Full Text:  <1831>

Summary:  <1391>

CLC number: Q255

On-line Access: 2019-06-06

Received: 2018-10-03

Revision Accepted: 2019-03-12

Crosschecked: 2019-06-05

Cited: 0

Clicked: 3066

Citations:  Bibtex RefMan EndNote GB/T7714

-   Go to

Article info.
Open peer comments

Journal of Zhejiang University SCIENCE B

Accepted manuscript available online (unedited version)


Epigenetics recording varied environment and complex cell events represents the origin of cellular aging


Author(s):  Xue-Jun Guo, Dong Yang, Xiang-Yuan Zhang

Affiliation(s):  State Key Laboratory of Environment Simulation, School of Environment, Beijing Normal University, Beijing 100875, China; more

Corresponding email(s):  guoxj@bnu.edu.cn

Key Words:  Epigenetics; Environment; Cell event; Cellular aging; Epigenome entropy; DNA methylation


Share this article to: More <<< Previous Paper|Next Paper >>>

Xue-Jun Guo, Dong Yang, Xiang-Yuan Zhang. Epigenetics recording varied environment and complex cell events represents the origin of cellular aging[J]. Journal of Zhejiang University Science B,in press.Frontiers of Information Technology & Electronic Engineering,in press.https://doi.org/10.1631/jzus.B1800507

@article{title="Epigenetics recording varied environment and complex cell events represents the origin of cellular aging",
author="Xue-Jun Guo, Dong Yang, Xiang-Yuan Zhang",
journal="Journal of Zhejiang University Science B",
year="in press",
publisher="Zhejiang University Press & Springer",
doi="https://doi.org/10.1631/jzus.B1800507"
}

%0 Journal Article
%T Epigenetics recording varied environment and complex cell events represents the origin of cellular aging
%A Xue-Jun Guo
%A Dong Yang
%A Xiang-Yuan Zhang
%J Journal of Zhejiang University SCIENCE B
%P 550-562
%@ 1673-1581
%D in press
%I Zhejiang University Press & Springer
doi="https://doi.org/10.1631/jzus.B1800507"

TY - JOUR
T1 - Epigenetics recording varied environment and complex cell events represents the origin of cellular aging
A1 - Xue-Jun Guo
A1 - Dong Yang
A1 - Xiang-Yuan Zhang
J0 - Journal of Zhejiang University Science B
SP - 550
EP - 562
%@ 1673-1581
Y1 - in press
PB - Zhejiang University Press & Springer
ER -
doi="https://doi.org/10.1631/jzus.B1800507"


Abstract: 
Although a relationship between epigenetics and aging phenotypic changes has been established, a theoretical explanation of the intrinsic connection between the epigenetics and aging is lacking. In this essay, we propose that epigenetic recording of varied cell environment and complex history could be an origin of cellular aging. Through epigenetic modifications, the environment and historical events can induce the chromatin template into an activated or repressive accessible structure, thereby shaping the DNA template into a spectrum of chromatin states. The inner nature of diversity and conflicts born by the cell environment and its historical events are hence recorded into the chromatin template. This could result in a dissipated spectrum of the chromatin state and chaos in overall gene expression. An unavoidable degradation of epigenome entropy, similar to Shannon entropy, would be consequently induced. The resultant disorder in epigenome, characterized by corrosion of epigenome entropy as reflected in chromatin template, can be stably memorized and propagated through cell division. Furthermore, the hysteretic nature of epigenetics responding to the emerging environment could exacerbate the degradation of epigenome entropy. As well as stochastic errors, we propose that outside entropy (or chaos) derived from the varied environment and complex cell history, gradually input and imprinted into the chromatin via epigenetic modifications, would lead inevitably to cellular aging, the extent of which could be aggravated by hysteresis of epigenetics without error erasing and correction.

