Epigenetics

Source: Wikipedia, the free encyclopedia.
(Redirected from
Epigenetically
)

Epigenetic mechanisms

In

environmental factors, or be part of normal development. They can lead to cancer.[4]

The term also refers to the mechanism of changes: functionally relevant alterations to the

silencer regions of the DNA. These epigenetic changes may last through cell divisions for the duration of the cell's life, and may also last for multiple generations, even though they do not involve changes in the underlying DNA sequence of the organism;[7] instead, non-genetic factors cause the organism's genes to behave (or "express themselves") differently.[8]

One example of an epigenetic change in

blood vessels, etc., by activating some genes while inhibiting the expression of others.[9]

Definitions

The term epigenesis has a generic meaning of "extra growth" that has been used in English since the 17th century.[10] In scientific publications, the term epigenetics started to appear in the 1930s (see Fig. on the right). However, its contemporary meaning emerged only in the 1990s.[11]

Number of patent families and non-patent documents with the term "epigenetic*" by publication year.

A definition of the concept of epigenetic trait as a "stably heritable phenotype resulting from changes in a chromosome without alterations in the DNA sequence" was formulated at a Cold Spring Harbor meeting in 2008,[12] although alternate definitions that include non-heritable traits are still being used widely.[13]

Waddington's canalisation, 1940s

The hypothesis of epigenetic changes affecting the expression of

embryonic development.[16]

When Waddington coined the term, the physical nature of

lowest local elevation.[17] Waddington suggested visualising increasing irreversibility of cell type differentiation as ridges rising between the valleys where the marbles (analogous to cells) are travelling.[18]

In recent times, Waddington's notion of the epigenetic landscape has been rigorously formalized in the context of the

strange attractor) or oscillatory.[20]

Contemporary

Robin Holliday defined in 1990 epigenetics as "the study of the mechanisms of temporal and spatial control of gene activity during the development of complex organisms."[21]

More recent usage of the word in biology follows stricter definitions. As defined by Arthur Riggs and colleagues, it is "the study of mitotically and/or meiotically heritable changes in gene function that cannot be explained by changes in DNA sequence."[22]

The term has also been used, however, to describe processes which have not been demonstrated to be heritable, such as some forms of histone modification. Consequently, there are attempts to redefine "epigenetics" in broader terms that would avoid the constraints of requiring

prions unless they impinge on chromosome function. Such redefinitions however are not universally accepted and are still subject to debate.[23] The NIH "Roadmap Epigenomics Project", which ran from 2008 to 2017, uses the following definition: "For purposes of this program, epigenetics refers to both heritable changes in gene activity and expression (in the progeny of cells or of individuals) and also stable, long-term alterations in the transcriptional potential of a cell that are not necessarily heritable."[24] In 2008, a consensus definition of the epigenetic trait, a "stably heritable phenotype resulting from changes in a chromosome without alterations in the DNA sequence," was made at a Cold Spring Harbor meeting.[12]

The similarity of the word to "genetics" has generated many parallel usages. The "

]

Mechanisms

Covalent modification of either DNA (e.g. cytosine methylation and hydroxymethylation) or of histone proteins (e.g. lysine acetylation, lysine and arginine methylation, serine and threonine phosphorylation, and lysine ubiquitination and sumoylation) play central roles in many types of epigenetic inheritance. Therefore, the word "epigenetics" is sometimes used as a synonym for these processes. However, this can be misleading. Chromatin remodeling is not always inherited, and not all epigenetic inheritance involves chromatin remodeling.[25] In 2019, a further lysine modification appeared in the scientific literature linking epigenetics modification to cell metabolism, i.e. Lactylation[26]

DNA associates with histone proteins to form chromatin.

Because the

transcription states can give rise to epigenetic effects. There are several layers of regulation of gene expression. One way that genes are regulated is through the remodeling of chromatin. Chromatin is the complex of DNA and the histone
proteins with which it associates. If the way that DNA is wrapped around the histones changes, gene expression can change as well. Chromatin remodeling is accomplished through two main mechanisms:

  1. The first way is
    post translational modification
    of the amino acids that make up histone proteins. Histone proteins are made up of long chains of amino acids. If the amino acids that are in the chain are changed, the shape of the histone might be modified. DNA is not completely unwound during replication. It is possible, then, that the modified histones may be carried into each new copy of the DNA. Once there, these histones may act as templates, initiating the surrounding new histones to be shaped in the new manner. By altering the shape of the histones around them, these modified histones would ensure that a lineage-specific transcription program is maintained after cell division.
  2. The second way is the addition of methyl groups to the DNA, mostly at
    5-methylcytosine. 5-Methylcytosine performs much like a regular cytosine, pairing with a guanine in double-stranded DNA. However, when methylated cytosines are present in CpG sites in the promoter and enhancer regions of genes, the genes are often repressed.[27][28] When methylated cytosines are present in CpG sites in the gene body (in the coding region excluding the transcription start site) expression of the gene is often enhanced. Transcription of a gene usually depends on a transcription factor binding to a (10 base or less) recognition sequence at the enhancer that interacts with the promoter region of that gene (Gene expression#Enhancers, transcription factors, mediator complex and DNA loops in mammalian transcription).[29] About 22% of transcription factors are inhibited from binding when the recognition sequence has a methylated cytosine. In addition, presence of methylated cytosines at a promoter region can attract methyl-CpG-binding domain (MBD) proteins. All MBDs interact with nucleosome remodeling and histone deacetylase complexes, which leads to gene silencing. In addition, another covalent modification involving methylated cytosine is its demethylation by TET enzymes. Hundreds of such demethylations occur, for instance, during learning and memory forming events in neurons.[30][31]

There is frequently a reciprocal relationship between DNA methylation and histone lysine methylation.

DNMT3A and DNMT3B and histone methylation so that there is a correlation between the genome-wide distribution of DNA methylation and histone methylation.[33]

Mechanisms of heritability of histone state are not well understood; however, much is known about the mechanism of heritability of DNA methylation state during cell division and differentiation. Heritability of methylation state depends on certain enzymes (such as DNMT1) that have a higher affinity for 5-methylcytosine than for cytosine. If this enzyme reaches a "hemimethylated" portion of DNA (where 5-methylcytosine is in only one of the two DNA strands) the enzyme will methylate the other half. However, it is now known that DNMT1 physically interacts with the protein UHRF1. UHRF1 has been recently recognized as essential for DNMT1-mediated maintenance of DNA methylation. UHRF1 is the protein that specifically recognizes hemi-methylated DNA, therefore bringing DNMT1 to its substrate to maintain DNA methylation.[33]

Some acetylations and some methylations of lysines (symbol K) are activation signals for transcription when present on a nucleosome, as shown in the top figure. Some methylations on lysines or arginine (R) are repression signals for transcription when present on a nucleosome, as shown in the bottom figure. Nucleosomes consist of four pairs of histone proteins in a tightly assembled core region plus up to 30% of each histone remaining in a loosely organized tail[34] (only one tail of each pair is shown). DNA is wrapped around the histone core proteins in chromatin. The lysines (K) are designated with a number showing their position as, for instance (K4), indicating lysine as the 4th amino acid from the amino (N) end of the tail in the histone protein. Methylations [Me], and acetylations [Ac] are common post-translational modifications on the lysines of the histone tails.

Although histone modifications occur throughout the entire sequence, the unstructured N-termini of histones (called histone tails) are particularly highly modified. These modifications include

sumoylation, ribosylation and citrullination. Acetylation is the most highly studied of these modifications. For example, acetylation of the K14 and K9 lysines of the tail of histone H3 by histone acetyltransferase enzymes (HATs) is generally related to transcriptional competence[35]
(see Figure).

