Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 254
Filter
1.
Br J Dermatol ; 185(1): 147-152, 2021 07.
Article in English | MEDLINE | ID: mdl-33393080

ABSTRACT

BACKGROUND: Ceramide kinase-like protein (CERKL) was originally described in retinal tissue. CERKL has been shown to protect cells from oxidative stress, and mutations in CERKL underlie the inherited disease retinitis pigmentosa. CERKL expression maintains cellular sphingolipids via an unknown mechanism. OBJECTIVES: To determine whether CERKL is expressed in epidermis and cutaneous squamous cell carcinoma (cSCC) and whether CERKL expression affects cSCC sphingolipid metabolism and susceptibility to oxidative stress. METHODS: CERKL expression was determined by RNA-Seq, quantitative polymerase chain reaction and immunohistochemistry. CERKL was knocked down in cSCC cells using small interfering RNA. Sphingolipid content was analysed by liquid chromatography-mass spectrometry. Oxidative stress was induced by treatment with H2 O2 , and apoptosis was measured using flow cytometry to determine annexin V binding. RESULTS: CERKL mRNA and protein are highly expressed in actinic keratosis and cSCC in comparison with normal epidermis. CERKL is also expressed in metabolically active epithelial cells in normal hair bulbs and sebaceous glands. CERKL knockdown in cultured cSCC cells reduces cellular sphingolipid content and enhances susceptibility to oxidative stress. CONCLUSIONS: These findings suggest that CERKL may be important in cSCC progression and could lead to novel strategies for prevention and treatment of cSCC.


Subject(s)
Carcinoma, Squamous Cell , Skin Neoplasms , Carcinoma, Squamous Cell/genetics , Humans , Oxidative Stress , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Skin Neoplasms/genetics , Sphingolipids
2.
Br J Dermatol ; 2020 Dec 03.
Article in English | MEDLINE | ID: mdl-33270220

ABSTRACT

BACKGROUND: Ceramide Kinase-Like Protein (CERKL) was originally described in retinal tissue. CERKL has been shown to protect cells from oxidative stress, and mutations in CERKL underlie the inherited disease, retinitis pigmentosa. CERKL expression maintains cellular sphingolipids via an unknown mechanism. OBJECTIVES: To determine whether CERKL is expressed in epidermis and cutaneous squamous cell carcinoma (cSCC) and whether CERKL expression affects cSCC sphingolipid metabolism and susceptibility to oxidative stress. METHODS: CERKL expression was determined by RNA-Seq, qPCR and immunohistochemistry. CERKL was knocked down in cSCC cells using siRNA. Sphingolipid content was analyzed by liquid chromatography-mass spectrometry (LC-MS). Oxidative stress was induced by treatment with H2 O2 , and apoptosis was measured using flow cytometry to determine annexin v binding. RESULTS: CERKL mRNA and protein are highly expressed in actinic keratosis and cSCC in comparison with normal epidermis. CERKL also is expressed in metabolically active epithelial cells in normal hair bulbs and sebaceous glands. CERKL knockdown in cultured cSCC cells reduces cellular sphingolipid content and enhances susceptibility to oxidative stress. CONCLUSIONS: These findings suggest that CERKL may be important in cSCC progression and could lead to novel strategies for prevention and treatment of cSCC.

3.
Mucosal Immunol ; 7(1): 78-88, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23632328

ABSTRACT

Infectious pneumonias are the leading cause of death worldwide, particularly among immunocompromised patients. Therapeutic stimulation of the lungs' intrinsic defenses with a unique combination of inhaled Toll-like receptor (TLR) agonists broadly protects mice against otherwise lethal pneumonias. As the survival benefit persists despite cytotoxic chemotherapy-related neutropenia, the cells required for protection were investigated. The inducibility of resistance was tested in mice with deficiencies of leukocyte lineages due to genetic deletions and in wild-type mice with leukocyte populations significantly reduced by antibodies or toxins. Surprisingly, these serial reductions in leukocyte lineages did not appreciably impair inducible resistance, but targeted disruption of TLR signaling in the lung epithelium resulted in complete abrogation of the protective effect. Isolated lung epithelial cells were also induced to kill pathogens in the absence of leukocytes. Proteomic and gene expression analyses of isolated epithelial cells and whole lungs revealed highly congruent antimicrobial responses. Taken together, these data indicate that lung epithelial cells are necessary and sufficient effectors of inducible resistance. These findings challenge conventional paradigms about the role of epithelia in antimicrobial defense and offer a novel potential intervention to protect patients with impaired leukocyte-mediated immunity from fatal pneumonias.


