101
|
Fleming AM, Burrows CJ. 8-Oxo-7,8-dihydroguanine, friend and foe: Epigenetic-like regulator versus initiator of mutagenesis. DNA Repair (Amst) 2017. [PMID: 28629775 DOI: 10.1016/j.dnarep.2017.06.009] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
A high flux of reactive oxygen species during oxidative stress results in oxidative modification of cellular components including DNA. Oxidative DNA "damage" to the heterocyclic bases is considered deleterious because polymerases may incorrectly read the modifications causing mutations. A prominent member in this class is the oxidized guanine base 8-oxo-7,8-dihydroguanine (OG) that is moderately mutagenic effecting G→T transversion mutations. Recent reports have identified that formation of OG in G-rich regulatory elements in the promoters of the VEGF, TNFα, and SIRT1 genes can increase transcription via activation of the base excision repair (BER) pathway. Work in our laboratory with the G-rich sequence in the promoter of VEGF concluded that BER drives a shift in structure to a G-quadruplex conformation leading to gene activation in mammalian cells. More specifically, removal of OG from the duplex context by 8-oxoguanine glycosylase 1 (OGG1) produces an abasic site (AP) that destabilizes the duplex, shifting the equilibrium toward the G-quadruplex fold because of preferential extrusion of the AP into a loop. The AP is bound but inefficiently cleaved by apurinic/apyrimidinic endoDNase I (APE1) that likely allows recruitment of activating transcription factors for gene induction. The ability of OG to induce transcription ascribes a regulatory or epigenetic-like role for this oxidatively modified base. We compare OG to the 5-methylcytosine (5mC) epigenetic pathway including its oxidized derivatives, some of which poise genes for transcription while also being substrates for BER. The mutagenic potential of OG to induce only ∼one-third the number of mutations (G→T) compared to deamination of 5mC producing C→T mutations is described. These comparisons blur the line between friendly epigenetic base modifications and those that are foes, i.e. DNA "damage," causing genetic mutations.
Collapse
Affiliation(s)
- Aaron M Fleming
- Department of Chemistry, University of Utah, 315 S 1400 East, Salt Lake City, UT 84112-0850, USA.
| | - Cynthia J Burrows
- Department of Chemistry, University of Utah, 315 S 1400 East, Salt Lake City, UT 84112-0850, USA.
| |
Collapse
|
102
|
Nakabeppu Y, Ohta E, Abolhassani N. MTH1 as a nucleotide pool sanitizing enzyme: Friend or foe? Free Radic Biol Med 2017; 107:151-158. [PMID: 27833032 DOI: 10.1016/j.freeradbiomed.2016.11.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 10/30/2016] [Accepted: 11/04/2016] [Indexed: 12/21/2022]
Abstract
8-Oxo-7,8-dihydroguanine (GO) can originate as 8-oxo-7,8-dihydro-2'-deoxyguanosine 5'-triphosphate (8-oxo-dGTP), an oxidized form of dGTP in the nucleotide pool, or by direct oxidation of guanine base in DNA. Accumulation of GO in cellular genomes can result in mutagenesis or programmed cell death, and is thus minimized by the actions of MutT homolog-1 (MTH1) with 8-oxo-dGTPase, OGG1 with GO DNA glycosylase and MutY homolog (MUTYH) with adenine DNA glycosylase. Studies on Mth1/Ogg1/Mutyh-triple knockout mice demonstrated that the defense systems efficiently minimize GO accumulation in cellular genomes, and thus maintain low incidences of spontaneous mutagenesis and tumorigenesis. Mth1/Ogg1-double knockout mice increased GO accumulation in the genome, but exhibited little susceptibility to spontaneous tumorigenesis, thus revealing that accumulation of GO in cellular genomes induces MUTYH-dependent cell death. Cancer cells are exposed to high oxidative stress levels and accumulate a high level of 8-oxo-dGTP in their nucleotide pools; cancer cells consequently express increased levels of MTH1 to eliminate 8-oxo-dGTP, indicating that increased expression of MTH1 in cancer cells may be detrimental for cancer patients. Mth1/Ogg1-double knockout mice are highly vulnerable to neurodegeneration under oxidative conditions, while transgenic expression of human MTH1 efficiently prevents neurodegeneration by avoiding GO accumulation in mitochondrial genomes of neurons and/or nuclear genomes of microglia, indicating that increased expression of MTH1 may be beneficial for neuronal tissues.
Collapse
Affiliation(s)
- Yusaku Nakabeppu
- Division of Neurofunctional Genomics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan.
| | - Eiko Ohta
- Division of Neurofunctional Genomics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Nona Abolhassani
- Division of Neurofunctional Genomics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| |
Collapse
|
103
|
Harris K, Pritchard JK. Rapid evolution of the human mutation spectrum. eLife 2017; 6. [PMID: 28440220 PMCID: PMC5435464 DOI: 10.7554/elife.24284] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 04/07/2017] [Indexed: 01/02/2023] Open
Abstract
DNA is a remarkably precise medium for copying and storing biological information. This high fidelity results from the action of hundreds of genes involved in replication, proofreading, and damage repair. Evolutionary theory suggests that in such a system, selection has limited ability to remove genetic variants that change mutation rates by small amounts or in specific sequence contexts. Consistent with this, using SNV variation as a proxy for mutational input, we report here that mutational spectra differ substantially among species, human continental groups and even some closely related populations. Close examination of one signal, an increased TCC→TTC mutation rate in Europeans, indicates a burst of mutations from about 15,000 to 2000 years ago, perhaps due to the appearance, drift, and ultimate elimination of a genetic modifier of mutation rate. Our results suggest that mutation rates can evolve markedly over short evolutionary timescales and suggest the possibility of mapping mutational modifiers. DOI:http://dx.doi.org/10.7554/eLife.24284.001 DNA is a molecule that contains the information needed to build an organism. This information is stored as a code made up of four chemicals: adenine (A), guanine (G), cytosine (C), and thymine (T). Every time a cell divides and copies its DNA, it accidentally introduces ‘typos’ into the code, known as mutations. Most mutations are harmless, but some can cause damage. All cells have ways to proofread DNA, and the more resources are devoted to proofreading, the less mutations occur. Simple organisms such as bacteria use less energy to reduce mutations, because their genomes may tolerate more damage. More complex organisms, from yeast to humans, instead need to proofread their genomes more thoroughly. Recent research has shown that humans have a lower mutation rate than chimpanzees and gorillas, their closest living relatives. Humans and other apes copy and proofread their DNA with basically the same biological machinery as yeast, which is about a billion years old. Yet, humans and apes have only existed for a small fraction of this time, a few million years. Why then do humans need to replicate and proofread their DNA differently from apes, and could it be that the way mutations arise is still evolving? Previous research revealed that European people experience more mutations within certain DNA motifs (specifically, the DNA sequences ‘TCC’, ‘TCT’, ‘CCC’ and ‘ACC’) than Africans or East Asians do. Now, Harris (who conducted the previous research) and Pritchard have compared how various human ethnic groups accumulate mutations and how these processes differ in different groups. Statistical analysis of the genomes of thousands of people from all over the world did indeed show that the mutation rates of many different three-letter DNA motifs have changed during the past 20,000 years of human evolution. Harris and Pritchard report that when groups of humans left Africa and settled in isolated populations across different continents, each population quickly became better at avoiding mutations in some genomic contexts, but worse in others. This suggests that the risk of passing on harmful mutations to future generations is changing and evolving at an even faster rate than was originally suspected. The results suggest that every human ethnic group carries specific variants of the genes which ensure that DNA replication and repair are accurate. These differences appear to influence which types of mutations are frequently passed down to future generations. An important next step will be to identify the genetic variants that could be controlling mutational patterns and how they affect human health. DOI:http://dx.doi.org/10.7554/eLife.24284.002
Collapse
Affiliation(s)
- Kelley Harris
- Department of Genetics, Stanford University, Stanford, United States
| | - Jonathan K Pritchard
- Department of Genetics, Stanford University, Stanford, United States.,Department of Biology, Stanford University, Stanford, United States.,Howard Hughes Medical Institute, Stanford University, Stanford, United States
| |
Collapse
|
104
|
Su Y, Chen L, Su Y, Li Z, Zhang C, Mu T. Spectroscopic evidences of toxic trans-crotonaldehyde trapped and transformed by resveratrol to prevent the damage of mitochondrial DNA. IUBMB Life 2017; 69:500-509. [DOI: 10.1002/iub.1631] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Accepted: 03/20/2017] [Indexed: 01/16/2023]
Affiliation(s)
- Yanbin Su
- College of Chemical and Pharmaceutical Engineering; Jilin University of Chemical Technology; Jilin China
