fbpx
Wikipedia

Transgenerational epigenetic inheritance

Transgenerational epigenetic inheritance is the transmission of epigenetic markers and modifications from one generation to multiple subsequent generations without altering the primary structure of DNA.[1] Thus, the regulation of genes via epigenetic mechanisms can be heritable; the amount of transcripts and proteins produced can be altered by inherited epigenetic changes. In order for epigenetic marks to be heritable, however, they must occur in the gametes in animals, but since plants lack a definitive germline and can propagate, epigenetic marks in any tissue can be heritable.

"Intergenerational" vs "Transgenerational" Inheritance

It is important to note that the inheritance of epigenetic marks in the immediate generation is referred to as intergenerational inheritance.[2] In male mice, the epigenetic signal is maintained through the F1 generation.[3] In female mice, the epigenetic signal is maintained through the F2 generation as a result of the exposure of the germline in the womb.[3] Many epigenetic signals are lost beyond the F2/F3 generation and are no longer inherited, because the subsequent generations were not exposed to the same environment as the parental generations.[2] The signals that are maintained beyond the F2/F3 generation are referred to as transgenerational epigenetic inheritance (TEI), because initial environmental stimuli resulted in inheritance of epigenetic modifications.[4] There are several mechanisms of TEI that have shown to affect germline reprogramming, such as transgenerational increases in susceptibility to diseases, mutations, and stress inheritance. During germline reprogramming and early embryogenesis in mice, methylation marks are removed to allow for development to commence, but the methylation mark is converted into hydroxymethyl-cytosine so that it is recognized and methylated once that area of the genome is no longer being used,[5] which serves as a memory for that TEI mark. Therefore, under lab conditions, inherited methyl marks are removed and restored to ensure TEI still occurs. However, observing TEI in wild populations is still in its infancy, as laboratory studies allow for more tractable systems.[6]

Environmental factors can induce the epigenetic marks (epigenetic tags) for some epigenetically influenced traits.[1] These can include, but are not limited to, changes in temperature, resources availability, exposure to pollutants, chemicals, and endocrine disruptors.[7] The dosage and exposure levels can affect the extent of the environmental factors' influence over the epigenome and its effect on later generations. The epigenetic marks can result in a wide range of effects, including minor phenotypic changes to complex diseases and disorders.[7] The complex cell signaling pathways of multicellular organisms such as plants and humans can make understanding the mechanisms of this inherited process very difficult.[8]

Epigenetic categories edit

There are mechanisms by which environmental exposures induce epigenetic changes by affecting regulation and gene expression. Four general categories of epigenetic modification are known.

  1. self-sustaining metabolic loops, in which an mRNA or protein product of a gene stimulates transcription of the gene; e.g. Wor1 gene in Candida albicans;
  2. Structural templating: structures are replicated using a template or scaffold structure of the parent. This can include, but is not limited to, the orientation and architecture of cytoskeletal structures, cilia and flagella. Ciliates provide a good example of this type of modification. In an experiment Beisson and Sonneborn in 1985, it was demonstrated in Paramecium that if a section of cilia was removed and inverted, then the progeny of that Paramecium would also display the modified cilia structure for several generations.[9] Another example is seen in prions, special proteins that are capable of changing the structure of normal proteins to match their own. The prions use themselves as a template and then edit the folding of normal proteins to match their own folding pattern. The changes in the protein folding results in an alteration in the normal protein's function. This transmission of programming can also alter the chromatin and histone of the DNA and can be passed through the cytosol from parent to offspring during meiosis.[9]
  3. Histone modifications in which the structure of chromatin and its transcriptional state is regulated. DNA is wrapped into a DNA-protein complex called chromatin in the nucleus of eukaryotic cells.[10] Chromatin consists of DNA and nucleosomes that comes together to form a histone octamer.[11] The N- and C- terminal of the histone proteins are post-translationally modified by the removal or addition of acetyl (acetylation), phosphate (phosphorylation), methyl (methylation), ubiquitin (ubiquitination), and ubiquitin-like modifier (SUMOylating) groups.[10] Histone modifications can be transgenerational epigenetic signals. For example, histone H3K4 trimethylation (H34me3) and a network of lipid metabolic genes interact to increase the transcription response to TEI obesogenic effects.[12] TEI can also be observed in Drosophila embryos through the exposure of heat stress over generations.[3] The induced heat stress resulted in the phosphorylation of ATF-2 (dATF-2) which is required for heterochromatin assembly.[13] This epigenetic event was maintained over multiple generations, but over time dATF-2 returned back to its normal state.[13]
  4. Non-coding and coding RNAs in which various classes of RNA is implicated in TEI through maternal stores of mRNA, translation of mRNA (miRNA), and small RNA strands interfering with transcription (piRNAs and siRNAs) via RNA interference pathways (RNAi).[2] There has been an increase in studies reporting noncoding RNA contributions to TEI. For example, altered miRNA in early trauma mice.[14] Early trauma mice with unpredictable maternal separation and maternal stress (MSUS) were used as a model to identify the effects of altered miRNA in sperm.[15] In MSUS mice, behavior responses were affected, insulin levels, and blood glucose levels were decreased.[15] Notably, these effects were more severe across the F2 and F3 generation. The expression of miRNA in MSUS mice was down regulated in the brain, serum, and sperm of the F1 generation.[15] However, the miRNA was not altered in the sperm of the F2 generation, and the miRNAs were normal in the F3 generation.[15] This provides supportive evidence that the initial alterations in miRNAs in sperm are transferred to epigenetic marks to maintain transmission.[16] In C.elegans, starvation is induced in which survival is dependent on the mechanisms of the RNAi pathway, repression of microRNAs, and regulation of small RNAs.[17] Thus, memorization of dietary history is inherited across generations.[17]

Inheritance of epigenetic marks edit

Although there are various forms of inheriting epigenetic markers, inheritance of epigenetic markers can be summarized as the dissemination of epigenetic information by means of the germline.[18] Furthermore, epigenetic variation typically takes one of four general forms, though there are other forms that have yet to be elucidated. Currently, self-sustaining feedback loops, spatial templating, chromatin marking, and RNA-mediated pathways modify epigenes of individual cells. Epigenetic variation within multicellular organisms is either endogenous or exogenous.[19] Endogenous is generated by cell–cell signaling (e.g. during cell differentiation early in development), while exogenous is a cellular response to environmental cues.[citation needed]

Removal vs. retention edit

In sexually reproducing organisms, much of the epigenetic modification within cells is reset during meiosis (e.g. marks at the FLC locus controlling plant vernalization[20]), though some epigenetic responses have been shown to be conserved (e.g. transposon methylation in plants[20]). Differential inheritance of epigenetic marks due to underlying maternal or paternal biases in removal or retention mechanisms may lead to the assignment of epigenetic causation to some parent of origin effects in animals[21] and plants.[22]

Reprogramming edit

In mammals, epigenetic marks are erased during two phases of the life cycle. Firstly just after fertilization and secondly, in the developing primordial germ cells, the precursors to future gametes.[23] During fertilization the male and female gametes join in different cell cycle states and with different configuration of the genome. The epigenetic marks of the male are rapidly diluted. First, the protamines associated with male DNA are replaced with histones from the female's cytoplasm, most of which are acetylated due to either higher abundance of acetylated histones in the female's cytoplasm or through preferential binding of the male DNA to acetylated histones.[24][25] Second, male DNA is systematically demethylated in many organisms,[26][27] possibly through 5-hydroxymethylcytosine. However, some epigenetic marks, particularly maternal DNA methylation, can escape this reprogramming; leading to parental imprinting.

In the primordial germ cells (PGC) there is a more extensive erasure of epigenetic information. However, some rare sites can also evade erasure of DNA methylation.[28] If epigenetic marks evade erasure during both zygotic and PGC reprogramming events, this could enable transgenerational epigenetic inheritance.[citation needed]

Recognition of the importance of epigenetic programming to the establishment and fixation of cell line identity during early embryogenesis has recently stimulated interest in artificial removal of epigenetic programming.[29] Epigenetic manipulations may allow for restoration of totipotency in stem cells or cells more generally, thus generalizing regenerative medicine.

Retention edit

Cellular mechanisms may allow for co-transmission of some epigenetic marks. During replication, DNA polymerases working on the leading and lagging strands are coupled by the DNA processivity factor proliferating cell nuclear antigen (PCNA), which has also been implicated in patterning and strand crosstalk that allows for copy fidelity of epigenetic marks.[30][31] Work on histone modification copy fidelity has remained in the model phase, but early efforts suggest that modifications of new histones are patterned on those of the old histones and that new and old histones randomly assort between the two daughter DNA strands.[32] With respect to transfer to the next generation, many marks are removed as described above. Emerging studies are finding patterns of epigenetic conservation across generations. For instance, centromeric satellites resist demethylation.[33] The mechanism responsible for this conservation is not known, though some evidence suggests that methylation of histones may contribute.[33][34] Dysregulation of the promoter methylation timing associated with gene expression dysregulation in the embryo was also identified.[35]

Decay edit

Whereas the mutation rate in a given 100-base gene may be 10−7 per generation, epigenes may "mutate" several times per generation or may be fixed for many generations.[36] This raises the question: do changes in epigene frequencies constitute evolution? Rapidly decaying epigenetic effects on phenotypes (i.e. lasting less than three generations) may explain some of the residual variation in phenotypes after genotype and environment are accounted for. However, distinguishing these short-term effects from the effects of the maternal environment on early ontogeny remains a challenge.[citation needed]

Examples of TEI edit

The relative importance of genetic and epigenetic inheritance is subject to debate. Though hundreds of examples of epigenetic modification of phenotypes have been published, few studies have been conducted outside of the laboratory setting. Therefore, the interactions of genes with the environment cannot be inferred despite the central role of environment in natural selection. Multiple epigenetic factors can influence the state of genes and alter the epigenetic state. Due to the multivariate nature of environmental factors, it is difficult for researchers to pinpoint the exact cause of epigenetic changes outside of a laboratory setting.[37]

In Plants edit

Studies concerning transgenerational epigenetic inheritance in plants have been reported as early as the 1950s.[38] One of the earliest and best characterized examples of this is b1 paramutation in maize.[38][39][40][41][42][43][44][45] The b1 gene encodes a basic helix-loop-helix transcription factor that is involved in the anthocyanin production pathway. When the b1 gene is expressed, the plant accumulates anthocyanin within its tissues, leading to a purple coloration of those tissues. The B-I allele (for B-Intense) has high expression of b1 resulting in the dark pigmentation of the sheath and husk tissues while the B' (pronounced B-prime) allele has low expression of b1 resulting in low pigmentation in those tissues.[46] When homozygous B-I parents are crossed to homozygous B', the resultant F1 offspring all display low pigmentation which is due to gene silencing of b1.[38][46] Unexpectedly, when F1 plants are self-crossed, the resultant F2 generation all display low pigmentation and have low levels of b1 expression. Furthermore, when any F2 plant (including those that are genetically homozygous for B-I) are crossed to homozygous B-I, the offspring will all display low pigmentation and expression of b1.[38][46] The lack of darkly pigmented individuals in the F2 progeny is an example of non-Mendelian inheritance and further research has suggested that the B-I allele is converted to B' via epigenetic mechanisms.[40][41] The B' and B-I alleles are considered to be epialleles because they are identical at the DNA sequence level but differ in the level of DNA methylation, siRNA production, and chromosomal interactions within the nucleus.[44][47][43][42] Additionally, plants defective in components of the RNA-directed DNA-methylation pathway show an increased expression of b1 in B' individuals similar to that of B-I, however, once these components are restored, the plant reverts to the low expression state.[45][48][49][50] Although spontaneous conversion from B-I to B' has been observed, a reversion from B' to B-I (green to purple) has never been observed over 50 years and thousands of plants in both greenhouse and field experiments.[51]

Examples of environmentally induced transgenerational epigenetic inheritance in plants has also been reported. In one case, rice plants that were exposed to drought-simulation treatments displayed increased tolerance to drought after 11 generations of exposure and propagation by single-seed descent as compared to non-drought treated plants. Differences in drought tolerance was linked to directional changes in DNA-methylation levels throughout the genome, suggesting that stress-induced heritable changes in DNA-methylation patterns may be important in adaptation to recurring stresses. In another study, plants that were exposed to moderate caterpillar herbivory over multiple generations displayed increased resistance to herbivory in subsequent generations (as measured by caterpillar dry mass) compared to plants lacking herbivore pressure. This increase in herbivore resistance persisted after a generation of growth without any herbivore exposure suggesting that the response was transmitted across generations. The report concluded that components of the RNA-directed DNA-methylation pathway are involved in the increased resistance across generations. Transgenerational epigenetic inheritance has also been observed in polyploid plants. Genetically identical reciprocal F1 hybrid triploids have been shown to display transgenerational epigenetic effects on viable F2 seed development.

It has been demonstrated in wild radish plants (Raphanus raphanistrum) that TEI can be induced when the plants are exposed to predators such as Pieris rapae, the cabbage white caterpillar. The radish plants will increase production of bristly leaf hairs and toxic mustard oil in response to caterpillar predation. The increased levels will also be seen in the next generation. Decreased levels of predation also results in decreased leaf hairs and toxins produced in the current and subsequent generations.[52]

In Animals edit

It is difficult to trace TEI in animals due to the reprogramming of genes during meiosis and embryogenesis, especially in wild populations that are not reared in a lab setting. Further studies must be conducted to strengthen the documentation of TEI in animals. However, a few examples do exist.

Induced transgenerational epigenetic inheritance has been demonstrated in animals, such as Daphnia cucullata. These tiny crustaceans will develop protective helmets as juveniles if exposed to kairomones, a type of hormone, secreted by predators while they are in utero. The helmet acts as a method of defense by decreasing the ability of predators to capture the Daphnia, thus induction of helmet presence will lower mortality rates. D. cucullata will develop a small helmet if no kairomones are present. However, depending upon the level of predator kairomones, the length of the helmet will almost double. The next generation of Daphnia will display a similar helmet size. If the kairomone levels decrease or disappear, then the third generation will revert to the original helmet size. These organisms display adaptive phenotypes that will affect the phenotype in the subsequent generations.[53]

Genetic analysis of coral reef fish, Acanthochromis polyacanthus, has proposed TEI in response to climate change. As climate change occurs, the ocean water temperature increases. When A. polyacanthus is exposed to higher water temperatures of up to +3 °C from normal ocean temperatures, the fish express increased DNA methylation levels on 193 genes, resulting in phenotypic changes in the function of oxygen consumption, metabolism, insulin response, energy production, and angiogenesis. The increase in DNA methylation and its phenotypic affects were carried over to multiple subsequent generations.[54]

Possible TEI has been studied in guinea pigs (Cavia aperea) by exposing males to increased ambient temperature for two months. In the lab, the males were allowed to mate with the same female before and after the heat exposure to determine if the high temperatures affected the offspring. Since it serves as a thermoregulatory organ, samples of the liver were studied in the father guinea pigs (F0 generation) and liver and testes of the male offspring (F1 generation). The F0 males experienced an immediate epigenetic response to the increase in temperature; the levels of hormones in the liver responsible for thermoregulation increased. The F1 generation also displayed the different methylated epigenetic response in their liver and testes, indicating that they could potentially pass on the epigenetic marks to the F2 generation.[55]

In Humans edit

Although genetic inheritance is important when describing phenotypic outcomes, it cannot entirely explain why offspring resemble their parents. Aside from genes, offspring come to inherit similar environmental conditions established by previous generations. One environment that human offspring commonly share with their maternal parent for nine months is the womb. Considering the duration of the fetal stages of development, the environment of the mother's womb can have long lasting effects on the health of offspring.

