to list all identified variants and to allow the inclusion of future reports (www.umd.be/CSA/ and www.umd.be/CSB/).


Subject(s)
Cockayne Syndrome/genetics , DNA Helicases/genetics , DNA Repair Enzymes/genetics , Mutation/genetics , Transcription Factors/genetics , Amino Acid Sequence , Cockayne Syndrome/diagnosis , DNA Helicases/chemistry , DNA Repair Enzymes/chemistry , Databases, Genetic , Genetic Association Studies , Humans , Molecular Sequence Data , Poly-ADP-Ribose Binding Proteins , Polymorphism, Genetic , Sequence Alignment , Structure-Activity Relationship , Transcription Factors/chemistry
4.
Br J Radiol ; 81(961): 51-8, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18079351

ABSTRACT

We have examined our ionizing radiation survival data for 33 xeroderma pigmentosum (XP) primary fibroblast lines and compared the data to that of 53 normal fibroblast lines, 7 Cockayne syndrome (CS) lines, 4 combined XP/CS lines and 8 ataxia-telangiectasia fibroblast lines. Although there are differences in radiosensitivity between cell lines within each class, we have no convincing evidence that XP lines as a group are more sensitive to ionizing radiation than the general population. However, because the XP phenotype may lead to premature ageing, especially of sun-exposed tissues, we would still advocate caution when XP patients come to radiotherapy. Our results confirm the extreme ionizing radiation hypersensitivity of ataxia-telangiectasia; they are also consistent with a tendency for slight hypersensitivity in CS, but not (necessarily) in combined XP/CS.


Subject(s)
Fibroblasts/radiation effects , Radiation Tolerance , Xeroderma Pigmentosum/pathology , Ataxia Telangiectasia/pathology , Cell Line , Cell Survival/radiation effects , Cockayne Syndrome/pathology , Dose-Response Relationship, Drug , Gamma Rays , Humans
5.
Br J Dermatol ; 155(1): 81-8, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16792756

ABSTRACT

BACKGROUND: Xeroderma pigmentosum (XP) is an autosomal recessive disorder of, in most cases, defective nucleotide excision repair (NER) of ultraviolet radiation (UV)- and chemical-induced DNA damage. The condition is characterized by an increased sensitivity of the skin to UV radiation, with early development of pigmentary changes and premalignant lesions in sun-exposed areas of the skin, signs of photoageing and a greatly increased incidence from a young age of skin tumours including melanoma. Approximately 20% of patients with XP show neurological abnormalities of varying severity due to primary neuronal degeneration. Genetic analysis by somatic cell hybridization has led to the identification in the NER-defective form of XP of seven complementation groups, designated XP-A to XP-G. These complementation groups correspond to different proteins involved in the NER process. XP-A classically includes some of the most severely affected patients. OBJECTIVES: We describe a 61-year-old Punjabi woman with XP. Remarkably she had only mild cutaneous abnormalities, minimal neurological features and unusual longevity, and developed a malignant spindle cell melanoma. There are few previous reports of spindle cell melanoma associated with XP. To gain insight into the aetiology of these unusual features, we sought to analyse the DNA repair properties of the patient and identify the complementation group and the causative mutation in the defective gene. METHODS: Unscheduled DNA synthesis and the inhibition of RNA synthesis were measured. The complementation group was assigned by fusing the cells of our patient with XP cells of known complementation groups and determining the ability to carry out unscheduled DNA repair. Molecular analysis of the cDNA was carried out by polymerase chain reaction and DNA sequencing. RESULTS: Levels of DNA repair were extremely low and complementation analysis assigned the defect to the XP-A group. Sequencing of the XPA gene revealed a novel homozygous mutation of A-->G at the eighth nucleotide of intron 4 causing aberrant splicing and a nonfunctional truncated XP-A protein. However, a small amount of normally spliced mRNA was detected at <5% the level in normal cells. CONCLUSIONS: The small amount of normally spliced mRNA detected may be sufficient to explain the relatively mild clinical features in our patient.


