Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 85
Filter
1.
Proc Natl Acad Sci U S A ; 117(25): 14412-14420, 2020 06 23.
Article in English | MEDLINE | ID: mdl-32513688

ABSTRACT

Nucleotide excision repair (NER) removes helix-destabilizing adducts including ultraviolet (UV) lesions, cyclobutane pyrimidine dimers (CPDs), and pyrimidine (6-4) pyrimidone photoproducts (6-4PPs). In comparison with CPDs, 6-4PPs have greater cytotoxicity and more strongly destabilizing properties of the DNA helix. It is generally believed that NER is the only DNA repair pathway that removes the UV lesions as evidenced by the previous data since no repair of UV lesions was detected in NER-deficient skin fibroblasts. Topoisomerase I (TOP1) constantly creates transient single-strand breaks (SSBs) releasing the torsional stress in genomic duplex DNA. Stalled TOP1-SSB complexes can form near DNA lesions including abasic sites and ribonucleotides embedded in chromosomal DNA. Here we show that base excision repair (BER) increases cellular tolerance to UV independently of NER in cancer cells. UV lesions irreversibly trap stable TOP1-SSB complexes near the UV damage in NER-deficient cells, and the resulting SSBs activate BER. Biochemical experiments show that 6-4PPs efficiently induce stable TOP1-SSB complexes, and the long-patch repair synthesis of BER removes 6-4PPs downstream of the SSB. Furthermore, NER-deficient cancer cell lines remove 6-4PPs within 24 h, but not CPDs, and the removal correlates with TOP1 expression. NER-deficient skin fibroblasts weakly express TOP1 and show no detectable repair of 6-4PPs. Remarkably, the ectopic expression of TOP1 in these fibroblasts led them to completely repair 6-4PPs within 24 h. In conclusion, we reveal a DNA repair pathway initiated by TOP1, which significantly contributes to cellular tolerance to UV-induced lesions particularly in malignant cancer cells overexpressing TOP1.


Subject(s)
DNA Breaks, Single-Stranded/radiation effects , DNA Repair , DNA Topoisomerases, Type I/metabolism , Ultraviolet Rays/adverse effects , CRISPR-Cas Systems/genetics , DNA Polymerase beta/genetics , DNA Polymerase beta/metabolism , Fibroblasts , Gene Knockout Techniques , Humans , MCF-7 Cells , Primary Cell Culture , Skin/cytology , Skin/pathology , Skin/radiation effects , X-ray Repair Cross Complementing Protein 1/genetics , X-ray Repair Cross Complementing Protein 1/metabolism , Xeroderma Pigmentosum/etiology , Xeroderma Pigmentosum/pathology , Xeroderma Pigmentosum Group A Protein/genetics , Xeroderma Pigmentosum Group A Protein/metabolism
2.
Nucleic Acids Res ; 48(4): 1941-1953, 2020 02 28.
Article in English | MEDLINE | ID: mdl-31853541

ABSTRACT

UVA-induced mutagenesis was investigated in human pol eta-deficient (XP-V) cells through whole-exome sequencing. In UVA-irradiated cells, the increase in the mutation frequency in deficient cells included a remarkable contribution of C>T transitions, mainly at potential pyrimidine dimer sites. A strong contribution of C>A transversions, potentially due to oxidized bases, was also observed in non-irradiated XP-V cells, indicating that basal mutagenesis caused by oxidative stress may be related to internal tumours in XP-V patients. The low levels of mutations involving T induced by UVA indicate that pol eta is not responsible for correctly replicating T-containing pyrimidine dimers, a phenomenon known as the 'A-rule'. Moreover, the mutation signature profile of UVA-irradiated XP-V cells is highly similar to the human skin cancer profile, revealing how studies involving cells deficient in DNA damage processing may be useful to understand the mechanisms of environmentally induced carcinogenesis.