多变环境和复杂事件的表观遗传记录是细胞的衰老之源

概要:虽然表观遗传和衰老的关联性已经被广泛接受,但是两者之间内在的因果关系需要深度的理论阐述和分析.本文通过分析环境与表观遗传相互作用及其过程的本质特征,证明表观遗传记录多变的细胞环境和复杂的细胞事件是细胞的衰老之源.细胞周围环境以及所经历事件可通过表观修饰激活或抑制染色体上各个基因的表达,从而 对染色体模板以及基因表达模式进行塑造.外在环境和细胞历史的多变性以及内在冲突性由此被逐渐记录在染色体模板上,整体上导致染色体模板以及基因表达模式的混乱和耗散.这将导致细胞表观熵(类似于Shannon信息熵)不可避免地逐渐衰减,其混乱度可通过细胞分裂而进一步被稳定积累、记忆和扩增.另外,表观修饰应对外在环境的变化存在滞后性和一定的不可逆性,加剧了表观熵的衰退.
关键词组:表观遗传;环境;细胞事件;细胞衰老;表观熵;DNA甲基化

Darkslateblue:Affiliate; Royal Blue:Author; Turquoise:Article

Reference

[1]Aguilera O, Fernández AF, Muñoz A, et al., 2010. Epigenetics and environment: a complex relationship. J Appl Physiol, 109(1):243-251.

[2]Alberts B, Johnson A, Lewis J, et al., 2008. Molecular Biology of the Cell, 5th Ed. Garland Science Taylor and Francis Group, New York, p.411-477.

[3]Allis CD, Caparros ML, Jenuwein T, et al., 2015. Overview and concepts. In: Allis CD, Caparros ML, Jenuwein T, et al. (Eds.), Epigenetics, 2nd Ed. Cold Spring Harbor, New York, p.47-115.

[4]Baccarelli A, Bollati V, 2009. Epigenetics and environmental chemicals. Curr Opin Pediatr, 21(2):243-251.

[5]Barros SP, Offenbacher S, 2009. Epigenetics: connecting environment and genotype to phenotype and disease. J Dent Res, 88(5):400-408.

[6]Ben-Porath I, Weinberg RA, 2005. The signals and pathways activating cellular senescence. Int J Biochem Cell Biol, 37(5):961-976.

[7]Berdyshev GD, Korotaev GK, Boiarskikh GV, et al., 1967. Nucleotide composition of DNA and RNA from somatic tissues of humpback and its changes during spawning. Biokhimiia, 32(5):988-993.

[8]Brandeis M, Frank D, Keshet I, et al., 1994. Sp1 elements protect a CpG island from de novo methylation. Nature, 371(6496):435-438.

[9]Brenner C, Deplus R, Didelot C, et al., 2005. Myc represses transcription through recruitment of DNA methyltransferase corepressor. EMBO J, 24(2):336-346.

[10]Brunet A, Berger SL, 2014. Epigenetics of aging and aging-related disease. J Gerontol Ser A, 69(S1):S17-S20.

[11]Calvanese V, Lara E, Kahn A, et al., 2009. The role of epigenetics in aging and age-related diseases. Ageing Res Rev, 8(4):268-276.

[12]Campisi J, 2013. Aging, cellular senescence, and cancer. Annu Rev Physiol, 75:685-705.

[13]Cavalli G, Paro R, 1999. Epigenetic inheritance of active chromatin after removal of the main transactivator. Science, 286(5441):955-958.

[14]Cooney CA, 2007. Epigenetics—DNA-based mirror of our environment? Dis Markers, 23(1-2):121-137.

[15]Darlington CD, 1937. Recent Advances in Cytology, 2nd Ed. Blakiston, Philadelphia.

[16]Feil R, Fraga MF, 2012. Epigenetics and the environment: emerging patterns and implications. Nat Rev Genet, 13(2):97-109.

[17]Feng Y, Tian JJ, Krylova I, et al., 2016. Chronic TCDD exposure results in the dysregulation of gene expression in splenic B-lymphocytes and in the impairments in T-cell and B-cell differentiation in mouse model. J Environ Sci, 39:218-227.