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, College of Life Sciences; Jilin University; Changchun China
| | - Lei Chen
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, College of Life Sciences; Jilin University; Changchun China
| | - Yanwen Su
- College of Chemical and Pharmaceutical Engineering; Jilin University of Chemical Technology; Jilin China
| | - Zhengqiang Li
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, College of Life Sciences; Jilin University; Changchun China
| | - Chuang Zhang
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, College of Life Sciences; Jilin University; Changchun China
| | - Tongxing Mu
- College of Chemical and Pharmaceutical Engineering; Jilin University of Chemical Technology; Jilin China
| |
Collapse
|
105
|
A Specific Mutational Signature Associated with DNA 8-Oxoguanine Persistence in MUTYH-defective Colorectal Cancer. EBioMedicine 2017; 20:39-49. [PMID: 28551381 PMCID: PMC5478212 DOI: 10.1016/j.ebiom.2017.04.022] [Citation(s) in RCA: 150] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 04/07/2017] [Accepted: 04/11/2017] [Indexed: 01/17/2023] Open
Abstract
8-Oxoguanine, a common mutagenic DNA lesion, generates G:C>T:A transversions via mispairing with adenine during DNA replication. When operating normally, the MUTYH DNA glycosylase prevents 8-oxoguanine-related mutagenesis by excising the incorporated adenine. Biallelic MUTYH mutations impair this enzymatic function and are associated with colorectal cancer (CRC) in MUTYH-Associated Polyposis (MAP) syndrome. Here, we perform whole-exome sequencing that reveals a modest mutator phenotype in MAP CRCs compared to sporadic CRC stem cell lines or bulk tumours. The excess G:C>T:A transversion mutations in MAP CRCs exhibits a novel mutational signature, termed Signature 36, with a strong sequence dependence. The MUTYH mutational signature reflecting persistent 8-oxoG:A mismatches occurs frequently in the APC, KRAS, PIK3CA, FAT4, TP53, FAT1, AMER1, KDM6A, SMAD4 and SMAD2 genes that are associated with CRC. The occurrence of Signature 36 in other types of human cancer indicates that DNA 8-oxoguanine-related mutations might contribute to the development of cancer in other organs.
Collapse
|
106
|
Stephenson AA, Taggart DJ, Suo Z. Noncatalytic, N-terminal Domains of DNA Polymerase Lambda Affect Its Cellular Localization and DNA Damage Response. Chem Res Toxicol 2017; 30:1240-1249. [PMID: 28380295 DOI: 10.1021/acs.chemrestox.7b00067] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Specialized DNA polymerases, such as DNA polymerase lambda (Polλ), are important players in DNA damage tolerance and repair pathways. Knowing how DNA polymerases are regulated and recruited to sites of DNA damage is imperative to understanding these pathways. Recent work has suggested that Polλ plays a role in several distinct DNA damage tolerance and repair pathways. In this paper, we report previously unknown roles of the N-terminal domains of human Polλ for modulating its involvement in DNA damage tolerance and repair. By using Western blot analysis, fluorescence microscopy, and cell survival assays, we found that the BRCA1 C-terminal (BRCT) and proline/serine-rich (PSR) domains of Polλ affect its cellular localization and DNA damage responses. The nuclear localization signal (NLS) of Polλ was necessary to overcome the impediment of its nuclear localization caused by its BRCT and PSR domains. Induction of DNA damage resulted in recruitment of Polλ to chromatin, which was controlled by its BRCT and PSR domains. In addition, the presence of both domains was required for Polλ-mediated tolerance of oxidative DNA damage but not DNA methylation damage. These findings suggest that the N-terminal domains of Polλ are important for regulating its responses to DNA damage.
Collapse
Affiliation(s)
- Anthony A Stephenson
- Department of Chemistry and Biochemistry and ‡The Ohio State Biochemistry Program, The Ohio State University , Columbus, Ohio 43210, United States
| | - David J Taggart
- Department of Chemistry and Biochemistry and ‡The Ohio State Biochemistry Program, The Ohio State University , Columbus, Ohio 43210, United States
| | - Zucai Suo
- Department of Chemistry and Biochemistry and ‡The Ohio State Biochemistry Program, The Ohio State University , Columbus, Ohio 43210, United States
| |
Collapse
|
107
|
Pilati C, Shinde J, Alexandrov LB, Assié G, André T, Hélias‐Rodzewicz Z, Ducoudray R, Le Corre D, Zucman‐Rossi J, Emile J, Bertherat J, Letouzé E, Laurent‐Puig P. Mutational signature analysis identifies
MUTYH
deficiency in colorectal cancers and adrenocortical carcinomas. J Pathol 2017; 242:10-15. [DOI: 10.1002/path.4880] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 01/09/2017] [Accepted: 01/16/2017] [Indexed: 12/12/2022]
Affiliation(s)
- Camilla Pilati
- INSERM UMR‐S1147, Personalized Medicine, Pharmacogenomics, Therapeutic Optimization Université Paris Descartes Paris France
| | - Jayendra Shinde
- INSERM Unité Mixte de Recherche (UMR) 1162 Génomique Fonctionnelle des Tumeurs Solides, Equipe Labellisée Ligue contre le Cancer Paris France
- Université Paris Descartes Labex Immuno‐Oncology Sorbonne Paris Cité Paris France
- Université Paris 13 Sorbonne Paris Cité, Unité de Formation et de Recherche (UFR) Santé, Médecine, Biologie Humaine (SMBH) Bobigny France
- Université Paris Diderot Institut Universitaire d'Hématologie Paris France
| | - Ludmil B Alexandrov
- Theoretical Biology and Biophysics (T‐6) Los Alamos National Laboratory Los Alamos NM USA
- Center for Nonlinear Studies Los Alamos National Laboratory Los Alamos NM USA
| | - Guillaume Assié
- INSERM U1016, CNRS UMR 8104 Paris Descartes University Institut Cochin, Paris France
- Center for Rare Adrenal Diseases, Department of Endocrinology, Assistance Publique‐Hôpitaux de Paris Hôpital Cochin, Paris France
| | - Thierry André
- Department of Medical Oncology AP‐HP, Hospital Saint‐Antoine Paris France
- Université Pierre et Marie Curie (UMPC) Paris VI Paris France
| | - Zofia Hélias‐Rodzewicz
- Department of Pathology AP‐HP, Hôpital Ambroise Paré Paris France
- EA 4340, Université de Versailles Versailles France
| | - Romain Ducoudray
- Department of Pathology AP‐HP, Hôpital Ambroise Paré Paris France
- EA 4340, Université de Versailles Versailles France
| | - Delphine Le Corre
- INSERM UMR‐S1147, Personalized Medicine, Pharmacogenomics, Therapeutic Optimization Université Paris Descartes Paris France
| | - Jessica Zucman‐Rossi
- INSERM Unité Mixte de Recherche (UMR) 1162 Génomique Fonctionnelle des Tumeurs Solides, Equipe Labellisée Ligue contre le Cancer Paris France
- Université Paris Descartes Labex Immuno‐Oncology Sorbonne Paris Cité Paris France
- Université Paris 13 Sorbonne Paris Cité, Unité de Formation et de Recherche (UFR) Santé, Médecine, Biologie Humaine (SMBH) Bobigny France
- Université Paris Diderot Institut Universitaire d'Hématologie Paris France
| | - Jean‐François Emile
- Department of Pathology AP‐HP, Hôpital Ambroise Paré Paris France
- EA 4340, Université de Versailles Versailles France
| | - Jérôme Bertherat
- INSERM U1016, CNRS UMR 8104 Paris Descartes University Institut Cochin, Paris France
- Center for Rare Adrenal Diseases, Department of Endocrinology, Assistance Publique‐Hôpitaux de Paris Hôpital Cochin, Paris France
| | - Eric Letouzé
- INSERM Unité Mixte de Recherche (UMR) 1162 Génomique Fonctionnelle des Tumeurs Solides, Equipe Labellisée Ligue contre le Cancer Paris France
- Université Paris Descartes Labex Immuno‐Oncology Sorbonne Paris Cité Paris France
- Université Paris 13 Sorbonne Paris Cité, Unité de Formation et de Recherche (UFR) Santé, Médecine, Biologie Humaine (SMBH) Bobigny France
- Université Paris Diderot Institut Universitaire d'Hématologie Paris France
| | - Pierre Laurent‐Puig
- INSERM UMR‐S1147, Personalized Medicine, Pharmacogenomics, Therapeutic Optimization Université Paris Descartes Paris France
| |
Collapse
|
108
|
Sharma A, Ansari AH, Kumari R, Pandey R, Rehman R, Mehani B, Varma B, Desiraju BK, Mabalirajan U, Agrawal A, Mukhopadhyay A. Human brain harbors single nucleotide somatic variations in functionally relevant genes possibly mediated by oxidative stress. F1000Res 2017; 5:2520. [PMID: 28149503 DOI: 10.12688/f1000research.9495.2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/08/2016] [Indexed: 12/11/2022] Open
Abstract
Somatic variation in DNA can cause cells to deviate from the preordained genomic path in both disease and healthy conditions. Here, using exome sequencing of paired tissue samples, we show that the normal human brain harbors somatic single base variations measuring up to 0.48% of the total variations. Interestingly, about 64% of these somatic variations in the brain are expected to lead to non-synonymous changes, and as much as 87% of these represent G:C>T:A transversion events. Further, the transversion events in the brain were mostly found in the frontal cortex, whereas the corpus callosum from the same individuals harbors the reference genotype. We found a significantly higher amount of 8-OHdG (oxidative stress marker) in the frontal cortex compared to the corpus callosum of the same subjects (p<0.01), correlating with the higher G:C>T:A transversions in the cortex. We found significant enrichment for axon guidance and related pathways for genes harbouring somatic variations. This could represent either a directed selection of genetic variations in these pathways or increased susceptibility of some loci towards oxidative stress. This study highlights that oxidative stress possibly influence single nucleotide somatic variations in normal human brain.