An example of how the environment within the womb can affect the health of an offspring is the Dutch hunger winter of 1944-45 and its causal effect on induced transgenerational epigenetic inherited diseases. During the Dutch hunger winter, the offspring exposed to famine conditions during the third trimester of development were smaller than those born the year before the famine. Moreover, the offspring born during the famine and their subsequent offspring were found to have an increased risk of metabolic diseases, cardiovascular diseases, glucose intolerance, diabetes, and obesity in adulthood. The effects of this famine on development lasted up to two generations.[8][56] The increased risk factors to the health of F1 and F2 generations during the Dutch hunger winter is a known phenomenon called “fetal programming,” which is caused by exposure to harmful environmental factors in utero.[56]

The loss of genetic expression which results in Prader–Willi syndrome or Angelman syndrome has in some cases been found to be caused by epigenetic changes (or "epimutations") on both the alleles, rather than involving any genetic mutation. In all 19 informative cases, the epimutations that, together with physiological imprinting and therefore silencing of the other allele, were causing these syndromes were localized on a chromosome with a specific parental and grandparental origin. Specifically, the paternally derived chromosome carried an abnormal maternal mark at the SNURF-SNRPN, and this abnormal mark was inherited from the paternal grandmother.[57]

Several cancers have been found to be influenced by transgenerational epigenetics. Epimutations on the MLH1 gene has been found in two individuals with a phenotype of hereditary nonpolyposis colorectal cancer, and without any frank MLH1 mutation which otherwise causes the disease. The same epimutations were also found on the spermatozoa of one of the individuals, indicating the potential to be transmitted to offspring.[57] In addition to epimutations to the MLH1 gene, it has been determined that certain cancers, such as breast cancer, can originate during the fetal stages within the uterus.[58] Furthermore, evidence collected in various studies utilizing model systems (i.e. animals) have found that exposure during parental generations can result in multigenerational and transgenerational inheritance of breast cancer.[58] More recently, studies have discovered a connection between the adaptation of male germinal cells via pre-conception paternal diets and the regulation of breast cancer in developing offspring.[58] More specifically, studies have begun to uncover new data that underscores a relationship between transgenerational epigenetic inheritance of breast cancer and ancestral alimentary components or associated markers, such as birth weight.[58] By utilizing model systems, such as mice, studies have shown that stimulated paternal obesity at the time of conception can epigenetically alter the paternal germ-line. The paternal germ-line is responsible for regulating their daughters’ weight at birth and the potential for their daughter to develop breast cancer.[59] Furthermore, it was found that modifications to the miRNA expression profile of the male germline is coupled with elevated body weight.[59] Additionally, paternal obesity resulted in an increase in the percentage of female offspring developing carcinogen-induced mammary tumors, which is caused by changes to mammary miRNA expression.[59]

Aside from cancer related afflictions associated with the effects of transgenerational epigenetic inheritance, transgenerational epigenetic inheritance has recently been implicated in the progression of pulmonary arterial hypertension (PAH).[60] Recent studies have found that transgenerational epigenetic inheritance is likely to be involved in the progression of PAH because current therapies for PAH do not repair the irregular phenotypes associated with this disease.[60] Current treatments for PAH have attempted to correct symptoms of PAH with vasodilators and antithrombotic protectors, but neither has effectively alleviated the complications related to the impaired phenotypes associated with PAH.[60] The inability of vasodilators and antithrombotic protectants to correct PAH suggests that the progression of PAH is dependent upon multiple variables, which is likely to be consequent of transgenerational epigenetic inheritance.[60] Specifically, it is thought that transgenerational epigenetics is linked to the phenotypic changes associated with vascular remodeling.[60] For example, hypoxia during gestation may induce transgenerational epigenetic alterations that could prove to be detrimental during the early phases of fetal development and increase the possibility of developing PAH as an adult.[60] Though hypoxic states could induce the transgenerational epigenetic variance associated with PAH, there is strong evidence to support that a variety of maternal risk factors are linked to the eventual progression of PAH.[60] Such maternal risk factors linked to late-onset PAH includes placental dysfunction, hypertension, obesity, and preeclampsia.[60] These maternal risk factors and environmental stressors coupled with transgenerational epigenetic changes can result in prolonged insult to the signaling pathways associated with the vascular development during fetal stages, thus increasing the likelihood of having PAH.[60]

One study has shown childhood abuse, which is defined as "sexual contact, severe physical abuse and/or severe neglect," leads to epigenetic modifications of glucocorticoid receptor expression.[61][62] Glucocorticoid receptor expression plays a vital role in hypothalamic-pituitary-adrenal (HPA) activity. Additionally, animal experiments have shown that epigenetic changes can depend on mother-infant interactions after birth.[63] Furthermore, a recent study investigating the correlations between maternal stress in pregnancy and methylation in teenagers/their mothers has found that children of women who were abused during pregnancy were more likely to have methylated glucocorticoid-receptor genes.[64] Thus, children with methylated glucocorticoid-receptor genes experience an altered response to stress, ultimately leading to a higher susceptibility of experiencing anxiety.[64]

Additional studies examining the effects of diethylstilbestrol (DES), which is an endocrine disruptor, have found that the grandchildren (third-generation) of women exposed to DES significantly increased the probability of their grandchildren developing attention-deficit/hyperactivity disorder (ADHD).[65] This is because women exposed to endocrine disruptors, such as DES, during gestation may be linked to multigenerational neurodevelopmental deficits.[65] Furthermore, animal studies indicate that endocrine disruptors have a profound impact on germline cells and neurodevelopment.[65] The cause of DES's multigenerational impact is postulated to be the result of biological processes associated with epigenetic reprogramming of the germline, though this has yet to be determined.[65]

Effects on fitness edit

Epigenetic inheritance may only affect fitness if it predictably alters a trait under selection. Evidence has been forwarded that environmental stimuli are important agents in the alteration of epigenes. Ironically, Darwinian evolution may act on these neo-Lamarckian acquired characteristics as well as the cellular mechanisms producing them (e.g. methyltransferase genes). Epigenetic inheritance may confer a fitness benefit to organisms that deal with environmental changes at intermediate timescales.[66] Short-cycling changes are likely to have DNA-encoded regulatory processes, as the probability of the offspring needing to respond to changes multiple times during their lifespans is high. On the other end, natural selection will act on populations experiencing changes on longer-cycling environmental changes. In these cases, if epigenetic priming of the next generation is deleterious to fitness over most of the interval (e.g. misinformation about the environment), these genotypes and epigenotypes will be lost. For intermediate time cycles, the probability of the offspring encountering a similar environment is sufficiently high without substantial selective pressure on individuals lacking a genetic architecture capable of responding to the environment. Naturally, the absolute lengths of short, intermediate, and long environmental cycles will depend on the trait, the length of epigenetic memory, and the generation time of the organism. Much of the interpretation of epigenetic fitness effects centers on the hypothesis that epigenes are important contributors to phenotypes, which remains to be resolved.

Deleterious effects edit

Inherited epigenetic marks may be important for regulating important components of fitness. In plants, for instance, the Lcyc gene in Linaria vulgaris controls the symmetry of the flower. Linnaeus first described radially symmetric mutants, which arise when Lcyc is heavily methylated.[67] Given the importance of floral shape to pollinators,[68] methylation of Lcyc homologues (e.g. CYCLOIDEA) may have deleterious effects on plant fitness. In animals, numerous studies have shown that inherited epigenetic marks can increase susceptibility to disease. Transgenerational epigenetic influences are also suggested to contribute to disease, especially cancer, in humans.[69] Tumor methylation patterns in gene promoters have been shown to correlate positively with familial history of cancer.[70] Furthermore, methylation of the MSH2 gene is correlated with early-onset colorectal and endometrial cancers.[71]

Putatively adaptive effects edit

Experimentally demethylated seeds of the model organism Arabidopsis thaliana have significantly higher mortality, stunted growth, delayed flowering, and lower fruit set,[72] indicating that epigenes may increase fitness. Furthermore, environmentally induced epigenetic responses to stress have been shown to be inherited and positively correlated with fitness.[73] In animals, communal nesting changes mouse behavior increasing parental care regimes[74] and social abilities[75] that are hypothesized to increase offspring survival and access to resources (such as food and mates), respectively.

Inheritance of Immunity edit

Epigenetics play a crucial role in regulation and development of the immune system.[76] In 2021, evidence of inheritance of trained immunity across generations to progeny of mice with a systemic infection of Candida albicans was provided.[77] The progeny of mice survived the Candida albicans infection via functional, transcriptional, and epigenetic changes linked to the immune gene loci.[77] The responsiveness of myeloid cells to the Candida albicans infection increased in inflammatory pathways, and resistance was increased to infections in the next generations.[77] Immunity in vertebrates can also be transferred from maternal through the passing of hormones, nutrients and antibodies.[78] In mammals, the maternal factors can be transferred via lactation or the placenta.[78] The transgenerational transmission of immune-related traits are also described in plants and invertebrates. Plants have a defense priming system which enables them to have an alternate defense response that can be accelerated upon exposure to stress actions or pathogens.[79] After the event of priming, priming stress clue information is stored, and the memory may be inherited in the offspring (intergenerational or transgenerational).[79] In studies, the progeny of Pseudomonas syringae infected Arabidopsis were primed during the expression of systemic acquired resistance (SAR).[80] The progeny showed to have resistance against (hemi)-biotrophic pathogens which is associated with salicylic dependent genes and the defense regulatory gene, non expressor of PR genes (NPR1).[80] Transgenerational SAR in the progeny was associated with increased acetylation of histone 3 at lysine 9, hypomethylation of genes, and chromatin marks on promoter regions of salicylic dependent genes.[80] Similarly in insects, the red flour beetle Trifolium castoreum is primed through the exposure of the pathogen Bacillus thuringiensis.[78] Double-mating experiments with the red flour beetle demonstrated that paternal transgenerational immune priming is mediated by sperm or seminal fluid which enhances survival upon exposure to pathogens and contribute to epigenetic changes.[78]

Feedback loops and TEI edit

Positive and negative feedback loops are commonly observed in molecular mechanisms and regulation of homeostatic processes. There is evidence that feedback loops interact to maintain epigenetic modifications within one generation, as well as contributing to TEI in various organisms, and these feedback loops can showcase putative adaptations to environmental perturbances. Feedback loops are truly a repercussion of any epigenetic modification, since it results in changes in expression. Even more so, the feedback loops seen across multiple generations because of TEI showcases a spatio-temporal dynamic that is associated with TEI alone. For example, elevated temperatures during embryogenesis and PIWI RNA (piRNA) establishment are directly proportional, providing a heritable outcome for repressing transposable elements via piRNA clusters.[81] Furthermore, subsequent generations retain an active locus to continue establishing piRNA, which its formation was previously enigmatic.[81] In another case, it was suggested that endocrine disruption had a feedback loop interaction with methylation of varying genomic sites in Menidia beryllina, which may have been a function of TEI.[82] When exposure was removed, and M. beryllina F2 offspring still retained these methylation marks, which caused a negative feedback loop on expression of various genes.[82] In another example, hybridization of eels can lead to feedback loops contributing to transposon demethylation and transposable element activation.[83] Because TE's are typically silenced in the genome, their presence and potential expression creates a feedback loop to prevent hybrids from reproducing with other hybrids or non-hybrid species, which eliminates the proliferation of TE expression and prevents TEI in this context. This phenomenon is known as a form of post-zygotic reproductive isolation.

Macroevolutionary patterns edit

Inherited epigenetic effects on phenotypes have been well documented in bacteria, protists, fungi, plants, nematodes, and fruit flies.[84][18] Though no systematic study of epigenetic inheritance has been conducted (most focus on model organisms), there is preliminary evidence that this mode of inheritance is more important in plants than in animals.[84] The early differentiation of animal germlines is likely to preclude epigenetic marking occurring later in development, while in plants and fungi somatic cells may be incorporated into the germ line.[85][86]

It is thought that transgenerational epigenetic inheritance can enable certain populations to readily adapt to variable environments.[18] Though there are well documented cases of transgenerational epigenetic inheritance in certain populations, there are questions to whether this same form of adaptability is applicable to mammals.[18] More specifically, it is questioned if it applies to humans.[18] As of late, most of the experimental models utilizing mice and limited observations in humans have only found epigenetically inherited traits that are detrimental to the health of both organisms.[18] These harmful traits range from increased risk of disease, such as cardiovascular disease, to premature death.[18] However, this may be based on the premise of limited reporting bias because it is easier to detect negative experimental effects, opposed to positive experimental effects.[18] Furthermore, considerable epigenetic reprogramming necessary for the evolutionary success of germlines and the initial phases of embryogenesis in mammals may be the potential cause limiting transgenerational inheritance of chromatin marks in mammals.[18]  

Life history patterns may also contribute to the occurrence of epigenetic inheritance. Sessile organisms, those with low dispersal capability, and those with simple behavior may benefit most from conveying information to their offspring via epigenetic pathways. Geographic patterns may also emerge, where highly variable and highly conserved environments might host fewer species with important epigenetic inheritance.[citation needed]

Controversies edit

Humans have long recognized that traits of the parents are often seen in offspring. This insight led to the practical application of selective breeding of plants and animals, but did not address the central question of inheritance: how are these traits conserved between generations, and what causes variation? Several positions have been held in the history of evolutionary thought.

Blending vs. particulate inheritance edit

 
Blending inheritance leads to the averaging out of every characteristic, which as the engineer Fleeming Jenkin pointed out, makes evolution by natural selection impossible.

Addressing these related questions, scientists during the time of the Enlightenment largely argued for the blending hypothesis, in which parental traits were homogenized in the offspring much like buckets of different colored paint being mixed together.[87] Critics of Charles Darwin's On the Origin of Species, pointed out that under this scheme of inheritance, variation would quickly be swamped by the majority phenotype.[88] In the paint bucket analogy, this would be seen by mixing two colors together and then mixing the resulting color with only one of the parent colors 20 times; the rare variant color would quickly fade.

Unknown to most of the European scientific community, the monk Gregor Mendel had resolved the question of how traits are conserved between generations through breeding experiments with pea plants.[89] Charles Darwin thus did not know of Mendel's proposed "particulate inheritance" in which traits were not blended but passed to offspring in discrete units that we now call genes. Darwin came to reject the blending hypothesis even though his ideas and Mendel's were not unified until the 1930s, a period referred to as the modern synthesis.

Inheritance of innate vs. acquired characteristics edit

In his 1809 book, Philosophie Zoologique,[90] Jean-Baptiste Lamarck recognized that each species experiences a unique set of challenges due to its form and environment. Thus, he proposed that the characters used most often would accumulate a "nervous fluid." Such acquired accumulations would then be transmitted to the individual's offspring. In modern terms, a nervous fluid transmitted to offspring would be a form of epigenetic inheritance.

Lamarckism, as this body of thought became known, was the standard explanation for change in species over time when Charles Darwin and Alfred Russel Wallace co-proposed a theory of evolution by natural selection in 1859. Responding to Darwin and Wallace's theory, a revised neo-Lamarckism attracted a small following of biologists,[91] though the Lamarckian zeal was quenched in large part due to Weismann's[92] famous experiment in which he cut off the tails of mice over several successive generations without having any effect on tail length. Thus the emergent consensus that acquired characteristics could not be inherited became canon.[23]

Revision of evolutionary theory edit

Non-genetic variation and inheritance, however, proved to be quite common. Concurrent with the 20th-century development of the modern evolutionary synthesis (unifying Mendelian genetics and natural selection), C. H. Waddington (1905-1975) was working to unify developmental biology and genetics. In so doing, he adopted the word "epigenetic"[93] to represent the ordered differentiation of embryonic cells into functionally distinct cell types despite having identical primary structure of their DNA.[94] Researchers discussed Waddington's epigenetics sporadically - it became more of a catch-all for puzzling non-genetic heritable characters rather than a concept advancing the body of inquiry.[95][96] Consequently, the definition of Waddington's word has itself evolved, broadening beyond the subset of developmentally signaled, inherited cell specialization.

Some scientists have questioned whether epigenetic inheritance compromises the foundation of the modern synthesis. Outlining the central dogma of molecular biology, Francis Crick[97] succinctly stated, "DNA is held in a configuration by histone[s] so that it can act as a passive template for the simultaneous synthesis of RNA and protein[s]. None of the detailed 'information' is in the histone." However, he closes the article stating, "this scheme explains the majority of the present experimental results!" Indeed, the emergence of epigenetic inheritance (in addition to advances in the study of evolutionary-development, phenotypic plasticity, evolvability, and systems biology) has strained the current framework of the modern evolutionary synthesis, and prompted the re-examination of previously dismissed evolutionary mechanisms.[98]

Furthermore, patterns in epigenetic inheritance and the evolutionary implications of the epigenetic codes in living organisms are connected to both Lamarck's and Darwin's theories of evolution.[99] For example, Lamarck postulated that environmental factors were responsible for modifying phenotypes hereditarily, which supports the constructs that exposure to environmental factors during critical stages of development can result in epimutations in germlines, thus augmenting phenotypic variance.[99] In contrast, Darwin's theory claimed that natural selection strengthened a populations ability to survive and remain reproductively fit by favoring populations that are able to readily adapt.[99] This theory is consistent with intergenerational plasticity and phenotypic variance resulting from heritable adaptivity.[99]

In addition, some epigenetic variability may provide beneficial plasticity, so that certain organisms can adapt to fluctuating environmental conditions. However, the exchange of epigenetic information between generations can result in epigenetic aberrations, which are epigenetic traits that deviate from the norm. Therefore, the offspring of the parental generations may be predisposed to specific diseases and reduced plasticity due to epigenetic aberrations. Though the ability to readily adapt when faced with a new environment may be beneficial to certain populations of species that can quickly reproduce, species with long generational gaps may not benefit from such an ability. If a species with a longer generational gap does not appropriately adapt to the anticipated environment, then the reproductive fitness of the offspring of that species will be diminished.

