<<

72 Current and Personalized , 2020, Vol. 17, No. 2 Editorial

EDITORIAL The PharmacoEpiGenetic Connection

Ramón Cacabelos1,*

1International Center of and Genomic Medicine, EuroEspes Biomedical Research Center, 15165, Corunna, Spain

Epigenetics is a discipline that studies heritable changes in expression without structural changes in the DNA sequence. is one of the most rapidly developing fields in the history of biology. The concept of epigenetics has evolved since Waddington defined it in the late 1930s, becoming a multifaceted contextual disci- pline with influence in , speciation, functional , transcriptomics, , , and obviously in species-specific health and disease [1]. Epigenetics plays an important role in phenotypic variation in different species of animal and vegetal kingdom [2]. Epigenetic memory can persist across generations. A stress- induced signal can be transmitted across multiple unexposed generations leading to persistent changes in epigenetic gene regulation [3]. Epigenetic mechanisms contribute to phenotypic variation and disparities in morbidity and mor- tality [4]. Epigenetics acts as an interface between the and the environment, and the mechanistic changes associated with the epigenetic phenomena can also be considered a sophisticated form of intracellular and intercel- lular communication [5]. Epigenetics is an adaptive mechanism of developmental plasticity, a phenomenon of rele- vance in evolutionary biology and human health and disease, which enables organisms to respond to their environ- ment based on previous experience without changes to the underlying sequence [6]. correlates with phenotypes depending upon -specific genetic changes linked to , DNA methyl- ation, histone marks, and miRNA regulation of proteomic and metabolomic processes [7]. Epigenomic modifications are involved in several pathological conditions; of major importance are those related with age and with significant health problems such as cardiovascular disorders, obesity, cancer, inflammatory dis- orders, asthma, allergy, and brain disorders. Pharmaceuticals, pesticides, air pollutants, industrial chemicals, heavy metals, hormones, nutrition, and behavior can change gene expression through a broad array of gene regulatory mechanisms which include regulation of gene translocation, histone modifications, DNA methylation, DNA repair, transcription, RNA stability, alternative RNA splicing, protein degradation, gene copy number, and transposon acti- vation [8]. Epigenetic modifications are reversible and can potentially be targeted by pharmacological and dietary interventions [9]. The effects of ( and ) and their therapeutic outcome in the treatment of a given disease are the result of a network of metabolomic events (genomics-transcriptomics-proteomics) associ- ated with the binomial interaction of a chemical or biological molecule with a living organism. The clusters of currently involved in a pharmacogenomic process include pathogenic, mechanistic, metabolic, transporter, and pleiotropic genes [10]. In practice, the expression of these genes is potentially modifiable (transcriptionally and/or post-transcriptionally) by epigenetic mechanisms which may alter (i) pathogenic events, (ii) - interactions, (iii) drug (phase I and II enzymatic reactions), (iv) drug transport (influx-efflux across membranes and cellular barriers), and (v) pleiotropic events leading to unexpected therapeutic outcomes. The un- derstanding of these mechanisms is the main focus of in order to optimize therapeutics and advance towards [11-16]. Unfortunately, the molecular mechanisms underlying the assembly, function and regulation of the epigenetic machinery are poorly understood, and most information in this regard is fragmented. This restrictive knowledge on epigenetic mechanisms represents an important limitation for defining the fundamentals of pharmacoepigenetics. Furthermore, the number of studies on pharmacogenetics and pharmacoepigenetics of current drugs for the treat- ment of common pathologies is still very limited; however, the available information is shedding light on the bene- fits that these complementary disciplines can provide to physicians and patients for the implementation of an effi- cient personalized medicine [16]. ______*Address correspondence to this author at the International Center of Neuroscience and Genomic Medicine, EuroEspes Biomedical Research Center, 15165, Corunna, Spain; E-mails: [email protected]; www.ramoncacabelos.org.