Subject(s)
Alveolar Epithelial Cells/metabolism , Disease Resistance/immunology , Pneumonia/immunology , Pneumonia/metabolism , Alveolar Epithelial Cells/drug effects , Animals , Disease Models, Animal , Disease Resistance/drug effects , Leukocytes/drug effects , Leukocytes/immunology , Leukocytes/metabolism , Lipopeptides/metabolism , Lipopeptides/pharmacology , Mice , Mice, Knockout , Pneumonia/genetics , Pneumonia/mortality , Signal Transduction , Toll-Like Receptors/metabolism
4.
Mech Ageing Dev ; 129(7-8): 492-7, 2008.
Article in English | MEDLINE | ID: mdl-18336867

ABSTRACT

Cancer, aging, and neurodegeneration are all associated with DNA damage and repair in complex fashions. Aging appears to be a cell and tissue-wide process linked to the insulin-dependent pathway in several DNA repair deficient disorders, especially in mice. Cancer and neurodegeneration appear to have complementary relationships to DNA damage and repair. Cancer arises from surviving cells, or even stem cells, that have down-regulated many pathways, including apoptosis, that regulate genomic stability in a multi-step process. Neurodegeneration however occurs in nondividing neurons in which the persistence of apoptosis in response to reactive oxygen species is, itself, pathological. Questions that remain open concern: sources and chemical nature of naturally occurring DNA damaging agents, especially whether mitochondria are the true source; the target tissues for DNA damage and repair; do the human DNA repair deficient diseases delineate specific pathways of DNA damage relevant to clinical outcomes; if naturally occurring reactive oxygen species are pathological in human repair deficient disease, would anti-oxidants or anti-apoptotic agents be feasible therapeutic agent?


Subject(s)
Aging/genetics , Cockayne Syndrome/genetics , DNA Damage , Neoplasms/genetics , Neurodegenerative Diseases/genetics , Xeroderma Pigmentosum/genetics , Animals , Cockayne Syndrome/diagnosis , Cockayne Syndrome/drug therapy , DNA Repair , Humans , Mice , Xeroderma Pigmentosum/diagnosis , Xeroderma Pigmentosum/drug therapy
5.
Oncogene ; 26(39): 5713-21, 2007 Aug 23.
Article in English | MEDLINE | ID: mdl-17369853

ABSTRACT

Hydroxyurea reduces DNA replication by nucleotide deprivation, whereas UV damage generates DNA photoproducts that directly block replication fork progression. We show that the low fidelity class Y polymerase Pol eta is recruited to proliferating cell nuclear antigen at replication forks both by hydroxyurea and UV light. Under nucleotide deprivation, Pol eta allows cells to accumulate at the G1/S boundary by facilitating slow S-phase progression and promotes apoptosis. Normal cells consequently enter apoptosis at a faster rate than Pol eta-deficient cells. Coincident with hydroxyurea-induced S-phase delay, Pol eta-deficient cells undergo more replication fork breakage and accumulate more foci of the Mre11/Rad50/Nbs1 complex and phosphorylated histone H2AX. We conclude that under conditions of nucleotide deprivation, Pol eta is required for S-phase progression but is proapoptotic. However, as Pol eta is reported to require higher nucleotide concentrations than class B replicative polymerases, its recruitment by hydroxyurea requires it to function under suboptimal conditions. Our results suggest that hydroxyurea-induced apoptosis occurs at the G1/S boundary and that initiation of the S-phase requires greater nucleotide concentrations than does S-phase progression.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/physiology , DNA Replication/drug effects , DNA-Directed DNA Polymerase/physiology , Hydroxyurea/pharmacology , Nucleotides/metabolism , S Phase/physiology , Apoptosis/radiation effects , Blotting, Western , Cell Cycle/drug effects , Cell Cycle/physiology , Cells, Cultured/enzymology , Cells, Cultured/radiation effects , DNA Damage , DNA Repair , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Flow Cytometry , Fluorescent Antibody Technique , Histones , Humans , MRE11 Homologue Protein , Proliferating Cell Nuclear Antigen/metabolism , Recombination, Genetic , S Phase/radiation effects , Ultraviolet Rays , Xeroderma Pigmentosum
6.
Proc Natl Acad Sci U S A ; 104(4): 1389-94, 2007 Jan 23.
Article in English | MEDLINE | ID: mdl-17229834

ABSTRACT

Cockayne syndrome (CS) is a rare recessive childhood-onset neurodegenerative disease, characterized by a deficiency in the DNA repair pathway of transcription-coupled nucleotide excision repair. Mice with a targeted deletion of the CSB gene (Csb-/-) exhibit a much milder ataxic phenotype than human patients. Csb-/- mice that are also deficient in global genomic repair [Csb-/-/xeroderma pigmentosum C (Xpc)-/-] are more profoundly affected, exhibiting whole-body wasting, ataxia, and neural loss by postnatal day 21. Cerebellar granule cells demonstrated high TUNEL staining indicative of apoptosis. Purkinje cells, identified by the marker calbindin, were severely depleted and, although not TUNEL-positive, displayed strong immunoreactivity for p53, indicating cellular stress. A subset of animals heterozygous for Csb and Xpc deficiencies was more mildly affected, demonstrating ataxia and Purkinje cell loss at 3 months of age. Mouse, Csb-/-, and Xpc-/- embryonic fibroblasts each exhibited increased sensitivity to UV light, which generates bulky DNA damage that is a substrate for excision repair. Whereas Csb-/-/Xpc-/- fibroblasts were more UV-sensitive than either single knockout, double-heterozygote fibroblasts had normal UV sensitivity. Csb-/- mice crossed with a strain defective in base excision repair (Ogg1) demonstrated no enhanced neurodegenerative phenotype. Complete deficiency in nucleotide excision repair therefore renders the brain profoundly sensitive to neurodegeneration in specific cell types of the cerebellum, possibly because of unrepaired endogenous DNA damage that is a substrate for nucleotide but not base excision repair.


Subject(s)
Apoptosis/physiology , Cerebellum/pathology , Cockayne Syndrome/physiopathology , DNA Repair , Neurons/pathology , Tumor Suppressor Protein p53/physiology , Up-Regulation , Animals , DNA Repair Enzymes/genetics , DNA Repair Enzymes/physiology , Immunohistochemistry , Mice , Poly-ADP-Ribose Binding Proteins , Ultraviolet Rays
7.
Neuroscience ; 145(4): 1300-8, 2007 Apr 14.
Article in English | MEDLINE | ID: mdl-17055654

ABSTRACT

Cockayne syndrome (CS) is a progressive childhood neurodegenerative disorder associated with a DNA repair defect caused by mutations in either of two genes, CSA and CSB. These genes are involved in nucleotide excision repair (NER) of DNA damage from ultraviolet (UV) light, other bulky chemical adducts and reactive oxygen in transcriptionally active genes (transcription-coupled repair, TCR). For a long period it has been assumed that the symptoms of CS patients are all due to reduced TCR of endogenous DNA damage in the brain, together with unexplained unique sensitivity of specific neural cells in the cerebellum. Not all the symptoms of CS patients are however easily related to repair deficiencies, so we hypothesize that there are additional pathways relevant to the disease, particularly those that are downstream consequences of a common defect in the E3 ubiquitin ligase associated with the CSA and CSB gene products. We have found that the CSB defect results in altered expression of anti-angiogenic and cell cycle genes and proteins at the level of both gene expression and protein lifetime. We find an over-abundance of p21 due to reduced protein turnover, possibly due to the loss of activity of the CSA/CSB E3 ubiquitylation pathway. Increased levels of p21 can result in growth inhibition, reduced repair from the p21-PCNA interaction, and increased generation of reactive oxygen. Consistent with increased reactive oxygen levels we find that CS-A and -B cells grown under ambient oxygen show increased DNA breakage, as compared with xeroderma pigmentosum cells. Thus the complex symptoms of CS may be due to multiple, independent downstream targets of the E3 ubiquitylation system that results in increased DNA damage, reduced transcription coupled repair, and inhibition of cell cycle progression and growth.


Subject(s)
Cockayne Syndrome/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Damage/genetics , DNA Repair/genetics , Gene Expression Regulation/genetics , Transcription, Genetic/genetics , Cell Cycle/genetics , Cell Line , Cockayne Syndrome/metabolism , Cockayne Syndrome/physiopathology , Cyclin-Dependent Kinase Inhibitor p21/genetics , DNA Damage/radiation effects , DNA Helicases/genetics , DNA Repair Enzymes/genetics , Humans , Oxidative Stress/physiology , Poly-ADP-Ribose Binding Proteins , Reactive Oxygen Species/metabolism , Signal Transduction/genetics , Transcription Factors/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ultraviolet Rays
8.
Oncogene ; 24(23): 3708-14, 2005 May 26.
Article in English | MEDLINE | ID: mdl-15750628

ABSTRACT

XP variant (XP-V) cells lack the damage-specific polymerase eta and exhibit prolonged replication arrest after UV irradiation due to impaired bypass of UV photoproducts. To analyse the outcome of the arrested replication forks, homologous recombination (HR, Rad51 events) and fork breakage (Rad50 events) were assayed by immunofluorescent detection of foci-positive cells. Within 1 h of irradiation, XP-V cells showed more Rad51-positive cells than normal cells, while neither cell type showed an increase in Rad50 foci. Beyond 1 h, the frequency of Rad51-positive cells reached similar levels in both cell types, then declined at higher UV doses. At these later times, Rad50-positive cells increased with dose and to a greater extent in XP-V cells. Few cells were simultaneously positive for both sets of foci, suggesting a mutually exclusive recruitment of recombination proteins, or that these pathways operate at different stages during S phase. Analysis of cells containing a vector of tandemly arranged enhanced green fluorescent protein genes also showed that UV-induced HR was higher in XP-V cells. These results suggest that cells make an early commitment to HR, and that at later times a subset of arrested forks degrade into double-strand breaks, two alternative pathways that are greater in XP-V cells.


Subject(s)
Recombination, Genetic , Ultraviolet Rays , Xeroderma Pigmentosum/genetics , Acid Anhydride Hydrolases , Cells, Cultured , DNA Repair Enzymes/analysis , DNA-Binding Proteins/analysis , Green Fluorescent Proteins/genetics , Humans , Rad51 Recombinase
9.
Br J Anaesth ; 94(4): 514-9, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15695544

ABSTRACT

BACKGROUND: Chronic sciatica can be managed by caudal steroid epidural or by targeted steroid placement during spinal endoscopy. Spinal endoscopy is a new unproven procedure. We aimed to compare the two pain management techniques and to investigate whether the site of steroid placement within the epidural space was significant. METHODS: We randomized 60 patients with a 6-18 months history of sciatica to either targeted epidural local anaesthetic and steroid placement with a spinal endoscope or caudal epidural local anaesthetic and steroid treatment. Pre-treatment and 6-week, 3-month, and 6-month SF-MPQ and HAD scores were recorded. RESULTS: No significant differences were found between the groups for any of the measures at any time. However, there were significant differences within both groups compared with pre-treatment values. For the caudal group, significant improvements were found for descriptive pain at 6 months (P=0.031), VAS at 6 weeks (P=0.036), 3 months (P=0.026), and 6 months (P=0.003), present pain intensity (PPI) at 3 months (P=0.013) and 6 months (P=0.01); anxiety at 6 weeks (P=0.008), 3 months (P=0.004), and 6 months (P=0.001) and depression at 6 months only (P=0.037). For the epiduroscopy group there were fewer significant changes. PPI was significantly reduced at 6 weeks (P=0.004) and at 6 months (P=0.02). Anxiety was reduced at 6 months only (P=0.03). CONCLUSION: The targeted placement of epidural steroid onto the affected nerve root causing sciatica does not significantly reduce pain intensity and anxiety and depression compared with untargeted caudal epidural steroid injection. When analysed individually, both techniques benefited patients.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Glucocorticoids/administration & dosage , Sciatica/drug therapy , Adult , Aged , Aged, 80 and over , Anesthetics, Local/administration & dosage , Chronic Disease , Double-Blind Method , Drug Therapy, Combination , Endoscopy , Female , Humans , Injections, Epidural , Injections, Intralesional , Lidocaine/administration & dosage , Male , Middle Aged , Pain Measurement , Prospective Studies , Treatment Outcome
10.
DNA Repair (Amst) ; 3(2): 183-87, 2004 Feb 03.
Article in English | MEDLINE | ID: mdl-15344228

ABSTRACT

Most forms of the human hereditary disease xeroderma pigmentation (XP) are due to a defect in nucleotide excision repair of DNA damage in skin cells associated with exposure to sunlight. This discovery by James Cleaver had an important impact on our understanding of nucleotide excision repair in mammals.


Subject(s)
DNA Repair , DNA Replication , Skin Neoplasms/history , Xeroderma Pigmentosum/history , DNA, Neoplasm/radiation effects , Genetics/history , History, 20th Century , Humans , Skin Neoplasms/genetics , Sunlight/adverse effects , Xeroderma Pigmentosum/genetics
11.
Genomics ; 82(5): 561-70, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14559213

ABSTRACT

POLH and POLI are paralogs encoding low-fidelity, class Y, DNA polymerases involved in replication of damaged DNA in the human disease xeroderma pigmentosum variant. Analysis of genomic regions for human and mouse homologs, employing the analytic tool Genome Cryptographer, detected low-repetitive or unique regions at exons and other potential control regions, especially within intron I of human POLH. The human and mouse homologs are structurally similar, but the paralogs have undergone evolutionary divergence. The information content of splice sites for human POLH, the probability that a base would contribute to splicing, was low only for the acceptor site of exon II, which is preceded by a region of high information content that could contain sequences controlling splicing. This analysis explains previous observations of tissue-specific skipping during mRNA processing, resulting in the loss of the transcription start site in exon II, in human tissues.


Subject(s)
DNA Damage/genetics , DNA Polymerase I/genetics , DNA Repair , DNA-Directed DNA Polymerase/genetics , RNA Splice Sites/genetics , Alternative Splicing , Animals , Base Sequence , DNA Replication , Exons , Humans , Introns , Mice , Molecular Sequence Data
12.
Proc Natl Acad Sci U S A ; 100(20): 11412-7, 2003 Sep 30.
Article in English | MEDLINE | ID: mdl-13679571

ABSTRACT

Epidermal stem cells play a central role in tissue homeostasis, wound repair, tumor initiation, and gene therapy. A major impediment to the purification and molecular characterization of epidermal stem cells is the lack of a quantitative assay for cells capable of long-term repopulation in vivo, such as exists for hematopoietic cells. The tremendous strides made in the characterization and purification of hematopoietic stem cells have been critically dependent on the availability of competitive transplantation assays, because these assays permit the accurate quantitation of long-term repopulating cells in vivo. We have developed an analogous functional assay for epidermal stem cells, and have measured the frequency of functional epidermal stem cells in interfollicular epidermis. These studies indicate that cells capable of long-term reconstitution of a squamous epithelium reside in the interfollicular epidermis. We find that the frequency of these long-term repopulating cells is 1 in 35,000 total epidermal cells, or in the order of 1 in 104 basal epidermal cells, similar to that of hematopoietic stem cells in the bone marrow, and much lower than previously estimated in epidermis. Furthermore, these studies establish a novel functional assay that can be used to validate immunophenotypic markers and enrichment strategies for epidermal stem cells, and to quantify epidermal stem cells in various keratinocyte populations. Thus further studies using this type of assay for epidermis should aid in the progress of cutaneous stem cell-targeted gene therapy, and in more basic studies of epidermal stem cell regulation and differentiation.


Subject(s)
Cell Lineage , Stem Cells/cytology , Animals , Animals, Newborn , Mice , Mice, SCID
13.
Cell Cycle ; 2(4): 310-5, 2003.
Article in English | MEDLINE | ID: mdl-12851481

ABSTRACT

We describe here a model for sequential recruitment of various enzymatic systems that maintain DNA replication fidelity in cells with damaged bases, especially those formed by ultraviolet (UV) irradiation. Systems of increasing complexity but decreasing fidelity are recruited to restore replication of damaged DNA. The first and most accurate response is nucleotide excision repair (NER) that is cell cycle-independent; next come various delaying cell cycle checkpoints that provide an extended time window for NER. These delay the onset of the S phase at the G1/S boundary, and inhibit the initiation of individual replicating units (replicons and clusters of replicons) within the S phase. When checkpoints fail to operate completely, DNA replication forks must negotiate damage and the loss of coding information on the parental DNA strands. Replication can be resumed using bypass polymerases, or alternative bypass mechanisms. Finally, if all else fails, replication forks may degrade to double strand breaks and recombinational processes then allow their reconstruction. A network of signaling kinases modulates the efficiency of many damage responsive proteins to tailor their activities and subcellular localizations by phosphorylation and dephosphorylation.


Subject(s)
DNA Damage/physiology , DNA Repair/physiology , DNA Replication/physiology , Recombination, Genetic/physiology , Animals , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , DNA Damage/genetics , DNA Repair/genetics , DNA Replication/genetics , DNA-Directed DNA Polymerase/genetics , DNA-Directed DNA Polymerase/metabolism , Humans , Mutation , Phosphorylation , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/metabolism , Recombination, Genetic/genetics , Replication Origin/genetics , Replication Origin/physiology , Replicon/genetics , Replicon/physiology , S Phase/physiology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Ultraviolet Rays
14.
Mutat Res ; 510(1-2): 121-9, 2002 Dec 29.
Article in English | MEDLINE | ID: mdl-12459448

ABSTRACT

Ultraviolet (UV) irradiation produces DNA photoproducts that are blocks to DNA replication by normal replicative polymerases. A specialized, damage-specific, distributive polymerase, Pol H or Pol h, that is the product of the hRad30A gene, is required for replication past these photoproducts. This polymerase is absent from XP variant (XP-V) cells that must employ other mechanisms to negotiate blocks to DNA replication. These mechanisms include the use of alternative polymerases or recombination between sister chromatids. Replication forks arrested by UV damage in virus transformed XP-V cells degrade into DNA double strand breaks that are sites for recombination, but in normal cells arrested forks may be protected from degradation by p53 protein. These breaks are sites for binding a protein complex, hMre11/hRad50/Nbs1, that colocalizes with H2AX and PCNA, and can be visualized as immunofluorescent foci. The protein complexes need phosphorylation to activate their DNA binding capacity. Incubation of UV irradiated XP-V cells with the irreversible kinase inhibitor wortmannin, however, increased the yield of Mre11 focus-positive cells. One interpretation of this observation is that two classes of kinases are involved after UV irradiation. One would be a wortmannin-resistant kinase that phosphorylates the Mre11 complex. The other would be a wortmannin-sensitive kinase that phosphorylates and activates the p53/large T in SV40 transformed XP-V cells. The sensitive class corresponds to the PI3-kinases of ATM, ATR, and DNA-PK, but the resistant class remains to be identified. Alternatively, the elevated yield of Mre11 foci positive cells following wortmannin treatment may reflect an overall perturbation to the signaling cascades regulated by wortmannin-sensitive PI3 related kinases. In this scenario, wortmannin could compromise damage inducible-signaling pathways that maintain the stability of stalled forks, resulting in a further destabilization of stalled forks that then degrade, with the formation of DNA double strand breaks.


Subject(s)
DNA Replication , Androstadienes/pharmacology , Cell Line , DNA Damage , DNA Replication/radiation effects , DNA-Binding Proteins/metabolism , Humans , MRE11 Homologue Protein , Recombination, Genetic , Ultraviolet Rays/adverse effects , Wortmannin , Xeroderma Pigmentosum/genetics , Xeroderma Pigmentosum/metabolism
15.
DNA Repair (Amst) ; 1(1): 41-57, 2002 Jan 22.
Article in English | MEDLINE | ID: mdl-12509296

ABSTRACT

Xeroderma pigmentosum variant (XPV) cells lack the damage-specific polymerase eta and undergo a protracted arrest at the S phase checkpoint(s) following UV damage. The S phase checkpoints encompass several qualitatively different processes, and stimulate downstream events that are dependent on the functional state of p53. Primary fibroblasts with wild-type p53 arrest in S, and require a functional polymerase eta (pol eta) to carry out bypass replication, but do not recruit recombination factors for recovery. XPV cells with non-functional p53, as a result of transformation by SV40 or HPV16 (E6/E7), recruit the hMre11/hRad50/Nbs1 complex to arrested replication forks, coincident with PCNA, whereas normal transformed cells preferentially use the pol eta bypass replication pathway. The formation of hMre11 foci implies that arrested replication forks rapidly undergo a collapse involving double strand breakage and rejoining. Apoptosis occurs after UV only in cells transformed by SV40, and not in normal or XPV fibroblasts or HPV16 (E6/E7) transformed cells. Conversely, ultimate cell survival in XPV cells was much less in HPV16 (E6/E7) transformed cells than in SV40 transformed cells, indicating that apoptosis was not a reliable predictor of cell survival. Inhibition of p53 transactivation by pifithrin-alpha or inhibition of protein synthesis by cycloheximide did not induce hMre11 foci or apoptosis in UV damaged fibroblasts. Inhibition of kinase activity with wortmannin did not increase killing by UV, unlike the large increase seen with caffeine. Since HPV16 (E6/E7) transformed XPV cells were highly UV sensitive and not further sensitized by caffeine, it appears likely that caffeine sensitization proceeds through a p53 pathway. The S phase checkpoints are therefore, a complex set of different checkpoints that are coordinated by p53 with the capacity to differentially modulate cell survival, apoptosis, bypass replication and hMre11 recombination.


Subject(s)
Apoptosis/physiology , Cell Survival/physiology , DNA Repair Enzymes , DNA-Binding Proteins/genetics , DNA-Directed DNA Polymerase/physiology , Fibroblasts/cytology , Recombination, Genetic/genetics , S Phase/physiology , Toluene/analogs & derivatives , Tumor Suppressor Protein p53/physiology , Acid Anhydride Hydrolases , Apoptosis/radiation effects , Benzothiazoles , Blotting, Western , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Transformed/radiation effects , Cell Survival/radiation effects , DNA Replication/radiation effects , DNA-Binding Proteins/metabolism , Fibroblasts/physiology , Fibroblasts/radiation effects , Humans , MRE11 Homologue Protein , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oncogene Proteins, Viral/genetics , Simian virus 40/genetics , Thiazoles/pharmacology , Toluene/pharmacology , Ultraviolet Rays , beta-Galactosidase/metabolism
16.
Environ Mol Mutagen ; 38(2-3): 122-31, 2001.
Article in English | MEDLINE | ID: mdl-11746745

ABSTRACT

Richard B. Setlow inspired the field of DNA repair. His demonstration that photoproducts could be quantified within cells and their excision examined experimentally pioneered the identification of nucleotide excision repair. His early work was associated with the discovery of many founding phenomena of photobiology and DNA repair: the concept of excision repair itself, correlations between DNA repair, life span and aging, variations in repair among mammalian species, caffeine sensitization to UV damage, and the xeroderma pigmentosum (XP) repair deficiencies. We may now have mapped thoroughly the landscape of DNA repair that Dick helped open to exploration, but questions persist of how comprehensively we have explored all its canyons and mesas. Research into nontraditional species and kingdoms may yet provide unexpected surprises. The signal transduction pathways and mechanisms of DNA replication arrest in damaged mammalian cells remain a challenge. The importance of repair in vivo also provides many difficult research questions. One problem of current interest is the role of endogenous DNA damage and repair in human pathology, especially neurodegeneration exemplified by many XP patients. Cancer and neurodegeneration may represent converse responses of dividing and nondividing cells to mutagenic and lethal effects of DNA damaging agents. Cell death from endogenous oxidative DNA damage (apoptosis) may be antagonistic to malignant transformation in dividing cells but may cause neurodegeneration in nondividing neural tissue. Small reductions in the efficiency of repair, especially transcription-coupled repair, may overemphasize carcinogenesis in mice, while minimizing neurodegeneration, as compared to human patients.


Subject(s)
DNA Repair , Radiobiology/history , Animals , History, 20th Century , Humans , Mice , United States
18.
Genes Chromosomes Cancer ; 32(3): 222-35, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11579462

ABSTRACT

Polymerase eta (pol eta) is a low-fidelity DNA polymerase that is the product of the gene, POLH, associated with the human XP variant disorder in which there is an extremely high level of solar-induced skin carcinogenesis. The complete human genomic sequence spans about 40 kb containing 10 coding exons and a cDNA of 2.14 kb; exon I is untranslated and is 6 kb upstream from the first coding exon. Using bacterial artificial chromosomes (BACs), the gene was mapped to human chromosome band 6p21 and mouse band 17D. The gene is expressed in most tissues, except for very low or undetectable levels in peripheral lymphocytes, fetal spleen, and adult muscle; exon II, however, is frequently spliced out in normal cells and in almost half the transcripts in the testis and fetal liver. Expression of POLH in a multicopy episomal vector proved nonviable, suggesting that overexpression is toxic. Expression from chromosomally integrated linear copies using either an EF1-alpha or CMV promoter was functional, resulting in cell lines with low or high levels of pol eta protein, respectively. Point mutations in the center of the gene and in a C-terminal cysteine and deletion of exon II resulted in inactivation, but addition of a terminal 3 amino acid C-terminal tag, or an N- or C-terminal green fluorescent protein, had no effect on function. A low level of expression of pol eta eliminated hMre11 recombination and partially restored UV survival, but did not prevent UV-induced apoptosis, which required higher levels of expression. Polymerase eta is therefore involved in S-phase checkpoint and signal transduction pathways that lead to arrest in S, apoptosis, and recombination. In normal cells, the predominant mechanism of replication of UV damage involves pol eta-dependent bypass, and Mre11-dependent recombination that acts is a secondary, backup mechanism when cells are severely depleted of pol eta.


Subject(s)
Alternative Splicing/genetics , Apoptosis/radiation effects , DNA-Binding Proteins/genetics , DNA-Directed DNA Polymerase/genetics , Radiation Tolerance/genetics , Recombination, Genetic/genetics , Ultraviolet Rays , Alternative Splicing/radiation effects , Animals , Artificial Gene Fusion , Base Composition/genetics , Cell Line , Chromosome Mapping , DNA Repair Enzymes , DNA-Binding Proteins/radiation effects , DNA-Directed DNA Polymerase/radiation effects , Gene Expression Regulation , Genetic Complementation Test , Green Fluorescent Proteins , Humans , Luminescent Proteins/analysis , MRE11 Homologue Protein , Mice , Organ Specificity/genetics , Radiation Tolerance/radiation effects , Recombinant Fusion Proteins/analysis , Recombination, Genetic/radiation effects
20.
Mutat Res ; 485(1): 23-36, 2001 Feb 25.
Article in English | MEDLINE | ID: mdl-11341991

ABSTRACT

The first half of the 20th century has seen an enormous growth in our knowledge of DNA repair, in no small part due to the work of Dirk Bootsma, Philip Hanawalt and Bryn Bridges; those honored by this issue. For the new millennium, we have asked three general questions: (A) Do we know all possible strategies of nucleotide excision repair (NER) in all organisms? (B) How is NER integrated and regulated in cells and tissues? (C) Does DNA replication represent a new frontier in the roles of DNA repair? We make some suggestions for the kinds of answers the next generation may provide. The kingdom of archea represents an untapped field for investigation of DNA repair in organisms with extreme lifestyles. NER appears to involve a similar strategy to the other kingdoms of prokaryotes and eukaryotes, but subtle differences suggest that individual components of the system may differ. NER appears to be regulated by several major factors, especially p53 and Rb which interact with transcription coupled repair and global genomic repair, respectively. Examples can be found of major regulatory changes in repair in testicular tissue and melanoma cells. Our understanding of replication of damaged DNA has undergone a revolution in recent years, with the discovery of multiple low-fidelity DNA polymerases that facilitate replicative bypass. A secondary mechanism of replication in the absence of NER or of one or more of these polymerases involves sister chromatid exchange and recombination (hMre11/hRad50/Nbs1). The relative importance of bypass and recombination is determined by the action of p53. We hypothesise that these polymerases may be involved in resolution of complex DNA structures during completion of replication and sister chromatid resolution. With these fascinating problems to investigate, the field of DNA repair will surely not disappoint the next generation.


Subject(s)
DNA Repair , Animals , DNA/genetics , DNA/metabolism , DNA/radiation effects , DNA Damage , DNA Repair/genetics , DNA Replication , DNA-Directed DNA Polymerase/metabolism , Humans , Recombination, Genetic , Ultraviolet Rays/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL
...