DOI QR코드

DOI QR Code

Pre-Natal Epigenetic Influences on Acute and Chronic Diseases Later in Life, such as Cancer: Global Health Crises Resulting from a Collision of Biological and Cultural Evolution

  • Trosko, James E. (Center for Integrative Toxicology, Institute of International Health, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University)
  • Received : 2011.11.01
  • Accepted : 2011.12.02
  • Published : 2011.12.31

Abstract

Better understanding of the complex factors leading to human diseases will be necessary for both long term prevention and for managing short and long-term health problems. The underlying causes, leading to a global health crisis in both acute and chronic diseases, include finite global health care resources for sustained healthy human survival, the population explosion, increased environmental pollution, decreased clean air, water, food distribution, diminishing opportunities for human self-esteem, increased median life span, and the interconnection of infectious and chronic diseases. The transition of our pre-human nutritional requirements for survival to our current culturally-shaped diet has created a biologically-mismatched human dietary experience. While individual genetic, gender, and developmental stage factors contribute to human diseases, various environmental and culturally-determined factors are now contributing to both acute and chronic diseases. The transition from the hunter-gatherer to an agricultural-dependent human being has brought about a global crisis in human health. Initially, early humans ate seasonally-dependent and calorically-restricted foods, during the day, in a "feast or famine" manner. Today, modern humans eat diets of caloric abundance, at all times of the day, with foods of all seasons and from all parts of the world, that have been processed and which have been contaminated by all kinds of factors. No longer can one view, as distinct, infectious agent-related human acute diseases from chronic diseases. Moreover, while dietary and environmental chemicals could, in principle, cause disease pathogenesis by mutagenic and cytotoxic mechanisms, the primary cause is via "epigenetic", or altered gene expression, modifications in the three types of cells (e.g., adult stem; progenitor and terminally-differentiated cells of each organ) during all stages of human development. Even more significantly, alteration in the quantity of adult stem cells during early development by epigenetic chemicals could either increase or decrease the risk to various stem cell-based diseases, such as cancer, later in life. A new concept, the Barker hypothesis, has emerged that indicates pre-natal maternal dietary exposures can now affect diseases later in life. Examples from the studies of the atomic bomb survivors should illustrate this insight.

Keywords

References

  1. Saul JM. 2008. Did detoxification processes cause complex life to emerge? Lethaia 42: 179-184.
  2. Kaput J. 2004. Diet-disease gene interactions. Nutrition 20: 26-31. https://doi.org/10.1016/j.nut.2003.09.005
  3. Kaput J, Rodriguez RL. 2004. Nutritional genomics: the next frontier in the postgenomic era. Physiol Genomics 16: 166-177. https://doi.org/10.1152/physiolgenomics.00107.2003
  4. Saul JM, Schwartz L. 2007. Cancer as a consequence of the rising level of oxygen in the Precambrian. Lethaia 40: 211-220. https://doi.org/10.1111/j.1502-3931.2007.00014.x