There has been critical discussion of mainstream evolutionary theory by Edward J Steele, Robyn A Lindley and colleagues,[100][101][102][103][104] Fred Hoyle and N. Chandra Wickramasinghe,[105][106][107] Yongsheng Liu[108][109] Denis Noble,[110][111] John Mattick[112] and others that the logical inconsistencies as well as Lamarckian Inheritance effects involving direct DNA modifications, as well as the just described indirect, viz. epigenetic, transmissions, challenge conventional thinking in evolutionary biology and adjacent fields.

See also edit

References edit

  1. ^ a b Moore, David Scott (2015). The developing genome : an introduction to behavioral epigenetics. Oxford. ISBN 978-0-19-992235-2. OCLC 899240120.{{cite book}}: CS1 maint: location missing publisher (link)
  2. ^ a b c Heard, Edith; Martienssen, Robert A. (2014-03-27). "Transgenerational Epigenetic Inheritance: Myths and Mechanisms". Cell. 157 (1): 95–109. doi:10.1016/j.cell.2014.02.045. ISSN 0092-8674. PMC 4020004. PMID 24679529.
  3. ^ a b c Fitz-James, Maximilian H.; Cavalli, Giacomo (June 2022). "Molecular mechanisms of transgenerational epigenetic inheritance". Nature Reviews Genetics. 23 (6): 325–341. doi:10.1038/s41576-021-00438-5. ISSN 1471-0064. PMID 34983971. S2CID 245703043.
  4. ^ Fitz-James, Maximilian H.; Cavalli, Giacomo (June 2022). "Molecular mechanisms of transgenerational epigenetic inheritance". Nature Reviews Genetics. 23 (6): 325–341. doi:10.1038/s41576-021-00438-5. ISSN 1471-0056. PMID 34983971. S2CID 245703043.
  5. ^ Iqbal, Khursheed; Jin, Seung-Gi; Pfeifer, Gerd P.; Szabó, Piroska E. (March 2011). "Reprogramming of the paternal genome upon fertilization involves genome-wide oxidation of 5-methylcytosine". Proceedings of the National Academy of Sciences. 108 (9): 3642–3647. Bibcode:2011PNAS..108.3642I. doi:10.1073/pnas.1014033108. ISSN 0027-8424. PMC 3048122. PMID 21321204.
  6. ^ Husby, Arild (2022-02-09). "Wild epigenetics: insights from epigenetic studies on natural populations". Proceedings of the Royal Society B: Biological Sciences. 289 (1968): 20211633. doi:10.1098/rspb.2021.1633. ISSN 0962-8452. PMC 8826306. PMID 35135348.
  7. ^ a b Ho, Shuk-Mei; Johnson, Abby; Tarapore, Pheruza; Janakiram, Vinothini; Zhang, Xiang; Leung, Yuet-Kin (December 2012). "Environmental Epigenetics and Its Implication on Disease Risk and Health Outcomes". ILAR Journal. 53 (3–4): 289–305. doi:10.1093/ilar.53.3-4.289. ISSN 1084-2020. PMC 4021822. PMID 23744968.
  8. ^ a b Emmanuel, DROUET (2016-09-30). "Epigenetics: How the environment influences our genes". Encyclopedia of the Environment. Retrieved 2023-02-22.
  9. ^ a b Jablonka, Eva; Raz, Gal (June 2009). "Transgenerational Epigenetic Inheritance: Prevalence, Mechanisms, and Implications for the Study of Heredity and Evolution". The Quarterly Review of Biology. 84 (2): 131–176. doi:10.1086/598822. ISSN 0033-5770. PMID 19606595. S2CID 7233550.
  10. ^ a b Li, Dong; Yang, Yan; Li, Youping; Zhu, Xiaohua; Li, Zeqin (2021-07-01). "Epigenetic regulation of gene expression in response to environmental exposures: From bench to model". Science of the Total Environment. 776: 145998. Bibcode:2021ScTEn.776n5998L. doi:10.1016/j.scitotenv.2021.145998. ISSN 0048-9697. S2CID 233548366.
  11. ^ "DNA Packaging: Nucleosomes and Chromatin | Learn Science at Scitable". www.nature.com. Retrieved 2023-02-26.
  12. ^ Wan, Qin-Li; Meng, Xiao; Wang, Chongyang; Dai, Wenyu; Luo, Zhenhuan; Yin, Zhinan; Ju, Zhenyu; Fu, Xiaodie; Yang, Jing; Ye, Qunshan; Zhang, Zhan-Hui; Zhou, Qinghua (2022-02-09). "Histone H3K4me3 modification is a transgenerational epigenetic signal for lipid metabolism in Caenorhabditis elegans". Nature Communications. 13 (1): 768. Bibcode:2022NatCo..13..768W. doi:10.1038/s41467-022-28469-4. ISSN 2041-1723. PMC 8828817. PMID 35140229.
  13. ^ a b Seong, Ki-Hyeon; Li, Dong; Shimizu, Hideyuki; Nakamura, Ryoichi; Ishii, Shunsuke (2011-06-24). "Inheritance of Stress-Induced, ATF-2-Dependent Epigenetic Change". Cell. 145 (7): 1049–1061. doi:10.1016/j.cell.2011.05.029. ISSN 0092-8674. PMID 21703449. S2CID 2918891.
  14. ^ Sen, Rwik; Barnes, Christopher (June 2021). "Do Transgenerational Epigenetic Inheritance and Immune System Development Share Common Epigenetic Processes?". Journal of Developmental Biology. 9 (2): 20. doi:10.3390/jdb9020020. ISSN 2221-3759. PMC 8162332. PMID 34065783.
  15. ^ a b c d Gapp, Katharina; Jawaid, Ali; Sarkies, Peter; Bohacek, Johannes; Pelczar, Pawel; Prados, Julien; Farinelli, Laurent; Miska, Eric; Mansuy, Isabelle M. (May 2014). "Implication of sperm RNAs in transgenerational inheritance of the effects of early trauma in mice". Nature Neuroscience. 17 (5): 667–669. doi:10.1038/nn.3695. ISSN 1546-1726. PMC 4333222. PMID 24728267.
  16. ^ Rodgers, Ali B.; Morgan, Christopher P.; Leu, N. Adrian; Bale, Tracy L. (2015-11-03). "Transgenerational epigenetic programming via sperm microRNA recapitulates effects of paternal stress". Proceedings of the National Academy of Sciences. 112 (44): 13699–13704. Bibcode:2015PNAS..11213699R. doi:10.1073/pnas.1508347112. ISSN 0027-8424. PMC 4640733. PMID 26483456.
  17. ^ a b Rechavi, Oded; Houri-Ze’evi, Leah; Anava, Sarit; Goh, Wee Siong Sho; Kerk, Sze Yen; Hannon, Gregory J.; Hobert, Oliver (2014-07-17). "Starvation-Induced Transgenerational Inheritance of Small RNAs in C. elegans". Cell. 158 (2): 277–287. doi:10.1016/j.cell.2014.06.020. ISSN 0092-8674. PMC 4377509. PMID 25018105.
  18. ^ a b c d e f g h i Horsthemke B (July 2018). "A critical view on transgenerational epigenetic inheritance in humans". Nature Communications. 9 (1): 2973. Bibcode:2018NatCo...9.2973H. doi:10.1038/s41467-018-05445-5. PMC 6065375. PMID 30061690.
  19. ^ Duclos KK, Hendrikse JL, Jamniczky HA (September 2019). "Investigating the evolution and development of biological complexity under the framework of epigenetics". Evolution & Development. 21 (5): 247–264. doi:10.1111/ede.12301. PMC 6852014. PMID 31268245.
  20. ^ a b Bond DM, Finnegan EJ (May 2007). "Passing the message on: inheritance of epigenetic traits". Trends in Plant Science. 12 (5): 211–216. doi:10.1016/j.tplants.2007.03.010. PMID 17434332.
  21. ^ Morison IM, Reeve AE (1998). "A catalogue of imprinted genes and parent-of-origin effects in humans and animals". Human Molecular Genetics. 7 (10): 1599–1609. doi:10.1093/hmg/7.10.1599. PMID 9735381.
  22. ^ Scott RJ, Spielman M, Bailey J, Dickinson HG (September 1998). "Parent-of-origin effects on seed development in Arabidopsis thaliana". Development. 125 (17): 3329–3341. doi:10.1242/dev.125.17.3329. PMID 9693137.
  23. ^ a b Moore DS (2015). The Developing Genome. Oxford University Press. ISBN 978-0-19-992234-5.[pages needed]
  24. ^ Adenot PG, Mercier Y, Renard JP, Thompson EM (November 1997). "Differential H4 acetylation of paternal and maternal chromatin precedes DNA replication and differential transcriptional activity in pronuclei of 1-cell mouse embryos". Development. 124 (22): 4615–4625. doi:10.1242/dev.124.22.4615. PMID 9409678.
  25. ^ Santos F, Hendrich B, Reik W, Dean W (January 2002). "Dynamic reprogramming of DNA methylation in the early mouse embryo". Developmental Biology. 241 (1): 172–182. doi:10.1006/dbio.2001.0501. PMID 11784103.
  26. ^ Oswald J, Engemann S, Lane N, Mayer W, Olek A, Fundele R, et al. (April 2000). "Active demethylation of the paternal genome in the mouse zygote". Current Biology. 10 (8): 475–478. doi:10.1016/S0960-9822(00)00448-6. PMID 10801417.
  27. ^ Fulka H, Mrazek M, Tepla O, Fulka J (December 2004). "DNA methylation pattern in human zygotes and developing embryos". Reproduction. 128 (6): 703–708. doi:10.1530/rep.1.00217. PMID 15579587. S2CID 28719804.
  28. ^ Hackett JA, Sengupta R, Zylicz JJ, Murakami K, Lee C, Down TA, Surani MA (January 2013). "Germline DNA demethylation dynamics and imprint erasure through 5-hydroxymethylcytosine". Science. 339 (6118): 448–452. Bibcode:2013Sci...339..448H. doi:10.1126/science.1229277. PMC 3847602. PMID 23223451.
  29. ^ Surani MA, Hajkova P (2010). "Epigenetic reprogramming of mouse germ cells toward totipotency". Cold Spring Harbor Symposia on Quantitative Biology. 75: 211–218. doi:10.1101/sqb.2010.75.010. PMID 21139069.
  30. ^ Zhang Z, Shibahara K, Stillman B (November 2000). "PCNA connects DNA replication to epigenetic inheritance in yeast". Nature. 408 (6809): 221–225. Bibcode:2000Natur.408..221Z. doi:10.1038/35041601. PMID 11089978. S2CID 205010657.
  31. ^ Henderson DS, Banga SS, Grigliatti TA, Boyd JB (March 1994). "Mutagen sensitivity and suppression of position-effect variegation result from mutations in mus209, the Drosophila gene encoding PCNA". The EMBO Journal. 13 (6): 1450–1459. doi:10.1002/j.1460-2075.1994.tb06399.x. PMC 394963. PMID 7907981.
  32. ^ Probst AV, Dunleavy E, Almouzni G (March 2009). "Epigenetic inheritance during the cell cycle". Nature Reviews. Molecular Cell Biology. 10 (3): 192–206. doi:10.1038/nrm2640. PMID 19234478. S2CID 205494340.
  33. ^ a b Morgan HD, Santos F, Green K, Dean W, Reik W (April 2005). "Epigenetic reprogramming in mammals". Human Molecular Genetics. 14 (Review Issue 1): R47–R58. doi:10.1093/hmg/ddi114. PMID 15809273.
  34. ^ Santos F, Peters AH, Otte AP, Reik W, Dean W (April 2005). "Dynamic chromatin modifications characterise the first cell cycle in mouse embryos". Developmental Biology. 280 (1): 225–236. doi:10.1016/j.ydbio.2005.01.025. PMID 15766761.
  35. ^ Taguchi YH (2015). "Identification of aberrant gene expression associated with aberrant promoter methylation in primordial germ cells between E13 and E16 rat F3 generation vinclozolin lineage". BMC Bioinformatics. 16 (Suppl 18): S16. doi:10.1186/1471-2105-16-S18-S16. PMC 4682393. PMID 26677731.
  36. ^ Richards EJ (May 2006). "Inherited epigenetic variation--revisiting soft inheritance". Nature Reviews. Genetics. 7 (5): 395–401. doi:10.1038/nrg1834. PMID 16534512. S2CID 21961242.
  37. ^ Day, Jeremy J. (2014-09-30). "New approaches to manipulating the epigenome". Dialogues in Clinical Neuroscience. 16 (3): 345–357. doi:10.31887/DCNS.2014.16.3/jday. ISSN 1958-5969. PMC 4214177. PMID 25364285.
  38. ^ a b c d Coe EH (June 1959). "A regular and continuing conversion-type phenomenon at the B locus in maize". Proceedings of the National Academy of Sciences of the United States of America. 45 (6): 828–832. Bibcode:1959PNAS...45..828C. doi:10.1073/pnas.45.6.828. PMC 222644. PMID 16590451.
  39. ^ Chandler VL (February 2007). "Paramutation: from maize to mice". Cell. 128 (4): 641–645. doi:10.1016/j.cell.2007.02.007. PMID 17320501.
  40. ^ a b Stam M, Belele C, Ramakrishna W, Dorweiler JE, Bennetzen JL, Chandler VL (October 2002). "The regulatory regions required for B' paramutation and expression are located far upstream of the maize b1 transcribed sequences". Genetics. 162 (2): 917–930. doi:10.1093/genetics/162.2.917. PMC 1462281. PMID 12399399.
  41. ^ a b Belele CL, Sidorenko L, Stam M, Bader R, Arteaga-Vazquez MA, Chandler VL (2013-10-17). "Specific tandem repeats are sufficient for paramutation-induced trans-generational silencing". PLOS Genetics. 9 (10): e1003773. doi:10.1371/journal.pgen.1003773. PMC 3798267. PMID 24146624.
  42. ^ a b Arteaga-Vazquez M, Sidorenko L, Rabanal FA, Shrivistava R, Nobuta K, Green PJ, et al. (July 2010). "RNA-mediated trans-communication can establish paramutation at the b1 locus in maize". Proceedings of the National Academy of Sciences of the United States of America. 107 (29): 12986–12991. Bibcode:2010PNAS..10712986A. doi:10.1073/pnas.1007972107. PMC 2919911. PMID 20616013.
  43. ^ a b Louwers M, Bader R, Haring M, van Driel R, de Laat W, Stam M (March 2009). "Tissue- and expression level-specific chromatin looping at maize b1 epialleles". The Plant Cell. 21 (3): 832–842. doi:10.1105/tpc.108.064329. PMC 2671708. PMID 19336692.
  44. ^ a b Haring M, Bader R, Louwers M, Schwabe A, van Driel R, Stam M (August 2010). "The role of DNA methylation, nucleosome occupancy and histone modifications in paramutation". The Plant Journal. 63 (3): 366–378. doi:10.1111/j.1365-313X.2010.04245.x. PMID 20444233.
  45. ^ a b Dorweiler JE, Carey CC, Kubo KM, Hollick JB, Kermicle JL, Chandler VL (November 2000). "mediator of paramutation1 is required for establishment and maintenance of paramutation at multiple maize loci". The Plant Cell. 12 (11): 2101–2118. doi:10.1105/tpc.12.11.2101. PMC 150161. PMID 11090212.
  46. ^ a b c Chandler V, Alleman M (April 2008). "Paramutation: epigenetic instructions passed across generations". Genetics. 178 (4): 1839–1844. doi:10.1093/genetics/178.4.1839. PMC 2323780. PMID 18430919.
  47. ^ Nobuta K, Lu C, Shrivastava R, Pillay M, De Paoli E, Accerbi M, et al. (September 2008). "Distinct size distribution of endogeneous siRNAs in maize: Evidence from deep sequencing in the mop1-1 mutant". Proceedings of the National Academy of Sciences of the United States of America. 105 (39): 14958–14963. Bibcode:2008PNAS..10514958N. doi:10.1073/pnas.0808066105. PMC 2567475. PMID 18815367.
  48. ^ Alleman M, Sidorenko L, McGinnis K, Seshadri V, Dorweiler JE, White J, et al. (July 2006). "An RNA-dependent RNA polymerase is required for paramutation in maize". Nature. 442 (7100): 295–298. Bibcode:2006Natur.442..295A. doi:10.1038/nature04884. PMID 16855589. S2CID 4419412.
  49. ^ Arteaga-Vazquez MA, Chandler VL (April 2010). "Paramutation in maize: RNA mediated trans-generational gene silencing". Current Opinion in Genetics & Development. 20 (2): 156–163. doi:10.1016/j.gde.2010.01.008. PMC 2859986. PMID 20153628.
  50. ^ Huang J, Lynn JS, Schulte L, Vendramin S, McGinnis K (2017-01-01). "Epigenetic Control of Gene Expression in Maize". International Review of Cell and Molecular Biology. 328: 25–48. doi:10.1016/bs.ircmb.2016.08.002. ISBN 9780128122204. PMID 28069135.
  51. ^ Chandler VL (October 2010). "Paramutation's properties and puzzles". Science. 330 (6004): 628–629. Bibcode:2010Sci...330..628C. doi:10.1126/science.1191044. PMID 21030647. S2CID 13248794.
  52. ^ Sobral, Mar; Sampedro, Luis; Neylan, Isabelle; Siemens, David; Dirzo, Rodolfo (2021-08-17). "Phenotypic plasticity in plant defense across life stages: Inducibility, transgenerational induction, and transgenerational priming in wild radish". Proceedings of the National Academy of Sciences. 118 (33): e2005865118. Bibcode:2021PNAS..11805865S. doi:10.1073/pnas.2005865118. ISSN 0027-8424. PMC 8379918. PMID 34389664.
  53. ^ Agrawal, Anurag A.; Laforsch, Christian; Tollrian, Ralph (1999-09-02). "Transgenerational induction of defences in animals and plants". Nature. 401 (6748): 60–63. Bibcode:1999Natur.401...60A. doi:10.1038/43425. ISSN 0028-0836. S2CID 4326322.