Collapse
Affiliation(s)
- Anchal Sharma
- Genomics & Molceular Medicine Unit, CSIR-Institute of Genomics & Integrative Biology, Delhi, 110020, India; Academy of Scientific and Innovative Research, CSIR-Institute of Genomics & Integrative Biology (AcSIR-IGIB), Delhi, 110020, India
| | - Asgar Hussain Ansari
- Genomics & Molceular Medicine Unit, CSIR-Institute of Genomics & Integrative Biology, Delhi, 110020, India; Academy of Scientific and Innovative Research, CSIR-Institute of Genomics & Integrative Biology (AcSIR-IGIB), Delhi, 110020, India
| | - Renu Kumari
- Genomics & Molceular Medicine Unit, CSIR-Institute of Genomics & Integrative Biology, Delhi, 110020, India; Academy of Scientific and Innovative Research, CSIR-Institute of Genomics & Integrative Biology (AcSIR-IGIB), Delhi, 110020, India
| | - Rajesh Pandey
- CSIR Ayurgenomics Unit- TRISUTRA, CSIR-Institute of Genomics & Integrative Biology, Delhi, 110020, India
| | - Rakhshinda Rehman
- Academy of Scientific and Innovative Research, CSIR-Institute of Genomics & Integrative Biology (AcSIR-IGIB), Delhi, 110020, India; Molecular Immunogenetics Unit, CSIR-Institute of Genomics & Integrative Biology, Delhi, 110020, India
| | - Bharati Mehani
- Genomics & Molceular Medicine Unit, CSIR-Institute of Genomics & Integrative Biology, Delhi, 110020, India; Academy of Scientific and Innovative Research, CSIR-Institute of Genomics & Integrative Biology (AcSIR-IGIB), Delhi, 110020, India
| | - Binuja Varma
- CSIR Ayurgenomics Unit- TRISUTRA, CSIR-Institute of Genomics & Integrative Biology, Delhi, 110020, India
| | - Bapu K Desiraju
- Academy of Scientific and Innovative Research, CSIR-Institute of Genomics & Integrative Biology (AcSIR-IGIB), Delhi, 110020, India; Molecular Immunogenetics Unit, CSIR-Institute of Genomics & Integrative Biology, Delhi, 110020, India
| | - Ulaganathan Mabalirajan
- Academy of Scientific and Innovative Research, CSIR-Institute of Genomics & Integrative Biology (AcSIR-IGIB), Delhi, 110020, India; Molecular Immunogenetics Unit, CSIR-Institute of Genomics & Integrative Biology, Delhi, 110020, India
| | - Anurag Agrawal
- Academy of Scientific and Innovative Research, CSIR-Institute of Genomics & Integrative Biology (AcSIR-IGIB), Delhi, 110020, India; Molecular Immunogenetics Unit, CSIR-Institute of Genomics & Integrative Biology, Delhi, 110020, India
| | - Arijit Mukhopadhyay
- Genomics & Molceular Medicine Unit, CSIR-Institute of Genomics & Integrative Biology, Delhi, 110020, India; Academy of Scientific and Innovative Research, CSIR-Institute of Genomics & Integrative Biology (AcSIR-IGIB), Delhi, 110020, India; School of Environment and Life Sciences, University of Salford, Manchester, UK
| |
Collapse
|
109
|
Understanding the molecular mechanism for the differential inhibitory activities of compounds against MTH1. Sci Rep 2017; 7:40557. [PMID: 28074893 PMCID: PMC5225434 DOI: 10.1038/srep40557] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 12/08/2016] [Indexed: 01/18/2023] Open
Abstract
MTH1 can hydrolyze oxidized nucleotides and is required for cancer survival. The IC50 values were 0.8 nM for TH287 with a methyl substitution, 5.0 nM for TH588 with a cyclopropyl substitution, and 2.1 μM for TH650 with an oxetanyl substitution. Thus, it is very significant to understand inhibitory mechanisms of these structurally similar compounds against MTH1 and influences of the substituent on the bioactivities. Our MD researches indicate that TH287 maintains significant hydrogen bonds with Asn33 and Asp119, stabilizes the binding site, and induces MTH1 adopt a closed motion, leading to a high inhibitory activity. When bound with TH588, the binding site can be partially stabilized and take a semi-closed state, which is because the cyclopropyl group in TH588 has larger steric hindrance than a methyl group in TH287. So TH588 has a slightly reduced inhibitory activity compared to TH287. TH650 induces greater conformation fluctuations than TH588 and the binding site adopts an opening state, which is caused by the large bulk of oxetanyl group and the interference of solvent on the oxetanyl substituent, leading to the lowest inhibitory activity. Thus, the inhibitory activity follows a TH287 > TH588 > TH650 trend, which well matches with the experimental finding.
Collapse
|
110
|
Molecular pathophysiology of impaired glucose metabolism, mitochondrial dysfunction, and oxidative DNA damage in Alzheimer's disease brain. Mech Ageing Dev 2017; 161:95-104. [DOI: 10.1016/j.mad.2016.05.005] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 05/20/2016] [Accepted: 05/22/2016] [Indexed: 02/07/2023]
|
111
|
Champroux A, Torres-Carreira J, Gharagozloo P, Drevet JR, Kocer A. Mammalian sperm nuclear organization: resiliencies and vulnerabilities. Basic Clin Androl 2016; 26:17. [PMID: 28031843 PMCID: PMC5175393 DOI: 10.1186/s12610-016-0044-5] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 11/12/2016] [Indexed: 01/07/2023] Open
Abstract
Sperm cells are remarkably complex and highly specialized compared to somatic cells. Their function is to deliver to the oocyte the paternal genomic blueprint along with a pool of proteins and RNAs so a new generation can begin. Reproductive success, including optimal embryonic development and healthy offspring, greatly depends on the integrity of the sperm chromatin structure. It is now well documented that DNA damage in sperm is linked to reproductive failures both in natural and assisted conception (Assisted Reproductive Technologies [ART]). This manuscript reviews recent important findings concerning - the unusual organization of mammalian sperm chromatin and its impact on reproductive success when modified. This review is focused on sperm chromatin damage and their impact on embryonic development and transgenerational inheritance.
