Next Article in Journal
Systems Medicine Design for Triple-Negative Breast Cancer and Non-Triple-Negative Breast Cancer Based on Systems Identification and Carcinogenic Mechanisms
Next Article in Special Issue
Roles of Plant Growth-Promoting Rhizobacteria (PGPR) in Stimulating Salinity Stress Defense in Plants: A Review
Previous Article in Journal
The Role of CTLA4 and Its Polymorphisms in Solid Organ and Haematopoietic Stem Cell Transplantation
Previous Article in Special Issue
Advances in Sensing, Response and Regulation Mechanism of Salt Tolerance in Rice
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Jasmonates and Plant Salt Stress: Molecular Players, Physiological Effects, and Improving Tolerance by Using Genome-Associated Tools

1
Institute of Biological Sciences, Campus Talca, Universidad de Talca, Talca 3465548, Chile
2
Department of Life Sciences, College of Science, National University of Kaohsiung, Kaohsiung 811, Taiwan
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(6), 3082; https://doi.org/10.3390/ijms22063082
Submission received: 12 February 2021 / Revised: 11 March 2021 / Accepted: 15 March 2021 / Published: 17 March 2021
(This article belongs to the Special Issue Molecular Aspects of Plant Salinity Stress and Tolerance)

Abstract

:
Soil salinity is one of the most limiting stresses for crop productivity and quality worldwide. In this sense, jasmonates (JAs) have emerged as phytohormones that play essential roles in mediating plant response to abiotic stresses, including salt stress. Here, we reviewed the mechanisms underlying the activation and response of the JA-biosynthesis and JA-signaling pathways under saline conditions in Arabidopsis and several crops. In this sense, molecular components of JA-signaling such as MYC2 transcription factor and JASMONATE ZIM-DOMAIN (JAZ) repressors are key players for the JA-associated response. Moreover, we review the antagonist and synergistic effects between JA and other hormones such as abscisic acid (ABA). From an applied point of view, several reports have shown that exogenous JA applications increase the antioxidant response in plants to alleviate salt stress. Finally, we discuss the latest advances in genomic techniques for the improvement of crop tolerance to salt stress with a focus on jasmonates.

1. Introduction

Salinity is a serious hazard for agriculture since most of the crop plants are salt-sensitive [1]. Current data show that global soil salinization increased by more than 100 Mha between 1986 and 2016 and it is expanding on a global scale, approximately at a rate of 2 Mha per year [2]. Thus, the future of food supplies for animals and humankind is threatened [3]. Natural processes such as geological deposits due to parent rock constituents, salinized groundwater, marine transgressions, storm flood events, tsunamis, and recurrent drought events and the general increase in temperature [4,5] cause soil salinization. Unfortunately, human interventions have also promoted the increment of saline lands. Wrong irrigation practices, poor drainage conditions [6], the use of fertilizers [7], mismanagement of treated wastewater [8], industrial [9], and mining operation effluents enriched with a salt contribute to the salinity increment in the soil and water [10].
Under salt stress conditions, physiological and metabolic activities are impaired by osmotic-, ionic-, and oxidative stresses, nutritional imbalance, or a combination of these factors [11]. In fact, plant growth and development are limited by salt stress due to the negative influences through the ionic and osmotic components on various biochemical reactions and physiological processes such as photosynthesis, antioxidant metabolism, mineral nutrient homeostasis, osmolyte accumulation, and hormonal signaling [12]. Most species, including crops, activate tolerance mechanisms only after exposure to salt stress. Activation of the tolerance program drives plants to acclimatize under the saline condition and involves altered physiological responses, redirection of metabolism, reinforcement of defense and repair, and changes in developmental programs to adapt morphological and anatomical characteristics [1,13]. Complex coordination of several signaling pathways is needed to activate the plant responses to salt stress. Phytohormone-mediated signaling, for instance, is crucial in the induction of gene networks related to salt tolerance [14].
Among abiotic stress-related hormones, abscisic acid (ABA) is well known to be the key phytohormone in endogenous signaling that allows plants to survive adverse environmental conditions [15]. The role of ABA for salinity adaptation has long been intensively studied and documented [16,17,18]. More recently, the biological relevance of jasmonate (JA) and its derivatives in the induction of tolerance to abiotic stresses has been demonstrated [19,20,21]. JAs are critical signaling molecules in various development and defense processes of plants [22,23] and play essential roles in plant response to salt stress [24,25,26]. Similar to ABA, accumulation of JAs has been reported in salt-tolerant crops compared to sensitive cultivars [27]. Salt stress has been observed to cause increased levels of JA in leaves and roots and the induction of JA biosynthesis-related genes [28,29,30]. In addition, exogenous application of JA significantly reduces the Na+ ion content in salinity-tolerant rice and wheat [27,31] and recovers salt-induced defects in seedling development and photosynthetic activity [32,33]. All this evidence is highly correlated with a positive role of JA in the plant response to salt stress. JA-crosstalk with other phytohormones has aroused the interest of researchers since the interaction among multiple plant hormone signaling integrates environmental and development cues. In this sense, JA has been proposed as a core signal in the phytohormone signaling network [34] because it regulates the balance between plant growth and defense [35,36]. The JA signaling components JASMONATE ZIM-DOMAIN (JAZ) and MYC2 have been identified as the main nodes in the orchestration of JA interplay with other hormone signaling pathways [37,38]. However, the cross-coordination between JA and the other phytohormone is far from being completely understood.
Currently, many strategies have been implemented to obtain tolerant plants to salt stress including classical plant breeding and genetic engineering approaches [39]. However, the complexity of the salt tolerance trait requires more in-depth studies to be understood. Exploring germplasm that possesses genetic variability across a wide spectrum of salt tolerance-related traits could provide valuable information. The study of biparental mapping populations and diversity panels has allowed the discovery of beneficial genetic variants (or alleles) that can be examined for traits that have significant components of salt stress tolerance and their associated quantitative trait loci (QTL) [40]. The analysis of these traits by phenotyping and genotyping techniques could reveal new insights into the biological mechanisms underlying the salt tolerance phenotypes [41]. A widely employed approach to identify the association between each genotyped marker and a phenotype of interest that has been scored across several individuals is the genome-wide association study (GWAS) [42]. It offers some advantages such as more accurate positioning and mapping, simultaneous assessments of multiple alleles at a locus, no requirement for linkage group construction [43,44] and can serve as a basic experiment to identify candidates for mutagenesis and transgenics [45].
With the possibility of performance-specific and predictable genetic modifications through genome (or gene) editing (GE) tools, the information obtained by GWASs becomes more valuable for plant breeding and crop improvement efforts. Among GE methods, the CRISPR-Cas9 system has been used to enhance salt tolerance in some crops like rice and tomato [46,47]. Given that JA regulates processes associated with growth, development, and salt response, the components of JA-biosynthesis, metabolism, and signaling can be targets for phytohormone engineering to produce salt-resilient crops with high yields. Furthermore, increased knowledge of the signals and molecular mechanisms involved in the plant salinity response would pave the path to obtain salt-tolerant crops without productivity penalties.
In this review, we update the information of the role of JA in the salt stress response, mainly in the topics related to JA-crosstalk with other phytohormones in salinity conditions, although limited information about JA-crosstalk during salt stress is available, here we bring together the latest studies on this topic. Finally, we reviewed the use of genome-associated tools to improve salt tolerance by manipulation of JA signaling.

2. Jasmonate Metabolism, Signaling, and Response during Salt Stress

2.1. JA Biosynthesis, Signaling, and Catabolism

The JA hormones have traditionally been studied in several plant species in the regulation of aspects regarding development, metabolism, and adaptation to biotic stress. However, JAs have lastly been associated with plant responses to several abiotic stresses, adding to JAs extra functions in plant adaptation [20,48]. The JA pathway consists mainly of the JA-associated biosynthesis, signaling, catabolism, and response. According to the information obtained in Arabidopsis, JAs are oxylipins, which biosynthesis begins in the plastids. The α-linolenic acid fatty acid, produced by the action of A1-type lipases (PLIP and PLA1), is transformed into several intermediates by reactions catalyzed by 13-lipoxygenase (13-LOX), allene oxide synthase (AOS), and allene oxide cyclase (AOC), leading to 12-oxo-phytodienoic acid (OPDA) [49,50]. OPDA is translocated to the peroxisome where 12-oxo-phytodienoate reductase 3 (OPR3) and several β-oxidation cycles give rise to jasmonic acid (JA) [51]. The biosynthesis of methyl jasmonate (MeJA) and jasmonoyl-isoleucine (JA-Ile) from JA is catalyzed by jasmonic acid methyltransferase (JMT) [52] and jasmonic acid-amide synthetase 1 (JAR1) [53], respectively. Meanwhile, the reconversion of MeJA and JA-Ile to JA is catalyzed by methyl jasmonate esterase (MJE) [54] and JA-Ile hydrolase 1 (JIH1) [55], respectively.
The JA-Ile molecule is the bioactive JA responsible for the activation of JA responses [56]. The physiological effects mediated by JA-Ile require activation of the JA signaling pathway, which has been well characterized in Arabidopsis [57]. The F-box CORONATINE INSENSITIVE1 protein (COI1) is part of the Skp-Cullin-F-box-type E3 ubiquitin ligase complex (SCFCOI1) and together with JAZ proteins form the JA-Ile receptor [58,59,60,61,62]. In Arabidopsis, when JA-Ile level is low, JAZ proteins repress MYC transcriptional activity by recruiting NOVEL INTERACTOR OF JAZ (NINJA) and TOPLESS corepressors [63,64] and by obstructing the association of the coactivator protein mediator complex subunit 25 (MED25) with the transcription initiation complex [65,66,67]. However, once the JA-Ile level rises, it mediates the COI1-JAZ interaction leading to JAZ proteins ubiquitination and degradation by the 26S proteasome. Then, MYC2 and additional transcription factors (TFs) induce the expression of early JA-responsive genes such as JAZs, MYCs, and JA biosynthetic ones [58,68]. MYC2 is a key transcription factor in the JA pathway and acts as a regulatory hub for several biotic and abiotic stress-related responses. Its homologs, MYC3, MYC4, and MYC5, also act alternatively in the JA pathway in Arabidopsis [57,69,70]. MYC2 contains a conserved basic helix-loop-helix (bHLH) domain [71], which is required to form homo- or heterodimers with MYC3 and MYC4 [69], although the strongest activation of JA-regulated genes is provided by the MYC2 tetramer [72]. The basic region of MYC2 protein is involved in binding to the sequence 5′-CACGTG-3′, known as G-box, which is present in the MYC2 target promoters [73]. The G-box contains many other variants and weaker binding sequences [74,75]. Furthermore, MYC2 N-terminal contains a putative transcriptional activation domain (TAD) [69], and a JAZ interaction domain (JID) through which interacts with the C-terminal Jas domain of JAZ proteins [58,69]. In several JA-dependent functions, MYC2 regulates many secondary TFs by binding to their promoters, which, in turn, activate downstream gene promoters, creating a hierarchical transcriptional network of JA-mediated response [76,77,78].
Recently, an OPR3-independent pathway for JA synthesis has been described. Chini et al. [79] isolated and characterized the complete knockout mutant of the opr3-3 allele. Similar to wild-type (WT) plants, the opr3-3 mutants were resistant to necrotrophic pathogens and insect feeding and activated COI1-dependent JA-mediated gene expression. Through OPDA derivatives analysis the 4,5-didehydro-JA (4,5-ddh-JA) was identified to act as a precursor for JA and JA-Ile biosynthesis in OPR3 absence. The authors demonstrated that in the lack of OPR3, OPDA could enter the peroxisomal β-oxidation pathway to produce 4,5-ddh-JA, which after leaving the peroxisome, is reduced by the cytosolic 12-oxo-phytodienoate reductase 2 (OPR2). This pathway takes place naturally in WT plants and is maximized in the opr3 mutant [79].
Together with JA biosynthetic and signaling genes, the JA-Ile catabolic genes are strongly coregulated indicating the importance to maintain JA homeostasis [80]. There are two JA-catabolic pathways: one is defined by CYP94B3/B1 and CYP94C1, members of the cytochrome P450 enzymes of the subfamily 94 (CYP94) [81,82,83], and the other one involves the IAA-alanine resistant 3 (IAR3) and IAA-amino acid hydrolase ILR1-like 6 (ILL6) enzymes, members of the amidohydrolase (AH) family [84]. CYP94B3 and CYP94B1 are JA-Ile ω-hydroxylases that generate 12OH-JA-Ile, and CYP94C1 catalyzes a more complete JA-Ile oxidation to 12COOH-JA-Ile [82]. Meanwhile, IAR3 and ILL6 prompt JA-Ile to deactivate by deconjugation reactions [55]. All these enzymes diminish specifically JA-Ile hormone pools to weaken JA signaling as many loss and gain-of-function experiments have demonstrated [85]. The proper regulation and termination of JA-mediated processes are essential to avoid the harmful metabolic effects of a JA amplified response trigger by biotic and abiotic stress. However, it is necessary to highlight that lately, a role of 12OH-JA-Ile in JA signaling has been described [86,87]. Exogenous application of 12OH-JA-Ile mimicked several JA-Ile effects including JA-marker gene expression, anthocyanin accumulation, and trichome induction in Arabidopsis [86]. In silico and in vitro assays showed that 12OH-JA-Ile could interact with some COI1-JAZs coreceptors and function as an active jasmonate signal, but more weakly comparing with JA-Ile. It has been proposed that after a strong immune response mediated by JA-Ile, 12OH-JA-Ile modulates JA-Ile activated processes contributing to wound and defense plant response [87].
In Figure 1, a schematic diagram of the biosynthesis, signaling, and catabolism of JAs is presented.

