Hostname: page-component-848d4c4894-5nwft Total loading time: 0 Render date: 2024-05-15T23:58:52.866Z Has data issue: false hasContentIssue false

Heritable responses to stress in plants

Published online by Cambridge University Press:  07 December 2023

Igor Kovalchuk*
Affiliation:
Department of Biological Sciences, University of Lethbridge, Lethbridge, AB, Canada
*
Corresponding author: Igor Kovalchuk; Email: Igor.kovalchuk@uleth.ca

Abstract

Most plants are adapted to their environments through generations of exposure to all elements. The adaptation process involves the best possible response to fluctuations in the environment based on the genetic and epigenetic make-up of the organism. Many plant species have the capacity to acclimate or adapt to certain stresses, allowing them to respond more efficiently, with fewer resources diverted from growth and development. However, plants can also acquire protection against stress across generations. Such a response is known as an intergenerational response to stress; typically, plants lose most of the tolerance in the subsequent generation when propagated without stress. Occasionally, the protection lasts for more than one generation after stress exposure and such a response is called transgenerational. In this review, we will summarize what is known about inter- and transgenerational responses to stress, focus on phenotypic and epigenetic events, their mechanisms and ecological and evolutionary meaning.

Type
Review
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (https://creativecommons.org/licenses/by/4.0), which permits unrestricted re-use, distribution and reproduction, provided the original article is properly cited.
Copyright
© The Author(s), 2023. Published by Cambridge University Press in association with The John Innes Centre

1. Introduction

Through millions of years of evolution, organisms developed mechanisms of stress avoidance, resistance and tolerance and became well-adapted to their environment. These responses are encoded by the genetic make-up of the organism and are fine-tuned by epigenetic regulations. To be able to respond to the environment in a manner similar to their ancestral generations, the progeny requires faithful replication of their genetic material and epigenetic marks. This is critically important for the survival of an organism in a stable environment. In contrast, survival under stressful conditions requires drastic measures that are implemented quickly. In such a situation, due to the rare nature of mutations, genetic mechanisms may not be able to provide swift and efficient responses for the survival of future generations. In contrast, the regulation at the epigenetic level represents a more versatile and flexible mechanism controlling gene expression and inheritance of old traits and the appearance of new traits (Chang et al., Reference Chang, Zhu, Jiang, Zhang, Zhu and Duan2020). Epigenetic mechanisms are frequently reversible because they do not represent permanent chemical changes (Tao et al., Reference Tao, Shen, Gu, Wang, Yu and He2017). Moreover, the rate and spectrum of epigenetic changes by far exceed those of genetic changes, allowing better phenotypic plasticity and faster adaptation (van der Graaf et al., Reference van der Graaf, Wardenaar, Neumann, Taudt, Shaw, Jansen, Schmitz, Colome-Tatche and Johannes2015).

When environmental conditions become substantially different from normal, the plant employs various mechanisms, including epigenetics, to pass the memory of responses to encountered stresses to the progeny (Nguyen et al., Reference Nguyen, Vu and Cheong2022). This information may be in the form of differentially accumulated metabolites, including primary and secondary metabolites (proteins, fatty acids, messenger ribonucleic acid (mRNA), non-coding RNA (ncRNA), etc.) or chromatin modifications in the form of deoxyribonucleic acid (DNA) methylation or histone modifications. The most well-known examples of such response to stress are known as adaptation and acclimation (Ding et al., Reference Ding, Shi and Yang2020). Changes observed in the progeny are often referred to as intergenerational (Lamke & Baurle, Reference Lamke and Baurle2017) or transgenerational stress response, but they also have several other names, including intergenerational inheritance, intergenerational resilience, plasticity, priming or tolerance. For the sake of this review, we will refer to the changes observed in the immediate progeny of exposed plants as intergenerational changes (IGCs) (Verhoeven et al., Reference Verhoeven, Verbon, van Gurp, Oplaat, Ferreira de Carvalho, Morse, Stahl, Macel and McIntyre2018). In contrast, when the changes persist to the ‘grand progeny’, without stress, we will refer to them as transgenerational changes (TGCs). Furthermore, in this review, we will not cover such classical transgenerational events as paramutations (Heard & Martienssen, Reference Heard and Martienssen2014).

Many parameters likely regulate the ability to establish IGC or TGC, including the species analysed, genetic and epigenetic composition, type of stress, severity of stress, length of the exposure and time during the development when plants were exposed. Also, IGC and TGC manifest themselves as changes in transcriptome, in DNA methylation pattern, in plant physiology and in plant response to stress. We will discuss these points in detail in this review and introduce potential mechanisms of the establishment of heritable memory of stress exposure.

Table 1 Naturally occurring epialleles.

2. IGCs and TGCs

2.1. Types of IGCs or TGCs

IGC and TGC may include alterations at many levels: DNA methylation and histone modifications, changes in transcriptome, including mRNA and ncRNA transcripts, and changes in metabolome and proteome (reviewed in Herman & Sultan, Reference Herman and Sultan2011; Kinoshita & Seki, Reference Kinoshita and Seki2014; Liu et al., Reference Liu, Feng, Gu, Deng, Qiu, Li, Zhang, Wang, Deng, Wang, He, Baurle, Li, Cao and He2019). These changes, when occurring in response to stress, typically lead to higher tolerance to the same or similar stresses but may also result in increased, and sometimes decreased, tolerance to other stresses, for example, higher tolerance to heat stress, but lower tolerance to pathogens. Such changes often disappear in consecutive generations, and most likely, they occur due to differential seed viability or quality caused by the accumulation of metabolites or nutrients, such as starch, hormones, such as abscisic acid and other primary and secondary metabolites that give a certain advantage to plants grown under specific environmental conditions (Donohue, Reference Donohue2009). Occasionally, especially in cases when the stressor persists for longer, TGCs persist, in the form of epialleles. The only well-documented types of so-called natural epialleles are those due to changes in DNA methylation (van der Graaf et al., Reference van der Graaf, Wardenaar, Neumann, Taudt, Shaw, Jansen, Schmitz, Colome-Tatche and Johannes2015). It is important to distinguish such naturally occurring epialleles from IGC and TGC events triggered experimentally. While IGC and TGC observed experimentally cover all the above-mentioned changes, the naturally occurring epialleles only retain changes in methylation patterns. It is possible that some naturally occurring epialleles are the result of spontaneous events, possibly mutations in the components of the epigenetic machinery, leading to heritable epigenetic change. We hypothesize, however, that most naturally occurring epialleles are the consequences of changes in the environment, ‘forcing’ an entire population or a sub-population of plants to acquire an epiallele. In this respect, the TGCs we observe when we conduct experiments are the initial steps towards the formation of epialleles.

2.2. Naturally occurring epialleles as evidence of TGCs

TGCs may be heritable and even persist for many generations, forming epialleles (Quadrana & Colot, Reference Quadrana and Colot2016; Tonosaki et al., Reference Tonosaki, Fujimoto, Dennis, Raboy and Osabe2022; van der Graaf et al., Reference van der Graaf, Wardenaar, Neumann, Taudt, Shaw, Jansen, Schmitz, Colome-Tatche and Johannes2015). Such epialleles typically consist of differentially methylated loci, where cytosines at various positions are hyper- or hypomethylated as compared to the parental alleles. Many known epialleles are believed to have occurred naturally (Table 1). In Linaria vulgaris, hypermethylation of linaria cycloidea-like gene (Lcyc), the gene responsible for flower symmetry, results in a stable phenotype, which reverts occasionally upon a loss of hypermethylation (Cubas et al., Reference Cubas, Vincent and Coen1999). Imprinting of the FLOWERING WAGENINGEN (FWA) gene in Arabidopsis results in female-specific expression, causing a stable late flowering phenotype (Fujimoto et al., Reference Fujimoto, Kinoshita, Kawabe, Kinoshita, Takashima, Nordborg, Nasrallah, Shimizu, Kudoh and Kakutani2008). Variations in methylation of a retrotransposon, named ‘NMR19’ (naturally occurring DNA methylation variation region 19), represent epialleles that control leaf senescence by regulating the expression of PHEOPHYTIN PHEOPHORBIDE HYDROLASE (PPH) in Arabidopsis (He et al., Reference He, Wu, Zinta, Yang, Wang, Liu, Zhang, Zheng, Huang, Zhang and Zhu2018). The NMR19-4 epiallele is heritable and correlates with local climates (He et al., Reference He, Wu, Zinta, Yang, Wang, Liu, Zhang, Zheng, Huang, Zhang and Zhu2018). In rice, heritable hypomethylation in the FERTILIZATION-INDEPENDENT ENDOSPERM1 (FIE1) gene results in the dwarf phenotype (Zhang et al., Reference Zhang, Cheng, Qin, Qiu, Wang, Cui, Gu, Zhang, Guo, Wang, Jiang, Wu, Wang, Cao and Wan2012). FIE1 encodes a component of the polycomb repressive complex 2 involved in H3K27me3-mediated gene repression; this naturally occurring gain-of-function hypomethylation results in the change in histone modifications of hundreds of genes (Zhang et al., Reference Zhang, Cheng, Qin, Qiu, Wang, Cui, Gu, Zhang, Guo, Wang, Jiang, Wu, Wang, Cao and Wan2012). Another case of heritable DNA hypermethylation involves the colourless non-ripening (Cnr) gene, responsible for the fruit ripening and colouring in tomatoes (Manning et al., Reference Manning, Tor, Poole, Hong, Thompson, King, Giovannoni and Seymour2006). In the perennial herb Helleborus foetidus, many heritable size- and fecundity-related traits are controlled by DNA methylation (Alonso et al., Reference Alonso, Perez, Bazaga, Medrano and Herrera2014). Yet, another example of heritable epigenetic changes includes the de novo-originated gene qua-quine starch (QQS) in Arabidopsis thaliana; Silveira et al. (Reference Silveira, Trontin, Cortijo, Barau, Del Bem, Loudet, Colot and Vincentz2013) found substantial variations in DNA methylation in natural accessions of Arabidopsis, with many hypomethylated states inherited for up to eight generations (Silveira et al., Reference Silveira, Trontin, Cortijo, Barau, Del Bem, Loudet, Colot and Vincentz2013).

Figure 1. Potential mechanism of establishment of transgenerational effects and development of new epialleles. In the proposed scenario, stress generates mobile response molecules, likely in the form of small interfering RNAs (siRNAs) or other types of ncRNAs, but could also include differential levels of proteins, metabolites and various histone modifications, which reach meiocytes and alter DNA methylation and gene expression patterns. Developing meiocytes may retain certain signals and pass new epigenetic patterns into gametes. It is possible that some of the differentially expressed ncRNAs, as well as mRNAs, especially from female gametes, are preserved and influence the developing progeny. The persistence of stress may further reinforce these signalling molecules, leading to the development of stable changes in DNA methylation that do not revert even when stress is absent. Such changes in DNA methylation and chromatin structure represent epimutations and could lead to the development of epialleles persisting for many generations.

2.3. IGCs or TGCs in the form of changes in the plant stress response

As stated above, IGC and TGC manifest themselves in various forms, with the most common being changes in phenotype, stress tolerance and epigenetic modifications. The most desired effect of IGC or TGC is an increased stress tolerance that does not affect the plant performance under normal conditions.

