5,523
Views
45
CrossRef citations to date
0
Altmetric
Extra View

Chemo brain: From discerning mechanisms to lifting the brain fog—An aging connection

&
Pages 1345-1349 | Received 12 May 2017, Accepted 18 May 2017, Published online: 30 Jun 2017

ABSTRACT

Mounting evidence indicates that cancer treatments cause numerous deleterious effects, including central nervous system (CNS) toxicity. Chemotherapy-caused CNS side effects encompass changes in cognitive function, memory, and attention, to name a few. Although chemotherapy treatment-induced side effects occur in 16–75% of all patients, the mechanisms of these effects are not well understood. We have recently proposed a new epigenetic theory of chemo brain and, in a pioneer study, determined that cytotoxic chemotherapy agents induce oxidative DNA damage and affect molecular and epigenetic processes in the brain, and may be associated with brain aging processes.

In this paper, we discuss the implications of chemo brain epigenetic effects and future perspectives, as well as outline potential links with brain aging and future translational research opportunities.

Introduction - neurotoxicity of chemotherapy

Elevated cancer rates have resulted in increased awareness and thus the outpouring of research seeking new ways to improve cancer prevention, achieve effective early detection and precise diagnostics, and, most important of all, develop effective treatment options. Ensuring that cancer patients have the best possible quality of life and suffer minimum side effects from their treatments is of utmost importance. Chemotherapy is a key cancer treatment strategy. The vast majority of cytotoxic chemotherapy agents target rapidly dividing cells, including both cancer cells and normal cells that are growing and dividing. As such, these agents can have numerous toxic side effects, such as hair loss, skin changes, gastro-intestinal syndromes, and dysfunction of the bone marrow, among many other effects.Citation1,2

The brain is the key coordinating organ that is responsible for every function of our bodies. Cancer treatment side effects also manifest in central nervous system (CNS) toxicity.Citation3 Recent research shows that chemotherapy agents are, in fact, more toxic to healthy brain cells than to the cancer cells they were designed to treat.Citation4 Chemotherapeutic drugs cause side effects in the cognitive domains of memory, attention, processing speed, and executive function, and these chemotherapy-induced persistent cognitive dysfunction.Citation5-12 This condition, often described by patients as brain fog, is called “chemo brain”.Citation13 The duration of chemo brain symptoms ranges from short to long,Citation14-16 with around one third of patients reporting side effects for months to as long as 5 to 10 y after the cessation of their treatments.Citation13,17

In breast cancer alone, more than 60 studies have investigated and found various degrees of association between chemotherapy and cognitive impairments.Citation18 Nevertheless, which cognitive domains are most affected and most vulnerable to chemotherapy treatment remain unclear. This knowledge gap is due to the multifactorial nature of the neuropsychological tests used in various clinical studies.Citation19 In a longitudinal study by O'Farrel et al., they found the following 4 cognitive factors that were affected in cases of chemo brain: processing speed, working memory, visual memory, and verbal memory. These test findings agree well with patients' self-reports of experiencing losses in cognitive function.Citation19 At the same time, other studies have found that self-reported cognitive function impairment is weakly correlated with testing performance on neurocognitive tasks.Citation20 However, this dichotomy may suggest that tests of neurocognitive tasks may be not fully accurate in assessing how well patients perform in their everyday lives. Subjective reports of impairment from patients, while providing grounds that issues occur in post-chemotherapy treatment, are based on assignments of cognitive tests that assess a particular cognitive domain. A recent elegant scoping review by Olson and colleaguesCitation21 focused on the comprehensive cognitive assessment of adult cancer chemotherapy patients concluded that while cognitive function is a constant and burning concern of individuals diagnosed with cancer, “additional research is needed to find an objective testing protocol that is more highly correlated with perceived cognitive changes”.Citation21 Nevertheless, current clinical reports do not provide any information on the molecular and cellular changes that go on in the brain and serve as a foundation for cognitive deficits.

