Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 175
Filter
1.
Nat Commun ; 11(1): 1120, 2020 02 28.
Article in English | MEDLINE | ID: mdl-32111838

ABSTRACT

The structure-specific endonuclease XPF-ERCC1 participates in multiple DNA damage repair pathways including nucleotide excision repair (NER) and inter-strand crosslink repair (ICLR). How XPF-ERCC1 is catalytically activated by DNA junction substrates is not currently understood. Here we report cryo-electron microscopy structures of both DNA-free and DNA-bound human XPF-ERCC1. DNA-free XPF-ERCC1 adopts an auto-inhibited conformation in which the XPF helical domain masks the ERCC1 (HhH)2 domain and restricts access to the XPF catalytic site. DNA junction engagement releases the ERCC1 (HhH)2 domain to couple with the XPF-ERCC1 nuclease/nuclease-like domains. Structure-function data indicate xeroderma pigmentosum patient mutations frequently compromise the structural integrity of XPF-ERCC1. Fanconi anaemia patient mutations in XPF often display substantial in-vitro activity but are resistant to activation by ICLR recruitment factor SLX4. Our data provide insights into XPF-ERCC1 architecture and catalytic activation.


Subject(s)
DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , DNA/metabolism , Endonucleases/chemistry , Endonucleases/metabolism , Binding Sites , Cryoelectron Microscopy , DNA-Binding Proteins/genetics , Endonucleases/genetics , Fanconi Anemia/enzymology , Fanconi Anemia/genetics , Humans , Models, Molecular , Mutation , Protein Conformation , Protein Domains , Protein Multimerization , Structure-Activity Relationship , Xeroderma Pigmentosum/enzymology , Xeroderma Pigmentosum/genetics
2.
RNA Biol ; 15(7): 845-848, 2018.
Article in English | MEDLINE | ID: mdl-29683386

ABSTRACT

Gene expression and DNA repair are fundamental processes for life. During the last decade, accumulating experimental evidence point towards different modes of coupling between these processes. Here we discuss the molecular mechanisms by which RNAPII-dependent transcription affects repair by the Nucleotide Excision Repair system (NER) and how NER activity, through the generation of single stranded DNA intermediates and activation of the DNA damage response kinase ATR, drives gene expression in a genotoxic scenario. Since NER-dependent repair is compromised in Xeroderma Pigmentosum (XP) patients, and having in mind that these patients present a high degree of clinical heterogeneity, we speculate that some of the clinical features of XP patients can be explained by misregulation of gene expression.


Subject(s)
DNA Repair/physiology , DNA, Single-Stranded/metabolism , Gene Expression/radiation effects , Xeroderma Pigmentosum/enzymology , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Cockayne Syndrome/enzymology , DNA Damage , DNA Helicases/genetics , Humans , Mutation , RNA Polymerase II/metabolism , Skin/radiation effects , Transcription, Genetic/physiology , Ultraviolet Rays/adverse effects
3.
Adv Exp Med Biol ; 996: 41-54, 2017.
Article in English | MEDLINE | ID: mdl-29124689

ABSTRACT

The sensitivity of Xeroderma pigmentosa (XP) patients to sunlight has spurred the discovery and genetic and biochemical analysis of the eight XP gene products (XPA-XPG plus XPV) responsible for this disorder. These studies also have served to elucidate the nucleotide excision repair (NER) process, especially the critical role played by the XPA protein. More recent studies have shown that NER also involves numerous other proteins normally employed in DNA metabolism and cell cycle regulation. Central among these is ataxia telangiectasia and Rad3-related (ATR), a protein kinase involved in intracellular signaling in response to DNA damage, especially DNA damage-induced replicative stresses. This review summarizes recent findings on the interplay between ATR as a DNA damage signaling kinase and as a novel ligand for intrinsic cell death proteins to delay damage-induced apoptosis, and on ATR's regulation of XPA and the NER process for repair of UV-induced DNA adducts. ATR's regulatory role in the cytosolic-to-nuclear translocation of XPA will be discussed. In addition, recent findings elucidating a non-NER role for XPA in DNA metabolism and genome stabilization at ds-ssDNA junctions, as exemplified in prematurely aging progeroid cells, also will be reviewed.