Subject(s)
Mutagenesis/genetics , Oxidative Stress/genetics , Pyrimidine Dimers/genetics , Xeroderma Pigmentosum/genetics , Cell Line , DNA Damage/radiation effects , DNA Repair/radiation effects , DNA Replication/radiation effects , Humans , Mutagenesis/radiation effects , Mutation/genetics , Mutation/radiation effects , Oxidative Stress/radiation effects , Pyrimidine Dimers/radiation effects , Ultraviolet Rays , Exome Sequencing , Xeroderma Pigmentosum/etiology
3.
Cell Biol Int ; 42(6): 643-650, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29271530

ABSTRACT

DNA is constantly being damaged, either by endogenous or exogenous genotoxins. In that regard, DNA repair activities are essential for maintaining genomic stability and to life itself. Mutations in genes encoding DNA repair proteins cause severe human syndromes, but DNA repair defects have also been linked to several other diseases, notably to cancer and normal aging. Recently, new evidence has emerged indicating that some DNA repair diseases display mitochondrial and metabolic dysfunction through mechanisms that are yet being uncovered. These results suggest that mitochondria play an import role in the DNA damage response pathways and that damage accumulation may lead to mitochondrial dysfunction via metabolic imbalance and mitophagy impairment. Here we review the recent findings linking mitochondrial impairment and cell death to DNA damage accumulation in the context of DNA repair defects. In addition, the general involvement of DNA damage in cellular dysfunction suggests that these phenomena may be also involved in other human pathologies in which mitochondrial dysfunction and metabolic disruption play causative roles.


Subject(s)
Congenital Abnormalities/etiology , DNA Repair , Mitochondria/metabolism , Animals , Ataxia Telangiectasia/etiology , Ataxia Telangiectasia/genetics , Cockayne Syndrome/etiology , Cockayne Syndrome/genetics , Congenital Abnormalities/genetics , DNA Damage , Humans , Mitophagy , Xeroderma Pigmentosum/etiology , Xeroderma Pigmentosum/genetics
5.
Hautarzt ; 68(5): 359-363, 2017 May.
Article in German | MEDLINE | ID: mdl-28401271

ABSTRACT

Xeroderma pigmentosum is a rare autosomal recessive disorder which is caused by germinal mutations responsible for the repair of ultraviolet (UV) radiation-induced DNA lesions. It is characterized by hypersensitivity to UV radiation, poikiloderma, ocular surface disease, and in some patients pronounced sunburn and neurological disease. Patients have a very high risk of developing ocular and skin cancer on exposed body sites. No cure is available for these patients except complete protection from all types of UV radiation.


Subject(s)
Antioxidants/administration & dosage , Radiation Protection/methods , Sunlight/adverse effects , Sunscreening Agents/administration & dosage , Xeroderma Pigmentosum/etiology , Xeroderma Pigmentosum/prevention & control , Antioxidants/chemistry , Dermatology/trends , Evidence-Based Medicine , Humans , Radiation-Protective Agents/administration & dosage , Radiation-Protective Agents/chemistry , Solar Energy , Sunscreening Agents/chemistry , Treatment Outcome
6.
Photochem Photobiol ; 93(1): 245-258, 2017 01.
Article in English | MEDLINE | ID: mdl-27645605

ABSTRACT

Melanoma is the deadliest form of skin cancer because of its propensity to spread beyond the primary site of disease and because it resists many forms of treatment. Incidence of melanoma has been increasing for decades. Although ultraviolet radiation (UV) has been identified as the most important environmental causative factor for melanoma development, UV-protective strategies have had limited efficacy in melanoma prevention. UV mutational burden correlates with melanoma development and tumor progression, underscoring the importance of UV in melanomagenesis. However, besides amount of UV exposure, melanocyte UV mutational load is influenced by the robustness of nucleotide excision repair, the genome maintenance pathway charged with removing UV photoproducts before they cause permanent mutations in the genome. In this review, we highlight the importance of the melanocortin hormonal signaling axis on regulating efficiency of nucleotide excision repair in melanocytes. By understanding the molecular mechanisms by which nucleotide excision repair can be increased, it may be possible to prevent many cases of melanoma by reducing UV mutational burden over time.