  5. Nursall JR. 1959. Oxygen as prerequisite to the origin of metazoan. Nature 183: 1170-1172.
  6. Ozbek S, Balayubramanian PG, Chiquet-Ehrismann R, Tucker RP, Adams JC. 2010. The evolution of extracellular matrix. Molec Biol Cell 21: 4300-4305. https://doi.org/10.1091/mbc.E10-03-0251
  7. Mohyeldin A, Garzon-Muvdi T, Quinones-Hinojosa A. 2010. Oxygen in stem cell biology: A critical component of stem cell niche. Cell Stem Cell 7: 150-161 https://doi.org/10.1016/j.stem.2010.07.007
  8. Pervaiz S, Taneja R, Ghaffan S. 2009. Oxidative stress regulation of stem and progenitor cells. Antioxidants & Redox Signaling 11: 2777-2789. https://doi.org/10.1089/ars.2009.2804
  9. Brigelius-Flohe R, Flohe L. 2011. Basic principles and emerging concepts in the redox control of transcription factors. Antioxidants & Redox Signaling 15: 763-780. https://doi.org/10.1089/ars.2010.3397
  10. Upham BL, Trosko JE. 2009. Carcinogenic tumor promotion, induced oxidative stress signaling, modulated gap junction function and altered gene expression. Antioxidants & Redox Signaling 11: 297-308. https://doi.org/10.1089/ars.2008.2146
  11. Rigoulet M, Yoboue ED, Devin A. 2011. Mitochondrial ROS generation and its regulation: Mechanisms involved in $H_2O_2$ signaling. Antioxidants& Redox Signaling 14: 459-468. https://doi.org/10.1089/ars.2010.3363
  12. Borek C, Sachs L. 1966. The difference in contact inhibition of cell replication between normal cells and cells transformed by different carcinogens. Proc Natl Acad Sci USA 56: 1705-1711. https://doi.org/10.1073/pnas.56.6.1705
  13. Trosko JE. 2011. The gap junction as a "Biological Rosetta Stone": Implications of evolution, stem cells to homeostatic regulation of health and disease in the Barker hypothesis. J Cell Commun Signal 5: 53-66. https://doi.org/10.1007/s12079-010-0108-9
  14. Fuchs E, Tumbar T, Guasch G. 2004. Socializing with the neighbors: stem cells and their niche. Cell 116: 769-778. https://doi.org/10.1016/S0092-8674(04)00255-7
  15. Tumbar T, Guasch G, Greco V, Blanpain C, Lowry WE, Rendl M, Fuchs E. 2004. Defining the epithelial stem cell niche in skin. Science 303: 359-363. https://doi.org/10.1126/science.1092436
  16. Kang KS, Trosko JE. 2011. Stem cells in toxicology: Fundamental biology and practical considerations". Tox Sci 120: 269-289. https://doi.org/10.1093/toxsci/kfr013
  17. Yach D, Leeder SR, Bell J, Kistnasamy B. 2005. Global chronic disease. Science 307: 317. https://doi.org/10.1126/science.307.5708.317
  18. Trosko JE. 1984. Scientific views of human nature: Implications for the ethics of technological intervention. In The Culture of Biomedicine. Brock DH, ed. University of Delaware Press, Newark, NJ, USA. Vol 1, p 70-97.
  19. Trosko JE. 2007. Stem cells and cell-cell communication in the understanding of the role of diet and nutrients in human diseases. J Food Hygiene & Safety 22: 1-14.
  20. Teaford MF, Ungar PS. 2000. Diet and the evolution of the earliest human ancestors. Proc Natl Acad Sci USA 97: 13506-13511. https://doi.org/10.1073/pnas.260368897
  21. Mariani-Costantini A. 2000. Natural and cultural influences on the evolution of the human diet: Background of the multifactorial processes that shaped the eating habits of Western societies. Nutrition 16: 483-486. https://doi.org/10.1016/S0899-9007(00)00296-3
  22. Muegge BD, Kuczynski J, Knights D, Clemente JC, Gonzalez A, Fontana L, Henrissat B, Knight R, Gordon JI. 2011. Diet drives convergence in gut microbiome functions across mammalian phylogeny and within humans. Science 332: 970-974. https://doi.org/10.1126/science.1198719
  23. Strober W. 2010. Gut microbes: Friends or fiends? Nature Medicine 16: 1195-1197. https://doi.org/10.1038/nm1110-1195
  24. Gareau MG, Sherman PM, Walker WA. 2010. Probiotics and the gut microbiota in intestinal health and disease. Nature Reviews Gastro Hepatol 7: 503-514. https://doi.org/10.1038/nrgastro.2010.117
  25. Sandoval DA, Seeley RJ. 2010. The microbes made me eat it. Science 328: 12.
  26. Niwa T, Tsukamoto T, Toyoda T, Mon A, Tanaka H, Maekita T, Ichinose M, Tatematsu M, Ushijima T. 2010. Inflammatory processes triggered by Heliobacter pylori infection cause aberrant DNA methylation in gastric epithelial cells. Cancer Res 70: 1430-1440. https://doi.org/10.1158/0008-5472.CAN-09-2755
  27. Turnbaugh PJ, Ridaura VK, Faith JJ, Rey FE, Knight R, Gordon JI. 2009. The effect of diet on the human gut microbiome: A metagenomic analysis in humanized gnotobiotic mice. Sci Transl Med 1: 5-8.
