130421 Wiki

http://en.wikipedia.org/wiki/Epigenetics This page was last modified on 18 April 2013 at 11:39. Here 21/4/13

Epigenetics

From Wikipedia, the free encyclopedia

For the unfolding of an organism or the theory that plants and animals (including humans) develop in this way, see epigenesis (biology). For epigenetics in robotics, see developmental robotics.

In biology, and specifically genetics, epigenetics is the study of changes in gene expression or cellularphenotype, caused by mechanisms other than changes in the underlying DNA sequence – hence the name epi- (Greek: επί- over, above, outer) -genetics, some of which are heritable.

It refers to functionally relevant modifications to the genome that do not involve a change in the nucleotide sequence. Examples of such modifications are DNA methylation and histone modification, both of which serve to regulate gene expression without altering the underlying DNA sequence. These changes may remain through cell divisions for the remainder of the cell's life and may also last for multiple generations. However, there is no change in the underlying DNA sequence of the organism;[1] instead, non-genetic factors cause the organism's genes to behave (or "express themselves") differently.[2] There are objections to the use of the term epigenetic to describe chemical modification of histone, since it remains unclear whether or not histone modifications are heritable.[3]

One example of epigenetic changes in eukaryotic biology is the process of cellular differentiation. Duringmorphogenesis, totipotent stem cells become the various pluripotent cell lines of the embryo, which in turn become fully differentiated cells. In other words, a single fertilized egg cell – the zygote – changes into the many cell types including neurons, muscle cells, epithelium, endothelium of blood vessels, etc. as it continues to divide. It does so by activating some genes while inhibiting others.[4]

In 2011, it was demonstrated that the methylation of mRNA has a critical role in human energy homeostasis. The obesity associated FTO gene is shown to be able to demethylate N6-methyladenosinein RNA. This opened the related field of RNA epigenetics.[5][6]

Etymology and definitions

There exist several definitions of epigenetics, and as a result, there are disagreements as to what epigenetics should mean. Epigenetics(as in "epigenetic landscape") was coined byC. H. Waddington in 1942 as a portmanteauof the words genetics and epigenesis.[7]Epigenesis is an old[8] word that has more recently been used (see preformationism for historical background) to describe the differentiation of cells from their initialtotipotent state in embryonic development. When Waddington coined the term the physical nature of genes and their role in heredity was not known; he used it as a conceptual model of how genes might interact with their surroundings to produce a phenotype.

Robin Holliday defined epigenetics as "the study of the mechanisms of temporal and spatial control of gene activity during the development of complex organisms."[9] Thus epigenetic can be used to describe anything other than DNA sequence that influences the development of an organism.

The more recent usage of the word in science has a stricter definition. It is, as defined by Arthur Riggs and colleagues, "the study of mitotically and/or meiotically heritable changes in gene function that cannot be explained by changes in DNA sequence."[10] The Greek prefix epi- in epigenetics implies features that are "on top of" or "in addition to" genetics; thus epigenetic traits exist on top of or in addition to the traditional molecular basis for inheritance.

The term "epigenetics", however, has been used to describe processes which haven't been demonstrated to be heritable such as histone modification, there are therefore attempts to redefine it in broader terms that would avoid the constraints of requiring heritability. For example, Adrian Bird defined epigenetics as "the structural adaptation of chromosomal regions so as to register, signal or perpetuate altered activity states."[1] This definition would be inclusive of transient modifications associated with DNA repair or cell-cycle phases as well as stable changes maintained across multiple cell generations, but exclude others such as templating of membrane architecture and prions unless they impinge on chromosome function. Such redefinitions however are not universally accepted and are still subject to dispute.[3]

In 2008, a consensus definition of the epigenetic trait, "stably heritable phenotype resulting from changes in a chromosome without alterations in the DNA sequence", was made at a Cold Spring Harbor meeting.[11]

The similarity of the word to "genetics" has generated many parallel usages. The "epigenome" is a parallel to the word "genome", and refers to the overall epigenetic state of a cell. The phrase "genetic code" has also been adapted—the "epigenetic code" has been used to describe the set of epigenetic features that create different phenotypes in different cells. Taken to its extreme, the "epigenetic code" could represent the total state of the cell, with the position of each molecule accounted for in anepigenomic map, a diagrammatic representation of the gene expression, DNA methylation and histone modification status of a particular genomic region. More typically, the term is used in reference to systematic efforts to measure specific, relevant forms of epigenetic information such as the histone codeor DNA methylation patterns.

The psychologist Erik Erikson used the term epigenetic in his book Identity: Youth and Crisis (1968). Erikson writes that the epigenetic principle is where "anything that grows has a ground plan, and that out of this ground plan, the parts arise, each part having its time of special ascendancy, until all parts have arisen to form a functioning whole."[12] That usage, however, is of primarily historical interest.[13]

Molecular basis of epigenetics

Epigenetic changes can modify the activation of certain genes, but not the sequence of DNA. Additionally, the chromatin proteins associated with DNA may be activated or silenced. This is why the differentiated cells in a multi-cellular organism 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, but, if gene disactivation occurs in a sperm or egg cell that results in fertilization, then some epigenetic changes can be transferred to the next generation.[14] This raises the question of whether or not epigenetic changes in an organism can alter the basic structure of its DNA (see Evolution, below), a form of Lamarckism.

Specific epigenetic processes include paramutation, bookmarking, imprinting, gene silencing, X chromosome inactivation, position effect, reprogramming, transvection, maternal effects, the progress ofcarcinogenesis, many effects of teratogens, regulation of histone modifications and heterochromatin, and technical limitations affecting parthenogenesis and cloning.

DNA damage can also cause epigenetic changes.[15][16][17] DNA damages are very frequent, occurring on average about 10,000 times a day per cell of the human body (see DNA damage (naturally occurring)). These damages are largely repaired, but at the site of a DNA repair, epigenetic changes can remain.[18]In particular, a double strand break in DNA can initiate unprogrammed epigenetic gene silencing both by causing DNA methylation as well as by promoting silencing types of histone modifications (chromatin remodeling) (see next section).[19] In addition, the enzyme Parp1 (poly(ADP)-ribose polymerase) and its product poly(ADP)-ribose (PAR) accumulate at sites of DNA damage as part of a repair process.[20] This accumulation, in turn, directs recruitment and activation of the chromatin remodeling protein ALC1 that can cause nucleosome remodeling.[21] Nucleosome remodeling has been found to cause, for instance, epigenetic silencing of DNA repair gene MLH1.[22] [11]. 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.[23]

Foods are known to alter the epigenetics of rats on different diets.[24] Some food components epigenetically increase the levels of DNA repair enzymes such as MGMT and MLH1[25] and p53.[26][27]Other foods components can reduce DNA damage, such as soy isoflavones[28][29] and bilberry anthocyanins.[30]

Epigenetic research uses a wide range of molecular biologic techniques to further our understanding of epigenetic phenomena, including chromatin immunoprecipitation (together with its large-scale variantsChIP-on-chip and ChIP-Seq), fluorescent in situ hybridization, methylation-sensitive restriction enzymes, DNA adenine methyltransferase identification (DamID) and bisulfite sequencing. Furthermore, the use ofbioinformatic methods is playing an increasing role (computational epigenetics).

Computer simulations and molecular dynamics approaches revealed the atomistic motions associated with the molecular recognition of the histone tail through an allosteric mechanism.[31]

Mechanisms

Several types of epigenetic inheritance systems may play a role in what has become known as cell memory,[32] note however that not all of these are universally accepted to be examples of epigenetics.

DNA methylation and chromatin remodeling

Because the phenotype of a cell or individual is affected by which of its genes are transcribed, heritable 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. Histone proteins are little spheres that DNA wraps around. 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 sphere 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 it, these modified histones would ensure that a differentiated cell would stay differentiated, and not convert back into being a stem cell.
    2. The second way is the addition of methyl groups to the DNA, mostly at CpG sites, to convertcytosine to 5-methylcytosine. 5-Methylcytosine performs much like a regular cytosine, pairing up with a guanine. However, some areas of the genome are methylated more heavily than others, and highly methylated areas tend to be less transcriptionally active, through a mechanism not fully understood. Methylation of cytosines can also persist from the germ line of one of the parents into the zygote, marking the chromosome as being inherited from this parent (genetic imprinting).

The way that the cells stay differentiated in the case of DNA methylation is clearer to us than it is in the case of histone shape. Basically, certain enzymes (such as DNMT1) have a higher affinity for the methylated cytosine. If this enzyme reaches a "hemimethylated" portion of DNA (where methylcytosine is in only one of the two DNA strands) the enzyme will methylate the other half.

Although histone modifications occur throughout the entire sequence, the unstructured N-termini of histones (called histone tails) are particularly highly modified. These modifications include acetylation,methylation, ubiquitylation, phosphorylation and sumoylation. 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 correlated with transcriptional competence.

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 epigenetic function. In other words, changes to the histone tails have a direct effect on the DNA itself.

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 (and basal transcription machinery as well). This Chromatin Remodeler can then cause changes to the state of the chromatin. Indeed, the bromodomain — a protein segment (domain) that specifically binds acetyl-lysine — is found in many enzymes that help activate transcription, including the SWI/SNFcomplex (on the protein polybromo). 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 histone methylation as well. Methylation of lysine 9 of histone H3 has long been associated with constitutively transcriptionally silent chromatin (constitutive heterochromatin). It has been determined that a chromodomain (a domain that specifically binds methyl-lysine) in the transcriptionally repressive protein 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. 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.[33]

Differing histone modifications are likely to function in differing ways; acetylation at one position is likely to function differently than 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 nucleosome. The idea that multiple dynamic modifications regulate gene transcription in a systematic and reproducible way is called the histone code.

DNA methylation frequently occurs in repeated sequences, and helps to suppress the expression and mobility of 'transposable elements':[34] Because 5-methylcytosine can be spontaneously deaminated (replacing nitrogen by oxygen) to thymidine, CpG sites are frequently mutated and become rare in the genome, except at CpG islands where they remain unmethylated. Epigenetic changes of this type thus have the potential to direct increased frequencies of permanent genetic mutation. DNA methylationpatterns 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.[35] DNMT1 is the most abundant methyltransferase in somatic cells,[36] localizes to replication foci,[37] has a 10–40-fold preference for hemimethylated DNA and interacts with the proliferating cell nuclear antigen (PCNA).[38]

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.[39] DNMT1 is essential for proper embryonic development, imprinting and X-inactivation.[35][40] 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.[41] 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.[42]

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.[43]

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 alternativecovalent modifications of histones.[44] The stability and heritability of states of larger chromosomal regions are often thought to involve positive feedback where modified nucleosomes recruit enzymes that similarly modify nearby nucleosomes. A simplified stochastic model for this type of epigenetics is found here.[45][46]

Because DNA methylation and chromatin remodeling play such a central role in many types of epigenic inheritance, 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.[47]

It has been suggested that the histone code could be mediated by the effect of small RNAs. The recent discovery and characterization of a vast array of small (21- to 26-nt), non-coding RNAs suggests that there is an RNA component, possibly involved in epigenetic gene regulation. Small interfering RNAs can modulate transcriptional gene expression via epigenetic modulation of targeted promoters.[48]

RNA transcripts and their encoded proteins

Sometimes a gene, after being turned on, transcribes a product that (either directly or indirectly) maintains the activity of that gene. For example, Hnf4 and MyoD enhance the transcription of many liver- and muscle-specific genes, respectively, including their own, through the transcription factor activity of the proteins they encode. RNA signalling includes differential recruitment of a hierarchy of generic chromatin modifying complexes and DNA methyltransferases to specific loci by RNAs during differentiation and development.[49] Other epigenetic changes are mediated by the production of different splice forms ofRNA, or by formation of double-stranded RNA (RNAi). Descendants of the cell in which the gene was turned on will inherit this activity, even if the original stimulus for gene-activation is no longer present. These genes are most often turned on or off by signal transduction, although in some systems wheresyncytia 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.[50]

MicroRNAs

MicroRNAs (miRNAs) are members of non-coding RNAs that range in size from 17 to 25 nucleotides. As indicated by Wang et al.,[51] miRNAs regulate a large variety of biological functions. So far, in 2013, about 2000 miRNAs have been discovered in humans and these can be found online in an miRNA database.[52]Each miRNA expressed in a cell may target about 100 to 200 messenger RNAs that it downregulates.[53]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.[54]

It appears that about 60% of human protein coding genes are regulated by miRNAs.[55] Many miRNAs are epigenetically regulated. About 50% of miRNA genes are associated with CpG islands,[51] that may be repressed by epigenetic methylation. Transcription from methylated CpG islands is strongly and heritably repressed.[56] Other miRNAs are epigenetically regulated by either histone modifications or by combined DNA methylation and histone modification.[51]

Prions

For more details on this topic, see Fungal 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.[57]

Fungal prions are considered epigenetic because the infectious phenotype caused by the prion can be inherited without modification of the genome. PSI+ and URE3, discovered in yeast in 1965 and 1971, are the two best studied of this type of prion.[58][59] Prions can have a phenotypic effect through the sequestration of protein in aggregates, thereby reducing that protein's activity. In PSI+ cells, the loss of the Sup35 protein (which is involved in termination of translation) causes ribosomes to have a higher rate of read-through of stop codons, an effect that results in suppression of nonsense mutations in other genes.[60] 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.[61][62][63][64]

Structural inheritance systems

For more details on this topic, see 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.[65][66][67]

Epigenetic mechanisms

DNA associates with histone proteins to form chromatin.

Functions and consequences

Development

Somatic epigenetic inheritance through epigenetic modifications, particularly through DNA methylation and chromatin remodeling, is very important in the development of multicellular eukaryotic organisms. The genome sequence is static (with some notable exceptions), but cells differentiate into many different types, which perform different functions, and respond differently to the environment and intercellular signalling. Thus, as individuals develop, morphogens activate or silence genes in an epigenetically heritable fashion, giving cells a "memory". In mammals, most cells terminally differentiate, with only stem cells retaining the ability to differentiate into several cell types ("totipotency" and "multipotency"). Inmammals, some stem cells continue producing new differentiated cells throughout life, 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. Unlike animals, plant cells do not terminally differentiate, remaining totipotent with the ability to give rise to a new individual plant. While plants do utilise many of the same epigenetic mechanisms as animals, such as chromatin remodeling, it has been hypothesised 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.[68]

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.[69]

Medicine

Epigenetics has many and varied potential medical applications as it tends to be multidimensional in nature.[70] Congenital genetic disease is well understood, and it is also clear that epigenetics can play a role, for example, in the case of Angelman syndrome and Prader-Willi syndrome. These are normal genetic diseases caused by gene deletions or inactivation of the genes, but are unusually common because individuals are essentially 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.[71]

Evolution

Epigenetics can impact evolution when epigenetic changes are heritable. A sequestered germ line orWeismann barrier is specific to animals, and epigenetic inheritance is more common in plants and microbes. Eva Jablonka and Marion Lamb have argued that these effects may require enhancements to the standard conceptual framework of the modern evolutionary synthesis.[72][73] Other evolutionary biologists have incorporated epigenetic inheritance into population genetics models[74] or are openly skeptical.[75]

Two important ways in which epigenetic inheritance can be different from traditional genetic inheritance, with important consequences for evolution, are that rates of epimutation can be much faster than rates of mutation[76] and the epimutations are more easily reversible.[77] An epigenetically inherited element such as the [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.[78] The existence of this possibility increases the evolvability of a species.

Examples

Epigenetic changes have been observed to occur in response to environmental exposure—for example, mice given some dietary supplements have epigenetic changes affecting expression of the agouti gene, which affects their fur color, weight, and propensity to develop cancer.[79][80]

In the case of humans with different environmental exposues, Fraga et al.[81] studied young monozygotic (identical) twins and older monozygotic twins. They found that although such twins were epigenetically indistinguishable during their early years, older twins had remarkable differences in the overall content and genomic distribution of 5-methylcytosine DNA and histone acetylation. The twin pairs who had spent less of their lifetime together and/or had greater differences in their medical histories where those who showed the largest differences in their levels of 5methylcytosine DNA and acetylation of histones H3 and H4.

More than 100 cases of transgenerational epigenetic inheritance phenomena have been reported in a wide range of organisms, including prokaryotes, plants, and animals.[82]

Recent analyses have suggested that members of the APOBEC/AID family of cytosine deaminases are capable of simultaneously mediating genetic and epigenetic inheritance using similar molecular mechanisms.[83]

Epigenetic effects in humans

Genomic imprinting and related disorders

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.[84]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.[85] 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.

Transgenerational epigenetic observations

See main article Transgenerational epigenetics

In the Överkalix study, Marcus Pembrey and colleagues observed that the paternal (but not maternal) grandsons[86] 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.[87] 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.[88]

Cancer and developmental abnormalities

A variety of compounds are considered as epigenetic carcinogens—they result in an increased incidence of tumors, but they do not show mutagen activity (toxic compounds or pathogens that cause tumors incident to increased regeneration should also be excluded). Examples include diethylstilbestrol, arsenite,hexachlorobenzene, and nickel compounds.

Many teratogens exert specific effects on the fetus by epigenetic mechanisms.[89][90] While epigenetic effects may preserve the effect of a teratogen such as diethylstilbestrol throughout the life of an affected child, the possibility of birth defects resulting from exposure of fathers or in second and succeeding generations of offspring has generally been rejected on theoretical grounds and for lack of evidence.[91]However, a range of male-mediated abnormalities have been demonstrated, and more are likely to exist.[92] FDA label information for Vidaza, a formulation of 5-azacitidine (an unmethylatable analog of cytidine that causes hypomethylation when incorporated into DNA) states that "men should be advised not to father a child" while using the drug, citing evidence in treated male mice of reduced fertility, increased embryo loss, and abnormal embryo development.[93] In rats, endocrine differences were observed in offspring of males exposed to morphine.[94] In mice, second generation effects of diethylstilbesterol have been described occurring by epigenetic mechanisms.[95]

Recent studies have shown that the mixed-lineage leukemia (MLL) gene causes leukemia by rearranging and fusing with other genes in different chromosomes, which is a process under epigenetic control.[96]

Other investigations have concluded that alterations in histone acetylation and DNA methylation occur in various genes influencing prostate cancer.[97] Gene expression in the prostate can be modulated by nutrition and lifestyle changes.[98]

In 2008, the National Institutes of Health announced that $190 million had been earmarked for epigenetics research over the next five years. In announcing the funding, government officials noted that epigenetics has the potential to explain mechanisms of aging, human development, and the origins of cancer, heart disease, mental illness, as well as several other conditions. Some investigators, like Randy Jirtle, PhD, of Duke University Medical Center, think epigenetics may ultimately turn out to have a greater role in disease than genetics.[99]

DNA methylation in cancer

DNA methylation is an important regulator of gene transcription and a large body of evidence has demonstrated that aberrant DNA methylation is associated with unscheduled gene silencing, and the genes with high levels of 5-methylcytosine in their promoter region are transcriptionally silent. DNA methylation is essential during embryonic development, and in somatic cells, patterns of DNA methylation are in general transmitted to daughter cells with a high fidelity. Aberrant DNA methylation patterns have been associated with a large number of human malignancies and found in two distinct forms: hypermethylation and hypomethylation compared to normal tissue. Hypermethylation is one of the major epigenetic modifications that repress transcription via promoter region of tumour suppressor genes. Hypermethylation typically occurs at CpG islands in the promoter region and is associated with gene inactivation. Global hypomethylation has also been implicated in the development and progression of cancer through different mechanisms.[100]

DNA repair epigenetics in cancer

Germ line (familial) mutations have been identified in 34 different DNA repair genes that cause a high risk of cancer, including, for example BRCA1 and ATM. These are listed in the article DNA repair-deficiency disorder. However, cancers caused by such germ line mutations make up only a very small proportion of cancers. For instance, germ line mutations cause only 2% to 5% of colon cancer cases.[101]

Epigenetic reductions in expression of DNA repair genes, however, are very frequent in sporadic (non-germ line) cancers, as shown among some representative cancers in the table in this section, while mutations in DNA repair genes in sporadic cancer are very rare.[102]

References in the table are given here: 1,[103] 2,[104] 3,[105] 4,[106] 5,[107] 6,[108] 7,[109] 8,[110] 9,[111]10,[112] 11,[113] 12,[114] 13,[115] 14,[116] 15,[117] 16,[118] 17,[119] 18,[120] 19,[121] 20[122]

Deficiencies in expression of DNA repair genes cause increased mutation rates. Mutations rates increase in mice defective for mismatch DNA repair genes PMS2, MLH1, MSH2, MSH3 orMSH6[123][124] or for DNA repair gene BRCA2,[125] while chromosomal rearrangements and aneuploidy are noted to increase in humans defective in DNA repair gene BLM.[126] Thus, deficiency in DNA repair causes genome instability and this genome instability is likely the main underlying cause of the genetic alterations leading to cancer. In fact, as indicated by Nowak et al.[127] through a mathematical calculation, the first event in many sporadic neoplasias is a heritable alteration that affects genetic instability, and we note that epigenetic defects in DNA repair are somatically heritable.

Variant histones H2A in cancer

The histone variants of the H2A family are highly conserved in mammals, playing critical roles in regulating many nuclear processes by altering chromatin structure. One of the key H2A variants, H2A.X, marks DNA damage, facilitating the recruitment of DNA repair proteins to restore genomic integrity. Another variant, H2A.Z, plays an important role in both gene activation and repression. A high level of H2A.Z expression is ubiquitously detected in many cancers and is significantly associated with cellular proliferation and genomic instability.[100]

Cancer treatment

Current research has shown that epigenetic pharmaceuticals could be a putative replacement or adjuvant therapy for currently accepted treatment methods such as radiation and chemotherapy, or could enhance the effects of these current treatments.[128] It has been shown that the epigenetic control of the proto-onco regions and the tumor suppressor sequences by conformational changes in histones directly affects the formation and progression of cancer.[129] Epigenetics also has the factor of reversibility, a characteristic that other cancer treatments do not offer.[97]

Drug development has focused mainly on histone acetyltransferase (HAT) and histone deacetylase(HDAC), and has included the introduction to the market of the new pharmaceutical vorinostat, an HDAC inhibitor.[130] HDAC has been shown to play an integral role in the progression of oral squamous cancer.[129]

Current front-runner candidates for new drug targets are histone lysine methyltransferases (KMT) and protein arginine methyltransferases (PRMT).[131]

A study conducted on AML patients showed that the use of chemotherapy together with a combination of a DNA methyltransferase (such as epigallocatechingallate [EGCG]) and a histone deacetylase inhibitor (such as valproic acid [VPA]) can distinctively modify the malignant transcriptome of AML blasts by inhibiting DNA hypermethylation and histone acetylation and can confer a positive prognostic impact on AML patients.[132]

Twin studies

Recent studies involving both dizygotic and monozygotic twins have produced some evidence of epigenetic influence in humans.[133][134][135]

Epigenetics in microorganisms

Bacteria make widespread use of postreplicative DNA methylation for the epigenetic control of DNA-protein interactions. Bacteria make use of DNA adenine methylation (rather than DNA cytosine methylation) as an epigenetic signal. DNA adenine methylation is important in bacteria virulence in organisms such as Escherichia coli, Salmonella, Vibrio,Yersinia, Haemophilus, and Brucella. In Alphaproteobacteria, methylation of adenine regulates the cell cycle and couples gene transcription to DNA replication. In Gammaproteobacteria, adenine methylation provides signals for DNA replication, chromosome segregation, mismatch repair, packaging of bacteriophage, transposase activity and regulation of gene expression.[136][137]

The filamentous fungus Neurospora crassa is a prominent model system for understanding the control and function of cytosine methylation. In this organisms, 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.[138]

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. This is an example of epigenetic regulation enabling 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.[137]

See also

Escherichia coli bacteria

Notes and references

    1. ^ a b Bird A (May 2007). "Perceptions of epigenetics". Nature 447 (7143): 396–8.doi:10.1038/nature05913. PMID 17522671.
    2. ^ "Special report: 'What genes remember' by Philip Hunter | [[Prospect Magazine]] May 2008 issue 146". Web.archive.org. 2008-05-01. Retrieved 2012-07-26. Wikilink embedded in URL title (help)
    3. ^ a b Ledford H. (2008). "Disputed definitions".Nature 455 (7216): 1023–8. PMID 18948925.
    4. ^ Reik W (May 2007). "Stability and flexibility of epigenetic gene regulation in mammalian development". Nature 447 (7143): 425–32.doi:10.1038/nature05918. PMID 17522676.
    5. ^ Jia, Guifang; Fu, Ye, Zhao, Xu, Dai, Qing, Zheng, Guanqun, Yang, Ying, Yi, Chengqi, Lindahl, Tomas, Pan, Tao, Yang, Yun-Gui, He, Chuan (16 October 2011). "N6-Methyladenosine in nuclear RNA is a major substrate of the obesity-associated FTO". Nature Chemical Biology 7 (12): 885–887.doi:10.1038/nchembio.687. PMC 3218240.PMID 22002720.
    6. ^ "New research links common RNA modification to obesity". Physorg.com. Retrieved 2012-07-26.
    7. ^ Waddington CH (1942). "The epigenotype".Endeavour 1: 18–20.
    8. ^ According to the Oxford English Dictionary:
        1. 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’.
    9. It is also worth quoting this adumbration of the definition given there (viz., "The formation of an organic germ as a new product"):
        1. theory of epigenesis: 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’.
    10. ^ Holliday R (November 1990). "Mechanisms for the control of gene activity during development".Biol Rev Camb Philos Soc 65 (4): 431–71.PMID 2265224.
    11. ^ Riggs AD, Russo VEA, Martienssen RA (1996).Epigenetic mechanisms of gene regulation. Plainview, N.Y: Cold Spring Harbor Laboratory Press. ISBN 0-87969-490-4.
    12. ^ Berger SL, Kouzarides T, Shiekhattar R, Shilatifard A (2009). "An operational definition of epigenetics". Genes Dev. 23 (7): 781–3.doi:10.1101/gad.1787609. PMID 19339683.
    13. ^ Erikson, Erik (1968). Identity: Youth and Crisis. Chapter 3: W.W. Norton and Company, Inc. p. 92.
    14. ^ "Epigenetics". Bio-Medicine.org. Retrieved 2011-05-21.
    15. ^ Chandler VL (February 2007). "Paramutation: from maize to mice". Cell 128 (4): 641–5.doi:10.1016/j.cell.2007.02.007.PMID 17320501.
    16. ^ Kovalchuk O, Baulch JE (2008). Epigenetic changes and nontargeted radiation effects--is there a link? Environ Mol Mutagen 49(1):16-25. doi: 10.1002/em.20361. PMID 18172877
    17. ^ Ilnytskyy Y, Kovalchuk O (2011). Non-targeted radiation effects-an epigenetic connection. Mutat Res 714(1-2):113-125. doi: 10.1016/j.mrfmmm.2011.06.014 Review. PMID 21784089
    18. ^ Friedl AA, Mazurek B, Seiler DM (2012). Radiation-induced alterations in histone modification patterns and their potential impact on short-term radiation effects. Front Oncol 2:117. doi: 10.3389/fonc.2012.00117. PMID 23050241
    19. ^ Cuozzo C, Porcellini A, Angrisano T, Morano A, Lee B, Di Pardo A, Messina S, Iuliano R, Fusco A, Santillo MR, Muller MT, Chiariotti L, Gottesman ME, Avvedimento EV (2007). DNA damage, homology-directed repair, and DNA methylation. PLoS Genet 3(7):e110. PMID 17616978
    20. ^ O'Hagan HM, Mohammad HP, Baylin SB. Double strand breaks can initiate gene silencing and SIRT1-dependent onset of DNA methylation in an exogenous promoter CpG island. PLoS Genet 2008;4(8) e1000155. PMID 18704159
    21. ^ Malanga M, Althaus FR (2005). The role of poly(ADP-ribose) in the DNA damage signaling network. Biochem Cell Biol 83(3):354-364. Review. PMID 15959561
    22. ^ Gottschalk AJ, Timinszky G, Kong SE, Jin J, Cai Y, Swanson SK, Washburn MP, Florens L, Ladurner AG, Conaway JW, Conaway RC (2009). Poly(ADP-ribosyl)ation directs recruitment and activation of an ATP-dependent chromatin remodeler. Proc Natl Acad Sci U S A 106(33):13770-4. doi: 10.1073/pnas.0906920106. PMID 19666485[PubMed - indexed for MEDLINE] PMCID: PMC2722505
    23. ^ Lin JC, Jeong S, Liang G, Takai D, Fatemi M, Tsai YC, Egger G, Gal-Yam EN, Jones PA (2007). Role of nucleosomal occupancy in the epigenetic silencing of the MLH1 CpG island. Cancer Cell 12(5):432-444. PMID 17996647
    24. ^ Tabish AM, Poels K, Hoet P, Godderis L (2012). Epigenetic factors in cancer risk: effect of chemical carcinogens on global DNA methylation pattern in human TK6 cells. PLoS One 7(4):e34674. doi: 10.1371/journal.pone.0034674. PMID 22509344
    25. ^ Burdge GC, Hoile SP, Uller T, Thomas NA, Gluckman PD, Hanson MA, Lillycrop KA (2011). Progressive, transgenerational changes in offspring phenotype and epigenotype following nutritional transition. PLoS One 6(11):e28282. doi: 10.1371/journal.pone.0028282. PMID 22140567
    26. ^ Fang M, Chen D, Yang CS (2007). Dietary polyphenols may affect DNA methylation. J Nutr 137(1 Suppl):223S-228S. PMID 17182830
    27. ^ Olaharski AJ, Rine J, Marshall BL, Babiarz J, Zhang L, Verdin E, Smith MT (2005). The flavoring agent dihydrocoumarin reverses epigenetic silencing and inhibits sirtuin deacetylases. PLoS Genet 1(6):e77. PMID 16362078 [PubMed - indexed for MEDLINE] PMCID: PMC1315280
    28. ^ Kikuno N, Shiina H, Urakami S, Kawamoto K, Hirata H, Tanaka Y, Majid S, Igawa M, Dahiya R (2008). Genistein mediated histone acetylation and demethylation activates tumor suppressor genes in prostate cancer cells. Int J Cancer 123(3):552-560. doi: 10.1002/ijc.23590. PMID 18431742
    29. ^ Davis JN, Kucuk O, Djuric Z, Sarkar FH (2001). Soy isoflavone supplementation in healthy men prevents NF-kappa B activation by TNF-alpha in blood lymphocytes. Free Radic Biol Med 30(11):1293-1302. PMID 11368927
    30. ^ Djuric Z, Chen G, Doerge DR, Heilbrun LK, Kucuk O (2001). Effect of soy isoflavone supplementation on markers of oxidative stress in men and women. Cancer Lett 172(1):1-6.PMID 11595123
    31. ^ Kropat C, Mueller D, Boettler U, Zimmermann K, Heiss EH, Dirsch VM, Rogoll D, Melcher R, Richling E, Marko D (2013). Modulation of Nrf2-dependent gene transcription by bilberry anthocyanins in vivo. Mol Nutr Food Res doi: 10.1002/mnfr.201200504. [Epub ahead of print]PMID 23349102
    32. ^ Baron R (2012). "LSD1/CoREST is an allosteric nanoscale clamp regulated by H3-histone-tail molecular recognition". Proc Natl Acad Sci U S A. 109 (31): 12509–14.doi:10.1073/pnas.1207892109.PMID 22802671.
    33. ^ Jablonka E, Lamb MJ, Lachmann M (September 1992). "Evidence, mechanisms and models for the inheritance of acquired characteristics". J. Theoret. Biol. 158 (2): 245–268. doi:10.1016/S0022-5193(05)80722-2.
    34. ^ Jenuwein T, Laible G, Dorn R, Reuter G (January 1998). "SET domain proteins modulate chromatin domains in eu- and heterochromatin".Cell. Mol. Life Sci. 54 (1): 80–93.doi:10.1007/s000180050127.PMID 9487389.
    35. ^ Slotkin RK, Martienssen R (April 2007). "Transposable elements and the epigenetic regulation of the genome". Nat. Rev. Genet. 8(4): 272–85. doi:10.1038/nrg2072.PMID 17363976.
    36. ^ a b Li E, Bestor TH, Jaenisch R (June 1992). "Targeted mutation of the DNA methyltransferase gene results in embryonic lethality". Cell 69 (6): 915–26. doi:10.1016/0092-8674(92)90611-F.PMID 1606615.
    37. ^ Robertson KD, Uzvolgyi E, Liang G, Talmadge C, Sumegi J, Gonzales FA, Jones PA (June 1999). "The human DNA methyltransferases (DNMTs) 1, 3a and 3b: coordinate mRNA expression in normal tissues and overexpression in tumors". Nucleic Acids Res. 27 (11): 2291–8. doi:10.1093/nar/27.11.2291.PMC 148793. PMID 10325416.
    38. ^ Leonhardt H, Page AW, Weier HU, Bestor TH (November 1992). "A targeting sequence directs DNA methyltransferase to sites of DNA replication in mammalian nuclei". Cell 71 (5): 865–73. doi:10.1016/0092-8674(92)90561-P.PMID 1423634.
    39. ^ Chuang LS, Ian HI, Koh TW, Ng HH, Xu G, Li BF (September 1997). "Human DNA-(cytosine-5) methyltransferase-PCNA complex as a target for p21WAF1". Science 277 (5334): 1996–2000.doi:10.1126/science.277.5334.1996.PMID 9302295.
    40. ^ Robertson KD, Wolffe AP (October 2000). "DNA methylation in health and disease". Nat. Rev. Genet. 1 (1): 11–9.doi:10.1038/35049533. PMID 11262868.
    41. ^ Li E, Beard C, Jaenisch R (November 1993). "Role for DNA methylation in genomic imprinting".Nature 366 (6453): 362–5.doi:10.1038/366362a0. PMID 8247133.
    42. ^ Viens A et al. "Analysis of human histone H2AZ deposition in vivo argues against its direct role in epigenetic templating mechanisms". Mol Cell Biol. 2006 26(14):5325-35.[1]
    43. ^ Ogryzko VV. Erwin Schroedinger, Francis Crick and epigenetic stability. Biol Direct. 2008 Apr 17;3:15. [2] doi: 10.1186/1745-6150-3-15
    44. ^ Nottke A, Colaiácovo MP, Shi Y (March 2009)."Developmental roles of the histone lysine demethylases". Development 136 (6): 879–89.doi:10.1242/dev.020966. PMC 2692332.PMID 19234061.
    45. ^ Rosenfeld JA, Wang Z, Schones DE, Zhao K, DeSalle R, Zhang MQ (2009). "Determination of enriched histone modifications in non-genic portions of the human genome". BMC Genomics 10: 143. doi:10.1186/1471-2164-10-143. PMC 2667539. PMID 19335899.
    46. ^ "Epigenetic cell memory". Cmol.nbi.dk. Retrieved 2012-07-26.
    47. ^ Dodd IB, Micheelsen MA, Sneppen K, Thon G (May 2007). "Theoretical analysis of epigenetic cell memory by nucleosome modification". Cell129 (4): 813–22.doi:10.1016/j.cell.2007.02.053.PMID 17512413.
    48. ^ Ptashne M (April 2007). "On the use of the word 'epigenetic'". Curr. Biol. 17 (7): R233–6.doi:10.1016/j.cub.2007.02.030.PMID 17407749.
    49. ^ Morris KL (2008). "Epigenetic Regulation of Gene Expression". RNA and the Regulation of Gene Expression: A Hidden Layer of Complexity. Norfolk, England: Caister Academic Press.ISBN 1-904455-25-5.
    50. ^ Mattick JS, Amaral PP, Dinger ME, Mercer TR, Mehler MF (January 2009). "RNA regulation of epigenetic processes". BioEssays 31 (1): 51–9.doi:10.1002/bies.080099. PMID 19154003.
    51. ^ Choi CQ (2006-05-25). "The Scientist: RNA can be hereditary molecule". The Scientist. Retrieved 2006.
    52. ^ a b c Wang Z, Yao H, Lin S, Zhu X, Shen Z, Lu G, Poon WS, Xie D, Lin MC, Kung HF (2012). Transcriptional and epigenetic regulation of human microRNAs. Cancer Lett 331(1):1-10. doi: 10.1016/j.canlet.2012.12.006. PMID 3246373
    53. ^ http://www.mirbase.org/cgi-bin/browse.pl
    54. ^ Lim LP, Lau NC, Garrett-Engele P, Grimson A, Schelter JM, Castle J, Bartel DP, Linsley PS, Johnson JM (2005). Microarray analysis shows that some microRNAs downregulate large numbers of target mRNAs. Nature 433(7027):769-773. PMID 15685193
    55. ^ Lee D, Shin C (2012). MicroRNA-target interactions: new insights from genome-wide approaches. Ann N Y Acad Sci 1271:118-28. doi: 10.1111/j.1749-6632.2012.06745.x. Review.PMID 23050973
    56. ^ Friedman RC, Farh KK, Burge CB, Bartel DP (2009). Most mammalian mRNAs are conserved targets of microRNAs. Genome Res 19(1):92-105. doi: 10.1101/gr.082701.108. PMID 18955434
    57. ^ Goll MG, Bestor TH (2005). Eukaryotic cytosine methyltransferases. Annu Rev Biochem 74:481-514. PMID 15952895
    58. ^ Yool A, Edmunds WJ (1998). "Epigenetic inheritance and prions". Journal of Evolutionary Biology 11 (2): 241–242.doi:10.1007/s000360050085.
    59. ^ Cox BS (1965). "[PSI], a cytoplasmic suppressor of super-suppression in yeast".Heredity 20 (4): 505–521.doi:10.1038/hdy.1965.65.
    60. ^ Lacroute F (May 1971). "Non-Mendelian mutation allowing ureidosuccinic acid uptake in yeast". J. Bacteriol. 106 (2): 519–22.PMC 285125. PMID 5573734.
    61. ^ Liebman SW, Sherman F (September 1979)."Extrachromosomal psi+ determinant suppresses nonsense mutations in yeast". J. Bacteriol.139 (3): 1068–71. PMC 218059.PMID 225301.
    62. ^ True HL, Lindquist SL (September 2000). "A yeast prion provides a mechanism for genetic variation and phenotypic diversity". Nature 407(6803): 477–83. doi:10.1038/35035005.PMID 11028992.
    63. ^ Shorter J, Lindquist S (June 2005). "Prions as adaptive conduits of memory and inheritance".Nat. Rev. Genet. 6 (6): 435–50.doi:10.1038/nrg1616. PMID 15931169.
    64. ^ Giacomelli M, Hancock AS, Masel J, (2007)."The conversion of 3′ UTRs into coding regions". Molecular Biology & Evolution 24 (2): 457–464. doi:10.1093/molbev/msl172.PMC 1808353. PMID 17099057.
    65. ^ Lancaster AK, Bardill JP, True HL, Masel J (2010). "The Spontaneous Appearance Rate of the Yeast Prion [PSI+] and Its Implications for the Evolution of the Evolvability Properties of the [PSI+] System". Genetics 184 (2): 393–400.doi:10.1534/genetics.109.110213.PMC 2828720. PMID 19917766.
    66. ^ Sapp J (1991). "Concepts of organization. The leverage of ciliate protozoa". Dev. Biol. (NY) 7: 229–58. PMID 1804215.
    67. ^ Sapp J (2003). Genesis: the evolution of biology. Oxford [Oxfordshire]: Oxford University Press. ISBN 0-19-515619-6.
    68. ^ Gray RD, Oyama S, Griffiths PE (2003). Cycles of Contingency: Developmental Systems and Evolution (Life and Mind: Philosophical Issues in Biology and Psychology). Cambridge, Mass: The MIT Press. ISBN 0-262-65063-0.
    69. ^ Costa S, Shaw P (March 2007). "'Open minded' cells: how cells can change fate" (PDF).Trends Cell Biol. 17 (3): 101–6.doi:10.1016/j.tcb.2006.12.005.PMID 17194589. "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)"
    70. ^ Griesemer J, Haber MH, Yamashita G, Gannett L (March 2005). "Critical Notice: Cycles of Contingency – Developmental Systems and Evolution". Biology & Philosophy 20 (2–3): 517–544. doi:10.1007/s10539-004-0836-4.
    71. ^ Chahwan R, Wontakal SN, Roa S (March 2011). "The multidimensional nature of epigenetic information and its role in disease".Discov Med 11 (58): 233–43. PMID 21447282.
    72. ^ Online 'Mendelian Inheritance in Man' (OMIM)105830
    73. ^ Lamb MJ, Jablonka E (2005). Evolution in four dimensions: genetic, epigenetic, behavioral, and symbolic variation in the history of life. Cambridge, Mass: MIT Press. ISBN 0-262-10107-6.
    74. ^ See also Denis Noble The Music of Life see esp pp. 93–8 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)
    75. ^ Maynard Smith, John (1990). "Models of a Dual Inheritance System". Journal of Theoretical Biology 143 (1): 41–53. doi:10.1016/S0022-5193(05)80287-5.
    76. ^ Lynch, M. (2007). "The frailty of adaptive hypotheses for the origins of organismal complexity". PNAS 104 (suppl. 1): 8597–8604.Bibcode:2007PNAS..104.8597L.doi:10.1073/pnas.0702207104.PMC 1876435. PMID 17494740.
    77. ^ Rando OJ, Verstrepen KJ (February 2007). "Timescales of genetic and epigenetic inheritance". Cell 128 (4): 655–68.doi:10.1016/j.cell.2007.01.023.PMID 17320504.
    78. ^ Lancaster, Alex K.; Masel, Joanna (1 September 2009). "The evolution of reversible switches in the presence of irreversible mimics".Evolution 63 (9): 2350–2362. doi:10.1111/j.1558-5646.2009.00729.x. PMID 19486147.
    79. ^ Griswold CK, Masel J (2009). "Complex Adaptations Can Drive the Evolution of the Capacitor PSI+, Even with Realistic Rates of Yeast Sex". In Úbeda, Francisco. PLoS Genetics 5 (6): e1000517.doi:10.1371/journal.pgen.1000517.PMC 2686163. PMID 19521499.
    80. ^ Cooney CA, Dave AA, Wolff GL (August 2002). "Maternal methyl supplements in mice affect epigenetic variation and DNA methylation of offspring". J. Nutr. 132 (8 Suppl): 2393S–2400S.PMID 12163699.
    81. ^ Waterland RA, Jirtle RL (August 2003)."Transposable elements: targets for early nutritional effects on epigenetic gene regulation". Mol. Cell. Biol. 23 (15): 5293–300.doi:10.1128/MCB.23.15.5293-5300.2003.PMC 165709. PMID 12861015.
    82. ^ Fraga MF, Ballestar E, Paz MF, Ropero S, Setien F, Ballestar ML, Heine-Suñer D, Cigudosa JC, Urioste M, Benitez J, Boix-Chornet M, Sanchez-Aguilera A, Ling C, Carlsson E, Poulsen P, Vaag A, Stephan Z, Spector TD, Wu YZ, Plass C, Esteller M (2005). Proc Natl Acad Sci U S A 102(30):10604-10609. PMID 16009939
    83. ^ Jablonka E, Raz G (June 2009). "Transgenerational epigenetic inheritance: prevalence, mechanisms, and implications for the study of heredity and evolution". Q Rev Biol 84(2): 131–76. doi:10.1086/598822.PMID 19606595.
    84. ^ Chahwan R, Wontakal SN, Roa S (October 2010). "Crosstalk between genetic and epigenetic information through cytosine deamination". Trends Genet. 26 (10): 443–8.doi:10.1016/j.tig.2010.07.005.PMID 20800313.
    85. ^ Wood AJ, Oakey RJ (November 2006)."Genomic imprinting in mammals: emerging themes and established theories". PLoS Genet. 2 (11): e147.doi:10.1371/journal.pgen.0020147.PMC 1657038. PMID 17121465.
    86. ^ Knoll JH, Nicholls RD, Magenis RE, Graham JM, Lalande M, Latt SA (February 1989). "Angelman and Prader-Willi syndromes share a common chromosome 15 deletion but differ in parental origin of the deletion". Am. J. Med. Genet. 32 (2): 285–90. doi:10.1002/ajmg.1320320235.PMID 2564739.
    87. ^ A person's paternal grandson is the son of a son of that person; a maternal grandson is the son of a daughter.
    88. ^ Pembrey ME, Bygren LO, Kaati G, Edvinsson S, Northstone K, Sjöström M, Golding J (February 2006). "Sex-specific, male-line transgenerational responses in humans". Eur. J. Hum. Genet. 14 (2): 159–66.doi:10.1038/sj.ejhg.5201538.PMID 16391557. Robert Winston refers to this study in a lecture; see also discussion atLeeds University, here [3]
    89. ^ "NOVA | Transcripts | Ghost in Your Genes". PBS. 2007-10-16. Retrieved 2012-07-26.
    90. ^ Bishop JB, Witt KL, Sloane RA (December 1997). "Genetic toxicities of human teratogens".Mutat. Res. 396 (1–2): 9–43.doi:10.1016/S0027-5107(97)00173-5.PMID 9434858.
    91. ^ Gurvich N, Berman MG, Wittner BS, Gentleman RC, Klein PS, Green JB (July 2005). "Association of valproate-induced teratogenesis with histone deacetylase inhibition in vivo". FASEB J. 19 (9): 1166–8. doi:10.1096/fj.04-3425fje.PMID 15901671.
    92. ^ Smithells D (November 1998). "Does thalidomide cause second generation birth defects?". Drug Saf 19 (5): 339–41.doi:10.2165/00002018-199819050-00001.PMID 9825947.
    93. ^ Friedler G (December 1996). "Paternal exposures: impact on reproductive and developmental outcome. An overview".Pharmacol. Biochem. Behav. 55 (4): 691–700.doi:10.1016/S0091-3057(96)00286-9.PMID 8981601.
    94. ^ WebCite query result
    95. ^ Cicero TJ, Adams ML, Giordano A, Miller BT, O'Connor L, Nock B (March 1991). "Influence of morphine exposure during adolescence on the sexual maturation of male rats and the development of their offspring". J. Pharmacol. Exp. Ther. 256 (3): 1086–93. PMID 2005573.
    96. ^ Newbold RR, Padilla-Banks E, Jefferson WN (June 2006). "Adverse effects of the model environmental estrogen diethylstilbestrol are transmitted to subsequent generations".Endocrinology 147 (6 Suppl): S11–7.doi:10.1210/en.2005-1164.PMID 16690809.
    97. ^ Mandal SS (April 2010). "Mixed lineage leukemia: versatile player in epigenetics and human disease". FEBS J. 277 (8): 1789.doi:10.1111/j.1742-4658.2010.07605.x.PMID 20236314.
    98. ^ a b Li LC, Carroll PR, Dahiya R (January 2005). "Epigenetic changes in prostate cancer: implication for diagnosis and treatment". J. Natl. Cancer Inst. 97 (2): 103–15.doi:10.1093/jnci/dji010. PMID 15657340.
    99. ^ Ornish D, Magbanua MJ, Weidner G, Weinberg V, Kemp C, Green C, Mattie MD, Marlin R, Simko J, Shinohara K, Haqq CM, Carroll PR (June 2008). "Changes in prostate gene expression in men undergoing an intensive nutrition and lifestyle intervention". Proc. Natl. Acad. Sci. U.S.A. 105 (24): 8369–74.doi:10.1073/pnas.0803080105.PMC 2430265. PMID 18559852.
    100. ^ Beil, Laura (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).
    101. ^ a b Wong NC, Craig JM (2011). Epigenetics: A Reference Manual. Norfolk, England: Caister Academic Press. ISBN 1-904455-88-3.
    102. ^ Jasperson KW, Tuohy TM, Neklason DW, Burt RW (2010). Hereditary and familial colon cancer. Gastroenterology 138(6):2044-2058. doi: 10.1053/j.gastro.2010.01.054. PMID 20420945
    103. ^ Wood LD, Parsons DW, Jones S, Lin J, Sjöblom T, Leary RJ, Shen D, Boca SM, Barber T, Ptak J, Silliman N, Szabo S, Dezso Z, Ustyanksky V, Nikolskaya T, Nikolsky Y, Karchin R, Wilson PA, Kaminker JS, Zhang Z, Croshaw R, Willis J, Dawson D, Shipitsin M, Willson JK, Sukumar S, Polyak K, Park BH, Pethiyagoda CL, Pant PV, Ballinger DG, Sparks AB, Hartigan J, Smith DR, Suh E, Papadopoulos N, Buckhaults P, Markowitz SD, Parmigiani G, Kinzler KW, Velculescu VE, Vogelstein B (2007). The genomic landscapes of human breast and colorectal cancers. Science 318(5853):1108-1113. PMID 17932254
    104. ^ Esteller M, Silva JM, Dominguez G, Bonilla F, Matias-Guiu X, Lerma E, Bussaglia E, Prat J, Harkes IC, Repasky EA, Gabrielson E, Schutte M, Baylin SB, Herman JG (2000). Promoter hypermethylation and BRCA1 inactivation in sporadic breast and ovarian tumors. J Natl Cancer Inst 92(7) 564-569. PMID 10749912
    105. ^ Agrelo R, Cheng WH, Setien F, Ropero S, Espada J, Fraga MF, Herranz M, Paz MF, Sanchez-Cespedes M, Artiga J, Guerrero D, Castells A, von Kobbe C, Bohr VA, Esteller M (2006). Epigenetic inactivation of the premature aging Werner syndrome gene in human cancer. Proc Natl Acad Sci U S A 2006;103(23) 8822-8827. PMID 16723399 PMCID: PMC1466544
    106. ^ Baldwin RL, Nemeth E, Tran H, Shvartsman H, Cass I, Narod S, Karlan BY (2000). BRCA1 promoter region hypermethylation in ovarian carcinoma: a population-based study. Cancer Res 60(19):5329-5333. PMID 11034065
    107. ^ Shen L, Kondo Y, Rosner GL, Xiao L, Hernandez NS, Vilaythong J, Houlihan PS, Krouse RS, Prasad AR, Einspahr JG, Buckmeier J, Alberts DS, Hamilton SR, Issa JP (2005). MGMT promoter methylation and field defect in sporadic colorectal cancer. J Natl Cancer Inst 97(18) 1330-1338. PMID 16174854
    108. ^ Psofaki V, Kalogera C, Tzambouras N, Stephanou D, Tsianos E, Seferiadis K, Kolios G (2010). Promoter methylation status of hMLH1, MGMT, and CDKN2A/p16 in colorectal adenomas. World J Gastroenterol 16(28) 3553-3560. PMID 20653064 PMCID: PMC2909555
    109. ^ Lee KH, Lee JS, Nam JH, Choi C, Lee MC, Park CS, Juhng SW, Lee JH (2011). Promoter methylation status of hMLH1, hMSH2, and MGMT genes in colorectal cancer associated with adenoma-carcinoma sequence. Langenbecks Arch Surg 396(7) 1017-1026. PMID 21706233
    110. ^ Amatu A, Sartore-Bianchi A, Moutinho C, Belotti A, Bencardino K, Chirico G, Cassingena A, Rusconi F, Esposito A, Nichelatti M, Esteller M, Siena S (2013). Promoter CpG Island Hypermethylation of the DNA Repair Enzyme MGMT Predicts Clinical Response to Dacarbazine in a Phase II Study for Metastatic Colorectal Cancer. Clin Cancer Res [Epub ahead of print] PMID 23422094
    111. ^ Mokarram P, Zamani M, Kavousipour S, Naghibalhossaini F, Irajie C, Moradi Sarabi M, Hosseini SV (2012). Different patterns of DNA methylation of the two distinct O6-methylguanine-DNA methyltransferase (O(6)-MGMT) promoter regions in colorectal cancer. Mol Biol Rep Dec 28. [Epub ahead of print] PMID 23271133
    112. ^ Truninger K, Menigatti M, Luz J, Russell A, Haider R, Gebbers JO, Bannwart F, Yurtsever H, Neuweiler J, Riehle HM, Cattaruzza MS, Heinimann K, Schär P, Jiricny J, Marra G. Immunohistochemical analysis reveals high frequency of PMS2 defects in colorectal cancer. Gastroenterology 2005;128(5) 1160-1171. PMID 15887099
    113. ^ Facista A, Nguyen H, Lewis C, Prasad AR, Ramsey L, Zaitlin B, Nfonsam V, Krouse RS, Bernstein H, Payne CM, Stern S, Oatman N, Banerjee B, Bernstein C (2012). Deficient expression of DNA repair enzymes in early progression to sporadic colon cancer. Genome Integr 3(1): 3. PMID 22494821
    114. ^ Rigakos G, Razis E (2012). BRCAness: finding the Achilles heel in ovarian cancer. Oncologist 17(7):956-962. doi: 10.1634/theoncologist.2012-0028. Review. PMID 22673632
    115. ^ Stefansson OA, Villanueva A, Vidal A, Martí L, Esteller M (2012). BRCA1 epigenetic inactivation predicts sensitivity to platinum-based chemotherapy in breast and ovarian cancer. Epigenetics 7(11):1225-1229. doi: 10.4161/epi.22561. PMID 23069641
    116. ^ Chaisaingmongkol J, Popanda O, Warta R, Dyckhoff G, Herpel E, Geiselhart L, Claus R, Lasitschka F, Campos B, Oakes CC, Bermejo JL, Herold-Mende C, Plass C, Schmezer P (2012). Epigenetic screen of human DNA repair genes identifies aberrant promoter methylation of NEIL1 in head and neck squamous cell carcinoma. Oncogene 31(49):5108-16. doi: 10.1038/onc.2011.660. PMID 22286769
    117. ^ Fan CY (2004). Epigenetic alterations in head and neck cancer: prevalence, clinical significance, and implications. Curr Oncol Rep 6(2):152-161. Review. PMID 14751093
    118. ^ Koutsimpelas D, Pongsapich W, Heinrich U, Mann S, Mann WJ, Brieger J (2012). Promoter methylation of MGMT, MLH1 and RASSF1A tumor suppressor genes in head and neck squamous cell carcinoma: pharmacological genome demethylation reduces proliferation of head and neck squamous carcinoma cells. Oncol Rep 27(4):1135-41. doi: 10.3892/or.2012.1624.PMID 22246327
    119. ^ Sun W, Zaboli D, Liu Y, Arnaoutakis D, Khan T, Wang H, Koch W, Khan Z, Califano JA (2012). Comparison of promoter hypermethylation pattern in salivary rinses collected with and without an exfoliating brush from patients with HNSCC. PLoS One 7(3):e33642. doi: 10.1371/journal.pone.0033642. PMID 22438973
    120. ^ Puri SK, Si L, Fan CY, Hanna E (2005). Aberrant promoter hypermethylation of multiple genes in head and neck squamous cell carcinoma. Am J Otolaryngol 26(1):12-17. PMID 15635575
    121. ^ Ai L, Vo QN, Zuo C, Li L, Ling W, Suen JY, Hanna E, Brown KD, Fan CY (2004). Ataxia-telangiectasia-mutated (ATM) gene in head and neck squamous cell carcinoma: promoter hypermethylation with clinical correlation in 100 cases. Cancer Epidemiol Biomarkers Prev (1):150-6. PMID 14744748
    122. ^ Zuo C, Zhang H, Spencer HJ, Vural E, Suen JY, Schichman SA, Smoller BR, Kokoska MS, Fan CY (2009). Increased microsatellite instability and epigenetic inactivation of the hMLH1 gene in head and neck squamous cell carcinoma. Otolaryngol Head Neck Surg 141(4):484-490. doi: 10.1016/j.otohns.2009.07.007. PMID 19786217
    123. ^ Tawfik HM, El-Maqsoud NM, Hak BH, El-Sherbiny YM (2011). Head and neck squamous cell carcinoma: mismatch repair immunohistochemistry and promoter hypermethylation of hMLH1 gene. Am J Otolaryngol 32(6):528-536. doi: 10.1016/j.amjoto.2010.11.005. PMID 21353335
    124. ^ Narayanan L, Fritzell JA, Baker SM, Liskay RM, Glazer PM. (1997). Elevated levels of mutation in multiple tissues of mice deficient in the DNA mismatch repair gene Pms2. Proc Natl Acad Sci U S A 94(7):3122-3127. PMID 9096356
    125. ^ Hegan DC, Narayanan L, Jirik FR, Edelmann W, Liskay RM, Glazer PM. (2006). Differing patterns of genetic instability in mice deficient in the mismatch repair genes Pms2, Mlh1, Msh2, Msh3 and Msh6. Carcinogenesis. 2006 Dec;27(12):2402-2408. PMID 16728433
    126. ^ Tutt AN, van Oostrom CT, Ross GM, van Steeg H, Ashworth A. (2002). Disruption of Brca2 increases the spontaneous mutation rate in vivo: synergism with ionizing radiation. EMBO Rep. 3(3):255-260. PMID 11850397 PMCID: PMC1084010
    127. ^ German J. (1969). Bloom's syndrome. I. Genetical and clinical observations in the first twenty-seven patients. Am J Hum Genet. 1969 Mar;21(2):196-227. PMID 5770175 PMCID: PMC1706430
    128. ^ Nowak MA, Komarova NL, Sengupta A, Jallepalli PV, Shih IeM, Vogelstein B, Lengauer C (2002). The role of chromosomal instability in tumor initiation. Proc Natl Acad Sci U S A 99(25):16226-16231. PMID 12446840
    129. ^ Wang LG, Chiao JW (September 2010). "Prostate cancer chemopreventive activity of phenethyl isothiocyanate through epigenetic regulation (review)". Int. J. Oncol. 37 (3): 533–9.PMID 20664922.
    130. ^ a b Iglesias-Linares A, Yañez-Vico RM, González-Moles MA (May 2010). "Potential role of HDAC inhibitors in cancer therapy: insights into oral squamous cell carcinoma". Oral Oncol.46 (5): 323–9.doi:10.1016/j.oraloncology.2010.01.009.PMID 20207580.
    131. ^ Spannhoff A, Sippl W, Jung M (January 2009). "Cancer treatment of the future: inhibitors of histone methyltransferases". Int. J. Biochem. Cell Biol. 41 (1): 4–11.doi:10.1016/j.biocel.2008.07.024.PMID 18773966.
    132. ^ Dowden J, Hong W, Parry RV, Pike RA, Ward SG (April 2010). "Toward the development of potent and selective bisubstrate inhibitors of protein arginine methyltransferases". Bioorg. Med. Chem. Lett. 20 (7): 2103–5.doi:10.1016/j.bmcl.2010.02.069.PMID 20219369.
  1. ^http://physiology.eajbs.eg.net/pdf/vol2.n2/13.pdf
    1. ^ O'Connor, Anahad (2008-03-11). "The Claim: Identical Twins Have Identical DNA". New York Times. Retrieved 2010-05-02.
    2. ^ Kaminsky ZA, Tang T, Wang SC, Ptak C, Oh GH, Wong AH, Feldcamp LA, Virtanen C, Halfvarson J, Tysk C, McRae AF, Visscher PM, Montgomery GW, Gottesman II, Martin NG, Petronis A (February 2009). "DNA methylation profiles in monozygotic and dizygotic twins". Nat. Genet. 41 (2): 240–5. doi:10.1038/ng.286.PMID 19151718.
    3. ^ Fraga MF, Ballestar E, Paz MF, Ropero S, Setien F, Ballestar ML, Heine-Suñer D, Cigudosa JC, Urioste M, Benitez J, Boix-Chornet M, Sanchez-Aguilera A, Ling C, Carlsson E, Poulsen P, Vaag A, Stephan Z, Spector TD, Wu YZ, Plass C, Esteller M (July 2005). "Epigenetic differences arise during the lifetime of monozygotic twins".Proc. Natl. Acad. Sci. U.S.A. 102 (30): 10604–9.doi:10.1073/pnas.0500398102.PMC 1174919. PMID 16009939.
    4. ^ Casadesús J, Low D (September 2006)."Epigenetic gene regulation in the bacterial world". Microbiol. Mol. Biol. Rev. 70 (3): 830–56. doi:10.1128/MMBR.00016-06.PMC 1594586. PMID 16959970.
    5. ^ a b Jorg Tost (2008). Epigenetics. Norfolk, England: Caister Academic Press. ISBN 1-904455-23-9.
    6. ^ Lewis ZA, Honda S, Khlafallah TK, Jeffress JK, Freitag M, Mohn F, Schübeler D, Selker EU (March 2009). "Relics of repeat-induced point mutation direct heterochromatin formation in Neurospora crassa". Genome Res. 19 (3): 427–37. doi:10.1101/gr.086231.108.PMC 2661801. PMID 19092133.