Biological effects of radiation on the epigenome

Last updated

Ionizing radiation can cause biological effects which are passed on to offspring through the epigenome. The effects of radiation on cells has been found to be dependent on the dosage of the radiation, the location of the cell in regards to tissue, and whether the cell is a somatic or germ line cell. Generally, ionizing radiation appears to reduce methylation of DNA in cells. [1]

Contents

Ionizing radiation has been known to cause damage to cellular components such as proteins, lipids, and nucleic acids. It has also been known to cause DNA double-strand breaks. Accumulation of DNA double strand breaks can lead to cell cycle arrest in somatic cells and cause cell death. Due to its ability to induce cell cycle arrest, ionizing radiation is used on abnormal growths in the human body such as cancer cells, in radiation therapy. Most cancer cells are fully treated with some type of radiotherapy, however some cells such as stem cell cancer cells show a reoccurrence when treated by this type of therapy. [1]

Radiation exposure in everyday life

Non-ionising radiations, electromagnetic fields (EMF) such as radiofrequency (RF), or power frequency radiation have become very common in everyday life. All of these exist as low frequency radiation which can come from wireless cellular devices or through electrical appliances which induce extremely low frequency radiation (ELF). Exposure to these radioactive frequencies has shown negative affects on the fertility of men by impacting the DNA of the sperm and deteriorating the testes [2] as well as an increased risk of tumor formation in salivary glands. [3] [4] The International Agency for Research on Cancer considers RF electromagnetic fields to be possibly carcinogenic to humans, however the evidence is limited. [5]

Radiation and medical imaging

Advances in medical imaging have resulted in increased exposure of humans to low doses of ionizing radiation. Radiation exposure in pediatrics has been shown to have a greater impact as children's cells are still developing. [2] The radiation obtained from medical imaging techniques is only harmful if consistently targeted multiple times in a short space of time. Safety measures have been introduced in order to limit the exposure of harmful ionizing radiation such as the usage of protective material during the use of these imaging tools. A lower dosage is also used in order to fully rid the possibility of a harmful effect from the medical imaging tools. The National Council on Radiation Protection and Measurements along with many other scientific committees have ruled in favor of continued use of medical imaging as the reward far outweighs the minimal risk obtained from these imaging techniques. If the safety protocols are not followed there is a potential increase in the risk of developing cancer. This is primarily due to the decreased methylation of cell cycle genes, such as those relating to apoptosis and DNA repair. The ionizing radiation from these techniques can cause many other detrimental effects in cells including changes in gene expression and halting the cell cycle. However, these results are extremely unlikely if the proper protocols are followed. [1] [4]

Target theory

Target theory concerns the models of how radiation kills biological cells and is based around two main postulates:

  1. "Radiation is considered to be a sequence of random projectiles;
  2. the components of the cell are considered as the targets bombarded by these projectiles" [6]

Several models have been based around the above two points. From the various proposed models three main conclusions were found:

  1. Physical hits obey a Poisson distribution
  2. Failure of radioactive particles to attack sensitive areas of cells allow for survival of the cell
  3. Cell death is an exponential function of the dose of radiation received as the number of hits received is directly proportional to the radiation dose; all hits are considered lethal [7]

Radiation exposure through ionizing radiation (IR) affects a variety of processes inside of an exposed cell. IR can cause changes in gene expression, disruption of cell cycle arrest, and apoptotic cell death. The extent of how radiation effects cells depends on the type of cell and the dosage of the radiation. Some irradiated cancer cells have been shown to exhibit DNA methylation patterns due to epigenetic mechanisms in the cell. In medicine, medical diagnostic methods such as CT scans and radiation therapy expose the individual to ionizing radiation. Irradiated cells can also induce genomic instability in neighboring un-radiated cells via the bystander effect. Radiation exposure could also occur via many other channels than just ionizing radiation.

The basic ballistic models

The single-target single-hit model

In this model a single hit on a target is sufficient to kill a cell [7] The equation used for this model is as follows:

Where k represents a hit on the cell and m represents the mass of the cell.