Several studies have found that exposure to elevated CO2 levels has a transgenerational effect on plant biomass (Bezemer et al., Reference Bezemer, Thompson and Jones2004; Lau et al., Reference Lau, Peiffer, Reich and Tiffin2008; Li et al., Reference Li, Li and Yu2017; Lv et al., Reference Lv, Hu, Wei and Wang2022). The immediate progeny of Poa pratensis exposed to high CO2 level exhibited higher biomass and produced more tillers (Bezemer et al., Reference Bezemer, Thompson and Jones2004). IGC in response to elevated CO2 and increased N (nitrogen) deposition was observed in Lupinus perennis, Poa pratensis and Schizachyrium scoparium (Lau et al., Reference Lau, Peiffer, Reich and Tiffin2008). In particular, the authors found increased biomass and higher seed weight in the progeny of plants exposed to high CO2 when they were grown in the presence of high CO2 or N; curiously, the progeny of plants exposed to high N did not perform better in response to high N, but did do better when grown on high CO2 as compared to the progeny of plants grown in normal CO2 and N (Lau et al., Reference Lau, Peiffer, Reich and Tiffin2008). Two generations of exposure of wheat (Triticum aestivum L.) to elevated CO2 resulted in increased stomatal conductance and drought tolerance (Li et al., Reference Li, Li and Yu2017). In contrast, Lv et al. (Reference Lv, Hu, Wei and Wang2022) found that five consecutive generations of rice exposure to elevated CO2 levels resulted in a decreased rate of photosynthesis and a negative effect on plant growth (Lv et al., Reference Lv, Hu, Wei and Wang2022).

Klironomos et al. (Reference Klironomos, Allen, Rillig, Piotrowski, Makvandi-Nejad, Wolfe and Powell2005) studied the effect of 21 generations of a perennial grass Bromus inermis exposure to elevated CO2; they analysed the response of mycorrhizal symbiotic system to abrupt (from ambient 350 p.p.m. to 550 p.p.m.) or gradual (10 p.p.m. increase per generation, from 350 p.p.m. to 550 p.p.m.) increase in CO2 concentration (Klironomos et al., Reference Klironomos, Allen, Rillig, Piotrowski, Makvandi-Nejad, Wolfe and Powell2005). The authors did not find any difference between the generation 21 and generation 1 plants in the biomass or photosynthesis rate, while they found that exposure to an abrupt change in CO2 resulted in a significant decrease in biodiversity as compared to ambient CO2 or a gradual change in progeny (Klironomos et al., Reference Klironomos, Allen, Rillig, Piotrowski, Makvandi-Nejad, Wolfe and Powell2005).

More recent data demonstrated that exposure of Arabidopsis thaliana and Physcomitrium patens to high CO2 resulted in accelerated growth rates in the immediate progeny (Panda et al., Reference Panda, Mohanasundaram, Gutierrez, McLain, Castillo, Sheng, Casto, Gratacos, Chakrabarti, Fahlgren, Pandey, Gehan and Slotkin2023). The authors showed that this intergenerational effect was dependent on DNA methylation, the function of RNA-dependent DNA methylation (RdDM) machinery and Chromomethyltransferase 2 (CMT2) and CMT3 DNA methyltransferases (Panda et al., Reference Panda, Mohanasundaram, Gutierrez, McLain, Castillo, Sheng, Casto, Gratacos, Chakrabarti, Fahlgren, Pandey, Gehan and Slotkin2023).

The progeny of Oryza sativa L. exposed to heavy metals was found to be more tolerant to the same stress (Ou et al., Reference Ou, Zhang, Xu, Lin, Zang, Zhuang, Jiang, von Wettstein and Liu2012). Increased tolerance to heavy metal stress was also found in the progeny of rice plants exposed to heavy metals; the authors also found changes in the expression of various transporters and these changes were also observed in the second generation, when plants were propagated in normal conditions (Cong et al., Reference Cong, Miao, Xu, Zhang, Yuan, Wang, Zhuang, Lin, Jiang, Wang, Ma, Sanguinet, Liu, Rustgi and Ou2019).

Arabidopsis plants were exposed to salt for five generations, and the authors found evidence of higher tolerance to salt only starting from the second generation, while no such adaptation was found in the first generation after stress exposure (Wibowo et al., Reference Wibowo, Becker, Marconi, Durr, Price, Hagmann, Papareddy, Putra, Kageyama, Becker, Weigel and Gutierrez-Marcos2016). They also noted that the removal of stress at any generation resulted in the loss of this tolerance in the progeny, indicating a transient nature of this change, or an IGC (Wibowo et al., Reference Wibowo, Becker, Marconi, Durr, Price, Hagmann, Papareddy, Putra, Kageyama, Becker, Weigel and Gutierrez-Marcos2016).

The progeny of oilseed rape exposed to drought showed lower quality of seeds but higher tolerance to drought (Hatzig et al., Reference Hatzig, Nuppenau, Snowdon and Schiessl2018). Similar results were found for rice; exposure to drought for 11 generations improved drought tolerance and oxidative stress resilience (Zheng et al., Reference Zheng, Chen, Xia, Wei, Lou, Li, Li and Luo2017). Also, the progeny of Polygonum persicaria plants exposed to drought had longer roots and larger biomass (Herman & Sultan, Reference Herman and Sultan2011). The authors found the effect of two successive generations of drought stress to be cumulative, resulting in greater provisioning, root growth and survivorship when the progeny was exposed to stress. A positive effect on seedling development was even observed when the progeny of stressed plants was propagated in normal conditions, indicating TGC at least for some traits (Herman et al., Reference Herman, Sultan, Horgan-Kobelski and Riggs2012).

The immediate progeny of ultraviolet C (UVC)-treated Arabidopsis plants exhibited an increase in the seed size, a decrease in the leaf number and an earlier bolting time (Migicovsky & Kovalchuk, Reference Migicovsky and Kovalchuk2014). Similar changes were found in the progeny of heat-stressed plants (Migicovsky et al., Reference Migicovsky, Yao and Kovalchuk2014). Earlier bolting, larger seeds and changes in the leaf number or size are likely some of the mechanisms of adaptation to UV stress.

Higher tolerance to stress was also observed in the progeny of plants infected with pathogens. Luna et al. (Reference Luna, Bruce, Roberts, Flors and Ton2012) found that the progeny of plants infected with Pseudomonas syringae exhibited reduced bacterial colonization when encountering similar infection and higher tolerance to a fungal pathogen Hyaloperonospora arabidopsidis (Luna et al., Reference Luna, Bruce, Roberts, Flors and Ton2012 ). This IGC became a TGC when plants were propagated for one more generation without stress—a higher pathogen tolerance was observed (Luna et al., Reference Luna, Bruce, Roberts, Flors and Ton2012). Slaughter et al. (Reference Slaughter, Daniel, Flors, Luna, Hohn and Mauch-Mani2012) confirmed the finding by Luna et al. (Reference Luna, Bruce, Roberts, Flors and Ton2012) in the establishment of IGC in response to infection with Pseudomonas syringae but found that the propagation without stress removed this tolerance; thus, no TGC was established (Slaughter et al., Reference Slaughter, Daniel, Flors, Luna, Hohn and Mauch-Mani2012). IGC events in the form of cross-tolerance to infection with various pathogens seem to be common. The progeny of tobacco plants infected with tobacco mosaic virus (TMV) not only exhibited higher tolerance to TMV infection but also to inoculation with the bacterial pathogen Pseudomonas syringae and the fungal pathogen Phytophthora nicotianae and higher tolerance to the chemical methyl methanesulfonate (MMS) (Kathiria et al., Reference Kathiria, Sidler, Golubov, Kalischuk, Kawchuk and Kovalchuk2010).

Insect grazing also led to heritable events. The progeny of wild radish exposed to herbivores was resistant to herbivory (Agrawal, Reference Agrawal2001). Also, yellow monkeyflower plants respond to herbivory with an increased trichome density in the progeny; trichome density positively correlates with tolerance to herbivores (Holeski et al., Reference Holeski, Chase-Alone and Kelly2010). Exposure of Arabidopsis and tomato plants to caterpillar herbivory resulted in enhanced resistance to two of three herbivores tested in the progeny (Rasmann et al., Reference Rasmann, De Vos, Casteel, Tian, Halitschke, Sun, Agrawal, Felton and Jander2012). This effect was partially transmitted to the next generation when plants were propagated in normal conditions but was lost when they were propagated to the third generation (Rasmann et al., Reference Rasmann, De Vos, Casteel, Tian, Halitschke, Sun, Agrawal, Felton and Jander2012). Also, exposure of Solanum carolinense to caterpillar herbivory led to greater emergence, earlier flowering and larger seed yield in the progeny (Nihranz et al., Reference Nihranz, Walker, Brown, Mescher, De Moraes and Stephenson2020). Wounding often mimics the attack by insect; the progeny of wounded plants exhibited higher trichome density and herbivore resistance (Colicchio, Reference Colicchio2017). Also, chemicals mimicking pathogen attack, such as jasmonic acid (JA), trigger heritable changes; dandelion plants treated with JA showed heritable changes in the transcriptomes and metabolomes; the intergenerational effect of treatment was very substantial—about 40% of changes in transcriptome and 10% of changes in metabolome were heritable (Verhoeven et al., Reference Verhoeven, Verbon, van Gurp, Oplaat, Ferreira de Carvalho, Morse, Stahl, Macel and McIntyre2018).

2.4. Changes in DNA methylation in the progeny of stressed plants

Heritable changes in DNA methylation in response to stress have been observed in many reports. A dose-dependent genome hypermethylation was found in the pine trees grown in the Chernobyl area—the progeny germinated from seeds of trees grown in areas with higher radiation load was more hypermethylated (Kovalchuk et al., Reference Kovalchuk, Burke, Arkhipov, Kuchma, James, Kovalchuk and Pogribny2003). More recently, it was shown that the exposure of Arabidopsis plants for three generations to different levels of radiation also resulted in an increase in DNA methylation, primarily in the CG context; the authors noted that the highest level of radiation was less efficient in the establishment of IGC in DNA methylation (Laanen et al., Reference Laanen, Saenen, Mysara, Van de Walle, Van Hees, Nauts, Van Nieuwerburgh, Voorspoels, Jacobs, Cuypers and Horemans2021).

Genome hypermethylation was observed in the progeny of Arabidopsis plants exposed to salt for five generations (Wibowo et al., Reference Wibowo, Becker, Marconi, Durr, Price, Hagmann, Papareddy, Putra, Kageyama, Becker, Weigel and Gutierrez-Marcos2016). They found that these methylation changes occurred primarily in CHG and CHH contexts and these changes correlated well with stress treatment, whereas changes in CG methylation patterns occurred stochastically (Wibowo et al., Reference Wibowo, Becker, Marconi, Durr, Price, Hagmann, Papareddy, Putra, Kageyama, Becker, Weigel and Gutierrez-Marcos2016).

In rice exposed to drought for 11 generations, changes in DNA methylation were not linear, with the largest change observed between generations 10 and 11 (Zheng et al., Reference Zheng, Chen, Xia, Wei, Lou, Li, Li and Luo2017). They found that hypomethylation occurred primarily at CG and CHG contexts at intergenic regions, while hypermethylation occurred mainly in CHH associated with transposable elements. The recurring methylation changes observed in all generations were predominantly at CHH. Finally, they found DNA methylation changes maintained in the progeny propagated in normal watering condition after 11 generations of draught exposure, a transgenerational event (Zheng et al., Reference Zheng, Chen, Xia, Wei, Lou, Li, Li and Luo2017).