Subject(s)
Melanoma/genetics , Point Mutation , Skin Neoplasms/genetics , Xeroderma Pigmentosum Group A Protein/genetics , Afghanistan/ethnology , DNA Repair , Female , Genetic Complementation Test , Homozygote , Humans , London , Melanoma/complications , Melanoma/pathology , Middle Aged , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Skin Neoplasms/complications , Skin Neoplasms/pathology , Xeroderma Pigmentosum/complications , Xeroderma Pigmentosum/genetics , Xeroderma Pigmentosum/pathology
6.
Br J Radiol ; 79(942): 510-7, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16714754

ABSTRACT

XP14BR is a cell line derived from a xeroderma pigmentosum (XP) patient from complementation group C. The patient was unusual in presenting with an angiosarcoma of the scalp, treated by surgical excision and radiotherapy. Following 38 Gy in 19 fractions with 6 MEV electrons, a severe desquamation and necrosis of the underlying bone ensued, and death followed 4 years later. The cell line was correspondingly hypersensitive to the lethal effects of gamma irradiation. We had previously shown that this sensitivity could be discriminated from that seen in ataxia-telangiectasia (A-T). The cellular response to ultraviolet radiation below 280 nm (UVC) was characteristic of XP cells, indicating the second instance, in our experience, of dual cellular UVC and ionizing radiation hypersensitivity in XP. We then set out to evaluate any defects in repair of ionizing radiation damage and to verify any direct contribution of the XPC gene. The cells were defective in repair of a fraction of double strand breaks, with a pattern reminiscent of A-T. The cell line was immortalized with the vector pSV3neo and the XPC cDNA transfected in to correct the defect. The progeny derived from this transfection showed the presence of the XPC gene product, as measured by immunoblotting. A considerable restoration of normal UVC, but not ionizing radiation, sensitivity was observed amongst the clones. This differential correction of cellular sensitivity is strong evidence for the presence of a defective radiosensitivity gene, distinct from XPC, which is responsible for the clinical hypersensitivity to ionizing radiation. It is important to resolve how widespread ionizing radiation sensitivity is amongst XP patients.


Subject(s)
Head and Neck Neoplasms/radiotherapy , Hemangiosarcoma/radiotherapy , Radiation Tolerance/genetics , Scalp , Skin Neoplasms/radiotherapy , Xeroderma Pigmentosum/complications , Cell Death/genetics , Cell Death/radiation effects , Cell Line, Tumor , DNA Damage/radiation effects , DNA Repair/radiation effects , DNA-Binding Proteins/genetics , Gamma Rays/adverse effects , Humans , Osteonecrosis/etiology , Parietal Bone/pathology , Parietal Bone/radiation effects , Radiation Injuries/genetics , Radiation Injuries/pathology , Transfection , Ultraviolet Rays/adverse effects , Xeroderma Pigmentosum/genetics
7.
J Biol Chem ; 278(52): 52223-30, 2003 Dec 26.
Article in English | MEDLINE | ID: mdl-14559922

ABSTRACT

Human papillomaviruses (HPV) are causative agents in a variety of human diseases; for example over 99% of cervical carcinomas contain HPV DNA sequences. Often in cervical carcinoma the HPV genome is integrated into the host genome resulting in unregulated expression of the viral transforming proteins E6 and E7. Therefore viral integration is a step toward HPV-induced carcinogenesis. Integration of the HPV genome could occur following double-strand DNA breaks that could arise during viral DNA replication. We investigated the fidelity of HPV 16 E1- and E2-mediated DNA replication of non-damaged and UVC-damaged templates in a variety of cell lines with different genetic backgrounds; C33a (derived from an HPV-negative cervical carcinoma), XP30RO (deficient in the by-pass polymerase eta (poleta)), XP30eta (expressing a restored wild-type poleta), XP12RO (nucleotide excision repair defective), and MRC5 (derived from a 14-week-old human fetus). The results demonstrate that the fidelity of E1- and E2-mediated DNA replication is reflective of the genetic background in which the assays are carried out. For example, restoring poleta to the XP30 cell line results in a 3-fold drop in the number of mutants obtained following replication of a UVC-damaged template. A relatively high percentage of the mutant-replicated molecules arise as a result of genetic rearrangement. This is the first time such studies have been carried out with an HPV replication system, and the results are discussed in the context of the HPV life cycle and what is known about HPV genomes in human cancers.