Subject(s)
DNA Repair , Melanocortins/metabolism , Melanocytes/metabolism , Pyrimidine Dimers/metabolism , Signal Transduction , Ultraviolet Rays/adverse effects , Cyclic AMP/metabolism , Humans , Melanoma/epidemiology , Receptor, Melanocortin, Type 1/agonists , Receptor, Melanocortin, Type 1/antagonists & inhibitors , Receptor, Melanocortin, Type 1/metabolism , United States/epidemiology , Xeroderma Pigmentosum/etiology
7.
Int J Mol Sci ; 17(11)2016 Nov 04.
Article in English | MEDLINE | ID: mdl-27827925

ABSTRACT

Ultraviolet (UV) radiation from sunlight represents a constant threat to genome stability by generating modified DNA bases such as cyclobutane pyrimidine dimers (CPD) and pyrimidine-pyrimidone (6-4) photoproducts (6-4PP). If unrepaired, these lesions can have deleterious effects, including skin cancer. Mammalian cells are able to neutralize UV-induced photolesions through nucleotide excision repair (NER). The NER pathway has multiple components including seven xeroderma pigmentosum (XP) proteins (XPA to XPG) and numerous auxiliary factors, including ataxia telangiectasia and Rad3-related (ATR) protein kinase and RCC1 like domain (RLD) and homologous to the E6-AP carboxyl terminus (HECT) domain containing E3 ubiquitin protein ligase 2 (HERC2). In this review we highlight recent data on the transcriptional and posttranslational regulation of NER activity.


Subject(s)
DNA Repair , Protein Processing, Post-Translational , Skin Neoplasms/genetics , Transcription, Genetic , Ultraviolet Rays/adverse effects , Xeroderma Pigmentosum/genetics , Animals , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , DNA Damage , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Humans , Protein Isoforms/genetics , Protein Isoforms/metabolism , Pyrimidine Dimers/metabolism , Signal Transduction , Skin Neoplasms/etiology , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Ubiquitin-Protein Ligases , Xeroderma Pigmentosum/etiology , Xeroderma Pigmentosum/metabolism , Xeroderma Pigmentosum/pathology , Xeroderma Pigmentosum Group A Protein/genetics , Xeroderma Pigmentosum Group A Protein/metabolism
8.
Sci Context ; 28(3): 427-64, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26256506

ABSTRACT

This paper brings together the history of risk and the history of DNA repair, a biological phenomenon that emerged as a research field in between molecular biology, genetics, and radiation research in the 1960s. The case of xeroderma pigmentosum (XP), an inherited hypersensitivity to UV light and, hence, a disposition to skin cancer will be the starting point to argue that, in the 1970s and 1980s, DNA repair became entangled in the creation of new models of the human body at risk - what is here conceptually referred to as the vulnerability aspect of body history - and new attempts at cancer prevention and enhancement of the body associated with the new flourishing research areas of antimutagenesis and anticarcinogenesis. The aim will be to demonstrate that DNA repair created special attempts at disease prevention: molecular enhancement, seeking to identify means to increase the self-repair abilities of the body at the molecular level. Prevention in this sense meant enhancing the body's ability to cope with the environmental hazards of an already toxic world. This strategy has recently been adopted by the beauty industry, which introduced DNA care as a new target for skin care research and anti-aging formulas.


Subject(s)
DNA Repair , Genetics/history , Molecular Biology/history , Radiologic Health/history , Xeroderma Pigmentosum/history , History, 20th Century , History, 21st Century , Humans , Risk Assessment/history , Xeroderma Pigmentosum/etiology
9.
Photochem Photobiol ; 91(2): 254-64, 2015.
Article in English | MEDLINE | ID: mdl-25534312

ABSTRACT

Ultraviolet (UV) radiation from sunlight is a major etiologic factor for skin cancer, the most prevalent cancer in the United States, as well as premature skin aging. In particular, UVB radiation causes formation of specific DNA damage photoproducts between pyrimidine bases. These DNA damage photoproducts are repaired by a process called nucleotide excision repair, also known as UV-induced DNA repair. When left unrepaired, UVB-induced DNA damage leads to accumulation of mutations, predisposing people to carcinogenesis as well as to premature aging. Genetic loss of nucleotide excision repair leads to severe disorders, namely, xeroderma pigmentosum (XP), trichothiodystrophy (TTD) and Cockayne syndrome (CS), which are associated with predisposition to skin carcinogenesis at a young age as well as developmental and neurological conditions. Regulation of nucleotide excision repair is an attractive avenue to preventing or reversing these detrimental consequences of impaired nucleotide excision repair. Here, we review recent studies on molecular mechanisms regulating nucleotide excision repair by extracellular cues and intracellular signaling pathways, with a special focus on the molecular regulation of individual repair factors.