Changes in CHG methylation were also inherited in rice exposed to heavy metals—hypomethylation of cytosines in the CHG context was found (Ou et al., Reference Ou, Zhang, Xu, Lin, Zang, Zhuang, Jiang, von Wettstein and Liu2012). Exposure of rice to various heavy metal salts showed a complex pattern of changes in DNA methylation in several transposons in the progeny, and these changes persisted to a second generation when plants were propagated in normal conditions, again, a TGC (Cong et al., Reference Cong, Miao, Xu, Zhang, Yuan, Wang, Zhuang, Lin, Jiang, Wang, Ma, Sanguinet, Liu, Rustgi and Ou2019).

The role of DNA methylation in the establishment of IGCs in Polygonum persicaria plants was also shown in the response to drought; while the progeny of drought-exposed plants showed IGC, treatment with the demethylation agent zebularine removed the adaptive advantage, indicating a critical role of methylation in the process of IGC establishment (Herman & Sultan, Reference Herman and Sultan2011).

The work by Zheng et al. (Reference Zheng, Chen, Li, Lou, Xia, Wang, Li, Liu and Luo2013) demonstrated subtle changes in DNA methylation in rice in response to drought for six generations; it was found that only the drought-sensitive variety responded in a meaningful way, while changes in the resistant variety were negligible (Zheng et al., Reference Zheng, Chen, Li, Lou, Xia, Wang, Li, Liu and Luo2013). Similarly, Arabidopsis plants exposed to drought exhibited only subtle stochastic changes in DNA methylation that did not accumulate in consecutive generations of drought exposure (Ganguly et al., Reference Ganguly, Crisp, Eichten and Pogson2017).

Exposure to many other stresses such as salt, flood, heat, cold and UVC also led to changes in DNA methylation in the progeny; in all these cases, global genome hypermethylation was observed (Boyko et al., Reference Boyko, Blevins, Yao, Golubov, Bilichak, Ilnytskyy, Hollunder, Meins and Kovalchuk2010). As we mentioned above, changes in methylation often persist for several generations after stress has been removed. In Arabidopsis plants exposed to salt, water or temperature stress, hypermethylation persisted to a second generation when plants were propagated under normal conditions (Boyko et al., Reference Boyko, Blevins, Yao, Golubov, Bilichak, Ilnytskyy, Hollunder, Meins and Kovalchuk2010).

Global genome hypermethylation in the progeny of stressed Arabidopsis plants does not reflect changes in the individual loci. Promoters of SUVH2, SUVH5 and SUVH8 genes involved in the regulation of the chromatin structure, and the promoter of ROS1, responsible for demethylation activities, were hypermethylated, while the promoters of stress-responsive genes UVH3, ERF1, TUBG1 and RAP2.7 were hypomethylated (Bilichak et al., Reference Bilichak, Ilnystkyy, Hollunder and Kovalchuk2012). As in the case of Polygonum persicaria plants described above, exposure of seeds of the progeny of salt-stressed plants to 5-azaC, a chemical compound that modifies cytosines by preventing methylation, removes the positive IGC in the form of stress tolerance and prevents the inheritance of hypermethylation (Boyko et al., Reference Boyko, Blevins, Yao, Golubov, Bilichak, Ilnytskyy, Hollunder, Meins and Kovalchuk2010).

Similar to the changes in methylation found in response to abiotic stresses, global genome hypermethylation was also observed in the progeny of TMV-infected tobacco plants; hypermethylation persisted in the second generation propagated in a normal environment (Boyko et al., Reference Boyko, Kathiria, Zemp, Yao, Pogribny and Kovalchuk2007). Loci that were undergoing rearrangements were found to be hypomethylated, while loci that were stable were either normally methylated or hypermethylated. It can be hypothesized that such differential methylation controls the rearrangements in the genome of stressed plants (Boyko et al., Reference Boyko, Kathiria, Zemp, Yao, Pogribny and Kovalchuk2007).

3. Possible mechanisms involved in the regulation of TGCs

What are the mechanisms that control heritable changes in response to stress? How is the specificity of changes established and how are they propagated? To understand it, we first need to understand how genetic information is normally inherited in plant gametes. Plant gametes are established late in development. Meiocytes differentiate from somatic meristematic cells. They differentiate into microspores and megaspores, and after several cell divisions, they give rise to pollen and ovum. Pollen consists of generative cell (GC) and vegetative cell (VC), and they differ in gene expression and the presence of siRNAs. While VC is hypomethylated and has considerably higher levels of expression of various genes, including those giving rise to siRNAs, the GC is fairly hypermethylated, with poor gene expression and low level of siRNAs. siRNAs expressed in VC can cross to GC where they are involved in the suppression of transposon activity (Martinez & Kohler, Reference Martinez and Kohler2017).

Several mechanisms may be involved, and research demonstrates the role of RdDM, ncRNAs, DNA methylation and demethylation processes and histone modifications. The accumulation of metabolites, proteins or certain coding and non-coding RNAs may also play a role in the establishment of IGC, as they may give an advantage to the developing embryo. While all the above-mentioned molecules may contribute to IGCs, for TGCs, the involvement of metabolites, proteins or transcripts is highly unlikely, unless there is a certain mechanism of amplification of such metabolites or proteins, which has not yet been ruled out. Accumulation of stress-induced molecules is likely to affect female gametes more than male gametes, simply due to the larger cytoplasmic content of the former. Indeed, it was shown that epigenetic memory of salt stress is primarily established through the female gametes, while in the male gametes, changes in the DNA methylation were erased by the activity of DNA glycosylases, demonstrating both that heritable events are controlled by methylation and that there is a specific mechanism to restrict transmission of these events through male gametes (Wibowo et al., Reference Wibowo, Becker, Marconi, Durr, Price, Hagmann, Papareddy, Putra, Kageyama, Becker, Weigel and Gutierrez-Marcos2016).

3.1. The role of epigenetic regulators

Epigenetics is the most plausible mechanism behind heritable changes in response to stress. DNA methylation is likely to play the most crucial role. In plants, DNA methylation occurs in various sequence contexts, including symmetrical methylation at CG and CHG sites and asymmetrical methylation at CHH sites. Control of DNA methylation in plants is complex, with symmetrical CpG and CpHpG and non-symmetrical CpHpH methylation established and maintained through multiple, partially redundant mechanisms. De novo symmetrical methylation is established by the DOMAINS REARRANGED METHYLTRANSFERASE 2 (DRM2) with the help of ncRNAs of the RdDM pathway, while maintained by the METHYLTRANSFERASE 1 (MET1) in the CpG context and CMT2/CMT3 proteins in the CpHpG context (Zhang et al., Reference Zhang, Lang and Zhu2018). CMT3 is recruited to the repressive histone mark H3K9me2 (Du et al., Reference Du, Johnson, Jacobsen and Patel2015), and in turn, CMT3 binding to DNA can facilitate the recruitment of H3K9me2 (Du et al., Reference Du, Johnson, Jacobsen and Patel2015). In contrast, CpHpH methylation is established by DRM2 and maintained by DRM2 at short transposons in euchromatic regions and by CHROMOMETHYLASE 2 (CMT2) at large transposons in heterochromatic regions (Zemach et al., Reference Zemach, Kim, Hsieh, Coleman-Derr, Eshed-Williams, Thao, Harmer and Zilberman2013). DRM2 uses 24-nt siRNAs to guide DNA methylation at euchromatic TEs (Law & Jacobsen, Reference Law and Jacobsen2010; Matzke & Mosher, Reference Matzke and Mosher2014), while Decreased DNA Methylation I (DDM1) mediates recruitment of CMT2 to pericentromeric H3K9me2 regions (Stroud et al., Reference Stroud, Do, Du, Zhong, Feng, Johnson, Patel and Jacobsen2014). The functionality of the RdDM pathway is also partially dependent on Dicer-like (DCL) proteins, DCL2, DCL3 and DCL4 (Yang et al., Reference Yang, Zhang, Tang, Li, Yang, Huang, Zhang and Zhu2016). It can thus be hypothesized that the RdDM pathway is responsible for heritable changes in phenotype.

Experiments in our laboratory and the work of the others partially confirmed this hypothesis. We found that dcl2 and dcl3 plants but not dcl4 plants exposed to UVC were impaired in IGCs in transposon activation, changes in leaf size, differential changes in the histone marks and expression of several repair genes (Migicovsky & Kovalchuk, Reference Migicovsky and Kovalchuk2014). The more prominent role of DCL2 and DCL3 as compared to DCL4 in the establishment of IGC and TGC was also confirmed in the progeny of Arabidopsis exposed to heat (Migicovsky et al., Reference Migicovsky, Yao and Kovalchuk2014).

Rasmann et al. (Reference Rasmann, De Vos, Casteel, Tian, Halitschke, Sun, Agrawal, Felton and Jander2012) obtained similar results—they found the Arabidopsis dcl2 dcl3 dcl4 triple mutant impaired in passing the memory of exposure to herbivory to the progeny (Rasmann et al., Reference Rasmann, De Vos, Casteel, Tian, Halitschke, Sun, Agrawal, Felton and Jander2012). Somewhat different results were reported by Ito et al. (Reference Ito, Gaubert, Bucher, Mirouze, Vaillant and Paszkowski2011); they found that the heat-induced expression of ONSEN was higher in the dcl3 plants compared with the wild-type plants and suggested that DCL3 may be partially restricting the accumulation of ONSEN in response to heat stress in somatic tissues (Ito et al., Reference Ito, Gaubert, Bucher, Mirouze, Vaillant and Paszkowski2011). They found a higher rate of transposition of ONSEN and new reinsertions in the progeny of heat-stressed dcl3 plants. Hence, despite the fact that the authors reported somewhat different results than the two above-mentioned studies, they still suggested the role of siRNA biogenesis in the regulation of heritable response to stress. Likewise, the potential role of RdDM was also suggested for the response to the elevated levels of CO2; changes in the plant physiology and changes in DNA methylation in the progeny were dependent on the function of RdDM machinery, specifically CMT2 and CMT3 DNA methyltransferases (Panda et al., Reference Panda, Mohanasundaram, Gutierrez, McLain, Castillo, Sheng, Casto, Gratacos, Chakrabarti, Fahlgren, Pandey, Gehan and Slotkin2023). It should be noted that the siRNAs may not be absolutely required for intergenerational memory, as the changes in the DNA methylation in the stressed plants can occur through the RDR6-RdDM pathway (Nuthikattu et al., Reference Nuthikattu, McCue, Panda, Fultz, DeFraia, Thomas and Slotkin2013) or through the activity of DNA glycosylases (Williams et al., Reference Williams, Bechen, Pohlmann and Gehring2022).