Subject(s)
DNA Replication , DNA-Binding Proteins , Oncogene Proteins, Viral/genetics , Blotting, Southern , Cell Line , Cell Line, Tumor , DNA/metabolism , DNA Damage , Escherichia coli/metabolism , Genome, Viral , Humans , Mutagenesis , Mutation , Plasmids/metabolism , Sequence Analysis, DNA , Transfection , Ultraviolet Rays
8.
Hum Mol Genet ; 10(24): 2797-802, 2001 Nov 15.
Article in English | MEDLINE | ID: mdl-11734544

ABSTRACT

The transcription factor TFIIH is involved in both basal transcription and DNA repair. Mutations in the XPD helicase component of TFIIH can result in the diverse clinical features associated with xeroderma pigmentosum (XP) and trichothiodystrophy (TTD). It is generally believed that the multi-system abnormalities associated with TTD are the result of a subtle deficiency in basal transcription. However, to date, there has been no clear demonstration of a defect in expression of any specific gene in individuals with these syndromes. Here we show that the specific mutations in XPD that cause TTD result in reduced expression of the beta-globin genes in these individuals. Eleven TTD patients with characterized mutations in the XPD gene have the haematological features of beta-thalassaemia trait, and reduced levels of beta-globin synthesis and beta-globin mRNA. All these parameters were normal in three patients with XP. These findings provide the first evidence for reduced expression of a specific gene in TTD. They support the hypothesis that many of the clinical features of TTD result from inadequate expression of a diverse set of highly expressed genes.


Subject(s)
Globins/genetics , Hair Diseases/complications , Hair Diseases/genetics , Mutation , Transcription Factors, TFII , Transcription Factors/genetics , beta-Thalassemia/genetics , Cells, Cultured , DNA Repair , Globins/biosynthesis , Haplotypes , Hematology , Humans , Reticulocytes , Transcription Factor TFIIH , Transcription Factors/physiology , Transcription, Genetic , Xeroderma Pigmentosum/genetics , beta-Thalassemia/complications
9.
Hum Mol Genet ; 10(22): 2539-47, 2001 Oct 15.
Article in English | MEDLINE | ID: mdl-11709541

ABSTRACT

The xeroderma pigmentosum group D (XPD) protein is a subunit of transcription factor TFIIH with DNA helicase activity. TFIIH has two functions, in basal transcription and nucleotide excision repair. Mutations in XPD that affect DNA repair but not transcription result in the skin cancer-prone disorder, xeroderma pigmentosum (XP). If transcription is also affected, the result is the multi-system disorder trichothiodystrophy (TTD), in which there is no skin cancer predisposition, or in rare cases, XP combined with Cockayne syndrome. Up till now there have been no reports of combined clinical features of XP and TTD. We have now identified two patients with some features of both these disorders. One of these, XP189MA, a 3-year-old girl with sun sensitivity, mental and physical developmental delay, has XPD mutations not previously reported, and barely detectable levels of nucleotide excision repair. The other, XP38BR, a 28-year-old woman with sun sensitivity, pigmentation changes and skin cancers typical of XP, has a mutation that has been identified previously, but only in TTD patients with no features of XP. The level of repair of UV damage in XP38BR is substantially higher than that in other patients with the same mutation. With both patients, polarized light microscopy revealed a 'tiger-tail' appearance of the hair, and amino acid analysis of the hair shafts show levels of sulfur-containing proteins intermediate between those of normal and TTD individuals. Our findings highlight the complexities of genotype-phenotype relationships in the XPD gene.


Subject(s)
DNA Helicases , DNA-Binding Proteins , Hair Diseases/genetics , Proteins/genetics , Transcription Factors , Xeroderma Pigmentosum/genetics , Adult , Amino Acid Sequence , Apoptosis/radiation effects , Base Sequence , Cell Survival/radiation effects , Cells, Cultured , Child, Preschool , DNA Mutational Analysis , DNA, Complementary/chemistry , DNA, Complementary/genetics , Dose-Response Relationship, Radiation , Female , Hair Diseases/pathology , Humans , Mutation , Photosensitivity Disorders/genetics , Photosensitivity Disorders/pathology , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Ultraviolet Rays , Xeroderma Pigmentosum/pathology , Xeroderma Pigmentosum Group D Protein
10.
Mol Biol Cell ; 12(6): 1583-94, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11408570