New insights into mechanisms of chemo brain

The underlying mechanisms of chemotherapy-related cognitive dysfunction need to be further elucidated.Citation22 Recently, increasing amounts of data have shown that chemotherapy imposes toxic effects on the cellular populations of the CNS.Citation22 Chemotherapy induces oxidative stress and apoptosis, inhibits neuronal proliferation and differentiation, activates microglia, and affects chromatin remodeling, leading to the aberrant expression of neurotrophic proteins in the brains of experimental animals.Citation5-11 These molecular changes are linked to altered neurogenesis and deficits in learning and memory.Citation12,23,24 Furthermore, the frequency and timing of chemo brain occurrence and persistence suggest that its origins may be epigenetic and associated with aberrant global gene expression patterns.Citation25 Epigenetic changes are defined as “meiotically heritable and mitotically stable alterations in gene expression” that “include DNA methylation, histone modification and RNA-associated silencing”.Citation26-28 Epigenetic changes play key roles in brain and behavior.Citation29,30

In a recent pioneer study in Aging (2016)Citation31 we have proposed a new theory of chemo brain in which the mechanisms that underlie the neurotoxic side effects of chemotherapy on the brain are epigenetically regulated and associated with altered gene expression.Citation31 Our analysis focused on the hippocampus and prefrontal cortex (PFC) and was based on their pivotal roles in memory, learning, and executive functions. The PFC is at the foremost section of the frontal lobes. It is involved in “executive functions,” such as decision making, planning and judgment, and working memory. It is also regulates abstract thinking and social behavior.Citation32,33 The PFC undergoes prolonged development and is extensively interconnected with other cortical, subcortical, and brain stem sites.Citation32 The hippocampus is a part of the limbic system and is located within the medial temporal lobe. It regulates several cognitive processes, including spatial navigation and memory processing.Citation34 It plays major roles in the storage of long-term memory and in declarative memory, which concerns things that can be recalled with purpose, such as facts or events.Citation35

We dissected the molecular mechanisms of chemo brain by using a murine model, and we analyzed epigenetic and gene expression changes in the hippocampus and PFC tissues of mice 24 hours and 3 weeks after treatment with cytotoxic chemotherapy agents mitomycin C (MMC) and cyclophosphomade (CPP), 2 agents that have been shown to cause chemo brain; however, the mechanisms of their effects remained elusive.Citation31 Our data showed that MMC and CPP treatments lead to drug-, sex-, and brain region-specific and persistent changes in global gene expression profiles. Overall, gene expression responses were much more profound for MCC than CPP exposure, and they were most prominent in the PFC tissues of female animals 3 weeks after MMC treatment, affecting pathways responsible for oxidative stress and other effects. Mitomycin C treatment caused oxidative stress, accumulation of 8-oxodG, decreased global DNA methylation, and increased DNA hydroxymethylation in the PFC tissues of female animals. The molecular changes caused by MMC exposure persisted for up to 3 weeks and were most pronounced in the PCF tissues of female animals. The results show that the PFCs of females may be more vulnerable than those of males in the long-term because the significant changes observed in females at 3 weeks post-exposure to MMC were not apparent in males. Moreover, the majority of the changes induced by MMC in the PFC tissues of female mice resembled those that occur during aging processes, suggesting that chemotherapy exposures may accelerate brain aging.

Reflections and future perspectives – from mechanisms to aging links

In our pioneer study,Citation31 we used Illumina mRNA profiling technology to determine that chemotherapy exposures cause gene expression changes in rodent brain, although mRNAs constitute only a small portion of cellular RNA makeup. Genome sequencing, as well as recent advances in non-coding RNA biology, has shown that more than 98% of our genes encode RNA molecules that are never translated into proteins.Citation36,37 These non-coding RNAs (ncRNAs) are structurally and functionally diverse, and many of them partake in the regulation of cellular proliferation, differentiation, apoptosis, stress responses, and control of genome stability.Citation38-40,41 Among the large repertoire of cellular ncRNAs, microRNAs and piwi-interacting RNAs are implicated as important players in the regulation of neuronal development and function, aging and neurodegeneration, and a variety of neurologic diseases, such as Alzheimer disease, Parkinson disease, amyotrophic lateral sclerosis, stroke, Huntington's disease, and brain cancers, as reviewed in.Citation42-47 Chemo brain has not been explored in terms of the small ncRNA domain. Future research that examines the effects of chemotherapy on non-coding RNAs in the brain is both interesting and important.