One mode of thinking is that this tendency of acetylation to be associated with "active" transcription is biophysical in nature. Because it normally has a positively charged nitrogen at its end, lysine can bind the negatively charged phosphates of the DNA backbone. The acetylation event converts the positively charged amine group on the side chain into a neutral amide linkage. This removes the positive charge, thus loosening the DNA from the histone. When this occurs, complexes like SWI/SNF and other transcriptional factors can bind to the DNA and allow transcription to occur. This is the "cis" model of the epigenetic function. In other words, changes to the histone tails have a direct effect on the DNA itself.[36]

Another model of epigenetic function is the "trans" model. In this model, changes to the histone tails act indirectly on the DNA. For example, lysine acetylation may create a binding site for chromatin-modifying enzymes (or transcription machinery as well). This chromatin remodeler can then cause changes to the state of the chromatin. Indeed, a bromodomain – a protein domain that specifically binds acetyl-lysine – is found in many enzymes that help activate transcription, including the SWI/SNF complex. It may be that acetylation acts in this and the previous way to aid in transcriptional activation.

The idea that modifications act as docking modules for related factors is borne out by

HP1
recruits HP1 to K9 methylated regions. One example that seems to refute this biophysical model for methylation is that tri-methylation of histone H3 at lysine 4 is strongly associated with (and required for full) transcriptional activation (see top Figure). Tri-methylation, in this case, would introduce a fixed positive charge on the tail.

It has been shown that the histone lysine methyltransferase (KMT) is responsible for this methylation activity in the pattern of histones H3 & H4. This enzyme utilizes a catalytically active site called the SET domain (Suppressor of variegation, Enhancer of Zeste, Trithorax). The SET domain is a 130-amino acid sequence involved in modulating gene activities. This domain has been demonstrated to bind to the histone tail and causes the methylation of the histone.[37]

Differing histone modifications are likely to function in differing ways; acetylation at one position is likely to function differently from acetylation at another position. Also, multiple modifications may occur at the same time, and these modifications may work together to change the behavior of the

SIR protein
based silencing of the yeast hidden mating-type loci HML and HMR.

DNA methylation

CpG islands where they remain unmethylated. Epigenetic changes of this type thus have the potential to direct increased frequencies of permanent genetic mutation. DNA methylation patterns are known to be established and modified in response to environmental factors by a complex interplay of at least three independent DNA methyltransferases, DNMT1, DNMT3A, and DNMT3B, the loss of any of which is lethal in mice.[39] In invertebrate of social honey bees, main enzymes are DNMT1 and DNMT3.[40] DNMT1 is the most abundant methyltransferase in somatic cells,[41] localizes to replication foci,[42] has a 10–40-fold preference for hemimethylated DNA and interacts with the proliferating cell nuclear antigen (PCNA).[43]

By preferentially modifying hemimethylated DNA, DNMT1 transfers patterns of methylation to a newly synthesized strand after DNA replication, and therefore is often referred to as the 'maintenance' methyltransferase.[44] DNMT1 is essential for proper embryonic development, imprinting and X-inactivation.[39][45] To emphasize the difference of this molecular mechanism of inheritance from the canonical Watson-Crick base-pairing mechanism of transmission of genetic information, the term 'Epigenetic templating' was introduced.[46] Furthermore, in addition to the maintenance and transmission of methylated DNA states, the same principle could work in the maintenance and transmission of histone modifications and even cytoplasmic (structural) heritable states.[47]

In invertebrates of honey bees, DNA methylation has been studied since the honey bee genome [48] was sequenced in 2006. DNA methylation is associated with alternative splicing and gene regulation based on functional genomic research published in 2013.[49] In addition, DNA methylation is associated with the changes of expression in immune genes when honey bees were under lethal viral infection in a timely manner.[50] Several review papers have been published on the topics of DNA methylation in social insects.[51]

RNA methylation

RNA methylation of N6-methyladenosine (m6A) as the most abundant eukaryotic RNA modification has recently been recognized as an important gene regulatory mechanism.[52]

In invertebrates such as social insects of honey bees, RNA methylation is studied to be a possible epigenetic mechanism underlying aggression via reciprocal crosses.[53]

Histone modifications

Histones H3 and H4 can also be manipulated through demethylation using histone lysine demethylase (KDM). This recently identified enzyme has a catalytically active site called the Jumonji domain (JmjC). The demethylation occurs when JmjC utilizes multiple cofactors to hydroxylate the methyl group, thereby removing it. JmjC is capable of demethylating mono-, di-, and tri-methylated substrates.[54]

Chromosomal regions can adopt stable and heritable alternative states resulting in bistable gene expression without changes to the DNA sequence. Epigenetic control is often associated with alternative

covalent modifications of histones.[55] The stability and heritability of states of larger chromosomal regions are suggested to involve positive feedback where modified nucleosomes recruit enzymes that similarly modify nearby nucleosomes.[56] A simplified stochastic model for this type of epigenetics is found here.[57][58]

It has been suggested that chromatin-based transcriptional regulation could be mediated by the effect of small RNAs.

RNA transcripts

Sometimes a gene, after being turned on, transcribes a product that (directly or indirectly) maintains the activity of that gene. For example,

syncytia or gap junctions are important, RNA may spread directly to other cells or nuclei by diffusion. A large amount of RNA and protein is contributed to the zygote by the mother during oogenesis or via nurse cells, resulting in maternal effect phenotypes. A smaller quantity of sperm RNA is transmitted from the father, but there is recent evidence that this epigenetic information can lead to visible changes in several generations of offspring.[61]

MicroRNAs

MicroRNAs (miRNAs) are members of non-coding RNAs that range in size from 17 to 25 nucleotides. miRNAs regulate a large variety of biological functions in plants and animals.[62] So far, in 2013, about 2000 miRNAs have been discovered in humans and these can be found online in a miRNA database.[63] Each miRNA expressed in a cell may target about 100 to 200 messenger RNAs(mRNAs) that it downregulates.[64] Most of the downregulation of mRNAs occurs by causing the decay of the targeted mRNA, while some downregulation occurs at the level of translation into protein.[65]

It appears that about 60% of human protein coding genes are regulated by miRNAs.

CpG islands,[62] that may be repressed by epigenetic methylation. Transcription from methylated CpG islands is strongly and heritably repressed.[67] Other miRNAs are epigenetically regulated by either histone modifications or by combined DNA methylation and histone modification.[62]

mRNA

In 2011, it was demonstrated that the

sRNAs

gene maps based on their targets in microbial genomes are also constructed.[72]

Long non-coding RNAs

Numerous investigations have demonstrated the pivotal involvement of long non-coding RNAs (lncRNAs) in the regulation of gene expression and chromosomal modifications, thereby exerting significant control over cellular differentiation. These long non-coding RNAs also contribute to genomic imprinting and the inactivation of the X chromosome.[73] In invertebrates such as social insects of honey bees, long non-coding RNAs are detected as a possible epigenetic mechanism via allele-specific genes underlying aggression via reciprocal crosses.[74]

Prions

Prions are infectious forms of proteins. In general, proteins fold into discrete units that perform distinct cellular functions, but some proteins are also capable of forming an infectious conformational state known as a prion. Although often viewed in the context of infectious disease, prions are more loosely defined by their ability to catalytically convert other native state versions of the same protein to an infectious conformational state. It is in this latter sense that they can be viewed as epigenetic agents capable of inducing a phenotypic change without a modification of the genome.[75]

codons, an effect that results in suppression of nonsense mutations in other genes.[78] The ability of Sup35 to form prions may be a conserved trait. It could confer an adaptive advantage by giving cells the ability to switch into a PSI+ state and express dormant genetic features normally terminated by stop codon mutations.[79][80][81][82]