Collapse
Affiliation(s)
- A. Champroux
- GReD “Genetics, Reproduction & Development” Laboratory, UMR CNRS 6293, INSERM U1103, Clermont Université, BP60026 - TSA60026, 63178 Aubière cedex, France
| | - J. Torres-Carreira
- Centro Universitário Rio Preto, UNIRP, Rodovia Br153, Km 69, CEP15093-450 São José do Rio Preto, São Paulo Brazil
| | - P. Gharagozloo
- CellOxess LLC, 830 Bear Tavern Road, Ewing, NJ 08628 USA
| | - J. R. Drevet
- GReD “Genetics, Reproduction & Development” Laboratory, UMR CNRS 6293, INSERM U1103, Clermont Université, BP60026 - TSA60026, 63178 Aubière cedex, France
| | - A. Kocer
- GReD “Genetics, Reproduction & Development” Laboratory, UMR CNRS 6293, INSERM U1103, Clermont Université, BP60026 - TSA60026, 63178 Aubière cedex, France
| |
Collapse
|
112
|
Raper AT, Reed AJ, Gadkari VV, Suo Z. Advances in Structural and Single-Molecule Methods for Investigating DNA Lesion Bypass and Repair Polymerases. Chem Res Toxicol 2016; 30:260-269. [PMID: 28092942 DOI: 10.1021/acs.chemrestox.6b00342] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Innovative advances in X-ray crystallography and single-molecule biophysics have yielded unprecedented insight into the mechanisms of DNA lesion bypass and damage repair. Time-dependent X-ray crystallography has been successfully applied to view the bypass of 8-oxo-7,8-dihydro-2'-deoxyguanine (8-oxoG), a major oxidative DNA lesion, and the incorporation of the triphosphate form, 8-oxo-dGTP, catalyzed by human DNA polymerase β. Significant findings of these studies are highlighted here, and their contributions to the current mechanistic understanding of mutagenic translesion DNA synthesis (TLS) and base excision repair are discussed. In addition, single-molecule Förster resonance energy transfer (smFRET) techniques have recently been adapted to investigate nucleotide binding and incorporation opposite undamaged dG and 8-oxoG by Sulfolobus solfataricus DNA polymerase IV (Dpo4), a model Y-family DNA polymerase. The mechanistic response of Dpo4 to a DNA lesion and the complex smFRET technique are described here. In this perspective, we also describe how time-dependent X-ray crystallography and smFRET can be used to achieve the spatial and temporal resolutions necessary to answer some of the mechanistic questions that remain in the fields of TLS and DNA damage repair.
Collapse
Affiliation(s)
- Austin T Raper
- Department of Chemistry and Biochemistry and The Ohio State Biochemistry Program, The Ohio State University , Columbus, Ohio 43210, United States
| | - Andrew J Reed
- Department of Chemistry and Biochemistry and The Ohio State Biochemistry Program, The Ohio State University , Columbus, Ohio 43210, United States
| | - Varun V Gadkari
- Department of Chemistry and Biochemistry and The Ohio State Biochemistry Program, The Ohio State University , Columbus, Ohio 43210, United States
| | - Zucai Suo
- Department of Chemistry and Biochemistry and The Ohio State Biochemistry Program, The Ohio State University , Columbus, Ohio 43210, United States
| |
Collapse
|
113
|
Sharma A, Ansari AH, Kumari R, Pandey R, Rehman R, Mehani B, Varma B, Desiraju BK, Mabalirajan U, Agrawal A, Mukhopadhyay A. Human brain harbors single nucleotide somatic variations in functionally relevant genes possibly mediated by oxidative stress. F1000Res 2016; 5:2520. [PMID: 28149503 PMCID: PMC5265704 DOI: 10.12688/f1000research.9495.3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/11/2017] [Indexed: 12/14/2022] Open
Abstract
Somatic variation in DNA can cause cells to deviate from the preordained genomic
path in both disease and healthy conditions. Here, using exome sequencing of
paired tissue samples, we show that the normal human brain harbors somatic
single base variations measuring up to 0.48% of the total variations.
Interestingly, about 64% of these somatic variations in the brain are expected
to lead to non-synonymous changes, and as much as 87% of these represent
G:C>T:A transversion events. Further, the transversion events in the brain
were mostly found in the frontal cortex, whereas the corpus callosum from the
same individuals harbors the reference genotype. We found a significantly higher
amount of 8-OHdG (oxidative stress marker) in the frontal cortex compared to the
corpus callosum of the same subjects (p<0.01), correlating with the higher
G:C>T:A transversions in the cortex. We found significant enrichment for axon
guidance and related pathways for genes harbouring somatic variations. This
could represent either a directed selection of genetic variations in these
pathways or increased susceptibility of some loci towards oxidative stress. This
study highlights that oxidative stress possibly influence single nucleotide
somatic variations in normal human brain.
Collapse
Affiliation(s)
- Anchal Sharma
- Genomics & Molceular Medicine Unit, CSIR-Institute of Genomics & Integrative Biology, Delhi, 110020, India; Academy of Scientific and Innovative Research, CSIR-Institute of Genomics & Integrative Biology (AcSIR-IGIB), Delhi, 110020, India
| | - Asgar Hussain Ansari
- Genomics & Molceular Medicine Unit, CSIR-Institute of Genomics & Integrative Biology, Delhi, 110020, India; Academy of Scientific and Innovative Research, CSIR-Institute of Genomics & Integrative Biology (AcSIR-IGIB), Delhi, 110020, India
| | - Renu Kumari
- Genomics & Molceular Medicine Unit, CSIR-Institute of Genomics & Integrative Biology, Delhi, 110020, India; Academy of Scientific and Innovative Research, CSIR-Institute of Genomics & Integrative Biology (AcSIR-IGIB), Delhi, 110020, India
| | - Rajesh Pandey
- CSIR Ayurgenomics Unit- TRISUTRA, CSIR-Institute of Genomics & Integrative Biology, Delhi, 110020, India
| | - Rakhshinda Rehman
- Academy of Scientific and Innovative Research, CSIR-Institute of Genomics & Integrative Biology (AcSIR-IGIB), Delhi, 110020, India; Molecular Immunogenetics Unit, CSIR-Institute of Genomics & Integrative Biology, Delhi, 110020, India
| | - Bharati Mehani
- Genomics & Molceular Medicine Unit, CSIR-Institute of Genomics & Integrative Biology, Delhi, 110020, India; Academy of Scientific and Innovative Research, CSIR-Institute of Genomics & Integrative Biology (AcSIR-IGIB), Delhi, 110020, India
| | - Binuja Varma
- CSIR Ayurgenomics Unit- TRISUTRA, CSIR-Institute of Genomics & Integrative Biology, Delhi, 110020, India
| | - Bapu K Desiraju
- Academy of Scientific and Innovative Research, CSIR-Institute of Genomics & Integrative Biology (AcSIR-IGIB), Delhi, 110020, India; Molecular Immunogenetics Unit, CSIR-Institute of Genomics & Integrative Biology, Delhi, 110020, India
| | - Ulaganathan Mabalirajan
- Academy of Scientific and Innovative Research, CSIR-Institute of Genomics & Integrative Biology (AcSIR-IGIB), Delhi, 110020, India; Molecular Immunogenetics Unit, CSIR-Institute of Genomics & Integrative Biology, Delhi, 110020, India
| | - Anurag Agrawal
- Academy of Scientific and Innovative Research, CSIR-Institute of Genomics & Integrative Biology (AcSIR-IGIB), Delhi, 110020, India; Molecular Immunogenetics Unit, CSIR-Institute of Genomics & Integrative Biology, Delhi, 110020, India
| | - Arijit Mukhopadhyay
- Genomics & Molceular Medicine Unit, CSIR-Institute of Genomics & Integrative Biology, Delhi, 110020, India; Academy of Scientific and Innovative Research, CSIR-Institute of Genomics & Integrative Biology (AcSIR-IGIB), Delhi, 110020, India; School of Environment and Life Sciences, University of Salford, Manchester, UK
| |
Collapse
|
114
|
Nakatake S, Murakami Y, Ikeda Y, Morioka N, Tachibana T, Fujiwara K, Yoshida N, Notomi S, Hisatomi T, Yoshida S, Ishibashi T, Nakabeppu Y, Sonoda KH. MUTYH promotes oxidative microglial activation and inherited retinal degeneration. JCI Insight 2016; 1:e87781. [PMID: 27699246 DOI: 10.1172/jci.insight.87781] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Oxidative stress is implicated in various neurodegenerative disorders, including retinitis pigmentosa (RP), an inherited disease that causes blindness. The biological and cellular mechanisms by which oxidative stress mediates neuronal cell death are largely unknown. In a mouse model of RP (rd10 mice), we show that oxidative DNA damage activates microglia through MutY homolog-mediated (MUYTH-mediated) base excision repair (BER), thereby exacerbating retinal inflammation and degeneration. In the early stage of retinal degeneration, oxidative DNA damage accumulated in the microglia and caused single-strand breaks (SSBs) and poly(ADP-ribose) polymerase activation. In contrast, Mutyh deficiency in rd10 mice prevented SSB formation in microglia, which in turn suppressed microglial activation and photoreceptor cell death. Moreover, Mutyh-deficient primary microglial cells attenuated the polarization to the inflammatory and cytotoxic phenotype under oxidative stress. Thus, MUTYH-mediated BER in oxidative microglial activation may be a novel target to dampen the disease progression in RP and other neurodegenerative disorders that are associated with oxidative stress.
