Obesity, DNA Damage, and Development of Obesity-Related Diseases
Abstract
:1. Introduction
2. Inflammation and Reactive Oxygen Species (ROS)-Induced DNA Damage
3. DNA Damage Repair
4. Obesity and DNA Damage
4.1. Obesity and DNA Damage Repair
4.2. Obesity and Mitochondrial DNA Damage
4.3. Effect of Parental Obesity-Related DNA Damage on Offspring
5. DNA Damage and Obesity-Related Metabolic Disorders
6. DNA Damage and Development of Obesity-Related Cancer
7. Conclusions
Author Contributions
Acknowledgments
Conflicts of Interest
Abbreviations
8-OHdG | 8-OH deoxyguanosine |
AGEs | advanced glycation end products |
APE | apurinic endonuclease |
ATM | ataxia telangiectasia–mutated protein |
BER | base excision repair |
BLM | bloom syndrome protein |
BMI | body mass index |
COX-2 | cyclooxygenase 2 |
CSA | cockayne syndrome protein A |
CSB | cockayne syndrome protein B |
DAPI | 4′,6-diamidino-2-phenylindole |
DDR | DNA damage response |
DFI | DNA fragmentation index |
DNA-PK | DNA-dependent kinase |
DR | direct repair |
DSB | double-strand breaks |
ELISA | enzyme-linked immunosorbent assay |
ENDS | electronic nicotine delivery devices |
EtBr | ethidium bromide |
EXO1 | human exonuclease 1 |
FANCF | fanconi anemia complementation group |
Fapy Ade | 4,6-diamino-5-formamidopyrimidine |
Fapy Gua | 2,6-diamino-4-hydroxy-5-formamidopyrimidine |
FEN1 | flap endonuclease 1 |
FFA | free fatty acids |
FISH | fluorescence in situ hybridization |
GC-MS | gas chromatography-mass spectrometry |
GGR | global genome repair |
H2O2 | hydrogen peroxide |
HPLC | high performance liquid chromatography |
HR | homologous recombination |
IL-6 | interleukin 6 |
LIG | DNA ligase |
MGMT | O6-methylguanine DNA methyltransferase |
MN | micronucleus assay |
MMR | mismatch repair |
NADPH | nicotinamide adenine dinucleotide phosphate |
NER | nucleotide excision repair |
NF-κB | nuclear factor κB |
NHEJ | non-homologous end-joining |
NO | nitric oxide |
NOS | nitric oxide synthase |
NOX4 | NADPH oxidase 4 |
OGG1 | 8-Oxoguanine glycosylase |
PAH | polycyclic aromatic hydrocarbons |
PARP | poly ADP-ribose polymerase 1 |
PCR | polymerase chain reaction |
PCNA | proliferating cell nuclear antigen |
PhIP | 2-amino-1-methyl-6-phenylimidazo(4,5-b)pyridine |
PTEN | phosphatase and tensin homolog |
ROS | reactive oxygen species |
RPA | replication protein A |
SCGE | Single cell gel electrophoresis |
SSB | single-strand breaks |
ssDNA | single-strand DNA |
SOD | superoxide dismutase |
TCR | transcription-coupled repair |
TFIIH | transcription factor complex IIH |
TNFα | tumor necrosis factor |
TUNEL | terminal deoxynucleotidyl transferase dUTP nick end labeling UV |
UV | ultraviolet |
WHO | World Health Organization |
WRN | Werner syndrome protein |
XP | xeroderma pigmentosium |
XRCC | X-ray repair cross-complementing group |
References
- Leung, Y.M.; Pollack, L.M.; Colditz, G.A.; Chang, S.H. Life years lost and lifetime health care expenditures associated with diabetes in the U.S., National Health Interview Survey, 1997–2000. Diabetes Care 2015, 38, 460–468. [Google Scholar] [CrossRef] [PubMed]
- Ligibel, J.A.; Alfano, C.M.; Courneya, K.S.; Demark-Wahnefried, W.; Burger, R.A.; Chlebowski, R.T.; Fabian, C.J.; Gucalp, A.; Hershman, D.L.; Hudson, M.M.; et al. American Society of Clinical Oncology position statement on obesity and cancer. J. Clin. Oncol. 2014, 32, 3568–3574. [Google Scholar] [CrossRef] [PubMed]
- Scherer, P.E.; Hill, J.A. Obesity, diabetes, and cardiovascular diseases: A compendium. Circ. Res. 2016, 118, 1703–1705. [Google Scholar] [CrossRef] [PubMed]
- Cerda, C.; Sanchez, C.; Climent, B.; Vazquez, A.; Iradi, A.; El Amrani, F.; Bediaga, A.; Saez, G.T. Oxidative stress and DNA damage in obesity-related tumorigenesis. Adv. Exp. Med. Biol. 2014, 824, 5–17. [Google Scholar] [PubMed]
- Wlodarczyk, M.; Jablonowska-Lietz, B.; Olejarz, W.; Nowicka, G. Anthropometric and dietary factors as predictors of DNA damage in obese women. Nutrients 2018, 10, 578. [Google Scholar] [CrossRef] [PubMed]
- Zaki, M.; Basha, W.; El-Bassyouni, H.T.; El-Toukhy, S.; Hussein, T. Evaluation of DNA damage profile in obese women and its association to risk of metabolic syndrome, polycystic ovary syndrome and recurrent preeclampsia. Genes Dis. 2018, 5, 367–373. [Google Scholar] [CrossRef] [PubMed]
- Irigaray, P.; Belpomme, D. Basic properties and molecular mechanisms of exogenous chemical carcinogens. Carcinogenesis 2010, 31, 135–148. [Google Scholar] [CrossRef] [PubMed]
- Heilbronn, L.K.; de Jonge, L.; Frisard, M.I.; DeLany, J.P.; Larson-Meyer, D.E.; Rood, J.; Nguyen, T.; Martin, C.K.; Volaufova, J.; Most, M.M.; et al. Effect of 6-month calorie restriction on biomarkers of longevity, metabolic adaptation, and oxidative stress in overweight individuals: A randomized controlled trial. JAMA 2006, 295, 1539–1548. [Google Scholar] [CrossRef] [PubMed]
- Chen, L.; Deng, H.; Cui, H.; Fang, J.; Zuo, Z.; Deng, J.; Li, Y.; Wang, X.; Zhao, L. Inflammatory responses and inflammation-associated diseases in organs. Oncotarget 2018, 9, 7204–7218. [Google Scholar] [CrossRef] [PubMed]
- Abou-Raya, S.; Abou-Raya, A.; Naim, A.; Abuelkheir, H. Chronic inflammatory autoimmune disorders and atherosclerosis. Ann. N. Y. Acad. Sci. 2007, 1107, 56–67. [Google Scholar] [CrossRef] [PubMed]