Prion-based epigenetics has also been observed in Saccharomyces cerevisiae.[83]

Molecular basis

Epigenetic changes modify the activation of certain genes, but not the genetic code sequence of DNA.[84] The microstructure (not code) of DNA itself or the associated chromatin proteins may be modified, causing activation or silencing. This mechanism enables differentiated cells in a multicellular organism to express only the genes that are necessary for their own activity. Epigenetic changes are preserved when cells divide. Most epigenetic changes only occur within the course of one individual organism's lifetime; however, these epigenetic changes can be transmitted to the organism's offspring through a process called transgenerational epigenetic inheritance. Moreover, if gene inactivation occurs in a sperm or egg cell that results in fertilization, this epigenetic modification may also be transferred to the next generation.[85]

Specific epigenetic processes include

teratogens, regulation of histone modifications and heterochromatin, and technical limitations affecting parthenogenesis and cloning.[86][87][88]

DNA damage

DNA damage can also cause epigenetic changes.

Parp1 (poly(ADP)-ribose polymerase) and its product poly(ADP)-ribose (PAR) accumulate at sites of DNA damage as part of the repair process.[94] This accumulation, in turn, directs recruitment and activation of the chromatin remodeling protein, ALC1, that can cause nucleosome remodeling.[95] Nucleosome remodeling has been found to cause, for instance, epigenetic silencing of DNA repair gene MLH1.[22][96] DNA damaging chemicals, such as benzene, hydroquinone, styrene, carbon tetrachloride and trichloroethylene, cause considerable hypomethylation of DNA, some through the activation of oxidative stress pathways.[97]

Foods are known to alter the epigenetics of rats on different diets.

isoflavones. In one study, markers for oxidative stress, such as modified nucleotides that can result from DNA damage, were decreased by a 3-week diet supplemented with soy.[102] A decrease in oxidative DNA damage was also observed 2 h after consumption of anthocyanin-rich bilberry (Vaccinium myrtillius L.) pomace extract.[103]

DNA repair

Damage to DNA is very common and is constantly being repaired. Epigenetic alterations can accompany DNA repair of oxidative damage or double-strand breaks. In human cells, oxidative DNA damage occurs about 10,000 times a day and DNA double-strand breaks occur about 10 to 50 times a cell cycle in somatic replicating cells (see DNA damage (naturally occurring)). The selective advantage of DNA repair is to allow the cell to survive in the face of DNA damage. The selective advantage of epigenetic alterations that occur with DNA repair is not clear.[citation needed]

Repair of oxidative DNA damage can alter epigenetic markers

In the steady state (with endogenous damages occurring and being repaired), there are about 2,400 oxidatively damaged guanines that form

8-oxo-2'-deoxyguanosine (8-OHdG) in the average mammalian cell DNA.[104] 8-OHdG constitutes about 5% of the oxidative damages commonly present in DNA.[105] The oxidized guanines do not occur randomly among all guanines in DNA. There is a sequence preference for the guanine at a methylated CpG site (a cytosine followed by guanine along its 5' → 3' direction and where the cytosine is methylated (5-mCpG)).[106] A 5-mCpG site has the lowest ionization potential for guanine oxidation.[citation needed
]

8-hydroxy-2'-deoxyguanosine (8-OHdG), and resulting in a 5mCp-8-OHdG dinucleotide site. The base excision repair enzyme OGG1 targets 8-OHdG and binds to the lesion without immediate excision. OGG1, present at a 5mCp-8-OHdG site recruits TET1 and TET1 oxidizes the 5mC adjacent to the 8-OHdG. This initiates demethylation of 5mC.[107]

Oxidized guanine has mispairing potential and is mutagenic.[108] Oxoguanine glycosylase (OGG1) is the primary enzyme responsible for the excision of the oxidized guanine during DNA repair. OGG1 finds and binds to an 8-OHdG within a few seconds.[109] However, OGG1 does not immediately excise 8-OHdG. In HeLa cells half maximum removal of 8-OHdG occurs in 30 minutes,[110] and in irradiated mice, the 8-OHdGs induced in the mouse liver are removed with a half-life of 11 minutes.[105]

When OGG1 is present at an oxidized guanine within a methylated CpG site it recruits TET1 to the 8-OHdG lesion (see Figure). This allows TET1 to demethylate an adjacent methylated cytosine. Demethylation of cytosine is an epigenetic alteration.[citation needed]

As an example, when human mammary epithelial cells were treated with H2O2 for six hours, 8-OHdG increased about 3.5-fold in DNA and this caused about 80% demethylation of the 5-methylcytosines in the genome.[107] Demethylation of CpGs in a gene promoter by TET enzyme activity increases transcription of the gene into messenger RNA.[111] In cells treated with H2O2, one particular gene was examined, BACE1.[107] The methylation level of the BACE1 CpG island was reduced (an epigenetic alteration) and this allowed about 6.5 fold increase of expression of BACE1 messenger RNA.[citation needed]

While six-hour incubation with H2O2 causes considerable demethylation of 5-mCpG sites, shorter times of H2O2 incubation appear to promote other epigenetic alterations. Treatment of cells with H2O2 for 30 minutes causes the mismatch repair protein heterodimer MSH2-MSH6 to recruit DNA methyltransferase 1 (DNMT1) to sites of some kinds of oxidative DNA damage.[112] This could cause increased methylation of cytosines (epigenetic alterations) at these locations.

Jiang et al.[113] treated HEK 293 cells with agents causing oxidative DNA damage, (potassium bromate (KBrO3) or potassium chromate (K2CrO4)). Base excision repair (BER) of oxidative damage occurred with the DNA repair enzyme polymerase beta localizing to oxidized guanines. Polymerase beta is the main human polymerase in short-patch BER of oxidative DNA damage. Jiang et al.[113] also found that polymerase beta recruited the DNA methyltransferase protein DNMT3b to BER repair sites. They then evaluated the methylation pattern at the single nucleotide level in a small region of DNA including the promoter region and the early transcription region of the BRCA1 gene. Oxidative DNA damage from bromate modulated the DNA methylation pattern (caused epigenetic alterations) at CpG sites within the region of DNA studied. In untreated cells, CpGs located at −189, −134, −29, −19, +16, and +19 of the BRCA1 gene had methylated cytosines (where numbering is from the messenger RNA transcription start site, and negative numbers indicate nucleotides in the upstream promoter region). Bromate treatment-induced oxidation resulted in the loss of cytosine methylation at −189, −134, +16 and +19 while also leading to the formation of new methylation at the CpGs located at −80, −55, −21 and +8 after DNA repair was allowed.