The n-target single-hit model

In this model the cell has a number of targets n. A single hit on one target is not sufficient to kill the cell but does disable the target. An accumulation of successful hits on various targets leads to cell death. [7] The equation used for this model is as follows:

Where n represents number of the targets in the cell.

The linear quadratic model

The equation used for this model is as follows: [7]

where αD represents a hit made by a one particle track and βD represents a hit made by a two particle track and S(D) represents the probability of survival of the cell.

The three lambda model

This model showed the accuracy of survival description for higher or repeated doses. [7]

The equation used for this model is as follows:

The linear-quadratic-cubic model

The equation used for this model is as follows: [7]

Sublesions hypothesis models

The repair-misrepair model

This model shows the mean number of lesions before any repair activations in a cell. [7]

The equation used for this model is as follows:

where Uo represents the yield of initially induced lesions, with λ being the linear self-repair coefficient, and T equaling time

The lethal-potentially lethal model

This equation explores the hypothesis of a lesion becoming fatal within a given of time if it is not repair by repair enzymes. [7]

The equation used for this model is as follows:

T is the radiation duration and tr is the available repair time.

The saturable repair model

This model illustrates the efficiency of the repair system decreasing as the dosage of radiation increases. This is due to the repair kinetics becoming increasingly saturated with the increase in radiation dosage. [7]

The equation used for this model is as follows:

n(t) is the number of unrepaired lesions, c(t) is the number of repair molecules or enzymes, k is the proportionality coefficient, and T is the time available for repair.

Cellular environment and radiation hormesis

Radiation hormesis

Hormesis is the hypothesis that low levels of disrupting stimulus can cause beneficial adaptations in an organism. [8] The ionizing radiation stimulates repair proteins that are usually not active. Cells use this new stimuli to adapt to the stressors they are being exposed to. [9]

Radiation-Induced Bystander Effect (RIBE)

In biology, the bystander effect is described as changes to nearby non-targeted cells in response to changes in an initially targeted cell by some disrupting agent. [10] In the case of Radiation-Induced Bystander Effect, the stress on the cell is caused by ionizing radiation.

The bystander effect can be broken down into two categories, long range bystander effect and short range bystander effect. In long range bystander effect, the effects of stress are seen further away from the initially targeted cell. In short range bystander, the effects of stress are seen in cells adjacent to the target cell. [10]

Both low linear energy transfer and high linear energy transfer photons have been shown to produce RIBE. Low linear energy transfer photons were reported to cause increases in mutagenesis and a reduction in the survival of cells in clonogenic assays. X-rays and gamma rays were reported to cause increases in DNA double strand break, methylation, and apoptosis. [10] Further studies are needed to reach a conclusive explanation of any epigenetic impact of the bystander effect.

Radiation and oxidative stress

Formation of ROS

Ionizing radiation produces fast moving particles which have the ability to damage DNA, and produce highly reactive free radicals known as reactive oxygen species (ROS). The production of ROS in cells radiated by LDIR (Low-Dose Ionizing Radiation) occur in two ways, by the radiolysis of water molecules or the promotion of nitric oxide synthesis (NOS) activity. The resulting nitric oxide formation reacts with superoxide radicals. This generates peroxynitrite which is toxic to biomolecules. Cellular ROS is also produced with the help of a mechanism involving nicotinamide adenosine dinucleotide phosphate (NADPH) oxidase. NADPH oxidase helps with the formation of ROS by generating a superoxide anion by transferring electrons from cytosolic NADPH across the cell membrane to the extracellular molecular oxygen. This process increases the potential for leakage of electrons and free radicals from the mitochondria. The exposure to the LDIR induces electron release from the mitochondria resulting in more electrons contributing to the superoxide formation in the cells.