  54. ^ Ryu, Taewoo; Veilleux, Heather D.; Donelson, Jennifer M.; Munday, Philip L.; Ravasi, Timothy (2018-04-30). "The epigenetic landscape of transgenerational acclimation to ocean warming". Nature Climate Change. 8 (6): 504–509. Bibcode:2018NatCC...8..504R. doi:10.1038/s41558-018-0159-0. ISSN 1758-678X. S2CID 90082460.
  55. ^ Hu, J.; Barrett, R. D. H. (2017-07-20). "Epigenetics in natural animal populations". Journal of Evolutionary Biology. 30 (9): 1612–1632. doi:10.1111/jeb.13130. ISSN 1010-061X. PMID 28597938. S2CID 20558647.
  56. ^ a b Stein, A. D (2004-07-28). "Intrauterine famine exposure and body proportions at birth: the Dutch Hunger Winter". International Journal of Epidemiology. 33 (4): 831–836. doi:10.1093/ije/dyh083. ISSN 1464-3685. PMID 15166208.
  57. ^ a b Wei Y, Schatten H, Sun QY (2014). "Environmental epigenetic inheritance through gametes and implications for human reproduction". Human Reproduction Update. 21 (2): 194–208. doi:10.1093/humupd/dmu061. PMID 25416302.
  58. ^ a b c d da Cruz, R. S., Chen, E., Smith, M., Bates, J., & de Assis, S. (2020). Diet and Transgenerational Epigenetic Inheritance of Breast Cancer: The Role of the Paternal Germline. Frontiers in nutrition, 7, 93. https://doi.org/10.3389/fnut.2020.0009
  59. ^ a b c Fontelles CC, Carney E, Clarke J, Nguyen NM, Yin C, Jin L, Cruz MI, Ong TP, Hilakivi-Clarke L, de Assis S (June 2016). "Paternal overweight is associated with increased breast cancer risk in daughters in a mouse model". Scientific Reports. 6: 28602. Bibcode:2016NatSR...628602F. doi:10.1038/srep28602. PMC 4919621. PMID 27339599.
  60. ^ a b c d e f g h i Napoli C, Benincasa G, Loscalzo J (April 2019). "Epigenetic Inheritance Underlying Pulmonary Arterial Hypertension". Arteriosclerosis, Thrombosis, and Vascular Biology. 39 (4): 653–664. doi:10.1161/ATVBAHA.118.312262. PMC 6436974. PMID 30727752.
  61. ^ Weaver IC, Cervoni N, Champagne FA, D'Alessio AC, Sharma S, Seckl JR, et al. (August 2004). "Epigenetic programming by maternal behavior". Nature Neuroscience. 7 (8): 847–854. doi:10.1038/nn1276. PMID 15220929. S2CID 1649281.
  62. ^ McGowan PO, Sasaki A, D'Alessio AC, Dymov S, Labonté B, Szyf M, et al. (March 2009). "Epigenetic regulation of the glucocorticoid receptor in human brain associates with childhood abuse". Nature Neuroscience. 12 (3): 342–348. doi:10.1038/nn.2270. PMC 2944040. PMID 19234457.
  63. ^ Meaney MJ, Szyf M (2005). "Environmental programming of stress responses through DNA methylation: life at the interface between a dynamic environment and a fixed genome". Dialogues in Clinical Neuroscience. 7 (2): 103–123. doi:10.31887/DCNS.2005.7.2/mmeaney. PMC 3181727. PMID 16262207.
  64. ^ a b Radtke KM, Ruf M, Gunter HM, Dohrmann K, Schauer M, Meyer A, Elbert T (July 2011). "Transgenerational impact of intimate partner violence on methylation in the promoter of the glucocorticoid receptor". Translational Psychiatry. 1 (July 19): e21. doi:10.1038/tp.2011.21. PMC 3309516. PMID 22832523.
  65. ^ a b c d Kioumourtzoglou MA, Coull BA, O'Reilly ÉJ, Ascherio A, Weisskopf MG (July 2018). "Association of Exposure to Diethylstilbestrol During Pregnancy With Multigenerational Neurodevelopmental Deficits". JAMA Pediatrics. 172 (7): 670–677. doi:10.1001/jamapediatrics.2018.0727. PMC 6137513. PMID 29799929.
  66. ^ Jablonka E, Lamb MJ (2005). Epigenetic inheritance and evolution: the Lamarckian dimension (Reprinted ed.). Oxford: Oxford University Press. ISBN 978-0-19-854063-2.
  67. ^ Cubas P, Vincent C, Coen E (September 1999). "An epigenetic mutation responsible for natural variation in floral symmetry". Nature. 401 (6749): 157–161. Bibcode:1999Natur.401..157C. doi:10.1038/43657. PMID 10490023. S2CID 205033495.
  68. ^ Dafni A, Kevan PG (1997). "Flower size and shape: implications in pollination". Israeli Journal of Plant Science. 45 (2–3): 201–211. doi:10.1080/07929978.1997.10676684.
  69. ^ Nilsson EE, Sadler-Riggleman I, Skinner MK (April 2018). "Environmentally induced epigenetic transgenerational inheritance of disease". Environmental Epigenetics. 4 (2): dvy016. doi:10.1093/eep/dvy016. PMC 6051467. PMID 30038800.
  70. ^ Frazier ML, Xi L, Zong J, Viscofsky N, Rashid A, Wu EF, et al. (August 2003). "Association of the CpG island methylator phenotype with family history of cancer in patients with colorectal cancer". Cancer Research. 63 (16): 4805–4808. PMID 12941799.
  71. ^ Chan TL, Yuen ST, Kong CK, Chan YW, Chan AS, Ng WF, et al. (October 2006). "Heritable germline epimutation of MSH2 in a family with hereditary nonpolyposis colorectal cancer". Nature Genetics. 38 (10): 1178–1183. doi:10.1038/ng1866. PMC 7097088. PMID 16951683.
  72. ^ Bossdorf O, Arcuri D, Richards CL, Pigliucci M (2010). "Experimental alteration of DNA methylation affects the phenotypic plasticity of ecologically relevant traits in Arabidopsis thaliana" (PDF). Evolutionary Ecology. 24 (3): 541–553. doi:10.1007/s10682-010-9372-7. S2CID 15763479.
  73. ^ Whittle CA, Otto SP, Johnston MO, Krochko JE (2009). "Adaptive epigenetic memory of ancestral temperature regime in Arabidopsis thaliana". Botany. 87 (6): 650–657. doi:10.1139/b09-030.
  74. ^ Curley, JP, FA Champagne, and P Bateson (2007) Communal nesting induces alternative emotional, social and maternal behavior in offspring. Society for Behavioral Neuroendocrinology 11th Annual Meeting Pacific Grove, CA, USA. Cited in Branchi I (April 2009). "The mouse communal nest: investigating the epigenetic influences of the early social environment on brain and behavior development". Neuroscience and Biobehavioral Reviews. 33 (4): 551–559. doi:10.1016/j.neubiorev.2008.03.011. PMID 18471879. S2CID 1592896.
  75. ^ Branchi I, D'Andrea I, Fiore M, Di Fausto V, Aloe L, Alleva E (October 2006). "Early social enrichment shapes social behavior and nerve growth factor and brain-derived neurotrophic factor levels in the adult mouse brain". Biological Psychiatry. 60 (7): 690–696. doi:10.1016/j.biopsych.2006.01.005. PMID 16533499. S2CID 16627324.
  76. ^ Sen, Rwik; Barnes, Christopher (2021-05-12). "Do Transgenerational Epigenetic Inheritance and Immune System Development Share Common Epigenetic Processes?". Journal of Developmental Biology. 9 (2): 20. doi:10.3390/jdb9020020. ISSN 2221-3759. PMC 8162332. PMID 34065783.
  77. ^ a b c Katzmarski, Natalie; Domínguez-Andrés, Jorge; Cirovic, Branko; Renieris, Georgios; Ciarlo, Eleonora; Le Roy, Didier; Lepikhov, Konstantin; Kattler, Kathrin; Gasparoni, Gilles; Händler, Kristian; Theis, Heidi; Beyer, Marc; van der Meer, Jos W. M.; Joosten, Leo A. B.; Walter, Jörn (November 2021). "Transmission of trained immunity and heterologous resistance to infections across generations". Nature Immunology. 22 (11): 1382–1390. doi:10.1038/s41590-021-01052-7. hdl:2066/241159. ISSN 1529-2916. PMID 34663978. S2CID 239026066.
  78. ^ a b c d Eggert, Hendrik; Kurtz, Joachim; Diddens-de Buhr, Maike F. (2014-12-22). "Different effects of paternal trans-generational immune priming on survival and immunity in step and genetic offspring". Proceedings of the Royal Society B: Biological Sciences. 281 (1797): 20142089. doi:10.1098/rspb.2014.2089. ISSN 0962-8452. PMC 4240996. PMID 25355479.
  79. ^ a b Singh, Krishna P.; Jahagirdar, Shamarao; Sarma, Birinchi Kumar, eds. (2021). Emerging Trends in Plant Pathology. doi:10.1007/978-981-15-6275-4. ISBN 978-981-15-6274-7. S2CID 228078200.
  80. ^ a b c Luna, Estrella; Ton, Jurriaan (June 2012). "The epigenetic machinery controlling transgenerational systemic acquired resistance". Plant Signaling & Behavior. 7 (6): 615–618. doi:10.4161/psb.20155. ISSN 1559-2324. PMC 3442853. PMID 22580690. S2CID 38372184.
  81. ^ a b Casier, Karine; Delmarre, Valérie; Gueguen, Nathalie; Hermant, Catherine; Viodé, Elise; Vaury, Chantal; Ronsseray, Stéphane; Brasset, Emilie; Teysset, Laure; Boivin, Antoine (2019-03-15). Nilsen, Timothy W; Manley, James L (eds.). "Environmentally-induced epigenetic conversion of a piRNA cluster". eLife. 8: e39842. doi:10.7554/eLife.39842. ISSN 2050-084X. PMC 6420265. PMID 30875295.
  82. ^ a b Major, Kaley M.; DeCourten, Bethany M.; Li, Jie; Britton, Monica; Settles, Matthew L.; Mehinto, Alvine C.; Connon, Richard E.; Brander, Susanne M. (2020). "Early Life Exposure to Environmentally Relevant Levels of Endocrine Disruptors Drive Multigenerational and Transgenerational Epigenetic Changes in a Fish Model". Frontiers in Marine Science. 7. doi:10.3389/fmars.2020.00471. ISSN 2296-7745.
  83. ^ Liu, Shenglin; Tengstedt, Aja Noersgaard Buur; Jacobsen, Magnus W.; Pujolar, Jose Martin; Jónsson, Bjarni; Lobón‐Cervià, Javier; Bernatchez, Louis; Hansen, Michael M. (August 2022). "Genome‐wide methylation in the panmictic European eel ( Anguilla anguilla )". Molecular Ecology. 31 (16): 4286–4306. doi:10.1111/mec.16586. ISSN 0962-1083. PMID 35767387. S2CID 250115270.
  84. ^ a b Jablonka E, Raz G (June 2009). "Transgenerational epigenetic inheritance: prevalence, mechanisms, and implications for the study of heredity and evolution". The Quarterly Review of Biology. 84 (2): 131–176. CiteSeerX 10.1.1.617.6333. doi:10.1086/598822. PMID 19606595. S2CID 7233550.
  85. ^ Whitham TG, Slobodchikoff CN (July 1981). "Evolution by individuals, plant-herbivore interactions, and mosaics of genetic variability: The adaptive significance of somatic mutations in plants". Oecologia. 49 (3): 287–292. Bibcode:1981Oecol..49..287W. doi:10.1007/BF00347587. PMID 28309985. S2CID 20411802.
  86. ^ Turian G (1979). "Sporogenesis in fungi". Annual Review of Phytopathology. 12: 129–137. doi:10.1146/annurev.py.12.090174.001021.
  87. ^ Vorzimmer P (1963). "Charles Darwin and blending inheritance". Isis. 54 (3): 371–390. doi:10.1086/349734. S2CID 143975567.
  88. ^ Jenkin F (1867). "Review of The Origin of Species". North British Review.
  89. ^ Mendel G (1866). "Versuche über Plflanzenhybriden. Verhandlungen des naturforschenden Vereines in Brünn" [Experiments in Plant Hybridization] (PDF). Read at the February 8th, and March 8th, 1865, meetings of the Brünn Natural History Society (in German).
  90. ^ Lamarck JB (1809). Philosophie zoologique: ou Exposition des considérations relative à l'histoire naturelle des animaux. Dentu et L'Auteur, Paris.
  91. ^ Bowler PJ (1989). Evolution, the history of an idea. Berkeley: University of California Press. ISBN 978-0-520-06386-0.
  92. ^ Weismann A (1891). Poulton EB, Schönland S, Shipley E (eds.). Essays upon heredity and kindred biological problems. Oxford: Clarendon Press. doi:10.5962/bhl.title.28066.
  93. ^ Goldberg AD, Allis CD, Bernstein E (February 2007). "Epigenetics: a landscape takes shape". Cell. 128 (4): 635–638. doi:10.1016/j.cell.2007.02.006. PMID 17320500.
  94. ^ Waddington CH (2016) [1939]. "Development as an Epigenetic Process". Introduction to Modern Genetics. London: Allen and Unwin. ISBN 9781317352037. One of the classical controversies in embryology was that between the preformationists and the epigenisists[sic]. [...] the interaction of these constituents gives rise to new types of tissue and organ which were not present originally, and in so far development must be considered as 'epigenetic.'
  95. ^ Holliday R (2006). "Epigenetics: a historical overview". Epigenetics. 1 (2): 76–80. doi:10.4161/epi.1.2.2762. PMID 17998809.
  96. ^ Nanney DL (July 1958). "Epigenetic Control Systems". Proceedings of the National Academy of Sciences of the United States of America. 44 (7): 712–717. Bibcode:1958PNAS...44..712N. doi:10.1073/pnas.44.7.712. PMC 528649. PMID 16590265.
  97. ^ Crick FH (1958). "On protein synthesis" (PDF). Symposia of the Society for Experimental Biology. 12: 138–163. PMID 13580867.
  98. ^ Pigliucci M (December 2007). "Do we need an extended evolutionary synthesis?". Evolution; International Journal of Organic Evolution. 61 (12): 2743–2749. doi:10.1111/j.1558-5646.2007.00246.x. PMID 17924956.
  99. ^ a b c d van Otterdijk SD, Michels KB (July 2016). "Transgenerational epigenetic inheritance in mammals: how good is the evidence?". FASEB Journal. 30 (7): 2457–65. doi:10.1096/fj.201500083. PMID 27037350. S2CID 11969347.
  100. ^ Steele EJ (1979). Somatic selection and adaptive evolution: on the inheritance of acquired characters (1st edit ed.). Toronto: Williams-Wallace.
  101. ^ Steele EJ, Lindley RA, Blanden RV (1998). Davies P (ed.). Lamarck's signature: how retrogenes are changing Darwin's natural selection paradigm. Frontiers of Science. Sydney: Allen & Unwin.
  102. ^ Lindley RA (2010). The Soma: how our genes really work and how that changes everything!. Piara Waters, CYO Foundation. ISBN 978-1451525649.
  103. ^ Steele EJ, Lloyd SS (May 2015). "Soma-to-germline feedback is implied by the extreme polymorphism at IGHV relative to MHC: The manifest polymorphism of the MHC appears greatly exceeded at Immunoglobulin loci, suggesting antigen-selected somatic V mutants penetrate Weismann's Barrier". BioEssays. 37 (5): 557–569. doi:10.1002/bies.201400213. PMID 25810320. S2CID 1270807.
  104. ^ Steele EJ (2016). Levin M, Adams DS (eds.). Origin of congenital defects: stable inheritance through the male line via maternal antibodies specific for eye lens antigens inducing autoimmune eye defects in developing rabbits in utero. Ahead of the Curve -Hidden breakthroughs in the biosciences. Bristol, UK: IOP Publishing. pp. Chapter 3.
  105. ^ Hoyle F, Wickramasinghe C (1982). Why neo-Darwinism does not work. Cardiff: University College Cardiff Press. ISBN 0-906449-50-2.
  106. ^ Hoyle F, Wickramasinghe NC (1979). Diseases from space. London: J.M. Dent.
  107. ^ Hoyle F, Wickramasinghe NC (1981). Evolution from space. London: J.M. Dent.
  108. ^ Liu Y (September 2007). "Like father like son. A fresh review of the inheritance of acquired characteristics". EMBO Reports. 8 (9): 798–803. doi:10.1038/sj.embor.7401060. PMC 1973965. PMID 17767188.
  109. ^ Liu Y, Li X (May 2016). "Darwin's Pangenesis as a molecular theory of inherited diseases". Gene. 582 (1): 19–22. doi:10.1016/j.gene.2016.01.051. PMID 26836487.
  110. ^ Noble D (February 2012). "A theory of biological relativity: no privileged level of causation". Interface Focus. 2 (1): 55–64. doi:10.1098/rsfs.2011.0067. PMC 3262309. PMID 23386960.
  111. ^ Noble D (August 2013). "Physiology is rocking the foundations of evolutionary biology". Experimental Physiology. 98 (8): 1235–1243. doi:10.1113/expphysiol.2012.071134. PMID 23585325. S2CID 19689192.
  112. ^ Mattick JS (October 2012). "Rocking the foundations of molecular genetics". Proceedings of the National Academy of Sciences of the United States of America. 109 (41): 16400–16401. Bibcode:2012PNAS..10916400M. doi:10.1073/pnas.1214129109. PMC 3478605. PMID 23019584.