We determined that chemotherapy exposure causes changes in global genome DNA methylation and hydroxymethylation.Citation48 These epigenetic phenomena are essential regulators of gene expression,Citation26,42 49 and are important in health and disease,Citation48 including cognitive regulation, memory and aging.Citation50-52 Our data show the overall net changes in the amount of 5 mC and 5 hmC in the genome but lack details on the genomic distribution and locus specificity of the observed changes. Alterations in DNA methylation have been shown to occur in defined regions.Citation53 Future studies should be conducted to determine the distribution and plasticity of DNA methylation and hydroxymethylation in a quantitative fashion and to correlate genome-wide and promoter-specific DNA methylation and hydroxymethylation patterns with the levels of gene expression.Citation49,54,55 This approach will help analyze the regulation of gene expression by chemotherapy exposure. In addition, looking into the role of transcription factors in the regulation of gene expression responses to chemotherapy drugs would likewise be important, especially in context of brain aging.

Our study focused on the effects of 2 cytotoxic chemotherapy agents, MMC and CPP, on the brain. Notably, CNS side effects have been reported to occur upon exposure to ‘targeted’ chemotherapy drugs, such as proteasome inhibitors (bortezomib), topoisomerase inhibitors, bevacizumab, trastuzumab, and small-molecule tyrosine kinase inhibitors (TKIs), to name a few.Citation3 Among these, bevacizumab is a recombinant monoclonal antibody that blocks angiogenesis by inhibiting vascular endothelial growth factor A. Trastuzumab (i.e., Herceptin) is a monoclonal antibody that interacts with HER2. Gefitinib is one of many oral small-molecule TKIs that block the ErbB-1 receptor.Citation56 The molecular targets of many of these agents are involved in cancer, but they may also be important for brain function. Little is known about the effects of targeted drugs on the brain or on the mechanisms of chemo brain induction by these new targeted chemotherapy agents, as well as any potential pro-aging effects of targeted chemotherapy. While new techniques are being developed to better tailor individual drugs to individual patients with the use of new platforms, such as the OncoFinder algorithm,Citation57-59 conducting individualized predictions of any possible side effects, especially severe ones that involve the CNS, will also be important. Recent modifications to the OncoFinder algorithm allow the personalized screening of nootropic drugs,Citation60 as well as the analysis of the effects of small RNA (MiRImpact) on signaling pathways.Citation61 With thorough animal studies, OncoFinder and MiRImpact may be further developed and enabled to predict possible targeted chemotherapy-induced brain side effects.

Given that chemotherapy exposure leads to molecular epigenetic changes, analyzing neuroanatomical and behavioral post-chemotherapy outcomes is an interesting area for future study. Moreover, our studies and the available data on chemo brain used healthy animal models that, while treated with chemotherapy drugs, lacked one important component—the presence of an actual tumor. Investigating chemo brain in tumor-bearing animals is essential to gain a full understanding of the molecular mechanisms and pathways affected in chemo brain. Chemo brain is hypothesized to manifest itself in tumor-bearing mice and is more pronounced in treated animals than in untreated ones, whereas the presence of a tumor itself also affects molecular networks in the brain.

On another note, the phenomenon of chemo brain has not been fully explored in the aging domain. Chemotherapy may cause changes that lead to neuroinflammation and brain aging.Citation21,62 As highlighted at the recent conference on biomedical innovations for healthy longevity, the mechanism and role of cancer treatment-caused aging-related changes need to be analyzed because it will allow the development of strategies for the prevention and mitigation of treatment-induced neurodegeneration and aging.Citation63 To that effect, a new computational tool, the GeroScope, may be used to determine pro-aging and anti-aging pathways altered by chemotherapy exposures in the brain.Citation64

Even more crucial would be the study of chemo-treatment side effects in adolescents and children. For children in developed countries, cancer is the second most common cause of death after accidents. In Canada, 10,000 children live with cancer today, and 1,500 new cases are diagnosed each year. Among these, leukemia is the most common pediatric cancer, accounting for 30% of all malignancies diagnosed annually in children aged younger than 15 (http://childhoodcancer.ca/education/facts_figures). In 1960, the survival rate of pediatric leukemia patients was very low at about 10%. Nowadays, 80–85% of leukemia patients survive, but many of them suffer debilitating side effects, including severe manifestations of chemo brain, leading to huge losses in productive years of life.Citation65-67 In the future, animal model studies can help shed light on the molecular mechanisms and behavioral repercussions of pediatric chemo brain effects.