Subject(s)
Cell Nucleus/radiation effects , DNA Damage , DNA Repair/radiation effects , Sunlight/adverse effects , Ultraviolet Rays/adverse effects , Xeroderma Pigmentosum Group A Protein/metabolism , Xeroderma Pigmentosum/enzymology , Animals , Apoptosis/radiation effects , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Nucleus/enzymology , Cell Nucleus/pathology , Genetic Predisposition to Disease , Humans , Phenotype , Signal Transduction/radiation effects , Xeroderma Pigmentosum/genetics , Xeroderma Pigmentosum/pathology , Xeroderma Pigmentosum Group A Protein/genetics
4.
J Am Acad Dermatol ; 75(5): 855-870, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27745641

ABSTRACT

Hereditary photodermatoses are a spectrum of rare photosensitive disorders that are often caused by genetic deficiency or malfunction of various components of the DNA repair pathway. This results clinically in extreme photosensitivity, with many syndromes exhibiting an increased risk of cutaneous malignancies. This review will focus specifically on the syndromes with malignant potential, including xeroderma pigmentosum, Bloom syndrome, and Rothmund-Thomson syndrome. The typical phenotypic findings of each disorder will be examined and contrasted, including noncutaneous identifiers to aid in diagnosis. The management of these patients will also be discussed. At this time, the mainstay of therapy remains strict photoprotection; however, genetic therapies are under investigation.


Subject(s)
DNA Repair-Deficiency Disorders/genetics , Neoplastic Syndromes, Hereditary/genetics , Photosensitivity Disorders/genetics , Skin Neoplasms/genetics , Bloom Syndrome/enzymology , Bloom Syndrome/epidemiology , Bloom Syndrome/genetics , Bloom Syndrome/therapy , DNA Repair , DNA Repair Enzymes/deficiency , DNA Repair Enzymes/genetics , DNA Repair-Deficiency Disorders/epidemiology , Genes, Recessive , Genetic Predisposition to Disease , Humans , Neoplasms, Radiation-Induced/etiology , Neoplasms, Radiation-Induced/genetics , Neoplastic Syndromes, Hereditary/epidemiology , Phenotype , Proliferating Cell Nuclear Antigen/genetics , Rothmund-Thomson Syndrome/enzymology , Rothmund-Thomson Syndrome/epidemiology , Rothmund-Thomson Syndrome/genetics , Rothmund-Thomson Syndrome/therapy , Skin Neoplasms/etiology , Sunlight/adverse effects , Ultraviolet Rays/adverse effects , Xeroderma Pigmentosum/enzymology , Xeroderma Pigmentosum/epidemiology , Xeroderma Pigmentosum/genetics , Xeroderma Pigmentosum/therapy
5.
Photodermatol Photoimmunol Photomed ; 29(3): 132-9, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23651273

ABSTRACT

BACKGROUND: Xeroderma pigmentosum (XP) is a photosensitive genodermatosis with increased susceptibility to skin cancers. Patients are typically diagnosed with XP when they consult a dermatologist for skin cancers. CASE/METHODS: The genetic analysis and 2-8 years of follow-up for three school-age patients with XP-V is described. The patients were referred to us because of increased pigmented freckles; they had not experienced abnormal sunburn or developed skin cancer at their first visit. All patients harbored a genetic mutation in the POLH gene. XPV9KO was diagnosed at age 13 with a homozygous del1661A that creates a stop codon in the non-catalytic domain of POLH. The patient practiced sun protection, effectively preventing the development of skin cancer by age 21. XPV19KO was diagnosed at age 11 with a compound heterozygous mutation of G490T and C1066T, causing POLH truncation in the catalytic domain. This patient developed basal cell carcinoma at ages 12 and 13. XPV18KO was referred to us at age 11 and diagnosed with compound heterozygous variants of c.1246_1311del66 (exon 9 skipping), a novel mutation, and c.661_764 del104 (exon 6 skipping). CONCLUSION: Freckle-like pigmentation on sun-exposed skin is sometimes the only sign of XP-V, and early diagnosis is extremely important for children.


Subject(s)
Codon, Terminator , DNA-Directed DNA Polymerase/genetics , Heterozygote , Point Mutation , Xeroderma Pigmentosum/genetics , Adolescent , Child , DNA-Directed DNA Polymerase/metabolism , Exons , Female , Follow-Up Studies , Humans , Male , Protein Structure, Tertiary , Xeroderma Pigmentosum/enzymology , Xeroderma Pigmentosum/pathology
6.
Am J Hum Genet ; 92(5): 807-19, 2013 May 02.
Article in English | MEDLINE | ID: mdl-23623389

ABSTRACT

Cockayne syndrome (CS) is a genetic disorder characterized by developmental abnormalities and photodermatosis resulting from the lack of transcription-coupled nucleotide excision repair, which is responsible for the removal of photodamage from actively transcribed genes. To date, all identified causative mutations for CS have been in the two known CS-associated genes, ERCC8 (CSA) and ERCC6 (CSB). For the rare combined xeroderma pigmentosum (XP) and CS phenotype, all identified mutations are in three of the XP-associated genes, ERCC3 (XPB), ERCC2 (XPD), and ERCC5 (XPG). In a previous report, we identified several CS cases who did not have mutations in any of these genes. In this paper, we describe three CS individuals deficient in ERCC1 or ERCC4 (XPF). Remarkably, one of these individuals with XP complementation group F (XP-F) had clinical features of three different DNA-repair disorders--CS, XP, and Fanconi anemia (FA). Our results, together with those from Bogliolo et al., who describe XPF alterations resulting in FA alone, indicate a multifunctional role for XPF.