The production of ROS in high quantity in cells results in the degradation of biomolecules such as proteins, DNA, and RNA. In one such instance the ROS are known to create double stranded and single stranded breaks in the DNA. This causes the DNA repair mechanisms to try to adapt to the increase in DNA strand breaks. Heritable changes to the DNA sequence have been seen although the DNA nucleotide sequence seems the same after the exposure with LDIR. [11]

Activation of NOS

The formation of ROS is coupled with the formation of nitric oxide synthase activity (NOS). NO reacts with O2 generating peroxynitrite. The increase in the NOS activity causes the production of peroxynitrite (ONOO-). Peroxynitrite is a strong oxidant radical and it reacts with a wide array of biomolecules such as DNA bases, proteins and lipids. Peroxynitrite affects biomolecules function and structure and therefore effectively destabilizes the cell. [11]

Mechanism of oxidative stress and epigenetic gene regulation

Ionizing radiation causes the cell to generate increased ROS and the increase of this species damages biological macromolecules. In order to compensate for this increased radical species, cells adapt to IR induced oxidative effects by modifying the mechanisms of epigenetic gene regulation. There are 4 epigenetic modifications that can take place:

  1. formation of protein adducts inhibiting epigenetic regulation
  2. alteration of genomic DNA methylation status
  3. modification of post translational histone interactions affecting chromatin compaction
  4. modulation of signaling pathways that control transcription factor expression

ROS-mediated protein adduct formation

ROS generated by ionizing radiation chemically modify histones which can cause a change in transcription. Oxidation of cellular lipid components result in an electrophilic molecule formation. The electrophilic molecule binds to the lysine residues of histones causing a ketoamide adduct formation. The ketoamide adduct formation blocks the lysine residues of histones from binding to acetylation proteins thus decreasing gene transcription. [11]

ROS-mediated DNA methylation changes

DNA hypermethylation is seen in the genome with DNA breaks at a gene-specific basis, such as promoters of regulatory genes, but the global methylation of the genome shows a hypomethylation pattern during the period of reactive oxygen species stress. [12]

DNA damage induced by reactive oxygen species results in increased gene methylation and ultimately gene silencing. Reactive oxygen species modify the mechanism of epigenetic methylation by inducing DNA breaks which are later repaired and then methylated by DNMTs. DNA damage response genes, such as GADD45A, recruit nuclear proteins Np95 to direct histone methyltransferase's towards the damaged DNA site. The breaks in DNA caused by the ionizing radiation then recruit the DNMTs in order to repair and further methylate the repair site.

Genome wide hypomethylation occurs due to reactive oxygen species hydroxylating methylcytosines to 5-hydroxymethylcytosine (5hmC). [13] The production of 5hmC serves as an epigenetic marker for DNA damage which is recognizable by DNA repair enzymes. The DNA repair enzymes attracted by the marker convert 5hmC to an unmethylated cytosine base resulting in the hypomethylation of the genome. [14]

Another mechanism that induces hypomethylation is the depletion of S-adenosyl methionine synthetase (SAM). The prevalence of super oxide species causes the oxidization of reduced glutathione (GSH) to GSSG. Due to this, synthesis of the cosubstrate SAM is stopped. SAM is an essential cosubstrate for the normal functioning of DNMTs and histone methyltransferase proteins.

ROS-mediated post-translation modification

Double stranded DNA breaks caused by exposure to ionizing radiation are known to alter chromatin structure. Double stranded breaks are primarily repaired by poly ADP (PAR) polymerases which accumulate at the site of the break leading to activation of the chromatin remodeling protein ALC1. ALC1 causes the nucleosome to relax resulting in the epigenetic up-regulation of genes. A similar mechanism involves the ataxia telangiectasia mutated (ATM) serine/threonine kinase which is an enzyme involved in the repair of double stranded breaks caused by ionizing radiation. ATM phosphorylates KAP1 which causes the heterochromatin to relax, allowing increased transcription to occur.

The DNA mismatch repair gene (MSH2) promoter has shown a hypermethylation pattern when exposed to ionizing radiation. Reactive oxygen species induce the oxidization of deoxyguanosine into 8-hydroxydeoxyguanosine (8-OHdG) causing a change in chromatin structure. Gene promoters that contain 8-OHdG deactivate the chromatin by inducing trimethyl-H3K27 in the genome. Other enzymes such as transglutaminases (TGs) control chromatin remodeling through proteins such as sirtuin1 (SIRT1). TGs cause transcriptional repression during reactive oxygen species stress by binding to the chromatin and inhibiting the sirtuin 1 histone deacetylase from performing its function. [11]