DDM1 can also play a role in the establishment of heritable response to stress as it regulates the recruitment of CMT2 to DNA (Stroud et al., Reference Stroud, Do, Du, Zhong, Feng, Johnson, Patel and Jacobsen2014). DDM1 mutant has substantial loss of DNA methylation and activation of transcription of many resistance genes. Furci et al. (Reference Furci, Jain, Stassen, Berkowitz, Whelan, Roquis, Baillet, Colot, Johannes and Ton2019) have analysed the pathogen tolerance of epigenetic recombinant inbred lines (epiRILs) obtained by crosses of ddm1 mutant and wild-type Arabidopsis plants; the progeny of the cross maintained hypomethylated status of many loci in the absence of ddm1 mutation for sixteen generations (Furci et al., Reference Furci, Jain, Stassen, Berkowitz, Whelan, Roquis, Baillet, Colot, Johannes and Ton2019). They found several epigenetic quantitative trait loci (epiQTLs) associated with the priming of defence-related genes rendering plants resistant to biotrophic downy mildew pathogen Hyaloperonospora arabidopsidis (Furci et al., Reference Furci, Jain, Stassen, Berkowitz, Whelan, Roquis, Baillet, Colot, Johannes and Ton2019). They further propagated these plants to F9 and F10 generations and confirmed that the resistance to this pathogen was retained, although it was lost in ~2.5% (2 of 40 families), and in the remaining families, considerable variations in the resistance were observed (Furci et al., Reference Furci, Jain, Stassen, Berkowitz, Whelan, Roquis, Baillet, Colot, Johannes and Ton2019).

The mechanism of IGC and TGC may involve several steps. First, on the level of somatic cells, stress response includes differential expression of mRNAs, ncRNAs and changes in DNA methylation and histone modifications. If stress occurs early during development and influences the whole plant, gamete cells that would derive from the meristem will acquire and propagate the signal. If stress occurs when gametes are established, they may also be altered in response to stress. Even if meristem cells or gametes are not altered directly, these cells may acquire information about stress from all other somatic cells through the active functions of plasmodesmata and phloem that circulate a variety of molecules, including ncRNAs (Maizel et al., Reference Maizel, Markmann, Timmermans and Wachter2020; Yang et al., Reference Yang, Cui, Feng, Hu, Liu and Duan2023). It is possible that changes in DNA methylation and histone modifications caused by the RdDM mechanism may already occur in meristem cells or early during gametogenesis. Second, changes that occur in meristem cells or in the developing gametes have to survive reprogramming, a mechanism that erases the epigenetic marks, such as changes in DNA methylation, histone modifications and degradation of mRNA in pollen (Borg et al., Reference Borg, Papareddy, Dombey, Axelsson, Nodine, Twell and Berger2021). Male and female gametes likely do not contribute to the heritable memory in an equal manner. It was shown that female gametes accumulate greater amount of polymerase IV (PolIV)-dependent ncRNAs than male gametes (Mosher et al., Reference Mosher, Melnyk, Kelly, Dunn, Studholme and Baulcombe2009). It is proposed that heritable response to stress is mainly under maternal control (Pecinka & Mittelsten Scheid, Reference Pecinka and Mittelsten Scheid2012). Although the evidence is scarce, at least one report by Wibowo et al. (Reference Wibowo, Becker, Marconi, Durr, Price, Hagmann, Papareddy, Putra, Kageyama, Becker, Weigel and Gutierrez-Marcos2016) demonstrates that enhanced tolerance to hyperosmotic stress in the progeny is passed through the female germline (Wibowo et al., Reference Wibowo, Becker, Marconi, Durr, Price, Hagmann, Papareddy, Putra, Kageyama, Becker, Weigel and Gutierrez-Marcos2016). One of the DNA glycosylases, DEMETER (DME), is known to be especially active during male gametogenesis and is suggested to play a critical role in the eraser of methylation marks during the reprogramming step (Khouider et al., Reference Khouider, Borges, LeBlanc, Ungru, Schnittger, Martienssen, Colot and Bouyer2021). The authors exposed dme-6 plants to hyperosmotic stress for two generations and found the progeny of these plants to be more tolerant to hyperosmotic stress as compared to the progeny of wild-type plants, suggesting that DME actively resetting the memory of stress in the male gametes (Wibowo et al., Reference Wibowo, Becker, Marconi, Durr, Price, Hagmann, Papareddy, Putra, Kageyama, Becker, Weigel and Gutierrez-Marcos2016). Also, much higher genome instability was observed in the progeny of UVC- and salt-stressed plants when the non-exposed pollen was used to pollinate the exposed ova, as compared to fertilization of the non-exposed ova with the exposed pollen (Boyko & Kovalchuk, Reference Boyko and Kovalchuk2010). It was also demonstrated in Arabidopsis that transgenerational phenotype aggravation in the Chromatin assembly factor-1 (CAF-1) mutant, impaired in chromatin assembly, was predominantly propagated by female gametes (Mozgova et al., Reference Mozgova, Wildhaber, Trejo-Arellano, Fajkus, Roszak, Kohler and Hennig2018).

Epigenetic changes caused by stress also need to survive the second level of reprogramming that occurs after the fertilization event. It is possible that changes in DNA methylation occur in mature gametes or early embryos and are caused by differential expression of ncRNAs produced in gametes or embryos, or even in the endosperm. Third, it is possible that some of the differentially expressed ncRNAs may survive all reprogramming steps and trigger changes directly in the progeny. Our recent work in Brassica rapa showed that heat stress induces changes in ncRNA and mRNA expression in meristem tissues and gametes; some of these changes were propagated into the developing embryo and even into the progeny (Byeon et al., Reference Byeon, Bilichak and Kovalchuk2019).

Fourth, the propagation of stress memory and the maintenance of phenotypic changes in the next generations may require continuous stress exposure (generation after generation). This is not surprising because if changes in DNA methylation and ncRNA expression that trigger it play an essential role, they need to be generated constantly to reinforce transgenerational memory and replenish the molecules depleted during reprogramming.

It is curious that DNA methylation changes represent the most common TGC in the papers we described above. We can assume that TGCs are triggered by differential expression of non-coding RNAs that target various genomic loci to establish differential methylation and differential gene expression, leading to changes in stress tolerance. DNA methylation is maintained more consistently regardless of whether plants are exposed to stress for the second time, while stress tolerance depends on the second stress exposure, which suggests that changes in DNA methylation are more robust and can persist in the absence of stress re-exposure.

4. Evolutionary significance of IGC and TGC, cost and benefits and maladaptation to stress

In this review, we have presented multiple examples of IGC and TGC in response to stress in plants and discussed the type of changes that occur and the potential mechanisms of their establishment. Are the IGCs or TGCs just examples of reprogramming escapes? Or is there a reason plants allow information about stress to be passed to the progeny?

When plants mount the defence against stress, they allocate resources from their growth and development programme to the response to stress. In this respect, the response to mild stress in the form of priming was developed as a mechanism to optimize the trade-offs of cost and benefit of higher tolerance to stress (Lopez Sanchez et al., Reference Lopez Sanchez, Pascual-Pardo, Furci, Roberts and Ton2021). The stressor may never appear again, and in this case, those plants that did not prime their defences have an advantage, as they have focused on growth and development instead of allocating resources to priming (Wilkinson et al., Reference Wilkinson, Mageroy, Lopez Sanchez, Smith, Furci, Cotton, Krokene and Ton2019). In contrast, those plants that mount priming will always be better off if stress is repeated during their growth or in the progeny. At the population level, some plants may receive more severe stress or be more genetically or epigenetically ‘primed’ to respond to stress with heritable change. It is even possible that there is a heterogeneous response within the same plant, where the level of response is gradual among all produced seeds. It would be interesting in the future to test this theory, focusing on the potential for the distance of dispersion of seeds to correlate with the degree of transgenerational response—the rate of changes may be proportional to the distance at which the seeds would land from their mothers.

The molecular mechanisms of somatic and transgenerational response have likely been established through thousands of generations of trial and error. There were likely cases when the cost of establishment of priming paid off because the stress repeated itself, and those plants that utilized it survived better and passed the genetic or epigenetic regulatory mechanisms to the progeny. Many theoretical papers were published attempting to correlate the response in the form of maternal effects (IGC or TGC) and changes in phenotypic plasticity with stress severity or intensity. It is proposed that maternal effects correlate with a periodicity of stress exposure. In a stable environment, maternal effects may have a slight negative influence on phenotypic plasticity, while in an abruptly changing environment that is maintained at a more or less constant level, maternal effects would have a strong positive influence allowing the progeny to adopt beneficial maternal phenotypes (Kuijper & Hoyle, Reference Kuijper and Hoyle2015). In contrast, when there are fluctuations in the presence or severity of a stressor, maternal effects fluctuate or autocorrect according to the presence of a stressor (Figure 2a).

Figure 2. Correlation between environmental changes, maternal effects and adaptation. (a) Environmental stability regulates maternal effects. When environmental conditions are stable, maternal effects have a slight negative influence on selection or evolution and TGC. When environmental changes are rapid and stable, there is a positive maternal effect on TGC and trait diversity. Finally, when the environment fluctuates from stressful to normal, there is an equilibrium in maternal effects, changing from negative to positive. (b) TGC and adaptation or maladaptation are different for different traits. Traits under weak selection tend to respond less effectively to maternal effects and demonstrate lower TGC. In contrast, traits under higher selection pressure respond strongly to maternal changes; thus, the TGC and adaptation or maladaptation are easier to observe.

Generally, the strongest TGCs and maternal effects occur for those traits that are under very strong selective pressure, while for the traits that are under weak selective pressure, the evolutionary scope of maternal effects is very low or limited (Figure 2b). As it appears, the vast majority of traits are under weak selection; therefore, it is more problematic to observe transgenerational phenomena in nature; in contrast, it may be easier to establish IGC or TGC in the laboratory, if you identify the trait under strong selective pressure (Kuijper & Hoyle, Reference Kuijper and Hoyle2015).

At the end of the day, since priming as a response to stress has been demonstrated for many species, we assume that this mechanism is adaptive in nature. However, is transgenerational priming truly adaptive? We presented many examples where the progeny of primed plants had higher tolerance to the same stress and sometimes to a different stress. Very few reports, however, studied whether the fitness of such plants is comparable to the fitness of naïve plants when there is no encounter of stress in the progeny. Moreover, often great resistance to the stress encountered by parents results in lower resistance to another type of stress, and this is especially true for biotic stress encounters. There are several reports demonstrating the evidence of transgenerational maladaptation.

Repeated exposure to ozone-sensitized grapevine made them more sensitive in the progeny (Soja et al., Reference Soja, Eid, Gangl and Redl1997). Differential response to drought was found among closely related species, Polygonum persicaria and Polygonum hydropiper; while the progeny of the former one were more fit as compared to the control, the progeny of the latter one exhibited maladaptive traits—smaller seedlings with slower-growing roots (Sultan et al., Reference Sultan, Barton and Wilczek2009). The progeny of Arabidopsis plants exposed to spider mites were more resistant to infection with spider mites and even aphids but developed higher sensitivity to the biotrophic bacteria Pseudomonas syringae (Singh et al., Reference Singh, Dave, Vaistij, Worrall, Holroyd, Wells, Kaminski, Graham and Roberts2017). The progeny of Arabidopsis plants exposed to the biotrophic pathogen P. syringae was more tolerant to infection with the biotrophic pathogen, Hyaloperonospora arabidopsidis, while being more sensitive to the necrotrophic fungus Alternaria brassicicola (Luna et al., Reference Luna, Bruce, Roberts, Flors and Ton2012). Likewise, the progeny of Arabidopsis plants infected with biotrophic pathogen P. syringae or necrotrophic pathogen Plectosphaerella cucumerina or exposed to high salinity were more tolerant to the same pathogen but were more sensitive to a different pathogen—the progeny of plants exposed to the biotrophic pathogen were more sensitive to necrotrophic pathogen and vice versa; curiously, the progeny of salt-stressed plants did not acquire higher salt tolerance but was slightly more tolerant to both pathogens (Lopez Sanchez et al., Reference Lopez Sanchez, Pascual-Pardo, Furci, Roberts and Ton2021). Another potential problem is that invasive species may have greater benefits from transgenerational plasticity, as it allows them to retain fitness in nutrient-rich environments and outperform other species in nutrient-poor environments; this was demonstrated for two invasive species, Cyperus esculentus and Aegilops triuncialis (Dyer et al., Reference Dyer, Brown, Espeland, McKay, Meimberg and Rice2010).