  28. Philpott DJ, Girardin SE. 2010. Gut microbes extend reaches to systemic innate immunity. Nat Med 16: 160-161. https://doi.org/10.1038/nm0210-160
  29. Flier JS, Mekalaanos JJ. 2009. Gut check: testing a role for the intestinal microbiome in human obesity. Sci Transl Med 1: 5-8.
  30. Kau AL, Ahern PP, Griffin NW, Goodman AL, Gordon JI. 2011. Human nutrition, the gut microbiome and the immune system. Nature 474: 327-336. https://doi.org/10.1038/nature10213
  31. Rak K, Rader DJ. 2011. The diet-microbe morbid union. Nature 472: 440-472.
  32. Bruce KD, Byrne CD. 2009. The metabolic syndrome: common origins of multifactorial disorder. Postgrad Med J 85: 614-621. https://doi.org/10.1136/pgmj.2008.078014
  33. Lusis AJ, Attie AD, Karen Reue K. 2008. Metabolic syndrome: from epidemiology to systems biology. Nat Rev Genet 9: 819-830. https://doi.org/10.1038/nrg2468
  34. Trosko JE, Tai MH. 2006. Adult stem cell theory of the multi-stage, multi-mechanism theory of carcinogenesis: Role of inflammation on the promotion of initiated cells. In Infections and Inflammation: Impacts on Oncogenesis. Dittmar T, Zaenker KS, Schmidt A, eds. S. Karger AG, Basel, Switzerland. Vol 13, p 45-65.
  35. Trosko JE, Upham BL. 2010. Commentary on "Toxicology testing in the 21st Century: A vision and a strategy": Stem cells and cell-cell communication as fundamental targets in assessing the potential toxicity of chemicals. Hum Exp Toxicol 29: 21-29. https://doi.org/10.1177/0960327109354663
  36. Trosko JE, Upham BL. 2005. The emperor wears no clothes in the field of carcinogen risk assessment: Ignored concepts in cancer risk assessment. Mutagenesis 20: 81-92. https://doi.org/10.1093/mutage/gei017
  37. Pitot H, Dragon YP. 1991. Facts and theories concerning the mechanisms of carcinogenesis. FASEB J 5: 2280-2286.
  38. Weinstein IB, Cattoni-Celli S, Kirschmeier P, Lambert M, Hsiao W, Backer J, Jeffrey A. 1984. Multistage carcinogenesis involves multiple genes and multiple mechanisms. J Cell Physiol 3: 127-137.
  39. Fialkow PJ. 1976. Clonal origin of human tumours. Am Rev Med 30: 135-176.
  40. Nowell PC. 1976. The clonal evolution of tumor cell populations. Science 194: 23-28. https://doi.org/10.1126/science.959840
  41. Brash DE, Rudolph JA, Simon A, Lin A, McKenna GJ, Baden HP, Halperin AJ, Ponten J. 1991. A role for sunlight in skin cancer: UV-induced p53 mutations in squamous cell carcinomas. Proc Natl Acad Sci USA 88: 10124-10128. https://doi.org/10.1073/pnas.88.22.10124
  42. Hanahan D, Weinberg RA. 2000. The hallmarks of cancer. Cell 100: 57-70. https://doi.org/10.1016/S0092-8674(00)81683-9
  43. Hanahan D, Weinberg RA. 2011. Hallmarks of cancer: The next generation. Cell 144: 646-674. https://doi.org/10.1016/j.cell.2011.02.013
  44. Trosko JE, Ruch RJ. 1998. Cell-cell communication in carcinogenesis. Front Biosci 3: 208-236. https://doi.org/10.2741/A275
  45. Trosko JE, Ruch RJ. 2002. Gap junctions as targets for cancer chemoprevention and chemotherapy. Curr Drug Targets 3: 465-482. https://doi.org/10.2174/1389450023347371
  46. Goodman JA. 2001. Operational reversibility is a key aspect of carcinogenesis. Toxicol Sci 64: 147-148. https://doi.org/10.1093/toxsci/64.2.147
  47. Boutwell RK. 1964. Some biological aspects of skin carcinogenesis. Prog Exp Tumor Res 4: 207-250.
  48. Pitot HC. 1989. Progression-the terminal stage in carcinogenesis. Jpn J Cancer Res 80: 599-607. https://doi.org/10.1111/j.1349-7006.1989.tb01683.x
  49. Trosko JE. 2008. Human adult stem cells as targets for cancer stem cells: Evolution; Oct-4 gene and cell-cell communication. In Stem Cells and Cancer. Dittmar T, Zaenkar K, eds. Nova Science Publishers, Hauppauge, NY, USA. p 147-187.