- Lopez-Candales, A.; Burgos, P.M.H.; Hernandez-Suarez, D.F.; Harris, D. Linking chronic inflammation with cardiovascular disease: from normal aging to the metabolic syndrome. J. Nat. Sci. 2017, 3, e341. [Google Scholar] [PubMed]
- Dan Dunn, J.; Alvarez, L.A.; Zhang, X.; Soldati, T. Reactive oxygen species and mitochondria: A nexus of cellular homeostasis. Redox Biol. 2015, 6, 472–485. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- De Bont, R.; van Larebeke, N. Endogenous DNA damage in humans: A review of quantitative data. Mutagenesis 2004, 19, 169–185. [Google Scholar] [CrossRef] [PubMed]
- Yu, Y.; Cui, Y.; Niedernhofer, L.J.; Wang, Y. Occurrence, biological consequences, and human health relevance of oxidative stress-induced DNA damage. Chem. Res. Toxicol. 2016, 29, 2008–2039. [Google Scholar] [CrossRef] [PubMed]
- Luczaj, W.; Skrzydlewska, E. DNA damage caused by lipid peroxidation products. Cell Mol. Biol. Lett. 2003, 8, 391–413. [Google Scholar] [PubMed]
- Jena, N.R. DNA damage by reactive species: Mechanisms, mutation and repair. J. Biosci. 2012, 37, 503–517. [Google Scholar] [CrossRef] [PubMed]
- Kehrer, J.P. The Haber-Weiss reaction and mechanisms of toxicity. Toxicology 2000, 149, 43–50. [Google Scholar] [CrossRef]
- Thomas, C.; Mackey, M.M.; Diaz, A.A.; Cox, D.P. Hydroxyl radical is produced via the Fenton reaction in submitochondrial particles under oxidative stress: Implications for diseases associated with iron accumulation. Redox Rep. 2009, 14, 102–108. [Google Scholar] [CrossRef] [PubMed]
- Dedon, P.C.; Tannenbaum, S.R. Reactive nitrogen species in the chemical biology of inflammation. Arch. Biochem. Biophys. 2004, 423, 12–22. [Google Scholar] [CrossRef] [PubMed]
- Kamiya, H. Mutagenic potentials of damaged nucleic acids produced by reactive oxygen/nitrogen species: Approaches using synthetic oligonucleotides and nucleotides: Survey and summary. Nucleic Acids Res. 2003, 31, 517–531. [Google Scholar] [CrossRef] [PubMed]
- Sova, H.; Jukkola-Vuorinen, A.; Puistola, U.; Kauppila, S.; Karihtala, P. 8-Hydroxydeoxyguanosine: A new potential independent prognostic factor in breast cancer. Br. J. Cancer 2010, 102, 1018–1023. [Google Scholar] [CrossRef] [PubMed]
- McGowan, C.H.; Russell, P. The DNA damage response: Sensing and signaling. Curr. Opin. Cell Biol. 2004, 16, 629–633. [Google Scholar] [CrossRef] [PubMed]
- Allione, A.; Guarrera, S.; Russo, A.; Ricceri, F.; Purohit, R.; Pagnani, A.; Rosa, F.; Polidoro, S.; Voglino, F.; Matullo, G. Inter-individual variation in nucleotide excision repair pathway is modulated by non-synonymous polymorphisms in ERCC4 and MBD4 genes. Mutat. Res. 2013, 751–752, 49–54. [Google Scholar] [CrossRef] [PubMed]
- Nagel, Z.D.; Chaim, I.A.; Samson, L.D. Inter-individual variation in DNA repair capacity: A need for multi-pathway functional assays to promote translational DNA repair research. DNA Repair 2014, 19, 199–213. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kabzinski, J.; Mucha, B.; Cuchra, M.; Markiewicz, L.; Przybylowska, K.; Dziki, A.; Dziki, L.; Majsterek, I. Efficiency of Base Excision Repair of Oxidative DNA Damage and Its Impact on the Risk of Colorectal Cancer in the Polish Population. Oxid Med. Cell Longev. 2016, 2016, 3125989. [Google Scholar] [CrossRef] [PubMed]
- Slyskova, J.; Lorenzo, Y.; Karlsen, A.; Carlsen, M.H.; Novosadova, V.; Blomhoff, R.; Vodicka, P.; Collins, A.R. Both genetic and dietary factors underlie individual differences in DNA damage levels and DNA repair capacity. DNA Repair 2014, 16, 66–73. [Google Scholar] [CrossRef] [PubMed]
- Hakem, R. DNA-damage repair; the good, the bad, and the ugly. EMBO J. 2008, 27, 589–605. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cleaver, J.E. Profile of Tomas Lindahl, Paul Modrich, and Aziz Sancar, 2015 Nobel Laureates in Chemistry. Proc. Natl. Acad. Sci. USA 2016, 113, 242–245. [Google Scholar] [CrossRef] [PubMed]
- Lindahl, T.; Karran, P.; Wood, R.D. DNA excision repair pathways. Curr. Opin. Genet. Dev. 1997, 7, 158–169. [Google Scholar] [CrossRef] [Green Version]
- Sancar, A. Excision repair in mammalian cells. J. Biol. Chem. 1995, 270, 15915–15918. [Google Scholar] [CrossRef] [PubMed]
- Bukhari, S.A.; Rajoka, M.I.; Ibrahim, Z.; Jalal, F.; Rana, S.M.; Nagra, S.A. Oxidative stress elevated DNA damage and homocysteine level in normal pregnant women in a segment of Pakistani population. Mol. Biol. Rep. 2011, 38, 2703–2710. [Google Scholar] [CrossRef] [PubMed]
- Tomasello, B.; Malfa, G.; Galvano, F.; Reins, M. DNA damage in normal-weight obese syndrome measured by Comet assay. Mediterr. J. Nutr. Metab. 2011, 2, 99–104. [Google Scholar] [CrossRef]
- Scarpato, R.; Verola, C.; Fabiani, B.; Bianchi, V.; Saggese, G.; Federico, G. Nuclear damage in peripheral lymphocytes of obese and overweight Italian children as evaluated by the gamma-H2AX focus assay and micronucleus test. FASEB J. 2011, 25, 685–693. [Google Scholar] [CrossRef] [PubMed]
- Azzara, A.; Pirillo, C.; Giovannini, C.; Federico, G.; Scarpato, R. Different repair kinetic of DSBs induced by mitomycin C in peripheral lymphocytes of obese and normal weight adolescents. Mutat. Res. 2016, 789, 9–14. [Google Scholar] [CrossRef] [PubMed]