ABSTRACT

The structural maintenance of chromosomes (SMC) protein encoded by the fission yeast rad18 gene is involved in several DNA repair processes and has an essential function in DNA replication and mitotic control. It has a heterodimeric partner SMC protein, Spr18, with which it forms the core of a multiprotein complex. We have now isolated the human orthologues of rad18 and spr18 and designated them hSMC6 and hSMC5. Both proteins are about 1100 amino acids in length and are 27-28% identical to their fission yeast orthologues, with much greater identity within their N- and C-terminal globular domains. The hSMC6 and hSMC5 proteins interact to form a tight complex analogous to the yeast Rad18/Spr18 heterodimer. In proliferating human cells the proteins are bound to both chromatin and the nucleoskeleton. In addition, we have detected a phosphorylated form of hSMC6 that localizes to interchromatin granule clusters. Both the total level of hSMC6 and its phosphorylated form remain constant through the cell cycle. Both hSMC5 and hSMC6 proteins are expressed at extremely high levels in the testis and associate with the sex chromosomes in the late stages of meiotic prophase, suggesting a possible role for these proteins in meiosis.


Subject(s)
Cell Cycle Proteins/chemistry , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/metabolism , Fungal Proteins/chemistry , Saccharomyces cerevisiae Proteins , Schizosaccharomyces pombe Proteins , Schizosaccharomyces/chemistry , Amino Acid Sequence , Animals , Blotting, Northern , Blotting, Western , Cell Cycle Proteins/metabolism , Cell Division , Cell Line , Cell Line, Transformed , Cell Nucleus/metabolism , Cells, Cultured , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/chemistry , Cloning, Molecular , DNA, Complementary/metabolism , DNA-Binding Proteins/chemistry , Dimerization , Fibroblasts/metabolism , Humans , Meiosis , Mice , Microscopy, Fluorescence , Molecular Sequence Data , Phosphoric Monoester Hydrolases/metabolism , Phosphorylation , Protein Binding , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Tissue Distribution , Ubiquitin-Protein Ligases
11.
Nat Immunol ; 2(6): 537-41, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11376341

ABSTRACT

To determine whether DNA polymerase eta plays a role in the hypermutation of immunoglobulin variable genes, we examined the frequency and pattern of substitutions in variable VH6 genes from the peripheral blood lymphocytes of three patients with xeroderma pigmentosum variant disease, whose polymerase eta had genetic defects. The frequency of mutation was normal but the types of base changes were different: there was a decrease in mutations at A and T and a concomitant rise in mutations at G and C. We propose that more than one polymerase contributes to hypermutation and that if one is absent, others compensate. The data indicate that polymerase eta is involved in generating errors that occur predominantly at A and T and that another polymerase(s) may preferentially generate errors opposite G and C.


Subject(s)
DNA-Directed DNA Polymerase/metabolism , Genes, Immunoglobulin , Mutation , Animals , B-Lymphocytes/enzymology , B-Lymphocytes/immunology , Base Pairing , Base Sequence , DNA/genetics , DNA-Directed DNA Polymerase/genetics , Humans , Immunoglobulin Variable Region/genetics , Mice , Molecular Sequence Data , Xeroderma Pigmentosum/enzymology , Xeroderma Pigmentosum/genetics , Xeroderma Pigmentosum/immunology
12.
Genes Dev ; 15(2): 158-72, 2001 Jan 15.
Article in English | MEDLINE | ID: mdl-11157773

ABSTRACT

DNA polymerase eta carries out translesion synthesis past UV photoproducts and is deficient in xeroderma pigmentosum (XP) variants. We report that poleta is mostly localized uniformly in the nucleus but is associated with replication foci during S phase. Following treatment of cells with UV irradiation or carcinogens, it accumulates at replication foci stalled at DNA damage. The C-terminal third of poleta is not required for polymerase activity. However, the C-terminal 70 aa are needed for nuclear localization and a further 50 aa for relocalization into foci. Poleta truncations lacking these domains fail to correct the defects in XP-variant cells. Furthermore, we have identified mutations in two XP variant patients that leave the polymerase motifs intact but cause loss of the localization domains.