Subject(s)
Aging/radiation effects , Cockayne Syndrome/metabolism , DNA Repair , Skin Neoplasms/metabolism , Trichothiodystrophy Syndromes/metabolism , Ultraviolet Rays/adverse effects , Xeroderma Pigmentosum/metabolism , Aging/genetics , Aging/metabolism , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cockayne Syndrome/etiology , Cockayne Syndrome/genetics , Cockayne Syndrome/pathology , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Humans , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Signal Transduction , Skin Neoplasms/etiology , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Trichothiodystrophy Syndromes/etiology , Trichothiodystrophy Syndromes/genetics , Trichothiodystrophy Syndromes/pathology , Xeroderma Pigmentosum/etiology , Xeroderma Pigmentosum/genetics , Xeroderma Pigmentosum/pathology
10.
J Environ Pathol Toxicol Oncol ; 33(4): 279-93, 2014.
Article in English | MEDLINE | ID: mdl-25404376

ABSTRACT

The objective of the present work was to study the fine kinetics of DNA repair in xeroderma pigmentosum (XP) syndrome, a complex disorder linked to a deficiency in repair that increases cancer susceptibility. The repair process was evaluated by the comet assay (CA) in cells from 2 XP patients and 9 controls exposed to UVA/B (UVA 366/UVB 280 nm) and H2O2 (150 µM) at temperatures of 4, 15, and 37°C. Samples were taken at 2-min intervals during the first 10 min to analyze the "fine kinetics" repair during the initial phase of the curve, and then at 15, 20, 25, 30, 45, 60, and 120 min. CA evaluation of DNA repair activity points to BER/NER initiation in the first 30 min with both inductors at 37°C and 15°C, but final comet length showed differences according to treatment. Repair kinetics during 120 min showed a good correlation with clinical features in both XP patients. Differences in final comet length were less pronounced in XP cells treated with H2O2 than with UVA/B, probably because the peroxide produces mainly base oxidation but less bulky lesions; UVA/B generates a mixture of both. These findings reinforce the value of CA in testing in DNA repair ability or exposure monitoring.


Subject(s)
DNA Repair , Hydrogen Peroxide/toxicity , Leukocytes/metabolism , Ultraviolet Rays/adverse effects , Xeroderma Pigmentosum/genetics , Adult , Child , Comet Assay , DNA Damage/drug effects , DNA Damage/radiation effects , Female , Humans , Kinetics , Leukocytes/drug effects , Leukocytes/pathology , Leukocytes/radiation effects , Male , Time Factors , Xeroderma Pigmentosum/chemically induced , Xeroderma Pigmentosum/etiology , Xeroderma Pigmentosum/metabolism
11.
Exp Dermatol ; 23 Suppl 1: 43-6, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25234836

ABSTRACT

The young facial skin of children with a smooth healthy appearance changes over time to photoaged skin having mottled pigmentation, solar lentigines, wrinkles, dry and rough skin, leathery texture, and benign and malignant tumors after exposure to chronic, repeated solar radiation. The first sign of photoaging in Japanese subjects is usually solar lentigines appearing around 20 years of age on the face. Fine wrinkles can then appear after 30 years of age, and benign skin tumors, seborrhoeic keratoses, can occur after 35 years of age in sun-exposed skin. We theoretically calculated the maximal daily exposure time to solar radiation, which could prevent the development of photoaged skin until 60 and 80 years of age, based on published data of personal solar UVB doses in sun-exposed skin. One MED (minimal erythema dose) was determined to be 20 mJ/cm(2) , and 200 MED was used as the average yearly dose of Japanese children. Further, we hypothesized that the annual dose of Japanese adults is the same as that of the children. The cumulative UVB dose at 20 years of age was thus calculated to be 4000 MED, and 22 MED was used as the maximal daily UVB dose based on data measured in Kobe, located in the central area of Japan. We used the solar UVB dose from 10:00 a.m. to 14:00 p.m. which occupies 60% of the total daily UV dose, to obtain the maximal UVB per hour in a day, and calculated the maximal daily UV exposure time that would delay the onset of solar lentigines until 60 or 80 years of age. The mean daily sun exposure time to maintain healthy skin until 80 years of age in the summer was calculated to be 2.54 min (0.14 MED) for unprotected skin and 127 min with the use of a sunscreen of SPF (sun protection factor) of 50. In this study, we did not evaluate the photoaging effect of UVA radiation, but findings of the adverse effects of UVA radiation on the skin have accumulated in the last decade. Therefore, it will be important to estimate the maximal dose of solar UV radiation to retard the onset of photoaging based on an evaluation of both solar UVB and UVA in the future. Finally, we expect that this study may contribute to keeping Japanese and other types of skin young and healthy by limiting the exposure of the skin to solar radiation outdoors during the day.