2.2. Salt Stress and JA Response

Earlier reports have shown the induction of some JA biosynthetic genes in Arabidopsis roots under salt stress conditions [88,89]. In sweet potato, Zhang et al. [90] studied the root transcriptomes of a salt-sensitive variety and a salt-tolerant line revealing a significant upregulation of the genes involved in the JA biosynthesis and signaling pathways under salt stress. The upregulation in the salt-tolerant line was greater than the sensitive line indicating the essential role of JA in the response of sweet potato to salt stress [90]. The Arabidopsis lipoxygenase3 (LOX3) was dramatically induced under salt treatment and the lox3 mutant exhibited salt hypersensitivity. The lox3 mutant salt sensitivity phenotype was rescued by the MeJA application indicating the association between JA and salt tolerance [91]. The TaAOC1 gene from bread wheat responds to salinity and its constitutive expression in both bread wheat and Arabidopsis enhanced their level of tolerance to salt stress [92]. On the contrary, the impaired function of AOC in the OPDA-deficient rice ALLENE OXIDE CYCLASE mutants (cpm2 and hebiba) conferred salt tolerance [93]. It is not clear what causes this tolerance, the lack of JA or JA-Ile, or the absence of their precursor 12-OPDA [19].
Several studies in Arabidopsis have demonstrated the essential role of MYC2 in salt response mediated by JA signaling [94,95]. The salt and ABA inducible gene responsive to dehydration 22 (RD22) is regulated by MYC2. The RD22 promoter region contains the MYC and MYB recognition sites [96] and the AtMYC2 and AtMYB2 TFs specifically interact with them, respectively. The atmyc2 mutant and MYC2 overexpressing (OE) plants treated with ABA showed different RD22 expression levels, the latter increased the RD22 expression at low ABA concentration (500 nM) while the former increased RD22 expression at higher ABA concentration (1 µM) [94]. MYC2 also has an important role in the activation of JA signaling by salt stress on the inhibition of cell elongation in Arabidopsis primary roots [95]. Additionally, the salt stress-mediated activation of MYC2 by the MAPK cascade regulates the proline biosynthesis through the delta1-pyrroline-5-carboxylate synthase1 (P5CS1) gene, which is a rate-limiting enzyme in the proline biosynthesis pathway [97]. Moreover, Seo et al. [98] demonstrated that the E3 ubiquitin ligase PLANT U-BOX PROTEIN 10 (PUB10), which regulates MYC2 stability [99], positively regulates salt and osmotic stress tolerance during seed germination. It was suggested that PUB10 acts as a negative regulator of ABA signaling through MYC2 and participates in the fine-tuning of ABA signaling and JA crosstalk in the abiotic stress tolerance in plants [98].
In turn, COI1, a core component of the JA-Ile coreceptor [59], is essential for JAZ transcript upregulation in the roots during the response to salt stress [95]. The JAZ upregulation mediated by salt stress in a COI1-dependent manner observed in the roots is likely to follow the canonical JA signaling pathway [100], with proteasome-mediated degradation of JAZ proteins. In this sense, JAZ proteins, the negative regulators of JA signaling, play an important role in the plant response to salt stress. Several JAZ homologous genes were upregulated by NaCl treatment in cotton, Arabidopsis roots, tomato, and wheat [95,101,102,103]. Moreover, the OsJAZ9 overexpression in rice resulted in a higher tolerance to salt stress [104] mainly through regulating the expression of ion transporters for K+ homeostasis [105]. Similarly, enhancing the expression of OsJAZ8 transcripts assured better performance of transgenic rice lines under salt stress [106]. Furthermore, the rice nuclear factor, RICE SALT SENSITIVE3 (RSS3), forms a ternary complex with class-C bHLH TFs and JAZ proteins and regulates root cell elongation during adaptation to salinity [107]. Remarkably, JAZ genes from Glycine soja (GsJAZ2), Malus domestica (MdJAZ2), Triticum durum (TdTIFY11a), and Pohlia nutans (PnJAZ1) introduced in Arabidopsis, granted greater tolerance to salinity [103,108,109,110]. The introduction of GaJAZ1 from Gossypium arboreum into Gossypium hirsutum (upland cotton) significantly increased salt tolerance in upland cotton compared to the WT strain. GaJAZ1-transgenic and WT plants showed many differentially expressed genes involved in JA signaling and biosynthesis, salt stress, and other hormone pathways. In GaJAZ-OE plants, the expression level of JAZ1/3/6/8 and JAZ10 were upregulated without NaCl treatment compared to the WT, but under salt conditions they were first downregulated (after 6 h and 12 h of treatment) and then upregulated again (24 h of treatment), indicating a sophisticated regulation of these genes in GaJAZ1-OE plants. Moreover, MYC2 was significantly upregulated while JAR1 was downregulated in GaJAZ1-OE plants. JA biosynthesis was also affected in GaJAZ1-OE plants since various JA synthesis-related genes (e.g., LOXs, AOS, AOC4, and JMT) changed their expression compared to the WT. Additionally, several salt stress-related genes encoding for a vacuolar-associated protein (VSR1), an osmotic protein (OSM34), and a plasma membrane ion exchanger (CHX18) among others showed significant downregulation resulting in the accumulation of osmolytes that protect the plant from salt stress damage. Furthermore, some phytohormone-related genes besides JA-related genes reprogrammed their expression in GaJAZ1-OE plants compared to the WT including those related with ethylene- (ACS, ERF2, and ERF4); ABA- (ABR1, ABA2, CBF4, and RD26); and auxin- (GH3.6) pathways. In general, these results suggested that ectopic overexpression of GaJAZ1 affects JA-related genes to increase salt tolerance in G. hirsutum plants albeit it also depends on other factors including hormone-crossing signal and salt-inducible genes [111].
Recently, JA-Ile catabolic genes have also been related to salt stress tolerance. Thus, OsCYP94C2b overexpression enhanced the viability of the transgenic rice under saline conditions and delayed the salt stress-induced leaf senescence [112]. Similarly, a higher CYP94C2b expression has been observed in some salt-tolerant rice varieties indicating that, at least in part, CYP94C2b may account for salt tolerance [113]. Hazman et al. [85] analyzed the accumulation of JAs and catabolic compounds in leaves from salt-exposed and control seedlings. OPDA and JA levels were increased by NaCl (100 mM) at most time evaluated points. Correspondingly, the JA-Ile catabolites 12OH-JA-Ile and 12COOH-JA-Ile were enhanced in response to salt exposure. Then, they explored the rice CYP94 and AH gene families and examined the transcriptional response of a gene subset under salt exposure. Among the evaluated CYP94 genes, only OsCYP94C2a was induced by salt stress while the transcripts of the AH genes fluctuated marginally. Apparently, OsCYP94C2a is the main player of JA-Ile oxidation upon salt stress in rice [85,112,113].
The apparent incongruity in the JA contribution to salt adaptation due to the positive effects of JA exogenous application (see Section 4), the upregulation of JA biosynthesis, and the induction of negative regulators of JA signaling and JA-Ile catabolism, indicates that timing and control of JA are maybe more important than its presence or absence [19,24]. Thus, detailed studies are required to reveal the underlying mechanism of efficient fine-tuning of jasmonate signaling in salt adaptation. Table 1 summarizes the findings regarding the main molecular components of the JA pathway associated with salt tolerance.
It is necessary to highlight that JA does not work independently in the improvement of plant tolerance to salt stress. Instead, its tightened coordination with other phytohormone signaling pathways allows the expression of multiple genes and flux of various metabolic pathways to adjust plant response to stress severity, specifically, in the appropriate time and tissue [18,34].

3. Crosstalk between JA and Other Plant Hormones during Salt Stress

In JA signaling, the proteins MYC, JAZ, and COI1 have been established as the core of the pathway and have been pointed to serve as a link between different hormone signaling [116]. Thus, the JA crosstalk with other phytohormones involves these components.

3.1. JA and ABA

Several studies have reported the combined action of JA and ABA in the plant response to salt stress. Although an earlier study described that ABA and JA antagonistically regulate the expression of transcripts inducible by salt stress in rice (Oryza sativa) [28], in more recent studies the synergistic action of both phytohormones has been observed. For instance, the application of ABA together with different concentrations of JA activated the protection mechanism against NaCl-associated stress in strawberry plants (Fragaria × ananassa) [117]. Likewise, ABA and JAs had a synergistic effect on the inhibition of seed germination under salinity conditions [110]. Moreover, Yang et al. [118] confirmed the cooperation between JA and ABA in the tolerance to salt stress mediated by phytochromes. Albeit the evidence of the concerted action of JA-ABA, the cross-coordination that exists between their signaling pathways is just beginning to be understood [119].
MYC2 seems to be an essential point for JA-ABA crosstalk as demonstrated in several studies. Accordingly, MYC2-OE Arabidopsis plants and the myc2 mutant show boosted and decreased ABA sensitivity, respectively [94,120]. Additionally, the expression of the salt- and ABA-responsive gene RD22 is promoted by MYC2 [94,96]. MYC2 and ABI5 (a transcription factor activated by the ABA signaling pathway) are modulated at the protein level through MED25, which is a multifunctional subunit of the Arabidopsis mediator complex [121]. In turn, the ABA receptor PYL6 directly interacts and alters the transcriptional activity of MYC2 [122]. Meanwhile, PYL4 is involved in the coregulatory effects of ABA and JA on plant growth and metabolism [123]. Furthermore, the induction of MYC2 by ABA seems to depend on the JA-Ile COI1 receptor according to Lorenzo et al. [120]. Recently, it has been reported that the application of ABA to strawberry plantlets involves the upregulation of the MYC2 gene (FabHLH80) from 0.5 to 12 h post-treatment [124].
Some studies highlight the importance of JAZ proteins as key nodes of JA-ABA crosstalk. The ubiquitin ligase E3 KEEP ON GOING (KEG), a known ABI5 repressor in the ABA signaling pathway, directly interacts with JAZ12 and modulates its stability [125]. Furthermore, JAZ3 interacts with ABI5 in vivo and represses its transcriptional activity [126]. The overexpression of PnJAZ1 (isolated from the moss P. nutans) in Arabidopsis plants inhibited the expression of genes of the ABA-dependent pathway related to seed germination and shoot growth under high salt conditions [110]. Furthermore, the transcription factor GbWRKY1 (from Gossypium barbadense) negatively regulated ABA signaling through an interaction network involving JAZ1 and ABI1 (the negative regulator of ABA signaling), in the response to salt and drought stress [115]. The evidence demonstrates the role of JAZs in the response to salt stress and JA-ABA crosstalk, however, the mechanisms of action in which these proteins participate in salt tolerance remain to be fully elucidated.

3.2. JA and Other Phytohormones

The nature of jasmonate crosstalk with other phytohormones in salt stress cannot be clearly described due to limited experimental data. At the molecular level, little is known about the convergence points of JA signaling and other phytohormone pathways. Due to the well-established central role of ABA under abiotic stress, more evidence of JA-ABA crosstalk is available [19]. In recent years, the crosstalk among different phytohormone mediating salt stress responses has been described [18]. However, it is necessary to deepen the JA crosstalk with other phytohormones to regulate salt stress tolerance in plants.

3.2.1. JA and Ethylene (ET)

JA–ET crosstalk can be complex and depends on the specific situation. A synergistic effect has been observed in promoting leaf senescence [127,128], but antagonistic interaction was found in controlling apical hook curvature [129]. JA–ET interactions in response to pathogen infections, herbivore attacks, and environmental stress are context-specific. The ET-stabilized transcription factor ETHYLENE-INSENSITIVE3 (EIN3) physically interacts with MYC2 and inhibits its DNA binding activity attenuating JA-regulated plant defense against generalist herbivores [129]. Since the jaz decuple mutant showed robust activation of insect and fungal pathogen defenses, the JA–ET crosstalk seems to be mediated via EIN3/ETHYLENE INSENSITIVE 3-like 1 (EIL1) along with JAZs-MYC2 [35]. The two TFs ETHYLENE RESPONSE FACTOR1 (ERF1) and OCTADECANOID-RESPONSIVE ARABIDOPSIS AP2/ERF 59 (ORA59) were previously implicated in integrating JA and ET signaling in Arabidopsis [130,131]. Recently, a new convergence-point of JA–ET signaling has been reported. OsLOX9, a JA biosynthetic pathway-related gene, is regulated by OsEIL1 in response to piercing-sucking insect attacks [132].
In salt stress, the synergistic activation of the Arabidopsis ERF1 by JA and ET is required for inducing tolerance [133]. Moreover, a rice root-specific pathogenesis-related protein (RSOsPR10), induced by high salt and other abiotic stresses, promotes root growth and root mass increasing salt tolerance [134]. JA and ET also induce RSOsPR10, while salicylic acid (SA) almost completely suppresses its induction [135,136]. JA-inducible and OsERF87-dependent expression of RSOsPR10 were strongly repressed by the SA-inducible OsWRKY76 transcription factor [137]. As OsERF87 and OsWRKY76 bind at the RSOsPR10 promoter, they antagonistically regulate RSOsPR10 expression. This mechanism represents a fine-tuning balance between JA/ET and SA signaling in plants under environmental challenges. The description of the molecular components of the synergistic JA–ET crosstalk in the regulation of RSOsPR10 expression requires investigation. Another gene activated by salt stress and treatment with MeJA or ethephon (an ethylene releasing compound) is GmCYP82A3, a gene from the soybean CYP82 family. Transgenic Nicotiana benthamiana plants overexpressing GmCYP82A3 exhibited strong pathogen resistance and enhanced salinity tolerance. Besides, an increased expression of the JA/ET signaling pathway-related genes was observed in the transgenic plants [138]. How GmCYP82A3 is involved in JA–ET crosstalk under salt stress could be an interesting topic to be approached.

3.2.2. JA and SA

The JA–SA antagonism is well known in plant defense pathways and key components of JA–SA crosstalk have been identified. Recently, many of the molecular components in the JA–SA crosstalk that regulate the plant immune network at transcriptional and protein levels are reviewed by Aerts et al. [139]. Among them, MAPKs are involved in the convergence of these phytohormone pathways [140,141,142]. AtMPK4 negatively regulates the activation of SA- and the repression of JA-mediated defenses under biotic stress [140]. Besides, MPK4 positively regulates the glutaredoxin GRX480 in the SA signaling pathway and negatively regulates MYC2 in the JA signaling pathway, which is necessary for JA responsive genes (PDF1.2 and THI2.1) [100]. SA-induced NON EXPRESSOR OF PR GENE (NPR1) activates GRX480, which can block the JA response gene expression mediated by TGA (a D group of Arabidopsis bZIP TFs), confirming SA–JA antagonism [143]. Additionally, SA-induced protein kinase (SIPK) and wound-induced protein kinase (WIPK), which are rapidly activated after perception of herbivory, regulate herbivory-induced JA levels and JA-mediated defense metabolite accumulations. WIPK-signaling is associated with large fitness costs in competing Nicotiana attenuata plants, while SIPK acts as an important regulator of plant fitness, possibly modulating SA-JA crosstalk through ethylene signaling [142].
The role of JA and SA in salt stress tolerance has been explored previously [144], but their relationship under salinity is not well known yet. Several reports show that SA and JA can alleviate the hazardous effect of salt stress on plants. In strawberry, improved physiological characters such as increased antioxidant activity and a reduced Na+/K+ ratio were observed on MeJA and SA treatments [145]. Likewise, soybean performance under salinity was improved by foliar spraying of JA and SA [146]. Treatment of soybean plants with SA plus JA stimulated H+-ATPase activity of tonoplast, nutrient uptake, and salt tolerance [147]. Additionally, exogenous applications of JA and SA decreased the concentration of Na+ in soybean under different salt stress levels [148]. Moreover, MeJA treatment can protect plants from salt-induced damage by acting role as an antioxidant and cooperating with SA [149]. All the evidence invites a deepening in the JA–SA interplay under salinity conditions for identifying the molecular components of their crosstalk.

3.2.3. JA and Gibberellins (GA)

JA and GA signaling pathway interaction has been described that occurs by the key GA signaling proteins DELLAs and JAZs since they directly interact [150]. The JAZ–DELLA interaction would interfere with the inhibition of MYC2 by JAZ proteins resulting in the activation of MYC2 downstream genes. In presence of GA, DELLA proteins are degraded and JAZs bind MYC2 to inhibit JA signaling [150,151]. On the other hand, the della mutant is less sensitive to the plant growth inhibition mediated by JA suggesting that JA delays the degradation of DELLA mediated by GA [152]. Curiously, JA signaling upregulates the transcription of RGL3, a DELLA gene, which the promotor is a target of MYC2 [153]. Additionally, RGL3 physically interacts with JAZ1 and JAZ8 [64,153] suggesting the JA-mediated degradation of JAZ1 and consequent release of MYC2 to induce the RGL3 expression, which in turn binds the non-JA degradable JAZ8 enhancing the MYC2-dependent JA responses [153]. Recently, it was reported that JA and GA synergistically promote fiber cell initiation of cotton (G. hirsutum) possibly mediated by the GhJAZ3 and GhSLR1 (a DELLA protein) interaction [154].
The interplay of JA and GA under salt conditions has not been studied in depth. However, there is evidence that in salt-stressed plants of basil (Ocimum basilicum) the GA concentration significantly decreased and in non-stressed plants treated with JA. Stressed plants treated with JA also showed a significant decrease in GA concentration [155] showing the antagonistic effect of JA in the GA level under salt stress. The effects of the combination of MeJA and NaCl treatment on the growth regulation and defense response of Nitraria tangutorum, a desert halophyte, have been recently investigated [156]. Compared with NaCl treatment alone, MeJA treatment aggravated the growth inhibition of seedlings by antagonizing growth-related hormones like GA. It was demonstrated that the transcript levels of GA-responsive genes NtPIF3, NtGAST1, and NtGSAT4 were suppressed by MeJA [156]. More studies are needed to find out the components of JA–GA crosstalk under saline conditions.

3.2.4. JA and Cytokinin (CK)

Interactions between JA and CK could grant some developmental flexibility under stress conditions since they mediate stress response and developmental processes. Although there are scarce reports on JA–CK crosstalk, some data indicate that their interaction could be negative or positive [157,158]. Antagonistic effects of JA and CK have been observed in different processes such as senescence, photosynthesis, RNA and protein synthesis, and vascular formation [157,159,160,161]. In contrast, a positive interplay between these phytohormones in delaying senescence of the Iris flower (Iris × hollandica) has been reported [158]. Moreover, CK treatments promoted gene expression of JA-amino synthetase in Arabidopsis and tomato plants [162] suggesting an interaction between the JA–CK pathway.
Recently, Avalbaev et al. [163] demonstrated the influence of exogenous MeJA on endogenous CK content in wheat plants under normal and salinity conditions. Low concentrations of MeJA (0.01–1 µM) increased wheat seedling growth while higher concentrations (10 and 100 µM) inhibited it. The hormonal balance of wheat seedlings was shifted by exogenous application of 0.1 µM MeJA. In response to salt stress, MeJA-untreated wheat plants increased the ABA level and gradually decreased indole acetic acid (IAA) and CK contents. Meanwhile, MeJA-pretreated seedlings were characterized by a diminution of ABA accumulation and IAA decreased level induced by salinity. Noticeably, a salinity-induced decline in the CK content was completely preventive by MeJA, which eventually resulted in the maintenance of the wheat seedlings growth rate under salt stress. It was suggested that MeJA application influences CK metabolism since cytokinin oxidase (CKX) gene expression and enzyme activity decreased after 1 h of MeJA treatment [163]. CKX catalyzes the degradation of CKs and controls the CK content in plants [164]. Similar results were obtained on almond rootstocks when MeJA application (0.025–0.05 mM) increased CK concentration in the leaf due to restriction of CKX activity and its gene expression [165]. MeJA-induced protection against salinity was found to be reached by modulating the activity of the antioxidant system and accumulation of osmoprotectants [165,166]. However, the mechanism by which exogenous MeJA mitigates the effect on the growth of salt-stressed plants as a result of the inhibition of CK decline under salt stress is largely unknown.