  50. Hayflick L. 1965. The limited in vitro lifespan of human diploid cell strains. Exp Cell Res 37: 614-636. https://doi.org/10.1016/0014-4827(65)90211-9
  51. Borek C, Sachs L. 1966. The difference in contact inhibition of cell replication between normal cells and cells transformed by different carcinogens. Proc Natl Acad Sci USA 56: 1705-1711. https://doi.org/10.1073/pnas.56.6.1705
  52. Loewenstein WR, Kanno Y. 1966. Intercellular communication and the control of tissue growth: lack of communication between cancer cells. Nature 209: 1248-1249. https://doi.org/10.1038/2091248a0
  53. King TJ, Fukushima LH, Donlon TA, Hieber AD, Shimabukuro KA, Bertram JS. 2000. Correlation between growth control, neoplastic potential and exogenous connexin43 expression in HeLa cell lines: Implications for tumor progression. Carcinogenesis 21: 311-315. https://doi.org/10.1093/carcin/21.2.311
  54. Momiyama M, Omori Y, Ishizaka Y, Nishikawa Y, Tokairin T, Ogawa J, Enomoto K. 2003. Connexin26-mediated gap junctional communication reverses the malignant phenotypic of MCF-7 breast cancer cells. Cancer Sci 94: 501-507. https://doi.org/10.1111/j.1349-7006.2003.tb01473.x
  55. Trosko JE. 2003. The role of stem cells and gap junctional intercellular communication in carcinogenesis. J Biochem Molec Biol 36: 43-48. https://doi.org/10.5483/BMBRep.2003.36.1.043
  56. Al Hajj M, Wicha MS, Benito-Hernandez A. 2003. Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci USA 100: 3983-3988. https://doi.org/10.1073/pnas.0530291100
  57. Ponti D, Costa A, Zaffaroni N, Pratesi G, Petrangolini G, Coradini D, Pilotti S, Pierotti MA, Daidone MA. 2005. Isolation and in vitro propagation of tumorigenic breast cancer cells with stem/ progenitor cell properties. Cancer Res 65: 5506-5511. https://doi.org/10.1158/0008-5472.CAN-05-0626
  58. Nakagawa M, Koyanagi M, Tanabe K, Takahashi K, Ichisaka T, Aoi T, Okita K, Mochiduki Y, Takizawa N, Yamanaka S. 2008. Generation of induced pluripotent stem cells without Myc from mouse and human fibroblasts. Nat Biotechnol 26: 101-106. https://doi.org/10.1038/nbt1374
  59. Weinberg RA. 1991. Tumor suppressor genes. Science 254: 1138-1146. https://doi.org/10.1126/science.1659741
  60. Land H, Parada IE, Weinberg RA. 1983. Tumorigenic conversion of primary embryo fibroblasts requires at least two cooperating oncogenes. Nature 304: 596-602. https://doi.org/10.1038/304596a0
  61. Trosko JE. 2008. Human adult stem cells as the target cell for the initiation of carcinogenesis and for the generation of "cancer stem cells". Intl J Stem Cells 1: 8-26. https://doi.org/10.15283/ijsc.2008.1.1.8
  62. Trosko JE. 2009. Cancer stem cells and cancer non-stem cells: From adult stem cells or from re-programming of differentiated somatic cells. Vet Pathol 46: 176-193. https://doi.org/10.1354/vp.46-2-176
  63. Trosko JE, Chang CC, Wilson MR, Upham BL, Hayashi T, Wade M. 2000. Gap junctions and the regulation of cellular function of stem cells during development and differentiation. Methods 20: 245-264. https://doi.org/10.1006/meth.1999.0941
  64. Chang CC, Sun W, Cruz A, Saitoh M, Tai MH, Trosko JE. 2001. A human breast epithelial cell type with stem cell characteristics as targets for carcinogenesis. Radiat Res 155: 201-207. https://doi.org/10.1667/0033-7587(2001)155[0201:AHBECT]2.0.CO;2
  65. Tai MH, Chang CC, Kiupel M, Webster JD, Trosko JE. 2005. Oct-4 expression in adult stem cells: evidence in support of the stem cell theory of carcinogenesis. Carcinogenesis 26: 495-502.
  66. Trosko JE, Chang CC. 2010. Factors to consider in the use of stem cells for pharmaceutics drug development and for chemical safety assessment. Toxicology 270: 18-34. https://doi.org/10.1016/j.tox.2009.11.019
  67. Yamanaka S. 2009. A fresh look at iPS cells. Cell 137: 13-17. https://doi.org/10.1016/j.cell.2009.03.034
  68. Trosko JE. 2008. Human adult stem cells as the target cell for the initiation of carcinogenesis and for the generation of "cancer stem cells". Intl J Stem Cells 1: 8-26. https://doi.org/10.15283/ijsc.2008.1.1.8
  69. Liu S. 2008. One factor dropped from the 'inducing' soup, one piece of evidence added against the 'introduction' claim. Logical Biology 8: 39-41.
  70. Liu S. 2008. iPS cells: A more critical review. Stem Cells Dev 17: 391-397. https://doi.org/10.1089/scd.2008.0062
  71. Trosko JE. 2008. Commentary: Re-programming or selecting adult stem cells. Stem Cell Reviews 4: 81-88. https://doi.org/10.1007/s12015-008-9017-1
  72. Stephen D, Hursting SD, Jackie A, Lavigne JA, Berrigan D, Perkins SN, Barrett JC. 2003. Caloric restriction ,aging and cancer prevention: Mechanisms of action and applicability to humans. Annu Rev Med 54: 131-152. https://doi.org/10.1146/annurev.med.54.101601.152156
  73. Sporn M, Newton DL, 1979. Chemoprevention of cancer by retinoic acid. Fed Proc 38: 2528-2534.
  74. Forbes PD, Urbach F, Davies RE. 1979. Enhancement of experimental photocarcinogenesis by topical retinoic acid. Cancer Lett 7: 85-90. https://doi.org/10.1016/S0304-3835(79)80100-7
  75. Pence BC. 1993. Role of calcium in colon cancer prevention: Experimental and clinical studies. Mutat Res Fundam Mol Mech Mutagen 290: 87-95. https://doi.org/10.1016/0027-5107(93)90036-F
  76. Kim YS, Young MR, Bobe G, Colburn NH, Milner JA. 2009. Bioactive food components, inflammatory targets, and cancer prevention. Cancer Rev Res 2: 200-208. https://doi.org/10.1158/1940-6207.CAPR-08-0141
  77. Butler LM, Yu MC. 2011. Fish oil exacerbates colitis in SMAD3 mice. Cancer Res 71: 287. https://doi.org/10.1158/0008-5472.CAN-10-3426
  78. Reddy BS. 2004. Omega-3 fatty acids in colorectal cancer prevention. Int J Cancer 122: 1-7.
  79. Lambert JD, Sang S, Yang CS. 2007. Possible controversy over dietary polyphenols: benefits vs risks. Chem Res Toxicol 20: 583-585. https://doi.org/10.1021/tx7000515
  80. El Tonny LH, Banerjee PP. 2009. Identification of a biphasic role for genistein in the regulation of prostate cancer growth and metastasis. Cancer Res 69: 3695-3703. https://doi.org/10.1158/0008-5472.CAN-08-2958
  81. Molseeva EP, Manson MM. 2009. Dietary chemopreventive phytochemicals: Too little or too much? Cancer Prev Res 2: 611-616. https://doi.org/10.1158/1940-6207.CAPR-08-0102
  82. Trosko JE, Chang CC, Upham BL, Tai MH. 2005. The role of human adult stem cells and cell-cell communication in cancer chemoprevention and chemotherapy strategies. Mutat Res 591: 187-197. https://doi.org/10.1016/j.mrfmmm.2005.01.034
  83. Trosko JE. 2006. Dietary modulation of multi-stage, multi-mechanisms of human carcinogenesis: Effects of initiated stem cells and cell-cell communication. Nutr Cancer 54: 102-110. https://doi.org/10.1207/s15327914nc5401_12
  84. Metha PP, Loewenstrein WR. 1991. Differential regulation of communication by retinoic acid in homologous and heterologous junctions between normal and transformed cells. J Cell Biol 113: 371-379. https://doi.org/10.1083/jcb.113.2.371
  85. Zhang LX, Cooney RV, Bertram JS. 1991. Carotenoids enhance gap junctional communication and inhibit lipid peroxidation in C3H/10T1/2 cells: relationship to their cancer chemopreventive action. Carcinogenesis 12: 2109-2114. https://doi.org/10.1093/carcin/12.11.2109
  86. Nakamura Y, Yoshikawa N, Hiroki I, Sato K, Ohtsuki K, Chang CC, Upham BL, Trosko JE. 2005. $\beta$-Sitosterol, from psyllium seed husk (Plantago ovata Forsk), restores gap junctional intercellular communication in haras transfected rat liver cells. Nutr Cancer 5: 218-225.
  87. Sai K, Kanno J, Hasegawa, R, Trosko JE, Inoue T. 2000. Prevention of the down-regulation of gap junctional intercellular communication by green tea in the liver of mice fed pentachlorophenol. Carcinogenesis 21: 1671-1676. https://doi.org/10.1093/carcin/21.9.1671
  88. Nielson M, Ruch RJ, Vang O. 2000. Resveratrol reverses tumor-promoter-induced inhibition of gap junctional intercellular communication. Biochem Biophys Res Commun 275: 804-809. https://doi.org/10.1006/bbrc.2000.3378
  89. Na HK, Wilson MR, Kang KS, Chang CC, Grunberger D, Trosko JE. 2000. Restoration of gap junctional intercellular communication by caffeic acid phenethyl ester (CAPE) in a ras-transformed rat liver epithelial cell line. Cancer Lett 157: 31-38. https://doi.org/10.1016/S0304-3835(00)00470-5
  90. Lee D, Shin BJ, Hur HJ, Kim JH, Kim J, Kang NJ, Kim DO, Lee CY, Lee KW, Lee HJ. 2010. Quercetin, the active phenolic compound in kiwifruit, prevents hydrogen peroxide-induced inhibition of gap junction intercellular communication. Br J Nutr 104: 164-170. https://doi.org/10.1017/S0007114510000346
  91. Wakao S, Kitada M, Kuroda Y, Shigemoto T, Matsuse D, Akashi H, Tanimura Y, Tsuchiyama K, Kikuchi T, Goda M, Nakahata T, Fujiyoshi Y, Dezawa M. 2011. Multilineage-differentiating stress-enduring (MUSE) cells are a primary source of induced pluripotent stem cells in human fibroblasts. Proc Natl Acad Sci USA 108: 9875-9880. https://doi.org/10.1073/pnas.1100816108
  92. Trosko JE, Chang CC, Madhukar BV, Dupont E. 1996. Intercellular communication: A paradigm for the interpretation of the initiation/promotion/progression model of carcinogenesis. In Chemical Induction of Cancer: Modulation and Combination Effects. Arcos JC, Argus MF, Woo YT, eds. Birkhauser Publisher, Boston, MA, USA. p 205-225.
  93. Trosko JE. Chang CC. 1989. Nongenotoxic mechanisms in carcinogenesis: Role of inhibited intercellular communication. In Banbury Report 31: New Directions in the Qualitative and Quantitative Aspects of Carcinogen Risk Assessment. Hart RFD, Hoerger FD, eds. Cold Spring Harbor Press, Cold Spring Harbor, NY, USA. p 139-170.