5. Engineering plants with heritable epigenetic modifications

The knowledge we obtain from all inter- or transgenerational studies will allow us to understand how the memory of stress is formed and passed to the progeny. Information about loci that undergo epigenetic changes would allow us to engineer plants with higher stress tolerance.

Targeted epigenetic changes in the form of changes in DNA methylation and chromatin structure, leading to activation of multiple genes, have been demonstrated in plants; the dCas9-SunTag system fused to the VP64 transcriptional activator was used to target multiple loci for DNA demethylation; activation of FWA locus remained heritable for several generations (Papikian et al., Reference Papikian, Liu, Gallego-Bartolome and Jacobsen2019). More recently, Wang et al. used Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) or dCas9 in combination with the TEN-ELEVEN TRANSLOCATION1 (TET1) demethylation domain (Wang et al., Reference Wang, He, Chen, Wang, Wang, Zhou, Zhang, Cao, Zhang, Xie and Zhang2022) to target a naturally occurring hypermethylation epiallele (He et al., Reference He, Wu, Zinta, Yang, Wang, Liu, Zhang, Zheng, Huang, Zhang and Zhu2018) in one of the Arabidopsis ecotypes. They were able to achieve hypomethylation of the PPH gene, resulting in accelerated leaf senescence, inherited for two generations (Wang et al., Reference Wang, He, Chen, Wang, Wang, Zhou, Zhang, Cao, Zhang, Xie and Zhang2022). Tang et al. also used the SunTag-dCas9-TET1cd system to target the FIE1 gene (Tang et al., Reference Tang, Yang, Wang, Deng, Cao and Song2022); they found that the dwarf phenotype associated with hypomethylation of the FIE1 gene was inherited for two generations. In the cases described above, the targeted locus was a locus with naturally occurring variations in methylation status. It remains to be shown whether targeted heritable DNA methylation changes can also be achieved in the other loci. Inheritance of DNA methylation pattern and associated phenotypes have also been demonstrated in mice; two metabolism-related genes, the ankyrin repeat domain 26 and the low-density lipoprotein receptor, were targeted in embryonic stem cells, and the progeny with heritable obese phenotype was obtained (Takahashi et al., Reference Takahashi, Morales Valencia, Yu, Ouchi, Takahashi, Shokhirev, Lande, Williams, Fresia, Kurita, Hishida, Shojima, Hatanaka, Nunez-Delicado, Esteban and Izpisua Belmonte2023).

6. Concluding remarks

In this review, we discussed the hypothesis that TGCs are caused by the differential expression of ncRNAs and RdDM mechanisms causing differential changes in DNA methylation and possibly histone modifications that escape reprogramming and give advantage to the progeny of stressed plants. Direct links between differentially expressed siRNAs causing changes in DNA methylation at specific loci and changes in stress tolerance remain to be established. It is unclear whether such siRNAs are passed from the progeny via gametes, or their expression is induced in the early developing embryo or the germinated plants by some other unknown mechanisms. It is also possible that such siRNAs are propagated in the cytoplasm through some amplification mechanisms, or by avoiding degradation, rather than through the activation of transcription. It remains to be shown whether differentially regulated siRNAs are stress-specific, are indeed directed towards specific loci in the genome and promote specific changes at epigenetic levels.

What is known, however, is that the expression of some of the ncRNAs and their fragments is heritable. It is also known that changes in the methylation pattern in the progeny occur at various hotspots, relevant to the encountered stress; in addition, the repetitive elements are hypermethylated to stabilize the genome, while many loci associated with stress tolerance are hypomethylated, likely to allow them to respond to stress more efficiently. It was documented that in most cases, changes in methylation in the progeny of stressed plants occur at CHH, suggesting the role of RdDM, as de novo methylation in this context and the maintenance of this methylation are assisted by RdDM. The potential role of RdDM was further supported by showing that several mutants impaired in RdDM were impaired in heritable changes in response to stress.

Despite the huge amount of work done, there are still a number of questions remaining.

It is still unclear whether heritable events, especially TGC, represent a true plant adaptive mechanism, or they are just ‘imperfections’, that escape from reprogramming. If RdDM and siRNAs are involved in the establishment of transgenerational events, why do we see so few reports implementing specific siRNAs in changes in methylation and phenotype? Also, why the changes in DNA methylation are frequently very massive, but the changes in phenotype are very subtle? All these questions remain to be answered by well-planned and carefully executed experiments.

Finally, we would like to apologize to all the scientists whose work we were not able to cite in the review.

Acknowledgements

The author thanks the reviewers for making this review better.

Financial support

This study was funded by the Natural Sciences and Engineering Research Council of Canada Discovery Grant RGPIN-2023-03267.

Competing interest

The author declares none.

Data availability statement

There are no data to share.