3.2.5. JA and Auxin (AUX)

The JA–AUX crosstalk was early reported by the identification of the auxin-resistant1 (axr1) mutants with altered jasmonate responsive gene expression [167]. Moreover, a link between JA signaling and AUX homeostasis was evidenced through the JA-mediated modulation of YUCCA8 and YUCCA9 gene expression, which are involved in AUX biosynthesis [168]. JA and AUX interaction also involves the ETHYLENE RESPONSE FACTOR109 (ERF109) during JA-induced lateral root formation [169]. Recently, Xu et al. [170] reviewed the novel progresses on the integration of JA and ethylene into AUX signaling in regulating root development of Arabidopsis thaliana [170]. Besides, Zhang et al. [171] provided an example of metabolic-level crosstalk between the JA and AUX signaling pathways by demonstrating that wounded leaves JA-inducible amidohydrolases (ILR1, ILL6, and IAR3) contribute to regulate active IAA and JA-Ile levels, promoting AUX signaling while attenuating JA signaling [171].
The transcription factor WRKY57, which is upregulated by IAA, but downregulated by JA, has been described as a convergence node of JA and IAA-mediated signaling. The JAZ4/8 and IAA29 (an AUX/IAA protein) repressors of JA and IAA signaling, respectively, have an opposite function in WRKY57 regulation since both competitively bind WRKY57 during JA-induced leaf senescence in Arabidopsis [172]. In earlier work, Jiang et al. [173] found that the Arabidopsis adt mutant, which is constitutive to expressing the WRKY57 gene, exhibited drought, osmotic, and salt tolerance. The enhanced tolerance of the adt mutant to these stresses was associated with an increment of ABA content consistently with the upregulation of RD29A, NCED3, and ABA3 genes. It was demonstrated that WRKY57 could directly bind the W-box of RD29A and NCED3 promoter sequences, suggesting that WRKY57 could regulate their expression. Since WRKY57 is regulated by JAZ4/8 and IAA29 repressors [172], it could be interesting to evaluate their role in the plant response to salt stress. Perhaps, it would shed light on the JA–AUX interplay under salinity conditions.
The main molecular/physiological effects related to phytohormone crosstalk are summarized in Table 2.

4. Effects of JA-Exogenous Applications for Improving Salt Stress Tolerance

Exogenous jasmonate applications have effects on different physiological aspects, including protection against biotic and abiotic stresses [174,175,176]. Methyl jasmonate (MeJA) application induces protection against oxidative stress as has been reported in different species and conditions [176,177,178,179].
In Arabidopsis thaliana, the total activities of catalase (CAT), peroxidase (POD/POX), superoxide dismutase (SOD), and glutathione reductase (GR) increased considerably in response to MeJA [180]. In crops, such as strawberry, MeJA applications during the preharvest period increase the anthocyanin and ascorbic acid contents, CAT, and ascorbate peroxidase (APX) activities in fruits at postharvest storage [176]. In this sense, increased activity of the antioxidant enzymes, together with higher levels of antioxidant compounds, as a result of MeJA treatment reinforces the antioxidant response of plants to reactive oxidative species (ROS) caused by abiotic stresses, like high salt.
From a physiological point of view, salt stress increases free proline content, photorespiration, and stomatal resistance among others, while decreasing net photosynthetic rates, transpiration, protein, and relative water content (RWC) [181,182]. Pretreatment with 0.1 mM MeJA helps the pea seedlings to counteract the salt stress since RWC and protein content of the treated seedlings were higher in comparison to NaCl-treated seedlings. Moreover, MeJA-treated pea seedlings present a decrease of Na+ and Cl accumulation in the shoot [183]. In another experiment, pretreatment with 0.1 mM MeJA for 3 days before salt treatment diminished the inhibitory effect of NaCl on the rate of 14CO2 fixation, and activity and content of ribulose-1,5-bisphosphate carboxylase/oxygenase [184]. Additionally, in rapeseed (Brassica napus), exogenously applied MeJA counteracted the inhibitory effects of NaCl by increasing RWC, soluble sugar content, and photosynthesis rate [174]. The application of 0.25 mM MeJA was the most effective treatment to enhance salt tolerance at a concentration of 60 mM NaCl in strawberry (F. × ananassa ‘Camarosa’) seedlings [145]. In almond rootstocks, application of MeJA in optimal concentrations of 0.025–0.05 mM alleviated the adverse effect of salt stress by increasing the photosynthetic rate, activity of antioxidant enzymes (APX, SOD, and POX), root and shoot dry mass, and cell membrane integrity [165]. Alleviation of moderate salinity (40 mM NaCl) by foliar application of 5 mM MeJA has also been reported in broccoli [185]. Pretreatment of cowpea seeds with 0.05 mM MeJA improves plant tolerance to salt stress [186].
In other plants, like Limonium bicolor, which is a typical recretohalophyte with salt glands in the epidermis, 300 mM NaCl led to a dramatic inhibition of seedling growth that was significantly alleviated by the application of 0.03 mM MeJA, resulting in biomass close to that of plants not subjected to salt stress [187]. Even in high salt concentrations such as 500 mM NaCl, MeJA applied at 0.1 mM has a protective role in the defense response of Robinia pseudoacacia especially with a marked increase in the activity of antioxidant enzymes and related gene expression [188]. In another study, foliar applications of 0.5 mM MeJA increased the essential oil content and the antioxidant activities of basil (O. basilicum ‘Genove’) on 30 mM NaCl and have noticeable effects on the main components of the oils [189]. In Glycyrrhiza uralensis exposed to 100 mM NaCl, 0.025 or 0.05 mM MeJA increased the root length of salt-stressed G. uralensis seedlings but decreased root diameter, stem length, and stem diameter, enhancing peroxidase activity and ascorbate content [190].
In wheat seedlings, NaCl salt stress caused a significant increase in the malondialdehyde (MDA) content and H2O2 concentrations and a concomitant decrease in SOD, POD, CAT, and APX activities. Exogenous JA pretreatment (2 mM) combined with NaCl treatment (150 mM) produced a significant decline in MDA and H2O2 concentrations and an increase in SOD, POD, CAT, and APX activities. Moreover, a marked upregulation of SOD, POD, CAT, and APX genes was observed in the JA–NaCl combined treatment in comparison to NaCl treatment alone. Additionally, exogenous JA remarkably increased glutathione (GSH) concentration in wheat seedlings treated with NaCl and decreased the deleterious effect of salt stress on the growth of wheat [31]. These results indicate that exogenous JA can effectively scavenge ROS by enhancing the activities of antioxidant enzymes and the concentration of antioxidant compounds in wheat seedlings under salt stress and consequently play an important role in decreasing lipid peroxidation and increase the ability of wheat to resist salt stress [31].
In roselle (Hibiscus sabdariffa), the exogenous JA treatment protected roselle seedlings against salt-induced harms through enhancing the activities of both enzymatic and non-enzymatic antioxidants, such as APX, pyrogallol peroxidase (PPX), and PPO, and the accumulation of metabolites non-reducing sugars, total phenols, anthocyanins, flavonoids, and proline. The JA-treated roselle exhibited a significant increase in growth parameters under salt conditions compared to the WT [191]. In forage sorghum (Sorghum bicolor), at a high salinity level (200 mM), seeds treated with 10 mM JA showed a positive effect on various growth and physiological parameters such as emergence percentage, emergence rate, shoot length, total fresh weight, salt tolerance index, and total chlorophyll among others [192]. Similarly, seed priming and foliar application with JA enhanced salinity stress tolerance of soybean (Glycine max) seedlings. Improved water and osmotic potentials, water use efficiency, net photosynthetic, transpiration rate, stomatal conductance, and total chlorophyll content were observed in JA-primed and treated soybean seedlings compared to the untreated ones. Besides, JA treatment resulted in a reduction of Na+ concentration and an increment of K+ concentrations in the leaf and root of the analyzed cultivars despite salinity stress [193].
Although the exogenous application of JA and its derivates constitute a suitable approach to improve the plant response to salt stress, this strategy has some limitations. Due to the JA–SA antagonism during the plant defense response [194], an increment in JA content by its exogenous application will negatively affect the SA-mediated response to biotrophic pathogens. In addition, JA can inhibit plant growth by reprogramming plant metabolism to produce diverse defense compounds [35], thus the growth-defense trade-off is also an issue. Moreover, the high economic cost of MeJA application in field treatments [176] could prevent its use to mitigate the salt-induced damage in crop plants. It is necessary to keep in mind that the activation of JA-beneficial effects in plant response to salt stress depends on the JA levels and therefore, field experiments will be required to analyze the cost-effective JA doses for exogenous applications.

5. Application of Genome-Associated Tools for Salt Tolerance Mediated by JA

Salt stress tolerance is a complex trait regulated by polygenes [195]. In this context, QTL mapping and GWASs provide a suitable opportunity to identify genes responsible for quantitative trait variation such as salt tolerance. In this regard, genetic factors associated with salt stress have been previously investigated in several crops, such as rice [41,196,197], barley [198,199], wheat [200,201], chickpea [202], sesame [203], cotton [204,205], and soybean [206].
GWAS and QTL mapping have been implemented to identify the genetic factors involved in both osmotic and ionic components of salinity stress. A typical GWAS workflow to identify genes related to salt stress (and genes conferring salt tolerance) in crops is represented in Figure 2. Four main steps can be distinguished: (1) plant genotyping; (2) plant phenotyping based on morphological and physiological traits related to salt stress; (3) identification of marker–trait associations (MTAs); and (4) identification of candidate genes involved in the salt stress response. A diverse panel is genotyped using DNA markers, i.e., single nucleotide polymorphisms (SNPs), diversity arrays technology (DArT), or RNA-sequencing (RNA-seq) for the identification of genetic variants that affect the gene expression level in a specific tissue. Parallelly, this panel is phenotyped for different traits regarding the objectives of the study. In the context of salt stress in plants, the phenotyping is generally carried out considering several morphological and physiological traits, such as leaf area, root and shoot dry weight, seed germination rate, salt stress index (SSI), Na+/K+ ratio, chlorophyll content, and MDA, and, remarkably, changes in the phytohormone levels such as JAs. Particularly, these traits are evaluated in plants or seedlings that have been subjected to salt treatment. Subsequently, the genotypic and phenotypic data are combined to associate alleles with particular traits using classical GWAS models, which significantly detect molecular markers associated with the studied traits. Posteriorly, the MTAs can serve as a starting point for the mining of candidate genes. Gene Ontology (GO) annotation of the putative candidate genes can be carried out using BLAST tools.
In a GWAS for salt tolerance (or salt stress), the candidate genes are usually associated with gene annotations, such as abiotic stress-related, anion transport, Ca+ binding and signaling, phytohormone response elements, and others [207,208,209]. In this way, many TFs and downstream genes related to phytohormones biosynthesis and signaling have been characterized, including ABA, SA, JA, ET, and others considered as growth promotion hormones, including AUX, GA, and brassinosteroids [41,205,209,210,211,212,213]. Additionally, the candidate genes associated with salt stress or tolerance can be validated by quantitative reverse transcription PCR (RT-qPCR) analysis.
To our knowledge, several studies have analyzed the JA response to different stresses, but few have explored JA-dependent genetic mechanisms taking into account natural genetic variation. To et al. [214] performed a GWAS to identify genetic variants associated with exogenous JA treatment responses in rice. They found a high natural variability for the shoot and root growth (in a panel of 150 rice accessions) in response to JA treatment. This GWAS revealed about 230 candidate genes, including several JA-responsive TFs known to play a stress response role. Several GWASs have elucidated the participation of JA in salt tolerance in plants. In this sense, Li et al. [203] reported potential candidate genes related to drought and salt-induced stress in sesame. In this GWAS, the SiOPR3 gene (detected for drought stress in sesame) is the ortholog of the Arabidopsis OPR3 gene an essential component of the JA biosynthesis.
Besides, Rohila et al. [41] detected several candidate genes associated with seedling stage salt tolerance by the GWAS approach in a rice core-collection. Particularly, one SNP (on chromosome 3) was located close to the ALLENE OXIDE CYCLASE 1 (AOC1) gene. This gene has been related to salt and other abiotic stresses, and it is a key gene in the JA biosynthetic pathway in Arabidopsis [215]. Yuan et al. [205] combined the association mapping and RNA-seq analyzes to explore candidate genes for salt-tolerance in cotton at the germination stage. At least nine genes were associated with signaling or a response to signal factors, such as SA, GA, and JA. In other GWASs, genes related to the JA biosynthesis and signaling pathways have been indirectly identified, in which upstream genes from the JA signaling events and JA-regulated genes during salt stress were detected. For instance, An et al. [216] carried out a GWAS to identify associations conferring salt tolerance in rice. In this study, a significant SNP for seedling length was located on the promoter of a salt stress-related gene (RSOsPR10), which has been proposed to be induced by biotic and abiotic stresses, via the JA signaling pathway [135]. Sun et al. [204] reported DNA polymorphisms associated with salt tolerance candidate genes at the cotton seedling stage. Additionally, the expression levels of six genes were reported using salt-tolerant and salt-sensitive varieties. Interestingly, the expression of Gh_A10G1756, a homolog of the Arabidopsis CALCIUM-DEPENDENT PROTEIN KINASE 1 (AtCPK1) gene, had higher expression in the salt-sensitive than salt-tolerant types [204]. The AtCPK1 gene mediates pathogen resistance in Arabidopsis and plays a positive role in salt/drought-stress response [217]. According to Coca and San Segundo [218], AtCPK1-OE plants showed activation of two components of the chloroplastic pathway for JA biosynthesis (AOS and AOC genes), which could support the idea that JA is a key element for triggering different responses to salt stress in plants. Patishtan et al. [208] implemented a GWAS using a diversity panel of 306 rice accessions treated with different salt concentrations. The authors characterized more than 30 candidate genes for short, medium, and long-term NaCl treatment. Notably, among these genes, a DNA polymorphism was associated with the WRKY70 gene, which plays a pivotal role in an antagonistic interaction between SA and JA responses [210,219].
Due to the above-mentioned studies, GWAS and association studies have successfully identified many novel genes associated with traits of interest. These findings will be useful towards the developing of varieties tolerant to salt stress, using new genomic techniques, such GE; an aspect that has not been addressed in depth. In this sense, GE systems provide the ability to modify genes and generate new possibilities for crop improvement precisely. The RNA-guided CRISPR/Cas9 technology is simple to use across different genome editing technologies [220] and has been generally used in major crops such as wheat [221], maize [222], soybean [223], and many others. Even more, the highly efficient multiplex editing toolkit based on an intron-optimized zCas9i gene, which allows assembly of nuclease constructs expressing up to 32 sgRNAs [224], can enable simultaneously targeting of multiple independent loci to generate complex genotypes or to functionally interrogate groups of candidate genes such as those involved in phytohormones signaling.
To improve the salt tolerance in crops employing the CRISPR/Cas9 system with a focus on phytohormones, an example is the targeting of the OsRR22 gene, which encodes a transcription factor involved in CK signaling, which has effectively enhanced salt tolerance in rice [46]. Recently, Liu et al. [225] enhanced drought tolerance in tomato (Solanum lycopersicum) by CRISPR/Cas9 targeted mutagenesis of the SlLBD40 gene. SlLBD40 encodes an organ boundaries domain transcription factor, which is highly induced by polyethylene glycol (PEG), salt, and MeJA treatments. The analysis of the SlLBD40 expression in the jasmonic acid-insensitive1 (jai1) mutant (a mutant in the JA-Ile tomato receptor COI1) and MYC2-silenced plants demonstrated that SlLBD40 depends on JA signaling for its activation and it might be downstream of SlMYC2 [225]. The previous works offer a good background for targeting key components of the JA pathway to obtain high salinity tolerance in crops using GE tools.