  94. Matesic DF, Rupp HL, Bonney WJ, Ruch RJ, Trosko JE. 1994. Changes in gap junction permeability phosphorylation, and number mediated by phorbol ester and nonphorbol ester tumor promoters in rat liver epithelial cells. Molec Carcinog 10: 226-236. https://doi.org/10.1002/mc.2940100407
  95. Ruch RJ, Madhukar BV, Trosko JE, Klaunig JE. 1993. Reversal of ras-induced inhibition of gap junctional intercellular communication, transformation, and tumorigenesis by lovastatin. Mol Carcinog 7: 50-59. https://doi.org/10.1002/mc.2940070109
  96. Ogawa T, Hayashi T, Tokunou M, Nakachi K, Trosko JE, Chang CC, Yorioka N. 2005. Suberoylanilide hydroxamic acid enhances gap junctional intercellular communication via acetylation of histone containing connexin43 gene locus. Cancer Res 65: 9771-9778. https://doi.org/10.1158/0008-5472.CAN-05-0227
  97. Trosko JE. 2005. The role of stem cells and gap junctions as targets for cancer chemoprevention and chemotherapy. Biomed Pharmacother 59: 326-331. https://doi.org/10.1016/S0753-3322(05)80065-4
  98. Lamartiniere CA, Moore JB, Brown NM, Thompson R, Hardin MJ, Barnes S. 1995. Genistein suppresses mammary cancers in rats. Carcinogenesis 16: 2833-2840. https://doi.org/10.1093/carcin/16.11.2833
  99. Ling WH, Jones PJH. 1995. Dietary phytosterols: a review of metabolism benefits and side effects. Life Sci 57: 195-206. https://doi.org/10.1016/0024-3205(95)00263-6
  100. Ju YH, Allred CD, Allred KF, Karko KL, Doerge DR, Helferich WG. 2001. Physiological concentrations of dietary genistein dose-dependently stimulate growth of estrogen- dependent human breast cancer (MCF-7) tumors implanted inathymic mice. J Nutr 131: 2957-2962.
  101. Baur A, Kevin J, Pearson KJ, Price NL, Jamieson HA, Lerin C, Kalra A, Prabhu VV, Allard JS, Lopez-Lluch G, Lewis K, Pistell PJ, Poosala S, Becker KG, Boss O, Gwinn D, Wang M, Ramaswamy S, Fishbein KW, Spencer RG, Lakatta EG, Le Couteur D, Shaw RJ, Navas P, Puigserve P, Ingram DK, de Cabo R, Sinclair DA. 2006. Resveratrol improves health and survival of mice on a high caloric diet. Nature 444: 337-342. https://doi.org/10.1038/nature05354
  102. Ahmad A, Syed FA, Singh S, Hadi SM. 2005. Prooxidant activity of reservatrol in the presence of copper ions: mutagenicity in plasmid DNA. Toxicol Lett 159: 1-12. https://doi.org/10.1016/j.toxlet.2005.04.001
  103. Sporn MB, Newton DL. 1979. Chemoprevention of cancer with retinoids. Fed Proc 38: 2528-2534.
  104. Welsch CW, Goodrich-Smith M, Brown CK, Crowe N. 1981. Enhancement by retinyl acetate of hormone-induced mammary tumorigenesis in female GR/A mice. J Natl Cancer Inst 67: 935-938.
  105. Shuin T, Nishimura R, Noda K, Umeda M, Ono T. 1983. Concentration-dependent differential effect of retinoic acid on intercellular metabolic cooperation. Gann 74: 100-105.
  106. Henning H, Wenk ML, Dohahoe R. 1982. Retinoic acid promotion of Papilloma formation in mouse skin. Cancer Lett 16: 1-5. https://doi.org/10.1016/0304-3835(82)90084-2
  107. Halliwell B. 2009. The wanders of a free radical. Free Radic Biol Chem 46:531-542. https://doi.org/10.1016/j.freeradbiomed.2008.11.008
  108. Perera RW, Bardeesy N. 2011. When antioxidants are bad. Nature 475: 43-45. https://doi.org/10.1038/475043a
  109. Moini H, Packer L, Saris NE. 2002. Antioxidant and prooxidant activities of alpha-lipoic acid and dihydrolipoic acid. Toxicol Appl Pharmacol 182: 84-90. https://doi.org/10.1006/taap.2002.9437
  110. He K, Nukada H, Urakami T, Murphy MP. 2003. Antioxidant and pro-oxidant properties of pyrroloquinoline quinine (PQQ): Implications for its function in biological systems. Biochem Pharmocol 65: 67-74. https://doi.org/10.1016/S0006-2952(02)01453-3
  111. Duffield-Lillico AJ, Begg CB. 2004. Reflections on the landmark studies of beta-carotene supplementation. J Natl Cancer Inst 96: 1729-1731. https://doi.org/10.1093/jnci/djh344
  112. Goodman GE, Thornquist MD, Balmes J, Cullen MR, Meyskens FL, Omenn GS, Valanis B, Williams JH. Jr. 2004. The Beta-Carotene and Retinol Efficacy Trial: incidence of lung cancer and cardiovascular disease mortality during 6-year follow-up after stopping beta-carotene and retinol supplements. J Natl Cancer Inst 96: 1743-1750. https://doi.org/10.1093/jnci/djh320