Homologous recombinational repair alters epigenetic markers

At least four articles report the recruitment of DNA methyltransferase 1 (DNMT1) to sites of DNA double-strand breaks.[114][115][116][117] During homologous recombinational repair (HR) of the double-strand break, the involvement of DNMT1 causes the two repaired strands of DNA to have different levels of methylated cytosines. One strand becomes frequently methylated at about 21 CpG sites downstream of the repaired double-strand break. The other DNA strand loses methylation at about six CpG sites that were previously methylated downstream of the double-strand break, as well as losing methylation at about five CpG sites that were previously methylated upstream of the double-strand break. When the chromosome is replicated, this gives rise to one daughter chromosome that is heavily methylated downstream of the previous break site and one that is unmethylated in the region both upstream and downstream of the previous break site. With respect to the gene that was broken by the double-strand break, half of the progeny cells express that gene at a high level and in the other half of the progeny cells expression of that gene is repressed. When clones of these cells were maintained for three years, the new methylation patterns were maintained over that time period.[118]

In mice with a CRISPR-mediated homology-directed recombination insertion in their genome there were a large number of increased methylations of CpG sites within the double-strand break-associated insertion.[119]

Non-homologous end joining can cause some epigenetic marker alterations

Non-homologous end joining (NHEJ) repair of a double-strand break can cause a small number of demethylations of pre-existing cytosine DNA methylations downstream of the repaired double-strand break.[115] Further work by Allen et al.[120] showed that NHEJ of a DNA double-strand break in a cell could give rise to some progeny cells having repressed expression of the gene harboring the initial double-strand break and some progeny having high expression of that gene due to epigenetic alterations associated with NHEJ repair. The frequency of epigenetic alterations causing repression of a gene after an NHEJ repair of a DNA double-strand break in that gene may be about 0.9%.[116]

Techniques used to study epigenetics

Epigenetic research uses a wide range of

DamID) and bisulfite sequencing.[121] Furthermore, the use of bioinformatics methods has a role in computational epigenetics.[121]

Chromatin Immunoprecipitation

Chromatin Immunoprecipitation (ChIP) has helped bridge the gap between DNA and epigenetic interactions.[122] With the use of ChIP, researchers are able to make findings in regards to gene regulation, transcription mechanisms, and chromatin structure.[122]

Fluorescent in situ hybridization

Fluorescent in situ hybridization (FISH) is very important to understand epigenetic mechanisms.[123] FISH can be used to find the location of genes on chromosomes, as well as finding noncoding RNAs.[123][124] FISH is predominantly used for detecting chromosomal abnormalities in humans.[124]

Methylation-sensitive restriction enzymes

Methylation sensitive restriction enzymes paired with PCR is a way to evaluate methylation in DNA - specifically the CpG sites.[125] If DNA is methylated, the restriction enzymes will not cleave the strand.[125] Contrarily, if the DNA is not methylated, the enzymes will cleave the strand and it will be amplified by PCR.[125]

Bisulfite sequencing

Bisulfite sequencing is another way to evaluate DNA methylation. Cytosine will be changed to uracil from being treated with sodium bisulfite, whereas methylated cytosines will not be affected.[125][50][49]

Nanopore sequencing

Certain sequencing methods, such as nanopore sequencing, allow sequencing of native DNA. Native (=unamplified) DNA retains the epigenetic modifications which would otherwise be lost during the amplification step. Nanopore basecaller models can distinguish between the signals obtained for epigenetically modified bases and unaltered based and provide an epigenetic profile in addition to the sequencing result.[126]

Structural inheritance

In ciliates such as Tetrahymena and Paramecium, genetically identical cells show heritable differences in the patterns of ciliary rows on their cell surface. Experimentally altered patterns can be transmitted to daughter cells. It seems existing structures act as templates for new structures. The mechanisms of such inheritance are unclear, but reasons exist to assume that multicellular organisms also use existing cell structures to assemble new ones.[127][128][129]

Nucleosome positioning

Eukaryotic genomes have numerous

protamines). Thus nucleosome positioning is to some degree inheritable. Recent studies have uncovered connections between nucleosome positioning and other epigenetic factors, such as DNA methylation and hydroxymethylation.[131]

Histone variants

Different histone variants are incorporated into specific regions of the genome non-randomly. Their differential biochemical characteristics can affect genome functions via their roles in gene regulation,[132] and maintenance of chromosome structures.[133]

Genomic architecture

The three-dimensional configuration of the genome (the 3D genome) is complex, dynamic and crucial for regulating genomic function and nuclear processes such as DNA replication, transcription and DNA-damage repair.[134]

Functions and consequences

In the brain

Memory

gene transcription that create and renew memory in neurons.[31]

An event can set off a chain of reactions that result in altered methylations of a large set of genes in neurons, which give a representation of the event, a memory.[31]

including medial prefrontal cortex (mPFC)

Areas of the brain important in the formation of memories include the hippocampus, medial prefrontal cortex (mPFC), anterior cingulate cortex and amygdala, as shown in the diagram of the human brain in this section.[135]

When a strong memory is created, as in a rat subjected to

immediate early genes (IEGs) that are important in memory formation, allowing their expression in mRNA, with peak mRNA transcription at seven to ten minutes after CFC.[136][137]

Two important IEGs in memory formation are EGR1[138] and the alternative promoter variant of DNMT3A, DNMT3A2.[139] EGR1 protein binds to DNA at its binding motifs, 5′-GCGTGGGCG-3′ or 5′-GCGGGGGCGG-3', and there are about 12,000 genome locations at which EGR1 protein can bind.[140] EGR1 protein binds to DNA in gene promoter and enhancer regions. EGR1 recruits the demethylating enzyme TET1 to an association, and brings TET1 to about 600 locations on the genome where TET1 can then demethylate and activate the associated genes.[140]

Cytosine and 5-methylcytosine

The DNA methyltransferases DNMT3A1, DNMT3A2 and DNMT3B can all methylate cytosines (see image this section) at CpG sites in or near the promoters of genes. As shown by Manzo et al.,[141] these three DNA methyltransferases differ in their genomic binding locations and DNA methylation activity at different regulatory sites. Manzo et al. located 3,970 genome regions exclusively enriched for DNMT3A1, 3,838 regions for DNMT3A2 and 3,432 regions for DNMT3B. When DNMT3A2 is newly induced as an IEG (when neurons are activated), many new cytosine methylations occur, presumably in the target regions of DNMT3A2. Oliviera et al.[139] found that the neuronal activity-inducible IEG levels of Dnmt3a2 in the hippocampus determined the ability to form long-term memories.

Rats form long-term associative memories after contextual fear conditioning (CFC).[142] Duke et al.[30] found that 24 hours after CFC in rats, in hippocampus neurons, 2,097 genes (9.17% of the genes in the rat genome) had altered methylation. When newly methylated cytosines are present in CpG sites in the promoter regions of genes, the genes are often repressed, and when newly demethylated cytosines are present the genes may be activated.[143] After CFC, there were 1,048 genes with reduced mRNA expression and 564 genes with upregulated mRNA expression. Similarly, when mice undergo CFC, one hour later in the hippocampus region of the mouse brain there are 675 demethylated genes and 613 hypermethylated genes.[144] However, memories do not remain in the hippocampus, but after four or five weeks the memories are stored in the anterior cingulate cortex.[145] In the studies on mice after CFC, Halder et al.[144] showed that four weeks after CFC there were at least 1,000 differentially methylated genes and more than 1,000 differentially expressed genes in the anterior cingulate cortex, while at the same time the altered methylations in the hippocampus were reversed.

The epigenetic alteration of methylation after a new memory is established creates a different pool of nuclear mRNAs. As reviewed by Bernstein,[31] the epigenetically determined new mix of nuclear mRNAs are often packaged into neuronal granules, or messenger RNP, consisting of mRNA, small and large ribosomal subunits, translation initiation factors and RNA-binding proteins that regulate mRNA function. These neuronal granules are transported from the neuron nucleus and are directed, according to 3′ untranslated regions of the mRNA in the granules (their "zip codes"), to neuronal dendrites. Roughly 2,500 mRNAs may be localized to the dendrites of hippocampal pyramidal neurons and perhaps 450 transcripts are in excitatory presynaptic nerve terminals (dendritic spines). The altered assortments of transcripts (dependent on epigenetic alterations in the neuron nucleus) have different sensitivities in response to signals, which is the basis of altered synaptic plasticity. Altered synaptic plasticity is often considered the neurochemical foundation of learning and memory.

Aging

Epigenetics play a major role in

brain aging and age-related cognitive decline, with relevance to life extension.[146][147][148][149][150]

Other and general

In adulthood, changes in the

cognitive processes.[152]

Early events, including during

embryonic development, can influence development, cognition, and health outcomes through epigenetic mechanisms.[153]

Epigenetic mechanisms have been proposed as "a potential molecular mechanism for effects of endogenous hormones on the organization of developing brain circuits".[154]

Nutrients could interact with the epigenome to "protect or boost cognitive processes across the lifespan".[155][156]

A review suggests neurobiological effects of physical exercise via epigenetics seem "central to building an 'epigenetic memory' to influence long-term brain function and behavior" and may even be heritable.[157]

With the axo-ciliary

plasma membrane (and longer-term).[158][159]

Epigenetics also play a major role in the brain evolution in and to humans.[160]

Development

Developmental epigenetics can be divided into predetermined and probabilistic epigenesis. Predetermined epigenesis is a unidirectional movement from structural development in DNA to the functional maturation of the protein. "Predetermined" here means that development is scripted and predictable. Probabilistic epigenesis on the other hand is a bidirectional structure-function development with experiences and external molding development.[161]

Somatic epigenetic inheritance, particularly through DNA and histone covalent modifications and

neurogenesis, but mammals are not able to respond to loss of some tissues, for example, the inability to regenerate limbs, which some other animals are capable of. Epigenetic modifications regulate the transition from neural stem cells to glial progenitor cells (for example, differentiation into oligodendrocytes is regulated by the deacetylation and methylation of histones.[162] Unlike animals, plant cells do not terminally differentiate, remaining totipotent with the ability to give rise to a new individual plant. While plants do utilize many of the same epigenetic mechanisms as animals, such as chromatin remodeling, it has been hypothesized that some kinds of plant cells do not use or require "cellular memories", resetting their gene expression patterns using positional information from the environment and surrounding cells to determine their fate.[163]

Epigenetic changes can occur in response to environmental exposure – for example, maternal dietary supplementation with

Controversial results from one study suggested that traumatic experiences might produce an epigenetic signal that is capable of being passed to future generations. Mice were trained, using foot shocks, to fear a cherry blossom odor. The investigators reported that the mouse offspring had an increased aversion to this specific odor.[168][169] They suggested epigenetic changes that increase gene expression, rather than in DNA itself, in a gene, M71, that governs the functioning of an odor receptor in the nose that responds specifically to this cherry blossom smell. There were physical changes that correlated with olfactory (smell) function in the brains of the trained mice and their descendants. Several criticisms were reported, including the study's low statistical power as evidence of some irregularity such as bias in reporting results.[170] Due to limits of sample size, there is a probability that an effect will not be demonstrated to within statistical significance even if it exists. The criticism suggested that the probability that all the experiments reported would show positive results if an identical protocol was followed, assuming the claimed effects exist, is merely 0.4%. The authors also did not indicate which mice were siblings, and treated all of the mice as statistically independent.[171] The original researchers pointed out negative results in the paper's appendix that the criticism omitted in its calculations, and undertook to track which mice were siblings in the future.[172]

Transgenerational

Epigenetic mechanisms were a necessary part of the evolutionary origin of

cell differentiation.[173][need quotation to verify] Although epigenetics in multicellular organisms is generally thought to be a mechanism involved in differentiation, with epigenetic patterns "reset" when organisms reproduce, there have been some observations of transgenerational epigenetic inheritance (e.g., the phenomenon of paramutation observed in maize). Although most of these multigenerational epigenetic traits are gradually lost over several generations, the possibility remains that multigenerational epigenetics could be another aspect to evolution
and adaptation. As mentioned above, some define epigenetics as heritable.

A sequestered germ line or Weismann barrier is specific to animals, and epigenetic inheritance is more common in plants and microbes. Eva Jablonka, Marion J. Lamb and Étienne Danchin have argued that these effects may require enhancements to the standard conceptual framework of the modern synthesis and have called for an extended evolutionary synthesis.[174][175][176] Other evolutionary biologists, such as John Maynard Smith, have incorporated epigenetic inheritance into population-genetics models[177] or are openly skeptical of the extended evolutionary synthesis (Michael Lynch).[178] Thomas Dickins and Qazi Rahman state that epigenetic mechanisms such as DNA methylation and histone modification are genetically inherited under the control of natural selection and therefore fit under the earlier "modern synthesis".[179]

Two important ways in which epigenetic inheritance can differ from traditional genetic inheritance, with important consequences for evolution, are:

  • rates of epimutation can be much faster than rates of mutation[180]
  • the epimutations are more easily reversible[181]

In plants, heritable DNA methylation mutations are 100,000 times more likely to occur compared to DNA mutations.

PSI+ system can act as a "stop-gap", good enough for short-term adaptation that allows the lineage to survive for long enough for mutation and/or recombination to genetically assimilate the adaptive phenotypic change.[183] The existence of this possibility increases the evolvability
of a species.

More than 100 cases of transgenerational epigenetic inheritance phenomena have been reported in a wide range of organisms, including prokaryotes, plants, and animals.[184] For instance, mourning-cloak butterflies will change color through hormone changes in response to experimentation of varying temperatures.[185]

The filamentous fungus Neurospora crassa is a prominent model system for understanding the control and function of cytosine methylation. In this organism, DNA methylation is associated with relics of a genome-defense system called RIP (repeat-induced point mutation) and silences gene expression by inhibiting transcription elongation.[186]

The

yeast prion PSI is generated by a conformational change of a translation termination factor, which is then inherited by daughter cells. This can provide a survival advantage under adverse conditions, exemplifying epigenetic regulation which enables unicellular organisms to respond rapidly to environmental stress. Prions can be viewed as epigenetic agents capable of inducing a phenotypic change without modification of the genome.[187]

Direct detection of epigenetic marks in microorganisms is possible with

single molecule real time sequencing, in which polymerase sensitivity allows for measuring methylation and other modifications as a DNA molecule is being sequenced.[188] Several projects have demonstrated the ability to collect genome-wide epigenetic data in bacteria.[189][190][191][192]

Epigenetics in bacteria

Escherichia coli bacteria

While epigenetics is of fundamental importance in

Clostridioides difficile, adenine methylation regulates sporulation, biofilm formation and host-adaptation.[196]

Medicine

Epigenetics has many and varied potential medical applications.

mental illness, as well as several other conditions. Some investigators, like Randy Jirtle, Ph.D., of Duke University Medical Center, think epigenetics may ultimately turn out to have a greater role in disease than genetics.[198]

Twins

Direct comparisons of identical twins constitute an optimal model for interrogating

histones H3 and H4.[199]

Dizygotic (fraternal) and monozygotic (identical) twins show evidence of epigenetic influence in humans.[199][200][201] DNA sequence differences that would be abundant in a singleton-based study do not interfere with the analysis. Environmental differences can produce long-term epigenetic effects, and different developmental monozygotic twin subtypes may be different with respect to their susceptibility to be discordant from an epigenetic point of view.[202]

A high-throughput study, which denotes technology that looks at extensive genetic markers, focused on epigenetic differences between monozygotic twins to compare global and locus-specific changes in DNA methylation and histone modifications in a sample of 40 monozygotic twin pairs.[199] In this case, only healthy twin pairs were studied, but a wide range of ages was represented, between 3 and 74 years. One of the major conclusions from this study was that there is an age-dependent accumulation of epigenetic differences between the two siblings of twin pairs. This accumulation suggests the existence of epigenetic "drift". Epigenetic drift is the term given to epigenetic modifications as they occur as a direct function with age. While age is a known risk factor for many diseases, age-related methylation has been found to occur differentially at specific sites along the genome. Over time, this can result in measurable differences between biological and chronological age. Epigenetic changes have been found to be reflective of lifestyle and may act as functional biomarkers of disease before clinical threshold is reached.[203]

A more recent study, where 114 monozygotic twins and 80 dizygotic twins were analyzed for the DNA methylation status of around 6000 unique genomic regions, concluded that epigenetic similarity at the time of blastocyst splitting may also contribute to phenotypic similarities in monozygotic co-twins. This supports the notion that

microenvironment at early stages of embryonic development can be quite important for the establishment of epigenetic marks.[200]
Congenital genetic disease is well understood and it is clear that epigenetics can play a role, for example, in the case of
hemizygous because of genomic imprinting, and therefore a single gene knock out is sufficient to cause the disease, where most cases would require both copies to be knocked out.[204]

Genomic imprinting

Some human disorders are associated with genomic imprinting, a phenomenon in mammals where the father and mother contribute different epigenetic patterns for specific genomic loci in their

germ cells.[205] The best-known case of imprinting in human disorders is that of Angelman syndrome and Prader–Willi syndrome – both can be produced by the same genetic mutation, chromosome 15q partial deletion, and the particular syndrome that will develop depends on whether the mutation is inherited from the child's mother or from their father.[206] This is due to the presence of genomic imprinting in the region. Beckwith–Wiedemann syndrome is also associated with genomic imprinting, often caused by abnormalities in maternal genomic imprinting of a region on chromosome 11.[citation needed
]

Methyl CpG-binding protein 2 (

BDNF is downregulated in the MECP2 mutant resulting in Rett syndrome, as well as the increase of early neural senescence and accumulation of damaged DNA.[207]

In the Överkalix study, paternal (but not maternal) grandsons[208] of Swedish men who were exposed during preadolescence to famine in the 19th century were less likely to die of cardiovascular disease. If food was plentiful, then diabetes mortality in the grandchildren increased, suggesting that this was a transgenerational epigenetic inheritance.[209] The opposite effect was observed for females – the paternal (but not maternal) granddaughters of women who experienced famine while in the womb (and therefore while their eggs were being formed) lived shorter lives on average.[210]

Diabetic wound healing

Epigenetic modifications have given insight into the understanding of the pathophysiology of different disease conditions. Though, they are strongly associated with cancer, their role in other pathological conditions are of equal importance. It appears that the

Macrophage Polarization is mostly associated with all the diabetic complications in a clinical set-up. As of 2018, several reports reveal the relevance of different epigenetic modifications with respect to diabetic complications. Sooner or later, with the advancements in biomedical tools, the detection of such biomarkers as prognostic and diagnostic tools in patients could possibly emerge out as alternative approaches. It is noteworthy to mention here that the use of epigenetic modifications as therapeutic targets warrant extensive preclinical as well as clinical evaluation prior to use.[211]

Examples of drugs altering gene expression from epigenetic events

The use of beta-lactam

antibiotics can alter glutamate receptor activity and the action of cyclosporine on multiple transcription factors. Additionally, lithium can impact autophagy of aberrant proteins, and opioid drugs via chronic use can increase the expression of genes associated with addictive phenotypes.[212]

In a groundbreaking 2003 report, Caspi and colleagues demonstrated that in a robust cohort of over one-thousand subjects assessed multiple times from preschool to adulthood, subjects who carried one or two copies of the short allele of the

suicidality when exposed to childhood maltreatment when compared to long allele homozygotes with equal ELS exposure.[213]

Parental nutrition, in utero exposure to stress or endocrine disrupting chemicals,[214] male-induced maternal effects such as the attraction of differential mate quality, and maternal as well as paternal age, and offspring gender could all possibly influence whether a germline epimutation is ultimately expressed in offspring and the degree to which intergenerational inheritance remains stable throughout posterity.[215] However, whether and to what extent epigenetic effects can be transmitted across generations remains unclear, particularly in humans.[216][217]

Addiction

phenotypes has been noted to occur in preclinical studies.[222][223] However, robust evidence in support of the persistence of epigenetic effects across multiple generations has yet to be established in humans; for example, an epigenetic effect of prenatal exposure to smoking that is observed in great-grandchildren who had not been exposed.[216]

Depression

Epigenetic inheritance of depression-related phenotypes has also been reported in a preclinical study.[224] Inheritance of paternal stress-induced traits across generations involved small non-coding RNA signals transmitted via the paternal germline.[citation needed]

SARS-CoV-2

Recent work had demonstrated that SARS-CoV-2 markedly disrupts host cell epigenetic regulation. Viral proteins dampen antiviral responses by mimicking critical regions of human histone proteins, according to the study. The paper showed that SARS-CoV-2 protein encoded by ORF8 (ORF8) functions as a histone mimic of the ARKS motifs in histone H3 to disrupt host cell epigenetic regulation. Histone mimicry allows viruses to disrupt the host cell's ability to regulate gene expression, and respond to infection effectively. The discovery might have implications for the treatment of COVID-19 since SARS-CoV-2, lacking ORF8, is associated with decreased severity of COVID-19.[225]

Research

The two forms of heritable information, namely genetic and epigenetic, are collectively called dual inheritance. Members of the APOBEC/AID family of cytosine deaminases may concurrently influence genetic and epigenetic inheritance using similar molecular mechanisms, and may be a point of crosstalk between these conceptually compartmentalized processes.[226]

dioxygenases that require iron as a co-factor.[227]

Various pharmacological agents are applied for the production of induced pluripotent stem cells (iPSC) or maintain the embryonic stem cell (ESC) phenotypic via epigenetic approach. Adult stem cells like bone marrow stem cells have also shown a potential to differentiate into cardiac competent cells when treated with G9a histone methyltransferase inhibitor BIX01294.[228][229]

Cell plasticity, which is the adaptation of cells to stimuli without changes in their genetic code, requires epigenetic changes. These have been observed in cell plasticity in cancer cells during epithelial-to-mesenchymal transition[230] and also in immune cells, such as macrophages.[231] Interestingly, metabolic changes underly these adaptations, since various metabolites play crucial roles in the chemistry of epigenetic marks. This includes for instance alpha-ketoglutarate, which is required for histone demethylation, and acetyl-Coenzyme A, which is required for histone acetylation.