Collapse
Affiliation(s)
- Shunji Nakatake
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Maidashi, Higashi-Ku, Fukuoka, Japan
| | - Yusuke Murakami
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Maidashi, Higashi-Ku, Fukuoka, Japan
| | - Yasuhiro Ikeda
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Maidashi, Higashi-Ku, Fukuoka, Japan
| | - Noriko Morioka
- Division of Neurofunctional Genomics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, Maidashi, Higashi-Ku, Fukuoka, Japan
| | - Takashi Tachibana
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Maidashi, Higashi-Ku, Fukuoka, Japan
| | - Kohta Fujiwara
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Maidashi, Higashi-Ku, Fukuoka, Japan.,Department of Ophthalmology, Graduate School of Medical Sciences, Akita University, Hondo, Akita, Japan
| | - Noriko Yoshida
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Maidashi, Higashi-Ku, Fukuoka, Japan
| | - Shoji Notomi
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Maidashi, Higashi-Ku, Fukuoka, Japan
| | - Toshio Hisatomi
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Maidashi, Higashi-Ku, Fukuoka, Japan
| | - Shigeo Yoshida
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Maidashi, Higashi-Ku, Fukuoka, Japan
| | - Tatsuro Ishibashi
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Maidashi, Higashi-Ku, Fukuoka, Japan
| | - Yusaku Nakabeppu
- Division of Neurofunctional Genomics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, Maidashi, Higashi-Ku, Fukuoka, Japan
| | - Koh-Hei Sonoda
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Maidashi, Higashi-Ku, Fukuoka, Japan
| |
Collapse
|
115
|
Shifting patterns of genomic variation in the somatic evolution of papillary thyroid carcinoma. BMC Cancer 2016; 16:646. [PMID: 27538953 PMCID: PMC4989347 DOI: 10.1186/s12885-016-2665-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 08/02/2016] [Indexed: 12/12/2022] Open
Abstract
Background Cancer is increasingly understood to arise in the context of dynamically evolving genomes with continuously generated variants subject to selective pressures. Diverse mutations have been identified in papillary thyroid carcinoma (PTC), but unifying theories underlying genomic change are lacking. Applying a framework of somatic evolution, we sought to broaden understanding of the PTC genome through identification of global trends that help explain risk of tumorigenesis. Methods Exome sequencing was performed on 53 PTC and matched adjacent non-tumor thyroid tissues (ANT). Single nucleotide substitution (SNS) signatures from each sample pair were divided into three subsets based on their presence in tumor, non-tumor thyroid, or both. Nine matched blood samples were sequenced and SNS signatures intersected with these three subsets. The intersected genomic signatures were used to define branch-points in the evolution of the tumor genome, distinguishing variants present in the tissues’ common ancestor cells from those unique to each tissue type and therefore acquired after genomic divergence of the tumor, non-tumor, and blood samples. Results Single nucleotide substitutions shared by the tumor and the non-tumor thyroid were dominated by C-to-T transitions, whereas those unique to either tissue type were enriched for C-to-A transversions encoding non-synonymous, predicted-deleterious variants. On average, SNSs of matched blood samples were 81 % identical to those shared by tumor and non-tumor thyroid, but only 12.5 % identical to those unique to either tissue. Older age and BRAF mutation were associated with increased SNS burden. Conclusions The current study demonstrates novel patterns of genomic change in PTC, supporting a theory of somatic evolution in which the zygote’s germline genome undergoes continuous remodeling to produce progressively differentiated, tissue-specific signatures. Late somatic events in thyroid tissue demonstrate shifted mutational spectra compared to earlier polymorphisms. These late events are enriched for predicted-deleterious variants, suggesting a mechanism of genomic instability in PTC tumorigenesis. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2665-7) contains supplementary material, which is available to authorized users.
Collapse
|
116
|
Mutational History of a Human Cell Lineage from Somatic to Induced Pluripotent Stem Cells. PLoS Genet 2016; 12:e1005932. [PMID: 27054363 PMCID: PMC4824386 DOI: 10.1371/journal.pgen.1005932] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 02/22/2016] [Indexed: 12/13/2022] Open
Abstract
The accuracy of replicating the genetic code is fundamental. DNA repair mechanisms protect the fidelity of the genome ensuring a low error rate between generations. This sustains the similarity of individuals whilst providing a repertoire of variants for evolution. The mutation rate in the human genome has recently been measured to be 50–70 de novo single nucleotide variants (SNVs) between generations. During development mutations accumulate in somatic cells so that an organism is a mosaic. However, variation within a tissue and between tissues has not been analysed. By reprogramming somatic cells into induced pluripotent stem cells (iPSCs), their genomes and the associated mutational history are captured. By sequencing the genomes of polyclonal and monoclonal somatic cells and derived iPSCs we have determined the mutation rates and show how the patterns change from a somatic lineage in vivo through to iPSCs. Somatic cells have a mutation rate of 14 SNVs per cell per generation while iPSCs exhibited a ten-fold lower rate. Analyses of mutational signatures suggested that deamination of methylated cytosine may be the major mutagenic source in vivo, whilst oxidative DNA damage becomes dominant in vitro. Our results provide insights for better understanding of mutational processes and lineage relationships between human somatic cells. Furthermore it provides a foundation for interpretation of elevated mutation rates and patterns in cancer. The mutation load of human tissues is unknown and represents the genetic divergence from the fertilised egg. Reprogramming of somatic cells generates induced pluripotent stem cells (iPSCs), a cell type being considered for clinical applications. We generated iPSCs from tissues of healthy individuals and used whole genome sequencing to identify in vivo mutations accrued in a somatic cell during the lifetime of the individual. Next we identified in vitro mutations introduced during reprogramming and cell culture. Each has a unique mutation signature suggesting different mutagenic processes. Our study demonstrates the use of reprogramming as a tool to elucidate mutational processes within normal cells and highlights the importance of genetic characterisation of iPSCs prior to clinical translation.
Collapse
|
117
|
Lewis SEM, Kumar K. The paternal genome and the health of the assisted reproductive technology child. Asian J Androl 2016; 17:616-22. [PMID: 25926606 PMCID: PMC4492053 DOI: 10.4103/1008-682x.153301] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
As a number of children born by assisted reproductive technology (ART) are increasing each year across the developed world, the health of such offspring is a matter of public concern. Does the integrity of the paternal genome impact on offspring health? In societal terms, as birth rates fall, and the Western population become unsustainable, do the benefits outweigh the costs of creating and providing for this ART conceived subpopulation? There are little data to date to answer these questions. The long-term health of such children has largely been ignored, and success measured only by early (prebirth) outcomes such as embryo quality or pregnancy. However, there are powerful paradigms such as ageing and smoking that give vital clues as to the potential impact of unhealthy spermatozoa on disease risk, mental and physical health, fertility and mortality of these offspring.