[18]Gebhard C, Benner C, Ehrich M, et al., 2010. General transcription factor binding at CpG islands in normal cells correlates with resistance to de novo DNA methylation in cancer cells. Cancer Res, 70(2):1398-1407.

[19]Hemberger M, Dean W, Reik W, 2009. Epigenetic dynamics of stem cells and cell lineage commitment: digging Waddington’s canal. Nat Rev Mol Cell Biol, 10(8):526-537.

[20]Hong L, Schroth GP, Matthews HR, et al., 1993. Studies of the DNA binding properties of histone H4 amino terminus. Thermal denaturation studies reveal that acetylation markedly reduces the binding constant of the H4 “tail” to DNA. J Biol Chem, 268(1):305-314.

[21]Horvath S, 2013. DNA methylation age of human tissues and cell types. Genome Biol, 14(10):R115.

[22]Huidobro C, Fernandez AF, Fraga MF, 2013. Aging epigenetics: causes and consequences. Mol Aspects Med, 34(4):765-781.

[23]Issa JP, 2014. Aging and epigenetic drift: a vicious cycle. J Clin Invest, 124(1):24-29.

[24]Johnson FB, Sinclair DA, Guarente L, 1999. Molecular biology of aging. Cell, 96(2):291-302.

[25]Kaati G, Bygren LO, Edvinsson S, 2002. Cardiovascular and diabetes mortality determined by nutrition during parents’ and grandparents’ slow growth period. Eur J Hum Genet, 10(11):682-688.

[26]Klaassen CD, 2008. Casarett and Doull’s Toxicology: The Basic Science of Poisons, 7th Ed. The McGraw-Hill Companies, New York, p.557-582.

[27]Kohli RM, Zhang Y, 2013. TET enzymes, TDG and the dynamics of DNA demethylation. Nature, 502(7472):472-479.

[28]Koschinsky T, He CJ, Mitsuhashi T, et al., 1997. Orally absorbed reactive glycation products (glycotoxins):an environmental risk factor in diabetic nephropathy. Proc Natl Acad Sci USA, 94(12):6474-6479.

[29]Lardenoije R, Iatrou A, Kenis G, et al., 2015. The epigenetics of aging and neurodegeneration. Prog Neurobiol, 131: 21-64.

[30]Laurent M, Charvin G, Guespin-Michel J, 2005. Bistability and hysteresis in epigenetic regulation of the lactose operon. Since Delbrück, a long series of ignored models. Cell Mol Biol (Noisy-le-Grand), 51(7):583-594.

[31]Lienert F, Wirbelauer C, Som I, et al., 2011. Identification of genetic elements that autonomously determine DNA methylation states. Nat Genet, 43(11):1091-1097.

[32]Macleod D, Charlton J, Mullins J, et al., 1994. Sp1 sites in the mouse aprt gene promoter are required to prevent methylation of the CpG island. Genes Dev, 8(19):2282-2292.

[33]Margueron R, Reinberg D, 2011. The Polycomb complex PRC2 and its mark in life. Nature, 469(7330):343-349.

[34]Martin GM, 2005. Epigenetic drift in aging identical twins. Proc Natl Acad Sci USA, 102(30):10413-10414.

[35]Nagaraj VH, Mukhopadhyay S, Dayarian A, et al., 2014. Breaking an epigenetic chromatin switch: curious features of hysteresis in Saccharomyces cerevisiae telomeric silencing. PLoS ONE, 9(12):e113516.

[36]Nakayama J, Rice JC, Strahl BD, et al., 2001. Role of histone H3 lysine 9 methylation in epigenetic control of heterochromatin assembly. Science, 292(5514):110-113.

[37]Narita M, Nuñez S, Heard E, et al., 2003. Rb-mediated heterochromatin formation and silencing of E2F target genes during cellular senescence. Cell, 113(6):703-716.