Subject(s)
Cockayne Syndrome/genetics , DNA-Binding Proteins/genetics , Endonucleases/genetics , Fanconi Anemia/genetics , Genetic Predisposition to Disease/genetics , Phenotype , Xeroderma Pigmentosum/genetics , Amino Acid Sequence , Base Sequence , Cockayne Syndrome/enzymology , Cockayne Syndrome/pathology , DNA Primers/genetics , Fanconi Anemia/enzymology , Fanconi Anemia/pathology , Fatal Outcome , Female , Humans , Male , Molecular Sequence Data , Sequence Analysis, DNA , Xeroderma Pigmentosum/enzymology , Xeroderma Pigmentosum/pathology
7.
Acta Crystallogr D Biol Crystallogr ; 69(Pt 2): 237-46, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23385459

ABSTRACT

XPB is a DNA-dependent helicase and a subunit of the TFIIH complex required for both transcription and DNA repair. XPB contains four domains: an N-terminal domain, two conserved helicase domains (HD1 and HD2) and a C-terminal extension. The C-terminal extension is important for DNA repair since the phosphorylation of Ser751 inhibits 5'-incision by ERCC1-XPF endonuclease. A disease-causing frameshift mutation (XP11BE) that changes the last 42 amino acids of XPB causes manifestations including impaired DNA repair and deficient transcription. Here, the crystal structure of the C-terminal half of XPB (residues 494-782) is reported at 1.8 Šresolution. The structure contained the conserved XPB HD2 and a C-terminal extension which shares structural similarity with RIG-I, leading to a structural model of the XPF-XPB-DNA complex for 5' incision during DNA repair. A mutation mimicking the XP11BE mutation produced the much less soluble mutant XPBm(494-781). Western blotting results confirmed that the intracellular levels of XPB and other TFIIH subunits in XP11BE patient cells were much lower than those from the healthy parents. Together, these results indicate that the XP11BE mutation not only divests the XPF-interaction motif, impairing DNA repair, but also reduces XPB solubility, leading to a lower intracellular level of TFIIH and deficient transcription.


Subject(s)
DNA Helicases/chemistry , DNA Repair/genetics , Frameshift Mutation , Peptide Fragments/chemistry , Xeroderma Pigmentosum/enzymology , Xeroderma Pigmentosum/genetics , Cells, Cultured , Crystallization , DEAD Box Protein 58 , DEAD-box RNA Helicases/chemistry , DEAD-box RNA Helicases/genetics , DNA Helicases/genetics , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Female , Humans , Male , Peptide Fragments/genetics , Protein Structure, Tertiary/genetics , Receptors, Immunologic , Xeroderma Pigmentosum/chemistry
8.
Nucleic Acids Res ; 41(5): 3079-93, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23345618

ABSTRACT

Trans-lesion DNA synthesis (TLS) is a DNA damage-tolerance mechanism that uses low-fidelity DNA polymerases to replicate damaged DNA. The inherited cancer-propensity syndrome xeroderma pigmentosum variant (XPV) results from error-prone TLS of UV-damaged DNA. TLS is initiated when the Rad6/Rad18 complex monoubiquitinates proliferating cell nuclear antigen (PCNA), but the basis for recruitment of Rad18 to PCNA is not completely understood. Here, we show that Rad18 is targeted to PCNA by DNA polymerase eta (Polη), the XPV gene product that is mutated in XPV patients. The C-terminal domain of Polη binds to both Rad18 and PCNA and promotes PCNA monoubiquitination, a function unique to Polη among Y-family TLS polymerases and dissociable from its catalytic activity. Importantly, XPV cells expressing full-length catalytically-inactive Polη exhibit increased recruitment of other error-prone TLS polymerases (Polκ and Polι) after UV irradiation. These results define a novel non-catalytic role for Polη in promoting PCNA monoubiquitination and provide a new potential mechanism for mutagenesis and genome instability in XPV individuals.