Subject(s)
Skin Aging , Skin/radiation effects , Ultraviolet Rays/adverse effects , Adult , Aged , Asian People , Child , Dose-Response Relationship, Radiation , Humans , Infant , Lentigo/etiology , Models, Biological , Skin/pathology , Skin Aging/pathology , Sunlight/adverse effects , Sunscreening Agents/administration & dosage , Xeroderma Pigmentosum/etiology , Xeroderma Pigmentosum/pathology
14.
J Biomed Sci ; 20: 70, 2013 Sep 24.
Article in English | MEDLINE | ID: mdl-24063568

ABSTRACT

BACKGROUND: Xeroderma Pigmentosum (XP) is a rare skin disorder characterized by skin hypersensitivity to sunlight and abnormal pigmentation. The aim of this study was to investigate the genetic cause of a severe XP phenotype in a consanguineous Pakistani family and in silico characterization of any identified disease-associated mutation. RESULTS: The XP complementation group was assigned by genotyping of family for known XP loci. Genotyping data mapped the family to complementation group A locus, involving XPA gene. Mutation analysis of the candidate XP gene by DNA sequencing revealed a novel deletion mutation (c.654del A) in exon 5 of XPA gene. The c.654del A, causes frameshift, which pre-maturely terminates protein and result into a truncated product of 222 amino acid (aa) residues instead of 273 (p.Lys218AsnfsX5). In silico tools were applied to study the likelihood of changes in structural motifs and thus interaction of mutated protein with binding partners. In silico analysis of mutant protein sequence, predicted to affect the aa residue which attains coiled coil structure. The coiled coil structure has an important role in key cellular interactions, especially with DNA damage-binding protein 2 (DDB2), which has important role in DDB-mediated nucleotide excision repair (NER) system. CONCLUSIONS: Our findings support the fact of genetic and clinical heterogeneity in XP. The study also predicts the critical role of DDB2 binding region of XPA protein in NER pathway and opens an avenue for further research to study the functional role of the mutated protein domain.


Subject(s)
Amino Acid Sequence , Sequence Deletion , Skin Diseases/genetics , Xeroderma Pigmentosum Group A Protein/genetics , Xeroderma Pigmentosum/genetics , Child, Preschool , Computer Simulation , DNA Mutational Analysis , Humans , Male , Models, Genetic , Molecular Sequence Data , Pakistan , Skin Diseases/etiology , Xeroderma Pigmentosum/etiology , Xeroderma Pigmentosum Group A Protein/metabolism
15.
Ophthalmology ; 120(7): 1324-36, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23601806