Subject(s)
DNA-Directed DNA Polymerase/genetics , DNA-Directed DNA Polymerase/metabolism , Xeroderma Pigmentosum/enzymology , Xeroderma Pigmentosum/genetics , Amino Acid Sequence , Base Sequence , Cell Line, Transformed , Cell Nucleus/enzymology , DNA/metabolism , DNA/radiation effects , DNA Damage , DNA Primers/genetics , DNA Repair , DNA-Binding Proteins/metabolism , DNA-Directed DNA Polymerase/chemistry , Genetic Variation , Green Fluorescent Proteins , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Molecular Sequence Data , Mutation , Proliferating Cell Nuclear Antigen/metabolism , Protein Structure, Tertiary , Rad51 Recombinase , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Deletion , Sequence Homology, Amino Acid , Ultraviolet Rays/adverse effects
14.
Adv Genet ; 43: 71-102, 2001.
Article in English | MEDLINE | ID: mdl-11037299

ABSTRACT

The genetic disorders xeroderma pigmentosum (XP), Cockayne syndrome (CS), and trichothiodystrophy (TTD) are all associated with defects in nucleotide excision repair (NER) of DNA damage. Their clinical features are very different, however, XP being a highly cancer-prone skin disorder, whereas CS and TTD are cancer-free multisystem disorders. All three are genetically complex, with at least eight complementation groups for XP (XP-A to -G and variant), five for CS (CS-A, CS-B, XP-B, XP-D, and XP-G), and three for TTD (XP-B, XP-D, and TTD-A). With the exception of the variant, the products of the XP genes are proteins involved in the different steps of NER, and comprise three damage-recognition proteins, two helicases, and two nucleases. The two helicases, XPB and XPD, are components of the basal transcription factor TFIIH, which has a dual role in NER and initiation of transcription. Different mutations in these genes can affect NER and transcription differentially, and this accounts for the different clinical phenotypes. Mutations resulting in defective repair without affecting transcription result in XP, whereas if transcription is also affected, TTD is the outcome. CS proteins are only involved in transcription-coupled repair, a subpathway of NER in which damage in the transcribed strands of active genes is rapidly and preferentially repaired. Current evidence suggests that they also have an important but not essential role in transcription. The variant form of XP is defective in a novel DNA polymerase, which is able to synthesise DNA past UV-damaged sites.


Subject(s)
DNA Repair , Transcription, Genetic , Xeroderma Pigmentosum/genetics , Cockayne Syndrome/genetics , Humans , Mutation , Oxygen/metabolism , Phenotype
15.
Teratog Carcinog Mutagen ; 20(6): 357-86, 2000.
Article in English | MEDLINE | ID: mdl-11074520

ABSTRACT

We report the results of a collaborative study aimed at developing reliable, direct assays for mutation in human cells. The project used common lymphoblastoid cell lines, both with and without mutagen treatment, as a shared resource to validate the development of new molecular methods for the detection of low-level mutations in the presence of a large excess of normal alleles. As the "gold standard, " hprt mutation frequencies were also measured on the same samples. The methods under development included i) the restriction site mutation (RSM) assay, in which mutations lead to the destruction of a restriction site; ii) minisatellite length-change mutation, in which mutations lead to alleles containing new numbers of tandem repeat units; iii) loss of heterozygosity for HLA epitopes, in which antibodies can be used to direct selection for mutant cells; iv) multiple fluorescence-based long linker arm nucleotides assay (mf-LLA) technology, for the detection of substitutional mutations; v) detection of alterations in the TP53 locus using a (CA) array as the target for the screening; and vi) PCR analysis of lymphocytes for the presence of the BCL2 t(14:18) translocation. The relative merits of these molecular methods are discussed, and a comparison made with more "traditional" methods.