Subject(s)
DNA Replication , DNA-Binding Proteins/metabolism , DNA-Directed DNA Polymerase/physiology , Proliferating Cell Nuclear Antigen/metabolism , Ubiquitination , Amino Acid Sequence , Cell Line , Checkpoint Kinase 1 , Consensus Sequence , DNA Damage , DNA-Directed DNA Polymerase/genetics , DNA-Directed DNA Polymerase/metabolism , Enzyme Induction , Genomic Instability , Humans , Molecular Sequence Data , Protein Binding , Protein Kinases/metabolism , Protein Transport , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases , Xeroderma Pigmentosum/enzymology , Xeroderma Pigmentosum/genetics
9.
Cancer Res ; 73(6): 1844-54, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23288511

ABSTRACT

Xeroderma pigmentosum (XP), a UV-sensitivity syndrome characterized by skin hyperpigmentation, premature aging, and increased skin cancer, is caused by defects in the nucleotide excision repair (NER) pathway. XP shares phenotypical characteristics with telomere-associated diseases like Dyskeratosis congenita and mouse models with dysfunctional telomeres, including mice deficient for telomerase (Terc(-/-) mice). Thus, we investigated a hypothesized role for telomerase and telomere dysfunction in the pathobiology of XP by comparing Xpc(-/-)-mutant mice and Xpc(-/-)G1-G3Terc(-/-) double-mutant mice and exposed them to UV radiation. Chronically UV-exposed Xpc(-/-) skin displayed shorter telomeres on an average compared with wild-type skin. Strikingly, this effect was reversed by an additional deficiency in the telomerase. Moreover, aberrantly long telomeres were observed in the double-mutant mice. Telomere lengthening in the absence of telomerase suggested activation of the alternative lengthening of telomeres (ALT) in the UV-exposed skin of the double mutants. Mechanistic investigations revealed an elevated susceptibility for UV-induced p53 patches, known to represent precursor lesions of carcinomas, in Xpc(-/-)G1-G3Terc(-/-) mice where a high number of UV-induced skin tumors occurred that were characterized by aggressive growth. Taken together, our results establish a role for xeroderma pigmentosum, complementation group C (XPC) in telomere stability, particularly upon UV exposure. In absence of telomerase, critically short telomeres in XP mutants seem to aggravate this pathology, associated with an increased tumor incidence, by activating the ALT pathway of telomere lengthening.


Subject(s)
Telomerase/metabolism , Telomere , Ultraviolet Rays , Xeroderma Pigmentosum/enzymology , Xeroderma Pigmentosum/genetics , Animals , Immunohistochemistry , In Situ Hybridization, Fluorescence , Mice , Mice, Knockout , Neoplasms, Radiation-Induced/genetics
10.
Mutat Res ; 752(2): 138-152, 2013.
Article in English | MEDLINE | ID: mdl-23276657

ABSTRACT

Helicases have important roles in nucleic acid metabolism, and their prominence is marked by the discovery of genetic disorders arising from disease-causing mutations. Missense mutations can yield unique insight to molecular functions and basis for disease pathology. XPB or XPD missense mutations lead to Xeroderma pigmentosum, Cockayne's syndrome, Trichothiodystrophy, or COFS syndrome, suggesting that DNA repair and transcription defects are responsible for clinical heterogeneity. Complex phenotypes are also observed for RECQL4 helicase mutations responsible for Rothmund-Thomson syndrome, Baller-Gerold syndrome, or RAPADILINO. Bloom's syndrome causing missense mutations are found in the conserved helicase and RecQ C-terminal domain of BLM that interfere with helicase function. Although rare, patient-derived missense mutations in the exonuclease or helicase domain of Werner syndrome protein exist. Characterization of WRN separation-of-function mutants may provide insight to catalytic requirements for suppression of phenotypes associated with the premature aging disorder. Characterized FANCJ missense mutations associated with breast cancer or Fanconi anemia interfere with FANCJ helicase activity required for DNA repair and the replication stress response. For example, a FA patient-derived mutation in the FANCJ Iron-Sulfur domain was shown to uncouple its ATPase and translocase activity from DNA unwinding. Mutations in DDX11 (ChlR1) are responsible for Warsaw Breakage syndrome, a recently discovered autosomal recessive cohesinopathy. Ongoing and future studies will address clinically relevant helicase mutations and polymorphisms, including those that interfere with key protein interactions or exert dominant negative phenotypes (e.g., certain mutant alleles of Twinkle mitochondrial DNA helicase). Chemical rescue may be an approach to restore helicase activity in loss-of-function helicase disorders. Genetic and biochemical analyses of disease-causing missense mutations in human helicase disorders have led to new insights to the molecular defects underlying aberrant cellular and clinical phenotypes.