References

Agrawal, A. A. (2001). Transgenerational consequences of plant responses to herbivory: An adaptive maternal effect? The American Naturalist, 157, 555569. https://doi.org/10.1086/319932 CrossRefGoogle ScholarPubMed
Alonso, C., Perez, R., Bazaga, P., Medrano, M., & Herrera, C. M. (2014). Individual variation in size and fecundity is correlated with differences in global DNA cytosine methylation in the perennial herb Helleborus foetidus (Ranunculaceae). American Journal of Botany, 101, 13091313. https://doi.org/10.3732/ajb.1400126 CrossRefGoogle ScholarPubMed
Bezemer, T. M., Thompson, L. J., & Jones, T. H. (2004). Poa annua shows inter-generational differences in response to elevated CO sub(2). Global Change Biology, 4, 687691. https://doi.org/10.1111/jipb.12901 CrossRefGoogle Scholar
Bilichak, A., Ilnystkyy, Y., Hollunder, J., & Kovalchuk, I. (2012). The progeny of Arabidopsis thaliana plants exposed to salt exhibit changes in DNA methylation, histone modifications and gene expression. PLoS One, 7, e30515. https://doi.org/10.1371/journal.pone.0030515 CrossRefGoogle ScholarPubMed
Borg, M., Papareddy, R. K., Dombey, R., Axelsson, E., Nodine, M. D., Twell, D., & Berger, F. (2021). Epigenetic reprogramming rewires transcription during the alternation of generations in Arabidopsis. eLife, 10, e61894. https://doi.org/10.7554/eLife.61894 CrossRefGoogle ScholarPubMed
Boyko, A., Blevins, T., Yao, Y., Golubov, A., Bilichak, A., Ilnytskyy, Y., Hollunder, J., Meins, F., & Kovalchuk, I. (2010). Transgenerational adaptation of Arabidopsis to stress requires DNA methylation and the function of dicer-like proteins. PLoS One, 5, e9514. https://doi.org/10.1371/journal.pone.0009514 CrossRefGoogle ScholarPubMed
Boyko, A., Kathiria, P., Zemp, F. J., Yao, Y., Pogribny, I., & Kovalchuk, I. (2007). Transgenerational changes in the genome stability and methylation in pathogen-infected plants: (virus-induced plant genome instability). Nucleic Acids Research, 35, 17141725. https://doi.org/10.1093/nar/gkm029 CrossRefGoogle ScholarPubMed
Boyko, A., & Kovalchuk, I. (2010). Transgenerational response to stress in Arabidopsis thaliana. Plant Signaling & Behavior, 5, 995998. https://doi.org/10.1371/journal.pone.0009514 CrossRefGoogle ScholarPubMed
Byeon, B., Bilichak, A., & Kovalchuk, I. (2019). Transgenerational response to heat stress in the form of differential expression of noncoding RNA fragments in Brassica rapa plants. Plant Genome, 12. https://doi.org/10.3835/plantgenome2018.04.0022 CrossRefGoogle ScholarPubMed
Chang, Y. N., Zhu, C., Jiang, J., Zhang, H., Zhu, J. K., & Duan, C. G. (2020). Epigenetic regulation in plant abiotic stress responses. Journal of Integrative Plant Biology, 62, 563580. https://doi.org/10.1111/jipb.12901 CrossRefGoogle ScholarPubMed
Colicchio, J. (2017). Transgenerational effects alter plant defence and resistance in nature. Journal of Evolutionary Biology, 30, 664680. https://doi.org/10.1111/jeb.13042 CrossRefGoogle ScholarPubMed
Cong, W., Miao, Y., Xu, L., Zhang, Y., Yuan, C., Wang, J., Zhuang, T., Lin, X., Jiang, L., Wang, N., Ma, J., Sanguinet, K. A., Liu, B., Rustgi, S., & Ou, X. (2019). Transgenerational memory of gene expression changes induced by heavy metal stress in rice (Oryza sativa L.). BMC Plant Biology, 19, 282. https://doi.org/10.1186/s12870-019-1887-7 CrossRefGoogle ScholarPubMed
Cubas, P., Vincent, C., & Coen, E. (1999). An epigenetic mutation responsible for natural variation in floral symmetry. Nature, 401, 157161. https://doi.org/10.1038/43657 CrossRefGoogle ScholarPubMed
Ding, Y., Shi, Y., & Yang, S. (2020). Molecular regulation of plant responses to environmental temperatures. Molecular Plant, 13, 544564. https://doi.org/10.1016/j.molp.2020.02.004 CrossRefGoogle ScholarPubMed
Donohue, K. (2009). Completing the cycle: Maternal effects as the missing link in plant life histories. Philosophical Transactions of the Royal Society of London. Series B, Biological Sciences, 364, 10591074. https://doi.org/10.1098/rstb.2008.0291 CrossRefGoogle ScholarPubMed
Du, J., Johnson, L. M., Jacobsen, S. E., & Patel, D. J. (2015). DNA methylation pathways and their crosstalk with histone methylation. Nature Reviews. Molecular Cell Biology, 16, 519532. https://doi.org/10.1038/nrm4043 CrossRefGoogle ScholarPubMed
Dyer, A. R., Brown, C. S., Espeland, E. K., McKay, J. K., Meimberg, H., & Rice, K. J. (2010). The role of adaptive trans-generational plasticity in biological invasions of plants. Evolutionary Applications, 3, 179192. https://doi.org/10.1111/j.1752-4571.2010.00118.x CrossRefGoogle ScholarPubMed
Fujimoto, R., Kinoshita, Y., Kawabe, A., Kinoshita, T., Takashima, K., Nordborg, M., Nasrallah, M. E., Shimizu, K. K., Kudoh, H., & Kakutani, T. (2008). Evolution and control of imprinted FWA genes in the genus Arabidopsis. PLoS Genetics, 4, e1000048. https://doi.org/10.1371/journal.pgen.1000048 CrossRefGoogle ScholarPubMed
Furci, L., Jain, R., Stassen, J., Berkowitz, O., Whelan, J., Roquis, D., Baillet, V., Colot, V., Johannes, F., & Ton, J. (2019). Identification and characterisation of hypomethylated DNA loci controlling quantitative resistance in Arabidopsis. Elife, 8, e40655. https://doi.org/10.7554/eLife.40655 CrossRefGoogle ScholarPubMed
Ganguly, D. R., Crisp, P. A., Eichten, S. R., & Pogson, B. J. (2017). The Arabidopsis DNA Methylome is stable under transgenerational drought stress. Plant Physiology, 175, 18931912. https://doi.org/10.1104/pp.17.00744 CrossRefGoogle ScholarPubMed
Hatzig, S. V., Nuppenau, J. N., Snowdon, R. J., & Schiessl, S. V. (2018). Drought stress has transgenerational effects on seeds and seedlings in winter oilseed rape (Brassica napus L.). BMC Plant Biology, 18, 297. https://doi.org/10.1186/s12870-018-1531-y CrossRefGoogle ScholarPubMed
He, L., Wu, W., Zinta, G., Yang, L., Wang, D., Liu, R., Zhang, H., Zheng, Z., Huang, H., Zhang, Q., & Zhu, J. K. (2018). A naturally occurring epiallele associates with leaf senescence and local climate adaptation in Arabidopsis accessions. Nature Communications, 9, 460. https://doi.org/10.1038/s41467-018-02839-3 CrossRefGoogle ScholarPubMed
Heard, E., & Martienssen, R. A. (2014). Transgenerational epigenetic inheritance: Myths and mechanisms. Cell, 157, 95109. https://doi.org/10.1016/j.cell.2014.02.045 CrossRefGoogle ScholarPubMed
Herman, J. J., & Sultan, S. E. (2011). Adaptive transgenerational plasticity in plants: Case studies, mechanisms, and implications for natural populations. Frontiers in Plant Science, 2, 102. https://doi.org/10.3389/fpls.2011.00102 CrossRefGoogle ScholarPubMed
Herman, J. J., Sultan, S. E., Horgan-Kobelski, T., & Riggs, C. (2012). Adaptive transgenerational plasticity in an annual plant: Grandparental and parental drought stress enhance performance of seedlings in dry soil. Integrative and Comparative Biology, 52, 7788. https://doi.org/10.1093/icb/ics041 CrossRefGoogle Scholar
Holeski, L. M., Chase-Alone, R., & Kelly, J. K. (2010). The genetics of phenotypic plasticity in plant defense: Trichome production in Mimulus guttatus. The American Naturalist, 175, 391400. https://doi.org/10.1086/651300 CrossRefGoogle ScholarPubMed
Ito, H., Gaubert, H., Bucher, E., Mirouze, M., Vaillant, I., & Paszkowski, J. (2011). An siRNA pathway prevents transgenerational retrotransposition in plants subjected to stress. Nature, 472, 115119. https://doi.org/10.1038/nature09861 CrossRefGoogle ScholarPubMed
Kathiria, P., Sidler, C., Golubov, A., Kalischuk, M., Kawchuk, L. M., & Kovalchuk, I. (2010). Tobacco mosaic virus infection results in an increase in recombination frequency and resistance to viral, bacterial, and fungal pathogens in the progeny of infected tobacco plants. Plant Physiology, 153, 18591870. https://doi.org/10.1104/pp.110.157263 CrossRefGoogle Scholar
Khouider, S., Borges, F., LeBlanc, C., Ungru, A., Schnittger, A., Martienssen, R., Colot, V., & Bouyer, D. (2021). Male fertility in Arabidopsis requires active DNA demethylation of genes that control pollen tube function. Nature Communications, 12, 410. https://doi.org/10.1038/s41467-020-20606-1 CrossRefGoogle ScholarPubMed
Kinoshita, T., & Seki, M. (2014). Epigenetic memory for stress response and adaptation in plants. Plant & Cell Physiology, 55, 18591863. https://doi.org/10.1093/pcp/pcu125 CrossRefGoogle ScholarPubMed
Klironomos, J. N., Allen, M. F., Rillig, M. C., Piotrowski, J., Makvandi-Nejad, S., Wolfe, B. E., & Powell, J. R. (2005). Abrupt rise in atmospheric CO2 overestimates community response in a model plant-soil system. Nature, 433, 621624. https://doi.org/10.1038/nature03268 CrossRefGoogle Scholar
Kovalchuk, O., Burke, P., Arkhipov, A., Kuchma, N., James, S. J., Kovalchuk, I., & Pogribny, I. (2003). Genome hypermethylation in Pinus silvestris of Chernobyl--a mechanism for radiation adaptation? Mutation Research, 529, 1320. https://doi.org/10.1016/s0027-5107(03)00103-9 CrossRefGoogle ScholarPubMed
Kuijper, B., & Hoyle, R. B. (2015). When to rely on maternal effects and when on phenotypic plasticity? Evolution, 69, 950968. https://doi.org/10.1111/evo.12635 CrossRefGoogle ScholarPubMed
Laanen, P., Saenen, E., Mysara, M., Van de Walle, J., Van Hees, M., Nauts, R., Van Nieuwerburgh, F., Voorspoels, S., Jacobs, G., Cuypers, A., & Horemans, N. (2021). Changes in DNA methylation in Arabidopsis thaliana plants exposed over multiple generations to gamma radiation. Frontiers in Plant Science, 12, 611783. https://doi.org/10.3389/fpls.2021.611783 CrossRefGoogle ScholarPubMed
Lamke, J., & Baurle, I. (2017). Epigenetic and chromatin-based mechanisms in environmental stress adaptation and stress memory in plants. Genome Biology, 18, 124. https://doi.org/10.1186/s13059-017-1263-6 CrossRefGoogle ScholarPubMed
Lau, J. A., Peiffer, J., Reich, P. B., & Tiffin, P. (2008). Transgenerational effects of global environmental change: Long-term CO(2) and nitrogen treatments influence offspring growth response to elevated CO(2). Oecologia, 158, 141150. https://doi.org/10.1007/s00442-008-1127-6 CrossRefGoogle Scholar
Law, J. A., & Jacobsen, S. E. (2010). Establishing, maintaining and modifying DNA methylation patterns in plants and animals. Nature Reviews. Genetics, 11, 204220. https://doi.org/10.1038/nrg2719 CrossRefGoogle ScholarPubMed
Li, Y., Li, X., & Yu, J. (2017). Effect of the transgenerational exposure to elevated CO2 on the drought response of winter wheat: Stomatal control and water use efficiency. Environmental and Experimental Botany, 136, 7884. https://doi.org/10.1111/jipb.12901 CrossRefGoogle Scholar
Liu, J., Feng, L., Gu, X., Deng, X., Qiu, Q., Li, Q., Zhang, Y., Wang, M., Deng, Y., Wang, E., He, Y., Baurle, I., Li, J., Cao, X., & He, Z. (2019). An H3K27me3 demethylase-HSFA2 regulatory loop orchestrates transgenerational thermomemory in Arabidopsis. Cell Research, 29, 379390. https://doi.org/10.1038/s41422-019-0145-8 CrossRefGoogle ScholarPubMed
Lopez Sanchez, A., Pascual-Pardo, D., Furci, L., Roberts, M. R., & Ton, J. (2021). Costs and benefits of transgenerational induced resistance in Arabidopsis. Frontiers in Plant Science, 12, 644999. https://doi.org/10.3389/fpls.2021.644999 CrossRefGoogle ScholarPubMed
Luna, E., Bruce, T. J., Roberts, M. R., Flors, V., & Ton, J. (2012). Next-generation systemic acquired resistance. Plant Physiology, 158, 844853. https://doi.org/10.1104/pp.111.187468 CrossRefGoogle ScholarPubMed
Lv, C., Hu, Z., Wei, J., & Wang, Y. (2022). Transgenerational effects of elevated CO(2) on rice photosynthesis and grain yield. Plant Molecular Biology, 110, 413424. https://doi.org/10.1007/s11103-022-01294-5 CrossRefGoogle ScholarPubMed
Maizel, A., Markmann, K., Timmermans, M., & Wachter, A. (2020). To move or not to move: Roles and specificity of plant RNA mobility. Current Opinion in Plant Biology, 57, 5260. https://doi.org/10.1016/j.pbi.2020.05.005 CrossRefGoogle ScholarPubMed