ABSTRACT

OBJECTIVE: Xeroderma pigmentosum (XP) is a rare autosomal recessive disease caused by mutations in DNA repair genes. Clinical manifestations of XP include mild to extreme sensitivity to ultraviolet radiation resulting in inflammation and neoplasia in sun-exposed areas of the skin, mucous membranes, and ocular surfaces. This report describes the ocular manifestations of XP in patients systematically evaluated in the Clinical Center at the National Institutes of Health. DESIGN: Retrospective observational case series. PARTICIPANTS: Eighty-seven participants, aged 1.3 to 63.4 years, referred to the National Eye Institute (NEI) for examination from 1964 to 2011. Eighty-three patients had XP, 3 patients had XP/Cockayne syndrome complex, and 1 patient had XP/trichothiodystrophy complex. METHODS: Complete age- and developmental stage-appropriate ophthalmic examination. MAIN OUTCOME MEASURES: Visual acuity; eyelid, ocular surface, and lens pathology; tear film and tear production measures; and cytologic analysis of conjunctival surface swabs. RESULTS: Of the 87 patients, 91% had at least 1 ocular abnormality. The most common abnormalities were conjunctivitis (51%), corneal neovascularization (44%), dry eye (38%), corneal scarring (26%), ectropion (25%), blepharitis (23%), conjunctival melanosis (20%), and cataracts (14%). Thirteen percent of patients had some degree of visual axis impingement, and 5% of patients had no light perception in 1 or both eyes. Ocular surface cancer or a history of ocular surface cancer was present in 10% of patients. Patients with an acute sunburning skin phenotype were less likely to develop conjunctival melanosis and ectropion but more likely to develop neoplastic ocular surface lesions than nonburning patients. Some patients also showed signs of limbal stem cell deficiency. CONCLUSIONS: Our longitudinal study reports the ocular status of the largest group of patients with XP systematically examined at 1 facility over an extended period of time. Structural eyelid abnormalities, neoplasms of the ocular surface and eyelids, tear film and tear production abnormalities, ocular surface disease and inflammation, and corneal abnormalities were present in this population. Burning and nonburning patients with XP exhibit different rates of important ophthalmologic findings, including neoplasia. In addition, ophthalmic characteristics can help refine diagnoses in the case of XP complex phenotypes. DNA repair plays a major role in protection of the eye from sunlight-induced damage.


Subject(s)
DNA Repair/physiology , DNA/radiation effects , Eye Diseases/diagnosis , Radiation Injuries/diagnosis , Sunlight/adverse effects , Xeroderma Pigmentosum/diagnosis , Adolescent , Adult , Child , Child, Preschool , Cockayne Syndrome/diagnosis , Cockayne Syndrome/etiology , Cockayne Syndrome/prevention & control , Eye Diseases/etiology , Eye Diseases/prevention & control , Female , Follow-Up Studies , Humans , Infant , Male , Middle Aged , Radiation Injuries/etiology , Radiation Injuries/prevention & control , Retrospective Studies , Trichothiodystrophy Syndromes/diagnosis , Trichothiodystrophy Syndromes/etiology , Trichothiodystrophy Syndromes/prevention & control , Ultraviolet Rays/adverse effects , Visual Acuity/physiology , Xeroderma Pigmentosum/etiology , Xeroderma Pigmentosum/prevention & control , Young Adult
16.
ScientificWorldJournal ; 2013: 534752, 2013.
Article in English | MEDLINE | ID: mdl-24459435

ABSTRACT

Xeroderma pigmentosum (XP) is a hereditary autosomal recessive disorder characterized by photo hypersensitivity of sun exposed tissues and subsequent several-fold increased risk for malignant changes resulting from impaired ability to repair UV-induced DNA damage. Estimated incidences vary from 1 in 20,000 in Japan to 1 in 250,000 in the USA, and approximately 2.3 per million live births in Western Europe. Diagnosis is made clinically by the presence of unusual sunburns or lentiginosis or onset of cancers at an early age. It is confirmed by cellular tests for defective DNA repair. Although there is no cure for XP as of now, skin problems can be ameliorated with the use of sunscreens, sun avoidance methods, and recurrent tumor excisions. Oral isotretinoin and topical application of 5-fluorouracil to treat actinic keratoses are other therapeutic options. T4N5 and photolyase liposomal lotions are innovations in the therapy of XP. Genetic counselling implicating the effect of consanguineous marriages should be considered in the management of XP patients.


