Psychology Wiki
Register
Advertisement

Assessment | Biopsychology | Comparative | Cognitive | Developmental | Language | Individual differences | Personality | Philosophy | Social |
Methods | Statistics | Clinical | Educational | Industrial | Professional items | World psychology |

Biological: Behavioural genetics · Evolutionary psychology · Neuroanatomy · Neurochemistry · Neuroendocrinology · Neuroscience · Psychoneuroimmunology · Physiological Psychology · Psychopharmacology (Index, Outline)


This article needs rewriting to enhance its relevance to psychologists..
Please help to improve this page yourself if you can..




In biology, and specifically genetics, epigenetics is the study of heritable changes in gene expression or cellular phenotype caused by mechanisms other than changes in the underlying DNA sequence – hence the name epi- (Greek: επί- over, above, outer) -genetics. It refers to functionally relevant modifications to the genome that do not involve a change in the nucleotide sequence. Examples of such changes are DNA methylation and histone modification, both of which serve to regulate gene expression without altering the underlying DNA sequence. Conclusive evidence supporting epigenetics show that these mechanisms can enable the effects of parents' experiences to be passed down to subsequent generations.

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]

One example of epigenetic changes in eukaryotic biology is the process of cellular differentiation. During morphogenesis, 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.[3]

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-methyladenosine in RNA. This opened the related field of RNA epigenetics.[4][5]

Etymology and definitions[]

File:Epigenetic mechanisms.jpg

Epigenetic mechanisms

Epigenetics (as in "epigenetic landscape") was coined by C. H. Waddington in 1942 as a portmanteau of the words genetics and epigenesis.[6] Epigenesis is an old[7] word that has more recently been used (see preformationism for historical background) to describe the differentiation of cells from their initial totipotent 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."[8] Thus epigenetic can be used to describe anything other than DNA sequence that influences the development of an organism.

The modern usage of the word in scientific discourse is more narrow, referring to heritable traits (over rounds of cell division and sometimes transgenerationally) that do not involve changes to the underlying DNA sequence.[9] 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 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 an epigenomic 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 code or 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."[10] That usage, however, is of primarily historical interest.[11]

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 a mutation in the DNA has been caused in sperm or egg cell that results in fertilization, then some epigenetic changes are inherited from one generation to the next.[12] 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 of carcinogenesis, many effects of teratogens, regulation of histone modifications and heterochromatin, and technical limitations affecting parthenogenesis and cloning.

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 variants ChIP-on-chip and ChIP-Seq), fluorescent in situ hybridization, methylation-sensitive restriction enzymes, DNA adenine methyltransferase identification (DamID) and bisulfite sequencing. Furthermore, the use of bioinformatic methods is playing an increasing role (computational epigenetics).

Mechanisms[]

Several types of epigenetic inheritance systems may play a role in what has become known as cell memory:[13]

DNA methylation and chromatin remodeling[]

Nucleosome 1KX5 2

DNA associates with histone proteins to form chromatin.

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 convert cytosine 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/SNF complex (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 acetylation 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.[14]

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':[15] 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 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.[16] DNMT1 is the most abundant methyltransferase in somatic cells,[17] localizes to replication foci,[18] has a 10–40-fold preference for hemimethylated DNA and interacts with the proliferating cell nuclear antigen (PCNA).[19]

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.[20] DNMT1 is essential for proper embryonic development, imprinting and X-inactivation.[16][21]

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.[22]

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.[23] 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.[24][25]

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.[26]

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.[27]

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.[28] Other epigenetic changes are mediated by the production of different splice forms of RNA, 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 where 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.[29]

Prions[]

For more details on this topic, see 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.[30]

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.[31][32] 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.[33] 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 premature stop codon mutations.[34][35]

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.[36][37][38]

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"). In mammals, 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 plant cells do not have "memories", resetting their gene expression patterns at each cell division using positional information from the environment and surrounding cells to determine their fate.[39]

Medicine[]

Epigenetics has many and varied potential medical applications as it tends to be multidimensional in nature.[40] 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.[41]

Evolution[]

Epigenetic mechanisms were a necessary part of the evolutionary origin of cell differentiation.[42] 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. A sequestered germ line or Weismann barrier is specific to animals, and epigenetic inheritance is expected to be far more common in plants and microbes. These effects may require enhancements to the standard conceptual framework of the modern evolutionary synthesis.[43][44]

Epigenetic features may play a role in short-term adaptation of species by allowing for reversible phenotype variability. The modification of epigenetic features associated with a region of DNA allows organisms, on a multigenerational time scale, to switch between phenotypes that express and repress that particular gene.[45] When the DNA sequence of the region is not mutated, this change is reversible. It has also been speculated that organisms may take advantage of differential mutation rates associated with epigenetic features to control the mutation rates of particular genes.[45] Interestingly, 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.[46]

File:Epigenesis.jpg

Evolutionary 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.[47]

Epigenetic changes have also 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.[48][49]

More than 100 cases of transgenerational epigenetic inheritance phenomena have been reported in a wide range of organisms, including prokaryotes, plants, and animals.[50]

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.[51] 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.[52] 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[53] 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.[54] 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.[55]


Twin studies[]

Recent studies involving both dizygotic and monozygotic twins have produced some evidence of epigenetic influence in humans.[56][57][58]


See also[]

External links[]

References[]

  1. Bird A (May 2007). Perceptions of epigenetics. Nature 447 (7143): 396–8.
  2. Special report: 'What genes remember' by Philip Hunter | Prospect Magazine May 2008 issue 146. Web.archive.org. URL accessed on 2012-07-26.
  3. Reik W (May 2007). Stability and flexibility of epigenetic gene regulation in mammalian development. Nature 447 (7143): 425–32.
  4. 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.
  5. New research links common RNA modification to obesity. Physorg.com. URL accessed on 2012-07-26.
  6. Waddington CH (1942). The epigenotype. Endeavour 1: 18–20.
  7. According to the 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’.

    It is also worth quoting this adumbration of the definition given there (viz., "The formation of an organic germ as a new product"):

    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’.

  8. Holliday R (November 1990). Mechanisms for the control of gene activity during development. Biol Rev Camb Philos Soc 65 (4): 431–71.
  9. Riggs AD, Russo VEA, Martienssen RA (1996). Epigenetic mechanisms of gene regulation, Plainview, N.Y: Cold Spring Harbor Laboratory Press.
  10. Erikson, Erik (1968). Identity: Youth and Crisis, Chapter 3: W.W. Norton and Company, Inc..
  11. Epigenetics. Bio-Medicine.org. URL accessed on 2011-05-21.
  12. Chandler VL (February 2007). Paramutation: from maize to mice. Cell 128 (4): 641–5.
  13. 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.
  14. 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.
  15. Slotkin RK, Martienssen R (April 2007). Transposable elements and the epigenetic regulation of the genome. Nat. Rev. Genet. 8 (4): 272–85.
  16. 16.0 16.1 Li E, Bestor TH, Jaenisch R (June 1992). Targeted mutation of the DNA methyltransferase gene results in embryonic lethality. Cell 69 (6): 915–26.
  17. 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.
  18. 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.
  19. 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.
  20. Robertson KD, Wolffe AP (October 2000). DNA methylation in health and disease. Nat. Rev. Genet. 1 (1): 11–9.
  21. Li E, Beard C, Jaenisch R (November 1993). Role for DNA methylation in genomic imprinting. Nature 366 (6453): 362–5.
  22. Nottke A, Colaiácovo MP, Shi Y (March 2009). Developmental roles of the histone lysine demethylases. Development 136 (6): 879–89.
  23. 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.
  24. Epigenetic cell memory. Cmol.nbi.dk. URL accessed on 2012-07-26.
  25. Dodd IB, Micheelsen MA, Sneppen K, Thon G (May 2007). Theoretical analysis of epigenetic cell memory by nucleosome modification. Cell 129 (4): 813–22.
  26. Ptashne M (April 2007). On the use of the word 'epigenetic'. Curr. Biol. 17 (7): R233–6.
  27. 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.
  28. Mattick JS, Amaral PP, Dinger ME, Mercer TR, Mehler MF (January 2009). RNA regulation of epigenetic processes. BioEssays 31 (1): 51–9.
  29. Choi CQ. The Scientist: RNA can be hereditary molecule. The Scientist. URL accessed on 2006.
  30. Yool A, Edmunds WJ (1998). Epigenetic inheritance and prions. Journal of Evolutionary Biology 11 (2): 241–242.
  31. Cox BS (1965). [PSI], a cytoplasmic suppressor of super-suppression in yeast. Heredity 20 (4): 505–521.
  32. Lacroute F (May 1971). Non-Mendelian mutation allowing ureidosuccinic acid uptake in yeast. J. Bacteriol. 106 (2): 519–22.
  33. Liebman SW, Sherman F (September 1979). Extrachromosomal psi+ determinant suppresses nonsense mutations in yeast. J. Bacteriol. 139 (3): 1068–71.
  34. True HL, Lindquist SL (September 2000). A yeast prion provides a mechanism for genetic variation and phenotypic diversity. Nature 407 (6803): 477–83.
  35. Shorter J, Lindquist S (June 2005). Prions as adaptive conduits of memory and inheritance. Nat. Rev. Genet. 6 (6): 435–50.
  36. Sapp J (1991). Concepts of organization. The leverage of ciliate protozoa. Dev. Biol. (NY) 7: 229–58.
  37. Sapp J (2003). Genesis: the evolution of biology, Oxford [Oxfordshire]: Oxford University Press.
  38. 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.
  39. Costa S, Shaw P (March 2007). 'Open minded' cells: how cells can change fate. Trends Cell Biol. 17 (3): 101–6.
  40. 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.
  41. OMIM 105830
  42. Hoekstra RF (2000). Evolution: an introduction, Oxford [Oxfordshire]: Oxford University Press.
  43. Lamb MJ, Jablonka E (2005). Evolution in four dimensions: genetic, epigenetic, behavioral, and symbolic variation in the history of life, Cambridge, Mass: MIT Press.
  44. 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)
  45. 45.0 45.1 Rando OJ, Verstrepen KJ (February 2007). Timescales of genetic and epigenetic inheritance. Cell 128 (4): 655–68.
  46. Chahwan R, Wontakal SN, Roa S (October 2010). Crosstalk between genetic and epigenetic information through cytosine deamination. Trends Genet. 26 (10): 443–8.
  47. 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.
  48. 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.
  49. Waterland RA, Jirtle RL (August 2003). Transposable elements: targets for early nutritional effects on epigenetic gene regulation. Mol. Cell. Biol. 23 (15): 5293–300.
  50. 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.
  51. Wood AJ, Oakey RJ (November 2006). Genomic imprinting in mammals: emerging themes and established theories. PLoS Genet. 2 (11): e147.
  52. 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.
  53. A person's paternal grandson is the son of a son of that person; a maternal grandson is the son of a daughter.
  54. 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. Robert Winston refers to this study in a lecture; see also discussion at Leeds University, here [1]
  55. NOVA | Transcripts | Ghost in Your Genes. PBS. URL accessed on 2012-07-26.
  56. includeonly>O'Connor, Anahad. "The Claim: Identical Twins Have Identical DNA", New York Times, 2008-03-11. Retrieved on 2010-05-02.
  57. 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.
  58. 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.
  • Oskar Hertwig, 1849-1922. Biological problem of today: preformation or epigenesis? The basis of a theory of organic development. W. Heinemann: London, 1896.
  • R. Jaenisch and A. Bird (2003) Epigenetic regulation of gene expression: how the genome integrates intrinsic and environmental signals. Nat. Genet. 33 (Suppl) 245-254.
  • Joshua Lederberg, "The Meaning of Epigenetics", The Scientist 15(18):6, Sep. 17, 2001.
  • R. J. Sims III, K. Nishioka and D. Reinberg (2003) Histone lysine methylation: a signature for chromatin function. Trends Genet. 19, 629-637.
  • B. D. Strahl and C. D. Allis (2000) The language of covalent histone modifications. Nature 403, 41-45.
  • C.H. Waddington (1942), "The epigenotype". Endeavour 1, 18–20.
  • R.A. Waterland, R.L. Jirtle, "Transposable elements: Targets for early nutritional effects on epigenetic gene regulation", Molecular and Cellular Biology 2003 August 1;23(15):5293-5300.
  • B. McClintock (1978) Mechanisms that Rapidly Reorganize the Genome. Stadler Symposium vol 10:25-48
  • G.W. Grimes; K.J. Aufderheide; Cellular Aspects of Pattern Formation: the Problem of Assembly. Monographs in Developmental Biology, Vol. 22. Karger, Basel (1991)
  • Li E, Bestor TH, and Jaenisch R. Targeted mutation of the DNA methyltransferase gene results in embryonic lethality. Cell 69 , 915-926 (1992)
  • Li E, Beard C, and Jaenisch R. Role for DNA methylation in genomic imprinting. Nature 366 , 362-365 (1993)
  • Robertson KD, Uzyolgyi E, Liang G, Talmadge C, Sumegi J, Gonzales FA, and Jones PA. The human DNA methyltransferases (DNMTs) 1, 3a, 3b: Coordinate mRNA expression in normal tissues and overexpression in tumors. Nucleic Acids Res. 27 , 2291-2298 (1999)
  • Leonhardt H, Page A, Weier H, and Bestor TH. A targeting sequence directs DNA methyltransferase to sites of DNA replication in mammalian nuclei. Cell 71 , 865-873 (1992)
  • Chuang, L. Human DNA-(cytosine-5) methyltransferase-PCNA complex is target for p21 Waf1 . Science 277 , 1996-2000 (1997)
  • Robertson, K.D. and Wolffe, A.P. DNA methylation in health and disease. Nature Rev. Genet. 1 , 11-19 (2000)
The development of phenotype
Key concepts: Genotype-phenotype distinction | Norms of reaction | Gene-environment interaction | Heritability | Quantitative genetics
Genetic architecture: Dominance relationship | Epistasis | Polygenic inheritance | Pleiotropy | Plasticity | Canalisation | Fitness landscape
Non-genetic influences: Epigenetic inheritance | Epigenetics | Maternal effect | dual inheritance theory
Developmental architecture: Segmentation | Modularity
Evolution of genetic systems: Evolvability | Mutational robustness | Evolution of sex
Influential figures: C. H. Waddington | Richard Lewontin
Debates: Nature versus nurture
List of evolutionary biology topics

de:Epigenetik es:Epigenética fr:Épigénétique he:אפיגנטיקה hu:Epigenetika nl:Epigenetica sl:Epigenetika fi:Epigeneettinen periytyminen sv:Epigenetik zh:表觀遺傳學

This page uses Creative Commons Licensed content from Wikipedia (view authors).
Advertisement