Collapse
Affiliation(s)
- Sheena E M Lewis
- Centre for Public Health, Queen's University Belfast, Grosvenor Road, BT12 6BJ,NI, UK
| | | |
Collapse
|
118
|
Leon J, Sakumi K, Castillo E, Sheng Z, Oka S, Nakabeppu Y. 8-Oxoguanine accumulation in mitochondrial DNA causes mitochondrial dysfunction and impairs neuritogenesis in cultured adult mouse cortical neurons under oxidative conditions. Sci Rep 2016; 6:22086. [PMID: 26912170 PMCID: PMC4766534 DOI: 10.1038/srep22086] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 02/05/2016] [Indexed: 01/20/2023] Open
Abstract
Oxidative stress and mitochondrial dysfunction are implicated in aging-related neurodegenerative disorders. 8-Oxoguanine (8-oxoG), a common oxidised base lesion, is often highly accumulated in brains from patients with neurodegenerative disorders. MTH1 hydrolyses 8-oxo-2'-deoxyguanosine triphosphate (8-oxo-dGTP) to 8-oxo-dGMP and pyrophosphate in nucleotide pools, while OGG1 excises 8-oxoG paired with cytosine in DNA, thereby minimising the accumulation of 8-oxoG in DNA. Mth1/Ogg1-double knockout (TO-DKO) mice are highly susceptible to neurodegeneration under oxidative conditions and show increased accumulation of 8-oxoG in mitochondrial DNA (mtDNA) in neurons, suggesting that 8-oxoG accumulation in mtDNA causes mitochondrial dysfunction. Here, we evaluated the contribution of MTH1 and OGG1 to the prevention of mitochondrial dysfunction during neuritogenesis in vitro. We isolated cortical neurons from adult wild-type and TO-DKO mice and maintained them with or without antioxidants for 2 to 5 days and then examined neuritogenesis. In the presence of antioxidants, both TO-DKO and wild-type neurons exhibited efficient neurite extension and arborisation. However, in the absence of antioxidants, the accumulation of 8-oxoG in mtDNA of TO-DKO neurons was increased resulting in mitochondrial dysfunction. Cells also exhibited poor neurite outgrowth with decreased complexity of neuritic arborisation, indicating that MTH1 and OGG1 are essential for neuritogenesis under oxidative conditions.
Collapse
Affiliation(s)
- Julio Leon
- Division of Neurofunctional Genomics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Kunihiko Sakumi
- Division of Neurofunctional Genomics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Erika Castillo
- Division of Neurofunctional Genomics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Zijing Sheng
- Division of Neurofunctional Genomics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Sugako Oka
- Division of Neurofunctional Genomics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Yusaku Nakabeppu
- Division of Neurofunctional Genomics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| |
Collapse
|
119
|
Aitken RJ, Gibb Z, Baker MA, Drevet J, Gharagozloo P. Causes and consequences of oxidative stress in spermatozoa. Reprod Fertil Dev 2016; 28:1-10. [DOI: 10.1071/rd15325] [Citation(s) in RCA: 218] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Spermatozoa are highly vulnerable to oxidative attack because they lack significant antioxidant protection due to the limited volume and restricted distribution of cytoplasmic space in which to house an appropriate armoury of defensive enzymes. In particular, sperm membrane lipids are susceptible to oxidative stress because they abound in significant amounts of polyunsaturated fatty acids. Susceptibility to oxidative attack is further exacerbated by the fact that these cells actively generate reactive oxygen species (ROS) in order to drive the increase in tyrosine phosphorylation associated with sperm capacitation. However, this positive role for ROS is reversed when spermatozoa are stressed. Under these conditions, they default to an intrinsic apoptotic pathway characterised by mitochondrial ROS generation, loss of mitochondrial membrane potential, caspase activation, phosphatidylserine exposure and oxidative DNA damage. In responding to oxidative stress, spermatozoa only possess the first enzyme in the base excision repair pathway, 8-oxoguanine DNA glycosylase. This enzyme catalyses the formation of abasic sites, thereby destabilising the DNA backbone and generating strand breaks. Because oxidative damage to sperm DNA is associated with both miscarriage and developmental abnormalities in the offspring, strategies for the amelioration of such stress, including the development of effective antioxidant formulations, are becoming increasingly urgent.
Collapse
|
120
|
Gao T, Gu S, Liu F, Li L, Wang Z, Yang J, Li G. Investigation of MTH1 activity via mismatch-based DNA chain elongation. Anal Chim Acta 2016; 905:66-71. [DOI: 10.1016/j.aca.2015.12.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 12/02/2015] [Accepted: 12/11/2015] [Indexed: 12/27/2022]
|
121
|
Liu L, Li Y, Li T, Xie J, Chen C, Liu Q, Zhang S, Wu HC. Selective Detection of 8-Oxo-2'-deoxyguanosine in Single-Stranded DNA via Nanopore Sensing Approach. Anal Chem 2015; 88:1073-7. [PMID: 26699617 DOI: 10.1021/acs.analchem.5b04102] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
We have developed a nanopore sensing approach for the selective detection of 8-oxo-2'-deoxyguanosine (8-oxoG) in single-stranded DNA. First, 1,12-dodecanediamine is coupled with 8-oxoG-containing DNA molecules in high yield which leaves a free amine group for subsequent attaching of an adamantane moiety. After incubation with cucurbit[7]uril, the host-guest complex-modified DNA hybrid is translocated through an α-hemolysin nanopore. Highly characteristic events can be recorded and used to quantify the 8-oxoG-DNA content in a DNA mixture. Compared with the existing methods, this study provides a reliable, quick, and low-cost approach for the detection of 8-oxoG site in single-stranded DNA at the single-molecule level, particularly suitable for high-throughput screening of a massive number of samples.
Collapse
Affiliation(s)
- Lei Liu
- Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences , Beijing 100049, China
| | - Yuru Li
- Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences , Beijing 100049, China
| | - Ting Li
- Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences , Beijing 100049, China
| | - Jiani Xie
- Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences , Beijing 100049, China
| | - Chaofei Chen
- Multidisciplinary Center, Institute of High Energy Physics, Chinese Academy of Sciences , Beijing 100049, China
| | - Quansheng Liu
- Multidisciplinary Center, Institute of High Energy Physics, Chinese Academy of Sciences , Beijing 100049, China
| | - Shouwen Zhang
- Epilepsy Department, Beijing ChaoYang Emergency Medical Center , Beijing 100021, China
| | - Hai-Chen Wu
- Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences , Beijing 100049, China
| |
Collapse
|
122
|
Zhu J, Fleming AM, Orendt AM, Burrows CJ. pH-Dependent Equilibrium between 5-Guanidinohydantoin and Iminoallantoin Affects Nucleotide Insertion Opposite the DNA Lesion. J Org Chem 2015; 81:351-9. [PMID: 26582419 DOI: 10.1021/acs.joc.5b02180] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Four-electron oxidation of 2'-deoxyguanosine (dG) yields 5-guanidinohydantoin (dGh) as a product. Previously, we hypothesized that dGh could isomerize to iminoallantoin (dIa) via a mechanism similar to the isomerization of allantoin. The isomerization reaction was monitored by HPLC and found to be pH dependent with a transition pH = 10.1 in which dGh was favored at low pH and dIa was favored at high pH. The structures for these isomers were confirmed by UV-vis, MS, and (1)H and (13)C NMR. Additionally, the UV-vis and NMR experimental results are supported by density functional theory calculations. A mechanism is proposed to support the pH dependency of the isomerization reaction. Next, we noted the hydantoin ring of dGh mimics thymine, while the iminohydantoin ring of dIa mimics cytosine; consequently, a dGh/dIa site was synthesized in a DNA template strand, and standing start primer extension studies were conducted with Klenow fragment exo(-). The dATP/dGTP insertion ratio opposite the dGh/dIa site as a function of pH was evaluated from pH 6.5-9.0. At pH 6.5, only dATP was inserted, but as the pH increased to 9.0, the amount of dGTP insertion steadily increased. This observation supports dGh to dIa isomerization in DNA with a transition pH of ∼8.2.
Collapse
Affiliation(s)
- Judy Zhu
- Department of Chemistry, University of Utah , 315 South 1400 East, Salt Lake City, Utah 84112-0850, United States
| | - Aaron M Fleming
- Department of Chemistry, University of Utah , 315 South 1400 East, Salt Lake City, Utah 84112-0850, United States
| | - Anita M Orendt
- Department of Chemistry, University of Utah , 315 South 1400 East, Salt Lake City, Utah 84112-0850, United States.,Center for High Performance Computing, University of Utah , Salt Lake City, Utah 84112-0190, United States
| | - Cynthia J Burrows
- Department of Chemistry, University of Utah , 315 South 1400 East, Salt Lake City, Utah 84112-0850, United States
| |
Collapse
|
123
|
Lord T, Aitken RJ. Fertilization stimulates 8-hydroxy-2′-deoxyguanosine repair and antioxidant activity to prevent mutagenesis in the embryo. Dev Biol 2015; 406:1-13. [DOI: 10.1016/j.ydbio.2015.07.024] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 07/26/2015] [Accepted: 07/29/2015] [Indexed: 12/19/2022]
|
124
|
Ozkosem B, Feinstein SI, Fisher AB, O'Flaherty C. Advancing age increases sperm chromatin damage and impairs fertility in peroxiredoxin 6 null mice. Redox Biol 2015; 5:15-23. [PMID: 25796034 PMCID: PMC4371547 DOI: 10.1016/j.redox.2015.02.004] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 02/19/2015] [Accepted: 02/22/2015] [Indexed: 12/23/2022] Open
Abstract
Due to socioeconomic factors, more couples are choosing to delay conception than ever. Increasing average maternal and paternal age in developed countries over the past 40 years has raised the question of how aging affects reproductive success of males and females. Since oxidative stress in the male reproductive tract increases with age, we investigated the impact of advanced paternal age on the integrity of sperm nucleus and reproductive success of males by using a Prdx6(-/-) mouse model. We compared sperm motility, cytoplasmic droplet retention sperm chromatin quality and reproductive outcomes of young (2-month-old), adult (8-month-old), and old (20-month-old) Prdx6(-/-) males with their age-matched wild type (WT) controls. Absence of PRDX6 caused age-dependent impairment of sperm motility and sperm maturation and increased sperm DNA fragmentation and oxidation as well as decreased sperm DNA compaction and protamination. Litter size, total number of litters and total number of pups per male were significantly lower in Prdx6(-/-) males compared to WT controls. These abnormal reproductive outcomes were severely affected by age in Prdx6(-/-) males. In conclusion, the advanced paternal age affects sperm chromatin integrity and fertility more severely in the absence of PRDX6, suggesting a protective role of PRDX6 in age-associated decline in the sperm quality and fertility in mice.
Collapse
Affiliation(s)
- Burak Ozkosem
- Urology Research Laboratory, Research Institute of the McGill University Health Centre, McGill University, Montréal, Québec, Canada; Department of Surgery (Urology Division), Research Institute of the McGill University Health Centre, McGill University, Montréal, Québec, Canada
| | - Sheldon I Feinstein
- Institute for Environmental Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Aron B Fisher
- Institute for Environmental Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Cristian O'Flaherty
- Urology Research Laboratory, Research Institute of the McGill University Health Centre, McGill University, Montréal, Québec, Canada; Department of Surgery (Urology Division), Research Institute of the McGill University Health Centre, McGill University, Montréal, Québec, Canada; Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada.
| |
Collapse
|
125
|
Uchimura A, Higuchi M, Minakuchi Y, Ohno M, Toyoda A, Fujiyama A, Miura I, Wakana S, Nishino J, Yagi T. Germline mutation rates and the long-term phenotypic effects of mutation accumulation in wild-type laboratory mice and mutator mice. Genome Res 2015; 25:1125-34. [PMID: 26129709 PMCID: PMC4509997 DOI: 10.1101/gr.186148.114] [Citation(s) in RCA: 121] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 05/30/2015] [Indexed: 12/19/2022]
Abstract
The germline mutation rate is an important parameter that affects the amount of genetic variation and the rate of evolution. However, neither the rate of germline mutations in laboratory mice nor the biological significance of the mutation rate in mammalian populations is clear. Here we studied genome-wide mutation rates and the long-term effects of mutation accumulation on phenotype in more than 20 generations of wild-type C57BL/6 mice and mutator mice, which have high DNA replication error rates. We estimated the base-substitution mutation rate to be 5.4 × 10−9 (95% confidence interval = 4.6 × 10−9–6.5 × 10−9) per nucleotide per generation in C57BL/6 laboratory mice, about half the rate reported in humans. The mutation rate in mutator mice was 17 times that in wild-type mice. Abnormal phenotypes were 4.1-fold more frequent in the mutator lines than in the wild-type lines. After several generations, the mutator mice reproduced at substantially lower rates than the controls, exhibiting low pregnancy rates, lower survival rates, and smaller litter sizes, and many of the breeding lines died out. These results provide fundamental information about mouse genetics and reveal the impact of germline mutation rates on phenotypes in a mammalian population.
Collapse
Affiliation(s)
- Arikuni Uchimura
- KOKORO-Biology Group, Laboratories for Integrated Biology, Graduate School of Frontier Biosciences, Osaka University, Suita 565-0871, Japan
| | - Mayumi Higuchi
- KOKORO-Biology Group, Laboratories for Integrated Biology, Graduate School of Frontier Biosciences, Osaka University, Suita 565-0871, Japan
| | - Yohei Minakuchi
- Comparative Genomics Laboratory, National Institute of Genetics, Mishima 411-8540, Japan
| | - Mizuki Ohno
- Department of Medical Biophysics and Radiation Biology, Faculty of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Atsushi Toyoda
- Comparative Genomics Laboratory, National Institute of Genetics, Mishima 411-8540, Japan
| | - Asao Fujiyama
- Comparative Genomics Laboratory, National Institute of Genetics, Mishima 411-8540, Japan
| | - Ikuo Miura
- Technology and Development Team for Mouse Phenotype Analysis, Japan Mouse Clinic, RIKEN BioResource Center, Tsukuba 305-0074, Japan
| | - Shigeharu Wakana
- Technology and Development Team for Mouse Phenotype Analysis, Japan Mouse Clinic, RIKEN BioResource Center, Tsukuba 305-0074, Japan
| | - Jo Nishino
- Department of Biostatistics, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Takeshi Yagi
- KOKORO-Biology Group, Laboratories for Integrated Biology, Graduate School of Frontier Biosciences, Osaka University, Suita 565-0871, Japan
| |
Collapse
|
126
|
Taniguchi Y, Kikukawa Y, Sasaki S. Discrimination Between 8-Oxo-2′-Deoxyguanosine and 2′-Deoxyguanosine in DNA by the Single Nucleotide Primer Extension Reaction with Adap Triphosphate. Angew Chem Int Ed Engl 2015. [DOI: 10.1002/ange.201412086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
127
|
Taniguchi Y, Kikukawa Y, Sasaki S. Discrimination between 8-oxo-2'-deoxyguanosine and 2'-deoxyguanosine in DNA by the single nucleotide primer extension reaction with adap triphosphate. Angew Chem Int Ed Engl 2015; 54:5147-51. [PMID: 25727406 DOI: 10.1002/anie.201412086] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 01/12/2015] [Indexed: 12/16/2022]
Abstract
The adenosine derivative of 2-oxo-1,3-diazaphenoxazine (Adap) exhibits a superb ability to recognize and form base pairs with 8-oxo-2'-deoxyguanosine (8-oxo-dG) in duplex DNA. In this study, the triphosphate of Adap (dAdapTP) was synthesized and tested for single nucleotide incorporation into primer strands using the Klenow Fragment. The efficiency of dAdapTP incorporation into 8-oxo-dG-containing templates was more than 36-fold higher than with dG-containing templates, and provides better discrimination than does the incorporation of natural 2'-deoxyadenosine triphosphate (dATP). The selective incorporation of dAdapTP into 8-oxo-dG templates was therefore applied to the detection of 8-oxo-dG in human telomeric DNA sequences extracted from H2 O2 -treated HeLa cells. The enzymatic incorporation of dAdapTP into 8-oxo-dG-containing templates may provide a novel basis for sequencing oxidative DNA damage in the genome.
Collapse
Affiliation(s)
- Yosuke Taniguchi
- Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582 (Japan).
| | | | | |
Collapse
|
128
|
Arime Y, Fukumura R, Miura I, Mekada K, Yoshiki A, Wakana S, Gondo Y, Akiyama K. Effects of background mutations and single nucleotide polymorphisms (SNPs) on the Disc1 L100P behavioral phenotype associated with schizophrenia in mice. Behav Brain Funct 2014; 10:45. [PMID: 25487992 PMCID: PMC4295473 DOI: 10.1186/1744-9081-10-45] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Accepted: 11/21/2014] [Indexed: 01/31/2023] Open
Abstract
Background Disrupted-in-schizophrenia 1 (DISC1) is a promising candidate susceptibility gene for psychiatric disorders, including schizophrenia, bipolar disorder and major depression. Several previous studies reported that mice with N-ethyl-N-nitrosourea (ENU)-induced L100P mutation in Disc1 showed some schizophrenia-related behavioral phenotypes. This line originally carried several thousands of ENU-induced point mutations in the C57BL/6 J strain and single nucleotide polymorphisms (SNPs) from the DBA/2 J inbred strain. Methods To investigate the effect of Disc1 L100P, background mutations and SNPs on phenotypic characterization, we performed behavioral analyses to better understand phenotypes of Disc1 L100P mice and comprehensive genetic analyses using whole-exome resequencing and SNP panels to map ENU-induced mutations and strain-specific SNPs, respectively. Results We found no differences in spontaneous or methamphetamine-induced locomotor activity, sociability or social novelty preference among Disc1 L100P/L100P, L100P/+ mutants and wild-type littermates. Whole-exome resequencing of the original G1 mouse identified 117 ENU-induced variants, including Disc1 L100P per se. Two females and three males from the congenic L100P strain after backcrossing to C57BL/6 J were deposited to RIKEN BioResource Center in 2008. We genotyped them with DBA/2 J × C57BL/6 J SNPs and found a number of the checked SNPs still remained. Conclusion These results suggest that causal attribution of the discrepancy in behavioral phenotypes to the Disc1 L100P mutant mouse line existing among different research groups needs to be cautiously investigated in further study by taking into account the effect(s) of other ENU-induced mutations and/or SNPs from DBA/2 J. Electronic supplementary material The online version of this article (doi:10.1186/1744-9081-10-45) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Kazufumi Akiyama
- Department of Biological Psychiatry and Neuroscience, Dokkyo Medical University School of Medicine, 800 Kitakobayashi, Mibu-machi, Shimotsuga-gun, Tochigi 321-0293, Japan.
| |
Collapse
|
129
|
Graupner A, Instanes C, Andersen JM, Brandt-Kjelsen A, Dertinger SD, Salbu B, Brunborg G, Olsen AK. Genotoxic effects of two-generational selenium deficiency in mouse somatic and testicular cells. Mutagenesis 2014; 30:217-25. [PMID: 25358475 DOI: 10.1093/mutage/geu059] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Many studies have investigated genotoxic effects of high Se diets but very few have addressed the genotoxicity of Se deprivation and its consequences in germ cells and none in somatic cells. To address these data gaps, C57BL/6 male mice were subjected to Se deprivation starting in the parental generation, i.e. before conception. Mice were given a diet of either low (0.01mg Se/kg diet) or normal (0.23mg Se/kg diet) Se content. Ogg1-deficient (Ogg1 (-/-) ) mice were used as a sensitive model towards oxidative stress due to their reduced capacity to repair oxidised purines. Ogg1 (-/-) mice also mimic the repair characteristics of human post-meiotic male germ cells which have a reduced ability to repair such lesions. The genotoxicity of Se deficiency was addressed by measuring DNA lesions with the alkaline single cell gel electrophoresis (+ Fpg to detect oxidised DNA lesions) in somatic cells (nucleated blood cells and lung cells) and male germ cells (testicular cells). Total Se concentration in liver and GPx activity in plasma and testicular cells were measured. Gene mutation was evaluated by an erythrocyte-based Pig-a assay. We found that Se deprivation of F1 from their conception and until early adulthood led to the induction of DNA lesions in testicular and lung cells expressed as significantly increased levels of DNA lesions, irrespective of the mouse genotype. In blood cells, Se levels did not appear to affect DNA lesions or mutant cell frequencies. The results suggest that the testis was the most sensitive tissue. Thus, genotoxicity induced by the low Se diet in the spermatozoal genome has potential implications for the offspring.
Collapse
Affiliation(s)
- Anne Graupner
- Department of Chemicals and Radiation, National Institute of Public Health, Oslo 0456, Norway, Department of Plant and Environmental Sciences, University of Life Sciences, Aas 1430, Norway, and Litron Laboratories, Rochester, NY 14623, USA
| | - Christine Instanes
- Department of Chemicals and Radiation, National Institute of Public Health, Oslo 0456, Norway, Department of Plant and Environmental Sciences, University of Life Sciences, Aas 1430, Norway, and Litron Laboratories, Rochester, NY 14623, USA
| | - Jill M Andersen
- Department of Chemicals and Radiation, National Institute of Public Health, Oslo 0456, Norway, Department of Plant and Environmental Sciences, University of Life Sciences, Aas 1430, Norway, and Litron Laboratories, Rochester, NY 14623, USA
| | - Anicke Brandt-Kjelsen
- Department of Plant and Environmental Sciences, University of Life Sciences, Aas 1430, Norway, and
| | | | - Brit Salbu
- Department of Plant and Environmental Sciences, University of Life Sciences, Aas 1430, Norway, and
| | - Gunnar Brunborg
- Department of Chemicals and Radiation, National Institute of Public Health, Oslo 0456, Norway, Department of Plant and Environmental Sciences, University of Life Sciences, Aas 1430, Norway, and Litron Laboratories, Rochester, NY 14623, USA
| | - Ann-Karin Olsen
- Department of Chemicals and Radiation, National Institute of Public Health, Oslo 0456, Norway, Department of Plant and Environmental Sciences, University of Life Sciences, Aas 1430, Norway, and Litron Laboratories, Rochester, NY 14623, USA.
| |
Collapse
|
130
|
Ségurel L, Wyman MJ, Przeworski M. Determinants of Mutation Rate Variation in the Human Germline. Annu Rev Genomics Hum Genet 2014; 15:47-70. [DOI: 10.1146/annurev-genom-031714-125740] [Citation(s) in RCA: 232] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Laure Ségurel
- Laboratoire Éco-Anthropologie et Ethnobiologie, UMR 7206, Muséum National d'Histoire Naturelle–Centre National de la Recherche Scientifique–Université Paris 7 Diderot, Paris 75231, France;
| | - Minyoung J. Wyman
- Department of Biological Sciences, Columbia University, New York, NY 10027;
| | - Molly Przeworski
- Department of Human Genetics and Howard Hughes Medical Institute, University of Chicago, Chicago, Illinois 60637;
| |
Collapse
|
131
|
Nakabeppu Y. Cellular levels of 8-oxoguanine in either DNA or the nucleotide pool play pivotal roles in carcinogenesis and survival of cancer cells. Int J Mol Sci 2014; 15:12543-57. [PMID: 25029543 PMCID: PMC4139859 DOI: 10.3390/ijms150712543] [Citation(s) in RCA: 145] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Revised: 06/23/2014] [Accepted: 07/08/2014] [Indexed: 01/06/2023] Open
Abstract
8-Oxoguanine, a major oxidized base lesion formed by reactive oxygen species, causes G to T transversion mutations or leads to cell death in mammals if it accumulates in DNA. 8-Oxoguanine can originate as 8-oxo-dGTP, formed in the nucleotide pool, or by direct oxidation of the DNA guanine base. MTH1, also known as NUDT1, with 8-oxo-dGTP hydrolyzing activity, 8-oxoguanine DNA glycosylase (OGG1) an 8-oxoG DNA glycosylase, and MutY homolog (MUTYH) with adenine DNA glycosylase activity, minimize the accumulation of 8-oxoG in DNA; deficiencies in these enzymes increase spontaneous and induced tumorigenesis susceptibility. However, different tissue types have different tumorigenesis susceptibilities. These can be reversed by combined deficiencies in the defense systems, because cell death induced by accumulation of 8-oxoG in DNA is dependent on MUTYH, which can be suppressed by MTH1 and OGG1. In cancer cells encountering high oxidative stress levels, a high level of 8-oxo-dGTP accumulates in the nucleotide pool, and cells therefore express increased levels of MTH1 in order to eliminate 8-oxo-dGTP. Suppression of MTH1 may be an efficient strategy for killing cancer cells; however, because MTH1 and OGG1 protect normal tissues from oxidative-stress-induced cell death, it is important that MTH1 inhibition does not increase the risk of healthy tissue degeneration.
Collapse
Affiliation(s)
- Yusaku Nakabeppu
- Division of Neurofunctional Genomics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, and Research Center for Nucleotide Pool, Kyushu University, 3-1-1 Maidashi, Fukuoka 812-8582, Japan.
| |
Collapse
|