One other poorly studied aspect of chemo brain is the possibility of treatments that might reverse, or at least reduce, its manifestations. Such treatments could be based upon strategies devised for rehabilitation after brain injuries in animal models, such as complex housing, exercise, tactile stimulation, and psychomotor stimulants, among others. Moreover, given a link between chemo brain and aging, some of the novel geroprotectors can be explored in the anti-chemo brain domain.Citation68

Preclinical animal model data can serve as a foundation for the research and development of new chemo brain biomarkers. Our studies can be used as a roadmap for the development of tests that will predict sensitivity to radiation and chemo brain side effects. Molecular changes observed in the brain must first be correlated with those observed in blood to find effective biomarkers. The markers (small RNAs or mRNAs) that will be correlated between blood and the brain in animal models may be further explored to determine their usefulness in human studies. Last but not the least, animal models may be used to develop future strategies and interventions that help prevent and mitigate chemo brain.

Disclosure of potential conflicts of interest

No potential conflicts of interest were received.

References

  • Longe JL. Gale Encyclopedia of /cancer: A guide to Cancer and its Treatments, 3rd edtion Detroit, MI: Gale/Cengage Learning, 2013.
  • DeVita VT, Hellman S, Rosenberg SA. Cancer, principles & practice of oncology. Philadelphia, PA: Lippincott Williams & Wilkins, 2005.
  • Soffietti R, Trevisan E, Ruda R. Neurologic complications of chemotherapy and other newer and experimental approaches. Handb Clin Neurol 2014; 121:1199-218; PMID:24365412
  • Han R, Yang YM, Dietrich J, Luebke A, Mayer-Proschel M, Noble M. Systemic 5-fluorouracil treatment causes a syndrome of delayed myelin destruction in the central nervous system. J Biol 2008; 7:12; PMID:18430259; https://doi.org/10.1186/jbiol69
  • Seigers R, Fardell JE. Neurobiological basis of chemotherapy-induced cognitive impairment: a review of rodent research. Neurosci Biobehav Rev 2011; 35:729-41; PMID:20869395; https://doi.org/10.1016/j.neubiorev.2010.09.006
  • Seigers R, Loos M, Van Tellingen O, Boogerd W, Smit AB, Schagen SB. Cognitive impact of cytotoxic agents in mice. Psychopharmacology 2015; 232:17-37; PMID:24894481; https://doi.org/10.1007/s00213-014-3636-9
  • Seigers R, Pourtau L, Schagen SB, van Dam FS, Koolhaas JM, Konsman JP, Buwalda B. Inhibition of hippocampal cell proliferation by methotrexate in rats is not potentiated by the presence of a tumor. Brain Res Bull 2010; 81:472-6; PMID:19828128; https://doi.org/10.1016/j.brainresbull.2009.10.006
  • Seigers R, Schagen SB, Beerling W, Boogerd W, van Tellingen O, van Dam FS, Koolhaas JM, Buwalda B. Long-lasting suppression of hippocampal cell proliferation and impaired cognitive performance by methotrexate in the rat. Behav Brain Res 2008; 186:168-75; PMID:17854921; https://doi.org/10.1016/j.bbr.2007.08.004
  • Seigers R, Schagen SB, Coppens CM, van der Most PJ, van Dam FS, Koolhaas JM, Buwalda B. Methotrexate decreases hippocampal cell proliferation and induces memory deficits in rats. Behav Brain Res 2009; 201:279-84; PMID:19428645; https://doi.org/10.1016/j.bbr.2009.02.025
  • Seigers R, Schagen SB, Van Tellingen O, Dietrich J. Chemotherapy-related cognitive dysfunction: current animal studies and future directions. Brain imaging and behavior 2013; 7:453-9; PMID:23949877; https://doi.org/10.1007/s11682-013-9250-3