Epigenome editing

Epigenetic regulation of gene expression that could be altered or used in

histone modification.[232][233][234]

CpG sites, SNPs and biological traits

Methylation is a widely characterized mechanism of genetic regulation that can determine biological traits. However, strong experimental evidences correlate methylation patterns in SNPs as an important additional feature for the classical activation/inhibition epigenetic dogma. Molecular interaction data, supported by colocalization analyses, identify multiple nuclear regulatory pathways, linking sequence variation to disturbances in DNA methylation and molecular and phenotypic variation.[235]

UBASH3B locus

UBASH3B encodes a protein with tyrosine phosphatase activity, which has been previously linked to advanced neoplasia.[236] SNP rs7115089 was identified as influencing DNA methylation and expression of this locus, as well as and Body Mass Index (BMI).[235] In fact, SNP rs7115089 is strongly associated with BMI[237] and with genetic variants linked to other cardiovascular and metabolic traits in GWASs.[238][239][240] New studies suggesting UBASH3B as a potential mediator of adiposity and cardiometabolic disease.[235] In addition, animal models demonstrated that UBASH3B expression is an indicator of caloric restriction that may drive programmed susceptibility to obesity and it is associated with other measures of adiposity in human peripherical blood.[241]

NFKBIE locus

SNP rs730775 is located in the first intron of NFKBIE and is a cis eQTL for NFKBIE in whole blood.[235] Nuclear factor (NF)-κB inhibitor ε (NFKBIE) directly inhibits NF-κB1 activity and is significantly co-expressed with NF-κB1, also, it is associated with rheumatoid arthritis.[242] Colocalization analysis supports that variants for the majority of the CpG sites in SNP rs730775 cause genetic variation at the NFKBIE locus which is suggestible linked to rheumatoid arthritis through trans acting regulation of DNA methylation by NF-κB.[235]

FADS1 locus

Fatty acid desaturase 1 (FADS1) is a key enzyme in the metabolism of fatty acids.[243] Moreover, rs174548 in the FADS1 gene shows increased correlation with DNA methylation in people with high abundance of CD8+ T cells.[235] SNP rs174548 is strongly associated with concentrations of arachidonic acid and other metabolites in fatty acid metabolism,[244][245] blood eosinophil counts.[246] and inflammatory diseases such as asthma.[247] Interaction results indicated a correlation between rs174548 and asthma, providing new insights about fatty acid metabolism in CD8+ T cells with immune phenotypes.[235]

Pseudoscience

As epigenetics is in the early stages of development as a science and is surrounded by

new age authors making unfounded suggestions that a person's genes and health can be manipulated by mind control. Misuse of the scientific term by quack authors has produced misinformation among the general public.[2][248]