Subject(s)
Bloom Syndrome/genetics , Cockayne Syndrome/genetics , DNA Helicases/genetics , Fanconi Anemia/genetics , Mutation, Missense/genetics , Xeroderma Pigmentosum/genetics , Bloom Syndrome/enzymology , Bloom Syndrome/pathology , Cockayne Syndrome/enzymology , Cockayne Syndrome/pathology , Fanconi Anemia/enzymology , Fanconi Anemia/pathology , Humans , Xeroderma Pigmentosum/enzymology , Xeroderma Pigmentosum/pathology
11.
Am J Dermatopathol ; 35(1): 45-9, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22722466

ABSTRACT

Xeroderma pigmentosum (XP) is a heterogenous group of genetic diseases in which basal cell carcinoma (BCC) is the most common nonmelanoma skin cancer (NMSC) followed by squamous cell carcinoma (SCC). The aim of this study was to investigate the expression of matrix metalloproteinase (MMP)-13 and Ki-67 in SCC and BCC from patients with and without XP to elucidate their roles in the pathogenesis of these highly aggressive tumors in patients with XP. Immunolabeling using MMP-13 and Ki-67 antibodies was performed on tissue sections derived from skin biopsies of SCC and BCC of 15 patients with XP and 40 non-XP patients. There was no significant difference between XP and non-XP patients as regards MMP-13 expression by epithelial and stromal cells of SCC or BCC. Ki-67 expression in SCC and BCC of patients with XP was significantly higher than in non-XP patients. We concluded that the higher expression of Ki-67 in NMSC of patients with XP than of non-XP patients may reflect the growth and invasive capacity of these tumors in patients with XP. MMP-13 is expressed by tumor epithelial cells, stromal and inflammatory cells of NMSC of both XP and non-XP patients.


Subject(s)
Carcinoma, Basal Cell/chemistry , Carcinoma, Squamous Cell/chemistry , Ki-67 Antigen/analysis , Matrix Metalloproteinase 13/analysis , Skin Neoplasms/chemistry , Xeroderma Pigmentosum/chemistry , Adolescent , Biopsy , Carcinoma, Basal Cell/enzymology , Carcinoma, Basal Cell/immunology , Carcinoma, Basal Cell/pathology , Carcinoma, Squamous Cell/enzymology , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/pathology , Cell Proliferation , Child , Child, Preschool , Epithelial Cells/chemistry , Female , Humans , Immunohistochemistry , Male , Neoplasm Invasiveness , Skin Neoplasms/enzymology , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Stromal Cells/chemistry , Xeroderma Pigmentosum/enzymology , Xeroderma Pigmentosum/immunology , Xeroderma Pigmentosum/pathology , Young Adult
12.
Chem Res Toxicol ; 24(11): 1876-81, 2011 Nov 21.
Article in English | MEDLINE | ID: mdl-22007867

ABSTRACT

XPF-ERCC1 is a structure-specific endonuclease that is essential for nucleotide excision repair and DNA interstrand cross-link repair in mammalian cells. The yeast counterpart of XPF-ERCC1, Rad1-Rad10, plays multiple roles in DNA repair. Rad1-Rad10 is implicated to be involved in the repair of oxidative DNA damage. To explore the role(s) of XPF-ERCC1 in the repair of DNA damage induced by reactive oxygen species (ROS), cellular sensitivity of the XPF-deficient Chinese hamster ovary cell line UV41 to ROS was investigated. The XPF-deficient UV41 showed sensitivity to hydrogen peroxide, bleomycin, and paraquat. Furthermore, XPF-ERCC1 showed an ability to remove 3'-blocked ends such as 3'-phosphoglycolate from the 3'-end of DNA in vitro. These data suggest that XPF-ERCC1 plays a role in the repair of ROS-induced DNA damage by trimming 3'-blocked ends. The accumulation of various types of DNA damage, including ROS-induced DNA damage due to defects in multiple XPF-ERCC1-mediated DNA repair pathways, could contribute to the accelerated aging phenotypes observed in an XPF-ERCC1-deficient patient.


Subject(s)
DNA Repair , DNA-Binding Proteins/genetics , Endonucleases/genetics , Recombinant Proteins/metabolism , Xeroderma Pigmentosum/genetics , Animals , Bleomycin/adverse effects , Bleomycin/pharmacology , CHO Cells , Cell Survival/drug effects , Cricetinae , Cricetulus , DNA/genetics , DNA/metabolism , DNA Damage , Endonucleases/deficiency , Gene Deletion , Glycolates/chemistry , Glycolates/metabolism , Humans , Hydrogen Peroxide/adverse effects , Hydrogen Peroxide/pharmacology , Oxidation-Reduction , Paraquat/adverse effects , Paraquat/pharmacology , Plasmids , Reactive Oxygen Species/metabolism , Recombinant Proteins/genetics , Spodoptera , Transfection , Xeroderma Pigmentosum/enzymology
13.
Exp Mol Pathol ; 91(2): 528-33, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21640722