transgenerational, epigenetic, inheritance, transmission, epigenetic, markers, modifications, from, generation, multiple, subsequent, generations, without, altering, primary, structure, thus, regulation, genes, epigenetic, mechanisms, heritable, amount, transc. Transgenerational epigenetic inheritance is the transmission of epigenetic markers and modifications from one generation to multiple subsequent generations without altering the primary structure of DNA 1 Thus the regulation of genes via epigenetic mechanisms can be heritable the amount of transcripts and proteins produced can be altered by inherited epigenetic changes In order for epigenetic marks to be heritable however they must occur in the gametes in animals but since plants lack a definitive germline and can propagate epigenetic marks in any tissue can be heritable Intergenerational vs Transgenerational InheritanceIt is important to note that the inheritance of epigenetic marks in the immediate generation is referred to as intergenerational inheritance 2 In male mice the epigenetic signal is maintained through the F1 generation 3 In female mice the epigenetic signal is maintained through the F2 generation as a result of the exposure of the germline in the womb 3 Many epigenetic signals are lost beyond the F2 F3 generation and are no longer inherited because the subsequent generations were not exposed to the same environment as the parental generations 2 The signals that are maintained beyond the F2 F3 generation are referred to as transgenerational epigenetic inheritance TEI because initial environmental stimuli resulted in inheritance of epigenetic modifications 4 There are several mechanisms of TEI that have shown to affect germline reprogramming such as transgenerational increases in susceptibility to diseases mutations and stress inheritance During germline reprogramming and early embryogenesis in mice methylation marks are removed to allow for development to commence but the methylation mark is converted into hydroxymethyl cytosine so that it is recognized and methylated once that area of the genome is no longer being used 5 which serves as a memory for that TEI mark Therefore under lab conditions inherited methyl marks are removed and restored to ensure TEI still occurs However observing TEI in wild populations is still in its infancy as laboratory studies allow for more tractable systems 6 Environmental factors can induce the epigenetic marks epigenetic tags for some epigenetically influenced traits 1 These can include but are not limited to changes in temperature resources availability exposure to pollutants chemicals and endocrine disruptors 7 The dosage and exposure levels can affect the extent of the environmental factors influence over the epigenome and its effect on later generations The epigenetic marks can result in a wide range of effects including minor phenotypic changes to complex diseases and disorders 7 The complex cell signaling pathways of multicellular organisms such as plants and humans can make understanding the mechanisms of this inherited process very difficult 8 Contents 1 Epigenetic categories 2 Inheritance of epigenetic marks 2 1 Removal vs retention 2 2 Reprogramming 2 3 Retention 2 4 Decay 3 Examples of TEI 3 1 In Plants 3 2 In Animals 3 3 In Humans 4 Effects on fitness 4 1 Deleterious effects 4 2 Putatively adaptive effects 4 3 Inheritance of Immunity 4 3 1 Feedback loops and TEI 5 Macroevolutionary patterns 6 Controversies 6 1 Blending vs particulate inheritance 6 2 Inheritance of innate vs acquired characteristics 6 3 Revision of evolutionary theory 7 See also 8 ReferencesEpigenetic categories editThere are mechanisms by which environmental exposures induce epigenetic changes by affecting regulation and gene expression Four general categories of epigenetic modification are known self sustaining metabolic loops in which an mRNA or protein product of a gene stimulates transcription of the gene e g Wor1 gene in Candida albicans Structural templating structures are replicated using a template or scaffold structure of the parent This can include but is not limited to the orientation and architecture of cytoskeletal structures cilia and flagella Ciliates provide a good example of this type of modification In an experiment Beisson and Sonneborn in 1985 it was demonstrated in Paramecium that if a section of cilia was removed and inverted then the progeny of that Paramecium would also display the modified cilia structure for several generations 9 Another example is seen in prions special proteins that are capable of changing the structure of normal proteins to match their own The prions use themselves as a template and then edit the folding of normal proteins to match their own folding pattern The changes in the protein folding results in an alteration in the normal protein s function This transmission of programming can also alter the chromatin and histone of the DNA and can be passed through the cytosol from parent to offspring during meiosis 9 Histone modifications in which the structure of chromatin and its transcriptional state is regulated DNA is wrapped into a DNA protein complex called chromatin in the nucleus of eukaryotic cells 10 Chromatin consists of DNA and nucleosomes that comes together to form a histone octamer 11 The N and C terminal of the histone proteins are post translationally modified by the removal or addition of acetyl acetylation phosphate phosphorylation methyl methylation ubiquitin ubiquitination and ubiquitin like modifier SUMOylating groups 10 Histone modifications can be transgenerational epigenetic signals For example histone H3K4 trimethylation H34me3 and a network of lipid metabolic genes interact to increase the transcription response to TEI obesogenic effects 12 TEI can also be observed in Drosophila embryos through the exposure of heat stress over generations 3 The induced heat stress resulted in the phosphorylation of ATF 2 dATF 2 which is required for heterochromatin assembly 13 This epigenetic event was maintained over multiple generations but over time dATF 2 returned back to its normal state 13 Non coding and coding RNAs in which various classes of RNA is implicated in TEI through maternal stores of mRNA translation of mRNA miRNA and small RNA strands interfering with transcription piRNAs and siRNAs via RNA interference pathways RNAi 2 There has been an increase in studies reporting noncoding RNA contributions to TEI For example altered miRNA in early trauma mice 14 Early trauma mice with unpredictable maternal separation and maternal stress MSUS were used as a model to identify the effects of altered miRNA in sperm 15 In MSUS mice behavior responses were affected insulin levels and blood glucose levels were decreased 15 Notably these effects were more severe across the F2 and F3 generation The expression of miRNA in MSUS mice was down regulated in the brain serum and sperm of the F1 generation 15 However the miRNA was not altered in the sperm of the F2 generation and the miRNAs were normal in the F3 generation 15 This provides supportive evidence that the initial alterations in miRNAs in sperm are transferred to epigenetic marks to maintain transmission 16 In C elegans starvation is induced in which survival is dependent on the mechanisms of the RNAi pathway repression of microRNAs and regulation of small RNAs 17 Thus memorization of dietary history is inherited across generations 17 Inheritance of epigenetic marks editMain article Epigenetics See also Transgenerational epigenetic inheritance in plants Although there are various forms of inheriting epigenetic markers inheritance of epigenetic markers can be summarized as the dissemination of epigenetic information by means of the germline 18 Furthermore epigenetic variation typically takes one of four general forms though there are other forms that have yet to be elucidated Currently self sustaining feedback loops spatial templating chromatin marking and RNA mediated pathways modify epigenes of individual cells Epigenetic variation within multicellular organisms is either endogenous or exogenous 19 Endogenous is generated by cell cell signaling e g during cell differentiation early in development while exogenous is a cellular response to environmental cues citation needed Removal vs retention edit In sexually reproducing organisms much of the epigenetic modification within cells is reset during meiosis e g marks at the FLC locus controlling plant vernalization 20 though some epigenetic responses have been shown to be conserved e g transposon methylation in plants 20 Differential inheritance of epigenetic marks due to underlying maternal or paternal biases in removal or retention mechanisms may lead to the assignment of epigenetic causation to some parent of origin effects in animals 21 and plants 22 Reprogramming edit In mammals epigenetic marks are erased during two phases of the life cycle Firstly just after fertilization and secondly in the developing primordial germ cells the precursors to future gametes 23 During fertilization the male and female gametes join in different cell cycle states and with different configuration of the genome The epigenetic marks of the male are rapidly diluted First the protamines associated with male DNA are replaced with histones from the female s cytoplasm most of which are acetylated due to either higher abundance of acetylated histones in the female s cytoplasm or through preferential binding of the male DNA to acetylated histones 24 25 Second male DNA is systematically demethylated in many organisms 26 27 possibly through 5 hydroxymethylcytosine However some epigenetic marks particularly maternal DNA methylation can escape this reprogramming leading to parental imprinting In the primordial germ cells PGC there is a more extensive erasure of epigenetic information However some rare sites can also evade erasure of DNA methylation 28 If epigenetic marks evade erasure during both zygotic and PGC reprogramming events this could enable transgenerational epigenetic inheritance citation needed Recognition of the importance of epigenetic programming to the establishment and fixation of cell line identity during early embryogenesis has recently stimulated interest in artificial removal of epigenetic programming 29 Epigenetic manipulations may allow for restoration of totipotency in stem cells or cells more generally thus generalizing regenerative medicine Retention edit Cellular mechanisms may allow for co transmission of some epigenetic marks During replication DNA polymerases working on the leading and lagging strands are coupled by the DNA processivity factor proliferating cell nuclear antigen PCNA which has also been implicated in patterning and strand crosstalk that allows for copy fidelity of epigenetic marks 30 31 Work on histone modification copy fidelity has remained in the model phase but early efforts suggest that modifications of new histones are patterned on those of the old histones and that new and old histones randomly assort between the two daughter DNA strands 32 With respect to transfer to the next generation many marks are removed as described above Emerging studies are finding patterns of epigenetic conservation across generations For instance centromeric satellites resist demethylation 33 The mechanism responsible for this conservation is not known though some evidence suggests that methylation of histones may contribute 33 34 Dysregulation of the promoter methylation timing associated with gene expression dysregulation in the embryo was also identified 35 Decay edit Whereas the mutation rate in a given 100 base gene may be 10 7 per generation epigenes may mutate several times per generation or may be fixed for many generations 36 This raises the question do changes in epigene frequencies constitute evolution Rapidly decaying epigenetic effects on phenotypes i e lasting less than three generations may explain some of the residual variation in phenotypes after genotype and environment are accounted for However distinguishing these short term effects from the effects of the maternal environment on early ontogeny remains a challenge citation needed Examples of TEI editSee also Contribution of epigenetic modifications to evolution The relative importance of genetic and epigenetic inheritance is subject to debate Though hundreds of examples of epigenetic modification of phenotypes have been published few studies have been conducted outside of the laboratory setting Therefore the interactions of genes with the environment cannot be inferred despite the central role of environment in natural selection Multiple epigenetic factors can influence the state of genes and alter the epigenetic state Due to the multivariate nature of environmental factors it is difficult for researchers to pinpoint the exact cause of epigenetic changes outside of a laboratory setting 37 In Plants edit Studies concerning transgenerational epigenetic inheritance in plants have been reported as early as the 1950s 38 One of the earliest and best characterized examples of this is b1 paramutation in maize 38 39 40 41 42 43 44 45 The b1 gene encodes a basic helix loop helix transcription factor that is involved in the anthocyanin production pathway When the b1 gene is expressed the plant accumulates anthocyanin within its tissues leading to a purple coloration of those tissues The B I allele for B Intense has high expression of b1 resulting in the dark pigmentation of the sheath and husk tissues while the B pronounced B prime allele has low expression of b1 resulting in low pigmentation in those tissues 46 When homozygous B I parents are crossed to homozygous B the resultant F1 offspring all display low pigmentation which is due to gene silencing of b1 38 46 Unexpectedly when F1 plants are self crossed the resultant F2 generation all display low pigmentation and have low levels of b1 expression Furthermore when any F2 plant including those that are genetically homozygous for B I are crossed to homozygous B I the offspring will all display low pigmentation and expression of b1 38 46 The lack of darkly pigmented individuals in the F2 progeny is an example of non Mendelian inheritance and further research has suggested that the B I allele is converted to B via epigenetic mechanisms 40 41 The B and B I alleles are considered to be epialleles because they are identical at the DNA sequence level but differ in the level of DNA methylation siRNA production and chromosomal interactions within the nucleus 44 47 43 42 Additionally plants defective in components of the RNA directed DNA methylation pathway show an increased expression of b1 in B individuals similar to that of B I however once these components are restored the plant reverts to the low expression state 45 48 49 50 Although spontaneous conversion from B I to B has been observed a reversion from B to B I green to purple has never been observed over 50 years and thousands of plants in both greenhouse and field experiments 51 Examples of environmentally induced transgenerational epigenetic inheritance in plants has also been reported In one case rice plants that were exposed to drought simulation treatments displayed increased tolerance to drought after 11 generations of exposure and propagation by single seed descent as compared to non drought treated plants Differences in drought tolerance was linked to directional changes in DNA methylation levels throughout the genome suggesting that stress induced heritable changes in DNA methylation patterns may be important in adaptation to recurring stresses In another study plants that were exposed to moderate caterpillar herbivory over multiple generations displayed increased resistance to herbivory in subsequent generations as measured by caterpillar dry mass compared to plants lacking herbivore pressure This increase in herbivore resistance persisted after a generation of growth without any herbivore exposure suggesting that the response was transmitted across generations The report concluded that components of the RNA directed DNA methylation pathway are involved in the increased resistance across generations Transgenerational epigenetic inheritance has also been observed in polyploid plants Genetically identical reciprocal F1 hybrid triploids have been shown to display transgenerational epigenetic effects on viable F2 seed development It has been demonstrated in wild radish plants Raphanus raphanistrum that TEI can be induced when the plants are exposed to predators such as Pieris rapae the cabbage white caterpillar The radish plants will increase production of bristly leaf hairs and toxic mustard oil in response to caterpillar predation The increased levels will also be seen in the next generation Decreased levels of predation also results in decreased leaf hairs and toxins produced in the current and subsequent generations 52 In Animals edit It is difficult to trace TEI in animals due to the reprogramming of genes during meiosis and embryogenesis especially in wild populations that are not reared in a lab setting Further studies must be conducted to strengthen the documentation of TEI in animals However a few examples do exist Induced transgenerational epigenetic inheritance has been demonstrated in animals such as Daphnia cucullata These tiny crustaceans will develop protective helmets as juveniles if exposed to kairomones a type of hormone secreted by predators while they are in utero The helmet acts as a method of defense by decreasing the ability of predators to capture the Daphnia thus induction of helmet presence will lower mortality rates D cucullata will develop a small helmet if no kairomones are present However depending upon the level of predator kairomones the length of the helmet will almost double The next generation of Daphnia will display a similar helmet size If the kairomone levels decrease or disappear then the third generation will revert to the original helmet size These organisms display adaptive phenotypes that will affect the phenotype in the subsequent generations 53 Genetic analysis of coral reef fish Acanthochromis polyacanthus has proposed TEI in response to climate change As climate change occurs the ocean water temperature increases When A polyacanthus is exposed to higher water temperatures of up to 3 C from normal ocean temperatures the fish express increased DNA methylation levels on 193 genes resulting in phenotypic changes in the function of oxygen consumption metabolism insulin response energy production and angiogenesis The increase in DNA methylation and its phenotypic affects were carried over to multiple subsequent generations 54 Possible TEI has been studied in guinea pigs Cavia aperea by exposing males to increased ambient temperature for two months In the lab the males were allowed to mate with the same female before and after the heat exposure to determine if the high temperatures affected the offspring Since it serves as a thermoregulatory organ samples of the liver were studied in the father guinea pigs F0 generation and liver and testes of the male offspring F1 generation The F0 males experienced an immediate epigenetic response to the increase in temperature the levels of hormones in the liver responsible for thermoregulation increased The F1 generation also displayed the different methylated epigenetic response in their liver and testes indicating that they could potentially pass on the epigenetic marks to the F2 generation 55 In Humans edit See also Dutch famine of 1944 1945 Although genetic inheritance is important when describing phenotypic outcomes it cannot entirely explain why offspring resemble their parents Aside from genes offspring come to inherit similar environmental conditions established by previous generations One environment that human offspring commonly share with their maternal parent for nine months is the womb Considering the duration of the fetal stages of development the environment of the mother s womb can have long lasting effects on the health of offspring An example of how the environment within the womb can affect the health of an offspring is the Dutch hunger winter of 1944 45 and its causal effect on induced transgenerational epigenetic inherited diseases During the Dutch hunger winter the offspring exposed to famine conditions during the third trimester of development were smaller than those born the year before the famine Moreover the offspring born during the famine and their subsequent offspring were found to have an increased risk of metabolic diseases cardiovascular diseases glucose intolerance diabetes and obesity in adulthood The effects of this famine on development lasted up to two generations 8 56 The increased risk factors to the health of F1 and F2 generations during the Dutch hunger winter is a known phenomenon called fetal programming which is caused by exposure to harmful environmental factors in utero 56 The loss of genetic expression which results in Prader Willi syndrome or Angelman syndrome has in some cases been found to be caused by epigenetic changes or epimutations on both the alleles rather than involving any genetic mutation In all 19 informative cases the epimutations that together with physiological imprinting and therefore silencing of the other allele were causing these syndromes were localized on a chromosome with a specific parental and grandparental origin Specifically the paternally derived chromosome carried an abnormal maternal mark at the SNURF SNRPN and this abnormal mark was inherited from the paternal grandmother 57 Several cancers have been found to be influenced by transgenerational epigenetics Epimutations on the MLH1 gene has been found in two individuals with a phenotype of hereditary nonpolyposis colorectal cancer and without any frank MLH1 mutation which otherwise causes the disease The same epimutations were also found on the spermatozoa of one of the individuals indicating the potential to be transmitted to offspring 57 In addition to epimutations to the MLH1 gene it has been determined that certain cancers such as breast cancer can originate during the fetal stages within the uterus 58 Furthermore evidence collected in various studies utilizing model systems i e animals have found that exposure during parental generations can result in multigenerational and transgenerational inheritance of breast cancer 58 More recently studies have discovered a connection between the adaptation of male germinal cells via pre conception paternal diets and the regulation of breast cancer in developing offspring 58 More specifically studies have begun to uncover new data that underscores a relationship between transgenerational epigenetic inheritance of breast cancer and ancestral alimentary components or associated markers such as birth weight 58 By utilizing model systems such as mice studies have shown that stimulated paternal obesity at the time of conception can epigenetically alter the paternal germ line The paternal germ line is responsible for regulating their daughters weight at birth and the potential for their daughter to develop breast cancer 59 Furthermore it was found that modifications to the miRNA expression profile of the male germline is coupled with elevated body weight 59 Additionally paternal obesity resulted in an increase in the percentage of female offspring developing carcinogen induced mammary tumors which is caused by changes to mammary miRNA expression 59 Aside from cancer related afflictions associated with the effects of transgenerational epigenetic inheritance transgenerational epigenetic inheritance has recently been implicated in the progression of pulmonary arterial hypertension PAH 60 Recent studies have found that transgenerational epigenetic inheritance is likely to be involved in the progression of PAH because current therapies for PAH do not repair the irregular phenotypes associated with this disease 60 Current treatments for PAH have attempted to correct symptoms of PAH with vasodilators and antithrombotic protectors but neither has effectively alleviated the complications related to the impaired phenotypes associated with PAH 60 The inability of vasodilators and antithrombotic protectants to correct PAH suggests that the progression of PAH is dependent upon multiple variables which is likely to be consequent of transgenerational epigenetic inheritance 60 Specifically it is thought that transgenerational epigenetics is linked to the phenotypic changes associated with vascular remodeling 60 For example hypoxia during gestation may induce transgenerational epigenetic alterations that could prove to be detrimental during the early phases of fetal development and increase the possibility of developing PAH as an adult 60 Though hypoxic states could induce the transgenerational epigenetic variance associated with PAH there is strong evidence to support that a variety of maternal risk factors are linked to the eventual progression of PAH 60 Such maternal risk factors linked to late onset PAH includes placental dysfunction hypertension obesity and preeclampsia 60 These maternal risk factors and environmental stressors coupled with transgenerational epigenetic changes can result in prolonged insult to the signaling pathways associated with the vascular development during fetal stages thus increasing the likelihood of having PAH 60 One study has shown childhood abuse which is defined as sexual contact severe physical abuse and or severe neglect leads to epigenetic modifications of glucocorticoid receptor expression 61 62 Glucocorticoid receptor expression plays a vital role in hypothalamic pituitary adrenal HPA activity Additionally animal experiments have shown that epigenetic changes can depend on mother infant interactions after birth 63 Furthermore a recent study investigating the correlations between maternal stress in pregnancy and methylation in teenagers their mothers has found that children of women who were abused during pregnancy were more likely to have methylated glucocorticoid receptor genes 64 Thus children with methylated glucocorticoid receptor genes experience an altered response to stress ultimately leading to a higher susceptibility of experiencing anxiety 64 Additional studies examining the effects of diethylstilbestrol DES which is an endocrine disruptor have found that the grandchildren third generation of women exposed to DES significantly increased the probability of their grandchildren developing attention deficit hyperactivity disorder ADHD 65 This is because women exposed to endocrine disruptors such as DES during gestation may be linked to multigenerational neurodevelopmental deficits 65 Furthermore animal studies indicate that endocrine disruptors have a profound impact on germline cells and neurodevelopment 65 The cause of DES s multigenerational impact is postulated to be the result of biological processes associated with epigenetic reprogramming of the germline though this has yet to be determined 65 Effects on fitness editSee also Neo Lamarckism Epigenetic inheritance may only affect fitness if it predictably alters a trait under selection Evidence has been forwarded that environmental stimuli are important agents in the alteration of epigenes Ironically Darwinian evolution may act on these neo Lamarckian acquired characteristics as well as the cellular mechanisms producing them e g methyltransferase genes Epigenetic inheritance may confer a fitness benefit to organisms that deal with environmental changes at intermediate timescales 66 Short cycling changes are likely to have DNA encoded regulatory processes as the probability of the offspring needing to respond to changes multiple times during their lifespans is high On the other end natural selection will act on populations experiencing changes on longer cycling environmental changes In these cases if epigenetic priming of the next generation is deleterious to fitness over most of the interval e g misinformation about the environment these genotypes and epigenotypes will be lost For intermediate time cycles the probability of the offspring encountering a similar environment is sufficiently high without substantial selective pressure on individuals lacking a genetic architecture capable of responding to the environment Naturally the absolute lengths of short intermediate and long environmental cycles will depend on the trait the length of epigenetic memory and the generation time of the organism Much of the interpretation of epigenetic fitness effects centers on the hypothesis that epigenes are important contributors to phenotypes which remains to be resolved Deleterious effects edit Inherited epigenetic marks may be important for regulating important components of fitness In plants for instance the Lcyc gene in Linaria vulgaris controls the symmetry of the flower Linnaeus first described radially symmetric mutants which arise when Lcyc is heavily methylated 67 Given the importance of floral shape to pollinators 68 methylation of Lcyc homologues e g CYCLOIDEA may have deleterious effects on plant fitness In animals numerous studies have shown that inherited epigenetic marks can increase susceptibility to disease Transgenerational epigenetic influences are also suggested to contribute to disease especially cancer in humans 69 Tumor methylation patterns in gene promoters have been shown to correlate positively with familial history of cancer 70 Furthermore methylation of the MSH2 gene is correlated with early onset colorectal and endometrial cancers 71 Putatively adaptive effects edit Experimentally demethylated seeds of the model organism Arabidopsis thaliana have significantly higher mortality stunted growth delayed flowering and lower fruit set 72 indicating that epigenes may increase fitness Furthermore environmentally induced epigenetic responses to stress have been shown to be inherited and positively correlated with fitness 73 In animals communal nesting changes mouse behavior increasing parental care regimes 74 and social abilities 75 that are hypothesized to increase offspring survival and access to resources such as food and mates respectively Inheritance of Immunity edit Epigenetics play a crucial role in regulation and development of the immune system 76 In 2021 evidence of inheritance of trained immunity across generations to progeny of mice with a systemic infection of Candida albicans was provided 77 The progeny of mice survived the Candida albicans infection via functional transcriptional and epigenetic changes linked to the immune gene loci 77 The responsiveness of myeloid cells to the Candida albicans infection increased in inflammatory pathways and resistance was increased to infections in the next generations 77 Immunity in vertebrates can also be transferred from maternal through the passing of hormones nutrients and antibodies 78 In mammals the maternal factors can be transferred via lactation or the placenta 78 The transgenerational transmission of immune related traits are also described in plants and invertebrates Plants have a defense priming system which enables them to have an alternate defense response that can be accelerated upon exposure to stress actions or pathogens 79 After the event of priming priming stress clue information is stored and the memory may be inherited in the offspring intergenerational or transgenerational 79 In studies the progeny of Pseudomonas syringae infected Arabidopsis were primed during the expression of systemic acquired resistance SAR 80 The progeny showed to have resistance against hemi biotrophic pathogens which is associated with salicylic dependent genes and the defense regulatory gene non expressor of PR genes NPR1 80 Transgenerational SAR in the progeny was associated with increased acetylation of histone 3 at lysine 9 hypomethylation of genes and chromatin marks on promoter regions of salicylic dependent genes 80 Similarly in insects the red flour beetle Trifolium castoreum is primed through the exposure of the pathogen Bacillus thuringiensis 78 Double mating experiments with the red flour beetle demonstrated that paternal transgenerational immune priming is mediated by sperm or seminal fluid which enhances survival upon exposure to pathogens and contribute to epigenetic changes 78 Feedback loops and TEI edit Positive and negative feedback loops are commonly observed in molecular mechanisms and regulation of homeostatic processes There is evidence that feedback loops interact to maintain epigenetic modifications within one generation as well as contributing to TEI in various organisms and these feedback loops can showcase putative adaptations to environmental perturbances Feedback loops are truly a repercussion of any epigenetic modification since it results in changes in expression Even more so the feedback loops seen across multiple generations because of TEI showcases a spatio temporal dynamic that is associated with TEI alone For example elevated temperatures during embryogenesis and PIWI RNA piRNA establishment are directly proportional providing a heritable outcome for repressing transposable elements via piRNA clusters 81 Furthermore subsequent generations retain an active locus to continue establishing piRNA which its formation was previously enigmatic 81 In another case it was suggested that endocrine disruption had a feedback loop interaction with methylation of varying genomic sites in Menidia beryllina which may have been a function of TEI 82 When exposure was removed and M beryllina F2 offspring still retained these methylation marks which caused a negative feedback loop on expression of various genes 82 In another example hybridization of eels can lead to feedback loops contributing to transposon demethylation and transposable element activation 83 Because TE s are typically silenced in the genome their presence and potential expression creates a feedback loop to prevent hybrids from reproducing with other hybrids or non hybrid species which eliminates the proliferation of TE expression and prevents TEI in this context This phenomenon is known as a form of post zygotic reproductive isolation Macroevolutionary patterns editInherited epigenetic effects on phenotypes have been well documented in bacteria protists fungi plants nematodes and fruit flies 84 18 Though no systematic study of epigenetic inheritance has been conducted most focus on model organisms there is preliminary evidence that this mode of inheritance is more important in plants than in animals 84 The early differentiation of animal germlines is likely to preclude epigenetic marking occurring later in development while in plants and fungi somatic cells may be incorporated into the germ line 85 86 It is thought