See also

References

  1. PMID 19711245
    . In the original sense of this definition, epigenetics referred to all molecular pathways modulating the expression of a genotype into a particular phenotype. Over the following years, with the rapid growth of genetics, the meaning of the word has gradually narrowed. Epigenetics has been defined and today is generally accepted as 'the study of changes in gene function that are mitotically and/or meiotically heritable and that do not entail a change in DNA sequence.'
  2. ^ a b Rutherford A (19 July 2015). "Beware the pseudo gene genies". The Guardian.
  3. PMID 25855649
    .
  4. .
  5. .
  6. .
  7. ^ .
  8. ^ Hunter P (1 May 2008). "What genes remember". Prospect Magazine. Archived from the original on 1 May 2008. Retrieved 26 July 2012.
  9. S2CID 11794102
    .
  10. ^ Oxford English Dictionary: "The word is used by W. Harvey, Exercitationes 1651, p. 148, and in the English Anatomical Exercitations 1653, p. 272. It is explained to mean ‘partium super-exorientium additamentum’, ‘the additament of parts budding one out of another’."
  11. ^ ]
  12. ^ .
  13. ^ a b "Overview". NIH Roadmap Epigenomics Project. Archived from the original on 21 November 2019. Retrieved 7 December 2013.
  14. ^ Morange M. La tentative de Nikolai Koltzoff (Koltsov) de lier génétique, embryologie et chimie physique, J. Biosciences. 2011. V. 36. P. 211-214
  15. ^ Waddington CH (1942). "The epigenotype". Endeavour. 1: 18–20. "For the purpose of a study of inheritance, the relation between phenotypes and genotypes [...] is, from a wider biological point of view, of crucial importance, since it is the kernel of the whole problem of development. Many geneticists have recognized this and attempted to discover the processes involved in the mechanism by which the genes of the genotype bring about phenotypic effects. The first step in such an enterprise is – or rather should be, since it is often omitted by those with undue respect for the powers of reason – to describe what can be seen of the developmental processes. For enquiries of this kind, the word 'phenogenetics' was coined by Haecker [1918, Phänogenetik]. The second and more important part of the task is to discover the causal mechanisms at work and to relate them as far as possible to what experimental embryology has already revealed of the mechanics of development. We might use the name 'epigenetics' for such studies, thus emphasizing their relation to the concepts, so strongly favourable to the classical theory of epigenesis, which have been reached by the experimental embryologists. We certainly need to remember that between genotype and phenotype, and connecting them to each other, there lies a whole complex of developmental processes. It is convenient to have a name for this complex: 'epigenotype' seems suitable."
  16. ^ See preformationism for historical background. Oxford English Dictionary: "the theory that the germ is brought into existence (by successive accretions), and not merely developed, in the process of reproduction. [...] The opposite theory was formerly known as the 'theory of evolution'; to avoid the ambiguity of this name, it is now spoken of chiefly as the 'theory of preformation', sometimes as that of 'encasement' or 'emboîtement'."
  17. ]
  18. .
  19. .
  20. ^ .
  21. .
  22. ^ ]
  23. .
  24. .
  25. .
  26. .
  27. .
  28. .
  29. .
  30. ^ .
  31. ^ .
  32. ^ .
  33. ^ .
  34. .
  35. .
  36. .
  37. .
  38. .
  39. ^ .
  40. .
  41. .
  42. .
  43. .
  44. .
  45. .
  46. .
  47. .
  48. ^ The Honeybee Genome Sequencing Consortium The Honeybee Genome Sequencing Consortium
  49. ^
    PMID 23852726
    .
  50. ^ .
  51. .
  52. ^ Barbieri I, Kouzarides T. Role of RNA modifications in cancer. Nat Rev Cancer. 2020;20(6):303–22.
  53. PMID 37308882
    .
  54. .
  55. .
  56. .
  57. ^ "Epigenetic cell memory". Cmol.nbi.dk. Archived from the original on 30 September 2011. Retrieved 26 July 2012.
  58. S2CID 16091877
    .
  59. ]
  60. .
  61. ^ Choi CQ (25 May 2006). "RNA can be hereditary molecule". The Scientist. Archived from the original on 8 February 2007.
  62. ^
    PMID 23246373
    .
  63. ^ "Browse miRBase by species".
  64. S2CID 4430576
    .
  65. .
  66. .
  67. .
  68. .
  69. ^ "New research links common RNA modification to obesity". Physorg.com. Retrieved 26 July 2012.
  70. PMID 23733475
    .
  71. ^ "sRNATarBase 2.0 A comprehensive database of bacterial SRNA targets verified by experiments". Archived from the original on 26 September 2013.
  72. ^ "Genomics maps for small non-coding RNA's and their targets in microbial genomes". Archived from the original on 8 June 2017. Retrieved 13 August 2013.
  73. ^ Ruffo, Paola, et al. "Long-noncoding RNAs as epigenetic regulators in neurodegenerative diseases." Neural Regeneration Research 18.6 (2023): 1243.
  74. PMID 37308882
    .
  75. .
  76. .
  77. .
  78. .
  79. .
  80. .
  81. .
  82. .
  83. .
  84. .
  85. .
  86. .
  87. .
  88. .
  89. .
  90. .
  91. .
  92. .
  93. .
  94. .
  95. .
  96. .
  97. .
  98. .
  99. .
  100. .
  101. .
  102. .
  103. .
  104. .
  105. ^ .
  106. .
  107. ^ .
  108. .
  109. .
  110. .
  111. .
  112. .
  113. ^ .
  114. .
  115. ^ .
  116. ^ .
  117. .
  118. .
  119. .
  120. .
  121. ^ .
  122. ^ a b "Studying epigenetics using ChIP". Abcam.
  123. ^
    PMID 18951174.{{cite book}}: CS1 maint: location missing publisher (link
    )
  124. ^ a b O'Connor C (2008). "Fluorescence in situ hybridization (FISH)". Nature Education. 1 (1): 171.
  125. ^
    S2CID 26728480
    .
  126. .
  127. .
  128. .
  129. .
  130. .
  131. ^ .
  132. .
  133. .
  134. ^ "The 3D genome". www.nature.com. 2 September 2019. Retrieved 26 September 2021.
  135. PMID 28386011
    .
  136. ^ .
  137. .
  138. .
  139. ^ .
  140. ^ .
  141. .
  142. .
  143. .
  144. ^ .
  145. .
  146. .
  147. .
  148. .
  149. .
  150. .
  151. .
  152. .
  153. .
  154. .
  155. .
  156. .
  157. .
  158. ^ Tamim B (4 September 2022). "New discovery: Synapse hiding in the mice brain may advance our understanding of neuronal communication". interestingengineering.com. Retrieved 19 October 2022.
  159. S2CID 251958800
    .
  160. PMID 22122280
    .
  161. .
  162. ^ Chapter: "Nervous System Development" in "Epigenetics," by Benedikt Hallgrimsson and Brian Hall
  163. PMID 17194589. Archived from the original
    (PDF) on 15 December 2013. This might suggest that plant cells do not use or require a cellular memory mechanism and just respond to positional information. However, it has been shown that plants do use cellular memory mechanisms mediated by PcG proteins in several processes, ... (p. 104)
  164. .
  165. .
  166. .
  167. .
  168. ^ Callaway E (1 December 2013). "Fearful Memories Passed Down to Mouse Descendants: Genetic imprint from traumatic experiences carries through at least two generations". Nature Magazine – via Scientific American.
  169. ^ Le Roux M (13 December 2013). "Mice can 'warn' sons, grandsons of dangers via sperm".
  170. PMID 25316784
    .
  171. . (see comment by Gonzalo Otazu)
  172. ^ "Epigenetics Paper Raises Questions".
  173. .
  174. .
  175. ^ See also Denis Noble: The Music of Life, esp pp. 93–98 and p. 48, where he cites Jablonka & Lamb and Massimo Pigliucci's review of Jablonka and Lamb in Nature 435, 565–566 (2 June 2005)
  176. S2CID 8837202
    .
  177. .
  178. .
  179. .
  180. .
  181. .
  182. .
  183. .
  184. S2CID 7233550. Archived from the original
    (PDF) on 15 July 2011. Retrieved 1 November 2017.
  185. ^ Davies, Hazel (2008). Do Butterflies Bite?: Fascinating Answers to Questions about Butterflies and Moths (Animals Q&A). Rutgers University Press.
  186. PMID 19092133
    .
  187. ^ .
  188. .
  189. .
  190. .
  191. .
  192. ^ Fang G, Munera D, Friedman DI, Mandlik A, Chao MC, Banerjee O, et al. (December 2012). "Genome-wide mapping of methylated adenine residues in pathogenic Escherichia coli using single-molecule real-time sequencing". Nature Biotechnology. 30 (12): 1232–9.
    PMID 23138224
    .
  193. .
  194. .
  195. .
  196. .
  197. .
  198. ^ Beil L (Winter 2008). "Medicine's New Epicenter? Epigenetics: New field of epigenetics may hold the secret to flipping cancer's "off" switch". CURE (Cancer Updates, Research and Education). Archived from the original on 29 May 2009.
  199. ^
    PMID 16009939
    .
  200. ^ .
  201. ^ O'Connor A (11 March 2008). "The Claim: Identical Twins Have Identical DNA". New York Times. Retrieved 2 May 2010.
  202. S2CID 25040280
    .
  203. .
  204. ^ Online Mendelian Inheritance in Man (OMIM): 105830
  205. PMID 17121465
    .
  206. .
  207. .
  208. ^ A person's paternal grandson is the son of a son of that person; a maternal grandson is the son of a daughter.
  209. Robert Winston refers to this study in a "Lecture". Archived from the original
    on 23 May 2007.
  210. ^ "NOVA | Transcripts | Ghost in Your Genes". PBS. 16 October 2007. Retrieved 26 July 2012.
  211. PMID 29530315
    .
  212. .
  213. .
  214. .
  215. .
  216. ^ .
  217. .
  218. .
  219. .
  220. . Conclusions
    ΔFosB is an essential transcription factor implicated in the molecular and behavioral pathways of addiction following repeated drug exposure. The formation of ΔFosB in multiple brain regions, and the molecular pathway leading to the formation of AP-1 complexes is well understood. The establishment of a functional purpose for ΔFosB has allowed further determination as to some of the key aspects of its molecular cascades, involving effectors such as GluR2 (87,88), Cdk5 (93) and NFkB (100). Moreover, many of these molecular changes identified are now directly linked to the structural, physiological and behavioral changes observed following chronic drug exposure (60,95,97,102). New frontiers of research investigating the molecular roles of ΔFosB have been opened by epigenetic studies, and recent advances have illustrated the role of ΔFosB acting on DNA and histones, truly as a molecular switch (34). As a consequence of our improved understanding of ΔFosB in addiction, it is possible to evaluate the addictive potential of current medications (119), as well as use it as a biomarker for assessing the efficacy of therapeutic interventions (121,122,124). Some of these proposed interventions have limitations (125) or are in their infancy (75). However, it is hoped that some of these preliminary findings may lead to innovative treatments, which are much needed in addiction.
  221. . For these reasons, ΔFosB is considered a primary and causative transcription factor in creating new neural connections in the reward centre, prefrontal cortex, and other regions of the limbic system. This is reflected in the increased, stable and long-lasting level of sensitivity to cocaine and other drugs, and tendency to relapse even after long periods of abstinence. These newly constructed networks function very efficiently via new pathways as soon as drugs of abuse are further taken ... In this way, the induction of CDK5 gene expression occurs together with suppression of the G9A gene coding for dimethyltransferase acting on the histone H3. A feedback mechanism can be observed in the regulation of these 2 crucial factors that determine the adaptive epigenetic response to cocaine. This depends on ΔFosB inhibiting G9a gene expression, i.e. H3K9me2 synthesis which in turn inhibits transcription factors for ΔFosB. For this reason, the observed hyper-expression of G9a, which ensures high levels of the dimethylated form of histone H3, eliminates the neuronal structural and plasticity effects caused by cocaine by means of this feedback which blocks ΔFosB transcription
  222. .
  223. .
  224. .
  225. PMCID: PMC9533993 DOI
    : 10.1038/s41586-022-05282-z (2022). Retrieved 7 October 2022.
  226. .
  227. .
  228. .
  229. .
  230. .
  231. .
  232. .
  233. .
  234. .
  235. ^
    S2CID 245654240. Archived from the original
    on 29 October 2022. Retrieved 20 January 2023.
  236. .
  237. .
  238. .
  239. .
  240. .
  241. .
  242. .
  243. .
  244. .
  245. .
  246. .
  247. .
  248. ^ Gorski D (4 February 2013). "Epigenetics: It doesn't mean what quacks think it means". Science-Based Medicine.

Further reading

  • Haque FN, Gottesman II, Wong AH (May 2009). "Not really identical: epigenetic differences in monozygotic twins and implications for twin studies in psychiatry". American Journal of Medical Genetics. Part C, Seminars in Medical Genetics. 151C (2): 136–41.
    S2CID 205327825
    .
  • "What is Epigenetics?". Centers for Disease Control and Prevention. 15 August 2022. Retrieved 11 September 2023.

External links