Subject(s)
DNA Mutational Analysis/methods , Mutagenicity Tests/methods , Mutation , Base Sequence , Cell Line , Fluorescent Dyes , Genes, p53 , HLA-A Antigens/genetics , Humans , Hypoxanthine Phosphoribosyltransferase/genetics , Loss of Heterozygosity , Lymphocytes/drug effects , Lymphocytes/radiation effects , Minisatellite Repeats , Molecular Sequence Data , Point Mutation , Proto-Oncogene Proteins c-bcl-2/genetics , Restriction Mapping , Sensitivity and Specificity , Translocation, Genetic
17.
Curr Biol ; 10(8): 479-82, 2000 Apr 20.
Article in English | MEDLINE | ID: mdl-10801416

ABSTRACT

Checkpoints of DNA integrity are conserved throughout evolution, as are the kinases ATM (Ataxia Telangiectasia mutated) and ATR (Ataxia- and Rad-related), which are related to phosphatidylinositol (PI) 3-kinase [1] [2] [3]. The ATM gene is not essential, but mutations lead to ataxia telangiectasia (AT), a pleiotropic disorder characterised by radiation sensitivity and cellular checkpoint defects in response to ionising radiation [4] [5] [6]. The ATR gene has not been associated with human syndromes and, structurally, is more closely related to the canonical yeast checkpoint genes rad3(Sp) and MEC1(Sc) [7] [8]. ATR has been implicated in the response to ultraviolet (UV) radiation and blocks to DNA synthesis [8] [9] [10] [11], and may phosphorylate p53 [12] [13], suggesting that ATM and ATR may have similar and, perhaps, complementary roles in cell-cycle control after DNA damage. Here, we report that targeted inactivation of ATR in mice by disruption of the kinase domain leads to early embryonic lethality before embryonic day 8.5 (E8.5). Heterozygous mice were fertile and had no aberrant phenotype, despite a lower ATR mRNA level. No increase was observed in the sensitivity of ATR(+/-) embryonic stem (ES) cells to a variety of DNA-damaging agents. Attempts to target the remaining wild-type ATR allele in heterozygous ATR(+/-) ES cells failed, supporting the idea that loss of both alleles of the ATR gene, even at the ES-cell level, is lethal. Thus, in contrast to the closely related checkpoint gene ATM, ATR has an essential function in early mammalian development.


Subject(s)
Cell Cycle Proteins/physiology , Embryo Loss , Alleles , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/analysis , Cell Cycle Proteins/genetics , Cell Line , Cells, Cultured , Chimera , Chromosomes/chemistry , DNA/radiation effects , DNA Damage , DNA-Binding Proteins , Gamma Rays , Mice , Mitomycin/pharmacology , Protein Serine-Threonine Kinases/analysis , Protein Serine-Threonine Kinases/genetics , Stem Cells/cytology , Tumor Suppressor Proteins , Ultraviolet Rays
18.
Hum Mol Genet ; 9(8): 1171-5, 2000 May 01.
Article in English | MEDLINE | ID: mdl-10767341

ABSTRACT

Xeroderma pigmentosum (XP) and Cockayne syndrome (CS) are two hereditary disorders in which photosensitivity is associated with distinct clinical and cellular phenotypes and results from genetically different defects. We have identified the primary molecular alteration in two patients in whom clinical manifestations strongly reminiscent of a severe form of XP were unexpectedly associated with the CS cellular phenotype and with a defect in the CSB gene. Sequencing of the CSB -coding region in both cDNA and genomic DNA showed that these patients had identical alterations to those in a patient with the clinical features of the classical form of CS. These data, together with fluorescence in situ hybridization analysis, demonstrated that the two siblings with XP as well as the CS patient were homozygous for the same CSB mutated allele, containing a silent C2830T change and a nonsense mutation C2282T converting Arg735 to a stop codon. The finding that the same inactivating mutation underlies different pathological phenotypes indicates that there is no simple correlation between the molecular defect and the clinical features. Therefore, alterations in the CSB gene give rise to the same repair defect at the cellular level but other genetic and/or environmental factors determine the pathological phenotype.