6. Concluding Remarks

Besides the role of development and abiotic stress responses, the jasmonate pathway is certainly involved in plant salt stress responses. Several JA-biosynthetic genes are induced under salt stress, although the lack of jasmonates is also related to this tolerance. Key JA-associated molecular components such as the transcription factor MYC2 and the repressor JAZ seem to be crucial in salt tolerance. Remarkably, overexpression of JAZ genes confers salt tolerance in transgenic plants. Regarding phytohormone crosstalk between JA and others in a salt stress context, certainly, more information exists about JA-ABA crosstalk, focused on MYC2 and JAZ interactions with the ABA signaling-components.
In terms of future applied perspectives, on the one hand, several reports demonstrate the positive effects of JA exogenous applications on the physiological status against salt stress. It should be considered as complementary crop management to face not only salinity-derived damages but also all abiotic stresses in a global change framework. On the other hand, genomic tools such as GWAS could help to reveal several JA pathway-associated genes that can serve as a guide in breeding programs and targets in genome editing systems, such as CRISPR/Cas9, to get salt-resistant crops. Surely, the next years will be promising in discoveries and applications related to the role of jasmonates against salt stress in plants.

Author Contributions

Conceptualization, C.R.F. and F.M.-P.; writing—original draft preparation, C.D., C.R.F., F.M.-P. and S.A.; writing—review and editing, C.D., C.R.F., F.M.-P. and J.-T.C.; visualization, C.D., C.R.F., F.M.-P. and S.A.; funding acquisition, C.R.F. All authors have read and agreed to the published version of the manuscript.

Funding

This research and the APC were funded by the National Research and Development Agency (ANID, Chile) grant FONDECYT/Regular 1181310 to C.R.F. Funding approval date: 1 April 2018.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

C.D. thanks ANID for a doctoral scholarship (grant “Beca Doctorado Nacional” No. 6647/2019).

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study, in the collection, analyses, or interpretation of data, in the writing of the manuscript, or in the decision to publish the results.