  • Seigers R, Timmermans J, van der Horn HJ, de Vries EF, Dierckx RA, Visser L, Schagen SB, van Dam FS, Koolhaas JM, Buwalda B. Methotrexate reduces hippocampal blood vessel density and activates microglia in rats but does not elevate central cytokine release. Behav Brain Res 2010; 207:265-72; PMID:19840821; https://doi.org/10.1016/j.bbr.2009.10.009
  • Christie LA, Acharya MM, Parihar VK, Nguyen A, Martirosian V, Limoli CL. Impaired cognitive function and hippocampal neurogenesis following cancer chemotherapy. Clin Cancer Res 2012; 18:1954-65; PMID:22338017; https://doi.org/10.1158/1078-0432.CCR-11-2000
  • Mitchell T, Turton P. ‘Chemobrain’: concentration and memory effects in people receiving chemotherapy - a descriptive phenomenological study. Eur J Cancer Care 2011; 20:539-48; PMID:21443746; https://doi.org/10.1111/j.1365-2354.2011.01244.x
  • Ahles TA, Saykin AJ, Furstenberg CT, Cole B, Mott LA, Skalla K, Whedon MB, Bivens S, Mitchell T, Greenberg ER, et al. Neuropsychologic impact of standard-dose systemic chemotherapy in long-term survivors of breast cancer and lymphoma. J Clin Oncol 2002; 20:485-93; PMID:11786578; https://doi.org/10.1200/JCO.2002.20.2.485
  • Ahles TA, Saykin AJ, Furstenberg CT, Cole B, Mott LA, Titus-Ernstoff L, Skalla K, Bakitas M, Silberfarb PM. Quality of life of long-term survivors of breast cancer and lymphoma treated with standard-dose chemotherapy or local therapy. J Clin Oncol 2005; 23:4399-405; PMID:15994149; https://doi.org/10.1200/JCO.2005.03.343
  • Ahles TA, Silberfarb PM, Herndon J, 2nd, Maurer LH, Kornblith AB, Aisner J, Perry MC, Eaton WL, Zacharski LL, Green MR, et al. Psychologic and neuropsychologic functioning of patients with limited small-cell lung cancer treated with chemotherapy and radiation therapy with or without warfarin: a study by the Cancer and Leukemia Group B. J Clin Oncol 1998; 16:1954-60; PMID:9586915; https://doi.org/10.1200/JCO.1998.16.5.1954
  • Vardy J, Wefel JS, Ahles T, Tannock IF, Schagen SB. Cancer and cancer-therapy related cognitive dysfunction: an international perspective from the Venice cognitive workshop. Ann Oncol 2008; 19:623-9; https://doi.org/10.1093/annonc/mdm500
  • Wefel JS, Schagen SB. Chemotherapy-related cognitive dysfunction. Curr Neurol Neurosci Rep 2012; 12:267-75; PMID:22453825; https://doi.org/10.1007/s11910-012-0264-9
  • O'Farrell E, MacKenzie J, Collins B. Clearing the air: a review of our current understanding of “chemo fog.” Curr Oncol Rep 2013; 15:260-9; PMID:23483375; https://doi.org/10.1007/s11912-013-0307-7
  • Castellon S, Ganz PA. Neuropsychological studies in breast cancer: in search of chemobrain. Breast Cancer Res and Treat 2009; 116:125-7; PMID:18923899; https://doi.org/10.1007/s10549-008-0211-2
  • Olson K, Hewit J, Slater LG, Chambers T, Hicks D, Farmer A, Grattan K, Steggles S, Kolb B. Assessing cognitive function in adults during or following chemotherapy: a scoping review. Support Care Cancer 2016; 24:3223-34; PMID:27067592
  • Kaiser J, Bledowski C, Dietrich J. Neural correlates of chemotherapy-related cognitive impairment. Cortex 2014; 54:33-50; PMID:24632463; https://doi.org/10.1016/j.cortex.2014.01.010
  • Mustafa S, Walker A, Bennett G, Wigmore PM. 5-Fluorouracil chemotherapy affects spatial working memory and newborn neurons in the adult rat hippocampus. Eur J Neurosci 2008; 28:323-30; PMID:18702703; https://doi.org/10.1111/j.1460-9568.2008.06325.x
  • Briones TL, Woods J. Chemotherapy-induced cognitive impairment is associated with decreases in cell proliferation and histone modifications. BMC Neurosci 2011; 12:124; PMID:22152030; https://doi.org/10.1186/1471-2202-12-124
  • Wang XM, Walitt B, Saligan L, Tiwari AF, Cheung CW, Zhang ZJ. Chemobrain: a critical review and causal hypothesis of link between cytokines and epigenetic reprogramming associated with chemotherapy. Cytokine 2015; 72:86-96; PMID:25573802; https://doi.org/10.1016/j.cyto.2014.12.006
  • Jaenisch R, Bird A. Epigenetic regulation of gene expression: how the genome integrates intrinsic and environmental signals. Nat Genet 2003; 33:245-54; PMID:12610534; https://doi.org/10.1038/ng1089
  • Kovalchuk O. Epigenetic effects of ionizing radiation. In: Jirtle RL, Tyson, F.L., ed. Environmental Epigenomics in Health and Disease: Epigenetics and Disease Origins: Springer Science & Business Media, 2013:99-126
  • Jirtle RL. Epigenetics: How genes and environment interact. In: Jirtle RL, Tyson, F.L., ed. Environmental Epigenomics in Health and Disease: Epigenetics and Disease Origins: Springer Science & Business Media, 2013:3-30
  • Qureshi IA, Mehler MF. An evolving view of epigenetic complexity in the brain. Philos Trans R Soc Lond B Biol Sci 2014; 369; pii: 20130506; PMID:25135967; https://doi.org/10.1098/rstb.2013.0506
  • Mehler MF. Epigenetic principles and mechanisms underlying nervous system functions in health and disease. Prog Neurobiol 2008; 86:305-41; PMID:18940229; https://doi.org/10.1016/j.pneurobio.2008.10.001
  • Kovalchuk A, Rodriguez-Juarez R, Ilnytskyy Y, Byeon B, Shpyleva S, Melnyk S, Pogribny I, Kolb B, Kovalchuk O. Sex-specific effects of cytotoxic chemotherapy agents cyclophosphamide and mitomycin C on gene expression, oxidative DNA damage, and epigenetic alterations in the prefrontal cortex and hippocampus - an aging connection. Aging (Albany NY) 2016; 8:697-711; PMID:27032448; https://doi.org/10.18632/aging.100920
  • Kolb B, Mychasiuk R, Muhammad A, Li Y, Frost DO, Gibb R. Experience and the developing prefrontal cortex. Proc Natl Acad Sci U S A 2012; 109 Suppl 2:17186-93; PMID:23045653; https://doi.org/10.1073/pnas.1121251109
  • Lara AH, Wallis JD. The Role of Prefrontal Cortex in Working Memory: A Mini Review. Front Syst Neurosci 2015; 9:173; PMID:26733825; https://doi.org/10.3389/fnsys.2015.00173
  • de Haan M, Mishkin M, Baldeweg T, Vargha-Khadem F. Human memory development and its dysfunction after early hippocampal injury. Trends Neurosci 2006; 19(R2):374-81; PMID:16750273; https://doi.org/10.1016/j.tins.2006.05.008
  • Kolb B, Whishaw IQ. An introduction to brain and behavior. New York, NY: Worth Publishers, 2014
  • Ponting CP, Belgard TG. Transcribed dark matter: meaning or myth? Hum Mol Genet 2010:ddq362; https://doi.org/10.1093/hmg/ddq362.
  • Stein LD. Human genome: end of the beginning. Nature 2004; 431:915-6; PMID:15496902; https://doi.org/10.1038/431915a
  • Gibb EA, Brown CJ, Lam WL. The functional role of long non-coding RNA in human carcinomas. Mol Cancer 2011; 10:38-55; PMID:21489289; https://doi.org/10.1186/1476-4598-10-38
  • Iorio MV, Croce CM. MicroRNA dysregulation in cancer: diagnostics, monitoring and therapeutics. EMBO Mol Med 2012; 4:143-59; https://doi.org/10.1002/emmm.201100209
  • Koturbash I, Zemp FJ, Pogribny I, Kovalchuk O. Small molecules with big effects: the role of the microRNAome in cancer and carcinogenesis. Mutat Res 2011; 722:94-105; https://doi.org/10.1016/j.mrgentox.2010.05.006
  • Mattick JS, Makunin IV. Non-coding RNA. Hum Mol Genet 2006; 15:R17-R29; PMID:16651366; https://doi.org/10.1093/hmg/ddl046
  • Lardenoije R, Iatrou A, Kenis G, Kompotis K, Steinbusch HW, Mastroeni D, Coleman P, Lemere CA, Hof PR, van den Hove DL, et al. The epigenetics of aging and neurodegeneration. Prog Neurobiol 2015; 131:21-64; PMID:26072273; https://doi.org/10.1016/j.pneurobio.2015.05.002
  • Szafranski K, Abraham KJ, Mekhail K. Non-coding RNA in neural function, disease, and aging. Front Genet 2015; 6:87; PMID:25806046; https://doi.org/10.3389/fgene.2015.00087
  • Iyengar BR, Choudhary A, Sarangdhar MA, Venkatesh KV, Gadgil CJ, Pillai B. Non-coding RNA interact to regulate neuronal development and function. Front Cell Neurosci 2014; 8:47; PMID:24605084; https://doi.org/10.3389/fncel.2014.00047
  • Kovalchuk I, Kovalchuk O. Epigenetics in health and disease. Upper Saddle River, N.J.: FT Press, 2012
  • Koch MW, Metz LM, Kovalchuk O. Epigenetics and miRNAs in the diagnosis and treatment of multiple sclerosis. Trends Mol Med 2013; 19:23-30; PMID:23153574; https://doi.org/10.1016/j.molmed.2012.10.008
  • Baulina NM, Kulakova OG, Favorova OO. MicroRNAs: The Role in Autoimmune Inflammation. Acta Naturae 2016; 8:21-33; PMID:27099782
  • Liang J, Yang F, Zhao L, Bi C, Cai B. Physiological and pathological implications of 5-hydroxymethylcytosine in diseases. Oncotarget 2016; 7(30):48813-31
  • Wen L, Tang F. Genomic distribution and possible functions of DNA hydroxymethylation in the brain. Genomics 2014; 104:341-6; PMID:25205307; https://doi.org/10.1016/j.ygeno.2014.08.020
  • Haberman RP, Quigley CK, Gallagher M. Characterization of CpG island DNA methylation of impairment-related genes in a rat model of cognitive aging. Epigenetics 2012; 7:1008-19; PMID:22869088; https://doi.org/10.4161/epi.21291
  • Xu X. DNA methylation and cognitive aging. Oncotarget 2015; 6:13922-32; PMID:26015403; https://doi.org/10.18632/oncotarget.4215
  • Halder R, Hennion M, Vidal RO, Shomroni O, Rahman RU, Rajput A, Centeno TP, van Bebber F, Capece V, Garcia Vizcaino JC, et al. DNA methylation changes in plasticity genes accompany the formation and maintenance of memory. Nat Neurosci 2016; 19:102-10; PMID:26656643
  • Weber M, Schubeler D. Genomic patterns of DNA methylation: targets and function of an epigenetic mark. Curr Opin Cell Biol 2007; 19:273-80; PMID:17466503; https://doi.org/10.1016/j.ceb.2007.04.011
  • Weber M, Hellmann I, Stadler MB, Ramos L, Paabo S, Rebhan M, Schubeler D. Distribution, silencing potential and evolutionary impact of promoter DNA methylation in the human genome. Nat Genet 2007; 39:457-66; PMID:17334365; https://doi.org/10.1038/ng1990
  • Wilson IM, Davies JJ, Weber M, Brown CJ, Alvarez CE, MacAulay C, Schubeler D, Lam WL. Epigenomics: mapping the methylome. Cell Cycle 2006; 5:155-8; PMID:16397413; https://doi.org/10.4161/cc.5.2.2367
  • Gupta S, El-Rayes BF. Small molecule tyrosine kinase inhibitors in pancreatic cancer. Biologics: targets & therapy 2008; 2:707-15
  • Borisov NM, Terekhanova NV, Aliper AM, Venkova LS, Smirnov PY, Roumiantsev S, Korzinkin MB, Zhavoronkov AA, Buzdin AA. Signaling pathway activation profiles make better markers of cancer than expression of individual genes. Oncotarget 2014; https://doi.org/10.18632/oncotarget.2548
  • Buzdin AA, Zhavoronkov AA, Korzinkin MB, Venkova LS, Zenin AA, Smirnov PY, Borisov NM. Oncofinder, a new method for the analysis of intracellular signaling pathway activation using transcriptomic data. Front Genet 2014; 5:55; PMID:24723936; https://doi.org/10.3389/fgene.2014.00055
  • Lezhnina K, Kovalchuk O, Zhavoronkov AA, Korzinkin MB, Zabolotneva AA, Shegay PV, Sokov DG, Gaifullin NM, Rusakov IG, Aliper AM, et al. Novel robust biomarkers for human bladder cancer based on activation of intracellular signaling pathways. Oncotarget 2014; 5:9022-32; PMID:25296972; https://doi.org/10.18632/oncotarget.2493
  • Jellen LC, Aliper A, Buzdin A, Zhavoronkov A. Screening and personalizing nootropic drugs and cognitive modulator regimens in silico. Front Syst Neurosci 2015; 9:4; PMID:25705179; https://doi.org/10.3389/fnsys.2015.00004
  • Artcibasova AV, Korzinkin MB, Sorokin MI, Shegay PV, Zhavoronkov AA, Gaifullin N, Alekseev BY, Vorobyev NV, Kuzmin DV, Kaprin capital A C, et al. MiRImpact, a new bioinformatic method using complete microRNA expression profiles to assess their overall influence on the activity of intracellular molecular pathways. Cell Cycle 2016; 15:689-98; PMID:27027999; https://doi.org/10.1080/15384101.2016.1147633
  • Wardill HR, Mander KA, Van Sebille YZ, Gibson RJ, Logan RM, Bowen JM, Sonis ST. Cytokine-mediated blood brain barrier disruption as a conduit for cancer/chemotherapy-associated neurotoxicity and cognitive dysfunction. Int J Cancer 2016; 139:2635-45; PMID:27367824; https://doi.org/10.1002/ijc.30252
  • Moskalev A, Anisimov V, Aliper A, Artemov A, Asadullah K, Belsky D, Baranova A, de Grey A, Dixit VD, Debonneuil E, et al. A review of the biomedical innovations for healthy longevity. Aging (Albany NY) 2017; 9:7-25; PMID:28132958; https://doi.org/10.18632/aging.101163
  • Aliper A, Belikov AV, Garazha A, Jellen L, Artemov A, Suntsova M, Ivanova A, Venkova L, Borisov N, Buzdin A, et al. In search for geroprotectors: in silico screening and in vitro validation of signalome-level mimetics of young healthy state. Aging (Albany NY) 2016; 8:2127-52; PMID:27677171; https://doi.org/10.18632/aging.101047
  • Follin C, Erfurth EM, Johansson A, Latt J, Sundgren PC, Osterberg K, Spulber G, Mannfolk P, Bjorkman-Burtscher IM. Impaired brain metabolism and neurocognitive function in childhood leukemia survivors despite complete hormone supplementation in adulthood. Psychoneuroendocrinology 2016; 73:157-65; PMID:27498291; https://doi.org/10.1016/j.psyneuen.2016.07.222
  • Hearps S, Seal M, Anderson V, McCarthy M, Connellan M, Downie P, De Luca C. The relationship between cognitive and neuroimaging outcomes in children treated for acute lymphoblastic leukemia with chemotherapy only: A systematic review. Pediatr Blood Cancer 2016; 64(2):225-233; PMID:27696698
  • Kunin-Batson A, Kadan-Lottick N, Neglia JP. The contribution of neurocognitive functioning to quality of life after childhood acute lymphoblastic leukemia. Psychooncology 2014; 23:692-9; PMID:24497266; https://doi.org/10.1002/pon.3470
  • Moskalev A, Chernyagina E, de Magalhaes JP, Barardo D, Thoppil H, Shaposhnikov M, Budovsky A, Fraifeld VE, Garazha A, Tsvetkov V, et al. Geroprotectors.org: a new, structured and curated database of current therapeutic interventions in aging and age-related disease. Aging (Albany NY) 2015; 7:616-28; PMID:26342919; https://doi.org/10.18632/aging.100799

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.