Subject(s)
Cockayne Syndrome/genetics , DNA Helicases/genetics , Mutation, Missense , Point Mutation , Xeroderma Pigmentosum/genetics , Cells, Cultured , Codon, Terminator , DNA Repair Enzymes , Genetic Variation , Humans , In Situ Hybridization, Fluorescence , Nuclear Family , Poly-ADP-Ribose Binding Proteins
19.
Cancer Res ; 60(7): 1974-82, 2000 Apr 01.
Article in English | MEDLINE | ID: mdl-10766188

ABSTRACT

Xeroderma pigmentosum (XP)-C is one of the more common complementation groups of XP, but causative mutations have thus far been reported for only six cases (S. G. Khan et al., J. Investig. Dermatol., 115: 791-796, 1998; L. Li et al., Nat. Genet., 5: 413-417, 1993). We have now extended this analysis by investigating the genomic and coding sequence of the XPC gene, the level of expression of the XPC transcript and the status of the XPC protein in 12 unrelated patients, including all of the 8 Italian XP-C cases identified thus far and in 13 of their parents. Eighteen mutations were detected in the open reading frame of the XPC gene, 13 of which are relevant for the pathological phenotype. The mutations are distributed across the gene, with no indication of any hotspots or founder effects. Only 1 of the 13 relevant changes is a missense mutation, the remainder causing protein truncations as a result of nonsense mutations (3), frameshifts (6), deletion (1) or splicing abnormalities (2). These findings indicate that the XPC gene is not essential for cell proliferation and viability and that mutations causing minor structural alterations may not give an XP phenotype and may not, therefore, be identified clinically. XP13PV was the only patient carrying a missense mutation (Trp690Ser on the paternal allele). This was also the only patient in which the XPC transcript was present at a normal level and the XPC protein was detectable, although at a lower than normal level. No quantitative alterations in the transcript or protein levels were detected in the XP-C heterozygous parents. However, the expression of the normal allele predominated in all of them, except the father of XP13PV, which suggests the existence of a possible mechanism for monitoring the amount of the XPC protein.


Subject(s)
DNA-Binding Proteins/genetics , Mutation , Xeroderma Pigmentosum/genetics , Adolescent , Adult , Aged , Amino Acid Substitution , Cells, Cultured , Child , Child, Preschool , DNA Repair , Female , Fibroblasts/cytology , Fibroblasts/pathology , Fibroblasts/radiation effects , Gene Expression Regulation , Humans , Italy , Male , Nuclear Family , Open Reading Frames , Point Mutation , Polymorphism, Genetic , Sequence Deletion , Skin/cytology , Skin/pathology , Skin/radiation effects , Transcription, Genetic , Ultraviolet Rays , Xeroderma Pigmentosum/pathology
20.
EMBO J ; 19(7): 1691-702, 2000 Apr 03.
Article in English | MEDLINE | ID: mdl-10747036

ABSTRACT

In Schizosaccharomyces pombe, rad18 is an essential gene involved in the repair of DNA damage produced by ionizing radiation and in tolerance of UV-induced DNA damage. The Rad18 protein is a member of the SMC (structural maintenance of chromosomes) superfamily, and we show that, like the other SMC proteins in condensin and cohesin, Rad18 is a component of a high-molecular-weight complex. This complex contains at least six other proteins, the largest of which is Spr18, a novel SMC family member closely related to Rad18, and likely to be its heterodimeric partner. SMC proteins have ATP-binding domains at the N- and C-termini, and two extended coiled-coil domains separated by a hinge in the middle. We show that the N-terminal ATP-binding domain of Rad18 is essential for all functions, and overexpression of an N-terminal mutant has a dominant-negative effect. We have identified an important mutation (S1045A) near the C-terminus of Rad18 that separates its repair and essential roles. Potential models for the role of the Rad18-Spr18 complex during DNA repair are discussed.


Subject(s)
Cell Cycle Proteins , Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Fungal Proteins/chemistry , Fungal Proteins/metabolism , Saccharomyces cerevisiae Proteins , Schizosaccharomyces pombe Proteins , Schizosaccharomyces/metabolism , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Amino Acid Sequence , Chromosomal Proteins, Non-Histone/genetics , DNA Repair , DNA-Binding Proteins/genetics , Fungal Proteins/genetics , Genes, Fungal , Genetic Complementation Test , Macromolecular Substances , Molecular Sequence Data , Molecular Weight , Mutagenesis, Site-Directed , Phenotype , Schizosaccharomyces/genetics , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL