Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Nat Rev Dis Primers ; 5(1): 64, 2019 09 19.
Article in English | MEDLINE | ID: mdl-31537806

ABSTRACT

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


Subject(s)
DNA Repair-Deficiency Disorders/diagnosis , DNA Repair-Deficiency Disorders/genetics , Ataxia Telangiectasia/diagnosis , Ataxia Telangiectasia/genetics , Ataxia Telangiectasia/physiopathology , Bloom Syndrome/diagnosis , Bloom Syndrome/genetics , Bloom Syndrome/physiopathology , DNA Damage/genetics , DNA Repair-Deficiency Disorders/physiopathology , Fanconi Anemia/diagnosis , Fanconi Anemia/genetics , Fanconi Anemia/physiopathology , Humans , Nijmegen Breakage Syndrome/diagnosis , Nijmegen Breakage Syndrome/genetics , Nijmegen Breakage Syndrome/physiopathology
3.
J Clin Invest ; 124(7): 3137-46, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24911150

ABSTRACT

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


Subject(s)
DNA Repair-Deficiency Disorders/genetics , Mutant Proteins/genetics , Mutation, Missense , Proliferating Cell Nuclear Antigen/genetics , Adolescent , Adult , Aging, Premature/genetics , Amino Acid Substitution , Child , Chromosomes, Human, Pair 20/genetics , DNA Mutational Analysis , DNA Repair-Deficiency Disorders/pathology , DNA Repair-Deficiency Disorders/physiopathology , Dwarfism/genetics , Female , Hearing Loss/genetics , Homozygote , Humans , Male , Models, Molecular , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Nerve Degeneration/genetics , Pedigree , Phenotype , Photosensitivity Disorders/genetics , Proliferating Cell Nuclear Antigen/chemistry , Proliferating Cell Nuclear Antigen/metabolism , Protein Structure, Quaternary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Syndrome , Telangiectasis/genetics
4.
DNA Repair (Amst) ; 12(8): 558-67, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23712058

ABSTRACT

A number of DNA repair disorders are known to cause neurological problems. These disorders can be broadly characterised into early developmental, mid-to-late developmental or progressive. The exact developmental processes that are affected can influence disease pathology, with symptoms ranging from early embryonic lethality to late-onset ataxia. The category these diseases belong to depends on the frequency of lesions arising in the brain, the role of the defective repair pathway, and the nature of the mutation within the patient. Using observations from patients and transgenic mice, we discuss the importance of double strand break repair during neuroprogenitor proliferation and brain development and the repair of single stranded lesions in neuronal function and maintenance.


Subject(s)
DNA Breaks, Single-Stranded , DNA Damage/genetics , Animals , Brain/growth & development , Brain/pathology , Cerebellar Ataxia/genetics , Cerebellar Ataxia/metabolism , DNA Repair-Deficiency Disorders/genetics , DNA Repair-Deficiency Disorders/metabolism , DNA Repair-Deficiency Disorders/physiopathology , Humans , Mutation , Neurogenesis/genetics , Neurons/metabolism , Neurons/pathology
5.
Ann Dermatol Venereol ; 139 Suppl 3: S78-82, 2012 Nov.
Article in French | MEDLINE | ID: mdl-23260522

ABSTRACT

The occurrence of abnormally pigmented skin lesions is a common phenomenon and often associated with the influence of ultraviolet radiation (UV) and other sources of DNA damage. Pigmentary lesions induced by UV radiation and other sources of DNA damage occur in healthy individuals, but human diseases with defective DNA repair represent important models which allow the investigation of possible underlying molecular mechanisms leading to hypo- and hyperpigmentations. There are several hereditary diseases which are known to go along with genetic defects of DNA repair mechanisms comprising Xeroderma pigmentosum (XP), Cockayne syndrome (CS), Trichothiodystrophy (TTD), Werner syndrome (WS), Bloom syndrome (BS), Fanconi anemia (FA) and Ataxia telangiectasia (AT). These diseases share clinical characteristics including poikilodermatic skin changes such as hypo-and hyperpigmentation. Since UV radiation is the most common source of DNA damage which can cause pigmentary lesions both in healthy individuals and in patients with genetic deficiency in DNA repair, in the present article, we focus on pigmentary lesions in patients with XP as an example of a disease associated with genetic defects in DNA repair.


Subject(s)
DNA Damage/physiology , DNA Repair-Deficiency Disorders/diagnosis , DNA Repair-Deficiency Disorders/genetics , Photosensitivity Disorders/diagnosis , Photosensitivity Disorders/genetics , Pigmentation Disorders/diagnosis , Pigmentation Disorders/genetics , Ultraviolet Rays/adverse effects , Xeroderma Pigmentosum/diagnosis , Xeroderma Pigmentosum/genetics , Adult , Carcinoma, Basal Cell/diagnosis , Carcinoma, Basal Cell/genetics , Carcinoma, Basal Cell/physiopathology , Carcinoma, Basal Cell/prevention & control , Carcinoma, Squamous Cell/diagnosis , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/physiopathology , Carcinoma, Squamous Cell/prevention & control , Child , DNA Damage/genetics , DNA Repair-Deficiency Disorders/physiopathology , DNA Repair-Deficiency Disorders/prevention & control , Facial Neoplasms/diagnosis , Facial Neoplasms/genetics , Facial Neoplasms/physiopathology , Facial Neoplasms/prevention & control , Female , Genetic Predisposition to Disease/genetics , Humans , Male , Neoplasms, Multiple Primary/diagnosis , Neoplasms, Multiple Primary/genetics , Neoplasms, Multiple Primary/physiopathology , Neoplasms, Multiple Primary/prevention & control , Neoplasms, Radiation-Induced/diagnosis , Neoplasms, Radiation-Induced/genetics , Neoplasms, Radiation-Induced/physiopathology , Neoplasms, Radiation-Induced/prevention & control , Photosensitivity Disorders/physiopathology , Photosensitivity Disorders/prevention & control , Pigmentation Disorders/physiopathology , Pigmentation Disorders/prevention & control , Skin/physiopathology , Skin/radiation effects , Sunscreening Agents/administration & dosage , Syndrome , Xeroderma Pigmentosum/physiopathology
6.
Article in English | MEDLINE | ID: mdl-23209155

ABSTRACT

Within the last decade, multiple novel congenital human disorders have been described with genetic defects in known and/or novel components of several well-known DNA repair and damage response pathways. Examples include disorders of impaired nucleotide excision repair, DNA double-strand and single-strand break repair, as well as compromised DNA damage-induced signal transduction including phosphorylation and ubiquitination. These conditions further reinforce the importance of multiple genome stability pathways for health and development in humans. Furthermore, these conditions inform our knowledge of the biology of the mechanics of genome stability and in some cases provide potential routes to help exploit these pathways therapeutically. Here, I will review a selection of these exciting findings from the perspective of the disorders themselves, describing how they were identified, how genotype informs phenotype, and how these defects contribute to our growing understanding of genome stability pathways.


Subject(s)
Breast Neoplasms/physiopathology , DNA End-Joining Repair/physiology , DNA Repair-Deficiency Disorders/physiopathology , Fanconi Anemia/physiopathology , Nephritis, Interstitial/physiopathology , Ovarian Neoplasms/physiopathology , Severe Combined Immunodeficiency/physiopathology , DNA Repair/physiology , DNA-Activated Protein Kinase , Female , Genomic Instability/genetics , Genomic Instability/physiology , Humans
7.
DNA Repair (Amst) ; 10(7): 781-91, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21612988

ABSTRACT

ERCC1-XPF is a structure-specific endonuclease required for nucleotide excision repair, interstrand crosslink repair, and the repair of some double-strand breaks. Mutations in ERCC1 or XPF cause xeroderma pigmentosum, XFE progeroid syndrome or cerebro-oculo-facio-skeletal syndrome, characterized by increased risk of cancer, accelerated aging and severe developmental abnormalities, respectively. This review provides a comprehensive overview of the health impact of ERCC1-XPF deficiency, based on these rare diseases and mouse models of them. This offers an understanding of the tremendous health impact of DNA damage derived from environmental and endogenous sources.


Subject(s)
DNA Repair-Deficiency Disorders/genetics , DNA Repair , DNA-Binding Proteins/metabolism , Endonucleases/metabolism , Animals , DNA/genetics , DNA/metabolism , DNA/radiation effects , DNA Damage , DNA Repair-Deficiency Disorders/metabolism , DNA Repair-Deficiency Disorders/physiopathology , DNA-Binding Proteins/genetics , Endonucleases/genetics , Gene Expression Regulation , Genotype , Humans , Mice , Mice, Knockout , Mutation , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/physiopathology , Rare Diseases/genetics , Rare Diseases/metabolism , Rare Diseases/physiopathology , Telomere/genetics , Telomere/metabolism , Ultraviolet Rays
8.
Prog Neurobiol ; 94(2): 166-200, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21550379

ABSTRACT

Deficiency in repair of nuclear and mitochondrial DNA damage has been linked to several neurodegenerative disorders. Many recent experimental results indicate that the post-mitotic neurons are particularly prone to accumulation of unrepaired DNA lesions potentially leading to progressive neurodegeneration. Nucleotide excision repair is the cellular pathway responsible for removing helix-distorting DNA damage and deficiency in such repair is found in a number of diseases with neurodegenerative phenotypes, including Xeroderma Pigmentosum and Cockayne syndrome. The main pathway for repairing oxidative base lesions is base excision repair, and such repair is crucial for neurons given their high rates of oxygen metabolism. Mismatch repair corrects base mispairs generated during replication and evidence indicates that oxidative DNA damage can cause this pathway to expand trinucleotide repeats, thereby causing Huntington's disease. Single-strand breaks are common DNA lesions and are associated with the neurodegenerative diseases, ataxia-oculomotor apraxia-1 and spinocerebellar ataxia with axonal neuropathy-1. DNA double-strand breaks are toxic lesions and two main pathways exist for their repair: homologous recombination and non-homologous end-joining. Ataxia telangiectasia and related disorders with defects in these pathways illustrate that such defects can lead to early childhood neurodegeneration. Aging is a risk factor for neurodegeneration and accumulation of oxidative mitochondrial DNA damage may be linked with the age-associated neurodegenerative disorders Alzheimer's disease, Parkinson's disease and amyotrophic lateral sclerosis. Mutation in the WRN protein leads to the premature aging disease Werner syndrome, a disorder that features neurodegeneration. In this article we review the evidence linking deficiencies in the DNA repair pathways with neurodegeneration.


Subject(s)
DNA Repair-Deficiency Disorders/physiopathology , Neurodegenerative Diseases/genetics , Aging/metabolism , Animals , Brain/metabolism , DNA Repair , DNA Repair-Deficiency Disorders/genetics , DNA Repair-Deficiency Disorders/metabolism , Genomic Instability , Humans , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/metabolism , Neurons/metabolism , Reactive Oxygen Species/metabolism
9.
Expert Rev Clin Immunol ; 7(2): 169-85, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21426255

ABSTRACT

DNA double-strand break (DSB) repair is an essential cellular process required to maintain genomic integrity in the face of potentially lethal genetic damage. Failure to repair a DSB can trigger cell death, whereas misrepair of the break can lead to the generation of chromosomal translocations, which is a known causative event in the development or progression of cancer. DSBs can be induced following exposure to certain exogenous agents, such as ionising radiation or radiomimetic chemicals, as well as occurring naturally as intermediates of normal physiological processes, in particular during B and T cell antigen receptor assembly. Human syndromes with deficiencies in DSB repair commonly exhibit immunodeficiency, highlighting the critical nature of this pathway for development and maturation of the immune system. In this article we review the different pathways utilized by the cell to repair DSBs and how an inherited defect in some of the genes that are critical regulators of this process can be the underlying cause of human disorders associated with genome instability and immune system dysfunction. We focus on a newly described human immunodeficiency disorder called radiosensitivity, immunodeficiency dysmorphic features and learning difficulties (RIDDLE) syndrome, with particular reference to the function of the defective gene, RNF168. We also consider the implications of this finding on the mechanisms controlling development of the immune system.


Subject(s)
DNA Repair-Deficiency Disorders/immunology , DNA Repair , Immune System/metabolism , Immunologic Deficiency Syndromes/immunology , Ubiquitin-Protein Ligases/metabolism , Body Dysmorphic Disorders , DNA Breaks, Double-Stranded , DNA Repair/genetics , DNA Repair/immunology , DNA Repair-Deficiency Disorders/complications , DNA Repair-Deficiency Disorders/genetics , DNA Repair-Deficiency Disorders/physiopathology , Genetic Predisposition to Disease , Humans , Immune System/embryology , Immune System/growth & development , Immunologic Deficiency Syndromes/complications , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/physiopathology , Learning Disabilities , Mutation/genetics , Radiation Tolerance , Ubiquitin-Protein Ligases/genetics
10.
Bone Marrow Transplant ; 41(11): 983-9, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18278071

ABSTRACT

Several human disorders mutated in core components of the major DNA double-strand break (DSB) repair pathway, non-homologous end joining (NHEJ), have been described. Cell lines from these patients are characterized by sensitivity to DSB-inducing agents. DNA ligase IV syndrome (LIG4) patients specifically, for unknown reasons, respond particularly badly following treatment for malignancy or BMT. We report the first systematic evaluation of the response of LIG4 syndrome to compounds routinely employed for BMT conditioning. We found human pre-B lymphocytes, a key target population for BMT conditioning, when deficient for DNA ligase IV, unexpectedly exhibit significant sensitivity to CsA the principal prophylaxis for GVHD. Furthermore, we found that CsA treatment alone or in combination with BU and fludarabine resulted in increased levels of DSBs specifically in LIG4 syndrome cells compared to wild-type or Artemis-deficient cells. Our study shows that CsA can induce DSBs and that LIG4 syndrome patient's fail to adequately repair this damage. These DSBs likely arise as a consequence of DNA replication in the presence of CsA. This work has implications for BMT and GVHD management in general and specifically for LIG4 syndrome.


Subject(s)
Bone Marrow Transplantation , Cyclosporine/adverse effects , DNA Breaks, Double-Stranded/drug effects , DNA Ligases/drug effects , DNA Repair-Deficiency Disorders/chemically induced , Immunosuppressive Agents/adverse effects , Precursor Cells, B-Lymphoid/drug effects , Transplantation Conditioning/adverse effects , Cell Line , DNA Ligase ATP , DNA Ligases/deficiency , DNA Repair-Deficiency Disorders/physiopathology , Humans , Syndrome
11.
Expert Rev Mol Med ; 9(31): 1-20, 2007 Nov 19.
Article in English | MEDLINE | ID: mdl-18021462

ABSTRACT

Adult stem cells have become the focus of intense research in recent years as a result of their role in the maintenance and repair of tissues. They exert this function through their extensive expansion (self-renewal) and multipotent differentiation capacity. Understanding whether adult stem cells retain this capacity throughout the lifespan of the individual, or undergo a process of ageing resulting in a decreased stem cell pool, is an important area of investigation. Progress in this area has been hampered by lack of suitable models and of appropriate markers and assays to identify stem cells. However, recent data suggest that an understanding of the mechanisms governing stem cell ageing can give insight into the mechanism of tissue ageing and, most importantly, advance our ability to use stem cells in cell and gene therapy strategies.


Subject(s)
Adult Stem Cells/physiology , Cellular Senescence , Stem Cells/physiology , Animals , Cell Count , Cell Differentiation , Cell Division , DNA Repair-Deficiency Disorders/pathology , DNA Repair-Deficiency Disorders/physiopathology , Humans , Models, Animal , Syndrome
12.
Exp Dermatol ; 16(6): 532-44, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17518994

ABSTRACT

Genomic instability is the driving force behind cancer development. Human syndromes with DNA repair deficiencies comprise unique opportunities to study the clinical consequences of faulty genome maintenance leading to premature aging and premature cancer development. These syndromes include chromosomal breakage syndromes with defects in DNA damage signal transduction and double-strand break repair, mismatch repair defective syndromes as well as nucleotide excision repair defective syndromes. The same genes that are severely affected in these model diseases may harbour more subtle variations in the 'healthy' normal population leading to genomic instability, cancer development, and accelerated aging at later stages of life. Thus, studying those syndromes and the molecular mechanisms behind can significantly contribute to our understanding of (skin) cancerogenesis as well as to the development of novel individualized preventive and therapeutic anticancer strategies. The establishment of centers of excellence for studying rare genetic model diseases may be helpful in this direction.


Subject(s)
DNA Repair-Deficiency Disorders/genetics , DNA Repair-Deficiency Disorders/physiopathology , DNA Repair/physiology , Skin Neoplasms/genetics , Skin Neoplasms/physiopathology , Humans , Skin Aging/physiology
13.
Nat Clin Pract Neurol ; 3(3): 162-72, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17342192

ABSTRACT

In this Review, familial and sporadic neurological disorders reported to have an etiological link with DNA repair defects are discussed, with special emphasis placed on the molecular link between the disease phenotype and the precise DNA repair defect. Of the 15 neurological disorders listed, some of which have symptoms of progeria, six--spinocerebellar ataxia with axonal neuropathy-1, Huntington's disease, Alzheimer's disease, Parkinson's disease, Down syndrome and amyotrophic lateral sclerosis--seem to result from increased oxidative stress, and the inability of the base excision repair pathway to handle the damage to DNA that this induces. Five of the conditions (xeroderma pigmentosum, Cockayne's syndrome, trichothiodystrophy, Down syndrome, and triple-A syndrome) display a defect in the nucleotide excision repair pathway, four (Huntington's disease, various spinocerebellar ataxias, Friedreich's ataxia and myotonic dystrophy types 1 and 2) exhibit an unusual expansion of repeat sequences in DNA, and four (ataxia-telangiectasia, ataxia-telangiectasia-like disorder, Nijmegen breakage syndrome and Alzheimer's disease) exhibit defects in genes involved in repairing double-strand breaks. The current overall picture indicates that oxidative stress is a major causative factor in genomic instability in the brain, and that the nature of the resulting neurological phenotype depends on the pathway through which the instability is normally repaired.


Subject(s)
DNA Repair-Deficiency Disorders/genetics , DNA Repair-Deficiency Disorders/physiopathology , Nervous System Diseases/genetics , Nervous System Diseases/physiopathology , Animals , DNA Damage , DNA Repair , Humans
14.
Neuroscience ; 145(4): 1439-48, 2007 Apr 14.
Article in English | MEDLINE | ID: mdl-17207936

ABSTRACT

Postmitotic neurons must survive for the entire life of the organism and be able to respond adaptively to adverse conditions of oxidative and genotoxic stress. Unrepaired DNA damage can trigger apoptosis of neurons which is typically mediated by the ataxia telangiectasia mutated (ATM)-p53 pathway. As in all mammalian cells, telomeres in neurons consist of TTAGGG DNA repeats and several associated proteins that form a nucleoprotein complex that prevents chromosome ends from being recognized as double strand breaks. Proteins that stabilize telomeres include TRF1 and TRF2, and proteins known to play important roles in DNA damage responses and DNA repair including ATM, Werner and the catalytic subunit of DNA-dependent protein kinase (DNA-PKcs). We have been performing studies of developing and adult neurons aimed at understanding the effects of global and telomere-directed DNA damage responses in neuronal plasticity and survival in the contexts of aging and neurodegenerative disorders. Deficits in specific DNA repair proteins, including DNA-PKcs and uracil DNA glycosylase (UDG), render neurons vulnerable to adverse conditions of relevance to the pathogenesis of neurodegenerative disorders such as Alzheimer's disease and stroke. Similarly, early postmitotic neurons with reduced telomerase activity exhibit accentuated responses to DNA damage and are prone to apoptosis demonstrating a pivotal role for telomere maintenance in both mitotic cells and postmitotic neurons. Our recent findings suggest key roles for TRF2 in regulating the differentiation and survival of neurons. TRF2 affects cell survival and differentiation by modulating DNA damage pathways, and gene expression. A better understanding of the molecular mechanisms by which neurons respond to global and telomere-specific DNA damage may reveal novel strategies for prevention and treatment of neurodegenerative disorders. Indeed, work in this and other laboratories has shown that dietary folic acid can protect neurons against Alzheimer's disease by keeping homocysteine levels low and thereby minimizing the misincorporation of uracil into DNA in neurons.


Subject(s)
Cellular Senescence/genetics , DNA Damage/genetics , Nervous System/metabolism , Neurodegenerative Diseases/genetics , Neurons/metabolism , Telomere/genetics , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/genetics , DNA Repair-Deficiency Disorders/genetics , DNA Repair-Deficiency Disorders/metabolism , DNA Repair-Deficiency Disorders/physiopathology , DNA-Binding Proteins/genetics , Humans , Nervous System/cytology , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/physiopathology , Nuclear Proteins/genetics , Protein Serine-Threonine Kinases/genetics , TATA Box Binding Protein-Like Proteins/genetics , Telomeric Repeat Binding Protein 2 , Tumor Suppressor Proteins/genetics
15.
Endocr Rev ; 27(4): 398-426, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16543383

ABSTRACT

Meiosis is a double-division process that is preceded by only one DNA replication event to produce haploid gametes. The defining event in meiosis is prophase I, during which chromosome pairs locate each other, become physically connected, and exchange genetic information. Although many aspects of this process have been elucidated in lower organisms, there has been scant information available until now about the process in mammals. Recent advances in genetic analysis, especially in mice and humans, have revealed many genes that play essential roles in meiosis in mammals. These include cell cycle-regulatory proteins that couple the exit from the premeiotic DNA synthesis to the progression through prophase I, the chromosome structural proteins involved in synapsis, and the repair and recombination proteins that process the recombination events. Failure to adequately repair the DNA damage caused by recombination triggers meiotic checkpoints that result in ablation of the germ cells by apoptosis. These analyses have revealed surprising sexual dimorphism in the requirements of different gene products and a much less stringent checkpoint regulation in females. This may provide an explanation for the 10-fold increase in meiotic errors in females compared with males. This review provides a comprehensive analysis of the use of genetic manipulation, particularly in mice, but also of the analysis of mutations in humans, to elucidate the mechanisms that are required for traverse through prophase I.


Subject(s)
Cell Cycle Proteins/physiology , Chromosome Pairing/physiology , Recombination, Genetic/physiology , Animals , BRCA1 Protein/genetics , BRCA1 Protein/physiology , Cell Cycle Proteins/genetics , Chromosome Pairing/genetics , DNA Helicases/genetics , DNA Helicases/physiology , DNA Repair/physiology , DNA Repair-Deficiency Disorders/genetics , DNA Repair-Deficiency Disorders/physiopathology , Humans , Meiotic Prophase I/genetics , Meiotic Prophase I/physiology , Mutation
16.
Radiat Res ; 165(1): 26-34, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16392959

ABSTRACT

Radiation-induced bystander factors have been shown to be more toxic if they are from medium harvested from irradiated repair-deficient cells. The aim of this study was to test the hypothesis that the radiosensitivity of repair-proficient cells can be increased by exposing them to medium-borne factors harvested from sensitive cells and vice versa. Cells from a mismatch repair (MMR)-deficient cell line (Raji 10) with a sensitive response to radiation or the wild-type parent cell line were irradiated to 0.5 Gy gamma rays and then monitored for growth rate in their own medium or in the alternative conditioned medium. In other experiments, cells or conditioned medium were added to reporter cells (HPV-G, which are relatively sensitive keratinocytes, or highly radioresistant HT29 cells). The subsequent responses of the two cell lines to a 0.5-Gy dose of (60)Co gamma rays were measured. The results show that prior exposure of resistant cells to medium from irradiated sensitive cells reduced the clonogenic survival of the subsequently irradiated resistant cells. The reverse is also true. Measurement of the apoptosis index and BCL2 expression confirmed that the harvested medium was capable of modulating apoptosis after irradiation. This may have important applications in tumor therapy and also in the understanding of mechanisms involved in induction of adaptive responses.


Subject(s)
Bystander Effect/radiation effects , Culture Media/metabolism , DNA Repair-Deficiency Disorders/physiopathology , Fibroblasts/metabolism , Fibroblasts/radiation effects , Radiation Tolerance/radiation effects , Apoptosis/radiation effects , Cell Line , Cell Proliferation/radiation effects , Cell Survival/radiation effects , Gamma Rays , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...