ABSTRACT

Deficiencies of DNA polymerase eta-an enzyme mediating replication past UV-induced DNA damage-predispose individuals to xeroderma pigmentosum variant (XPV) and result in a high incidence of skin cancers. We designed, developed and assessed several complementary molecular approaches to detect a genetically inherited deletion within DNA polymerase eta. RNA was reverse transcribed from XPV fibroblasts and from normal human cells, and standard polymerase chain reaction (PCR) was conducted on the cDNA targeting a region with a 13 base pair deletion within the polymerase eta gene. PCR products were subjected to restriction fragment length polymorphism (RFLP) analysis and cycle DNA sequencing. The deletion was found to eliminate a BsrGI restriction site and affected the number of resultant fragments visualized after gel electrophoresis. Cycle sequencing of polymerase eta-specific amplicons from XPV and normal cells provided a second approach for detecting the mutation. Additionally, the use of a fluorescent nucleic acid dye-EvaGreen-in real-time PCR and melt curve analysis distinguished normal and XPV patient-derived amplicons as well as heteroduplexes that represent heterozygotic carriers without the need for high resolution melt analysis-compatible software. Our approaches are easily adaptable by diagnostic laboratories that screen for or verify genetically inherited disorders and identify carriers of a defective gene.


Subject(s)
Base Pairing/genetics , DNA-Directed DNA Polymerase/genetics , Polymorphism, Genetic , Sequence Deletion/genetics , Xeroderma Pigmentosum/enzymology , Xeroderma Pigmentosum/genetics , Base Sequence , DNA Mutational Analysis , DNA, Complementary/genetics , Humans , Male , Molecular Sequence Data , Nucleic Acid Denaturation/genetics , Polymorphism, Restriction Fragment Length , Prostate/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Reverse Transcription/genetics
15.
Nature ; 465(7301): 1044-8, 2010 Jun 24.
Article in English | MEDLINE | ID: mdl-20577208

ABSTRACT

The variant form of the human syndrome xeroderma pigmentosum (XPV) is caused by a deficiency in DNA polymerase eta (Poleta), a DNA polymerase that enables replication through ultraviolet-induced pyrimidine dimers. Here we report high-resolution crystal structures of human Poleta at four consecutive steps during DNA synthesis through cis-syn cyclobutane thymine dimers. Poleta acts like a 'molecular splint' to stabilize damaged DNA in a normal B-form conformation. An enlarged active site accommodates the thymine dimer with excellent stereochemistry for two-metal ion catalysis. Two residues conserved among Poleta orthologues form specific hydrogen bonds with the lesion and the incoming nucleotide to assist translesion synthesis. On the basis of the structures, eight Poleta missense mutations causing XPV can be rationalized as undermining the molecular splint or perturbing the active-site alignment. The structures also provide an insight into the role of Poleta in replicating through D loop and DNA fragile sites.


Subject(s)
DNA-Directed DNA Polymerase/chemistry , DNA-Directed DNA Polymerase/metabolism , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/metabolism , Base Sequence , Biocatalysis , Catalytic Domain , Crystallography, X-Ray , DNA/chemistry , DNA/metabolism , DNA Damage , DNA-Directed DNA Polymerase/genetics , Humans , Kinetics , Models, Molecular , Mutation, Missense/genetics , Pyrimidine Dimers/genetics , Pyrimidine Dimers/metabolism , Structure-Activity Relationship , Xeroderma Pigmentosum/enzymology , Xeroderma Pigmentosum/genetics
16.
Nature ; 465(7301): 1039-43, 2010 Jun 24.
Article in English | MEDLINE | ID: mdl-20577207

ABSTRACT

DNA polymerase eta (Poleta) is unique among eukaryotic polymerases in its proficient ability for error-free replication through ultraviolet-induced cyclobutane pyrimidine dimers, and inactivation of Poleta (also known as POLH) in humans causes the variant form of xeroderma pigmentosum (XPV). We present the crystal structures of Saccharomyces cerevisiae Poleta (also known as RAD30) in ternary complex with a cis-syn thymine-thymine (T-T) dimer and with undamaged DNA. The structures reveal that the ability of Poleta to replicate efficiently through the ultraviolet-induced lesion derives from a simple and yet elegant mechanism, wherein the two Ts of the T-T dimer are accommodated in an active site cleft that is much more open than in other polymerases. We also show by structural, biochemical and genetic analysis that the two Ts are maintained in a stable configuration in the active site via interactions with Gln 55, Arg 73 and Met 74. Together, these features define the basis for Poleta's action on ultraviolet-damaged DNA that is crucial in suppressing the mutagenic and carcinogenic consequences of sun exposure, thereby reducing the incidence of skin cancers in humans.