1875-6913/20 $65.00+.00 © 2020 Bentham Science Publishers Editorial Current Pharmacogenomics and Personalized Medicine, 2020, Vol. 17, No. 2 73

The epigenetic machinery is integrated by a cluster of interconnected elements that, in a coordinated manner, contribute to regulate gene expression at transcriptional and post-transcriptional levels. Classical epigenetic mecha- nisms include DNA methylation, histone modifications, and microRNA (miRNA) regulation. Canonical DNA me- thyltransferases (DNMTs)(DNMT1, DNMT3A and DNMT3B) are responsible for maintaining DNA methylation patterns, whereas DNA demethylation can be produced by at least 3 enzyme families: (i) the ten-eleven transloca- tion (TET) family, mediating the conversion of 5mC into 5hmC; (ii) the AID/APOBEC family, acting as mediators of 5mC or 5hmC deamination; and (iii) the BER (base excision repair) glycosylase family involved in DNA repair. Chromatin remodeling and histone post-translational modifications are under the regulatory control of a pleiad of effectors. Post-translational modifications include methylation, acetylation, ubiquitylation, sumoylation, phosphory- lation, acylation (propionylation, butyrylation, 2-hydroxyisobutyrylation, succinylation, malonylation, glutarylation, crotonylation and β-hydroxybutyrylation), N-Glycosylation and O-GlcNAcylation, chaperonization, glutathionyla- tion, poly ADP-ribosylation, and peroxidation. Histone methylation is catalyzed by histone lysine methyltransferas- es (HKMT) and histone demethylation by histone lysine demethylases. Histone acetylation is catalyzed by 5 fami- lies of histone lysine acetyltransferases (KATs)(KAT2A/GCN5, KAT2B/PCAF, KAT6-8, CREBBP/CBP, EP300). Histone deacetylation is catalyzed by 4 classes of HDACs (class I, II, III, IV). O-GlcNAcylation is controlled by O- linked β-N-acetylglucosamine (O-GlcNAc) transferase (OGT) and the glycoside hydrolase O-GlcNAcase (OGA). Ubiquitination consists of post-translational deposition of ubiquitin on proteins for degradation by proteases, and monoubiquitylation can be reversed by histone deubiquitinases [16]. Regulatory include two major categories: (i)(<200 nucleotides) microRNAs (miRNA), small interfering RNAs (siRNA), small nuclear RNAs (snRNA), piwi-interacting RNAs (piRNA), splice junction-associated RNAs; and (ii) long non-coding RNAs (lncRNAs) (>200 ), present in >8000 loci in the human genome: large intergenic non-coding RNAs (lincRNA), natural antisense transcripts (NATs), non-coding RNA expansion repeats, -associated RNAs (PARs), enhancer RNAs (eRNAs), small activating RNAs (saRNAs, RNAa). Other im- portant elements of the epigenetic machinery are transcription factors, transcriptional repressors, enhancers, trans- posable elements, and thousands of novel proteins involved in gene expression. This complex apparatus controls the genomic expression and orchestrates fecundity, embryo-fetal development, tissue maturation, organ conformation, body homeostasis, and aging. Epigenetic aberrations in the epigenetic machinery can lead to disease and in readers, writers or erasers causing epigenetic Mendelian disorders. Since epigenetics is a reversible process, pharmacoepigenetics opens new avenues for epigenetic drug development and epitherapeutics [16]. Epigenetics is evolving at a very fast step for the past few years, creating great expectations in biology and med- icine. It is very likely that many of our present concepts and interpretations on pathogenesis, molecular diagnosis and therapeutics of current human disorders will change in the near future with the advent of novel epigenetic data. Epigenetics adds complexity, diversity and evolutionary clues to the central dogma of biology by which gene ex- pression entails the flow of genetic information from DNA to RNA and proteins. Epigenetic states help to shape differential utilization of genetic information and to preserve a long-lived memory of past signals [17]. The development of new techniques and procedures for epigenome editing and for simplification and interpreta- tion of results is necessary for a rapid translation of epigenetic knowledge into the clinic and for drug development. The completion of genome, epigenome, and mapping demands precision biomolecular tools for DNA manipulation, chromatin structure reconstruction, and reshaping of gene expression patterns. Some molecular plat- forms for epigenetic editing have been developed [18]. Massive DNA has generated a large body of ge- nomic, transcriptomic and epigenomic information that has provided clues for establishing three-dimensional (3D) genomic landscapes in various cells and tissues [19]. Epigenetics challenges conventional understanding of gene-environment interaction and intergenerational inher- itance and, probably, it might contribute to change some modern political ideologies and/or philosophical beliefs (atomistic individualism) [20]. Experiences of racial discrimination have been associated with poor health out- comes. Significant epigenetic associations between disease-associated genes and perceived discrimination, as meas- ured by the Major Life Discrimination (MLD) Scale, have been reported. Consequently, future health disparities research should include epigenetics in high-risk populations to elucidate functional consequences induced by the psychosocial environment [21]. Advances in the field of epigenetics will contribute to apply this knowledge in environmental health risk assess- ment [22]. Pathogens pose serious threats to human health, agricultural investment, and biodiversity conservation through the emergence of zoonoses, spillover to domestic livestock, and epizootic outbreaks. Understanding epige- netics of wildlife disease will enable more accurate risk assessment, reconstruction of transmission pathways, dis- 74 Current Pharmacogenomics and Personalized Medicine, 2020, Vol. 17, No. 2 Editorial cernment of optimal intervention strategies, and development of more effective and ecologically sound treatments in order to minimize damage to the host population and the environment [23]. Despite significant technological advances for epigenetic profiling, there is still a need for a systematic under- standing of how epigenetics shapes cellular circuitry and disease pathogenesis. The development of accurate com- putational approaches for analyzing complex epigenetic profiles is essential for disentangling the mechanisms un- derlying cellular development, and the intricate interaction networks determining and sensing chromatin modifica- tions and DNA methylation to control gene expression. Computational epigenetics is an essential aid for the imple- mentation of epigenetic procedures in the clinical setting and pharmacoepigenetics [24]. The physicians of the 21st century have to adapt their mentality and aptitude to understand new concepts, new in- terpretations of disease pathology, new for an early (or presymptomatic) diagnosis, and new strategies for personalized medicine in order to efficiently serve their patients and preserve the health conditions of the popu- lation [16,25,26].

REFERENCES [1] Nicoglou A, Merlin F. Epigenetics: A way to bridge the gap between biological fields. Stud Hist Philos Biol Biomed Sci 2017; 66: 73-82. https://doi.org/10.1016/j.shpsc.2017.10.002. [2] Ma K, Sun L, Cheng T, Pan H, Wang J, Zhang Q. Epigenetic variance, performing cooperative structure with , is associated with leaf shape traits in widely distributed populations of ornamental Tree Prunus mume. Front Plant Sci 2018; 9: 41. https://doi.org/10.3389/fpls.2018.00041. eCollection 2018. [3] Morgado L, Preite V, Oplaat C, Anava S, Ferreira de Carvalho J, Rechavi O, Johannes F, J.F. Verhoeven K. Small RNAs reflect grandparental environments in apomictic dandelion. Mol Biol Evol 2017; 34(8) :2035-2040. https://doi.org/10.1093/molbev/msx150. [4] Vick AD, Burris HH. Epigenetics and health disparities. Curr Epidemiol Rep. 2017; 4(1): 31-37. https://doi.org/10.1007/s40471-017- 0096-x. [5] Huang B, Jiang C, Zhang R. Epigenetics: The language of the ? . 2014; 6(1): 73-88. https://doi.org/10.2217/epi.13.72. [6] Laubach ZM, Perng W, Dolinoy DC, et al. Epigenetics and the maintenance of developmental plasticity: Extending the signalling theory framework. Biol Rev Camb Philos Soc 2018; 93(3): 1323-1338. https://doi.org/10.1111/brv.12396. [7] Cheung WA, Shao X, Morin A, Siroux V, Kwan T, Ge B, Aïssi D, Chen L, Vasquez L, Allum F, Guénard F, Bouzigon E, Simon MM, Boulier E, Redensek A, Watt S, Datta A, Clarke L, Flicek P, Mead D, Paul DS, Beck S, Bourque G, Lathrop M, Tchernof A, Vohl MC, Demenais F, Pin I, Downes K, Stunnenberg HG, Soranzo N, Pastinen T, Grundberg E. Functional variation in allelic methylomes underscores a strong genetic contribution and reveals novel epigenetic alterations in the human epigenome. Genome Biol 2017; 18(1): 50. https://doi.org/10.1186/s13059-017-1173-7. [8] Heyn H, Moran S, Hernando-Herraez I, Sayols S, Gomez A, Sandoval J, Monk D, Hata K, Marques-Bonet T, Wang L, Esteller M. DNA methylation contributes to natural human variation. Genome Res 2013;23:1363-72. https://doi.org/10.1101/gr.154187.112 [9] Kubota T, Takae H, Miyake K. Epigenetic mechanisms and therapeutic perspectives for neurodevelopmental disorders. Pharmaceuti- cals (Basel) 2012; 5: 369-83. https://doi.org/10.3390/ph5040369 [10] Cacabelos R, Cacabelos P, Torrellas C, Tellado I, Carril JC. Pharmacogenomics of Alzheimer's disease: Novel therapeutic strategies for drug development. Methods Mol Biol 2014;1175:323-556. https://doi.org/10.1007/978-1-4939-0956-8_13 [11] Cacabelos R. Epigenomic networking in drug development: From pathogenic mechanisms to pharmacogenomics. Drug Dev Res 2014; 75: 348-65. https://doi.org/10.1002/ddr.21219 [12] Rasool M, Malik A, Naseer MI, Manan A, Ansari SA, Begum I, Qazi MH, Pushparaj PN, Abuzenadah AM, Al-Qahtani MH, Kamal MA, Pushparaj PN, Gan SH. The role of epigenetics in personalized medicine: Challenges and opportunities. BMC Med Genomics 2015; 8 Suppl 1:S5. https://doi.org/10.1186/1755-8794-8-S1-S5. [13] Cacabelos R, Torrellas C. Pharmacoepigenomics. In: Medical Epigenetics. T. Tollefsbol (Ed). United States: Elsevier. http://dx.doi.org/10.1016/B978-0-12-803239-8.00032-6. [14] Cacabelos R, Torrellas C. Epigenetics of aging and Alzheimer’s disease: Implications for pharmacogenomics and drug response. Int J Mol Sci 2015; 16: 30483-543. https://doi.org/10.3390/ijms161226236. [15] Cacabelos R. World guide for drug use and pharmacogenomics. EuroEspes Publishing, Corunna, Spain, 2012. [16] Cacabelos R (Ed). Pharmacoepigenetics. Translational Epigenetics. Vol. 10. San Diego: Elsevier/Academic Press 2019. [17] Yung PYK, Elsässer SJ. Evolution of epigenetic chromatin states. Curr Opin Chem Biol 2017; 41: 36-42. https://doi.org/10.1016/j.cbpa.2017.10.001. [18] Waryah CB, Moses C, Arooj M, Blancafort P. Zinc Fingers, TALEs, and CRISPR Systems: A Comparison of Tools for Epigenome Editing. Methods Mol Biol 2018;1767:19-63. https://doi.org/10.1007/978-1-4939-7774-1_2. [19] Ouimette JF, Rougeulle C, Veitia RA. Three-dimensional genome architecture in health and disease. Clin Genet 2018; 95(2): 187- 341. Doi: https://doi.org/10.1111/cge.13219 [20] Robison SK. The political implications of epigenetics. Politics Life Sci 2016;35(2):30-53. Editorial Current Pharmacogenomics and Personalized Medicine, 2020, Vol. 17, No. 2 75