  113. Barker DJP. 1998. Mothers, babies, and health in later life. 2nd ed. Churchill Livingstone, Edinburgh, UK.
  114. Barker DJ. The developmental origins of adult disease. J Am Coll Nutr 236: 588s-595s.
  115. Cousins L, Karp W, Lacey C, Lucas WE. 1980. Reproductive outcome of women exposed to diethylstilbestrol in utero. Obstet Gynecol 56: 70-76.
  116. Ho SM, Tang WY, de Frauto JB, Prins GS. 2006. Developmental exposures to estradiol and bisphenol: A increases susceptibility to prostate carcinogenesis and epigenetically regulates phosphodiesterase type 4 variant 4. Cancer Res 66: 5624-5632. https://doi.org/10.1158/0008-5472.CAN-06-0516
  117. Willcox BJ, Willcox DC, Todoriki H, Fujiyoshi A, Yano K, He Q, Curb JD, Suzuki M. 2007. Caloric restriction, the traditional Okinawan diet, and healthy aging: the diet of the world's longest-lived people and its potential impact on morbidity and life span. Ann N Y Acad Sci 1114: 434-455. https://doi.org/10.1196/annals.1396.037
  118. Trosko JE, Suzuki K. 2009. Adult stem cells, the Barker Hypothesis, epigenetic events and low level radiation effects. In Radiatiation Health Risk Sciences. Nakashima M, Takamura N, Tsukasaki K, Nagayama Yamashita YS, eds. Springer Publisher, Tokyo, Japan. p 216-226.
  119. Hsieh CY, Chang CC. 1999. Stem cell differentiation and reduction as a potential mechanism for chemoprevention of breast cancer. Chin Pharm J 51: 1530.
  120. Milton K. 2000. Back to basics: Why foods of wild primates have relevance for modern human health. Nutrition 16: 480-483. https://doi.org/10.1016/S0899-9007(00)00293-8
  121. Kiple KF. 2000. The Cambridge world history of food. Cambridge University Press, Cambridge, UK.
  122. Teaford MF, Ungar PS. 2000. Diet and the evolution of the earliest human ancestors. Proc Natl Acad Sci USA 97: 13506-13511. https://doi.org/10.1073/pnas.260368897
  123. Mariani-Costantini A. 2000. Natural and cultural influences on the evolution of the human diet: Background of the multifactorial processes that shaped the eating habits of Western societies. Nutrition 16: 483-486. https://doi.org/10.1016/S0899-9007(00)00296-3
  124. Cordain L, Miller JB, Eaton SB, Mann N, Holt SHA, Speth JD. 2000. Plant-animal subsistence ratios and micronutrient energy estimates in worldwide hunter-gather diets. Am J Clin Nutr 71: 682-692.
  125. Paoloni-Giacobino AR, Grimble R, Pichard C. 2003. Genetics and nutrition. Clin Nutr 22: 429-435. https://doi.org/10.1016/S0261-5614(03)00064-5
  126. Benzie IF, Wachtel-Galor S. 2010. Vegetarian diets and public health: Biomarker and redox connections. Antioxid Redox Signal 13: 1575-1591. https://doi.org/10.1089/ars.2009.3024

Cited by

  1. Evolution of energy metabolism, stem cells and cancer stem cells: how the Warburg and Barker hypothesis might be linked vol.7, pp.Suppl 2, 2013, https://doi.org/10.1186/1753-6561-7-S2-K8
  2. Science, innovation and society vol.14, 2016, https://doi.org/10.2903/j.efsa.2016.s0502
  3. Embryonic stem cells and the next generation of developmental toxicity testing vol.13, pp.8, 2017, https://doi.org/10.1080/17425255.2017.1351548
  4. Evolution of Microbial Quorum Sensing to Human Global Quorum Sensing: An Insight into How Gap Junctional Intercellular Communication Might Be Linked to the Global Metabolic Disease Crisis vol.5, pp.2, 2016, https://doi.org/10.3390/biology5020029