Subject(s)
DNA-Directed DNA Polymerase/chemistry , DNA-Directed DNA Polymerase/metabolism , Saccharomyces cerevisiae/enzymology , Skin Neoplasms/enzymology , Biocatalysis , Catalytic Domain , Crystallography, X-Ray , DNA/chemistry , DNA/metabolism , DNA Damage , DNA-Directed DNA Polymerase/genetics , Humans , Kinetics , Models, Molecular , Mutation, Missense , Nucleic Acid Conformation , Protein Structure, Tertiary , Pyrimidine Dimers/chemistry , Pyrimidine Dimers/metabolism , Saccharomyces cerevisiae/genetics , Skin Neoplasms/genetics , Structure-Activity Relationship , Xeroderma Pigmentosum/enzymology , Xeroderma Pigmentosum/genetics
17.
PLoS Genet ; 6(3): e1000871, 2010 Mar 05.
Article in English | MEDLINE | ID: mdl-20221251

ABSTRACT

Xeroderma pigmentosum (XP) is caused by defects in the nucleotide excision repair (NER) pathway. NER removes helix-distorting DNA lesions, such as UV-induced photodimers, from the genome. Patients suffering from XP exhibit exquisite sun sensitivity, high incidence of skin cancer, and in some cases neurodegeneration. The severity of XP varies tremendously depending upon which NER gene is mutated and how severely the mutation affects DNA repair capacity. XPF-ERCC1 is a structure-specific endonuclease essential for incising the damaged strand of DNA in NER. Missense mutations in XPF can result not only in XP, but also XPF-ERCC1 (XFE) progeroid syndrome, a disease of accelerated aging. In an attempt to determine how mutations in XPF can lead to such diverse symptoms, the effects of a progeria-causing mutation (XPF(R153P)) were compared to an XP-causing mutation (XPF(R799W)) in vitro and in vivo. Recombinant XPF harboring either mutation was purified in a complex with ERCC1 and tested for its ability to incise a stem-loop structure in vitro. Both mutant complexes nicked the substrate indicating that neither mutation obviates catalytic activity of the nuclease. Surprisingly, differential immunostaining and fractionation of cells from an XFE progeroid patient revealed that XPF-ERCC1 is abundant in the cytoplasm. This was confirmed by fluorescent detection of XPF(R153P)-YFP expressed in Xpf mutant cells. In addition, microinjection of XPF(R153P)-ERCC1 into the nucleus of XPF-deficient human cells restored nucleotide excision repair of UV-induced DNA damage. Intriguingly, in all XPF mutant cell lines examined, XPF-ERCC1 was detected in the cytoplasm of a fraction of cells. This demonstrates that at least part of the DNA repair defect and symptoms associated with mutations in XPF are due to mislocalization of XPF-ERCC1 into the cytoplasm of cells, likely due to protein misfolding. Analysis of these patient cells therefore reveals a novel mechanism to potentially regulate a cell's capacity for DNA repair: by manipulating nuclear localization of XPF-ERCC1.


Subject(s)
DNA Repair , DNA-Binding Proteins/metabolism , Endonucleases/metabolism , Xeroderma Pigmentosum/enzymology , Amino Acid Substitution/genetics , Animals , CHO Cells , Cell Survival , Cricetinae , Cricetulus , DNA-Binding Proteins/genetics , Endonucleases/genetics , Fluorescent Antibody Technique , Humans , Mutation/genetics , Protein Transport , Recombinant Fusion Proteins/metabolism , Xeroderma Pigmentosum/genetics , Xeroderma Pigmentosum/pathology
18.
Proc Natl Acad Sci U S A ; 106(28): 11552-7, 2009 Jul 14.
Article in English | MEDLINE | ID: mdl-19564618

ABSTRACT

Human cells tolerate UV-induced cyclobutane pyrimidine dimers (CPD) by translesion DNA synthesis (TLS), carried out by DNA polymerase eta, the POLH gene product. A deficiency in DNA polymerase eta due to germ-line mutations in POLH causes the hereditary disease xeroderma pigmentosum variant (XPV), which is characterized by sunlight sensitivity and extreme predisposition to sunlight-induced skin cancer. XPV cells are UV hypermutable due to the activity of mutagenic TLS across CPD, which explains the cancer predisposition of the patients. However, the identity of the backup polymerase that carries out this mutagenic TLS was unclear. Here, we show that DNA polymerase zeta cooperates with DNA polymerases kappa and iota to carry out error-prone TLS across a TT CPD. Moreover, DNA polymerases zeta and kappa, but not iota, protect XPV cells against UV cytotoxicity, independently of nucleotide excision repair. This presents an extreme example of benefit-risk balance in the activity of TLS polymerases, which provide protection against UV cytotoxicity at the cost of increased mutagenic load.