[38]Noori HR, 2014. Hysteresis Phenomena in Biology. Springer, Berlin, Heidelberg, p.35-43.

[39]Oakes CC, Smiraglia DJ, Plass C, et al., 2003. Aging results in hypermethylation of ribosomal DNA in sperm and liver of male rats. Proc Natl Acad Sci USA, 100(4):1775-1780.

[40]Pirrotta V, 2016. The necessity of chromatin: a view in perspective. Cold Spring Harb Perspect Biol, 8(1):a019547.

[41]Reiter RJ, Calvo JR, Karbownik M, et al., 2000. Melatonin and its relation to the immune system and inflammation. Ann N Y Acad Sci, 917:376-386.

[42]Sims RJ 3rd, Belotserkovskaya R, Reinberg D, 2004. Elongation by RNA polymerase II: the short and long of it. Genes Dev, 18(20):2437-2468.

[43]Sims RJ 3rd, Millhouse S, Chen CF, et al., 2007. Recognition of trimethylated histone H3 lysine 4 facilitates the recruitment of transcription postinitiation factors and pre-mRNA splicing. Mol Cell, 28(4):665-676.

[44]Smith E, Shilatifard A, 2013. Transcriptional elongation checkpoint control in development and disease. Genes Dev, 27(10):1079-1088.

[45]Song JK, Rechkoblit O, Bestor TH, et al., 2011. Structure of DNMT1-DNA complex reveals a role for autoinhibition in maintenance DNA methylation. Science, 331(6020):1036-1040.

[46]Steves CJ, Spector TD, Jackson SHD, 2012. Ageing, genes, environment and epigenetics: what twin studies tell us now, and in the future? Age Ageing, 41(5):581-586.

[47]Straussman R, Nejman D, Roberts D, et al., 2009. Developmental programming of CpG island methylation profiles in the human genome. Nat Struct Mol Biol, 16(5):564-571.

[48]Struhl K, 1998. Histone acetylation and transcriptional regulatory mechanisms. Genes Dev, 12(5):599-606.

[49]Sutherland JE, Costa M, 2003. Epigenetics and the environment. Ann N Y Acad Sci, 983(1):151-160.

[50]Takahashi K, Yamanaka S, 2006. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell, 126(4):663-676.

[51]Tchkonia T, Zhu Y, van Deursen J, et al., 2013. Cellular senescence and the senescent secretory phenotype: therapeutic opportunities. J Clin Invest, 123(3):966-972.

[52]Teschendorff AE, West J, Beck S, 2013. Age-associated epigenetic drift: implications, and a case of epigenetic thrift? Hum Mol Genet, 22(R1):R7-R15.

[53]Toyokawa S, Uddin M, Koenen KC, et al., 2012. How does the social environment ‘get into the mind’? Epigenetics at the intersection of social and psychiatric epidemiology. Soc Sci Med, 74(1):67-74.

[54]Weber CM, Henikoff S, 2014. Histone variants: dynamic punctuation in transcription. Genes Dev, 28(7):672-682.

[55]Wysocka J, Swigut T, Xiao H, et al., 2006. A PHD finger of NURF couples histone H3 lysine 4 trimethylation with chromatin remodelling. Nature, 442(7098):86-90.

[56]Yan LJ, Levine RL, Sohal RS, 1997. Oxidative damage during aging targets mitochondrial aconitase. Proc Natl Acad Sci USA, 94(21):11168-11172.

[57]Yang D, Guo XJ, Xie T, et al., 2018. Reactive oxygen species may play an essential role in driving biological evolution: the Cambrian Explosion as an example. J Environ Sci, 63:218-226.

Open peer comments: Debate/Discuss/Question/Opinion

<1>

Please provide your name, email address and a comment





Journal of Zhejiang University-SCIENCE, 38 Zheda Road, Hangzhou 310027, China
Tel: +86-571-87952783; E-mail: cjzhang@zju.edu.cn
Copyright © 2000 - 2024 Journal of Zhejiang University-SCIENCE