ROS-mediated loss of epigenetic imprinting

Epigenetic imprinting is lost during reactive oxygen species stress. This type of oxidative stress causes a loss of NF- κB signaling. Enhancer blocking element CCCTC-binding factor (CTCF) binds to the imprint control region of insulin-like growth factor 2 (IGF2) preventing the enhancers from allowing the transcription of the gene. The NF- κB proteins interact with IκB inhibitory proteins, but during oxidative stress IκB proteins are degraded in the cell. The loss of IκB proteins for NF- κB proteins to bind to results in NF- κB proteins entering the nucleus to bind to specific response elements to counter the oxidative stress. The binding of NF- κB and corepressor HDAC1 to response elements such as the CCCTC-binding factor causes a decrease in expression of the enhancer blocking element. This decrease in expression hinders the binding to the IGF2 imprint control region therefore causing the loss of imprinting and biallelic IGF2 expression. [11]

Mechanisms of epigenetic modifications

After the initial exposure to ionizing radiation, cellular changes are prevalent in the unexposed offspring of irradiated cells for many cell divisions. One way this non-Mendelian mode of inheritance can be explained is through epigenetic mechanisms. [11]

Ionizing radiation and DNA methylation

Genomic instability via hypomethylation of LINE1

Ionizing radiation exposure affects patterns of DNA methylation. Breast cancer cells treated with fractionated doses of ionizing radiation showed DNA hypomethylation at the various gene loci; dose fractionation refers to breaking down one dose of radiation into separate, smaller doses. [15] Hypomethylation of these genes correlated with decreased expression of various DNMTs and methyl CpG binding proteins. LINE1 transposable elements have been identified as targets for ionizing radiation. The hypomethylation of LINE1 elements results in activation of the elements and thus an increase in LINE1 protein levels. Increased transcription of LINE1 transposable elements results in greater mobilization of the LINE1 loci and therefore increases genomic instability. [11]

Ionizing radiation and histone modification

Irradiated cells can be linked to a variety of histone modifications. Ionizing radiation in breast cancer cell inhibits H4 lysine tri-methylation. Mouse models exposed to high levels of X-ray irradiation exhibited a decrease in both the tri-methylation of H4-Lys20 and the compaction of the chromatin. With the loss of tri-methylation of H4-Lys20, DNA hypomethylation increased resulting in DNA damage and increased genomic instability. [11]

Loss of methylation via repair mechanisms

Breaks in DNA due to ionizing radiation can be repaired. New DNA synthesis by DNA polymerases is one of the ways radiation induced DNA damage can be repaired. However, DNA polymerases do not insert methylated bases which leads to a decrease in methylation of the newly synthesized strand. Reactive oxygen species also inhibit DNMT activity which would normally add the missing methyl groups. This increases the chance that the demethylated state of DNA will eventually become permanent. [16]

Clinical consequences and applications

MGMT- and LINE1- specific DNA methylation

DNA methylation influences tissue responses to ionizing radiation. Modulation of methylation in the gene MGMT or in transposable elements such as LINE1 could be used to alter tissue responses to ionizing radiation and potentially opening new areas for cancer treatment.

MGMT serves as a prognostic marker in glioblastoma. Hypermethylation of MGMT is associated with the regression of tumors. Hypermethylation of MGMT silences its transcription inhibiting alkylating agents in tumor killing cells. Studies have shown patients who received radiotherapy, but no chemotherapy after tumor extraction, had an improved response to radiotherapy due to the methylation of the MGMT promoter.

Almost all human cancers include hypomethylation of LINE1 elements. Various studies depict that the hypomethylation of LINE1 correlates with a decrease in survival after both chemotherapy and radiotheraphy.

Treatment by DNMT inhibitors

DMNT inhibitors are being explored in the treatment of malignant tumors. Recent in-vitro studies show that DNMT inhibitors can increase the effects of other anti-cancer drugs. Knowledge of in-vivo effect of DNMT inhibitors are still being investigated. The long term effects of the use of DNMT inhibitors are still unknown. [16]

Related Research Articles

<span class="mw-page-title-main">Epigenetics</span> Study of DNA modifications that do not change its sequence

In biology, epigenetics is the study of stable changes in cell function that do not involve alterations in the DNA sequence. The Greek prefix epi- in epigenetics implies features that are "on top of" or "in addition to" the traditional genetic basis for inheritance. Epigenetics most often involves changes that affect the regulation of gene expression, and that persist through cellular division. Such effects on cellular and physiological phenotypic traits may result from external or environmental factors, or be part of normal development. It can also lead to diseases such as cancer.