References

  1. Zörb, C.; Geilfus, C.M.; Dietz, K.J. Salinity and crop yield. Plant Biol. 2019, 21, 31–38. [Google Scholar] [CrossRef] [PubMed]
  2. Ivushkin, K.; Bartholomeus, H.; Bregt, A.K.; Pulatov, A.; Kempen, B.; De Sousa, L. Global mapping of soil salinity change. Remote Sens. Environ. 2019, 231, 111260. [Google Scholar] [CrossRef]
  3. Kumar, K.; Kumar, M.; Kim, S.-R.; Ryu, H.; Cho, Y.-G. Insights into genomics of salt stress response in rice. Rice 2013, 6, 1–15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Dasgupta, S.; Hossain, M.M.; Huq, M.; Wheeler, D. Climate change and soil salinity: The case of coastal Bangladesh. Ambio 2015, 44, 815–826. [Google Scholar] [CrossRef] [Green Version]
  5. Daliakopoulos, I.; Tsanis, I.; Koutroulis, A.; Kourgialas, N.; Varouchakis, A.; Karatzas, G.; Ritsema, C. The threat of soil salinity: A European scale review. Sci. Total Environ. 2016, 573, 727–739. [Google Scholar] [CrossRef]
  6. Fan, X.; Pedroli, B.; Liu, G.; Liu, Q.; Liu, H.; Shu, L. Soil salinity development in the yellow river delta in relation to groundwater dynamics. Land Degrad. Dev. 2012, 23, 175–189. [Google Scholar] [CrossRef]
  7. Moreira Barradas, J.; Abdelfattah, A.; Matula, S.; Dolezal, F. Effect of fertigation on soil salinization and aggregate stability. J. Irrig. Drain. Eng. 2015, 141, 05014010. [Google Scholar] [CrossRef]
  8. Moral, R.; Perez-Murcia, M.; Perez-Espinosa, A.; Moreno-Caselles, J.; Paredes, C.; Rufete, B. Salinity, organic content, micronutrients and heavy metals in pig slurries from South-eastern Spain. Waste Manag. 2008, 28, 367–371. [Google Scholar] [CrossRef]
  9. Lefebvre, O.; Moletta, R. Treatment of organic pollution in industrial saline wastewater: A literature review. Water Res. 2006, 40, 3671–3682. [Google Scholar] [CrossRef]
  10. Mateo-Sagasta, J.; Burke, J. Agriculture and water quality interactions: A global overview. Solaw Backgr. Themat. Rep. 2011, 46. [Google Scholar]
  11. Yang, Y.; Guo, Y. Elucidating the molecular mechanisms mediating plant salt-stress responses. N. Phytol. 2018, 217, 523–539. [Google Scholar] [CrossRef] [Green Version]
  12. Arif, Y.; Singh, P.; Siddiqui, H.; Bajguz, A.; Hayat, S. Salinity induced physiological and biochemical changes in plants: An omic approach towards salt stress tolerance. Plant Physiol. Biochem. 2020, 156, 64–77. [Google Scholar] [CrossRef]
  13. Acosta-Motos, J.R.; Ortuño, M.F.; Bernal-Vicente, A.; Diaz-Vivancos, P.; Sanchez-Blanco, M.J.; Hernandez, J.A. Plant responses to salt stress: Adaptive mechanisms. Agronomy 2017, 7, 18. [Google Scholar] [CrossRef] [Green Version]
  14. Ali, Q.; Shahid, S.; Nazar, N.; Hussain, A.I.; Ali, S.; Chatha, S.A.S.; Perveen, R.; Naseem, J.; Haider, M.Z.; Hussain, B. Use of phytohormones in conferring tolerance to environmental stress. In Plant Ecophysiology and Adaptation under Climate Change: Mechanisms and Perspectives II; Springer: Berlin/Heidelberg, Germany, 2020; pp. 245–355. [Google Scholar] [CrossRef]
  15. Raghavendra, A.S.; Gonugunta, V.K.; Christmann, A.; Grill, E. ABA perception and signalling. Trends Plant Sci. 2010, 15, 395–401. [Google Scholar] [CrossRef]
  16. Suzuki, N.; Bassil, E.; Hamilton, J.S.; Inupakutika, M.A.; Zandalinas, S.I.; Tripathy, D.; Luo, Y.; Dion, E.; Fukui, G.; Kumazaki, A. ABA is required for plant acclimation to a combination of salt and heat stress. PLoS ONE 2016, 11, e0147625. [Google Scholar] [CrossRef] [Green Version]
  17. Llanes, A.; Andrade, A.; Alemano, S.; Luna, V. Metabolomic approach to understand plant adaptations to water and salt stress. In Plant Metabolites and Regulation under Environmental Stress; Elsevier: Amsterdam, The Netherlands, 2018; pp. 133–144. [Google Scholar] [CrossRef]
  18. Yu, Z.; Duan, X.; Luo, L.; Dai, S.; Ding, Z.; Xia, G. How Plant Hormones Mediate Salt Stress Responses. Trends Plant Sci. 2020, 25, 1117–1130. [Google Scholar] [CrossRef]
  19. Riemann, M.; Dhakarey, R.; Hazman, M.; Miro, B.; Kohli, A.; Nick, P. Exploring jasmonates in the hormonal network of drought and salinity responses. Front. Plant Sci. 2015, 6, 1077. [Google Scholar] [CrossRef] [Green Version]
  20. Ahmad, P.; Rasool, S.; Gul, A.; Sheikh, S.A.; Akram, N.A.; Ashraf, M.; Kazi, A.; Gucel, S. Jasmonates: Multifunctional roles in stress tolerance. Front. Plant Sci. 2016, 7, 813. [Google Scholar] [CrossRef] [Green Version]
  21. Wang, J.; Song, L.; Gong, X.; Xu, J.; Li, M. Functions of jasmonic acid in plant regulation and response to abiotic stress. Int. J. Mol. Sci. 2020, 21, 1446. [Google Scholar] [CrossRef] [Green Version]
  22. Campos, M.L.; Kang, J.-H.; Howe, G.A. Jasmonate-triggered plant immunity. J. Chem. Ecol. 2014, 40, 657–675. [Google Scholar] [CrossRef]
  23. Wang, J.; Wu, D.; Wang, Y.; Xie, D. Jasmonate action in plant defense against insects. J. Exp. Bot. 2019, 70, 3391–3400. [Google Scholar] [CrossRef]
  24. Ismail, A.; Seo, M.; Takebayashi, Y.; Kamiya, Y.; Eiche, E.; Nick, P. Salt adaptation requires efficient fine-tuning of jasmonate signalling. Protoplasma 2014, 251, 881–898. [Google Scholar] [CrossRef]
  25. Abouelsaad, I.; Renault, S. Enhanced oxidative stress in the jasmonic acid-deficient tomato mutant def-1 exposed to NaCl stress. J. Plant Physiol. 2018, 226, 136–144. [Google Scholar] [CrossRef]
  26. Ahmad, B.; Raina, A.; Naikoo, M.I.; Khan, S. Role of methyl jasmonates in salt stress tolerance in crop plants. In Plant Signaling Molecules; Elsevier: Amsterdam, The Netherlands, 2019; pp. 371–384. [Google Scholar] [CrossRef]
  27. Kang, D.J.; Seo, Y.J.; Lee, J.D.; Ishii, R.; Kim, K.; Shin, D.; Park, S.; Jang, S.; Lee, I.J. Jasmonic acid differentially affects growth, ion uptake and abscisic acid concentration in salt-tolerant and salt-sensitive rice cultivars. J. Agron. Crop. Sci. 2005, 191, 273–282. [Google Scholar] [CrossRef]
  28. Moons, A.; Prinsen, E.; Bauw, G.; Van Montagu, M. Antagonistic effects of abscisic acid and jasmonates on salt stress-inducible transcripts in rice roots. Plant Cell 1997, 9, 2243–2259. [Google Scholar] [CrossRef] [Green Version]
  29. Tani, T.; Sobajima, H.; Okada, K.; Chujo, T.; Arimura, S.-I.; Tsutsumi, N.; Nishimura, M.; Seto, H.; Nojiri, H.; Yamane, H. Identification of the OsOPR7 gene encoding 12-oxophytodienoate reductase involved in the biosynthesis of jasmonic acid in rice. Planta 2008, 227, 517. [Google Scholar] [CrossRef]
  30. Du, H.; Liu, H.; Xiong, L. Endogenous auxin and jasmonic acid levels are differentially modulated by abiotic stresses in rice. Front. Plant Sci. 2013, 4, 397. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Qiu, Z.; Guo, J.; Zhu, A.; Zhang, L.; Zhang, M. Exogenous jasmonic acid can enhance tolerance of wheat seedlings to salt stress. Ecotoxicol. Environ. Saf. 2014, 104, 202–208. [Google Scholar] [CrossRef] [PubMed]
  32. Yoon, J.Y.; Hamayun, M.; Lee, S.-K.; Lee, I.-J. Methyl jasmonate alleviated salinity stress in soybean. J. Crop. Sci. Biotechnol. 2009, 12, 63–68. [Google Scholar] [CrossRef]
  33. Javid, M.G.; Sorooshzadeh, A.; Moradi, F.; Modarres Sanavy, S.A.M.; Allahdadi, I. The role of phytohormones in alleviating salt stress in crop plants. Aust. J. Crop. Sci. 2011, 5, 726. [Google Scholar]
  34. Yang, J.; Duan, G.; Li, C.; Liu, L.; Han, G.; Zhang, Y.; Wang, C. The crosstalks between jasmonic acid and other plant hormone signaling highlight the involvement of jasmonic acid as a core component in plant response to biotic and abiotic stresses. Front. Plant Sci. 2019, 10, 1349. [Google Scholar] [CrossRef] [Green Version]
  35. Guo, Q.; Yoshida, Y.; Major, I.T.; Wang, K.; Sugimoto, K.; Kapali, G.; Havko, N.E.; Benning, C.; Howe, G.A. JAZ repressors of metabolic defense promote growth and reproductive fitness in Arabidopsis. Proc. Natl. Acad. Sci. USA 2018, 115, E10768–E10777. [Google Scholar] [CrossRef] [Green Version]
  36. Jang, G.; Yoon, Y.; Choi, Y.D. Crosstalk with jasmonic acid integrates multiple responses in plant development. Int. J. Mol. Sci. 2020, 21, 305. [Google Scholar] [CrossRef] [Green Version]
  37. Kazan, K.; Manners, J.M. JAZ repressors and the orchestration of phytohormone crosstalk. Trends Plant Sci. 2012, 17, 22–31. [Google Scholar] [CrossRef]
  38. Zander, M.; Lewsey, M.G.; Clark, N.M.; Yin, L.; Bartlett, A.; Guzmán, J.P.S.; Hann, E.; Langford, A.E.; Jow, B.; Wise, A. Integrated multi-omics framework of the plant response to jasmonic acid. Nat. Plants 2020, 6, 290–302. [Google Scholar] [CrossRef]
  39. Wani, S.H.; Kumar, V.; Khare, T.; Guddimalli, R.; Parveda, M.; Solymosi, K.; Suprasanna, P.; Kishor, P.K. Engineering salinity tolerance in plants: Progress and prospects. Planta 2020, 251, 1–29. [Google Scholar] [CrossRef]
  40. Thomson, M.J.; De Ocampo, M.; Egdane, J.; Rahman, M.A.; Sajise, A.G.; Adorada, D.L.; Tumimbang-Raiz, E.; Blumwald, E.; Seraj, Z.I.; Singh, R.K. Characterizing the Saltol quantitative trait locus for salinity tolerance in rice. Rice 2010, 3, 148–160. [Google Scholar] [CrossRef] [Green Version]
  41. Rohila, J.S.; Edwards, J.D.; Tran, G.D.; Jackson, A.K.; McClung, A.M. Identification of Superior Alleles for Seedling Stage Salt Tolerance in the USDA Rice Mini-Core Collection. Plants 2019, 8, 472. [Google Scholar] [CrossRef] [Green Version]
  42. Korte, A.; Farlow, A. The advantages and limitations of trait analysis with GWAS: A review. Plant Methods 2013, 9, 1–9. [Google Scholar] [CrossRef] [Green Version]
  43. Flint-Garcia, S.A.; Thuillet, A.C.; Yu, J.; Pressoir, G.; Romero, S.M.; Mitchell, S.E.; Doebley, J.; Kresovich, S.; Goodman, M.M.; Buckler, E.S. Maize association population: A high-resolution platform for quantitative trait locus dissection. Plant J. 2005, 44, 1054–1064. [Google Scholar] [CrossRef]
  44. Huang, X.; Han, B. Natural variations and genome-wide association studies in crop plants. Annu. Rev. Plant Biol. 2014, 65, 531–551. [Google Scholar] [CrossRef]
  45. Zhao, K.; Aranzana, M.J.; Kim, S.; Lister, C.; Shindo, C.; Tang, C.; Toomajian, C.; Zheng, H.; Dean, C.; Marjoram, P. An Arabidopsis example of association mapping in structured samples. PLoS Genet. 2007, 3, e4. [Google Scholar] [CrossRef] [Green Version]
  46. Zhang, A.; Liu, Y.; Wang, F.; Li, T.; Chen, Z.; Kong, D.; Bi, J.; Zhang, F.; Luo, X.; Wang, J. Enhanced rice salinity tolerance via CRISPR/Cas9-targeted mutagenesis of the OsRR22 gene. Mol. Breed. 2019, 39, 1–10. [Google Scholar] [CrossRef] [Green Version]
  47. Tran, M.T.; Doan, D.T.H.; Kim, J.; Song, Y.J.; Sung, Y.W.; Das, S.; Kim, E.J.; Son, G.H.; Kim, S.H.; Van Vu, T. CRISPR/Cas9-based precise excision of SlHyPRP1 domain (s) to obtain salt stress-tolerant tomato. Plant Cell Rep. 2020, 1–13. [Google Scholar] [CrossRef]
  48. Kazan, K. Diverse roles of jasmonates and ethylene in abiotic stress tolerance. Trends Plant Sci. 2015, 20, 219–229. [Google Scholar] [CrossRef] [PubMed]
  49. Wasternack, C.; Strnad, M. Jasmonates: News on occurrence, biosynthesis, metabolism and action of an ancient group of signaling compounds. Int. J. Mol. Sci. 2018, 19, 2539. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Wasternack, C.; Feussner, I. The oxylipin pathways: Biochemistry and function. Annu. Rev. Plant Biol. 2018, 69, 363–386. [Google Scholar] [CrossRef] [PubMed]
  51. Wasternack, C.; Kombrink, E. Jasmonates: Structural requirements for lipid-derived signals active in plant stress responses and development. ACS Chem. Biol. 2010, 5, 63–77. [Google Scholar] [CrossRef] [PubMed]
  52. Seo, H.S.; Song, J.T.; Cheong, J.-J.; Lee, Y.-H.; Lee, Y.-W.; Hwang, I.; Lee, J.S.; Do Choi, Y. Jasmonic acid carboxyl methyltransferase: A key enzyme for jasmonate-regulated plant responses. Proc. Natl. Acad. Sci. USA 2001, 98, 4788–4793. [Google Scholar] [CrossRef] [Green Version]
  53. Staswick, P.E.; Tiryaki, I. The oxylipin signal jasmonic acid is activated by an enzyme that conjugates it to isoleucine in Arabidopsis. Plant Cell 2004, 16, 2117–2127. [Google Scholar] [CrossRef] [Green Version]
  54. Koo, Y.J.; Yoon, E.S.; Seo, J.S.; Kim, J.-K.; Do Choi, Y. Characterization of a methyl jasmonate specific esterase in Arabidopsis. J. Korean Soc. Appl. Biol. Chem. 2013, 56, 27–33. [Google Scholar] [CrossRef]
  55. Woldemariam, M.G.; Onkokesung, N.; Baldwin, I.T.; Galis, I. Jasmonoyl-l-isoleucine hydrolase 1 (JIH1) regulates jasmonoyl-l-isoleucine levels and attenuates plant defenses against herbivores. Plant J. 2012, 72, 758–767. [Google Scholar] [CrossRef]
  56. Fonseca, S.; Chini, A.; Hamberg, M.; Adie, B.; Porzel, A.; Kramell, R.; Miersch, O.; Wasternack, C.; Solano, R. (+)-7-iso-Jasmonoyl-L-isoleucine is the endogenous bioactive jasmonate. Nat. Chem. Biol. 2009, 5, 344–350. [Google Scholar] [CrossRef]
  57. Chini, A.; Gimenez-Ibanez, S.; Goossens, A.; Solano, R. Redundancy and specificity in jasmonate signalling. Curr. Opin. Plant Biol. 2016, 33, 147–156. [Google Scholar] [CrossRef]
  58. Chini, A.; Fonseca, S.; Fernandez, G.; Adie, B.; Chico, J.; Lorenzo, O.; Garcia-Casado, G.; López-Vidriero, I.; Lozano, F.; Ponce, M. The JAZ family of repressors is the missing link in jasmonate signalling. Nature 2007, 448, 666–671. [Google Scholar] [CrossRef]
  59. Sheard, L.B.; Tan, X.; Mao, H.; Withers, J.; Ben-Nissan, G.; Hinds, T.R.; Kobayashi, Y.; Hsu, F.-F.; Sharon, M.; Browse, J. Jasmonate perception by inositol-phosphate-potentiated COI1–JAZ co-receptor. Nature 2010, 468, 400–405. [Google Scholar] [CrossRef]
  60. Thines, B.; Katsir, L.; Melotto, M.; Niu, Y.; Mandaokar, A.; Liu, G.; Nomura, K.; He, S.Y.; Howe, G.A.; Browse, J. JAZ repressor proteins are targets of the SCF COI1 complex during jasmonate signalling. Nature 2007, 448, 661–665. [Google Scholar] [CrossRef]
  61. Garrido-Bigotes, A.; Valenzuela-Riffo, F.; Figueroa, C.R. Evolutionary analysis of JAZ proteins in plants: An approach in search of the ancestral sequence. Int. J. Mol. Sci. 2019, 20, 5060. [Google Scholar] [CrossRef] [Green Version]
  62. Garrido-Bigotes, A.; Valenzuela-Riffo, F.; Torrejón, M.; Solano, R.; Morales-Quintana, L.; Figueroa, C.R. A new functional JAZ degron sequence in strawberry JAZ1 revealed by structural and interaction studies on the COI1–JA-Ile/COR–JAZs complexes. Sci. Rep. 2020, 10, 1–17. [Google Scholar] [CrossRef]
  63. Pauwels, L.; Barbero, G.F.; Geerinck, J.; Tilleman, S.; Grunewald, W.; Pérez, A.C.; Chico, J.M.; Bossche, R.V.; Sewell, J.; Gil, E. NINJA connects the co-repressor TOPLESS to jasmonate signalling. Nature 2010, 464, 788–791. [Google Scholar] [CrossRef] [Green Version]
  64. Shyu, C.; Figueroa, P.; De Pew, C.L.; Cooke, T.F.; Sheard, L.B.; Moreno, J.E.; Katsir, L.; Zheng, N.; Browse, J.; Howe, G.A. JAZ8 lacks a canonical degron and has an EAR motif that mediates transcriptional repression of jasmonate responses in Arabidopsis. Plant Cell 2012, 24, 536–550. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Zhang, F.; Yao, J.; Ke, J.; Zhang, L.; Lam, V.Q.; Xin, X.-F.; Zhou, X.E.; Chen, J.; Brunzelle, J.; Griffin, P.R. Structural basis of JAZ repression of MYC transcription factors in jasmonate signalling. Nature 2015, 525, 269–273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Çevik, V.; Kidd, B.N.; Zhang, P.; Hill, C.; Kiddle, S.; Denby, K.J.; Holub, E.B.; Cahill, D.M.; Manners, J.M.; Schenk, P.M. MEDIATOR25 acts as an integrative hub for the regulation of jasmonate-responsive gene expression in Arabidopsis. Plant Physiol. 2012, 160, 541–555. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. An, C.; Li, L.; Zhai, Q.; You, Y.; Deng, L.; Wu, F.; Chen, R.; Jiang, H.; Wang, H.; Chen, Q. Mediator subunit MED25 links the jasmonate receptor to transcriptionally active chromatin. Proc. Natl. Acad. Sci. USA 2017, 114, E8930–E8939. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Chung, H.S.; Koo, A.J.; Gao, X.; Jayanty, S.; Thines, B.; Jones, A.D.; Howe, G.A. Regulation and function of Arabidopsis JASMONATE ZIM-domain genes in response to wounding and herbivory. Plant Physiol. 2008, 146, 952–964. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Fernández-Calvo, P.; Chini, A.; Fernández-Barbero, G.; Chico, J.-M.; Gimenez-Ibanez, S.; Geerinck, J.; Eeckhout, D.; Schweizer, F.; Godoy, M.; Franco-Zorrilla, J.M. The Arabidopsis bHLH transcription factors MYC3 and MYC4 are targets of JAZ repressors and act additively with MYC2 in the activation of jasmonate responses. Plant Cell 2011, 23, 701–715. [Google Scholar] [CrossRef] [Green Version]