Subject(s)
Xeroderma Pigmentosum , DNA Damage , DNA Repair/genetics , Eye Diseases/etiology , Female , Humans , Male , Mouth Diseases/etiology , Mutation , Nervous System Diseases/etiology , Oral Health , Pregnancy , Skin Neoplasms/etiology , Xeroderma Pigmentosum/diagnosis , Xeroderma Pigmentosum/etiology , Xeroderma Pigmentosum/therapy
17.
J Fr Ophtalmol ; 33(7): 495.e1-4, 2010 Sep.
Article in French | MEDLINE | ID: mdl-20674086

ABSTRACT

Patients with the genetic disease xeroderma pigmentosum (XP) lack the ability to carry out a specific type of DNA repair process called nucleotide excision repair (NER). The NER pathway plays a critical role in the repair of DNA damage resulting from ultraviolet (UV) radiation. We report a case of a patient presenting a cutaneous form of XP. She presented acute paralysis of the third cranial nerve with cutaneous anesthesia. Nuclear magnetic resonance imaging revealed an intracranial tumor. She underwent surgery with incomplete tumor resection; anatomopathological study showed a schwannoma. Complementary radiosurgery was performed. The association between XP and neurological cancer is rare but not impossible. A priori, it would be assumed that the major medical outcome of hereditary deficiencies in DNA repair processes would be an increased risk of cancer. Indeed, there is an increased risk of cancer in several known DNA repair deficiencies including XP.


Subject(s)
Brain Neoplasms/complications , Neurilemmoma/complications , Xeroderma Pigmentosum/etiology , Adult , Female , Humans
19.
J Investig Dermatol Symp Proc ; 14(1): 8-14, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19675546

ABSTRACT

Progeroid syndromes are a group of diseases characterized by signs of premature aging. These syndromes comprise diseases such as Werner syndrome, Bloom syndrome, Rothmund-Thomson syndrome, Hutchinson-Gilford syndrome, Fanconi anemia, and ataxia-telangiectasia, as well as xeroderma pigmentosum, trichothiodystrophy, and Cockayne syndrome. Clinical symptoms of premature aging are skin atrophy with loss of cutaneous elasticity, dysfunction of cutaneous appendices, degeneration of the central nervous system and an increased susceptibility for malignant tumors. Genetic defects in the repair of DNA damage can lead to progeroid syndromes, and it is becoming increasingly evident that direct DNA damage and indirect damage by highly reactive oxygen species play central roles in aging. The clinical signs of progeroid syndromes and the molecular aspects of UV (ultraviolet radiation)-induced oxidative stress in aging are discussed.Journal of Investigative Dermatology Symposium Proceedings (2009) 14, 8-14; doi:10.1038/jidsymp.2009.6.


Subject(s)
Cockayne Syndrome/etiology , DNA Damage , Ultraviolet Rays/adverse effects , Ataxia Telangiectasia/etiology , Bloom Syndrome/etiology , Cockayne Syndrome/genetics , Cockayne Syndrome/metabolism , DNA Repair , Fanconi Anemia/etiology , Female , Humans , Male , Models, Biological , Oxidative Stress/radiation effects , Progeria/etiology , Rothmund-Thomson Syndrome/etiology , Trichothiodystrophy Syndromes/etiology , Werner Syndrome/etiology , Xeroderma Pigmentosum/etiology
20.
Mech Ageing Dev ; 128(2): 229-35, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17361460

ABSTRACT

Researchers and clinicians interested in human diseases of DNA repair deficiency and premature aging gathered at the National Conference Center in Lansdowne, Virginia on 5-8 September 2006 to attend a workshop co-organized by Vilhelm Bohr (National Institute of Aging) and Kenneth Kraemer (National Cancer Institute). An important feature of this workshop was the participation of representatives from xeroderma pigmentosum (XP), Cockayne Syndrome (CS) and trichothiodystrophy (TTD) family support groups. Studies presented at the workshop described important new insights into the phenotypic complexity of XP, CS and TTD, renewed focus on the neurological manifestations of each of these diseases, as well as keen interest in the role of oxidative stress and mitochondrial dysfunction in neurodegenerative processes and normal and/or premature aging. This workshop report summarizes some of the presentations and outcomes of the workshop.


Subject(s)
Aging, Premature/etiology , DNA Repair-Deficiency Disorders/etiology , Aging , Cockayne Syndrome/etiology , Cockayne Syndrome/genetics , DNA Repair-Deficiency Disorders/epidemiology , DNA Repair-Deficiency Disorders/genetics , Hair Diseases/etiology , Hair Diseases/genetics , Humans , Oxidative Stress , Prevalence , Xeroderma Pigmentosum/etiology , Xeroderma Pigmentosum/genetics
SELECTION OF CITATIONS
SEARCH DETAIL