Manning, K., Tor, M., Poole, M., Hong, Y., Thompson, A. J., King, G. J., Giovannoni, J. J., & Seymour, G. B. (2006). A naturally occurring epigenetic mutation in a gene encoding an SBP-box transcription factor inhibits tomato fruit ripening. Nature Genetics, 38, 948952. https://doi.org/10.1038/ng1841 CrossRefGoogle Scholar
Martinez, G., & Kohler, C. (2017). Role of small RNAs in epigenetic reprogramming during plant sexual reproduction. Current Opinion in Plant Biology, 36, 2228. https://doi.org/10.1016/j.pbi.2016.12.006 CrossRefGoogle ScholarPubMed
Matzke, M. A., & Mosher, R. A. (2014). RNA-directed DNA methylation: An epigenetic pathway of increasing complexity. Nature Reviews. Genetics, 15, 394408. https://doi.org/10.1038/nrg3683 CrossRefGoogle ScholarPubMed
Migicovsky, Z., & Kovalchuk, I. (2014). Transgenerational changes in plant physiology and in transposon expression in response to UV-C stress in Arabidopsis thaliana. Plant Signaling & Behavior, 9, e976490. https://doi.org/10.4161/15592324.2014.976490 CrossRefGoogle ScholarPubMed
Migicovsky, Z., Yao, Y., & Kovalchuk, I. (2014). Transgenerational phenotypic and epigenetic changes in response to heat stress in Arabidopsis thaliana. Plant Signaling & Behavior, 9, e27971. https://doi.org/10.4161/psb.27971 CrossRefGoogle ScholarPubMed
Mosher, R. A., Melnyk, C. W., Kelly, K. A., Dunn, R. M., Studholme, D. J., & Baulcombe, D. C. (2009). Uniparental expression of PolIV-dependent siRNAs in developing endosperm of Arabidopsis. Nature, 460, 283286. https://doi.org/10.1038/nature08084 CrossRefGoogle ScholarPubMed
Mozgova, I., Wildhaber, T., Trejo-Arellano, M. S., Fajkus, J., Roszak, P., Kohler, C., & Hennig, L. (2018). Transgenerational phenotype aggravation in CAF-1 mutants reveals parent-of-origin specific epigenetic inheritance. The New Phytologist, 220, 908921. https://doi.org/10.1111/nph.15082 CrossRefGoogle ScholarPubMed
Nguyen, N. H., Vu, N. T., & Cheong, J. J. (2022). Transcriptional stress memory and transgenerational inheritance of drought tolerance in plants. International Journal of Molecular Sciences, 23, 12918. https://doi.org/10.3390/ijms232112918 CrossRefGoogle ScholarPubMed
Nihranz, C. T., Walker, W. S., Brown, S. J., Mescher, M. C., De Moraes, C. M., & Stephenson, A. G. (2020). Transgenerational impacts of herbivory and inbreeding on reproductive output in Solanum carolinense. American Journal of Botany, 107, 286297. https://doi.org/10.1002/ajb2.1402 CrossRefGoogle ScholarPubMed
Nuthikattu, S., McCue, A. D., Panda, K., Fultz, D., DeFraia, C., Thomas, E. N., & Slotkin, R. K. (2013). The initiation of epigenetic silencing of active transposable elements is triggered by RDR6 and 21-22 nucleotide small interfering RNAs. Plant Physiology, 162, 116131. https://doi.org/10.1104/pp.113.216481 CrossRefGoogle ScholarPubMed
Ou, X., Zhang, Y., Xu, C., Lin, X., Zang, Q., Zhuang, T., Jiang, L., von Wettstein, D., & Liu, B. (2012). Transgenerational inheritance of modified DNA methylation patterns and enhanced tolerance induced by heavy metal stress in rice (Oryza sativa L.). PLoS One, 7, e41143. https://doi.org/10.1371/journal.pone.0041143 CrossRefGoogle ScholarPubMed
Panda, K., Mohanasundaram, B., Gutierrez, J., McLain, L., Castillo, S. E., Sheng, H., Casto, A., Gratacos, G., Chakrabarti, A., Fahlgren, N., Pandey, S., Gehan, M. A., & Slotkin, R. K. (2023). The plant response to high CO(2) levels is heritable and orchestrated by DNA methylation. The New Phytologist, 238(6), 24272439. https://doi.org/10.1111/nph.18876 CrossRefGoogle ScholarPubMed
Papikian, A., Liu, W., Gallego-Bartolome, J., & Jacobsen, S. E. (2019). Site-specific manipulation of Arabidopsis loci using CRISPR-Cas9 SunTag systems. Nature Communications, 10, 729. https://doi.org/10.1038/s41467-019-08736-7 CrossRefGoogle ScholarPubMed
Pecinka, A., & Mittelsten Scheid, O. (2012). Stress-induced chromatin changes: A critical view on their heritability. Plant & Cell Physiology, 53, 801808. https://doi.org/10.1093/pcp/pcs044 CrossRefGoogle ScholarPubMed
Quadrana, L., & Colot, V. (2016). Plant transgenerational epigenetics. Annual Review of Genetics, 50, 467491. https://doi.org/10.1146/annurev-genet-120215-035254 CrossRefGoogle ScholarPubMed
Rasmann, S., De Vos, M., Casteel, C. L., Tian, D., Halitschke, R., Sun, J. Y., Agrawal, A. A., Felton, G. W., & Jander, G. (2012). Herbivory in the previous generation primes plants for enhanced insect resistance. Plant Physiology, 158, 854863. https://doi.org/10.1104/pp.111.187831 CrossRefGoogle ScholarPubMed
Silveira, A. B., Trontin, C., Cortijo, S., Barau, J., Del Bem, L. E., Loudet, O., Colot, V., & Vincentz, M. (2013). Extensive natural epigenetic variation at a de novo originated gene. PLoS Genetics, 9, e1003437. https://doi.org/10.1371/journal.pgen.1003437 CrossRefGoogle Scholar
Singh, P., Dave, A., Vaistij, F. E., Worrall, D., Holroyd, G. H., Wells, J. G., Kaminski, F., Graham, I. A., & Roberts, M. R. (2017). Jasmonic acid-dependent regulation of seed dormancy following maternal herbivory in Arabidopsis. The New Phytologist, 214, 17021711. https://doi.org/10.1111/nph.14525 CrossRefGoogle ScholarPubMed
Slaughter, A., Daniel, X., Flors, V., Luna, E., Hohn, B., & Mauch-Mani, B. (2012). Descendants of primed Arabidopsis plants exhibit resistance to biotic stress. Plant Physiology, 158, 835843. https://doi.org/10.1104/pp.111.191593 CrossRefGoogle ScholarPubMed
Soja, G., Eid, M., Gangl, H., & Redl, H. (1997). Ozone sensitivity of grapevine (Vitis vinifera L.): Evidence for a memory effect in a perennial crop plant? Physical Chemistry Chemical Physics, 37, 265270. https://doi.org/10.1016/j.molimm.2021.08.015 Google Scholar
Stroud, H., Do, T., Du, J., Zhong, X., Feng, S., Johnson, L., Patel, D. J., & Jacobsen, S. E. (2014). Non-CG methylation patterns shape the epigenetic landscape in Arabidopsis. Nature Structural & Molecular Biology, 21, 6472. https://doi.org/10.1038/nsmb.2735 CrossRefGoogle ScholarPubMed
Sultan, S. E., Barton, K., & Wilczek, A. M. (2009). Contrasting patterns of transgenerational plasticity in ecologically distinct congeners. Ecology, 90, 18311839. https://doi.org/10.1890/08-1064.1 CrossRefGoogle ScholarPubMed
Takahashi, Y., Morales Valencia, M., Yu, Y., Ouchi, Y., Takahashi, K., Shokhirev, M. N., Lande, K., Williams, A. E., Fresia, C., Kurita, M., Hishida, T., Shojima, K., Hatanaka, F., Nunez-Delicado, E., Esteban, C. R., & Izpisua Belmonte, J. C. (2023). Transgenerational inheritance of acquired epigenetic signatures at CpG islands in mice. Cell, 186(4), 715731.e19. https://doi.org/10.1016/j.cell.2022.12.047 CrossRefGoogle ScholarPubMed
Tang, S., Yang, C., Wang, D., Deng, X., Cao, X., & Song, X. (2022). Targeted DNA demethylation produces heritable epialleles in rice. Science China. Life Sciences, 65, 753756. https://doi.org/10.1007/s11427-021-1974-7 CrossRefGoogle ScholarPubMed
Tao, Z., Shen, L., Gu, X., Wang, Y., Yu, H., & He, Y. (2017). Embryonic epigenetic reprogramming by a pioneer transcription factor in plants. Nature, 551, 124128. https://doi.org/10.1038/nature24300 CrossRefGoogle ScholarPubMed
Tonosaki, K., Fujimoto, R., Dennis, E. S., Raboy, V., & Osabe, K. (2022). Will epigenetics be a key player in crop breeding? Frontiers in Plant Science, 13, 958350. https://doi.org/10.3389/fpls.2022.958350 CrossRefGoogle ScholarPubMed
van der Graaf, A., Wardenaar, R., Neumann, D. A., Taudt, A., Shaw, R. G., Jansen, R. C., Schmitz, R. J., Colome-Tatche, M., & Johannes, F. (2015). Rate, spectrum, and evolutionary dynamics of spontaneous epimutations. Proceedings of the National Academy of Sciences of the United States of America, 112, 66766681. https://doi.org/10.1073/pnas.1424254112 CrossRefGoogle ScholarPubMed
Verhoeven, K. J. F., Verbon, E. H., van Gurp, T. P., Oplaat, C., Ferreira de Carvalho, J., Morse, A. M., Stahl, M., Macel, M., & McIntyre, L. M. (2018). Intergenerational environmental effects: Functional signals in offspring transcriptomes and metabolomes after parental jasmonic acid treatment in apomictic dandelion. The New Phytologist, 217, 871882. https://doi.org/10.1111/nph.14835 CrossRefGoogle ScholarPubMed
Wang, M., He, L., Chen, B., Wang, Y., Wang, L., Zhou, W., Zhang, T., Cao, L., Zhang, P., Xie, L., & Zhang, Q. (2022). Transgenerationally transmitted DNA demethylation of a spontaneous Epialleles using CRISPR/dCas9-TET1cd targeted epigenetic editing in Arabidopsis. International Journal of Molecular Sciences, 23, 10492. https://doi.org/10.3390/ijms231810492 CrossRefGoogle ScholarPubMed
Wibowo, A., Becker, C., Marconi, G., Durr, J., Price, J., Hagmann, J., Papareddy, R., Putra, H., Kageyama, J., Becker, J., Weigel, D., & Gutierrez-Marcos, J. (2016). Hyperosmotic stress memory in Arabidopsis is mediated by distinct epigenetically labile sites in the genome and is restricted in the male germline by DNA glycosylase activity. eLife, 5, 13546. https://doi.org/10.7554/eLife.13546 CrossRefGoogle ScholarPubMed
Wilkinson, S. W., Mageroy, M. H., Lopez Sanchez, A., Smith, L. M., Furci, L., Cotton, T. E. A., Krokene, P., & Ton, J. (2019). Surviving in a hostile world: Plant strategies to resist pests and diseases. Annual Review of Phytopathology, 57, 505529. https://doi.org/10.1146/annurev-phyto-082718-095959 CrossRefGoogle Scholar
Williams, B. P., Bechen, L. L., Pohlmann, D. A., & Gehring, M. (2022). Somatic DNA demethylation generates tissue-specific methylation states and impacts flowering time. Plant Cell, 34, 11891206. https://doi.org/10.1093/plcell/koab319 CrossRefGoogle ScholarPubMed
Yang, D. L., Zhang, G., Tang, K., Li, J., Yang, L., Huang, H., Zhang, H., & Zhu, J. K. (2016). Dicer-independent RNA-directed DNA methylation in Arabidopsis. Cell Research, 26, 1264. https://doi.org/10.1038/cr.2016.122 CrossRefGoogle ScholarPubMed
Yang, H., Cui, Y., Feng, Y., Hu, Y., Liu, L., & Duan, L. (2023). Long non-coding RNAs of plants in response to abiotic stresses and their regulating roles in promoting environmental adaption. Cell, 12, 729. https://doi.org/10.3390/cells12050729 CrossRefGoogle ScholarPubMed
Zemach, A., Kim, M. Y., Hsieh, P. H., Coleman-Derr, D., Eshed-Williams, L., Thao, K., Harmer, S. L., & Zilberman, D. (2013). The Arabidopsis nucleosome remodeler DDM1 allows DNA methyltransferases to access H1-containing heterochromatin. Cell, 153, 193205. https://doi.org/10.1016/j.cell.2013.02.033 CrossRefGoogle ScholarPubMed
Zhang, H., Lang, Z., & Zhu, J. K. (2018). Dynamics and function of DNA methylation in plants. Nature Reviews. Molecular Cell Biology, 19, 489506. https://doi.org/10.1038/s41580-018-0016-z CrossRefGoogle ScholarPubMed
Zhang, L., Cheng, Z., Qin, R., Qiu, Y., Wang, J. L., Cui, X., Gu, L., Zhang, X., Guo, X., Wang, D., Jiang, L., Wu, C. Y., Wang, H., Cao, X., & Wan, J. (2012). Identification and characterization of an epi-allele of FIE1 reveals a regulatory linkage between two epigenetic marks in rice. Plant Cell, 24, 44074421. https://doi.org/10.1105/tpc.112.102269 CrossRefGoogle ScholarPubMed
Zheng, X., Chen, L., Li, M., Lou, Q., Xia, H., Wang, P., Li, T., Liu, H., & Luo, L. (2013). Transgenerational variations in DNA methylation induced by drought stress in two rice varieties with distinguished difference to drought resistance. PLoS One, 8, e80253. https://doi.org/10.1371/journal.pone.0080253 CrossRefGoogle ScholarPubMed
Zheng, X., Chen, L., Xia, H., Wei, H., Lou, Q., Li, M., Li, T., & Luo, L. (2017). Transgenerational epimutations induced by multi-generation drought imposition mediate rice plant’s adaptation to drought condition. Scientific Reports, 7, 39843. https://doi.org/10.1038/srep39843 CrossRefGoogle ScholarPubMed
Figure 0