  70. Figueroa, P.; Browse, J. Male sterility in A rabidopsis induced by overexpression of a MYC 5-SRDX chimeric repressor. Plant J. 2015, 81, 849–860. [Google Scholar] [CrossRef] [Green Version]
  71. Pires, N.; Dolan, L. Origin and diversification of basic-helix-loop-helix proteins in plants. Mol. Biol. Evol. 2010, 27, 862–874. [Google Scholar] [CrossRef] [Green Version]
  72. Lian, T.-F.; Xu, Y.-P.; Li, L.-F.; Su, X.-D. Crystal structure of tetrameric Arabidopsis MYC2 reveals the mechanism of enhanced interaction with DNA. Cell Rep. 2017, 19, 1334–1342. [Google Scholar] [CrossRef] [Green Version]
  73. Toledo-Ortiz, G.; Huq, E.; Quail, P.H. The Arabidopsis basic/helix-loop-helix transcription factor family. Plant Cell 2003, 15, 1749–1770. [Google Scholar] [CrossRef] [Green Version]
  74. Figueroa, P.; Browse, J. The Arabidopsis JAZ2 promoter contains a G-Box and thymidine-rich module that are necessary and sufficient for jasmonate-dependent activation by MYC transcription factors and repression by JAZ proteins. Plant Cell Physiol. 2012, 53, 330–343. [Google Scholar] [CrossRef] [Green Version]
  75. Godoy, M.; Franco-Zorrilla, J.M.; Pérez-Pérez, J.; Oliveros, J.C.; Lorenzo, Ó.; Solano, R. Improved protein-binding microarrays for the identification of DNA-binding specificities of transcription factors. Plant J. 2011, 66, 700–711. [Google Scholar] [CrossRef]
  76. Dombrecht, B.; Xue, G.P.; Sprague, S.J.; Kirkegaard, J.A.; Ross, J.J.; Reid, J.B.; Fitt, G.P.; Sewelam, N.; Schenk, P.M.; Manners, J.M. MYC2 differentially modulates diverse jasmonate-dependent functions in Arabidopsis. Plant Cell 2007, 19, 2225–2245. [Google Scholar] [CrossRef] [Green Version]
  77. Van Moerkercke, A.; Duncan, O.; Zander, M.; Šimura, J.; Broda, M.; Bossche, R.V.; Lewsey, M.G.; Lama, S.; Singh, K.B.; Ljung, K. A MYC2/MYC3/MYC4-dependent transcription factor network regulates water spray-responsive gene expression and jasmonate levels. Proc. Natl. Acad. Sci. USA 2019, 116, 23345–23356. [Google Scholar] [CrossRef]
  78. Hickman, R.; Van Verk, M.C.; Van Dijken, A.J.; Mendes, M.P.; Vroegop-Vos, I.A.; Caarls, L.; Steenbergen, M.; Van der Nagel, I.; Wesselink, G.J.; Jironkin, A. Architecture and dynamics of the jasmonic acid gene regulatory network. Plant Cell 2017, 29, 2086–2105. [Google Scholar] [CrossRef] [Green Version]
  79. Chini, A.; Monte, I.; Zamarreño, A.M.; Hamberg, M.; Lassueur, S.; Reymond, P.; Weiss, S.; Stintzi, A.; Schaller, A.; Porzel, A.; et al. An OPR3-independent pathway uses 4,5-didehydrojasmonate for jasmonate synthesis. Nat. Chem. Biol. 2018, 14, 171–178. [Google Scholar] [CrossRef] [Green Version]
  80. Heitz, T.; Smirnova, E.; Marquis, V.; Poirier, L. Metabolic control within the jasmonate biochemical pathway. Plant Cell Physiol. 2019, 60, 2621–2628. [Google Scholar] [CrossRef] [PubMed]
  81. Kitaoka, N.; Matsubara, T.; Sato, M.; Takahashi, K.; Wakuta, S.; Kawaide, H.; Matsui, H.; Nabeta, K.; Matsuura, H. Arabidopsis CYP94B3 encodes jasmonyl-L-isoleucine 12-hydroxylase, a key enzyme in the oxidative catabolism of jasmonate. Plant Cell Physiol. 2011, 52, 1757–1765. [Google Scholar] [CrossRef] [Green Version]
  82. Koo, A.J.; Cooke, T.F.; Howe, G.A. Cytochrome P450 CYP94B3 mediates catabolism and inactivation of the plant hormone jasmonoyl-L-isoleucine. Proc. Natl. Acad. Sci. USA 2011, 108, 9298–9303. [Google Scholar] [CrossRef] [Green Version]
  83. Heitz, T.; Widemann, E.; Lugan, R.; Miesch, L.; Ullmann, P.; Désaubry, L.; Holder, E.; Grausem, B.; Kandel, S.; Miesch, M. Cytochromes P450 CYP94C1 and CYP94B3 catalyze two successive oxidation steps of plant hormone jasmonoyl-isoleucine for catabolic turnover. J. Biol. Chem. 2012, 287, 6296–6306. [Google Scholar] [CrossRef] [Green Version]
  84. Widemann, E.; Miesch, L.; Lugan, R.; Holder, E.; Heinrich, C.; Aubert, Y.; Miesch, M.; Pinot, F.; Heitz, T. The amidohydrolases IAR3 and ILL6 contribute to jasmonoyl-isoleucine hormone turnover and generate 12-hydroxyjasmonic acid upon wounding in Arabidopsis leaves. J. Biol. Chem. 2013, 288, 31701–31714. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Hazman, M.; Sühnel, M.; Schäfer, S.; Zumsteg, J.; Lesot, A.; Beltran, F.; Marquis, V.; Herrgott, L.; Miesch, L.; Riemann, M. Characterization of jasmonoyl-isoleucine (JA-Ile) hormonal catabolic pathways in rice upon wounding and salt stress. Rice 2019, 12, 1–14. [Google Scholar] [CrossRef] [PubMed]
  86. Poudel, A.N.; Holtsclaw, R.E.; Kimberlin, A.; Sen, S.; Zeng, S.; Joshi, T.; Lei, Z.; Sumner, L.W.; Singh, K.; Matsuura, H. 12-Hydroxy-jasmonoyl-L-isoleucine is an active jasmonate that signals through CORONATINE INSENSITIVE 1 and contributes to the wound response in Arabidopsis. Plant Cell Physiol. 2019, 60, 2152–2166. [Google Scholar] [CrossRef]
  87. Jimenez-Aleman, G.H.; Almeida-Trapp, M.; Fernández-Barbero, G.; Gimenez-Ibanez, S.; Reichelt, M.; Vadassery, J.; Mithöfer, A.; Caballero, J.; Boland, W.; Solano, R. Omega hydroxylated JA-Ile is an endogenous bioactive jasmonate that signals through the canonical jasmonate signaling pathway. Biochim. Biophys. Acta Mol. Cell Biol. Lipids 2019, 1864, 158520. [Google Scholar] [CrossRef]
  88. Jiang, Y.; Deyholos, M.K. Comprehensive transcriptional profiling of NaCl-stressed Arabidopsis roots reveals novel classes of responsive genes. BMC Plant Biol. 2006, 6, 1–20. [Google Scholar] [CrossRef] [Green Version]
  89. Ma, S.; Gong, Q.; Bohnert, H.J. Dissecting salt stress pathways. J. Exp. Bot. 2006, 57, 1097–1107. [Google Scholar] [CrossRef] [Green Version]
  90. Zhang, H.; Zhang, Q.; Zhai, H.; Li, Y.; Wang, X.; Liu, Q.; He, S. Transcript profile analysis reveals important roles of jasmonic acid signalling pathway in the response of sweet potato to salt stress. Sci. Rep. 2017, 7, 1–12. [Google Scholar] [CrossRef] [Green Version]
  91. Ding, H.; Lai, J.; Wu, Q.; Zhang, S.; Chen, L.; Dai, Y.-S.; Wang, C.; Du, J.; Xiao, S.; Yang, C. Jasmonate complements the function of Arabidopsis lipoxygenase3 in salinity stress response. Plant Sci. 2016, 244, 1–7. [Google Scholar] [CrossRef]
  92. Zhao, Y.; Dong, W.; Zhang, N.; Ai, X.; Wang, M.; Huang, Z.; Xiao, L.; Xia, G. A wheat allene oxide cyclase gene enhances salinity tolerance via jasmonate signaling. Plant Physiol. 2014, 164, 1068–1076. [Google Scholar] [CrossRef] [Green Version]
  93. Hazman, M.; Hause, B.; Eiche, E.; Nick, P.; Riemann, M. Increased tolerance to salt stress in OPDA-deficient rice ALLENE OXIDE CYCLASE mutants is linked to an increased ROS-scavenging activity. J. Exp. Bot. 2015, 66, 3339–3352. [Google Scholar] [CrossRef] [Green Version]
  94. Abe, H.; Urao, T.; Ito, T.; Seki, M.; Shinozaki, K.; Yamaguchi-Shinozaki, K. Arabidopsis AtMYC2 (bHLH) and AtMYB2 (MYB) function as transcriptional activators in abscisic acid signaling. Plant Cell 2003, 15, 63–78. [Google Scholar] [CrossRef] [Green Version]
  95. Valenzuela, C.E.; Acevedo-Acevedo, O.; Miranda, G.S.; Vergara-Barros, P.; Holuigue, L.; Figueroa, C.R.; Figueroa, P.M. Salt stress response triggers activation of the jasmonate signaling pathway leading to inhibition of cell elongation in Arabidopsis primary root. J. Exp. Bot. 2016, 67, 4209–4220. [Google Scholar] [CrossRef] [Green Version]
  96. Iwasaki, T.; Yamaguchi-Shinozaki, K.; Shinozaki, K. Identification of a cis-regulatory region of a gene in Arabidopsis thaliana whose induction by dehydration is mediated by abscisic acid and requires protein synthesis. Mol. Gen. Genet. MGG 1995, 247, 391–398. [Google Scholar] [CrossRef]
  97. Verma, D.; Jalmi, S.K.; Bhagat, P.K.; Verma, N.; Sinha, A.K. A bHLH transcription factor, MYC2, imparts salt intolerance by regulating proline biosynthesis in Arabidopsis. FEBS J. 2020, 287, 2560–2576. [Google Scholar] [CrossRef]
  98. Seo, J.S.; Zhao, P.; Jung, C.; Chua, N.-H. Plant U-box protein 10 negatively regulates abscisic acid response in Arabidopsis. Appl. Biol. Chem. 2019, 62, 39. [Google Scholar] [CrossRef] [Green Version]
  99. Jung, C.; Zhao, P.; Seo, J.S.; Mitsuda, N.; Deng, S.; Chua, N.-H. Plant U-box protein10 regulates MYC2 stability in Arabidopsis. Plant Cell 2015, 27, 2016–2031. [Google Scholar] [CrossRef] [Green Version]
  100. Wasternack, C.; Hause, B. Jasmonates: Biosynthesis, perception, signal transduction and action in plant stress response, growth and development. An update to the 2007 review in Annals of Botany. Ann. Bot. 2013, 111, 1021–1058. [Google Scholar] [CrossRef]
  101. Yao, D.; Zhang, X.; Zhao, X.; Liu, C.; Wang, C.; Zhang, Z.; Zhang, C.; Wei, Q.; Wang, Q.; Yan, H. Transcriptome analysis reveals salt-stress-regulated biological processes and key pathways in roots of cotton (Gossypium hirsutum L.). Genomics 2011, 98, 47–55. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Chini, A.; Ben-Romdhane, W.; Hassairi, A.; Aboul-Soud, M.A. Identification of TIFY/JAZ family genes in Solanum lycopersicum and their regulation in response to abiotic stresses. PLoS ONE 2017, 12, e0177381. [Google Scholar] [CrossRef]
  103. Ebel, C.; BenFeki, A.; Hanin, M.; Solano, R.; Chini, A. Characterization of wheat (Triticum aestivum) TIFY family and role of Triticum Durum Td TIFY11a in salt stress tolerance. PLoS ONE 2018, 13, e0200566. [Google Scholar] [CrossRef] [Green Version]
  104. Ye, H.; Du, H.; Tang, N.; Li, X.; Xiong, L. Identification and expression profiling analysis of TIFY family genes involved in stress and phytohormone responses in rice. Plant Mol. Biol. 2009, 71, 291–305. [Google Scholar] [CrossRef] [PubMed]
  105. Wu, H.; Ye, H.; Yao, R.; Zhang, T.; Xiong, L. OsJAZ9 acts as a transcriptional regulator in jasmonate signaling and modulates salt stress tolerance in rice. Plant Sci. 2015, 232, 1–12. [Google Scholar] [CrossRef] [PubMed]
  106. Peethambaran, P.K.; Glenz, R.; Höninger, S.; Islam, S.S.; Hummel, S.; Harter, K.; Kolukisaoglu, Ü.; Meynard, D.; Guiderdoni, E.; Nick, P. Salt-inducible expression of OsJAZ8 improves resilience against salt-stress. BMC Plant Biol. 2018, 18, 1–15. [Google Scholar] [CrossRef]
  107. Toda, Y.; Tanaka, M.; Ogawa, D.; Kurata, K.; Kurotani, K.-I.; Habu, Y.; Ando, T.; Sugimoto, K.; Mitsuda, N.; Katoh, E. Rice salt sensitive 3 forms a ternary complex with JAZ and class-C bHLH factors and regulates jasmonate-induced gene expression and root cell elongation. Plant Cell 2013, 25, 1709–1725. [Google Scholar] [CrossRef] [Green Version]
  108. Zhu, D.; Cai, H.; Luo, X.; Bai, X.; Deyholos, M.K.; Chen, Q.; Chen, C.; Ji, W.; Zhu, Y. Over-expression of a novel JAZ family gene from Glycine soja, increases salt and alkali stress tolerance. Biochem. Biophys. Res. Commun. 2012, 426, 273–279. [Google Scholar] [CrossRef]
  109. An, X.-H.; Hao, Y.-J.; Li, E.-M.; Xu, K.; Cheng, C.-G. Functional identification of apple MdJAZ2 in Arabidopsis with reduced JA-sensitivity and increased stress tolerance. Plant Cell Rep. 2017, 36, 255–265. [Google Scholar] [CrossRef]
  110. Liu, S.; Zhang, P.; Li, C.; Xia, G. The moss jasmonate ZIM-domain protein PnJAZ1 confers salinity tolerance via crosstalk with the abscisic acid signalling pathway. Plant Sci. 2019, 280, 1–11. [Google Scholar] [CrossRef]
  111. Zhao, G.; Song, Y.; Wang, Q.; Yao, D.; Li, D.; Qin, W.; Ge, X.; Yang, Z.; Xu, W.; Su, Z. Gossypium hirsutum Salt Tolerance is Enhanced by Overexpression of G. arboreum JAZ1. Front. Bioeng. Biotechnol. 2020, 8, 157. [Google Scholar] [CrossRef]
  112. Kurotani, K.-I.; Hayashi, K.; Hatanaka, S.; Toda, Y.; Ogawa, D.; Ichikawa, H.; Ishimaru, Y.; Tashita, R.; Suzuki, T.; Ueda, M. Elevated levels of CYP94 family gene expression alleviate the jasmonate response and enhance salt tolerance in rice. Plant Cell Physiol. 2015, 56, 779–789. [Google Scholar] [CrossRef] [Green Version]
  113. Kurotani, K.-I.; Yamanaka, K.; Toda, Y.; Ogawa, D.; Tanaka, M.; Kozawa, H.; Nakamura, H.; Hakata, M.; Ichikawa, H.; Hattori, T. Stress tolerance profiling of a collection of extant salt-tolerant rice varieties and transgenic plants overexpressing abiotic stress tolerance genes. Plant Cell Physiol. 2015, 56, 1867–1876. [Google Scholar] [CrossRef] [Green Version]
  114. Zhu, D.; Bai, X.; Luo, X.; Chen, Q.; Cai, H.; Ji, W.; Zhu, Y. Identification of wild soybean (Glycine soja) TIFY family genes and their expression profiling analysis under bicarbonate stress. Plant Cell Rep. 2013, 32, 263–272. [Google Scholar] [CrossRef] [PubMed]
  115. Luo, X.; Li, C.; He, X.; Zhang, X.; Zhu, L. ABA signaling is negatively regulated by GbWRKY1 through JAZ1 and ABI1 to affect salt and drought tolerance. Plant Cell Rep. 2020, 39, 181–194. [Google Scholar] [CrossRef] [PubMed]
  116. Howe, G.A.; Major, I.T.; Koo, A.J. Modularity in jasmonate signaling for multistress resilience. Annu. Rev. Plant Biol. 2018, 69, 387–415. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Jamalian, S.; Truemper, C.; Pawelzik, E. Jasmonic and Abscisic Acid Contribute to Metabolism Re-adjustment in Strawberry Leaves under NaCl Stress. Int. J. Fruit Sci. 2020, 20, 1–22. [Google Scholar] [CrossRef]
  118. Yang, T.; Lv, R.; Li, J.; Lin, H.; Xi, D. Phytochrome A and B negatively regulate salt stress tolerance of Nicotiana tobacum via ABA-jasmonic acid synergistic cross-talk. Plant Cell Physiol. 2018, 59, 2381–2393. [Google Scholar] [CrossRef]
  119. De Lucas, M.; Brady, S.M. Gene regulatory networks in the Arabidopsis root. Curr. Opin. Plant Biol. 2013, 16, 50–55. [Google Scholar] [CrossRef]
  120. Lorenzo, O.; Chico, J.M.; Sánchez-Serrano, J.J.; Solano, R. Jasmonate-insensitive 1 encodes a MYC transcription factor essential to discriminate between different jasmonate-regulated defense responses in Arabidopsis. Plant Cell 2004, 16, 1938–1950. [Google Scholar] [CrossRef] [Green Version]
  121. Chen, R.; Jiang, H.; Li, L.; Zhai, Q.; Qi, L.; Zhou, W.; Liu, X.; Li, H.; Zheng, W.; Sun, J. The Arabidopsis mediator subunit MED25 differentially regulates jasmonate and abscisic acid signaling through interacting with the MYC2 and ABI5 transcription factors. Plant Cell 2012, 24, 2898–2916. [Google Scholar] [CrossRef] [Green Version]
  122. Aleman, F.; Yazaki, J.; Lee, M.; Takahashi, Y.; Kim, A.Y.; Li, Z.; Kinoshita, T.; Ecker, J.R.; Schroeder, J.I. An ABA-increased interaction of the PYL6 ABA receptor with MYC2 transcription factor: A putative link of ABA and JA signaling. Sci. Rep. 2016, 6, 1–10. [Google Scholar] [CrossRef]
  123. Lackman, P.; González-Guzmán, M.; Tilleman, S.; Carqueijeiro, I.; Pérez, A.C.; Moses, T.; Seo, M.; Kanno, Y.; Häkkinen, S.T.; Van Montagu, M.C. Jasmonate signaling involves the abscisic acid receptor PYL4 to regulate metabolic reprogramming in Arabidopsis and tobacco. Proc. Natl. Acad. Sci. USA 2011, 108, 5891–5896. [Google Scholar] [CrossRef] [Green Version]
  124. Zhao, F.; Li, G.; Hu, P.; Zhao, X.; Li, L.; Wei, W.; Feng, J.; Zhou, H. Identification of basic/helix-loop-helix transcription factors reveals candidate genes involved in anthocyanin biosynthesis from the strawberry white-flesh mutant. Sci. Rep. 2018, 8, 1–15. [Google Scholar] [CrossRef] [Green Version]
  125. Pauwels, L.; Ritter, A.; Goossens, J.; Durand, A.N.; Liu, H.; Gu, Y.; Geerinck, J.; Boter, M.; Bossche, R.V.; De Clercq, R. The ring e3 ligase keep on going modulates jasmonate zim-domain12 stability. Plant Physiol. 2015, 169, 1405–1417. [Google Scholar] [CrossRef] [Green Version]
  126. Ju, L.; Jing, Y.; Shi, P.; Liu, J.; Chen, J.; Yan, J.; Chu, J.; Chen, K.M.; Sun, J. JAZ proteins modulate seed germination through interaction with ABI 5 in bread wheat and Arabidopsis. N. Phytol. 2019, 223, 246–260. [Google Scholar] [CrossRef]
  127. Kim, J.H.; Chung, K.M.; Woo, H.R. Three positive regulators of leaf senescence in Arabidopsis, ORE1, ORE3 and ORE9, play roles in crosstalk among multiple hormone-mediated senescence pathways. Genes Genom. 2011, 33, 373–381. [Google Scholar] [CrossRef]
  128. Lim, C.; Kang, K.; Shim, Y.; Sakuraba, Y.; An, G.; Paek, N.-C. Rice ethylene response factor 101 promotes leaf senescence through Jasmonic acid-mediated regulation of OsNAP and OsMYC2. Front. Plant Sci. 2020, 11, 1096. [Google Scholar] [CrossRef] [PubMed]