Subject(s)
DNA Repair/genetics , DNA-Directed DNA Polymerase/metabolism , DNA/biosynthesis , Pyrimidine Dimers/metabolism , Xeroderma Pigmentosum/genetics , Cells, Cultured , Humans , Models, Genetic , Ultraviolet Rays , Xeroderma Pigmentosum/enzymology
19.
Mutat Res ; 663(1-2): 69-76, 2009 Apr 26.
Article in English | MEDLINE | ID: mdl-19428372

ABSTRACT

The p53 tumour suppressor is a transcription factor that can either activate or repress the expression of specific genes in response to cellular stresses such as exposure to ultraviolet light. The p53 protein can exert both pro- and anti-apoptotic effects depending on cellular context. In primary human fibroblasts, p53 protects cells from UV-induced apoptosis at moderate doses but this is greatly affected by the nucleotide excision repair (NER) capacity of the cells. The damage-specific DNA binding protein 2 (DDB2) is involved in NER and is associated with xeroderma pigmentosum subgroup E (XP-E). Importantly, DDB2 is also positively regulated by the p53 protein. To study the potential interplay between DDB2 and p53 in determining the apoptotic response of primary fibroblasts exposed to UV light, the expression of these proteins was manipulated in primary normal and XP-E fibroblast strains using human papillomavirus E6 protein (HPV-E6), RNA interference and recombinant adenoviruses expressing either p53 or DDB2. Normal and XP-E fibroblast strains were equally sensitive to UV-induced apoptosis over a broad range of doses and disruption of p53 in these strains using HPV-E6 or RNA interference led to a similar increase in apoptosis following exposure to UV light. In contrast, forced expression of p53 or DDB2 did not affect UV-induced apoptosis greatly in these normal or XP-E fibroblast strains. Collectively, these results indicate that p53 is primarily protective against UV-induced apoptosis in primary human fibroblasts and this activity of p53 does not require DDB2.


Subject(s)
Apoptosis/radiation effects , DNA-Binding Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Ultraviolet Rays , Caspase 3/metabolism , Caspase 9/metabolism , Enzyme Activation/radiation effects , Fibroblasts/enzymology , Fibroblasts/pathology , Fibroblasts/radiation effects , Humans , Male , RNA Interference/radiation effects , Viral Proteins/metabolism , Xeroderma Pigmentosum/enzymology , Xeroderma Pigmentosum/pathology
20.
J Immunol ; 182(10): 6353-9, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19414788

ABSTRACT

Patients with the variant form of xeroderma pigmentosum (XPV) syndrome have a genetic deficiency in DNA polymerase (Pol) eta, and display accordingly an increased skin sensitivity to UV light, as well as an altered mutation pattern of their Ig V genes in memory B cells, alteration that consists in a reduced mutagenesis at A/T bases. We previously suggested that another polymerase with a different mutation signature, Pol kappa, is used as backup for Ig gene hypermutation in both humans and mice in cases of complete Pol eta deficiency, a proposition supported in this study by the analysis of Pol eta x Pol kappa double-deficient mice. We also describe a new XPV case, in which a splice site mutation of the first noncoding exon results in a decreased mRNA expression, a mRNA that otherwise encodes a normal Pol eta protein. Whereas the Pol eta mRNA level observed in patient's fibroblasts is one-twentieth the value of healthy controls, it is only reduced to one-fourth of the normal level in activated B cells. Memory B cells from this patient showed a 50% reduction in A/T mutations, with a spectrum that still displays a strict Pol eta signature. Pol eta thus appears as a dominant enzyme in hypermutation, its presence precluding the use of a substitute enzyme even in conditions of reduced availability. Such a dominant behavior may explain the lack of Pol kappa signature in Ig gene mutations of some XPV patients previously described, for whom residual Pol eta activity might exist.


Subject(s)
B-Lymphocytes/enzymology , DNA-Directed DNA Polymerase/genetics , Somatic Hypermutation, Immunoglobulin/genetics , Xeroderma Pigmentosum/genetics , Adult , Animals , B-Lymphocytes/immunology , DNA Mutational Analysis , DNA-Directed DNA Polymerase/immunology , Female , Humans , Mice , Mice, Knockout , Mutation , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Xeroderma Pigmentosum/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...