https://doi.org/10.1017/pls.2016.14. [21] de Mendoza VB, Huang Y, Crusto CA, Sun YV, Taylor JY. Perceived racial discrimination and methylation among african amer- ican women in the intergen study. Biol Res Nurs 2018; 20(2): 145-52. https://doi.org/10.1177/1099800417748759. [22] Cote IL, McCullough SD, Hines RN, Vandenberg JJ. Application of epigenetic data in human health risk assessment. Curr Opin Tox- icol 2017; 6: 71-8. https://doi.org/10.1016/j.cotox.2017.09.002. [23] DeCandia AL, Dobson AP, vonHoldt BM. Toward an integrative molecular approach to wildlife disease. Conserv Biol 2018; 32(4):798-807. doi: https://doi.org/10.1111/cobi.13083. [24] Angarica VE, Del Sol A. tools for genome-wide epigenetic research. Adv Exp Med Biol 2017; 978: 489-512. doi: https://doi.org/10.1007/978-3-319-53889-1_25. [25] Guest FL, Guest PC. Point-of-Care testing and personalized medicine for metabolic disorders. Methods Mol Biol 2018; 1735: 105- 114. https://doi.org/10.1007/978-1-4939-7614-0_6. [26] van Karnebeek CDM, Wortmann SB, Tarailo-Graovac M, Langeveld M, Ferreira CR, van de Kamp JM, Hollak CE, Wasserman WW, Waterham HM, Wevers RA, Haack TB, Wanders RJA, Boycott KM. The role of the clinician in the multi- era: Are you ready? J Inherit Metab Dis 2018; 41(3): 571-82. https://doi.org/10.1007/s10545-017-0128-1.