  129. Song, S.; Huang, H.; Gao, H.; Wang, J.; Wu, D.; Liu, X.; Yang, S.; Zhai, Q.; Li, C.; Qi, T. Interaction between MYC2 and ethylene insensitive 3 modulates antagonism between jasmonate and ethylene signaling in Arabidopsis. Plant Cell 2014, 26, 263–279. [Google Scholar] [CrossRef] [Green Version]
  130. Lorenzo, O.; Piqueras, R.; Sánchez-Serrano, J.J.; Solano, R. Ethylene response factor 1 integrates signals from ethylene and jasmonate pathways in plant defense. Plant Cell 2003, 15, 165–178. [Google Scholar] [CrossRef] [Green Version]
  131. Pré, M.; Atallah, M.; Champion, A.; De Vos, M.; Pieterse, C.M.; Memelink, J. The AP2/ERF domain transcription factor ORA59 integrates jasmonic acid and ethylene signals in plant defense. Plant Physiol. 2008, 147, 1347–1357. [Google Scholar] [CrossRef] [Green Version]
  132. Ma, F.; Yang, X.; Shi, Z.; Miao, X. Novel crosstalk between ethylene-and jasmonic acid-pathway responses to a piercing-sucking insect in rice. N. Phytol. 2020, 225, 474–487. [Google Scholar] [CrossRef] [Green Version]
  133. Cheng, M.-C.; Liao, P.-M.; Kuo, W.-W.; Lin, T.-P. The Arabidopsis ethylene response factor 1 regulates abiotic stress-responsive gene expression by binding to different cis-acting elements in response to different stress signals. Plant Physiol. 2013, 162, 1566–1582. [Google Scholar] [CrossRef] [Green Version]
  134. Takeuchi, K.; Hasegawa, H.; Gyohda, A.; Komatsu, S.; Okamoto, T.; Okada, K.; Terakawa, T.; Koshiba, T. Overexpression of RSOsPR10, a root-specific rice PR10 gene, confers tolerance against drought stress in rice and drought and salt stresses in bentgrass. Plant Cell Tissue Organ Cult. 2016, 127, 35–46. [Google Scholar] [CrossRef]
  135. Hashimoto, M.; Kisseleva, L.; Sawa, S.; Furukawa, T.; Komatsu, S.; Koshiba, T. A novel rice PR10 protein, RSOsPR10, specifically induced in roots by biotic and abiotic stresses, possibly via the jasmonic acid signaling pathway. Plant Cell Physiol. 2004, 45, 550–559. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Takeuchi, K.; Gyohda, A.; Tominaga, M.; Kawakatsu, M.; Hatakeyama, A.; Ishii, N.; Shimaya, K.; Nishimura, T.; Riemann, M.; Nick, P. RSOsPR10 expression in response to environmental stresses is regulated antagonistically by jasmonate/ethylene and salicylic acid signaling pathways in rice roots. Plant Cell Physiol. 2011, 52, 1686–1696. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Yamamoto, T.; Yoshida, Y.; Nakajima, K.; Tominaga, M.; Gyohda, A.; Suzuki, H.; Okamoto, T.; Nishimura, T.; Yokotani, N.; Minami, E. Expression of RSOsPR10 in rice roots is antagonistically regulated by jasmonate/ethylene and salicylic acid via the activator OsERF87 and the repressor OsWRKY76, respectively. Plant Direct 2018, 2, e00049. [Google Scholar] [CrossRef] [Green Version]
  138. Yan, Q.; Cui, X.; Lin, S.; Gan, S.; Xing, H.; Dou, D. GmCYP82A3, a soybean cytochrome P450 family gene involved in the jasmonic acid and ethylene signaling pathway, enhances plant resistance to biotic and abiotic stresses. PLoS ONE 2016, 11, e0162253. [Google Scholar] [CrossRef] [Green Version]
  139. Aerts, N.; Pereira Mendes, M.; Van Wees, S.C. Multiple levels of crosstalk in hormone networks regulating plant defense. Plant J. 2021, 105, 489–504. [Google Scholar] [CrossRef]
  140. Brodersen, P.; Petersen, M.; Bjørn Nielsen, H.; Zhu, S.; Newman, M.A.; Shokat, K.M.; Rietz, S.; Parker, J.; Mundy, J. Arabidopsis MAP kinase 4 regulates salicylic acid-and jasmonic acid/ethylene-dependent responses via EDS1 and PAD4. Plant J. 2006, 47, 532–546. [Google Scholar] [CrossRef] [Green Version]
  141. Leon-Reyes, A.; Van der Does, D.; De Lange, E.S.; Delker, C.; Wasternack, C.; Van Wees, S.C.M.; Ritsema, T.; Pieterse, C.M.J. Salicylate-mediated suppression of jasmonate-responsive gene expression in Arabidopsis is targeted downstream of the jasmonate biosynthesis pathway. Planta 2010, 232, 1423–1432. [Google Scholar] [CrossRef] [Green Version]
  142. Meldau, S.; Ullman-Zeunert, L.; Govind, G.; Bartram, S.; Baldwin, I.T. MAPK-dependent JA and SA signalling in Nicotiana attenuataaffects plant growth and fitness during competition with conspecifics. BMC Plant Biol. 2012, 12, 213. [Google Scholar] [CrossRef] [Green Version]
  143. Zander, M.; Chen, S.; Imkampe, J.; Thurow, C.; Gatz, C. Repression of the Arabidopsis thaliana jasmonic acid/ethylene-induced defense pathway by TGA-interacting glutaredoxins depends on their C-terminal ALWL motif. Mol. Plant 2012, 5, 831–840. [Google Scholar] [CrossRef] [Green Version]
  144. Khan, M.I.R.; Syeed, S.; Nazar, R.; Anjum, N.A. An insight into the role of salicylic acid and jasmonic acid in salt stress tolerance. Phytohorm. Abiotic Stress Toler. Plants 2012, 277–300. [Google Scholar] [CrossRef]
  145. Faghih, S.; Ghobadi, C.; Zarei, A. Response of strawberry plant cv‘Camarosa’to salicylic acid and methyl jasmonate application under salt stress condition. J. Plant Growth Regul. 2017, 36, 651–659. [Google Scholar] [CrossRef]
  146. Sheokand, M.; Jakhar, S.; Singh, V.; Sikerwal, V. Effect of salicylic acid, 24-Epibrassinolide and jasmonic acid in modulating morpho-physiological and biochemical constituents in Glycine max L. merill under salt stress. Int. J. Res. Anal. Rev. 2018, 5, i434–i442. [Google Scholar]
  147. Ghassemi-Golezani, K.; Farhangi-Abriz, S. Foliar sprays of salicylic acid and jasmonic acid stimulate H+-ATPase activity of tonoplast, nutrient uptake and salt tolerance of soybean. Ecotoxicol. Environ. Saf. 2018, 166, 18–25. [Google Scholar] [CrossRef]
  148. Farhangi-Abriz, S.; Tavasolee, A.; Ghassemi-Golezani, K.; Torabian, S.; Monirifar, H.; Rahmani, H.A. Growth-promoting bacteria and natural regulators mitigate salt toxicity and improve rapeseed plant performance. Protoplasma 2020, 257, 1035–1047. [Google Scholar] [CrossRef]
  149. Seckin-Dinler, B.; Tasci, E.; Sarisoy, U.; Gul, V. The cooperation between methyl jasmonate and salicylic acid to protect soybean (Glycine max L.) from salinity. Fresenius Environ. Bull. 2018, 27, 1618–1626. [Google Scholar]
  150. Hou, X.; Lee, L.Y.C.; Xia, K.; Yan, Y.; Yu, H. Dellas modulate jasmonate signaling via competitive binding to JAZs. Dev. Cell 2010, 19, 884–894. [Google Scholar] [CrossRef] [Green Version]
  151. Boter, M.; Ruíz-Rivero, O.; Abdeen, A.; Prat, S. Conserved MYC transcription factors play a key role in jasmonate signaling both in tomato and Arabidopsis. Genes Dev. 2004, 18, 1577–1591. [Google Scholar] [CrossRef] [Green Version]
  152. Yang, D.-L.; Yao, J.; Mei, C.-S.; Tong, X.-H.; Zeng, L.-J.; Li, Q.; Xiao, L.-T.; Sun, T.-P.; Li, J.; Deng, X.-W. Plant hormone jasmonate prioritizes defense over growth by interfering with gibberellin signaling cascade. Proc. Natl. Acad. Sci. USA 2012, 109, E1192–E1200. [Google Scholar] [CrossRef] [Green Version]
  153. Wild, M.; Davière, J.-M.; Cheminant, S.; Regnault, T.; Baumberger, N.; Heintz, D.; Baltz, R.; Genschik, P.; Achard, P. The Arabidopsis DELLA RGA-LIKE3 is a direct target of MYC2 and modulates jasmonate signaling responses. Plant Cell 2012, 24, 3307–3319. [Google Scholar] [CrossRef] [Green Version]
  154. Xia, X.-C.; Hu, Q.-Q.; Li, W.; Chen, Y.; Han, L.-H.; Tao, M.; Wu, W.-Y.; Li, X.-B.; Huang, G.-Q. Cotton (Gossypium hirsutum) JAZ3 and SLR1 function in jasmonate and gibberellin mediated epidermal cell differentiation and elongation. Plant Cell Tissue Organ Cult. 2018, 133, 249–262. [Google Scholar] [CrossRef]
  155. Al Timman, W.M.A.; Kamil, T.A. Alleviation of salt stress in Ocimum basilicum plants by jasmonic acid treatment. Plant Arch. 2019, 19, 1550–1557. [Google Scholar]
  156. Gao, Z.; Gao, S.; Li, P.; Zhang, Y.; Ma, B.; Wang, Y. Exogenous methyl jasmonate promotes salt stress-induced growth inhibition and prioritizes defense response of Nitraria tangutorum Bobr. Physiol. Plant 2021, 1–14. [Google Scholar] [CrossRef]
  157. Stoynova-Bakalova, E.; Petrov, P.; Gigova, L.; Baskin, T. Differential effects of methyl jasmonate on growth and division of etiolated zucchini cotyledons. Plant Biol. 2008, 10, 476–484. [Google Scholar] [CrossRef] [Green Version]
  158. Van Doorn, W.G.; Çelikel, F.G.; Pak, C.; Harkema, H. Delay of Iris flower senescence by cytokinins and jasmonates. Physiol. Plant 2013, 148, 105–120. [Google Scholar] [CrossRef]
  159. Mukherjee, I.; Reid, D.M.; Naik, G.R. Influence of cytokinins on the methyl jasmonate-promoted senescence in Helianthus annuus cotyledons. Plant Growth Regul. 2002, 38, 61–68. [Google Scholar] [CrossRef]
  160. Jang, G.; Chang, S.H.; Um, T.Y.; Lee, S.; Kim, J.-K.; Choi, Y.D. Antagonistic interaction between jasmonic acid and cytokinin in xylem development. Sci. Rep. 2017, 7, 10212. [Google Scholar] [CrossRef] [Green Version]
  161. Ananieva, K.; Malbeck, J.; Kamínek, M.; Van Staden, J. Methyl jasmonate down-regulates endogenous cytokinin levels in cotyledons of Cucurbita pepo (zucchini) seedlings. Physiol. Plant 2004, 122, 496–503. [Google Scholar] [CrossRef]
  162. Shi, X.; Gupta, S.; Lindquist, I.E.; Cameron, C.T.; Mudge, J.; Rashotte, A.M. Transcriptome analysis of cytokinin response in tomato leaves. PLoS ONE 2013, 8, e55090. [Google Scholar] [CrossRef] [Green Version]
  163. Avalbaev, A.; Yuldashev, R.; Fedorova, K.; Somov, K.; Vysotskaya, L.; Allagulova, C.; Shakirova, F. Exogenous methyl jasmonate regulates cytokinin content by modulating cytokinin oxidase activity in wheat seedlings under salinity. J. Plant Physiol. 2016, 191, 101–110. [Google Scholar] [CrossRef] [PubMed]
  164. Vysotskaya, L.B.; Korobova, A.V.; Veselov, S.Y.; Dodd, I.C.; Kudoyarova, G.R. ABA mediation of shoot cytokinin oxidase activity: Assessing its impacts on cytokinin status and biomass allocation of nutrient-deprived durum wheat. Funct. Plant Biol. 2009, 36, 66–72. [Google Scholar] [CrossRef]
  165. Tavallali, V.; Karimi, S. Methyl jasmonate enhances salt tolerance of almond rootstocks by regulating endogenous phytohormones, antioxidant activity and gas-exchange. J. Plant Physiol. 2019, 234, 98–105. [Google Scholar] [CrossRef]
  166. Avalbaev, A.; Allagulova, C.; Maslennikova, D.; Fedorova, K.; Shakirova, F. Methyl jasmonate and cytokinin mitigate the salinity-induced oxidative injury in wheat seedlings. J. Plant Growth Regul. 2020, 1–12. [Google Scholar] [CrossRef]
  167. Tiryaki, I.; Staswick, P.E. An Arabidopsis mutant defective in jasmonate response is allelic to the auxin-signaling mutant axr1. Plant Physiol. 2002, 130, 887–894. [Google Scholar] [CrossRef] [Green Version]
  168. Hentrich, M.; Böttcher, C.; Düchting, P.; Cheng, Y.; Zhao, Y.; Berkowitz, O.; Masle, J.; Medina, J.; Pollmann, S. The jasmonic acid signaling pathway is linked to auxin homeostasis through the modulation of YUCCA 8 and YUCCA 9 gene expression. Plant J. 2013, 74, 626–637. [Google Scholar] [CrossRef] [Green Version]
  169. Cai, X.-T.; Xu, P.; Zhao, P.-X.; Liu, R.; Yu, L.-H.; Xiang, C.-B. Arabidopsis ERF109 mediates cross-talk between jasmonic acid and auxin biosynthesis during lateral root formation. Nat. Commun. 2014, 5, 1–13. [Google Scholar] [CrossRef] [PubMed]
  170. Xu, P.; Zhao, P.-X.; Cai, X.-T.; Mao, J.-L.; Miao, Z.-Q.; Xiang, C.-B. Integration of jasmonic acid and ethylene into auxin signaling in root development. Front. Plant Sci. 2020, 11, 271. [Google Scholar] [CrossRef]
  171. Zhang, T.; Poudel, A.N.; Jewell, J.B.; Kitaoka, N.; Staswick, P.; Matsuura, H.; Koo, A.J. Hormone crosstalk in wound stress response: Wound-inducible amidohydrolases can simultaneously regulate jasmonate and auxin homeostasis in Arabidopsis thaliana. J. Exp. Bot. 2016, 67, 2107–2120. [Google Scholar] [CrossRef] [Green Version]
  172. Jiang, Y.; Liang, G.; Yang, S.; Yu, D. Arabidopsis WRKY57 functions as a node of convergence for jasmonic acid-and auxin-mediated signaling in jasmonic acid-induced leaf senescence. Plant Cell 2014, 26, 230–245. [Google Scholar] [CrossRef] [Green Version]
  173. Jiang, Y.; Liang, G.; Yu, D. Activated expression of WRKY57 confers drought tolerance in Arabidopsis. Mol. Plant 2012, 5, 1375–1388. [Google Scholar] [CrossRef] [Green Version]
  174. Ahmadi, F.; Karimi, K.; Struik, P. Effect of exogenous application of methyl jasmonate on physiological and biochemical characteristics of Brassica napus L. cv. Talaye under salinity stress. South. Afr. J. Bot. 2018, 115, 5–11. [Google Scholar] [CrossRef]
  175. Valenzuela-Riffo, F.; Zúñiga, P.E.; Morales-Quintana, L.; Lolas, M.; Cáceres, M.; Figueroa, C.R. Priming of defense systems and upregulation of MYC2 and JAZ1 genes after botrytis cinerea inoculation in methyl Jasmonate-treated strawberry fruits. Plants 2020, 9, 447. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Zuñiga, P.E.; Castañeda, Y.; Arrey-Salas, O.; Fuentes, L.; Aburto, F.; Figueroa, C.R. Methyl Jasmonate Applications from Flowering to Ripe Fruit Stages of Strawberry (Fragaria × ananassa ‘Camarosa’) Reinforce the Fruit Antioxidant Response at Post-harvest. Front. Plant Sci. 2020, 11, 538. [Google Scholar] [CrossRef] [PubMed]
  177. Maksymiec, W.; Krupa, Z. The in vivo and in vitro influence of methyl jasmonate on oxidative processes in Arabidopsis thaliana leaves. Acta Physiol. Plant 2002, 24, 351–357. [Google Scholar] [CrossRef]
  178. Popova, L.; Ananieva, E.; Hristova, V.; Christov, K.; Georgieva, K.; Alexieva, V.; Stoinova, Z. Salicylic acid-and methyl jasmonate-induced protection on photosynthesis to paraquat oxidative stress. Bulg J. Plant Physiol. 2003, 133, 152. [Google Scholar]
  179. Singh, I.; Shah, K. Exogenous application of methyl jasmonate lowers the effect of cadmium-induced oxidative injury in rice seedlings. Phytochemistry 2014, 108, 57–66. [Google Scholar] [CrossRef]
  180. Jung, S. Effect of chlorophyll reduction in Arabidopsis thaliana by methyl jasmonate or norflurazon on antioxidant systems. Plant Physiol. Biochem. 2004, 42, 225–231. [Google Scholar] [CrossRef]
  181. Turan, M.A.; Elkarim, A.H.A.; Taban, N.; Taban, S. Effect of salt stress on growth, stomatal resistance, proline and chlorophyll concentrations on maize plant. Afr. J. Agric. Res. 2009, 4, 893–897. [Google Scholar] [CrossRef]
  182. Shahbaz, M.; Ashraf, M.; Akram, N.A.; Hanif, A.; Hameed, S.; Joham, S.; Rehman, R. Salt-induced modulation in growth, photosynthetic capacity, proline content and ion accumulation in sunflower (Helianthus annuus L.). Acta Physiol. Plant 2011, 33, 1113–1122. [Google Scholar] [CrossRef]
  183. Fedina, I.; Tsonev, T. Effect of pretreatment with methyl jasmonate on the response of Pisum sativum to salt stress. J. Plant Physiol. 1997, 151, 735–740. [Google Scholar] [CrossRef]
  184. Velitchkova, M.; Fedina, I. Response of photosynthesis of Pisum sativum to salt stress as affected by methyl jasmonate. Photosynthetica 1998, 35, 89–97. [Google Scholar] [CrossRef]
  185. Del Amor, F.M.; Cuadra-Crespo, P. Alleviation of salinity stress in broccoli using foliar urea or methyl-jasmonate: Analysis of growth, gas exchange, and isotope composition. Plant Growth Regul. 2011, 63, 55–62. [Google Scholar] [CrossRef]
  186. Sadeghipour, O. Amelioration of salinity tolerance in cowpea plants by seed treatment with methyl jasmonate. Legume Res. Int. J. 2017, 40. [Google Scholar] [CrossRef] [Green Version]
  187. Yuan, F.; Liang, X.; Li, Y.; Yin, S.; Wang, B. Methyl jasmonate improves tolerance to high salt stress in the recretohalophyte Limonium bicolor. Funct. Plant Biol. 2019, 46, 82–92. [Google Scholar] [CrossRef] [PubMed]
  188. Jiang, M.; Xu, F.; Peng, M.; Huang, F.; Meng, F. Methyl jasmonate regulated diploid and tetraploid black locust (Robinia pseudoacacia L.) tolerance to salt stress. Acta Physiol. Plant 2016, 38, 106. [Google Scholar] [CrossRef]
  189. Talebi, M.; Moghaddam, M.; Pirbalouti, A.G. Methyl jasmonate effects on volatile oil compounds and antioxidant activity of leaf extract of two basil cultivars under salinity stress. Acta Physiol. Plant 2018, 40, 1–11. [Google Scholar] [CrossRef]
  190. Yu, X.; Fei, P.; Xie, Z.; Zhang, W.; Zhao, Q.; Zhang, X. Effects of methyl jasmonate on growth, antioxidants, and carbon and nitrogen metabolism of Glycyrrhiza uralensis under salt stress. Biol. Plant 2019, 63, 89–96. [Google Scholar] [CrossRef]