- Donmez-Altuntas, H.; Sahin, F.; Bayram, F.; Bitgen, N.; Mert, M.; Guclu, K.; Hamurcu, Z.; Aribas, S.; Gundogan, K.; Diri, H. Evaluation of chromosomal damage, cytostasis, cytotoxicity, oxidative DNA damage and their association with body-mass index in obese subjects. Mutat. Res. Genet. Toxicol. Environ. Mutagen. 2014, 771, 30–36. [Google Scholar] [CrossRef] [PubMed]
- Karbownik-Lewinska, M.; Szosland, J.; Kokoszko-Bilska, A.; Stepniak, J.; Zasada, K.; Gesing, A.; Lewinski, A. Direct contribution of obesity to oxidative damage to macromolecules. Neuro Endocrinol. Lett. 2012, 33, 453–461. [Google Scholar] [PubMed]
- Kocael, A.; Erman, H.; Zengin, K.; Kocael, P.C.; Korkmaz, G.G.; Gelisgen, R.; Taskin, M.; Ersan, Y.; Uzun, H. The effects on oxidative DNA damage of laparoscopic gastric band applications in morbidly obese patients. Can. J. Surg. 2014, 57, 183–187. [Google Scholar] [CrossRef] [PubMed]
- Setayesh, T.; Nersesyan, A.; Misik, M.; Ferk, F.; Langie, S.; Andrade, V.M.; Haslberger, A.; Knasmuller, S. Impact of obesity and overweight on DNA stability: Few facts and many hypotheses. Mutat. Res. 2018, 777, 64–91. [Google Scholar] [CrossRef] [PubMed]
- Iyer, A.; Fairlie, D.P.; Prins, J.B.; Hammock, B.D.; Brown, L. Inflammatory lipid mediators in adipocyte function and obesity. Nat. Rev. Endocrinol. 2010, 6, 71–82. [Google Scholar] [CrossRef] [PubMed]
- Han, C.Y. Roles of Reactive Oxygen Species on Insulin Resistance in Adipose Tissue. Diabetes Metab. J. 2016, 40, 272–279. [Google Scholar] [CrossRef] [PubMed]
- Han, C.Y.; Umemoto, T.; Omer, M.; den Hartigh, L.J.; Chiba, T.; LeBoeuf, R.; Buller, C.L.; Sweet, I.R.; Pennathur, S.; Abel, E.D.; et al. NADPH oxidase-derived reactive oxygen species increases expression of monocyte chemotactic factor genes in cultured adipocytes. J. Biol. Chem. 2012, 287, 10379–10393. [Google Scholar] [CrossRef] [PubMed]
- Weisberg, S.P.; McCann, D.; Desai, M.; Rosenbaum, M.; Leibel, R.L.; Ferrante, A.W., Jr. Obesity is associated with macrophage accumulation in adipose tissue. J. Clin. Investig. 2003, 112, 1796–1808. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gao, C.L.; Zhu, C.; Zhao, Y.P.; Chen, X.H.; Ji, C.B.; Zhang, C.M.; Zhu, J.G.; Xia, Z.K.; Tong, M.L.; Guo, X.R. Mitochondrial dysfunction is induced by high levels of glucose and free fatty acids in 3T3-L1 adipocytes. Mol. Cell Endocrinol. 2010, 320, 25–33. [Google Scholar] [CrossRef] [PubMed]
- Heo, J.W.; No, M.H.; Park, D.H.; Kang, J.H.; Seo, D.Y.; Han, J.; Neufer, P.D.; Kwak, H.B. Effects of exercise on obesity-induced mitochondrial dysfunction in skeletal muscle. Korean J. Physiol. Pharmacol. 2017, 21, 567–577. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fehsel, K.; Kolb-Bachofen, V.; Kolb, H. Analysis of TNF alpha-induced DNA strand breaks at the single cell level. Am. J. Pathol. 1991, 139, 251–254. [Google Scholar] [PubMed]
- Arango Duque, G.; Descoteaux, A. Macrophage cytokines: Involvement in immunity and infectious diseases. Front. Immunol. 2014, 5, 491. [Google Scholar] [CrossRef] [PubMed]
- Rastogi, S.; Boylan, M.; Wright, E.G.; Coates, P.J. Interactions of apoptotic cells with macrophages in radiation-induced bystander signaling. Radiat. Res. 2013, 179, 135–145. [Google Scholar] [CrossRef] [PubMed]
- Speed, N.; Blair, I.A. Cyclooxygenase- and lipoxygenase-mediated DNA damage. Cancer Metastasis Rev. 2011, 30, 437–447. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Faber, T.J.; Japink, D.; Leers, M.P.; Sosef, M.N.; von Meyenfeldt, M.F.; Nap, M. Activated macrophages containing tumor marker in colon carcinoma: Immunohistochemical proof of a concept. Tumour Biol. 2012, 33, 435–441. [Google Scholar] [CrossRef] [PubMed]
- O’Callaghan, N.J.; Clifton, P.M.; Noakes, M.; Fenech, M. Weight loss in obese men is associated with increased telomere length and decreased abasic sites in rectal mucosa. Rejuvenation Res. 2009, 12, 169–176. [Google Scholar] [CrossRef] [PubMed]
- Fejfer, K.; Buczko, P.; Niczyporuk, M.; Ladny, J.R.; Hady, H.R.; Knas, M.; Waszkiel, D.; Klimiuk, A.; Zalewska, A.; Maciejczyk, M. Oxidative modification of biomolecules in the nonstimulated and stimulated saliva of patients with morbid obesity treated with bariatric surgery. Biomed. Res. Int. 2017, 2017, 4923769. [Google Scholar] [CrossRef] [PubMed]
- Himbert, C.; Thompson, H.; Ulrich, C.M. Effects of intentional weight loss on markers of oxidative stress, DNA repair and telomere length—A systematic review. Obes. Facts 2017, 10, 648–665. [Google Scholar] [CrossRef] [PubMed]
- Tyson, J.; Caple, F.; Spiers, A.; Burtle, B.; Daly, A.K.; Williams, E.A.; Hesketh, J.E.; Mathers, J.C. Inter-individual variation in nucleotide excision repair in young adults: Effects of age, adiposity, micronutrient supplementation and genotype. Br. J. Nutr. 2009, 101, 1316–1323. [Google Scholar] [CrossRef] [PubMed]
- Adcock, I.M.; Cosio, B.; Tsaprouni, L.; Barnes, P.J.; Ito, K. Redox regulation of histone deacetylases and glucocorticoid-mediated inhibition of the inflammatory response. Antioxid. Redox Signal. 2005, 7, 144–152. [Google Scholar] [CrossRef] [PubMed]
- Kidane, D.; Chae, W.J.; Czochor, J.; Eckert, K.A.; Glazer, P.M.; Bothwell, A.L.; Sweasy, J.B. Interplay between DNA repair and inflammation, and the link to cancer. Crit. Rev. Biochem. Mol. Biol. 2014, 49, 116–139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, R.H.; Hotchkiss, J.H. Potential genotoxicity of chronically elevated nitric oxide: A review. Mutat. Res. 1995, 339, 73–89. [Google Scholar] [CrossRef]
- McAdam, E.; Brem, R.; Karran, P. Oxidative stress-induced protein damage inhibits DNA repair and determines mutation risk and therapeutic efficacy. Mol. Cancer Res. 2016, 14, 612–622. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nikodemova, M.; Yee, J.; Carney, P.R.; Bradfield, C.A.; Malecki, K.M. Transcriptional differences between smokers and non-smokers and variance by obesity as a risk factor for human sensitivity to environmental exposures. Environ. Int. 2018, 113, 249–258. [Google Scholar] [CrossRef] [PubMed]
- Zwamborn, R.A.; Slieker, R.C.; Mulder, P.C.; Zoetemelk, I.; Verschuren, L.; Suchiman, H.E.; Toet, K.H.; Droog, S.; Slagboom, P.E.; Kooistra, T.; et al. Prolonged high-fat diet induces gradual and fat depot-specific DNA methylation changes in adult mice. Sci. Rep. 2017, 7, 43261. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Keleher, M.R.; Zaidi, R.; Hicks, L.; Shah, S.; Xing, X.; Li, D.; Wang, T.; Cheverud, J.M. A high-fat diet alters genome-wide DNA methylation and gene expression in SM/J mice. BMC Genom. 2018, 19, 888. [Google Scholar] [CrossRef] [PubMed]
- Wang, D.; Zhao, R.; Qu, Y.Y.; Mei, X.Y.; Zhang, X.; Zhou, Q.; Li, Y.; Yang, S.B.; Zuo, Z.G.; Chen, Y.M.; et al. Colonic lysine homocysteinylation induced by high-fat diet suppresses DNA damage repair. Cell Rep. 2018, 25, 398–412. [Google Scholar] [CrossRef] [PubMed]
- Harreus, U.; Baumeister, P.; Zieger, S.; Matthias, C. The influence of high doses of vitamin C and zinc on oxidative DNA damage. Anticancer Res. 2005, 25, 3197–3201. [Google Scholar] [PubMed]
- Huang, H.Y.; Helzlsouer, K.J.; Appel, L.J. The effects of vitamin C and vitamin E on oxidative DNA damage: Results from a randomized controlled trial. Cancer Epidemiol. Biomark. Prev. 2000, 9, 647–652. [Google Scholar]
- Remely, M.; Ferk, F.; Sterneder, S.; Setayesh, T.; Roth, S.; Kepcija, T.; Noorizadeh, R.; Rebhan, I.; Greunz, M.; Beckmann, J.; et al. EGCG prevents high fat diet-induced changes in gut microbiota, decreases of DNA strand breaks, and changes in expression and DNA methylation of Dnmt1 and MLH1 in C57BL/6J male mice. Oxid Med. Cell Longev. 2017, 2017, 3079148. [Google Scholar] [CrossRef] [PubMed]
- Remely, M.; Ferk, F.; Sterneder, S.; Setayesh, T.; Kepcija, T.; Roth, S.; Noorizadeh, R.; Greunz, M.; Rebhan, I.; Wagner, K.H.; et al. Vitamin E modifies high-fat diet-induced increase of DNA strand breaks, and changes in expression and DNA methylation of Dnmt1 and MLH1 in C57BL/6J male mice. Nutrients 2017, 9, 607. [Google Scholar] [CrossRef] [PubMed]
- Van Houten, B.; Hunter, S.E.; Meyer, J.N. Mitochondrial DNA damage induced autophagy, cell death, and disease. Front. Biosci. 2016, 21, 42–54. [Google Scholar] [CrossRef]
- Stein, A.; Sia, E.A. Mitochondrial DNA repair and damage tolerance. Front. Biosci. 2017, 22, 920–943. [Google Scholar]
- Taanman, J.W. The mitochondrial genome: Structure, transcription, translation and replication. Biochim. Biophys. Acta 1999, 1410, 103–123. [Google Scholar] [CrossRef]
- Wang, J.; Xiong, S.; Xie, C.; Markesbery, W.R.; Lovell, M.A. Increased oxidative damage in nuclear and mitochondrial DNA in Alzheimer’s disease. J. Neurochem. 2005, 93, 953–962. [Google Scholar] [CrossRef] [PubMed]
- Nishikawa, M.; Oshitani, N.; Matsumoto, T.; Nishigami, T.; Arakawa, T.; Inoue, M. Accumulation of mitochondrial DNA mutation with colorectal carcinogenesis in ulcerative colitis. Br. J. Cancer 2005, 93, 331–337. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bensch, K.G.; Mott, J.L.; Chang, S.W.; Hansen, P.A.; Moxley, M.A.; Chambers, K.T.; de Graaf, W.; Zassenhaus, H.P.; Corbett, J.A. Selective mtDNA mutation accumulation results in beta-cell apoptosis and diabetes development. Am. J. Physiol. Endocrinol. Metab. 2009, 296, E672–E680. [Google Scholar] [CrossRef] [PubMed]
- Chinnery, P.F.; Hudson, G. Mitochondrial genetics. Br. Med. Bull. 2013, 106, 135–159. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sutherland, L.N.; Capozzi, L.C.; Turchinsky, N.J.; Bell, R.C.; Wright, D.C. Time course of high-fat diet-induced reductions in adipose tissue mitochondrial proteins: Potential mechanisms and the relationship to glucose intolerance. Am. J. Physiol. Endocrinol. Metab. 2008, 295, E1076–E1083. [Google Scholar] [CrossRef] [PubMed]
- Turner, N.; Bruce, C.R.; Beale, S.M.; Hoehn, K.L.; So, T.; Rolph, M.S.; Cooney, G.J. Excess lipid availability increases mitochondrial fatty acid oxidative capacity in muscle: Evidence against a role for reduced fatty acid oxidation in lipid-induced insulin resistance in rodents. Diabetes 2007, 56, 2085–2092. [Google Scholar] [CrossRef] [PubMed]
- Lee, H.; Oh, S.; Yang, W.; Park, R.; Kim, H.; Jeon, J.S.; Noh, H.; Han, D.C.; Cho, K.W.; Kim, Y.J.; et al. Bariatric surgery reduces elevated urinary mitochondrial DNA copy number in obese patients. J. Clin. Endocrinol. Metab. 2019. [Google Scholar] [CrossRef] [PubMed]
- Yuzefovych, L.V.; Musiyenko, S.I.; Wilson, G.L.; Rachek, L.I. Mitochondrial DNA damage and dysfunction, and oxidative stress are associated with endoplasmic reticulum stress, protein degradation and apoptosis in high fat diet-induced insulin resistance mice. PLoS ONE 2013, 8, e54059. [Google Scholar] [CrossRef] [PubMed]
- Pazmandi, K.; Agod, Z.; Kumar, B.V.; Szabo, A.; Fekete, T.; Sogor, V.; Veres, A.; Boldogh, I.; Rajnavolgyi, E.; Lanyi, A.; et al. Oxidative modification enhances the immunostimulatory effects of extracellular mitochondrial DNA on plasmacytoid dendritic cells. Free Radic. Biol. Med. 2014, 77, 281–290. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shimada, K.; Crother, T.R.; Karlin, J.; Dagvadorj, J.; Chiba, N.; Chen, S.; Ramanujan, V.K.; Wolf, A.J.; Vergnes, L.; Ojcius, D.M.; et al. Oxidized mitochondrial DNA activates the NLRP3 inflammasome during apoptosis. Immunity 2012, 36, 401–414. [Google Scholar] [CrossRef] [PubMed]
- Patro, B.; Liber, A.; Zalewski, B.; Poston, L.; Szajewska, H.; Koletzko, B. Maternal and paternal body mass index and offspring obesity: A systematic review. Ann. Nutr. Metab. 2013, 63, 32–41. [Google Scholar] [CrossRef] [PubMed]
- Wahl, S.; Drong, A.; Lehne, B.; Loh, M.; Scott, W.R.; Kunze, S.; Tsai, P.C.; Ried, J.S.; Zhang, W.; Yang, Y.; et al. Epigenome-wide association study of body mass index, and the adverse outcomes of adiposity. Nature 2017, 541, 81–86. [Google Scholar] [CrossRef] [PubMed]
- Campbell, J.M.; Lane, M.; Owens, J.A.; Bakos, H.W. Paternal obesity negatively affects male fertility and assisted reproduction outcomes: A systematic review and meta-analysis. Reprod. Biomed. Online 2015, 31, 593–604. [Google Scholar] [CrossRef] [PubMed]
- Kort, H.I.; Massey, J.B.; Elsner, C.W.; Mitchell-Leef, D.; Shapiro, D.B.; Witt, M.A.; Roudebush, W.E. Impact of body mass index values on sperm quantity and quality. J. Androl. 2006, 27, 450–452. [Google Scholar] [CrossRef] [PubMed]
- Evenson, D.P.; Wixon, R. Clinical aspects of sperm DNA fragmentation detection and male infertility. Theriogenology 2006, 65, 979–991. [Google Scholar] [CrossRef] [PubMed]
- Abdelbaki, S.A.; Sabry, J.H.; Al-Adl, A.M.; Sabry, H.H. The impact of coexisting sperm DNA fragmentation and seminal oxidative stress on the outcome of varicocelectomy in infertile patients: A prospective controlled study. Arab. J. Urol. 2017, 15, 131–139. [Google Scholar] [CrossRef] [PubMed]
- Tunc, O.; Bakos, H.W.; Tremellen, K. Impact of body mass index on seminal oxidative stress. Andrologia 2011, 43, 121–128. [Google Scholar] [CrossRef] [PubMed]
- Lezaja, A.; Altmeyer, M. Inherited DNA lesions determine G1 duration in the next cell cycle. Cell Cycle 2018, 17, 24–32. [Google Scholar] [CrossRef] [PubMed]
- Soubry, A. Epigenetic inheritance and evolution: A paternal perspective on dietary influences. Prog. Biophys. Mol. Biol. 2015, 118, 79–85. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hammoud, S.S.; Nix, D.A.; Hammoud, A.O.; Gibson, M.; Cairns, B.R.; Carrell, D.T. Genome-wide analysis identifies changes in histone retention and epigenetic modifications at developmental and imprinted gene loci in the sperm of infertile men. Hum. Reprod. 2011, 26, 2558–2569. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- McPherson, N.O.; Fullston, T.; Aitken, R.J.; Lane, M. Paternal obesity, interventions, and mechanistic pathways to impaired health in offspring. Ann. Nutr. Metab. 2014, 64, 231–238. [Google Scholar] [CrossRef] [PubMed]
- Noblanc, A.; Damon-Soubeyrand, C.; Karrich, B.; Henry-Berger, J.; Cadet, R.; Saez, F.; Guiton, R.; Janny, L.; Pons-Rejraji, H.; Alvarez, J.G.; et al. DNA oxidative damage in mammalian spermatozoa: Where and why is the male nucleus affected? Free Radic. Biol. Med. 2013, 65, 719–723. [Google Scholar] [CrossRef] [PubMed]
- Palmer, N.O.; Fullston, T.; Mitchell, M.; Setchell, B.P.; Lane, M. SIRT6 in mouse spermatogenesis is modulated by diet-induced obesity. Reprod. Fertil. Dev. 2011, 23, 929–939. [Google Scholar] [CrossRef] [PubMed]
- Van Gaal, L.F.; Mertens, I.L.; de Block, C.E. Mechanisms linking obesity with cardiovascular disease. Nature 2006, 444, 875–880. [Google Scholar] [CrossRef] [PubMed]
- Shimizu, I.; Yoshida, Y.; Suda, M.; Minamino, T. DNA damage response and metabolic disease. Cell Metab. 2014, 20, 967–977. [Google Scholar] [CrossRef] [PubMed]
- Vousden, K.H.; Lane, D.P. p53 in health and disease. Nat. Rev. Mol. Cell Biol. 2007, 8, 275–283. [Google Scholar] [CrossRef] [PubMed]
- Maier, B.; Gluba, W.; Bernier, B.; Turner, T.; Mohammad, K.; Guise, T.; Sutherland, A.; Thorner, M.; Scrable, H. Modulation of mammalian life span by the short isoform of p53. Genes Dev. 2004, 18, 306–319. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tyner, S.D.; Venkatachalam, S.; Choi, J.; Jones, S.; Ghebranious, N.; Igelmann, H.; Lu, X.; Soron, G.; Cooper, B.; Brayton, C.; et al. p53 mutant mice that display early ageing-associated phenotypes. Nature 2002, 415, 45–53. [Google Scholar] [CrossRef] [PubMed]
- Petitjean, A.; Mathe, E.; Kato, S.; Ishioka, C.; Tavtigian, S.V.; Hainaut, P.; Olivier, M. Impact of mutant p53 functional properties on TP53 mutation patterns and tumor phenotype: Lessons from recent developments in the IARC TP53 database. Hum. Mutat. 2007, 28, 622–629. [Google Scholar] [CrossRef] [PubMed]
- Ochs-Balcom, H.M.; Marian, C.; Nie, J.; Brasky, T.M.; Goerlitz, D.S.; Trevisan, M.; Edge, S.B.; Winston, J.; Berry, D.L.; Kallakury, B.V.; et al. Adiposity is associated with p53 gene mutations in breast cancer. Breast Cancer Res. Treat. 2015, 153, 635–645. [Google Scholar] [CrossRef] [PubMed]
- Minamino, T.; Orimo, M.; Shimizu, I.; Kunieda, T.; Yokoyama, M.; Ito, T.; Nojima, A.; Nabetani, A.; Oike, Y.; Matsubara, H.; et al. A crucial role for adipose tissue p53 in the regulation of insulin resistance. Nat. Med. 2009, 15, 1082–1087. [Google Scholar] [CrossRef] [PubMed]
- Vergoni, B.; Cornejo, P.J.; Gilleron, J.; Djedaini, M.; Ceppo, F.; Jacquel, A.; Bouget, G.; Ginet, C.; Gonzalez, T.; Maillet, J.; et al. DNA Damage and the Activation of the p53 Pathway Mediate Alterations in Metabolic and Secretory Functions of Adipocytes. Diabetes 2016, 65, 3062–3074. [Google Scholar] [CrossRef] [PubMed]
- Hinault, C.; Kawamori, D.; Liew, C.W.; Maier, B.; Hu, J.; Keller, S.R.; Mirmira, R.G.; Scrable, H.; Kulkarni, R.N. Delta40 Isoform of p53 controls beta-cell proliferation and glucose homeostasis in mice. Diabetes 2011, 60, 1210–1222. [Google Scholar] [CrossRef] [PubMed]
- Tavana, O.; Zhu, C. Too many breaks (brakes): Pancreatic beta-cell senescence leads to diabetes. Cell Cycle 2011, 10, 2471–2484. [Google Scholar] [CrossRef] [PubMed]
- Castro, A.V.; Kolka, C.M.; Kim, S.P.; Bergman, R.N. Obesity, insulin resistance and comorbidities? Mechanisms of association. Arq. Bras. Endocrinol. Metabol. 2014, 58, 600–609. [Google Scholar] [CrossRef] [PubMed]
- Al-Aubaidy, H.A.; Jelinek, H.F. Oxidative DNA damage and obesity in type 2 diabetes mellitus. Eur. J. Endocrinol. 2011, 164, 899–904. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, S.C.; Chan, J.C. Evidence for DNA damage as a biological link between diabetes and cancer. Chin. Med. J. 2015, 128, 1543–1548. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Y.; Zhou, J.; Wang, T.; Cai, L. High level glucose increases mutagenesis in human lymphoblastoid cells. Int. J. Biol. Sci. 2007, 3, 375–379. [Google Scholar] [CrossRef] [PubMed]
- Stopper, H.; Schinzel, R.; Sebekova, K.; Heidland, A. Genotoxicity of advanced glycation end products in mammalian cells. Cancer Lett. 2003, 190, 151–156. [Google Scholar] [CrossRef]
- Fukami, K.; Yamagishi, S.; Kaifu, K.; Matsui, T.; Kaida, Y.; Ueda, S.; Takeuchi, M.; Asanuma, K.; Okuda, S. Telmisartan inhibits AGE-induced podocyte damage and detachment. Microvasc. Res. 2013, 88, 79–83. [Google Scholar] [CrossRef] [PubMed]
- Simone, S.; Gorin, Y.; Velagapudi, C.; Abboud, H.E.; Habib, S.L. Mechanism of oxidative DNA damage in diabetes: Tuberin inactivation and downregulation of DNA repair enzyme 8-oxo-7,8-dihydro-2′-deoxyguanosine-DNA glycosylase. Diabetes 2008, 57, 2626–2636. [Google Scholar] [CrossRef] [PubMed]
- Habib, S.L.; Liang, S. Hyperactivation of Akt/mTOR and deficiency in tuberin increased the oxidative DNA damage in kidney cancer patients with diabetes. Oncotarget 2014, 5, 2542–2550. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xu, N.; Lao, Y.; Zhang, Y.; Gillespie, D.A. Akt: A double-edged sword in cell proliferation and genome stability. J. Oncol. 2012, 2012, 951724. [Google Scholar] [CrossRef] [PubMed]
- Habib, S.L.; Phan, M.N.; Patel, S.K.; Li, D.; Monks, T.J.; Lau, S.S. Reduced constitutive 8-oxoguanine-DNA glycosylase expression and impaired induction following oxidative DNA damage in the tuberin deficient Eker rat. Carcinogenesis 2003, 24, 573–582. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Habib, S.L. Molecular mechanism of regulation of OGG1: Tuberin deficiency results in cytoplasmic redistribution of transcriptional factor NF-YA. J. Mol. Signal. 2009, 4, 8. [Google Scholar] [CrossRef] [PubMed]
- Piscitello, D.; Varshney, D.; Lilla, S.; Vizioli, M.G.; Reid, C.; Gorbunova, V.; Seluanov, A.; Gillespie, D.A.; Adams, P.D. AKT overactivation can suppress DNA repair via p70S6 kinase-dependent downregulation of MRE11. Oncogene 2018, 37, 427–438. [Google Scholar] [CrossRef] [PubMed]
- Merkel, P.; Khoury, N.; Bertolotto, C.; Perfetti, R. Insulin and glucose regulate the expression of the DNA repair enzyme XPD. Mol. Cell Endocrinol. 2003, 201, 75–85. [Google Scholar] [CrossRef]
- Komakula, S.S.B.; Tumova, J.; Kumaraswamy, D.; Burchat, N.; Vartanian, V.; Ye, H.; Dobrzyn, A.; Lloyd, R.S.; Sampath, H. The DNA Repair Protein OGG1 Protects Against Obesity by Altering Mitochondrial Energetics in White Adipose Tissue. Sci. Rep. 2018, 8, 14886. [Google Scholar] [CrossRef] [PubMed]
- Dizdaroglu, M. Substrate specificities and excision kinetics of DNA glycosylases involved in base-excision repair of oxidative DNA damage. Mutat. Res. 2003, 531, 109–126. [Google Scholar] [CrossRef] [PubMed]
- Dou, H.; Mitra, S.; Hazra, T.K. Repair of oxidized bases in DNA bubble structures by human DNA glycosylases NEIL1 and NEIL2. J. Biol. Chem. 2003, 278, 49679–49684. [Google Scholar] [CrossRef] [PubMed]
- Vartanian, V.; Lowell, B.; Minko, I.G.; Wood, T.G.; Ceci, J.D.; George, S.; Ballinger, S.W.; Corless, C.L.; McCullough, A.K.; Lloyd, R.S. The metabolic syndrome resulting from a knockout of the NEIL1 DNA glycosylase. Proc. Natl. Acad. Sci. USA 2006, 103, 1864–1869. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sampath, H.; Vartanian, V.; Rollins, M.R.; Sakumi, K.; Nakabeppu, Y.; Lloyd, R.S. 8-Oxoguanine DNA glycosylase (OGG1) deficiency increases susceptibility to obesity and metabolic dysfunction. PLoS ONE 2012, 7, e51697. [Google Scholar] [CrossRef] [PubMed]
- Bankoglu, E.E.; Arnold, C.; Hering, I.; Hankir, M.; Seyfried, F.; Stopper, H. Decreased Chromosomal Damage in Lymphocytes of Obese Patients After Bariatric Surgery. Sci. Rep. 2018, 8, 11195. [Google Scholar] [CrossRef] [PubMed]
- Gavande, N.S.; VanderVere-Carozza, P.S.; Hinshaw, H.D.; Jalal, S.I.; Sears, C.R.; Pawelczak, K.S.; Turchi, J.J. DNA repair targeted therapy: The past or future of cancer treatment? Pharmacol. Ther. 2016, 160, 65–83. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Turgeon, M.O.; Perry, N.J.S.; Poulogiannis, G. DNA Damage, Repair, and Cancer Metabolism. Front. Oncol. 2018, 8, 15. [Google Scholar] [CrossRef] [PubMed]
- Lord, C.J.; Ashworth, A. BRCAness revisited. Nat. Rev. Cancer 2016, 16, 110–120. [Google Scholar] [CrossRef] [PubMed]
- Chae, Y.K.; Anker, J.F.; Carneiro, B.A.; Chandra, S.; Kaplan, J.; Kalyan, A.; Santa-Maria, C.A.; Platanias, L.C.; Giles, F.J. Genomic landscape of DNA repair genes in cancer. Oncotarget 2016, 7, 23312–23321. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chan, D.S.; Vieira, A.R.; Aune, D.; Bandera, E.V.; Greenwood, D.C.; McTiernan, A.; Rosenblatt, D.N.; Thune, I.; Vieira, R.; Norat, T. Body mass index and survival in women with breast cancer-systematic literature review and meta-analysis of 82 follow-up studies. Ann. Oncol. 2014, 25, 1901–1914. [Google Scholar] [CrossRef] [PubMed]
- Kocarnik, J.M.; Chan, A.T.; Slattery, M.L.; Potter, J.D.; Meyerhardt, J.; Phipps, A.; Nan, H.; Harrison, T.; Rohan, T.E.; Qi, L.; et al. Relationship of prediagnostic body mass index with survival after colorectal cancer: Stage-specific associations. Int. J. Cancer 2016, 139, 1065–1072. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nimptsch, K.; Pischon, T. Body fatness, related biomarkers and cancer risk: An epidemiological perspective. Horm. Mol. Biol. Clin. Investig. 2015, 22, 39–51. [Google Scholar] [CrossRef] [PubMed]
- Pischon, T.; Nimptsch, K. Obesity and Risk of Cancer: An Introductory Overview. Recent Results Cancer Res. 2016, 208, 1–15. [Google Scholar] [PubMed]
- Secord, A.A.; Hasselblad, V.; von Gruenigen, V.E.; Gehrig, P.A.; Modesitt, S.C.; Bae-Jump, V.; Havrilesky, L.J. Body mass index and mortality in endometrial cancer: A systematic review and meta-analysis. Gynecol. Oncol. 2016, 140, 184–190. [Google Scholar] [CrossRef] [PubMed]
- The, L. The link between cancer and obesity. Lancet 2017, 390, 1716. [Google Scholar]
- Ning, Y.; Wang, L.; Giovannucci, E.L. A quantitative analysis of body mass index and colorectal cancer: Findings from 56 observational studies. Obes. Rev. 2010, 11, 19–30. [Google Scholar] [CrossRef] [PubMed]
- Colotta, F.; Allavena, P.; Sica, A.; Garlanda, C.; Mantovani, A. Cancer-related inflammation, the seventh hallmark of cancer: Links to genetic instability. Carcinogenesis 2009, 30, 1073–1081. [Google Scholar] [CrossRef] [PubMed]
- Fernandez-Sanchez, A.; Madrigal-Santillan, E.; Bautista, M.; Esquivel-Soto, J.; Morales-Gonzalez, A.; Esquivel-Chirino, C.; Durante-Montiel, I.; Sanchez-Rivera, G.; Valadez-Vega, C.; Morales-Gonzalez, J.A. Inflammation, oxidative stress, and obesity. Int. J. Mol. Sci. 2011, 12, 3117–3132. [Google Scholar] [CrossRef] [PubMed]
- Jung, U.J.; Choi, M.S. Obesity and its metabolic complications: The role of adipokines and the relationship between obesity, inflammation, insulin resistance, dyslipidemia and nonalcoholic fatty liver disease. Int. J. Mol. Sci. 2014, 15, 6184–6223. [Google Scholar] [CrossRef] [PubMed]
- Sun, B.; Karin, M. Obesity, inflammation, and liver cancer. J. Hepatol. 2012, 56, 704–713. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ramos-Nino, M.E. The role of chronic inflammation in obesity-associated cancers. ISRN Oncol. 2013, 2013, 697521. [Google Scholar] [CrossRef] [PubMed]
- Riondino, S.; Roselli, M.; Palmirotta, R.; Della-Morte, D.; Ferroni, P.; Guadagni, F. Obesity and colorectal cancer: Role of adipokines in tumor initiation and progression. World J. Gastroenterol. 2014, 20, 5177–5190. [Google Scholar] [CrossRef] [PubMed]
- Otani, K.; Ishihara, S.; Yamaguchi, H.; Murono, K.; Yasuda, K.; Nishikawa, T.; Tanaka, T.; Kiyomatsu, T.; Hata, K.; Kawai, K.; et al. Adiponectin and colorectal cancer. Surg. Today 2017, 47, 151–158. [Google Scholar] [CrossRef] [PubMed]
- Barb, D.; Pazaitou-Panayiotou, K.; Mantzoros, C.S. Adiponectin: A link between obesity and cancer. Expert Opin. Investig. Drugs 2006, 15, 917–931. [Google Scholar] [CrossRef] [PubMed]
- Stattin, P.; Lukanova, A.; Biessy, C.; Soderberg, S.; Palmqvist, R.; Kaaks, R.; Olsson, T.; Jellum, E. Obesity and colon cancer: Does leptin provide a link? Int. J. Cancer 2004, 109, 149–152. [Google Scholar] [CrossRef] [PubMed]
- Stattin, P.; Palmqvist, R.; Soderberg, S.; Biessy, C.; Ardnor, B.; Hallmans, G.; Kaaks, R.; Olsson, T. Plasma leptin and colorectal cancer risk: A prospective study in Northern Sweden. Oncol. Rep. 2003, 10, 2015–2021. [Google Scholar] [CrossRef] [PubMed]
- Orecchioni, S.; Reggiani, F.; Talarico, G.; Bertolini, F. Mechanisms of obesity in the development of breast cancer. Discov. Med. 2015, 20, 121–128. [Google Scholar] [PubMed]
- Karin, M.; Greten, F.R. NF-kappaB: Linking inflammation and immunity to cancer development and progression. Nat. Rev. Immunol. 2005, 5, 749–759. [Google Scholar] [CrossRef] [PubMed]
- McCool, K.W.; Miyamoto, S. DNA damage-dependent NF-kappaB activation: NEMO turns nuclear signaling inside out. Immunol. Rev. 2012, 246, 311–326. [Google Scholar] [CrossRef] [PubMed]
- Wang, W.; Mani, A.M.; Wu, Z.H. DNA damage-induced nuclear factor-kappa B activation and its roles in cancer progression. J. Cancer Metastasis Treat. 2017, 3, 45–59. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Valentino, E.; Bellazzo, A.; di Minin, G.; Sicari, D.; Apollonio, M.; Scognamiglio, G.; di Bonito, M.; Botti, G.; del Sal, G.; Collavin, L. Mutant p53 potentiates the oncogenic effects of insulin by inhibiting the tumor suppressor DAB2IP. Proc. Natl. Acad. Sci. USA 2017, 114, 7623–7628. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [PubMed]
- Macheda, M.L.; Rogers, S.; Best, J.D. Molecular and cellular regulation of glucose transporter (GLUT) proteins in cancer. J. Cell Physiol. 2005, 202, 654–662. [Google Scholar] [CrossRef] [PubMed]
- Koppenol, W.H.; Bounds, P.L.; Dang, C.V. Otto Warburg’s contributions to current concepts of cancer metabolism. Nat. Rev. Cancer 2011, 11, 325–337. [Google Scholar] [CrossRef] [PubMed]
- Levine, A.J.; Puzio-Kuter, A.M. The control of the metabolic switch in cancers by oncogenes and tumor suppressor genes. Science 2010, 330, 1340–1344. [Google Scholar] [CrossRef] [PubMed]
- Rose, D.P.; Gracheck, P.J.; Vona-Davis, L. The Interactions of Obesity, Inflammation and Insulin Resistance in Breast Cancer. Cancers 2015, 7, 2147–2168. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Marnett, L.J. Chemistry and biology of DNA damage by malondialdehyde. IARC Sci. Publ. 1999, 150, 17–27. [Google Scholar]
- Voulgaridou, G.P.; Anestopoulos, I.; Franco, R.; Panayiotidis, M.I.; Pappa, A. DNA damage induced by endogenous aldehydes: Current state of knowledge. Mutat. Res. 2011, 711, 13–27. [Google Scholar] [CrossRef] [PubMed]
- Wen, C.; Wu, L.; Fu, L.; Wang, B.; Zhou, H. Unifying mechanism in the initiation of breast cancer by metabolism of estrogen (Review). Mol. Med. Rep. 2017, 16, 1001–1006. [Google Scholar] [CrossRef] [PubMed]
- Fussell, K.C.; Udasin, R.G.; Smith, P.J.; Gallo, M.A.; Laskin, J.D. Catechol metabolites of endogenous estrogens induce redox cycling and generate reactive oxygen species in breast epithelial cells. Carcinogenesis 2011, 32, 1285–1293. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yager, J.D.; Liehr, J.G. Molecular mechanisms of estrogen carcinogenesis. Annu. Rev. Pharmacol. Toxicol. 1996, 36, 203–232. [Google Scholar] [CrossRef] [PubMed]
- Yasuda, M.T.; Sakakibara, H.; Shimoi, K. Estrogen- and stress-induced DNA damage in breast cancer and chemoprevention with dietary flavonoid. Genes Environ. 2017, 39, 10. [Google Scholar] [CrossRef] [PubMed]
- Caldon, C.E. Estrogen signaling and the DNA damage response in hormone dependent breast cancers. Front. Oncol. 2014, 4, 106. [Google Scholar] [CrossRef] [PubMed]
- Wysham, W.Z.; Mhawech-Fauceglia, P.; Li, H.; Hays, L.; Syriac, S.; Skrepnik, T.; Wright, J.; Pande, N.; Hoatlin, M.; Pejovic, T. BRCAness profile of sporadic ovarian cancer predicts disease recurrence. PLoS ONE 2012, 7, e30042. [Google Scholar] [CrossRef] [PubMed]
- Mhawech-Fauceglia, P.; Wang, D.; Kim, G.; Sharifian, M.; Chen, X.; Liu, Q.; Lin, Y.G.; Liu, S.; Pejovic, T. Expression of DNA repair proteins in endometrial cancer predicts disease outcome. Gynecol. Oncol. 2014, 132, 593–598. [Google Scholar] [CrossRef] [PubMed]
- Godoy, H.; Mhawech-Fauceglia, P.; Beck, A.; Miller, A.; Lele, S.; Odunsi, K. Expression of poly (adenosine diphosphate-ribose) polymerase and p53 in epithelial ovarian cancer and their role in prognosis and disease outcome. Int. J. Gynecol. Pathol. 2011, 30, 139–144. [Google Scholar] [CrossRef] [PubMed]
© 2019 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Włodarczyk, M.; Nowicka, G. Obesity, DNA Damage, and Development of Obesity-Related Diseases. Int. J. Mol. Sci. 2019, 20, 1146. https://doi.org/10.3390/ijms20051146
Włodarczyk M, Nowicka G. Obesity, DNA Damage, and Development of Obesity-Related Diseases. International Journal of Molecular Sciences. 2019; 20(5):1146. https://doi.org/10.3390/ijms20051146
Chicago/Turabian StyleWłodarczyk, Marta, and Grażyna Nowicka. 2019. "Obesity, DNA Damage, and Development of Obesity-Related Diseases" International Journal of Molecular Sciences 20, no. 5: 1146. https://doi.org/10.3390/ijms20051146
APA StyleWłodarczyk, M., & Nowicka, G. (2019). Obesity, DNA Damage, and Development of Obesity-Related Diseases. International Journal of Molecular Sciences, 20(5), 1146. https://doi.org/10.3390/ijms20051146