Table 1 Naturally occurring epialleles.

Figure 1

Figure 1. Potential mechanism of establishment of transgenerational effects and development of new epialleles. In the proposed scenario, stress generates mobile response molecules, likely in the form of small interfering RNAs (siRNAs) or other types of ncRNAs, but could also include differential levels of proteins, metabolites and various histone modifications, which reach meiocytes and alter DNA methylation and gene expression patterns. Developing meiocytes may retain certain signals and pass new epigenetic patterns into gametes. It is possible that some of the differentially expressed ncRNAs, as well as mRNAs, especially from female gametes, are preserved and influence the developing progeny. The persistence of stress may further reinforce these signalling molecules, leading to the development of stable changes in DNA methylation that do not revert even when stress is absent. Such changes in DNA methylation and chromatin structure represent epimutations and could lead to the development of epialleles persisting for many generations.

Figure 2

Figure 2. Correlation between environmental changes, maternal effects and adaptation. (a) Environmental stability regulates maternal effects. When environmental conditions are stable, maternal effects have a slight negative influence on selection or evolution and TGC. When environmental changes are rapid and stable, there is a positive maternal effect on TGC and trait diversity. Finally, when the environment fluctuates from stressful to normal, there is an equilibrium in maternal effects, changing from negative to positive. (b) TGC and adaptation or maladaptation are different for different traits. Traits under weak selection tend to respond less effectively to maternal effects and demonstrate lower TGC. In contrast, traits under higher selection pressure respond strongly to maternal changes; thus, the TGC and adaptation or maladaptation are easier to observe.

Author comment: Heritable responses to stress in plants — R0/PR1

Comments

It is my pleasure to submit this review that provides a comprehensive coverage of somatic and heritable changes in the response to stress. We discuss evolutionary meaning of heritable changes and provide many examples of heritable response to stress. We covered details of epigenetic response to stress. The review is not under consideration anywhere and I declare no conflict of interest.

Sincerely,

Igor Kovalchuk

Review: Heritable responses to stress in plants — R0/PR2

Conflict of interest statement

"None"

Comments

The author reviews “heritable responses to stress in the plants”. The abstract and introduction clarify that the type of “heritable responses” the review analyzes are what the literature refers to as “IGC” and “TGC”. A hallmark of IGC and TGC are that they are transient and reversible responses that occur in a more less systematic fashion among plants in a population, rather than being random events affecting individual plants. In that sense, IGC and TGC are evolved acclimation responses to stress. An good example is “priming”, where exposure to stressor in one generation can confer more effective stress responses in the direct offspring generation and sometimes even in generations following the offspring generation.

I have several major comments about this review.

- First, the review does not deliver on the topic that is laid out in the abstract and the introduction. It spends a lot of time talking about the impact of stress on genome stability and the rate of HR, which are non-systematic events that lead to non-reversible genomic change, can can therefore not explain the reversible nature of IGC and TGC, nor the fact that the IGC and TGC are are systematically induced in population under stress.

- Second, I don’t understand the rationale for the structure of the review. It starts with possible molecular mechanisms, then moves to a description of the phenotypic examples of IGC and TGC and then returns to molecular considerations. In my view, the review could benefit tremendously from providing a clearer structure for the reader.

- Third, there are numerous reviews on IGC and TGC. The author needs to lay out in the beginning what makes his review unique. What unique aspect is he tackling? How is this different from existing reviews?

- Forth, the authors often confuses stable epigenetic changes that have been reported with transient ones.

In sum, in my assessment, the review requires a clearer focus, a more logical structure and a clear demarcation from existing reviews on that topic. The review needs to decide if it is about general, heritable, molecular changes in response to stress (which would include changes in mutation rates, HR etc.), or if it is about IGC and TGC as they are used in the literature.

Additionally:

- The author writes: “However, it should be noted that no experiment comparing the efficiency of response to environmental stimuli of genetic variants vs epigenetic variants was ever conducted, thus our assumption is strictly a hypothesis.” What type of experiment would that be?

- The author writes: “Such changes in DNA methylation and chromatin structure represent

epimutations and could lead to the development of epialleles if stabilized.” What does the author mean by “stabilize”, mechanistically?

- The section “2.2 IGCs/TGCs in the form of changes in plant physiology and stress response” should come first. Here the author establishes the phenotypic relevance / evidence of IGC and TGC. Considerations regarding molecular mechanisms should come later. Also, the second heading is a bit of a misnomer, as discussion is mainly about stress phenotypes or types of stressors rather than about physiology.

- The author writes “Furthermore, in this review, we will not cover such transgenerational events as gene silencing..”, but spends time describing experiments that show exactly that (e.g. the work on epiRILs).

- The whole section “2.3 IGCs/TGCs in the form of changes in the genome stability” refers to molecular consequences of stress that cannot explain the reversibility of IGC and TGC at the phenotypic level.

- What does the author mean by “inheritance of recombination events”?

- The author writes “Genome destabilization in the progeny is likely not liner”. “Liner”?

- The review could benefit from some English editing. The main concern here is the omission of “the” in many places.

Review: Heritable responses to stress in plants — R0/PR3

Conflict of interest statement

Reviewer declares none.

Comments

The manuscript “Heritable responses to stress in plants” by Kovalchuk reviews the recent literature about stress responses in plants that are inherited across generations, and this inheritance is often mediated by epigenetic mechanism. I think the topic of this review is interesting, that being said there are a reviews that have covered this topic in recent years. Moreover, from a more broad perspective epigenetic inheritance has been reviewed extensively over the past years.

This manuscript does a fair job of reviewing the recent literature specific to plants. However, there is a certain lack of focus that I would like to see corrected. I think currently the manuscript groups some phenomena that are separate and it would benefit either having clear distinctions these phenomena or dropping some of them altogether.

In section 2. the manuscript discusses epialleles that have been observed to occur in nature. However, these presumably spontaneously occurring epialleles (as in van der Graaf et al. 2015) are a different phenomenon from phenotypic plasticity that is inherited across generations (as in Wibowo et al. 2016). Currently these phenomena are not clearly enough separeted in the manuscript. But even inherited plastic responses are based on an underlying genetic program (utilizing the plant RdDM pathway for example as a response to specific stress) while some of these epialleles can be spontenous changes independent of DNA sequence.

The third phenomena that is discussed in section 2.3 is stress induced increases in mutation and recombination rates. Some research suggest that this increase in these rates is propagated into following generations even in the absence of the original stress. Again, this phenomena is quite different from phenotypic plasticity from an evolutionary perspective. Of course increased mutation or recombination rates can be beneficial for adaptation to a new environment.

I would suggest dropping the section about epialleles from this review and focusing on plastic responses and making a clear distinction between phenotypic plasticity and increases in mutation and recombination rates, as they have different evolutionary consequences.

I also think that section 4 does not review what is known about the evolutionary significance of across generation phenotypic plasticity very thoroughly. There has been a lot of theoretical work done to investigate in which conditions such plasticity should evolve and what are its evolutionary consequences, but none of this work is cited. Please consider citing some of this work. For example: the work of Bram Kuijper and others.

Kuijper, B. & Hoyle, R. B. 2015. When to rely on maternal effects and when on phenotypic plasticity? Evolution 69:, 950-968

There are also other theoretical papers that have investigated these issues.

I would also consider the evolutionary consequences of increased mutation and recombination rates separately, as they have different consequences on adaptation than phenotypic plasticity. Of course conditionally increased mutation and recombination rates can be adaptive.

Minor points

In the future, please include line numbers in the manuscript. This makes making comments easier.

The text has some points where terminology is needlessly complicated. The text speaks about immediate progeny (when discussing intergenerational inheritance) and then in the case of transgenerational inheritance speaks about the next generation (next generation of immediate progeny). It would be better to call them just progeny and grand progeny to distinguish between intergenerational and transgenerational.

Page 13, somewhat repetitive, these examples were discussed before and now are again the the perspective of the progeny.

Page 19, I think there is more evidence that maternal inheritance is more common. Check work from the lab of Sonia Sultan.

Herman, J. J.; Spencer, H. G.; Donohue, K. & Sultan, S. E. 2014. How stable ‘should’ epigenetic modifications be? Insights from adaptive plasticity and bet-hedging. Evolution 68: 632-643

Page 14, paragraph beginning “An interesting paper...” Wibowo is cited twice at the end of first sentence.

Same problem in page 22, when citing Elsalahy et al. 2020.

References

van der Graaf, A.; Wardenaar, R.; Neumann, D. A.; Taudt, A.; Shaw, R. G.; Jansen, R. C.; Schmitz, R. J.; Colomé-Tatché, M. & Johannes, F. 2015. Rate, spectrum, and evolutionary dynamics of spontaneous epimutations. Proceedings of the National Academy of Sciences, 112: 6676-6681

Wibowo, A.; Becker, C.; Marconi, G.; Durr, J.; Price, J.; Hagmann, J.; Papareddy, R.; Putra, H.; Kageyama, J.; Becker, J.; Weigel, D. & Gutierrez-Marcos, J. 2016. Hyperosmotic stress memory in Arabidopsis is mediated by distinct epigenetically labile sites in the genome and is restricted in the male germline by DNA glycosylase activity. eLife 5: e13546

Recommendation: Heritable responses to stress in plants — R0/PR4

Comments

Dear authors,

Both reviewers and myself acknowledge that the topic of your review is interesting and timely. However, the reviewers emit strong criticisms on the structure and content of the manuscript. Both reviewers suggest to have different focus and ideas for re-organizing (removing some parts), in order to make the review unique (and different from the large amount of published reviews on this topic). Please do address in a revised version fewer topics than intended which can then be covered in greater depth for the benefit of the readers. The reviewers also suggest to carefully edit the English writing and especially to be more precise in your definitions.

Best regards,

Aurelien Tellier

Decision: Heritable responses to stress in plants — R0/PR5

Comments

No accompanying comment.

Author comment: Heritable responses to stress in plants — R1/PR6

Comments

No accompanying comment.

Review: Heritable responses to stress in plants — R1/PR7

Conflict of interest statement

Reviewer declares none.

Comments

The manuscript has been revised and the structure is now more clear. Although I disagree with the choice to drop the genome instability part and keep the epiallele part, but fair enough.

The issue that there are a lot of recent reviews about this topic remains. While I have no scientific objections, I leave the decision whether the review is novel enough for the editor to decide.

My other points have been adressed in the revised manuscript.

Minor points (note that line numbers refer to the PDF proof.)

line 376: Is van der Graaf et al. 2015 cited correctly here? That paper is not about heritable responses to stress but abuot spontaneous epimutations.

Line 446 sentence should have the word “are”

Review: Heritable responses to stress in plants — R1/PR8

Conflict of interest statement

Reviewer declares none.

Comments

The author has substantially refocused and streamlined his manuscript based on the reviewer’s comments. I think this has improved the manuscript. Still, several conceptualizations / discussions throughout the work remain debatable (e.g. the distinction between stable epialleles and plasticity), and I occasionally do not share the author’s view on things. But the purpose of a review / opinion paper is to partly stimulate discussion, as it attempts to connect a diversity of empirical observations into a single perspective.

Recommendation: Heritable responses to stress in plants — R1/PR9

Comments

Dear authors,

Both reviewers are now pleased by your revised version. It reads much better and is more focused.

One reviewer asked for some last minor corrections, please update your last submitted version accordingly.

Congratulations and thank you for your contribution.

Aurelien Tellier

Decision: Heritable responses to stress in plants — R1/PR10

Comments

No accompanying comment.

Author comment: Heritable responses to stress in plants — R2/PR11

Comments

No accompanying comment.

Recommendation: Heritable responses to stress in plants — R2/PR12

Comments

No accompanying comment.

Decision: Heritable responses to stress in plants — R2/PR13

Comments

No accompanying comment.