that transgenerational epigenetic inheritance can enable certain populations to readily adapt to variable environments 18 Though there are well documented cases of transgenerational epigenetic inheritance in certain populations there are questions to whether this same form of adaptability is applicable to mammals 18 More specifically it is questioned if it applies to humans 18 As of late most of the experimental models utilizing mice and limited observations in humans have only found epigenetically inherited traits that are detrimental to the health of both organisms 18 These harmful traits range from increased risk of disease such as cardiovascular disease to premature death 18 However this may be based on the premise of limited reporting bias because it is easier to detect negative experimental effects opposed to positive experimental effects 18 Furthermore considerable epigenetic reprogramming necessary for the evolutionary success of germlines and the initial phases of embryogenesis in mammals may be the potential cause limiting transgenerational inheritance of chromatin marks in mammals 18 Life history patterns may also contribute to the occurrence of epigenetic inheritance Sessile organisms those with low dispersal capability and those with simple behavior may benefit most from conveying information to their offspring via epigenetic pathways Geographic patterns may also emerge where highly variable and highly conserved environments might host fewer species with important epigenetic inheritance citation needed Controversies editHumans have long recognized that traits of the parents are often seen in offspring This insight led to the practical application of selective breeding of plants and animals but did not address the central question of inheritance how are these traits conserved between generations and what causes variation Several positions have been held in the history of evolutionary thought Blending vs particulate inheritance edit nbsp Blending inheritance leads to the averaging out of every characteristic which as the engineer Fleeming Jenkin pointed out makes evolution by natural selection impossible Addressing these related questions scientists during the time of the Enlightenment largely argued for the blending hypothesis in which parental traits were homogenized in the offspring much like buckets of different colored paint being mixed together 87 Critics of Charles Darwin s On the Origin of Species pointed out that under this scheme of inheritance variation would quickly be swamped by the majority phenotype 88 In the paint bucket analogy this would be seen by mixing two colors together and then mixing the resulting color with only one of the parent colors 20 times the rare variant color would quickly fade Unknown to most of the European scientific community the monk Gregor Mendel had resolved the question of how traits are conserved between generations through breeding experiments with pea plants 89 Charles Darwin thus did not know of Mendel s proposed particulate inheritance in which traits were not blended but passed to offspring in discrete units that we now call genes Darwin came to reject the blending hypothesis even though his ideas and Mendel s were not unified until the 1930s a period referred to as the modern synthesis Inheritance of innate vs acquired characteristics edit In his 1809 book Philosophie Zoologique 90 Jean Baptiste Lamarck recognized that each species experiences a unique set of challenges due to its form and environment Thus he proposed that the characters used most often would accumulate a nervous fluid Such acquired accumulations would then be transmitted to the individual s offspring In modern terms a nervous fluid transmitted to offspring would be a form of epigenetic inheritance Lamarckism as this body of thought became known was the standard explanation for change in species over time when Charles Darwin and Alfred Russel Wallace co proposed a theory of evolution by natural selection in 1859 Responding to Darwin and Wallace s theory a revised neo Lamarckism attracted a small following of biologists 91 though the Lamarckian zeal was quenched in large part due to Weismann s 92 famous experiment in which he cut off the tails of mice over several successive generations without having any effect on tail length Thus the emergent consensus that acquired characteristics could not be inherited became canon 23 Revision of evolutionary theory edit Non genetic variation and inheritance however proved to be quite common Concurrent with the 20th century development of the modern evolutionary synthesis unifying Mendelian genetics and natural selection C H Waddington 1905 1975 was working to unify developmental biology and genetics In so doing he adopted the word epigenetic 93 to represent the ordered differentiation of embryonic cells into functionally distinct cell types despite having identical primary structure of their DNA 94 Researchers discussed Waddington s epigenetics sporadically it became more of a catch all for puzzling non genetic heritable characters rather than a concept advancing the body of inquiry 95 96 Consequently the definition of Waddington s word has itself evolved broadening beyond the subset of developmentally signaled inherited cell specialization Some scientists have questioned whether epigenetic inheritance compromises the foundation of the modern synthesis Outlining the central dogma of molecular biology Francis Crick 97 succinctly stated DNA is held in a configuration by histone s so that it can act as a passive template for the simultaneous synthesis of RNA and protein s None of the detailed information is in the histone However he closes the article stating this scheme explains the majority of the present experimental results Indeed the emergence of epigenetic inheritance in addition to advances in the study of evolutionary development phenotypic plasticity evolvability and systems biology has strained the current framework of the modern evolutionary synthesis and prompted the re examination of previously dismissed evolutionary mechanisms 98 Furthermore patterns in epigenetic inheritance and the evolutionary implications of the epigenetic codes in living organisms are connected to both Lamarck s and Darwin s theories of evolution 99 For example Lamarck postulated that environmental factors were responsible for modifying phenotypes hereditarily which supports the constructs that exposure to environmental factors during critical stages of development can result in epimutations in germlines thus augmenting phenotypic variance 99 In contrast Darwin s theory claimed that natural selection strengthened a populations ability to survive and remain reproductively fit by favoring populations that are able to readily adapt 99 This theory is consistent with intergenerational plasticity and phenotypic variance resulting from heritable adaptivity 99 In addition some epigenetic variability may provide beneficial plasticity so that certain organisms can adapt to fluctuating environmental conditions However the exchange of epigenetic information between generations can result in epigenetic aberrations which are epigenetic traits that deviate from the norm Therefore the offspring of the parental generations may be predisposed to specific diseases and reduced plasticity due to epigenetic aberrations Though the ability to readily adapt when faced with a new environment may be beneficial to certain populations of species that can quickly reproduce species with long generational gaps may not benefit from such an ability If a species with a longer generational gap does not appropriately adapt to the anticipated environment then the reproductive fitness of the offspring of that species will be diminished There has been critical discussion of mainstream evolutionary theory by Edward J Steele Robyn A Lindley and colleagues 100 101 102 103 104 Fred Hoyle and N Chandra Wickramasinghe 105 106 107 Yongsheng Liu 108 109 Denis Noble 110 111 John Mattick 112 and others that the logical inconsistencies as well as Lamarckian Inheritance effects involving direct DNA modifications as well as the just described indirect viz epigenetic transmissions challenge conventional thinking in evolutionary biology and adjacent fields See also editContribution of epigenetic modifications to evolution Dutch famine of 1944 45 Legacy Epigenetics of anxiety and stress related disorders Overkalix study Transgenerational epigenetic inheritance in plants Transgenerational stress inheritance Transgenerational trauma Inducible plant defenses against herbivoryReferences edit a b Moore David Scott 2015 The developing genome an introduction to behavioral epigenetics Oxford ISBN 978 0 19 992235 2 OCLC 899240120 a href Template Cite book html title Template Cite book cite book a CS1 maint location missing publisher link a b c Heard Edith Martienssen Robert A 2014 03 27 Transgenerational Epigenetic Inheritance Myths and Mechanisms Cell 157 1 95 109 doi 10 1016 j cell 2014 02 045 ISSN 0092 8674 PMC 4020004 PMID 24679529 a b c Fitz James Maximilian H Cavalli Giacomo June 2022 Molecular mechanisms of transgenerational epigenetic inheritance Nature Reviews Genetics 23 6 325 341 doi 10 1038 s41576 021 00438 5 ISSN 1471 0064 PMID 34983971 S2CID 245703043 Fitz James Maximilian H Cavalli Giacomo June 2022 Molecular mechanisms of transgenerational epigenetic inheritance Nature Reviews Genetics 23 6 325 341 doi 10 1038 s41576 021 00438 5 ISSN 1471 0056 PMID 34983971 S2CID 245703043 Iqbal Khursheed Jin Seung Gi Pfeifer Gerd P Szabo Piroska E March 2011 Reprogramming of the paternal genome upon fertilization involves genome wide oxidation of 5 methylcytosine Proceedings of the National Academy of Sciences 108 9 3642 3647 Bibcode 2011PNAS 108 3642I doi 10 1073 pnas 1014033108 ISSN 0027 8424 PMC 3048122 PMID 21321204 Husby Arild 2022 02 09 Wild epigenetics insights from epigenetic studies on natural populations Proceedings of the Royal Society B Biological Sciences 289 1968 20211633 doi 10 1098 rspb 2021 1633 ISSN 0962 8452 PMC 8826306 PMID 35135348 a b Ho Shuk Mei Johnson Abby Tarapore Pheruza Janakiram Vinothini Zhang Xiang Leung Yuet Kin December 2012 Environmental Epigenetics and Its Implication on Disease Risk and Health Outcomes ILAR Journal 53 3 4 289 305 doi 10 1093 ilar 53 3 4 289 ISSN 1084 2020 PMC 4021822 PMID 23744968 a b Emmanuel DROUET 2016 09 30 Epigenetics How the environment influences our genes Encyclopedia of the Environment Retrieved 2023 02 22 a b Jablonka Eva Raz Gal June 2009 Transgenerational Epigenetic Inheritance Prevalence Mechanisms and Implications for the Study of Heredity and Evolution The Quarterly Review of Biology 84 2 131 176 doi 10 1086 598822 ISSN 0033 5770 PMID 19606595 S2CID 7233550 a b Li Dong Yang Yan Li Youping Zhu Xiaohua Li Zeqin 2021 07 01 Epigenetic regulation of gene expression in response to environmental exposures From bench to model Science of the Total Environment 776 145998 Bibcode 2021ScTEn 776n5998L doi 10 1016 j scitotenv 2021 145998 ISSN 0048 9697 S2CID 233548366 DNA Packaging Nucleosomes and Chromatin Learn Science at Scitable www nature com Retrieved 2023 02 26 Wan Qin Li Meng Xiao Wang Chongyang Dai Wenyu Luo Zhenhuan Yin Zhinan Ju Zhenyu Fu Xiaodie Yang Jing Ye Qunshan Zhang Zhan Hui Zhou Qinghua 2022 02 09 Histone H3K4me3 modification is a transgenerational epigenetic signal for lipid metabolism in Caenorhabditis elegans Nature Communications 13 1 768 Bibcode 2022NatCo 13 768W doi 10 1038 s41467 022 28469 4 ISSN 2041 1723 PMC 8828817 PMID 35140229 a b Seong Ki Hyeon Li Dong Shimizu Hideyuki Nakamura Ryoichi Ishii Shunsuke 2011 06 24 Inheritance of Stress Induced ATF 2 Dependent Epigenetic Change Cell 145 7 1049 1061 doi 10 1016 j cell 2011 05 029 ISSN 0092 8674 PMID 21703449 S2CID 2918891 Sen Rwik Barnes Christopher June 2021 Do Transgenerational Epigenetic Inheritance and Immune System Development Share Common Epigenetic Processes Journal of Developmental Biology 9 2 20 doi 10 3390 jdb9020020 ISSN 2221 3759 PMC 8162332 PMID 34065783 a b c d Gapp Katharina Jawaid Ali Sarkies Peter Bohacek Johannes Pelczar Pawel Prados Julien Farinelli Laurent Miska Eric Mansuy Isabelle M May 2014 Implication of sperm RNAs in transgenerational inheritance of the effects of early trauma in mice Nature Neuroscience 17 5 667 669 doi 10 1038 nn 3695 ISSN 1546 1726 PMC 4333222 PMID 24728267 Rodgers Ali B Morgan Christopher P Leu N Adrian Bale Tracy L 2015 11 03 Transgenerational epigenetic programming via sperm microRNA recapitulates effects of paternal stress Proceedings of the National Academy of Sciences 112 44 13699 13704 Bibcode 2015PNAS 11213699R doi 10 1073 pnas 1508347112 ISSN 0027 8424 PMC 4640733 PMID 26483456 a b Rechavi Oded Houri Ze evi Leah Anava Sarit Goh Wee Siong Sho Kerk Sze Yen Hannon Gregory J Hobert Oliver 2014 07 17 Starvation Induced Transgenerational Inheritance of Small RNAs in C elegans Cell 158 2 277 287 doi 10 1016 j cell 2014 06 020 ISSN 0092 8674 PMC 4377509 PMID 25018105 a b c d e f g h i Horsthemke B July 2018 A critical view on transgenerational epigenetic inheritance in humans Nature Communications 9 1 2973 Bibcode 2018NatCo 9 2973H doi 10 1038 s41467 018 05445 5 PMC 6065375 PMID 30061690 Duclos KK Hendrikse JL Jamniczky HA September 2019 Investigating the evolution and development of biological complexity under the framework of epigenetics Evolution amp Development 21 5 247 264 doi 10 1111 ede 12301 PMC 6852014 PMID 31268245 a b Bond DM Finnegan EJ May 2007 Passing the message on inheritance of epigenetic traits Trends in Plant Science 12 5 211 216 doi 10 1016 j tplants 2007 03 010 PMID 17434332 Morison IM Reeve AE 1998 A catalogue of imprinted genes and parent of origin effects in humans and animals Human Molecular Genetics 7 10 1599 1609 doi 10 1093 hmg 7 10 1599 PMID 9735381 Scott RJ Spielman M Bailey J Dickinson HG September 1998 Parent of origin effects on seed development in Arabidopsis thaliana Development 125 17 3329 3341 doi 10 1242 dev 125 17 3329 PMID 9693137 a b Moore DS 2015 The Developing Genome Oxford University Press ISBN 978 0 19 992234 5 pages needed Adenot PG Mercier Y Renard JP Thompson EM November 1997 Differential H4 acetylation of paternal and maternal chromatin precedes DNA replication and differential transcriptional activity in pronuclei of 1 cell mouse embryos Development 124 22 4615 4625 doi 10 1242 dev 124 22 4615 PMID 9409678 Santos F Hendrich B Reik W Dean W January 2002 Dynamic reprogramming of DNA methylation in the early mouse embryo Developmental Biology 241 1 172 182 doi 10 1006 dbio 2001 0501 PMID 11784103 Oswald J Engemann S Lane N Mayer W Olek A Fundele R et al April 2000 Active demethylation of the paternal genome in the mouse zygote Current Biology 10 8 475 478 doi 10 1016 S0960 9822 00 00448 6 PMID 10801417 Fulka H Mrazek M Tepla O Fulka J December 2004 DNA methylation pattern in human zygotes and developing embryos Reproduction 128 6 703 708 doi 10 1530 rep 1 00217 PMID 15579587 S2CID 28719804 Hackett JA Sengupta R Zylicz JJ Murakami K Lee C Down TA Surani MA January 2013 Germline DNA demethylation dynamics and imprint erasure through 5 hydroxymethylcytosine Science 339 6118 448 452 Bibcode 2013Sci 339 448H doi 10 1126 science 1229277 PMC 3847602 PMID 23223451 Surani MA Hajkova P 2010 Epigenetic reprogramming of mouse germ cells toward totipotency Cold Spring Harbor Symposia on Quantitative Biology 75 211 218 doi 10 1101 sqb 2010 75 010 PMID 21139069 Zhang Z Shibahara K Stillman B November 2000 PCNA connects DNA replication to epigenetic inheritance in yeast Nature 408 6809 221 225 Bibcode 2000Natur 408 221Z doi 10 1038 35041601 PMID 11089978 S2CID 205010657 Henderson DS Banga SS Grigliatti TA Boyd JB March 1994 Mutagen sensitivity and suppression of position effect variegation result from mutations in mus209 the Drosophila gene encoding PCNA The EMBO Journal 13 6 1450 1459 doi 10 1002 j 1460 2075 1994 tb06399 x PMC 394963 PMID 7907981 Probst AV Dunleavy E Almouzni G March 2009 Epigenetic inheritance during the cell cycle Nature Reviews Molecular Cell Biology 10 3 192 206 doi 10 1038 nrm2640 PMID 19234478 S2CID 205494340 a b Morgan HD Santos F Green K Dean W Reik W April 2005 Epigenetic reprogramming in mammals Human Molecular Genetics 14 Review Issue 1 R47 R58 doi 10 1093 hmg ddi114 PMID 15809273 Santos F Peters AH Otte AP Reik W Dean W April 2005 Dynamic chromatin modifications characterise the first cell cycle in mouse embryos Developmental Biology 280 1 225 236 doi 10 1016 j ydbio 2005 01 025 PMID 15766761 Taguchi YH 2015 Identification of aberrant gene expression associated with aberrant promoter methylation in primordial germ cells between E13 and E16 rat F3 generation vinclozolin lineage BMC Bioinformatics 16 Suppl 18 S16 doi 10 1186 1471 2105 16 S18 S16 PMC 4682393 PMID 26677731 Richards EJ May 2006 Inherited epigenetic variation revisiting soft inheritance Nature Reviews Genetics 7 5 395 401 doi 10 1038 nrg1834 PMID 16534512 S2CID 21961242 Day Jeremy J 2014 09 30 New approaches to manipulating the epigenome Dialogues in Clinical Neuroscience 16 3 345 357 doi 10 31887 DCNS 2014 16 3 jday ISSN 1958 5969 PMC 4214177 PMID 25364285 a b c d Coe EH June 1959 A regular and continuing conversion type phenomenon at the B locus in maize Proceedings of the National Academy of Sciences of the United States of America 45 6 828 832 Bibcode 1959PNAS 45 828C doi 10 1073 pnas 45 6 828 PMC 222644 PMID 16590451 Chandler VL February 2007 Paramutation from maize to mice Cell 128 4 641 645 doi 10 1016 j cell 2007 02 007 PMID 17320501 a b Stam M Belele C Ramakrishna W Dorweiler JE Bennetzen JL Chandler VL October 2002 The regulatory regions required for B paramutation and expression are located far upstream of the maize b1 transcribed sequences Genetics 162 2 917 930 doi 10 1093 genetics 162 2 917 PMC 1462281 PMID 12399399 a b Belele CL Sidorenko L Stam M Bader R Arteaga Vazquez MA Chandler VL 2013 10 17 Specific tandem repeats are sufficient for paramutation induced trans generational silencing PLOS Genetics 9 10 e1003773 doi 10 1371 journal pgen 1003773 PMC 3798267 PMID 24146624 a b Arteaga Vazquez M Sidorenko L Rabanal FA Shrivistava R Nobuta K Green PJ et al July 2010 RNA mediated trans communication can establish paramutation at the b1 locus in maize Proceedings of the National Academy of Sciences of the United States of America 107 29 12986 12991 Bibcode 2010PNAS 10712986A doi 10 1073 pnas 1007972107 PMC 2919911 PMID 20616013 a b Louwers M Bader R Haring M van Driel R de Laat W Stam M March 2009 Tissue and expression level specific chromatin looping at maize b1 epialleles The Plant Cell 21 3 832 842 doi 10 1105 tpc 108 064329 PMC 2671708 PMID 19336692 a b Haring M Bader R Louwers M Schwabe A van Driel R Stam M August 2010 The role of DNA methylation nucleosome occupancy and histone modifications in paramutation The Plant Journal 63 3 366 378 doi 10 1111 j 1365 313X 2010 04245 x PMID 20444233 a b Dorweiler JE Carey CC Kubo KM Hollick JB Kermicle JL Chandler VL November 2000 mediator of paramutation1 is required for establishment and maintenance of paramutation at multiple maize loci The Plant Cell 12 11 2101 2118 doi 10 1105 tpc 12 11 2101 PMC 150161 PMID 11090212 a b c Chandler V Alleman M April 2008 Paramutation epigenetic instructions passed across generations Genetics 178 4 1839 1844 doi 10 1093 genetics 178 4 1839 PMC 2323780 PMID 18430919 Nobuta K Lu C Shrivastava R Pillay M De Paoli E Accerbi M et al September 2008 Distinct size distribution of endogeneous siRNAs in maize Evidence from deep sequencing in the mop1 1 mutant Proceedings of the National Academy of Sciences of the United States of America 105 39 14958 14963 Bibcode 2008PNAS 10514958N doi 10 1073 pnas 0808066105 PMC 2567475 PMID 18815367 Alleman M Sidorenko L McGinnis K Seshadri V Dorweiler JE White J et al July 2006 An RNA dependent RNA polymerase is required for paramutation in maize Nature 442 7100 295 298 Bibcode 2006Natur 442 295A doi 10 1038 nature04884 PMID 16855589 S2CID 4419412 Arteaga Vazquez MA Chandler VL April 2010 Paramutation in maize RNA mediated trans generational gene silencing Current Opinion in Genetics amp Development 20 2 156 163 doi 10 1016 j gde 2010 01 008 PMC 2859986 PMID 20153628 Huang J Lynn JS Schulte L Vendramin S McGinnis K 2017 01 01 Epigenetic Control of Gene Expression in Maize International Review of Cell and Molecular Biology 328 25 48 doi 10 1016 bs ircmb 2016 08 002 ISBN 9780128122204 PMID 28069135 Chandler VL October 2010 Paramutation s properties and puzzles Science 330 6004 628 629 Bibcode 2010Sci 330 628C doi 10 1126 science 1191044 PMID 21030647 S2CID 13248794 Sobral Mar Sampedro Luis Neylan Isabelle Siemens David Dirzo Rodolfo 2021 08 17 Phenotypic plasticity in plant defense across life stages Inducibility transgenerational induction and transgenerational priming in wild radish Proceedings of the National Academy of Sciences 118 33 e2005865118 Bibcode 2021PNAS 11805865S doi 10 1073 pnas 2005865118 ISSN 0027 8424 PMC 8379918 PMID 34389664 Agrawal Anurag A Laforsch Christian Tollrian Ralph 1999 09 02 Transgenerational induction of defences in animals and plants Nature 401 6748 60 63 Bibcode 1999Natur 401 60A doi 10 1038 43425 ISSN 0028 0836 S2CID 4326322 Ryu Taewoo Veilleux Heather D Donelson Jennifer M Munday Philip L Ravasi Timothy 2018 04 30 The epigenetic landscape of transgenerational acclimation to ocean warming Nature Climate Change 8 6 504 509 Bibcode 2018NatCC 8 504R doi 10 1038 s41558 018 0159 0 ISSN 1758 678X S2CID 90082460 Hu J Barrett R D H 2017 07 20 Epigenetics in natural animal populations Journal of Evolutionary Biology 30 9 1612 1632 doi 10 1111 jeb 13130 ISSN 1010 061X PMID 28597938 S2CID 20558647 a b Stein A D 2004 07 28 Intrauterine famine exposure and body proportions at birth the Dutch Hunger Winter International Journal of Epidemiology 33 4 831 836 doi 10 1093 ije dyh083 ISSN 1464 3685 PMID 15166208 a b Wei Y Schatten H Sun QY 2014 Environmental epigenetic inheritance through gametes and implications for human reproduction Human Reproduction Update 21 2 194 208 doi 10 1093 humupd dmu061 PMID 25416302 a b c d da Cruz R S Chen E Smith M Bates J amp de Assis S 2020 Diet and Transgenerational Epigenetic Inheritance of Breast Cancer The Role of the Paternal Germline Frontiers in nutrition 7 93 https doi org 10 3389 fnut 2020 0009 a b c Fontelles CC Carney E Clarke J Nguyen NM Yin C Jin L Cruz MI Ong TP Hilakivi Clarke L de Assis S June 2016 Paternal overweight is associated with increased breast cancer risk in daughters in a mouse model Scientific Reports 6 28602 Bibcode 2016NatSR 628602F doi 10 1038 srep28602 PMC 4919621 PMID 27339599 a b c d e f g h i Napoli C Benincasa G Loscalzo J April 2019 Epigenetic Inheritance Underlying Pulmonary Arterial Hypertension Arteriosclerosis Thrombosis and Vascular Biology 39 4 653 664 doi 10 1161 ATVBAHA 118 312262 PMC 6436974 PMID 30727752 Weaver IC Cervoni N Champagne FA D Alessio AC Sharma S Seckl JR et al August 2004 Epigenetic programming by maternal behavior Nature Neuroscience 7 8 847 854 doi 10 1038 nn1276 PMID 15220929 S2CID 1649281 McGowan PO Sasaki A D Alessio AC Dymov S Labonte B Szyf M et al March 2009 Epigenetic regulation of the glucocorticoid receptor in human brain associates with childhood abuse Nature Neuroscience 12 3 342 348 doi 10 1038 nn 2270 PMC 2944040 PMID 19234457 Meaney MJ Szyf M 2005 Environmental programming of stress responses through DNA methylation life at the interface between a dynamic environment and a fixed genome Dialogues in Clinical Neuroscience 7 2 103 123 doi 10 31887 DCNS 2005 7 2 mmeaney PMC 3181727 PMID 16262207 a b Radtke KM Ruf M Gunter HM Dohrmann K Schauer M Meyer A Elbert T July 2011 Transgenerational impact of intimate partner violence on methylation in the promoter of the glucocorticoid receptor Translational Psychiatry 1 July 19 e21 doi 10 1038 tp 2011 21 PMC 3309516 PMID 22832523 a b c d Kioumourtzoglou MA Coull BA O Reilly EJ Ascherio A Weisskopf MG July 2018 Association of Exposure to Diethylstilbestrol During Pregnancy With Multigenerational Neurodevelopmental Deficits JAMA Pediatrics 172 7 670 677 doi 10 1001 jamapediatrics 2018 0727 PMC 6137513 PMID 29799929 Jablonka E Lamb MJ 2005 Epigenetic inheritance and evolution the Lamarckian dimension Reprinted ed Oxford Oxford University Press ISBN 978 0 19 854063 2 Cubas P Vincent C Coen E September 1999 An epigenetic mutation responsible for natural variation in floral symmetry Nature 401 6749 157 161 Bibcode 1999Natur 401 157C doi 10 1038 43657 PMID 10490023 S2CID 205033495 Dafni A Kevan PG 1997 Flower size and shape implications in pollination Israeli Journal of Plant Science 45 2 3 201 211 doi 10 1080 07929978 1997 10676684 Nilsson EE Sadler Riggleman I Skinner MK April 2018 Environmentally induced epigenetic transgenerational inheritance of disease Environmental Epigenetics 4 2 dvy016 doi 10 1093 eep dvy016 PMC 6051467 PMID 30038800 Frazier ML Xi L Zong J Viscofsky N Rashid A Wu EF et al August 2003 Association of the CpG island methylator phenotype with family history of cancer in patients with colorectal cancer Cancer Research 63 16 4805 4808 PMID 12941799 Chan TL Yuen ST Kong CK Chan YW Chan AS Ng WF et al October 2006 Heritable germline epimutation of MSH2 in a family with hereditary nonpolyposis colorectal cancer Nature Genetics 38 10 1178 1183 doi 10 1038 ng1866 PMC 7097088 PMID 16951683 Bossdorf O Arcuri D Richards CL Pigliucci M 2010 Experimental alteration of DNA methylation affects the phenotypic plasticity of ecologically relevant traits in Arabidopsis thaliana PDF Evolutionary Ecology 24 3 541 553 doi 10 1007 s10682 010 9372 7 S2CID 15763479 Whittle CA Otto SP Johnston MO Krochko JE 2009 Adaptive epigenetic memory of ancestral temperature regime in Arabidopsis thaliana Botany 87 6 650 657 doi 10 1139 b09 030 Curley JP FA Champagne and P Bateson 2007 Communal nesting induces alternative emotional social and maternal behavior in offspring Society for Behavioral Neuroendocrinology 11th Annual Meeting Pacific Grove CA USA Cited in Branchi I April 2009 The mouse communal nest investigating the epigenetic influences of the early social environment on brain and behavior development Neuroscience and Biobehavioral Reviews 33 4 551 559 doi 10 1016 j neubiorev 2008 03 011 PMID 18471879 S2CID 1592896 Branchi I D Andrea I Fiore M Di Fausto V Aloe L Alleva E October 2006 Early social enrichment shapes social behavior and nerve growth factor and brain derived neurotrophic factor levels in the adult mouse brain Biological Psychiatry 60 7 690 696 doi 10 1016 j biopsych 2006 01 005 PMID 16533499 S2CID 16627324 Sen Rwik Barnes Christopher 2021 05 12 Do Transgenerational Epigenetic Inheritance and Immune System Development Share Common Epigenetic Processes Journal of Developmental Biology 9 2 20 doi 10 3390 jdb9020020 ISSN 2221 3759 PMC 8162332 PMID 34065783 a b c Katzmarski Natalie Dominguez Andres Jorge Cirovic Branko Renieris Georgios Ciarlo Eleonora Le Roy Didier Lepikhov Konstantin Kattler Kathrin Gasparoni Gilles Handler Kristian Theis Heidi Beyer Marc van der Meer Jos W M Joosten Leo A B Walter Jorn November 2021 Transmission of trained immunity and heterologous resistance to infections across generations Nature Immunology 22 11 1382 1390 doi 10 1038 s41590 021 01052 7 hdl 2066 241159 ISSN 1529 2916 PMID 34663978 S2CID 239026066 a b c d Eggert Hendrik Kurtz Joachim Diddens de Buhr Maike F 2014 12 22 Different effects of paternal trans generational immune priming on survival and immunity in step and genetic offspring Proceedings of the Royal Society B Biological Sciences 281 1797 20142089 doi 10 1098 rspb 2014 2089 ISSN 0962 8452 PMC 4240996 PMID 25355479 a b Singh Krishna P Jahagirdar Shamarao Sarma Birinchi Kumar eds 2021 Emerging Trends in Plant Pathology doi 10 1007 978 981 15 6275 4 ISBN 978 981 15 6274 7 S2CID 228078200 a b c Luna Estrella Ton Jurriaan June 2012 The epigenetic machinery controlling transgenerational systemic acquired resistance Plant Signaling amp Behavior 7 6 615 618 doi 10 4161 psb 20155 ISSN 1559 2324 PMC 3442853 PMID 22580690 S2CID 38372184 a b Casier Karine Delmarre Valerie Gueguen Nathalie Hermant Catherine Viode Elise Vaury Chantal Ronsseray Stephane Brasset Emilie Teysset Laure Boivin Antoine 2019 03 15 Nilsen Timothy W Manley James L eds Environmentally induced epigenetic conversion of a piRNA cluster eLife 8 e39842 doi 10 7554 eLife 39842 ISSN 2050 084X PMC 6420265 PMID 30875295 a b Major Kaley M DeCourten Bethany M Li Jie Britton Monica Settles Matthew L Mehinto Alvine C Connon Richard E Brander Susanne M 2020 Early Life Exposure to Environmentally Relevant Levels of Endocrine Disruptors Drive Multigenerational and Transgenerational Epigenetic Changes in a Fish Model Frontiers in Marine Science 7 doi 10 3389 fmars 2020 00471 ISSN 2296 7745 Liu Shenglin Tengstedt Aja Noersgaard Buur Jacobsen Magnus W Pujolar Jose Martin Jonsson Bjarni Lobon Cervia Javier Bernatchez Louis Hansen Michael M August 2022 Genome wide methylation in the panmictic European eel Anguilla anguilla Molecular Ecology 31 16 4286 4306 doi 10 1111 mec 16586 ISSN 0962 1083 PMID 35767387 S2CID 250115270 a b Jablonka E Raz G June 2009 Transgenerational epigenetic inheritance prevalence mechanisms and implications for the study of heredity and evolution The Quarterly Review of Biology 84 2 131 176 CiteSeerX 10 1 1 617 6333 doi 10 1086 598822 PMID 19606595 S2CID 7233550 Whitham TG Slobodchikoff CN July 1981 Evolution by individuals plant herbivore interactions and mosaics of genetic variability The adaptive significance of somatic mutations in plants Oecologia 49 3 287 292 Bibcode 1981Oecol 49 287W doi 10 1007 BF00347587 PMID 28309985 S2CID 20411802 Turian G 1979 Sporogenesis in fungi Annual Review of Phytopathology 12 129 137 doi 10 1146 annurev py 12 090174 001021 Vorzimmer P 1963 Charles Darwin and blending inheritance Isis 54 3 371 390 doi 10 1086 349734 S2CID 143975567 Jenkin F 1867 Review of The Origin of Species North British Review Mendel G 1866 Versuche uber Plflanzenhybriden Verhandlungen des naturforschenden Vereines in Brunn Experiments in Plant Hybridization PDF Read at the February 8th and March 8th 1865 meetings of the Brunn Natural History Society in German Lamarck JB 1809 Philosophie zoologique ou Exposition des considerations relative a l histoire naturelle des animaux Dentu et L Auteur Paris Bowler PJ 1989 Evolution the history of an idea Berkeley University of California Press ISBN 978 0 520 06386 0 Weismann A 1891 Poulton EB Schonland S Shipley E eds Essays upon heredity and kindred biological problems Oxford Clarendon Press doi 10 5962 bhl title 28066 Goldberg AD Allis CD Bernstein E February 2007 Epigenetics a landscape takes shape Cell 128 4 635 638 doi 10 1016 j cell 2007 02 006 PMID 17320500 Waddington CH 2016 1939 Development as an Epigenetic Process Introduction to Modern Genetics London Allen and Unwin ISBN 9781317352037 One of the classical controversies in embryology was that between the preformationists and the epigenisists sic the interaction of these constituents gives rise to new types of tissue and organ which were not present originally and in so far development must be considered as epigenetic Holliday R 2006 Epigenetics a historical overview Epigenetics 1 2 76 80 doi 10 4161 epi 1 2 2762 PMID 17998809 Nanney DL July 1958 Epigenetic Control Systems Proceedings of the National Academy of Sciences of the United States of America 44 7 712 717 Bibcode 1958PNAS 44 712N doi 10 1073 pnas 44 7 712 PMC 528649 PMID 16590265 Crick FH 1958 On protein synthesis PDF Symposia of the Society for Experimental Biology 12 138 163 PMID 13580867 Pigliucci M December 2007 Do we need an extended evolutionary synthesis Evolution International Journal of Organic Evolution 61 12 2743 2749 doi 10 1111 j 1558 5646 2007 00246 x PMID 17924956 a b c d van Otterdijk SD Michels KB July 2016 Transgenerational epigenetic inheritance in mammals how good is the evidence FASEB Journal 30 7 2457 65 doi 10 1096 fj 201500083 PMID 27037350 S2CID 11969347 Steele EJ 1979 Somatic selection and adaptive evolution on the inheritance of acquired characters 1st edit ed Toronto Williams Wallace Steele EJ Lindley RA Blanden RV 1998 Davies P ed Lamarck s signature how retrogenes are changing Darwin s natural selection paradigm Frontiers of Science Sydney Allen amp Unwin Lindley RA 2010 The Soma how our genes really work and how that changes everything Piara Waters CYO Foundation ISBN 978 1451525649 Steele EJ Lloyd SS May 2015 Soma to germline feedback is implied by the extreme polymorphism at IGHV relative to MHC The manifest polymorphism of the MHC appears greatly exceeded at Immunoglobulin loci suggesting antigen selected somatic V mutants penetrate Weismann s Barrier BioEssays 37 5 557 569 doi 10 1002 bies 201400213 PMID 25810320 S2CID 1270807 Steele EJ 2016 Levin M Adams DS eds Origin of congenital defects stable inheritance through the male line via maternal antibodies specific for eye lens antigens inducing autoimmune eye defects in developing rabbits in utero Ahead of the Curve Hidden breakthroughs in the biosciences Bristol UK IOP Publishing pp Chapter 3 Hoyle F Wickramasinghe C 1982 Why neo Darwinism does not work Cardiff University College Cardiff Press ISBN 0 906449 50 2 Hoyle F Wickramasinghe NC 1979 Diseases from space London J M Dent Hoyle F Wickramasinghe NC 1981 Evolution from space London J M Dent Liu Y September 2007 Like father like son A fresh review of the inheritance of acquired characteristics EMBO Reports 8 9 798 803 doi 10 1038 sj embor 7401060 PMC 1973965 PMID 17767188 Liu Y Li X May 2016 Darwin s Pangenesis as a molecular theory of inherited diseases Gene 582 1 19 22 doi 10 1016 j gene 2016 01 051 PMID 26836487 Noble D February 2012 A theory of biological relativity no privileged level of causation Interface Focus 2 1 55 64 doi 10 1098 rsfs 2011 0067 PMC 3262309 PMID 23386960 Noble D August 2013 Physiology is rocking the foundations of evolutionary biology Experimental Physiology 98 8 1235 1243 doi 10 1113 expphysiol 2012 071134 PMID 23585325 S2CID 19689192 Mattick JS October 2012 Rocking the foundations of molecular genetics Proceedings of the National Academy of Sciences of the United States of America 109 41 16400 16401 Bibcode 2012PNAS 10916400M doi 10 1073 pnas 1214129109 PMC 3478605 PMID 23019584 Retrieved from https en wikipedia org w index php title Transgenerational epigenetic inheritance amp oldid 1182811391, wikipedia, wiki, book, books, library,

article

, read, download, free, free download, mp3, video, mp4, 3gp, jpg, jpeg, gif, png, picture, music, song, movie, book, game, games.