<span class="mw-page-title-main">DNA repair</span> Cellular mechanism

DNA repair is a collection of processes by which a cell identifies and corrects damage to the DNA molecules that encode its genome. In human cells, both normal metabolic activities and environmental factors such as radiation can cause DNA damage, resulting in tens of thousands of individual molecular lesions per cell per day. Many of these lesions cause structural damage to the DNA molecule and can alter or eliminate the cell's ability to transcribe the gene that the affected DNA encodes. Other lesions induce potentially harmful mutations in the cell's genome, which affect the survival of its daughter cells after it undergoes mitosis. As a consequence, the DNA repair process is constantly active as it responds to damage in the DNA structure. When normal repair processes fail, and when cellular apoptosis does not occur, irreparable DNA damage may occur, including double-strand breaks and DNA crosslinkages. This can eventually lead to malignant tumors, or cancer as per the two hit hypothesis.

<span class="mw-page-title-main">DNA methylation</span> Biological process

DNA methylation is a biological process by which methyl groups are added to the DNA molecule. Methylation can change the activity of a DNA segment without changing the sequence. When located in a gene promoter, DNA methylation typically acts to repress gene transcription. In mammals, DNA methylation is essential for normal development and is associated with a number of key processes including genomic imprinting, X-chromosome inactivation, repression of transposable elements, aging, and carcinogenesis.

Histone methylation is a process by which methyl groups are transferred to amino acids of histone proteins that make up nucleosomes, which the DNA double helix wraps around to form chromosomes. Methylation of histones can either increase or decrease transcription of genes, depending on which amino acids in the histones are methylated, and how many methyl groups are attached. Methylation events that weaken chemical attractions between histone tails and DNA increase transcription because they enable the DNA to uncoil from nucleosomes so that transcription factor proteins and RNA polymerase can access the DNA. This process is critical for the regulation of gene expression that allows different cells to express different genes.

DNA oxidation is the process of oxidative damage of deoxyribonucleic acid. As described in detail by Burrows et al., 8-oxo-2'-deoxyguanosine (8-oxo-dG) is the most common oxidative lesion observed in duplex DNA because guanine has a lower one-electron reduction potential than the other nucleosides in DNA. The one electron reduction potentials of the nucleosides are guanine 1.29, adenine 1.42, cytosine 1.6 and thymine 1.7. About 1 in 40,000 guanines in the genome are present as 8-oxo-dG under normal conditions. This means that >30,000 8-oxo-dGs may exist at any given time in the genome of a human cell. Another product of DNA oxidation is 8-oxo-dA. 8-oxo-dA occurs at about 1/10 the frequency of 8-oxo-dG. The reduction potential of guanine may be reduced by as much as 50%, depending on the particular neighboring nucleosides stacked next to it within DNA.

<span class="mw-page-title-main">Methyltransferase</span> Group of methylating enzymes

Methyltransferases are a large group of enzymes that all methylate their substrates but can be split into several subclasses based on their structural features. The most common class of methyltransferases is class I, all of which contain a Rossmann fold for binding S-Adenosyl methionine (SAM). Class II methyltransferases contain a SET domain, which are exemplified by SET domain histone methyltransferases, and class III methyltransferases, which are membrane associated. Methyltransferases can also be grouped as different types utilizing different substrates in methyl transfer reactions. These types include protein methyltransferases, DNA/RNA methyltransferases, natural product methyltransferases, and non-SAM dependent methyltransferases. SAM is the classical methyl donor for methyltransferases, however, examples of other methyl donors are seen in nature. The general mechanism for methyl transfer is a SN2-like nucleophilic attack where the methionine sulfur serves as the leaving group and the methyl group attached to it acts as the electrophile that transfers the methyl group to the enzyme substrate. SAM is converted to S-Adenosyl homocysteine (SAH) during this process. The breaking of the SAM-methyl bond and the formation of the substrate-methyl bond happen nearly simultaneously. These enzymatic reactions are found in many pathways and are implicated in genetic diseases, cancer, and metabolic diseases. Another type of methyl transfer is the radical S-Adenosyl methionine (SAM) which is the methylation of unactivated carbon atoms in primary metabolites, proteins, lipids, and RNA.

<span class="mw-page-title-main">Histone-modifying enzymes</span> Type of enzymes

Histone-modifying enzymes are enzymes involved in the modification of histone substrates after protein translation and affect cellular processes including gene expression. To safely store the eukaryotic genome, DNA is wrapped around four core histone proteins, which then join to form nucleosomes. These nucleosomes further fold together into highly condensed chromatin, which renders the organism's genetic material far less accessible to the factors required for gene transcription, DNA replication, recombination and repair. Subsequently, eukaryotic organisms have developed intricate mechanisms to overcome this repressive barrier imposed by the chromatin through histone modification, a type of post-translational modification which typically involves covalently attaching certain groups to histone residues. Once added to the histone, these groups elicit either a loose and open histone conformation, euchromatin, or a tight and closed histone conformation, heterochromatin. Euchromatin marks active transcription and gene expression, as the light packing of histones in this way allows entry for proteins involved in the transcription process. As such, the tightly packed heterochromatin marks the absence of current gene expression.

<span class="mw-page-title-main">HBx</span>

HBx is a hepatitis B viral protein. It is 154 amino acids long and interferes with transcription, signal transduction, cell cycle progress, protein degradation, apoptosis and chromosomal stability in the host. It forms a heterodimeric complex with its cellular target protein, and this interaction dysregulates centrosome dynamics and mitotic spindle formation. It interacts with DDB1 redirecting the ubiquitin ligase activity of the CUL4-DDB1 E3 complexes, which are intimately involved in the intracellular regulation of DNA replication and repair, transcription and signal transduction.

<span class="mw-page-title-main">Biomarkers of aging</span> Type of biomarkers

Biomarkers of aging are biomarkers that could predict functional capacity at some later age better than chronological age. Stated another way, biomarkers of aging would give the true "biological age", which may be different from the chronological age.

<span class="mw-page-title-main">Cancer epigenetics</span> Field of study in cancer research

Cancer epigenetics is the study of epigenetic modifications to the DNA of cancer cells that do not involve a change in the nucleotide sequence, but instead involve a change in the way the genetic code is expressed. Epigenetic mechanisms are necessary to maintain normal sequences of tissue specific gene expression and are crucial for normal development. They may be just as important, if not even more important, than genetic mutations in a cell's transformation to cancer. The disturbance of epigenetic processes in cancers, can lead to a loss of expression of genes that occurs about 10 times more frequently by transcription silencing than by mutations. As Vogelstein et al. points out, in a colorectal cancer there are usually about 3 to 6 driver mutations and 33 to 66 hitchhiker or passenger mutations. However, in colon tumors compared to adjacent normal-appearing colonic mucosa, there are about 600 to 800 heavily methylated CpG islands in the promoters of genes in the tumors while these CpG islands are not methylated in the adjacent mucosa. Manipulation of epigenetic alterations holds great promise for cancer prevention, detection, and therapy. In different types of cancer, a variety of epigenetic mechanisms can be perturbed, such as the silencing of tumor suppressor genes and activation of oncogenes by altered CpG island methylation patterns, histone modifications, and dysregulation of DNA binding proteins. There are several medications which have epigenetic impact, that are now used in a number of these diseases.

While the cellular and molecular mechanisms of learning and memory have long been a central focus of neuroscience, it is only in recent years that attention has turned to the epigenetic mechanisms behind the dynamic changes in gene transcription responsible for memory formation and maintenance. Epigenetic gene regulation often involves the physical marking of DNA or associated proteins to cause or allow long-lasting changes in gene activity. Epigenetic mechanisms such as DNA methylation and histone modifications have been shown to play an important role in learning and memory.

Embryonic stem cells are capable of self-renewing and differentiating to the desired fate depending on their position in the body. Stem cell homeostasis is maintained through epigenetic mechanisms that are highly dynamic in regulating the chromatin structure as well as specific gene transcription programs. Epigenetics has been used to refer to changes in gene expression, which are heritable through modifications not affecting the DNA sequence.

Epigenetic regulation of neurogenesis is the role that epigenetics plays in the regulation of neurogenesis.

Epigenetic therapy is the use of drugs or other epigenome-influencing techniques to treat medical conditions. Many diseases, including cancer, heart disease, diabetes, and mental illnesses are influenced by epigenetic mechanisms. Epigenetic therapy offers a potential way to influence those pathways directly.

Epigenetics of physical exercise is the study of epigenetic modifications to the cell genome resulting from physical exercise. Environmental factors, including physical exercise, have been shown to have a beneficial influence on epigenetic modifications. Generally, it has been shown that acute and long-term exercise has a significant effect on DNA methylation, an important aspect of epigenetic modifications.

Neuroepigenetics is the study of how epigenetic changes to genes affect the nervous system. These changes may effect underlying conditions such as addiction, cognition, and neurological development.

Pharmacoepigenetics is an emerging field that studies the underlying epigenetic marking patterns that lead to variation in an individual's response to medical treatment.

Epigenetic effects of smoking concerns how epigenetics contributes to the deleterious effects of smoking. Cigarette smoking has been found to affect global epigenetic regulation of transcription across tissue types. Studies have shown differences in epigenetic markers like DNA methylation, histone modifications and miRNA expression between smokers and non-smokers. Similar differences exist in children whose mothers smoked during pregnancy. These epigenetic effects are thought to be linked to many of negative health effects associated with smoking.

<span class="mw-page-title-main">Epigenetic priming</span> Type of modification to a cells epigenome

Epigenetic priming is the modification to a cell's epigenome whereby specific chromatin domains within a cell are converted from a closed state to an open state, usually as the result of an external biological trigger or pathway, allowing for DNA access by transcription factors or other modification mechanisms. The action of epigenetic priming for a certain region of DNA dictates how other gene regulation mechanisms will be able to act on the DNA later in the cell’s life. Epigenetic priming has been chiefly investigated in neuroscience and cancer research, as it has been found to play a key role in memory formation within neurons and tumor-suppressor gene activation in cancer treatment respectively.

Epigenetics of autoimmune disorders is the role that epigenetics play in autoimmune diseases. Autoimmune disorders are a diverse class of diseases that share a common origin. These diseases originate when the immune system becomes dysregulated and mistakenly attacks healthy tissue rather than foreign invaders. These diseases are classified as either local or systemic based upon whether they affect a single body system or if they cause systemic damage.

References

  1. 1 2 3 Kim, Joong-Gook; Park, Moon-Taek; Heo, Kyu; Yang, Kwang-Mo; Yi, Joo Mi (2013-07-18). "Epigenetics Meets Radiation Biology as a New Approach in Cancer Treatment". International Journal of Molecular Sciences. 14 (7): 15059–15073. doi: 10.3390/ijms140715059 . ISSN   1422-0067. PMC   3742287 . PMID   23873297.
  2. 1 2 Sage, Cindy; Burgio, Ernesto (2018-01-01). "Electromagnetic Fields, Pulsed Radiofrequency Radiation, and Epigenetics: How Wireless Technologies May Affect Childhood Development". Child Development. 89 (1): 129–136. doi:10.1111/cdev.12824. ISSN   1467-8624. PMID   28504324.
  3. de Siqueira, Elisa Carvalho; de Souza, Fabrício Tinoco Alvim; Gomez, Ricardo Santiago; Gomes, Carolina Cavalieri; de Souza, Renan Pedra (2017-01-24). "Does cell phone use increase the chances of parotid gland tumor development? A systematic review and meta-analysis". Journal of Oral Pathology & Medicine. 46 (7): 480–483. doi:10.1111/jop.12531. ISSN   0904-2512. PMID   27935126. S2CID   3680709.
  4. 1 2 "JCDR - Non ionizing radiations, Oral cavity, Salivary gland, Tumour". jcdr.net. Retrieved 2018-05-10.
  5. IARC (2013). Non-Ionizing Radiation, Part 2: Radiofrequency Electromagnetic Fields. IARC Monographs on the Evaluation of Carcinogenic Risks to Humans. Vol. 102. World Health Organisation. ISBN   978-92-832-1325-3.
  6. Summers, William C. "Physics and Genes: Einstein to Delbrück". Creating a Physical Biology:The Three-Man Paper and Early Molecular Biology: 39–68.
  7. 1 2 3 4 5 6 7 8 9 Bodgi, Larry; Canet, Aurelien; Pujo-Menjouet, Laurent; Lesne, Annick; Victor, Jean-Marc; Foray, Nicolas (2016-04-07). "Mathematical models of radiation action on living cells: From the target theory to the modern approaches. A historical and critical review" (PDF). Journal of Theoretical Biology. 394: 93–101. doi:10.1016/j.jtbi.2016.01.018. ISSN   0022-5193. PMID   26807808. S2CID   12664478.
  8. Vaiserman, Alexander M. (2011). "Hormesis and epigenetics: Is there a link?". Ageing Research Reviews. 10 (4): 413–21. doi:10.1016/j.arr.2011.01.004. PMID   21292042. S2CID   13321847.
  9. Kim, Se-A.; Lee, Yu-Mi; Choi, Je-Yong; Jacobs, David R.; Lee, Duk-Hee (2018). "Evolutionarily adapted hormesis-inducing stressors can be a practical solution to mitigate harmful effects of chronic exposure to low dose chemical mixtures". Environmental Pollution. 233: 725–734. doi:10.1016/j.envpol.2017.10.124. PMID   29126094.
  10. 1 2 3 Verma, Neha; Tiku, Ashu Bhan (Jul 2017). "Significance and nature of bystander responses induced by various agents". Mutation Research/Reviews in Mutation Research. 773: 104–121. doi:10.1016/j.mrrev.2017.05.003. PMID   28927522.
  11. 1 2 3 4 5 6 7 8 Tharmalingam, Sujeenthar; Sreetharan, Shayenthiran; Kulesza, Adomas V.; Boreham, Douglas R.; Tai, T. C. (Jul 28, 2017). "Low-Dose Ionizing Radiation Exposure, Oxidative Stress and Epigenetic Programing of Health and Disease". Radiation Research. 188 (4.2): 525–538. Bibcode:2017RadR..188..525T. doi:10.1667/RR14587.1. ISSN   0033-7587. PMID   28753061. S2CID   207253006.
  12. Moran, Bruce; Silva, Romina; Perry, Antoinette S.; Gallagher, William M. (2017-10-23). "Epigenetics of malignant melanoma". Seminars in Cancer Biology. 51: 80–88. doi:10.1016/j.semcancer.2017.10.006. ISSN   1044-579X. PMID   29074395. S2CID   205146352.
  13. Ni, Qihan Wu and Xiaohua (2014-12-31). "ROS-Mediated DNA Methylation Pattern Alterations in Carcinogenesis". Current Drug Targets. 16 (1).
  14. Kafer, Georgia Rose; Li, Xuan; Horii, Takuro; Suetake, Isao; Tajima, Shoji; Hatada, Izuho; Carlton, Peter Mark (Feb 4, 2016). "5-Hydroxymethylcytosine Marks Sites of DNA Damage and Promotes Genome Stability". Cell Reports. 14 (6): 1283–1292. doi: 10.1016/j.celrep.2016.01.035 . hdl: 2433/207261 . ISSN   2211-1247. PMID   26854228.
  15. "NCI Dictionary of Cancer Terms". National Cancer Institute. 2011-02-02. Retrieved 2018-05-10.
  16. 1 2 Miousse, Isabelle R.; Kutanzi, Kristy R.; Koturbash, Igor (Feb 21, 2017). "Effects of Ionizing Radiation on DNA Methylation: From Experimental Biology to Clinical Applications". International Journal of Radiation Biology. 93 (5): 457–469. doi:10.1080/09553002.2017.1287454. ISSN   0955-3002. PMC   5411327 . PMID   28134023.