  191. Sheyhakinia, S.; Bamary, Z.; Einali, A.; Valizadeh, J. The induction of salt stress tolerance by jasmonic acid treatment in roselle (Hibiscus sabdariffa L.) seedlings through enhancing antioxidant enzymes activity and metabolic changes. Biologia 2020, 75, 681–692. [Google Scholar] [CrossRef]
  192. Ali, A.Y.A.; Ibrahim, M.E.H.; Zhou, G.; Nimir, N.E.A.; Jiao, X.; Zhu, G.; Elsiddig, A.M.I.; Suliman, M.S.E.; Elradi, S.B.M.; Yue, W. Exogenous jasmonic acid and humic acid increased salinity tolerance of sorghum. Agron. J. 2020, 112, 871–884. [Google Scholar] [CrossRef]
  193. Sheteiwy, M.S.; Shao, H.; Qi, W.; Daly, P.; Sharma, A.; Shaghaleh, H.; Hamoud, Y.A.; El-Esawi, M.A.; Pan, R.; Wan, Q. Seed priming and foliar application with jasmonic acid enhance salinity stress tolerance of soybean (Glycine max L.) seedlings. J. Sci. Food Agric. 2021, 101, 2027–2041. [Google Scholar] [CrossRef]
  194. Takahashi, H.; Kanayama, Y.; Zheng, M.S.; Kusano, T.; Hase, S.; Ikegami, M.; Shah, J. Antagonistic interactions between the SA and JA signaling pathways in Arabidopsis modulate expression of defense genes and gene-for-gene resistance to cucumber mosaic virus. Plant Cell Physiol. 2004, 45, 803–809. [Google Scholar] [CrossRef] [PubMed]
  195. Yasir, M.; He, S.; Sun, G.; Geng, X.; Pan, Z.; Gong, W.; Jia, Y.; Du, X. A Genome-Wide Association Study revealed key SNPs/genes associated with salinity stress tolerance in upland cotton. Genes 2019, 10, 829. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  196. Hossain, H.; Rahman, M.; Alam, M.; Singh, R. Mapping of quantitative trait loci associated with reproductive-stage salt tolerance in rice. J. Agron. Crop. Sci. 2015, 201, 17–31. [Google Scholar] [CrossRef]
  197. Quan, R.; Wang, J.; Hui, J.; Bai, H.; Lyu, X.; Zhu, Y.; Zhang, H.; Zhang, Z.; Li, S.; Huang, R. Improvement of salt tolerance using wild rice genes. Front. Plant Sci. 2018, 8, 2269. [Google Scholar] [CrossRef] [Green Version]
  198. Ahmadi-Ochtapeh, H.; Soltanloo, H.; Ramezanpour, S.; Naghavi, M.; Nikkhah, H.; Rad, S.Y. QTL mapping for salt tolerance in barley at seedling growth stage. Biol. Plant 2015, 59, 283–290. [Google Scholar] [CrossRef]
  199. Fan, Y.; Zhou, G.; Shabala, S.; Chen, Z.-H.; Cai, S.; Li, C.; Zhou, M. Genome-wide association study reveals a new QTL for salinity tolerance in barley (Hordeum vulgare L.). Front. Plant Sci. 2016, 7, 946. [Google Scholar] [CrossRef] [Green Version]
  200. Díaz De León, J.L.; Escoppinichi, R.; Geraldo, N.; Castellanos, T.; Mujeeb-Kazi, A.; Röder, M.S. Quantitative trait loci associated with salinity tolerance in field grown bread wheat. Euphytica 2011, 181, 371–383. [Google Scholar] [CrossRef]
  201. Xu, Y.; Li, S.; Li, L.; Zhang, X.; Xu, H.; An, D. Mapping QTL s for salt tolerance with additive, epistatic and QTL × treatment interaction effects at seedling stage in wheat. Plant Breed. 2013, 132, 276–283. [Google Scholar] [CrossRef]
  202. Vadez, V.; Krishnamurthy, L.; Thudi, M.; Anuradha, C.; Colmer, T.D.; Turner, N.C.; Siddique, K.H.M.; Gaur, P.M.; Varshney, R.K. Assessment of ICCV 2 × JG 62 chickpea progenies shows sensitivity of reproduction to salt stress and reveals QTL for seed yield and yield components. Mol. Breed. 2012, 30, 9–21. [Google Scholar] [CrossRef] [Green Version]
  203. Li, D.; Dossa, K.; Zhang, Y.; Wei, X.; Wang, L.; Zhang, Y.; Liu, A.; Zhou, R.; Zhang, X. GWAS uncovers differential genetic bases for drought and salt tolerances in sesame at the germination stage. Genes 2018, 9, 87. [Google Scholar] [CrossRef] [Green Version]
  204. Sun, Z.; Li, H.; Zhang, Y.; Li, Z.; Ke, H.; Wu, L.; Zhang, G.; Wang, X.; Ma, Z. Identification of SNPs and candidate genes associated with salt tolerance at the seedling stage in cotton (Gossypium hirsutum L.). Front. Plant Sci. 2018, 9, 1011. [Google Scholar] [CrossRef] [Green Version]
  205. Yuan, Y.; Xing, H.; Zeng, W.; Xu, J.; Mao, L.; Wang, L.; Feng, W.; Tao, J.; Wang, H.; Zhang, H.; et al. Genome-wide association and differential expression analysis of salt tolerance in Gossypium hirsutum L at the germination stage. BMC Plant Biol. 2019, 19, 394. [Google Scholar] [CrossRef]
  206. Do, T.D.; Vuong, T.D.; Dunn, D.; Clubb, M.; Valliyodan, B.; Patil, G.; Chen, P.; Xu, D.; Nguyen, H.T.; Shannon, J.G. Identification of new loci for salt tolerance in soybean by high-resolution genome-wide association mapping. BMC Genom. 2019, 20, 318. [Google Scholar] [CrossRef]
  207. Hazzouri, K.M.; Khraiwesh, B.; Amiri, K.; Pauli, D.; Blake, T.; Shahid, M.; Mullath, S.K.; Nelson, D.; Mansour, A.L.; Salehi-Ashtiani, K. Mapping of HKT1; 5 gene in barley using GWAS approach and its implication in salt tolerance mechanism. Front. Plant Sci. 2018, 9, 156. [Google Scholar] [CrossRef] [Green Version]
  208. Patishtan, J.; Hartley, T.N.; Fonseca de Carvalho, R.; Maathuis, F.J. Genome-wide association studies to identify rice salt-tolerance markers. Plant Cell Environ. 2018, 41, 970–982. [Google Scholar] [CrossRef]
  209. Luo, X.; Wang, B.; Gao, S.; Zhang, F.; Terzaghi, W.; Dai, M. Genome-wide association study dissects the genetic bases of salt tolerance in maize seedlings. J. Integr. Plant Biol. 2019, 61, 658–674. [Google Scholar] [CrossRef] [Green Version]
  210. Shim, J.S.; Jung, C.; Lee, S.; Min, K.; Lee, Y.W.; Choi, Y.; Lee, J.S.; Song, J.T.; Kim, J.K.; Choi, Y.D. A t MYB 44 regulates WRKY 70 expression and modulates antagonistic interaction between salicylic acid and jasmonic acid signaling. Plant J. 2013, 73, 483–495. [Google Scholar] [CrossRef]
  211. Kan, G.; Zhang, W.; Yang, W.; Ma, D.; Zhang, D.; Hao, D.; Hu, Z.; Yu, D. Association mapping of soybean seed germination under salt stress. Mol. Genet. Genom. 2015, 290, 2147–2162. [Google Scholar] [CrossRef]
  212. Pantalião, G.F.; Narciso, M.; Guimarães, C.; Castro, A.; Colombari, J.M.; Breseghello, F.; Rodrigues, L.; Vianello, R.P.; Borba, T.O.; Brondani, C. Genome wide association study (GWAS) for grain yield in rice cultivated under water deficit. Genetica 2016, 144, 651–664. [Google Scholar] [CrossRef]
  213. Hu, S.; Sanchez, D.L.; Wang, C.; Lipka, A.E.; Yin, Y.; Gardner, C.A.; Lübberstedt, T. Brassinosteroid and gibberellin control of seedling traits in maize (Zea mays L.). Plant Sci. 2017, 263, 132–141. [Google Scholar] [CrossRef] [Green Version]
  214. To, H.T.M.; Nguyen, H.T.; Dang, N.T.M.; Nguyen, N.H.; Bui, T.X.; Lavarenne, J.; Phung, N.T.P.; Gantet, P.; Lebrun, M.; Bellafiore, S.; et al. Unraveling the Genetic Elements Involved in Shoot and Root Growth Regulation by Jasmonate in Rice Using a Genome-Wide Association Study. Rice 2019, 12, 69. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  215. Delker, C.; Stenzel, I.; Hause, B.; Miersch, O.; Feussner, I.; Wasternack, C. Jasmonate biosynthesis in Arabidopsis thaliana-enzymes, products, regulation. Plant Biol. 2006, 8, 297–306. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  216. An, H.; Liu, K.; Wang, B.; Tian, Y.; Ge, Y.; Zhang, Y.; Tang, W.; Chen, G.; Yu, J.; Wu, W. Genome-wide association study identifies QTLs conferring salt tolerance in rice. Plant Breed. 2020, 139, 73–82. [Google Scholar] [CrossRef]
  217. Huang, K.; Peng, L.; Liu, Y.; Yao, R.; Liu, Z.; Li, X.; Yang, Y.; Wang, J. Arabidopsis calcium-dependent protein kinase AtCPK1 plays a positive role in salt/drought-stress response. Biochem. Biophys. Res. Commun. 2018, 498, 92–98. [Google Scholar] [CrossRef] [PubMed]
  218. Coca, M.; San Segundo, B. AtCPK1 calcium-dependent protein kinase mediates pathogen resistance in Arabidopsis. Plant J. 2010, 63, 526–540. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  219. Li, J.; Brader, G.; Palva, E.T. The WRKY70 transcription factor: A node of convergence for jasmonate-mediated and salicylate-mediated signals in plant defense. Plant Cell 2004, 16, 319–331. [Google Scholar] [CrossRef] [Green Version]
  220. Chen, K.; Wang, Y.; Zhang, R.; Zhang, H.; Gao, C. CRISPR/Cas genome editing and precision plant breeding in agriculture. Annu. Rev. Plant Biol. 2019, 70, 667–697. [Google Scholar] [CrossRef]
  221. Wang, Y.; Cheng, X.; Shan, Q.; Zhang, Y.; Liu, J.; Gao, C.; Qiu, J.-L. Simultaneous editing of three homoeoalleles in hexaploid bread wheat confers heritable resistance to powdery mildew. Nat. Biotechnol. 2014, 32, 947–951. [Google Scholar] [CrossRef]
  222. Svitashev, S.; Schwartz, C.; Lenderts, B.; Young, J.K.; Mark Cigan, A. Genome editing in maize directed by CRISPR—Cas9 ribonucleoprotein complexes. Nat. Commun. 2016, 7, 13274. [Google Scholar] [CrossRef]
  223. Cai, Y.; Chen, L.; Liu, X.; Sun, S.; Wu, C.; Jiang, B.; Han, T.; Hou, W. CRISPR/Cas9-mediated genome editing in soybean hairy roots. PLoS ONE 2015, 10, e0136064. [Google Scholar] [CrossRef]
  224. Stuttmann, J.; Barthel, K.; Martin, P.; Ordon, J.; Erickson, J.L.; Herr, R.; Ferik, F.; Kretschmer, C.; Berner, T.; Keilwagen, J. Highly efficient multiplex editing: One-shot generation of 8x Nicotiana benthamiana and 12x Arabidopsis mutants. Plant J. 2021. [Google Scholar] [CrossRef]
  225. Liu, L.; Zhang, J.; Xu, J.; Li, Y.; Guo, L.; Wang, Z.; Zhang, X.; Zhao, B.; Guo, Y.-D.; Zhang, N. CRISPR/Cas9 targeted mutagenesis of SlLBD40, a lateral organ boundaries domain transcription factor, enhances drought tolerance in tomato. Plant Sci. 2020, 301, 110683. [Google Scholar] [CrossRef]
Figure 1. Overview of the jasmonate (JA) pathway including the major molecular players involved in biosynthesis, signaling, and catabolism. Black, blue, and green fonts show the main metabolites, enzymes, and proteins, respectively, for each section of the pathway. Scheme made based on [49,58,80]. For more details, see the text.
Figure 1. Overview of the jasmonate (JA) pathway including the major molecular players involved in biosynthesis, signaling, and catabolism. Black, blue, and green fonts show the main metabolites, enzymes, and proteins, respectively, for each section of the pathway. Scheme made based on [49,58,80]. For more details, see the text.
Ijms 22 03082 g001
Figure 2. Genome-wide association study (GWAS) pipeline to identify jasmonate pathway-associated genes involved in the salt stress response and salt tolerance in crops. GBS, SSI, GLM, and MLM correspond to genotyping by sequencing, salt stress index, general linear model, and mixed linear model, respectively. For more details, see the text.
Figure 2. Genome-wide association study (GWAS) pipeline to identify jasmonate pathway-associated genes involved in the salt stress response and salt tolerance in crops. GBS, SSI, GLM, and MLM correspond to genotyping by sequencing, salt stress index, general linear model, and mixed linear model, respectively. For more details, see the text.
Ijms 22 03082 g002
Table 1. Participation of jasmonate (JA) pathway-associated molecular components in salt stress responses of different plants.
Table 1. Participation of jasmonate (JA) pathway-associated molecular components in salt stress responses of different plants.
ProteinFunctionSalt Stress ResponseSpeciesReferences
Lipoxygenase3 (LOX3)JA biosynthesisInduced under salt stress. Methyl jasmonate (MeJA) rescued the salt sensitivity phenotype of the lox3 mutantArabidopsis thaliana[91]
Allene oxide cyclase (AOC)JA biosynthesisConstitutive expression enhances tolerance to salt stressTriticum aestivum
A. thaliana
[92]
cpm2 and hebiba mutants display salt toleranceOryza sativa[93]
MYC2JA signalingTranscriptional activator of the salt- and abscisic acid(ABA)-responsive gene RD22A. thaliana[94,96]
An important role in salt-mediated JA-dependent inhibition of cell elongation in the elongation zone of primary rootA. thaliana[95]
Its salt stress-mediated activation by MAPK cascade regulates proline biosynthesisA. thaliana[97]
Mediates the negative regulation of ABA signaling by PUB10, which acts as a positive regulator for salt and osmotic stress toleranceA. thaliana[98,99]
Jasmonate ZIM-domain (JAZ)JA signalingInduced under salt stressA. thaliana
Gossypium hirsutum
Solanum lycopersicum
[95]
PnJAZ1 inhibited expression of ABA-dependent genes related to seed germination and shoot growth under high salt conditionsA. thaliana
Physcomitrella patens
[110]
OsJAZ9 and OsJAZ8 overexpression enhanced salt toleranceO. sativa[104]
Heterologous expression of GsJAZ2 and MdJAZ2 enhanced tolerance to salinityA. thaliana[114]
Overexpression of TdTIFY11a variants confer salt tolerance to Arabidopsis seedlingsA. thaliana[103]
GaJAZ1 overexpression significantly increased salt toleranceG. hirsutum[111]
GbWRKY1 overexpression negatively affects salt tolerance through an interaction network involving JAZ1 and ABI1A. thaliana[115]
Cytochrome P450 family (CYP94C2b)JA catabolismOsCYP94C2b overexpression enhanced viability under salt conditions and delayed the salt stress-induced leaf senescenceO. sativa[112]
Table 2. Molecular and physiological effects of the jasmonate (JA) crosstalk with abscisic acid (ABA), ethylene (ET), salicylic acid (SA), gibberellins (GA), cytokinin (CK), and auxin (AUX) in different plants under salt stress conditions. For more details, see the text.
Table 2. Molecular and physiological effects of the jasmonate (JA) crosstalk with abscisic acid (ABA), ethylene (ET), salicylic acid (SA), gibberellins (GA), cytokinin (CK), and auxin (AUX) in different plants under salt stress conditions. For more details, see the text.
CrosstalkMolecular/Physiological EffectsSpeciesReferences
JA-ABAJA and ABA applications in conjunction activate the antioxidant mechanism against salt stressFragaria × ananassa[117]
Synergistic effect on the inhibition of seed germination under salinity conditionsArabidopsis thaliana[110]
Synergism in the salt tolerance mediated by phytochrome A and BNicotiana tabacum[118]
JA–ETSynergistic upregulation of AtERF1 required to induce salt toleranceA. thaliana[133]
Synergistic upregulation of RSOsPR10 which promotes root growth and increases salt toleranceOryza sativa[136]
Synergistic upregulation of GmCYP82A3 which enhances salinity toleranceGlycine max
Nicotiana benthamiana
[138]
JA–SAMethyl jasmonate (MeJA) and SA application increases antioxidant activity and reduced the Na+/K+ ratioF. × ananassa[145]
JA and SA application protects plants from salt-induced damage and improves plant performance under salt conditionsG. max[146]
JA and SA application stimulates H+-ATPase activity of tonoplast, nutrient uptake, and salt toleranceG. max[147]
JA-GAJA application decreases GA content in salt-stressed plantsOcimum basilicum[155]
MeJA application suppresses the transcript levels of the GA-responsive genes NtPIF3, NtGAST1, and NtGSAT4Nitraria tangutorum[156]
JA-CKMeJA application prevents the salinity-induced decline of endogenous CK by reducing the cytokinin oxidase enzymatic activity and its related gene expressionTriticum aestivum
Prunus dulcis
[163]
JA-AUXAn opposite function of JAZ4/8 and IAA29 repressors on the regulation of WRKY57. Constitutive activation of WRKY57 in adt mutant confers salt toleranceA. thaliana[172,173]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Delgado, C.; Mora-Poblete, F.; Ahmar, S.; Chen, J.-T.; Figueroa, C.R. Jasmonates and Plant Salt Stress: Molecular Players, Physiological Effects, and Improving Tolerance by Using Genome-Associated Tools. Int. J. Mol. Sci. 2021, 22, 3082. https://doi.org/10.3390/ijms22063082

AMA Style

Delgado C, Mora-Poblete F, Ahmar S, Chen J-T, Figueroa CR. Jasmonates and Plant Salt Stress: Molecular Players, Physiological Effects, and Improving Tolerance by Using Genome-Associated Tools. International Journal of Molecular Sciences. 2021; 22(6):3082. https://doi.org/10.3390/ijms22063082

Chicago/Turabian Style

Delgado, Celia, Freddy Mora-Poblete, Sunny Ahmar, Jen-Tsung Chen, and Carlos R. Figueroa. 2021. "Jasmonates and Plant Salt Stress: Molecular Players, Physiological Effects, and Improving Tolerance by Using Genome-Associated Tools" International Journal of Molecular Sciences 22, no. 6: 3082. https://doi.org/10.3390/ijms22063082

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop