You are on page 1of 13

The Crop Journal 10 (2022) 13–25

Contents lists available at ScienceDirect

The Crop Journal


journal homepage: www.elsevier.com/locate/cj

Salt tolerance in rice: Physiological responses and molecular


mechanisms
Citao Liu a, Bigang Mao b, Dingyang Yuan b, Chengcai Chu c, Meijuan Duan a,⇑
a
College of Agriculture, Hunan Agricultural University, Changsha 410128, Hunan, China
b
State Key Laboratory of Hybrid Rice, Hunan Hybrid Rice Research Center, Changsha 410125, Hunan, China
c
State Key Laboratory of Plant Genomics, Institute of Genetics and Developmental Biology, The Innovative Academy for Seed Design, Chinese Academy of Sciences, Beijing
100101, China

a r t i c l e i n f o a b s t r a c t

Article history: Crop yield loss due to soil salinization is an increasing threat to agriculture worldwide. Salt stress dras-
Received 30 November 2020 tically affects the growth, development, and grain productivity of rice (Oryza sativa L.), and the improve-
Revised 21 January 2021 ment of rice tolerance to salt stress is a desirable approach for meeting increasing food demand. The main
Accepted 17 March 2021
contributors to salt toxicity at a global scale are Na+ and Cl ions, which affect up to 50% of irrigated soils.
Available online 6 April 2021
Plant responses to salt stress occur at the organismic, cellular, and molecular levels and are pleiotropic,
involving (1) maintenance of ionic homeostasis, (2) osmotic adjustment, (3) ROS scavenging, and (4)
Keywords:
nutritional balance. In this review, we discuss recent research progress on these four aspects of plant
Salt stress
Rice (Oryza sativa L.)
physiological response, with particular attention to hormonal and gene expression regulation and salt
Salt tolerance genes tolerance signaling pathways in rice. The information summarized here will be useful for accelerating
Physiological response the breeding of salt-tolerant rice.
Salt signal transduction Ó 2021 Crop Science Society of China and Institute of Crop Science, CAAS. Production and hosting by
Elsevier B.V. on behalf of KeAi Communications Co., Ltd. This is an open access article under the CC BY-NC-
ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14
2. Effects of salt stress on rice . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14
3. Mechanisms underlying rice salt stress tolerance . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14
3.1. Regulation of ionic balance. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14
3.2. Osmotic adjustment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16
3.3. ROS scavenging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16
3.4. Nutrient imbalance . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16
3.5. Developmental adjustment. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16
4. Salt stress signaling in rice . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17
4.1. The SOS signaling pathway. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17
4.2. Hormonal regulation during salt stress . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17
4.2.1. Abscisic acid . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17
4.2.2. Ethylene . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19
4.2.3. Gibberellic acid . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19
4.2.4. Auxin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19
4.2.5. Cytokinins . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19
4.2.6. Brassinosteroids . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20
4.2.7. Jasmonic acid . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20
4.2.8. Salicylic acid . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20
4.3. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20
5. Future perspectives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20

⇑ Corresponding author.
E-mail address: duanmeijuan@163.com (M. Duan).

https://doi.org/10.1016/j.cj.2021.02.010
2214-5141/Ó 2021 Crop Science Society of China and Institute of Crop Science, CAAS. Production and hosting by Elsevier B.V. on behalf of KeAi Communications Co., Ltd.
This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
C. Liu, B. Mao, D. Yuan et al. The Crop Journal 10 (2022) 13–25

Declaration of competing interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20


CRediT authorship contribution statement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 21
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 21

1. Introduction Calvin cycle and other pathways [27,28]. Excess Na+ in the cyto-
plasm also interferes with the uptake and transport of potassium
Soil salinity is an environmental factor that has severe negative (K+) and other macro- and micronutrients, including nitrogen (N),
effects on seed germination, crop growth, and productivity [1], and phosphorus (P), potassium (K+), calcium (Ca2+) and zinc (Zn2+)
it is becoming a serious problem worldwide [1,2]. Of the 230 mil- [13,29–32]. In addition to osmotic and ionic stresses, salt stress
lion hectares of farmland currently in use around the world, 20% is causes the cellular accumulation of reactive oxygen species
affected by salt, and this percentage increases every year as a result (ROS), which can severely damage cellular structures and macro-
of inappropriate crop irrigation practices, overfertilization, and molecules such as DNA, lipids, and enzymes [33,34].
excessive plowing, as well as natural causes such as salt intrusion Under salt stress, plants must adjust the physiological and bio-
into coastal zones resulting from rising sea levels [2–4]. The threats chemical processes involved in the regulation of ion and osmotic
of salinity are more obvious in arid and semiarid regions where homeostasis, oxidative stress, and nutritional balance [1,25].
limited rainfall, high evapotranspiration, and extreme tempera-
tures coupled with poor water and soil management are the main 3. Mechanisms underlying rice salt stress tolerance
contributing factors [5,6]. In China, there are as many as 100 mil-
lion hectares of saline farmland [7]. Globally, salinity is a signifi- 3.1. Regulation of ionic balance
cant abiotic stress that affects one-quarter to one-third of the
crop productivity of on agricultural soils [8]. As the world popula- Salt stress is commonly caused by high concentrations of Na+
tion increases, more food is needed to meet the rising demand, and and Cl in the soil [9,35]. Na+ and K+ are imported into the cell
the use of saline land to make up for the resulting food shortage is using the same suite of transporters, and the two cations compete
becoming more urgent [9]. Scientists are attempting to increase with each other [36]. K+ is important for the catalytic activities of
food production by 70% in order to avoid the risk of food shortages many central enzymes, and excess Na+ competes with K+ for
for 9.3 billion people by 2050 [10]. Rice, an important cereal crop uptake across the plasma membranes of plant cells [37]. K+ is also
that feeds half of the world’s population, is sensitive to salt stress necessary for osmoregulation and protein synthesis, the preserva-
[11,12]. Improvement of rice salt stress tolerance is therefore of tion of cell turgor, and optimal photosynthetic activity [6,38,39].
paramount importance for global food security. The maintenance of cellular Na+/K+ homeostasis is therefore a cru-
Combating salt stress to increase rice production requires a cial factor that determines the plant’s ability to survive during salt
thorough understanding of physiology, biochemistry, metabolism, stress [1,25].
and gene expression under salt stress conditions. In this review, we Mechanisms to reduce cytoplasmic Na+ include restriction of
systematically summarize the current understanding of salt stress Na+ uptake, increase of Na+ efflux, and compartmentalization of
effects on rice, the mechanisms that underlie rice salt stress toler- Na+ in the vacuole [1,40]. The rice plasma membrane Na+/H+ anti-
ance, and salt stress signaling in rice. This information may be porter (OsSOS1) excludes Na+ from the shoot, promoting a lower
helpful for developing salt-tolerant rice varieties. cellular Na+/K+ ratio and increasing salt tolerance [41,42]. The vac-
uolar Na+/H+ antiporters OsNHX1, OsNHX2, OsNHX3, OsNHX4,
2. Effects of salt stress on rice OsNHX5, and OsARP/OsCTP play important roles in the vacuolar
compartmentalization of Na+ and K+ that accumulate in the cyto-
Plant morphological changes under high salt conditions include plasm and thereby determine rice salt tolerance [43–45]. Members
impaired root system establishment, leaf rolling, chlorosis, fewer of the rice high-affinity K+ transporter (HKT) family, OsHKT1;1,
tillers per plant, reduced biomass, shorter plant height, lower thou- OsHKT1;4/OsHKT7, and SKC1/OsHKT1;5/OsHKT8, help to reduce
sand grain weight, fewer spikelets per panicle, and more sterile flo- Na+ accumulation in leaves during salt stress [46–48]. OsHKT1;1,
rets, ultimately leading to reductions in harvest index and grain OsHKT2;1, OsHKT2;3/OsHKT3, OsKAT1, OsKAT2, OsHAK5, and
yield [13–16]. OsHAK21/qSE3 transport Na+ or both Na+ and K+, helping to main-
Salt stress affects plant physiology and biochemistry at all tain Na+/K+ homeostasis in the cytoplasm and regulate rice
developmental stages from germination to senescence [6,17,18]. response to salt stress [46,49–58]. The vacuolar H+-translocating
Salt stress has both osmotic and ionic or ion-toxicity effects on inorganic pyrophosphatase OsVP1 pumps H+ from the cytosol into
plants, ultimately causing oxidative stress and nutrient depletion the vacuole, increasing the electrochemical potential gradient of H+
in plant cells [6,19]. Continuous salt stress reduces plant cell turgor between the cytoplasm and vacuole, thereby promoting the
pressure, which in turn reduces cell growth, and plants must exchange of Na+/H+ and improving rice tolerance to salt stress
osmotically adjust to maintain cell expansion and growth [20– [43]. Researchers have demonstrated that Cl induces deficiencies
22]. Osmotic stress (caused by lower water potential of the exter- in key macronutrients (e.g., N and S) because the uptake of NO 3
nal solution) is rapidly sensed by the plant soon after exposure to and SO2 4 is mediated by the same (non-selective) anion trans-
saline conditions and leads to plant water and solute deficits [9,21– porters as Cl [23,59]. OsCLC1, OsCLC-1, and OsCLC-2 encode rice
23]. Osmotic stress also results in rapid stomatal closure, which chloride channel proteins that promote plant growth under ionic
reduces the plant’s ability to assimilate CO2 and inhibits photosyn- stress and improve salt stress tolerance [60,61] (Table 1; Fig. 1).
thesis [24]. Ionic stress causes sodium (Na+) and chlorine (Cl) A number of genes influence rice salt tolerance by regulating
accumulation in plant cells, eventually resulting in premature leaf the expression of HKT, NHX, and CLC genes. These include tran-
senescence and even plant death [1,25,26]. The most common scription factors such as OsMYBc, OsbZIP71, OsNF-YC13, and the
explanation for Na+ toxicity is that it has an inhibitory effect on sucrose nonfermenting 1-related protein kinase2 (SnRK2) SAPK4,
enzyme activities, negatively affecting metabolism, including the which directly activate the expression of K+/Na+ transporters,
14
C. Liu, B. Mao, D. Yuan et al. The Crop Journal 10 (2022) 13–25

Table 1
Proteins that function in Na+/K+ homeostasis during salt stress.

Family Effect on Selectivity Relevant genes Cellular References


tolerance localization
Na+/H+ antiporter Positive Na+ OsNHX1, OsNHX2, OsNHX3, OsNHX4, Tonoplast Liu et al. [43], Fukuda et al. [44], Uddin et al. [45]
OsNHX5, OsARP/OsCTP
+
Positive Na OsSOS1 Plasma El Mahi et al. [41], Martínez-Atienza et al. [42]
membrane
Na+-transporter Positive Na+ OsHKT1;1, OsHKT2;3/OsHKT3, OsHKT1;4/ Plasma Wang et al. [46], Suzuki et al. [47], Ren et al. [48],
OsHKT7, SKC1/OsHKT1;5/OsHKT8 membrane Huang et al. [51]
Negative Na+ OsHKT2;1 Plasma Ardie et al. [49], Horie et al. [50]
membrane
Potassium transporter Positive Na+, K+ OsKAT1, OsKAT2, OsHAK5, OsHAK21/qSE3 Plasma Obata et al. [53], Hwang et al. [54], Yang et al. [55],
membrane Horie et al. [56], He et al. [58]
+ +
H -pyrophosphatase Positive H OsVP1 Tonoplast Liu et al. [53]
Cl channel Positive Cl OsCLC1, OsCLC-1, OsCLC-2 Tonoplast Nakamura et al. [60], Diedhiou and Golldack. [61]
Transcription factor Positive – OsMYBc, OsbZIP71, OsNF-YC13 Nucleus Wang et al. [46], Liu et al. [62], Manimaran et al.
[63]
Protein kinase Positive – OsSAPK4, OsSOS2/OsCIPK24, OsSOS3/OsCBL4 – El Mahi et al. [41], Diédhiou et al. [64]
Negative – OsMAPK33 – Lee et al. [65]

‘‘–” Not reported.

Fig. 1. Genes involved in the regulation of salt tolerance in rice, including Na+/K+ ion homeostasis (upper right), osmotic adjustment (upper left), ROS scavenging (lower left),
and nutritional balance (lower right). The middle yellow layer contains functional genes whose encoded proteins directly protect membranes and macromolecules under
stress conditions. The outer gray layer contains regulatory genes whose products protect plants from adversity by regulating the expression of functional genes under stress
conditions.

thereby improving the K+/Na+ ratio and positively regulating salt vating the Na+/H+ antiporter OsSOS1 and promoting rice tolerance
tolerance [46,62–64]. The myristoylated Ca2+-binding protein to salt stress [41,66]. The mitogen-activated protein kinase (MAPK)
OsSOS3/OsCBL4 phosphorylates OsSOS2/OsCIPK24, thereby acti- OsMAPK33 plays a negative role in salt tolerance by promoting

15
C. Liu, B. Mao, D. Yuan et al. The Crop Journal 10 (2022) 13–25

higher sodium uptake into cells and thereby lowering the K+/Na+ (hydrogen peroxide), and OH (hydroxyl radical) [9,34]. Although
ratio [65] (Table 1; Fig. 1). The results above suggest that the mod- low ROS concentrations can function as a signal to activate salt
ulation of Na+/K+ homeostasis under salt stress may provide an stress responses, excess ROS accumulation causes phytotoxic reac-
effective way to improve salt tolerance in rice. tions including DNA mutation, protein degradation, and the perox-
idation of carbohydrates and lipids [1,34]. Plants use enzymatic
3.2. Osmotic adjustment and nonenzymatic antioxidants to mitigate ROS stress [34,83,84].
Enzymatic scavengers include nicotinamide adenine dinucleotide
Salt stress also causes osmotic stress and promotes the biosyn- phosphate oxidases (NOXs, also called respiratory burst oxidase
thesis and accumulation of compatible osmolytes such as sugar, homologs [Rbohs]), superoxide dismutase (SOD), ascorbate perox-
proline, glycine betaine, polyamines, and proteins from the late idase (APX), catalase (CAT), glutaredoxin (GRX), glutathione perox-
embryogenesis abundant (LEA) superfamily. These osmolytes play idase (GR), glutathione S-transferase (GST), and glutathione
a dominant role in osmotic adjustment under salt stress by reduc- peroxidases (GPXs) [1,85–90]. Nonenzymatic scavengers include
ing cell osmotic potential and stabilizing proteins and cellular ascorbic acid (ASH), alkaloids, carotenoids, flavonoids, glutathione
structures [1,23]. The proline synthesis genes OsP5CS1 and OsP5CR (GSH), phenolic compounds, and tocopherol [83,91,92]. OsRbohA
increase proline accumulation and improve rice tolerance to salt and OsRbohI are induced by salt treatment, whereas OsRbohB, OsR-
stress [67]. OsTPS1, OsTPS2, OsTPS4, OsTPS5, OsTPS8, and OsTPS9, bohC, OsRbohE, and OsNox6 are repressed [93]. OsMn-SOD1 and
which encode trehalose-6-phosphate synthase (TPS), enhance rice OsCu/Zn-SOD overexpression lines show lower accumulation of
tolerance to cold, salt, and drought stresses by increasing trehalose O2 and H2O2 under salt stress [94,95]. OsAPX2, OsAPX7, OsAPx8,
and proline contents under abiotic stress [68]. OsGMST1, which OsAPXa, and OsAPXb increase APX activity, lower H2O2 and malon-
encodes a monosaccharide transporter, increases monosaccharide dialdehyde (MDA) levels, decrease oxidative stress damage, and
accumulation and improves salt tolerance in plants [69]. The enhance rice tolerance to salt stress [96–99]. OsGST4 is induced
Sugars Will Eventually be Exported Transporters (SWEETs) OsS- by heavy metals, hypoxia, and salt stress in rice [100]. Glutathione
WEET13 and OsSWEET15 regulate the transport and distribution responsive rice glyoxalase II (OsGLYII-2) functions in salinity adap-
of sucrose and maintain sugar homeostasis in rice under drought tation by maintaining better photosynthetic efficiency and increas-
and salinity stresses [70]. Rice glycine betaine is synthesized by ing the antioxidant pool [101]. OsGR2, OsGR3, and OsGRX8 increase
the choline monooxygenase OsCMO and the betaine aldehyde GSH content and enhance tolerance to various abiotic stresses,
dehydrogenase OsBADH1, which enhance rice tolerance to salt including salinity, osmotic, and oxidative stress [99,102–106].
stress by promoting glycine betaine accumulation [71,72]. The The cytosolic dihydroorotate dehydrogenase gene OsDHODH1
LEA genes OsLEA3-2, OsLEA4, OsLEA5, and OsEm1 significantly improves rice tolerance to salt and osmotic stresses [107]. Rice
improve plant tolerance to salt and osmotic stresses [73–76]. GDP-mannose pyrophosphorylase OsVTC1-1 and dehydroascor-
The salt-regulated gene OsSALP1 encodes a small plant-specific bate reductase (OsDHAR) play critical roles in plant salt tolerance
membrane protein that improves salt tolerance by increasing by promoting the ASH scavenging of excess ROS [108,109]. OsGSA1
OsP5CS expression and free proline content under salt stress [77]. encodes a UDP-glucosyltransferase that causes plants to accumu-
The transcription factors OsZFP252, OsZFP179, OsZFP182, OsNF- late flavonoid glycosides, which protect rice against abiotic stress
YC13, OsCOIN, and OsNAC5 increase the amount of free proline [110] (Table 3; Fig. 1).
and soluble sugars, upregulate the expression of stress tolerant A number of genes improve the salt tolerance of rice by regulat-
genes, and enhance rice tolerance to salt stress [63,78–82] (Table 2; ing genes involved in ROS biosynthesis and scavenging pathways.
Fig. 1). The calcium-dependent protein kinases OsCPK4 and OsCPK12
and the receptor-like kinase OsSIK1 promote tolerance to salt
3.3. ROS scavenging stress by reducing the accumulation of ROS [111–113]. The tran-
scription factors OsZFP179, OsZFP182, OsZFP213, OsHBP1b/Osb-
Plant exposure to salt stress can upregulate the production of ZIP3, OsMADS25, OsMyb2, and OsMyb6 positively regulate salt
ROS such as 1O2 (singlet oxygen), O2 (superoxide radical), H2O2
tolerance by increasing ROS-scavenging ability [79,80,114–118],
whereas the zinc-finger proteins DST and DCA1 negatively affect
rice salt tolerance by regulating the transcription of ROS-
Table 2 scavenging genes [119,120] (Table 3; Fig. 1). All these studies sug-
Proteins and metabolites that function in osmotic adjustment during salt stress.
gest that enhancing ROS-scavenging ability can efficiently increase
Group Effect on Relevant genes References the salt tolerance of rice.
tolerance
Proline Positive OsP5CS1, OsP5CR, Sripinyowanich et al.
OsSALP1 [67], Yuan et al. [77]
Trehalose Positive OsTPS1, OsTPS2, Li et al. [68] 3.4. Nutrient imbalance
OsTPS4, OsTPS5,
OsTPS8, OsTPS9
Monosaccharides Positive OsGMST1 Cao et al. [69] Salt stress causes plant nutritional deficiencies due to reduced
SWEETs Positive OsSWEET13, Mathan et al. [70] transport efficiency and decreased uptake of nutrients such as N,
OsSWEET15 P, K, Ca, Zn, and magnesium (Mg) [6,13,29,121]. Mg2+ transport
Glycine betaine Positive OsCMO, OsBADH1 Hasthanasombut et al.
by OsMGT1 in the mature root zone synchronously enhances
[71], Tang et al. [72]
LEA Positive OsLEA3-2, OsLEA4, Duan and Cai. [73], Hu OsHKT1;5 activity, which restricts Na+ accumulation in the shoots
OsLEA5, OsEm1 et al. [74], He et al. [75], and improves salt tolerance [122]. The rice aminotransferase
Yu et al. [76] OsAMTR1 interacts with stress associated protein 1 (OsSAP1) and
Membrane Positive OsSALP1 Yuan et al. [77] regulates abiotic stress responses [123]. The cytokinin type-B
protein
Transcription Positive OsZFP252, Manimaran et al. [63],
response regulator (RR) OsRR22 regulates Zn acquisition by
factor OsZFP179, Xu et al. [78], Sun et al. directly modulating the expression of Zn-regulated transporter
OsZFP182, OsNF- [79], Huang et al. [80], genes and the sensitivity to salt stress [124,125]. OsMADS27 regu-
YC13, OsCOIN, Liu et al. [81], Song et al. lates root development in a NO 3 -dependent manner and modu-
OsNAC5 [82]
lates salt tolerance in rice [126] (Table 4, Fig. 1).
16
C. Liu, B. Mao, D. Yuan et al. The Crop Journal 10 (2022) 13–25

Table 3
Proteins and metabolites that function in ROS homeostasis during salt stress.

Group Effect on Relevant genes References


tolerance
Respiratory burst oxidase homolog (Rboh) Induced OsRbohA, OsRbohI Wang et al. [93]
Repressed OsRbohB, OsRbohC, OsRbohE, OsNox6 Wang et al. [93]
Superoxide dismutase (SOD) Positive OsMn-SOD1, OsCu/Zn-SOD Li et al. [94], Guan et al. [95]
Ascorbate peroxidase (APX) Positive OsAPX2, OsAPX7, OsAPx8, OsAPXa, OsAPXb Zhang et al. [96], Lu et al. [97], Hong et al. [98],
Chou et al. [99]
Glutathione S-transferase (GST) Induced OsGST4 Lee et al. [100]
Positive OsGLYII-2 Lee et al. [100]
Glutathione reductase (GR) Positive OsGR2, OsGR3 Chou et al. [99], Wu et al. [105], Kaminaka et al. [106]
Glutathione peroxidase (GRX) Positive OsGPX1, OsGRX8 Sharma et al. [102], Wu et al. [103], Lima Melo et al. [104]
UDP-glucosyltransferase Positive OsGSA1 Dong et al. [110]
Dihydroorotate dehydrogenase Positive OsDHODH1 Liu et al. [107]
Ascorbic acid (ASH) Positive OsVTC1-1, OsDHAR Qin et al. [108], Ushimaru et al. [109]
Protein kinase Positive OsCPK4, OsCPK12, OsSIK1 Asano et al. [111], Campo et al. [112], Ouyang et al. [113]
Transcription factor Positive OsZFP179, OsZFP182, OsZFP213, Sun et al. [79], Huang et al. [80], Zhang et al. [114],
OsHBP1b/OsbZIP3, OsMADS25, Lakra et al. [115], Xu et al. [116], Yang et al. [117],
OsMyb2, OsMyb6 Tang et al. [118]
Negative DST, DCA1 Cui et al. [119], Huang et al. [120]

4.1. The SOS signaling pathway


Table 4
Proteins that function in nutritional adjustment during salt stress.
Plants use a calcium-dependent protein kinase pathway known
Group Effect on Relevant References as the salt overly sensitive (SOS) pathway for salt stress signaling
tolerance genes
and the development of Na+ tolerance [138,139]. The SOS pathway
Mg transporter Positive OsMGT1 Chen et al. [122]) was the first abiotic stress signaling pathway to be characterized in
Aminotransferase Positive OsAMTR1 Kothari et al. [123]
Regulator of Zn Negative OsRR22 Gao et al. [124], Zhang
plants [139]. It is a major mechanism for the exclusion of Na+ from
transport et al. [125] the cytosol, and its elucidation was a milestone in our understand-
Regulator of NO3 Positive OsMADS27 Chen et al. [126] ing of how plants deal with salt load [140,141]. In this pathway, the
transport calcium-binding protein OsSOS3/CBL4 senses the cytosolic calcium
signal elicited by salt stress, then interacts with and activates a
3.5. Developmental adjustment sucrose non-fermenting-1-related protein kinase-3 (SnRK3),
OsSOS2/OsCIPK24. Activated OsSOS2/OsCIPK24 phosphorylates
Plant response to salinity is the collective outcome of intricate and activates OsSOS1, a Na+/H+ antiporter at the plasma mem-
communications among various processes linked to plant mor- brane, which in turn controls Na+ homeostasis and improves rice
phology, biochemistry, and physiology, as well as the inhibition tolerance to salt stress [41] (Fig. 2).
of growth and photosynthesis and the reduction of grain yield Other Ca2+ signal response proteins also play a role in salt stress
[1,127,128]. Roots take up water and nutrients and have a key role signaling. The calmodulin protein OsMSR2, OsCam1-1, OsCCD1,
in plant growth, development, and survival. The root system is the OsCam1, and the CML protein OsCBL8 act as Ca2+ sensors and
first tissue to perceive salt stress [129]. Transgenic rice lines that improve rice tolerance to salt stress [142–146]. The CDPK/CPK pro-
overexpressed OsAHL1, OsHAL3, and OsMADS25 had greater root teins OsCPK4, OsCPK7, and OsCPK12 help to lower Na+ accumula-
volume under saline conditions and exhibited enhanced salt avoid- tion and maintain osmotic balance under salt stress conditions
ance [116,130,131]. Transgenic lines that overexpressed OsiSAP8 [112,147,148].
and OsCFM2 had higher chlorophyll contents and photosynthetic
rates than wild-type plants under abiotic stress [132,133]. 4.2. Hormonal regulation during salt stress

Downstream of the early salt signaling phase, phytohormone


4. Salt stress signaling in rice
levels change, and the salt-induced signaling cascade ultimately
result in adaptive responses [14]. Phytohormones are crucial
As sessile organisms, plants must cope with abiotic stresses
endogenous chemical signals that coordinate plant growth and
such as soil salinity [19]. To adapt to salt stress, plants have devel-
development under both optimal conditions and environmental
oped various strategies to integrate exogenous salinity stress sig-
challenge [14]. Response and adaptation to salt stress require the
nals with endogenous developmental cues to optimize the
integration and coordination of multiple phytohormones, includ-
balance between growth and stress response [134]. Na+ influx
ing abscisic acid (ABA), ethylene (ETH), jasmonic acid (JA), gib-
occurs through voltage-insensitive monovalent cation channels
berellic acid (GA), cytokinin (CK), and salicylic acid (SA), which
(VICs) [135,136]. These channels are regulated using bivalent
regulate normal growth and mediate responses to abiotic stress
cations such as Ca2+ to maintain ion homeostasis [31]. Early salt
[14,149–151].
signaling responses are induced, including Ca2+ signaling, and
these early Na+-induced signals reduce Na+ import [14]. Ca2+ acts
as a second messenger, and its levels are rapidly elevated when 4.2.1. Abscisic acid
plants encounter abiotic stresses [31]. An initial Ca2+-dependent ABA is the central regulator of abiotic stress tolerance in plants
signaling network is involved in salt stress responses and includes [151–153]. It has a major role in the salt stress response, regulating
both Ca2+ transport and downstream targets such as calmodulin stomatal closure, ion homeostasis, salt stress–responsive gene
(CaM), CMLs (CaM-like proteins), CDPK/CPK (calcium-dependent expression, and metabolic changes [14]. Salt stress causes an
protein kinase), CBLs (calcineurin B-like proteins), and CIPKs increase in ABA accumulation, and exogenous ABA may alleviate
(CBL-interacting protein kinases) [31,137]. the deleterious effects of salt stress [154,155]. ABA signal transduc-
17
C. Liu, B. Mao, D. Yuan et al. The Crop Journal 10 (2022) 13–25

Fig. 2. Known salt stress signaling pathways in rice. (1) The most classic pathway is the SOS signaling pathway (left), which consists of OsSOS3/OsCBL4, OsSOS2/OsCIPK24,
and OsSOS1. It is important for sensing salt-induced Ca2+ signals and regulating ion homeostasis by removing excess Na+ from cells or compartmentalizing it in the vacuole.
(2) In the ABA-dependent pathway (middle), salt stress induces ABA accumulation and promotes ABA binding to PYR/PYL ABA receptors such as OsPYR/RCAR5, leading to the
inactivation of core ABA-signaling components (PP2Cs such as PP108). The PYL-ABA-PP2C complex forms, releasing SnRK2s such as SAPK4 from association with and
inhibition by PP2Cs. The released SnRK2s are activated through autophosphorylation and in turn activate the expression of many downstream effectors. (3) In one typical
ABA-independent pathway (right), ethylene regulates stress responses. OsSIT1 mediates salt sensitivity by activating MPK3/6, thereby promoting ethylene biosynthesis and
ROS accumulation. The ethylene-response genes MHZ6/OsEIL1 and OsEIL2 negatively regulate salt tolerance by controlling OsHKT2;1 expression and Na+ uptake in rice roots.

tion requires three core components: PYR/PYL/RCAR ABA receptors ABA receptor gene OsPYL/RCAR5 is a positive regulator of the ABA
(PYLs), type 2C protein phosphatases (PP2Cs), and class III SNF-1- signal transduction pathway in abiotic stress tolerance [165,166]
related protein kinase 2 s (SnRK2s) [156]. Under salt stress condi- (Fig. 2). The PP2C genes (such as OsPP108) negatively regulate
tions, ABA binds to PYR/PYL/RCAR receptors, which then interact ABA signaling but positively regulate abiotic stress signaling
with PP2Cs and inhibit their activity, thus releasing SnRK2s from [167] (Table 5; Fig. 2). The transcription factors OsABF1/OsbZIP12,
repression [157–159]. SnRK2s phosphorylate various ABA- OsABF2/OsbZIP46, OsBZ8/OsbZIP05, OsbZIP71, OsWRKY45, SNAC1,
responsive element (ABRE)-binding protein/ABRE-binding factor OsNAC2, ONAC022, OsMADS25, OsMYB48-1, OsMYB91, and
(AREB/ABF) transcription factors, which further regulate ROS scav- OsRHP1 play positive roles in ABA-mediated salt tolerance of rice
enging, ion homeostasis, and stomatal closure in response to salt [62,116,168–179], whereas OsABI5/OsbZIP10, ZFP185, and
stress [14,160–162]. The ABA biosynthesis genes OsNCED3 and Oshox22 play negative roles [180–183]. OsbZIP23 directly targets
OsNCED5, encoding 9-cis-epoxycarotenoid dioxygenases, increase the ABA synthesis gene OsNCED4 and the ABA signaling component
ABA levels and enhance salt stress tolerance [163,164]. The rice gene OsPP2C49, which act as positive and negative regulators,
18
C. Liu, B. Mao, D. Yuan et al. The Crop Journal 10 (2022) 13–25

Table 5
Phytohormones that mediate salt stress responses.

Family Group Effect on Relevant genes References


tolerance
ABA Synthesis Positive OsNCDE3, OsNCED5 Huang et al. [163], Huang et al. [164]
Signal Positive OsPYL/RCAR5, OsABF1/OsbZIP12, OsABF2/OsbZIP46, OsBZ8/ Liu et al. [62], Xu et al. [116], Kim et al. [166], Hossain et al. [168],
OsbZIP05, OsbZIP71, OsWRKY45, SNAC1, OsNAC2, ONAC022, Hossain et al. [169], RoyChoudhury et al. [170], Mukherjee et al.
OsMADS25, OsMYB48-1, OsMYB91, OsRHP1 [171], Nakagawa et al. [172], Amir Tao et al. [173], Hu et al. [174],
Shen et al. [175], Hong et al. [176], Zhu et al. [177], Xiong et al.
[178], Zeng et al. [179]
Negative OsPP108, OsABI5/OsbZIP10, ZFP185, Oshox22 Singh et al. [167], Zou et al. [180], Bhattacharjee et al. [182], Zhang
et al. [183]
Feedback OsbZIP23 Zong et al. [184]
regulation
ETH Signal Negative MHZ6/OsEIL1, OsEIN2, OsSIT1, OsAP23, OsERF922, OsEREBP1, Yang et al. [185], Yang et al. [186], Li et al. [187], Serra et al. [188],
OsEREBP2 Zhuang et al. [189], Liu et al. [190], Jisha et al. [191]
Positive OsDOF15, OsBIERF3 Qin et al. [192], Cao et al. [193], Cao et al. [194]
GA Catabolism Positive OsGA2ox5, OsCYP71D8L Shan et al. [195]
Signal Negative OsSLR1 Mo et al. [196]
Auxin Signal Positive RBG1 Lo et al. [197]
Negative OsNAC2, OsTIR1, OsAFB2 Shen et al. [175], Xia et al. [198], Mao et al. [199])
CK Catabolism Negative OsCKX2 Joshi et al. [200]
Signal Negative OsRR9, OsRR10, OsRR22 Gao et al. [124], Zhang et al. [125], Wang et al. [201]
Positive OsAGO2, OsBG3, OsAHP1, OsAHP2, OsNAC2 Shen et al. [175], Mao et al. [199], Yin et al. [202], Xiao et al. [203],
Sun et al. [204]
BR Signal Negative GW5L, osa-miR1848 Tian et al. [205], Xia et al. [206]
Positive GSK2, OsCYP51G3 Xia et al. [206], Koh et al. [207]
JA Catabolism Positive OsCYP94C2b Kurotani et al. [208]
Signal Positive OsJAZ8, OsJAZ9 Wu et al. [209], Peethambaran et al. [210]
SA Signal Positive RSOsPR10 Yamamoto et al. [211], Takeuchi et al. [212]

respectively, and it can therefore feedback-regulate ABA signaling log of the Arabidopsis DELLA gene Slender rice1, OsSLR1, encodes a
and biosynthesis in response to abiotic stress [184] (Table 5). GA signaling component that regulates mesocotyl and root growth,
specifically in the dark and under salt stress [196] (Table 5). There-
4.2.2. Ethylene fore, rice growth inhibition under salt stress may be an active
Etiolated seedlings treated with ethylene exhibit a double adaptation mechanism whereby rice reduces GA levels or GA sig-
response, i.e. the promotion of coleoptile growth and the inhibition naling in response to changing environmental conditions [134].
of root elongation [213]. The function of ethylene in rice salt toler-
ance has been studied extensively. Promotion of ethylene biosyn-
4.2.4. Auxin
thesis and signal transduction can enhance plant salinity
Auxin plays an important role in root system growth in
tolerance, whereas their inhibition increases sensitivity to salinity
response to local soil conditions [134,222]. Rice Big Grain 1
in Arabidopsis and soybean [214,215]. However, in tomato, the
(RBG1) regulates cell division, auxin accumulation, and tolerance
direct ethylene precursor ACC causes Na+ accumulation and oxida-
to drought, salt, and heat stresses [197]. The two rice auxin recep-
tive stress in leaves and promotes leaf senescence under salinity
tor genes OsTIR1 and OsAFB2 are targeted by OsmiR393, downreg-
stress [216–218]. Ethylene treatment of Nipponbare rice seedlings
ulated the expression of OsAUX1 (an auxin transporter) and OsTB1
increased their sensitivity to salinity, whereas treatment with 1-
(a tillering inhibitor), repressing auxin signaling and leading to
MCP (a blocker of ethylene perception) enhanced salinity tolerance
more tillers, earlier flowering, and reduced salt and drought toler-
[185,219]. Salt Intolerance 1 (OsSIT1), a lectin receptor-like kinase,
ance [198]. The NAC transcription factor OsNAC2 decreases auxin
is an ethylene sensor that positively mediates salt sensitivity by
biosynthesis, content, and gene responses while increasing cytoki-
activating MITOGEN-ACTIVATED PROTEIN KINASE3/6 (MPK3/6),
nin (CK) biosynthesis gene expression and CK content, thereby
thereby promoting ethylene biosynthesis, ROS accumulation, and
integrating the auxin and CK pathways to modulate rice root devel-
sensitivity to salt stress [187] (Table 5; Fig. 2). The ethylene
opment under normal and saline conditions [175,199] (Table 5).
response factors (AP2/ERFs), such as OsAP23, OsERF922, OsEREBP1,
and OsEREBP2, as well as the ethylene signaling component genes
MHZ6/OsEIL1 and OsEIL2, are positively regulated by ethylene sig- 4.2.5. Cytokinins
naling and negatively affect salt tolerance in rice [185,188–191] CKs function in the control of plant adaptation to environmental
(Table 5; Fig. 2). The DOF transcription factor OsDOF15 and the stress. In Arabidopsis, plants with reduced levels of various CKs
ethylene-responsive element binding protein (EREBP) transcrip- exhibit enhanced salt tolerance [223], whereas exogenous CK
tion factor OsBIERF3 positively regulate primary root elongation application increases salt tolerance in Solanum melongena [224].
under salt stress by restricting ethylene biosynthesis [192–194] The cytokinin oxidase OsCKX2, an enzyme of the CK degradation
(Table 5). pathway, negatively regulates salt stress tolerance by controlling
CK levels [200]. The CK response regulators OsRR9, OsRR10, and
4.2.3. Gibberellic acid OsRR22 inhibit the response to CK signaling by a negative feedback
GA is an important hormone that regulates plant growth and mechanism and improve rice tolerance to salt stress [124,125,201].
has been linked to the regulation of growth under abiotic stress OsAGO2 increases salt tolerance by activating the expression of
[220]. The GA catabolic pathway genes gibberellin 2-oxidase 5 OsBG3 and altering CK distribution [202,203]. Two rice authentic
(OsGA2ox5) and OsCYP71D8L reduced GA accumulation and histidine phosphotransferases, OsAHP1 and OsAHP2, and the NAC
enhanced plant salt tolerance by retarding growth [195,221], sug- transcription factor OsNAC2 mediate CK signaling and enhance
gesting a negative role for GA in rice salt tolerance. The rice homo- plant tolerance to salt stress [199,204] (Table 5).
19
C. Liu, B. Mao, D. Yuan et al. The Crop Journal 10 (2022) 13–25

4.2.6. Brassinosteroids the most effective means for overcoming this environmental chal-
Brassinosteroids (BRs) are plant steroid hormones that play lenge [23]. Many salt response genes have been cloned and identi-
essential roles in plant growth, development, and abiotic stress fied in the past three decades, and mechanisms of rice salt stress
responses [225]. Exogenous application of BR enhances salt toler- tolerance have been characterized. Two major approaches have
ance in rice [226]. GW5-Like (GW5L) positively regulates BR sig- been used to improve crop salt tolerance: (1) exploitation of natu-
naling by repressing the phosphorylation activity of GSK2 and ral genetic variation between tolerant and sensitive varieties, and
confers salt stress sensitivity [205,207]. The rice microRNA osa- (2) generation of transgenic plants with novel genes or altered
miR1848 targets the obtusifoliol 14a-demethylase gene OsCYP51G3 expression levels of existing genes [234]. However, the application
and mediates the biosynthesis of phytosterols and BR during of this fundamental knowledge to improve salt stress tolerance of
development and in response to stress [206] (Table 5). crops in the field is a slow and challenging process.
Because salt tolerance is a complex quantitative trait, only a few
4.2.7. Jasmonic acid haplotypes of genes or loci have been reported that could be used
Jasmonic acid (JA) is required for the inhibition of root elonga- for molecular marker–assisted breeding. For example, the T67K
tion and the activation of antioxidant enzymes upon exposure to and P140A mutations may destabilize the transmembrane domain
high salinity [227]. Endogenous JA accumulates in rice roots under or alter the phosphorylation probability, respectively, of
salt stress, and exogenous JA improves salt stress tolerance in both OsHKT1;5/SKC1, thereby influencing rice tolerance to salt stress
rice and wheat [228,229]. Overexpression of the Cyt P450 family [235–237]. The K24E mutation in the SalT protein appears to alter
gene OsCYP94C2b, encoding a JA-catabolizing enzyme, reduces JA its interactions with other proteins, affecting its function and influ-
content and improves performance at high salt concentrations encing salinity tolerance [235,238]. The Saltol quantitative trait
[208]. The plant-specific TIFY transcription factors OsJAZ8 and locus (QTL) contributes to salinity tolerance and has been fine
OsJAZ9 act as negative regulators of jasmonic signaling but posi- mapped on chromosome 1 using a Pokkali/IR29 recombinant
tively modulate salt stress tolerance in rice [209,210] (Table 5). inbred line (RIL) population, and the simple sequence repeat
(SSR) markers RM493 and RM3412b have been used effectively
in marker-assisted breeding [239–243].
4.2.8. Salicylic acid
However, marker-assisted breeding has two problems: first,
Salicylic acid (SA) plays an important role in plant salt toler-
QTL mapping results based on specific parents are not usually uni-
ance. Exogenous SA application enhances the antioxidant system,
versal and cannot readily be applied to breeding populations. Sec-
increases the synthesis of osmolytes, and promotes photosynthesis
ond, salt tolerance is controlled by multiple minor genes, and these
and nitrogen fixation under salt stress [230–232]. The
genes or loci cannot be used effectively to improve quantitative
pathogenesis-related protein RSOsPR10 (root specific rice PR10)
traits. Genome selection (GS) using SNPs or alleles of the whole rice
is regulated antagonistically by JA/ethylene and SA signaling and
genome can be used to predict genomic estimated breeding value
improves plant tolerance to salt stress [211,212] (Table 5).
(GEBV) of individuals in rice populations based on allele effects
using a genetic relationship matrix. GS is time-efficient and can
4.3. Conclusions combine pedigree and genotype information, making it superior
to other methods of plant breeding [244,245].
Over the course of evolution, plants have developed the capac- Improvements in crop salt stress tolerance will become more
ity to detoxify the excessive Na+ levels that accompany high salt feasible with the aid of gene editing technologies and advances
concentrations and to utilize multiple strategies for the alleviation in the efficient genetic transformation of different species [246].
of salt stress damage [31]. Plant salt responses caused by hyperos- Genes that confer salt sensitivity, such as DST, OsRR22, OsEIL1,
motic tension are closely linked to Ca2+ channels and Ca2+ signaling OsEIN2, and OsSIT1, could be knocked out to improve rice salt
[31,233]. (1) The most typical Ca2+ signaling pathway is the SOS tolerance [120,125,185,187]. The OsbZIP23 promoter has a
pathway, in which OsSOS3/OsCBL4 senses the cytosolic Ca2+ signal 35-nucleotide deletion in the 50 -UTR in the drought-sensitive
elicited by salt stress and phosphorylates OsSOS2/OsCIPK24, which IR20 cultivar, and the deleted sequence is associated with
activates OsSOS1, a plasma membrane Na+/H+ antiporter that reg- decreased OsbZIP23 expression and reduced tolerance to abiotic
ulates ion homeostasis by transporting excess Na+ out of cells or stress relative to Oryza rufipogon [247]. We can also use genome
compartmentalizing it in the vacuole [41] (Fig. 2). (2) Phytohor- editing technology for knock-in of the deleted sequence in
mones mediate the response to various environmental stresses, cultivated rice.
including salt stress, and thus regulate plant growth adaptation An understanding of the molecular mechanisms of rice salt tol-
[9,134]. ABA is the hormone usually associated with major plant erance can now be combined with the development of molecular
responses to stress [153]. In the ABA-dependent pathway, the salt tolerance markers, the complete sequencing of plant genomes,
PYL-ABA-PP2C complex forms, releasing SnRK2s such as SAPK4 and the widespread use of microarray analysis and genome editing
to phosphorylate many downstream effectors (Fig. 2). (3) Ethylene technology. These powerful techniques offer advantages and pro-
is also a widely known abiotic stress signal. For example, the vide solutions to the complex and intriguing trait of plant salt
receptor-like kinase OsSIT1 mediates salt sensitivity by activating resistance.
MPK3/6, which promotes ethylene biosynthesis and ROS accumu-
lation [187] (Fig. 2). OsEIL1 and OsEIL2 are ethylene response genes Declaration of competing interest
that repress the expression of K+ transporters [185] (Fig. 2). In brief,
plants have developed various signaling cascades for sustaining ion The authors declare that they have no known competing finan-
homeostasis and cell turgor pressure, avoiding cellular oxidative cial interests or personal relationships that could have appeared
damage or nutritional imbalances, and optimizing the balance to influence the work reported in this paper.
between growth and stress responses under salt stress.
CRediT authorship contribution statement
5. Future perspectives
Citao Liu wrote the manuscript. Bigang Mao, Dingyang Yuan,
Ongoing soil salinity threatens rice production and future food Chengcai Chu, and Meijuan Duan revised and edited the
security, and breeding and cultivation of salt-tolerant varieties is manuscript.
20
C. Liu, B. Mao, D. Yuan et al. The Crop Journal 10 (2022) 13–25

Acknowledgments [26] R. Munns, M. Tester, Mechanisms of salinity tolerance, Annu. Rev. Plant Biol.
59 (2008) 651–681.
[27] H. Wu, X. Zhang, J.P. Giraldo, S. Shabala, It is not all about sodium: revealing
This work was supported by the Research Initiation Fund of tissue specificity and signalling roles of potassium in plant responses to salt
Hunan Agricultural University (20154/5407419002), the Open stress, Plant Soil 431 (2018) 1–17.
[28] J.M. Cheeseman, The integration of activity in saline environments: problems
Research Fund of the State Key Laboratory of Hybrid Rice, Hunan
and perspectives, Funct. Plant Biol. 40 (2013) 759–774.
Hybrid Rice Research Center (2020KF05), the Hunan Science and [29] M.N. Iqbal, R. Rasheed, M.Y. Ashraf, M.A. Ashraf, I. Hussain, Exogenously
Technology Major Project (2018NK1010), and the Hunan Science applied zinc and copper mitigate salinity effect in maize (Zea mays L.) by
improving key physiological and biochemical attributes, Environ. Sci. Pollut.
and Technology Talents Support Project (2019TJ-Q08). The authors
Res. Int. 25 (2018) 23883–23896.
would like to thank TopEdit (www.topeditsci.com) for its linguistic [30] S. Shabala, I. Pottosin, Regulation of potassium transport in plants under
assistance during the preparation of this manuscript. hostile conditions: implications for abiotic and biotic stress tolerance,
Physiol. Plant. 151 (2014) 257–279.
[31] M. Seifikalhor, S. Aliniaeifard, A. Shomali, N. Azad, B. Hassani, O. Lastochkina,
T. Li, Calcium signaling and salt tolerance are diversely entwined in plants,
References Plant Signal. Behav. 14 (2019) 1665455.
[32] R. Munns, R.A. James, M. Gilliham, T.J. Flowers, T.D. Colmer, Tissue tolerance:
an essential but elusive trait for salt-tolerant crops, Funct. Plant Biol. 43
[1] Y. Yang, Y. Guo, Elucidating the molecular mechanisms mediating plant salt-
(2016) 1103–1113.
stress responses, New Phytol. 217 (2018) 523–539.
[33] M.A. Ahanger, N.S. Tomar, M. Tittal, S. Argal, R.M. Agarwal, Plant growth
[2] C.J. Ruan, J.A.T. da Silva, S. Mopper, P. Qin, S. Lutts, Halophyte improvement
under water/salt stress: ROS production; antioxidants and significance of
for a salinized world, Crit. Rev. Plant Sci. 29 (2010) 329–359.
added potassium under such conditions, Physiol. Mol. Biol. Plants. 23 (2017)
[3] S. Shabala, Learning from halophytes: physiological basis and strategies to
731–744.
improve abiotic stress tolerance in crops, Ann. Bot. 112 (2013) 1209–1221.
[34] G. Miller, N. Suzuki, S. Ciftci-Yilmaz, R. Mittler, Reactive oxygen species
[4] M. Kamran, A. Parveen, S. Ahmar, Z. Malik, S. Hussain, M.S. Chattha, M.H.
homeostasis and signalling during drought and salinity stresses, Plant Cell
Saleem, M. Adil, P. Heidari, J.T. Chen, An overview of hazardous impacts of soil
Environ. 33 (2010) 453–467.
salinity in crops, tolerance mechanisms, and amelioration through selenium
[35] A. Ismail, S. Takeda, P. Nick, Life and death under salt stress: same players,
supplementation, Int. J. Mol. Sci. 21 (2019) 148.
different timing?, J Exp. Bot. 65 (2014) 2963–2979.
[5] A.D. de Azevedo Neto, J.T. Prisco, J. Enéas-Filho, C.E.B. de Abreu, E. Gomes-
[36] H. Greenway, R. Munns, Mechanisms of salt tolerance in nonhalophytes,
Filho, Effect of salt stress on antioxidative enzymes and lipid peroxidation in
Annu. Rev. Plant Physiol. 31 (1980) 149–190.
leaves and roots of salt-tolerant and salt-sensitive maize genotypes, Environ.
[37] H.H. Fu, S. Luan, AtKUP1: a dual-affinity K+ transporter from Arabidopsis, Plant
Exp. Bot. 56 (2006) 87–94.
Cell 10 (1998) 63–73.
[6] A.F. Lodeyro, N. Carrillo, Salt stress in higher plants: mechanisms of toxicity
[38] M. Ashraf, P. Harris, Potential biochemical indicators of salinity tolerance in
and defensive responses, in: B. Nath, T.M. Müller (Eds.), Stress Responses in
plants, Plant Sci. 166 (2004) 3–16.
Plants, Springer, Cham, Switzerland, 2015.
[39] J. Freitas, R. Chagas, I. Almeida, F. Cavalcanti, J. Silveira, Expression of
[7] S.A. Naveed, F. Zhang, J. Zhang, T.Q. Zheng, L.J. Meng, Y.L. Pang, J.L. Xu, Z.K. Li,
physiological traits related to salt tolerance in two contrasting cowpea
Identification of QTN and candidate genes for salinity tolerance at the
cultivars, Documentos Embrapa Meio Norte. 56 (2001) 115–118.
germination and seedling stages in rice by genome-wide association analyses,
[40] M. Tester, R. Davenport, Na+ tolerance and Na+ transport in higher plants,
Sci. Rep. 8 (2018) 6505.
Ann. Bot. 91 (2003) 503–527.
[8] R. Munns, Comparative physiology of salt and water stress, Plant Cell Environ.
[41] H. El Mahi, J. Pérez-Hormaeche, A. De Luca, I. Villalta, J. Espartero, F. Gámez-
25 (2002) 239–250.
Arjona, J.L. Fernández, M. Bundó, I. Mendoza, D. Mieulet, E. Lalanne, S.Y. Lee,
[9] H. Qin, R. Huang, The phytohormonal regulation of Na+/K+ and reactive
D.J. Yun, E. Guiderdoni, M. Aguilar, E.O. Leidi, J.M. Pardo, F.J. Quintero, A
oxygen species homeostasis in rice salt response, Mol. Breed. 40 (2020) 1–13.
critical role of sodium flux via the plasma membrane Na+/H+ exchanger SOS1
[10] S. Shabala, T.A. Cuin, Potassium transport and plant salt tolerance, Physiol.
in the salt tolerance of rice, Plant Physiol. 180 (2019) 1046–1065.
Plant. 133 (2008) 651–669.
[42] J. Martínez-Atienza, X. Jiang, B. Garciadeblas, I. Mendoza, J.K. Zhu, J.M. Pardo,
[11] X. Ma, F. Feng, H. Wei, H. Mei, K. Xu, S. Chen, T. Li, X. Liang, H. Liu, L. Luo,
F.J. Quintero, Conservation of the salt overly sensitive pathway in rice, Plant
Genome-wide association study for plant height and grain yield in rice under
Physiol. 143 (2007) 1001–1012.
contrasting moisture regimes, Front. Plant Sci. 7 (2016) 1801.
[43] S. Liu, L. Zheng, Y. Xue, Q. Zhang, L. Wang, H. Shou, Overexpression of OsVP1
[12] M. Hussain, S. Ahmad, S. Hussain, R. Lal, S. Ul-Allah, A. Nawaz, Rice in saline
and OsNHX1 increases tolerance to drought and salinity in rice, J. Plant Biol.
soils: physiology, biochemistry, genetics, and management, Adv. Agron. 148
53 (2010) 444–452.
(2018) 231–287.
[44] A. Fukuda, A. Nakamura, N. Hara, S. Toki, Y. Tanaka, Molecular and functional
[13] A. Razzaq, A. Ali, L.B. Safdar, M.M. Zafar, Y. Rui, A. Shakeel, A. Shaukat, M.
analyses of rice NHX-type Na+/H+ antiporter genes, Planta 233 (2011) 175–
Ashraf, W. Gong, Y. Yuan, Salt stress induces physiochemical alterations in
188.
rice grain composition and quality, J. Food Sci. 85 (2020) 14–20.
[45] M.I. Uddin, Y. Qi, S. Yamada, I. Shibuya, X.P. Deng, S.S. Kwak, H. Kaminaka, K.
[14] E. van Zelm, Y. Zhang, C. Testerink, Salt tolerance mechanisms of plants, Annu.
Tanaka, Overexpression of a new rice vacuolar antiporter regulating protein
Rev. Plant Biol. 71 (2020) 403–433.
OsARP improves salt tolerance in tobacco, Plant Cell Physiol. 49 (2008) 880–
[15] M.A. Hakim, A.S. Juraimi, M.M. Hanafi, M.R. Ismail, A. Selamat, M.Y. Rafii, M.A.
890.
Latif, Biochemical and anatomical changes and yield reduction in rice (Oryza
[46] R. Wang, W. Jing, L. Xiao, Y. Jin, L. Shen, W. Zhang, The rice high-affinity
sativa L.) under varied salinity regimes, BioMed Res. Int. 2014 (2014) 208584.
potassium transporter1;1 is involved in salt tolerance and regulated by an
[16] R.M.A. Machado, R.P. Serralheiro, Soil salinity: effect on vegetable crop
MYB-type transcription factor, Plant Physiol. 168 (2015) 1076–1090.
growth, Management practices to prevent and mitigate soil salinization,
[47] K. Suzuki, N. Yamaji, A. Costa, E. Okuma, N.I. Kobayashi, T. Kashiwagi, M.
Horticulturae 3 (2017) 30.
Katsuhara, C. Wang, K. Tanoi, Y. Murata, J.I. Schroeder, J.F. Ma, T. Horie,
[17] S. Muranaka, K. Shimizu, M. Kato, Ionic and osmotic effects of salinity on
OsHKT1;4 mediated Na+ transport in stems contributes to Na+ exclusion from
single-leaf photosynthesis in two wheat cultivars with different drought
leaf blades of rice at the reproductive growth stage upon salt stress, BMC
tolerance, Photosynthetica 40 (2002) 201–207.
Plant Biol. 16 (2016) 22.
[18] L.R. Murphy, S.T. Kinsey, M. Durako, Physiological effects of short-term
[48] Z.H. Ren, J.P. Gao, L.G. Li, X.L. Cai, W. Huang, D.Y. Chao, M.Z. Zhu, Z.Y. Wang, S.
salinity changes on Ruppia maritima, Aquat. Bot. 75 (2003) 293–309.
Luan, H.X. Lin, A rice quantitative trait locus for salt tolerance encodes a
[19] J.K. Zhu, Abiotic stress signaling and responses in plants, Cell 167 (2016) 313–
sodium transporter, Nat. Genet. 37 (2005) 1141–1146.
324.
[49] S.W. Ardie, L. Xie, R. Takahashi, S. Liu, T. Takano, Cloning of a high-affinity K+
[20] M. Hakim, A.S. Juraimi, M.M. Hanafi, M.R. Ismail, A. Selamat, The effect of
transporter gene PutHKT2;1 from Puccinellia tenuiflora and its functional
salinity on growth, ion accumulation and yield of rice varieties, J. Anim Plant
comparison with OsHKT2;1 from rice in yeast and Arabidopsis, J. Exp. Bot. 60
Sci. 24 (2014) 874–885.
(2009) 3491–3502.
[21] R. Munns, J. Guo, J.B. Passioura, G.R. Cramer, Leaf water status controls day-
[50] T. Horie, A. Costa, T.H. Kim, M.J. Han, R. Horie, H.Y. Leung, A. Miyao, H.
time but not daily rates of leaf expansion in salt-treated barley, Funct. Plant
Hirochika, G. An, J.I. Schroeder, Rice OsHKT2;1 transporter mediates large Na+
Biol. 27 (2000) 949–957.
influx component into K+-starved roots for growth, EMBO J. 26 (2007) 3003–
[22] W. Fricke, G. Akhiyarova, D. Veselov, G. Kudoyarova, Rapid and tissue-specific
3014.
changes in ABA and in growth rate in response to salinity in barley leaves, J.
[51] S. Huang, W. Spielmeyer, E.S. Lagudah, R. Munns, Comparative mapping of
Exp. Bot. 55 (2004) 1115–1123.
HKT genes in wheat, barley, and rice, key determinants of Na+ transport, and
[23] C. Zhao, H. Zhang, C. Song, J.K. Zhu, S. Shabala, Mechanisms of plant responses
salt tolerance, J. Exp. Bot. 59 (2008) 927–937.
and adaptation to soil salinity, Innovation 1 (2020) 100017.
[52] B. Garciadeblas, M.E. Senn, M.A. Banuelos, A. Rodriguez-Navarro, Sodium
[24] L.H. Wegner, G. Stefano, L. Shabala, M. Rossi, S. Mancuso, S. Shabala,
transport and HKT transporters: the rice model, Plant J. 34 (2003) 788–801.
Sequential depolarization of root cortical and stelar cells induced by an
[53] T. Obata, H.K. Kitamoto, A. Nakamura, A. Fukuda, Y. Tanaka, Rice shaker
acute salt shock-implications for Na+ and K+ transport into xylem vessels,
potassium channel OsKAT1 confers tolerance to salinity stress on yeast and
Plant Cell Environ. 34 (2011) 859–869.
rice cells, Plant Physiol. 144 (2007) 1978–1985.
[25] Y. Yang, Y. Guo, Unraveling salt stress signaling in plants, J. Integr. Plant Biol.
60 (2018) 796–804.

21
C. Liu, B. Mao, D. Yuan et al. The Crop Journal 10 (2022) 13–25

[54] H. Hwang, J.Y. Yoon, H. Cho, B.G. Kim, OsKAT2 is the prevailing functional [80] J. Huang, S. Sun, D. Xu, H. Lan, H. Sun, Z. Wang, Y. Bao, J. Wang, H. Tang, H.
inward rectifier potassium channels in rice guard cell, Plant Signal. Behav. 8 Zhang, A TFIIIA-type zinc finger protein confers multiple abiotic stress
(2013) e26643. tolerances in transgenic rice (Oryza sativa L.), Plant Mol, Biol. 80 (2012) 337–
[55] T. Yang, S. Zhang, Y. Hu, F. Wu, Q. Hu, G. Chen, J. Cai, T. Wu, N. Moran, L. Yu, G. 350.
Xu, The role of a potassium transporter OsHAK5 in potassium acquisition and [81] K. Liu, L. Wang, Y. Xu, N. Chen, Q. Ma, F. Li, K. Chong, Overexpression of
transport from roots to shoots in rice at low potassium supply levels, Plant OsCOIN, a putative cold inducible zinc finger protein, increased tolerance to
Physiol. 166 (2014) 945–959. chilling, salt and drought, and enhanced proline level in rice, Planta 226
[56] T. Horie, M. Sugawara, T. Okada, K. Taira, P. Kaothien-Nakayama, M. (2007) 1007–1016.
Katsuhara, A. Shinmyo, H. Nakayama, Rice sodium-insensitive potassium [82] S.Y. Song, Y. Chen, J. Chen, X.Y. Dai, W.H. Zhang, Physiological mechanisms
transporter, OsHAK5, confers increased salt tolerance in tobacco BY2 cells, J. underlying OsNAC5-dependent tolerance of rice plants to abiotic stress, Planta
Biosci. Bioeng. 111 (2011) 346–356. 234 (2011) 331–345.
[57] Y. Shen, L. Shen, Z. Shen, W. Jing, H. Ge, J. Zhao, W. Zhang, The potassium [83] S.S. Gill, N. Tuteja, Reactive oxygen species and antioxidant machinery in
transporter OsHAK21 functions in the maintenance of ion homeostasis and abiotic stress tolerance in crop plants, Plant Physiol. Bochem. 48 (2010) 909–
tolerance to salt stress in rice, Plant Cell Environ. 38 (2015) 2766–2779. 930.
[58] Y. He, B. Yang, Y. He, C. Zhan, Y. Cheng, J. Zhang, H. Zhang, J. Cheng, Z. Wang, A [84] J. You, Z. Chan, ROS regulation during abiotic stress responses in crop plants,
quantitative trait locus, qSE3, promotes seed germination and seedling Front. Plant Sci. 6 (2015) 1092.
establishment under salinity stress in rice, Plant J. 97 (2019) 1089–1104. [85] J.C. Begara-Morales, B. Sánchez-Calvo, M. Chaki, C. Mata-Pérez, R. Valderrama,
[59] N. Bazihizina, T.D. Colmer, T.A. Cuin, S. Mancuso, S. Shabala, Friend or foe?, M.N. Padilla, J. López-Jaramillo, F. Luque, F.J. Corpas, J.B. Barroso, Differential
Chloride patterning in halophytes, Trends Plant Sci 24 (2019) 142–151. molecular response of monodehydroascorbate reductase and glutathione
[60] A. Nakamura, A. Fukuda, S. Sakai, Y. Tanaka, Molecular cloning, functional reductase by nitration and S-nitrosylation, J. Exp. Bot. 66 (2015) 5983–
expression and subcellular localization of two putative vacuolar voltage- 5996.
gated chloride channels in rice (Oryza sativa L.), Plant Cell Physiol. 47 (2006) [86] X. Wang, G. Fang, J. Yang, Y. Li, A thioredoxin-dependent glutathione
32–42. peroxidase (OsGPX5) is required for rice normal development and salt
[61] C.J. Diedhiou, D. Golldack, Salt-dependent regulation of chloride channel stress tolerance, Plant Mol. Biol. Rep. 35 (2017) 333–342.
transcripts in rice, Plant Sci. 170 (2006) 793–800. [87] K. Das, A. Roychoudhury, Reactive oxygen species (ROS) and response of
[62] C.T. Liu, B.G. Mao, S.J. Ou, W. Wang, L.C. Liu, Y.B. Wu, C.C. Chu, X.P. Wang, antioxidants as ROS-scavengers during environmental stress in plants, Front.
OsbZIP71, a bZIP transcription factor, confers salinity and drought tolerance Environ. Sci. 2 (2014) 53.
in rice, Plant Mol. Biol. 84 (2014) 19–36. [88] A.E. Postiglione, G.K. Muday, The role of ROS homeostasis in ABA-induced
[63] P. Manimaran, S. Venkata Reddy, M. Moin, M. Raghurami Reddy, P. guard cell signaling, Front. Plant Sci. 11 (2020) 968.
Yugandhar, S.S. Mohanraj, S.M. Balachandran, P.B. Kirti, Activation-tagging [89] R. Mittler, ROS are good, Trends Plant Sci. 22 (2017) 11–19.
in indica rice identifies a novel transcription factor subunit, NF-YC13 [90] M.A. Torres, J.L. Dangl, Functions of the respiratory burst oxidase in biotic
associated with salt tolerance, Sci. Rep. 7 (2017) 9341. interactions, abiotic stress and development, Curr. Opin. Plant Biol. 8 (2005)
[64] C.J. Diédhiou, O.V. Popova, K.J. Dietz, D. Golldack, The SNF1-type serine- 397–403.
threonine protein kinase SAPK4 regulates stress-responsive gene expression [91] S.B. Abdallah, B. Aung, L. Amyot, I. Lalin, M. Lachâal, N. Karray-Bouraoui, A.
in rice, BMC Plant Biol. 8 (2008) 49. Hannoufa, Salt stress (NaCl) affects plant growth and branch pathways of
[65] S.K. Lee, B.G. Kim, T.R. Kwon, M.J. Jeong, S.R. Park, J.W. Lee, M.O. Byun, H.B. carotenoid and flavonoid biosyntheses in Solanum nigrum, Acta Physiol. Plant.
Kwon, B.F. Matthews, C.B. Hong, S.C. Park, Overexpression of the mitogen- 38 (2016) 72.
activated protein kinase gene OsMAPK33 enhances sensitivity to salt stress in [92] E. Borghesi, M.L. Gonzalez-Miret, M.L. Escudero-Gilete, F. Malorgio, F.J.
rice (Oryza sativa L.), J. Biosci. 36 (2011) 139–151. Heredia, A.J. Melendez-Martinez, Effects of salinity stress on carotenoids,
[66] G. Kumar, H.R. Kushwaha, R.S. Purty, S. Kumari, S.L. Singla-Pareek, A. Pareek, anthocyanins, and color of diverse tomato genotypes, J. Agric. Food Chem. 59
Cloning, structural and expression analysis of OsSOS2 in contrasting cultivars (2011) 11676–11682.
of rice under salinity stress, Genes Genomes Genomics 6 (2012) 34–41. [93] G.F. Wang, W.Q. Li, W.Y. Li, G.L. Wu, C.Y. Zhou, K.M. Chen, Characterization of
[67] S. Sripinyowanich, P. Klomsakul, B. Boonburapong, T. Bangyeekhun, T. Asami, rice NADPH oxidase genes and their expression under various environmental
H. Gu, T. Buaboocha, S. Chadchawan, Exogenous ABA induces salt tolerance in conditions, Int. J. Mol. Sci. 14 (2013) 9440–9458.
indica rice (Oryza sativa L.): The role of OsP5CS1 and OsP5CR gene expression [94] C.R. Li, D.D. Liang, J. Li, Y.B. Duan, H. Li, Y.C. Yang, R.Y. Qin, L. Li, P.C. Wei, J.B.
during salt stress, Environ. Exp. Bot. 86 (2013) 94–105. Yang, Unravelling mitochondrial retrograde regulation in the abiotic stress
[68] H.W. Li, B.S. Zang, X.W. Deng, X.P. Wang, Overexpression of the trehalose-6- induction of rice ALTERNATIVE OXIDASE 1 genes, Plant Cell Environ. 36 (2013)
phosphate synthase gene OsTPS1 enhances abiotic stress tolerance in rice, 775–788.
Planta 234 (2011) 1007–1018. [95] Q. Guan, X. Liao, M. He, X. Li, Z. Wang, H. Ma, S. Yu, S. Liu, Tolerance analysis of
[69] H. Cao, S. Guo, Y. Xu, K. Jiang, A.M. Jones, K. Chong, Reduced expression of a chloroplast OsCu/Zn-SOD overexpressing rice under NaCl and NaHCO3 stress,
gene encoding a Golgi localized monosaccharide transporter (OsGMST1) PLoS ONE 12 (2017) e0186052.
confers hypersensitivity to salt in rice (Oryza sativa), J. Exp. Bot. 62 (2011) [96] Z. Zhang, Q. Zhang, J. Wu, X. Zheng, S. Zheng, X. Sun, Q. Qiu, T. Lu, Gene
4595–4604. knockout study reveals that cytosolic ascorbate peroxidase 2(OsAPX2) plays a
[70] J. Mathan, A. Singh, A. Ranjan, Sucrose transport in response to drought and critical role in growth and reproduction in rice under drought, salt and cold
salt stress involves ABA-mediated induction of OsSWEET13 and OsSWEET15 in stresses, PLoS ONE 8 (2013) e57472.
rice, Physiol. Plant. (2020), https://doi.org/10.1111/ppl.13210. [97] Z. Lu, D. Liu, S. Liu, Two rice cytosolic ascorbate peroxidases differentially
[71] S. Hasthanasombut, K. Supaibulwatana, M. Mii, I. Nakamura, Genetic improve salt tolerance in transgenic Arabidopsis, Plant Cell Rep. 26 (2007)
manipulation of Japonica rice using the OsBADH1 gene from Indica rice to 1909–1917.
improve salinity tolerance, Plant Cell Tissue Organ. Cult. 104 (2011) 79–89. [98] C.Y. Hong, Y.T. Hsu, Y.C. Tsai, C.H. Kao, Expression of ASCORBATE PEROXIDASE
[72] W. Tang, J. Sun, J. Liu, F. Liu, J. Yan, X. Gou, B.R. Lu, Y. Liu, RNAi-directed 8 in roots of rice (Oryza sativa L.) seedlings in response to NaCl, J. Exp. Bot. 58
downregulation of betaine aldehyde dehydrogenase 1 (OsBADH1) results in (2007) 3273–3283.
decreased stress tolerance and increased oxidative markers without affecting [99] T.S. Chou, Y.Y. Chao, C.H. Kao, Involvement of hydrogen peroxide in heat
glycine betaine biosynthesis in rice (Oryza sativa), Plant Mol. Biol. 86 (2014) shock- and cadmium-induced expression of ascorbate peroxidase and
443–454. glutathione reductase in leaves of rice seedlings, J. Plant Physiol. 169
[73] J. Duan, W. Cai, OsLEA3-2, an abiotic stress induced gene of rice plays a key (2012) 478–486.
role in salt and drought tolerance, PLoS ONE 7 (2012) e45117. [100] K.W. Lee, M.A. Rahman, G. Choi, H. Ji, T. Hwang, S. Lee, Identification of
[74] T. Hu, H. Zeng, S. He, Y. Wu, G. Wang, X. Huang, Molecular analysis of OsLEA4 differentially expressed abiotic stress-induced genes in teff grass (Eragrostis
and its contributions to improve E. coli viability, Appl. Biochem. Biotechnol. tef) leaves, J. Anim. Plant Sci. 28 (2018) 1189–1193.
166 (2012) 222–233. [101] A. Ghosh, A. Pareek, S.K. Sopory, S.L. Singla Pareek, A glutathione responsive
[75] S. He, L. Tan, Z. Hu, G. Chen, G. Wang, T. Hu, Molecular characterization and rice glyoxalase II, OsGLYII-2, functions in salinity adaptation by maintaining
functional analysis by heterologous expression in E. coli under diverse abiotic better photosynthesis efficiency and anti-oxidant pool, Plant J. 80 (2014) 93–
stresses for OsLEA5, the atypical hydrophobic LEA protein from Oryza sativa L, 105.
Mol. Genet. Genomics 287 (2012) 39–54. [102] R. Sharma, P. Priya, M. Jain, Modified expression of an auxin-responsive rice
[76] J. Yu, Y. Lai, X. Wu, G. Wu, C. Guo, Overexpression of OsEm1 encoding a group I CC-type glutaredoxin gene affects multiple abiotic stress responses, Planta
LEA protein confers enhanced drought tolerance in rice, Biochem. Biophys. 238 (2013) 871–884.
Res. Commun. 478 (2016) 703–709. [103] T.M. Wu, W.R. Lin, Y.T. Kao, Y.T. Hsu, C.H. Yeh, C.Y. Hong, C.H. Kao,
[77] X. Yuan, H. Sun, Z. Tang, H. Tang, H. Zhang, J. Huang, A novel little membrane Identification and characterization of a novel chloroplast/mitochondria co-
protein confers salt tolerance in rice (Oryza sativa L.), Plant Mol, Biol. Rep. 34 localized glutathione reductase 3 involved in salt stress response in rice,
(2016) 524–532. Plant Mol. Biol. 83 (2013) 379–390.
[78] D.Q. Xu, J. Huang, S.Q. Guo, X. Yang, Y.M. Bao, H.J. Tang, H.S. Zhang, [104] Y. Lima-Melo, F.E. Carvalho, M.O. Martins, G. Passaia, R.H. Sousa, M.C. Neto,
Overexpression of a TFIIIA-type zinc finger protein gene ZFP252 enhances M. Margis-Pinheiro, J.A. Silveira, Mitochondrial GPX1 silencing triggers
drought and salt tolerance in rice (Oryza sativa L.), FEBS Lett. 582 (2008) differential photosynthesis impairment in response to salinity in rice
1037–1043. plants, J. Integr. Plant Biol. 58 (2016) 737–748.
[79] S.J. Sun, S.Q. Guo, X. Yang, Y.M. Bao, H.J. Tang, H. Sun, J. Huang, H.S. Zhang, [105] T.M. Wu, W.R. Lin, C.H. Kao, C.Y. Hong, Gene knockout of glutathione reductase
Functional analysis of a novel Cys2/His2-type zinc finger protein involved in 3 results in increased sensitivity to salt stress in rice, Plant Mol. Biol. 87
salt tolerance in rice, J. Exp. Bot. 61 (2010) 2807–2818. (2015) 555–564.

22
C. Liu, B. Mao, D. Yuan et al. The Crop Journal 10 (2022) 13–25

[106] H. Kaminaka, S. Morita, M. Nakajima, T. Masumura, K. Tanaka, Gene cloning [130] L. Zhou, Z. Liu, Y. Liu, D. Kong, T. Li, S. Yu, H. Mei, X. Xu, H. Liu, L. Chen, L. Luo,
and expression of cytosolic glutathione reductase in rice (Oryza sativa L.), A novel gene OsAHL1 improves both drought avoidance and drought
Plant Cell Physiol. 39 (1998) 1269–1280. tolerance in rice, Sci. Rep. 6 (2016) 30264.
[107] W.Y. Liu, M.M. Wang, J. Huang, H.J. Tang, H.X. Lan, H.S. Zhang, The OsDHODH1 [131] S.Y. Sun, D.Y. Chao, X.M. Li, M. Shi, J.P. Gao, M.Z. Zhu, H.Q. Yang, S. Luan, H.X.
gene is involved in salt and drought tolerance in rice, J. Integr. Plant Biol. 51 Lin, OsHAL3 mediates a new pathway in the light-regulated growth of rice,
(2009) 825–833. Nat. Cell Biol. 11 (2009) 845–851.
[108] H. Qin, Y. Wang, J. Wang, H. Liu, H. Zhao, Z. Deng, Z. Zhang, R. Huang, Z. Zhang, [132] Q. Zhang, L. Shen, D. Ren, J. Hu, L. Zhu, Z. Gao, G. Zhang, L. Guo, D. Zeng, Q.
Knocking down the expression of gmpase gene OsVTC1-1 decreases salt Qian, Characterization of the CRM gene family and elucidating the function of
tolerance of rice at seedling and reproductive stages, PLoS ONE 11 (2016) OsCFM2 in rice, Biomolecules 10 (2020) 327.
e0168650. [133] V. Kanneganti, A.K. Gupta, Overexpression of OsiSAP8, a member of stress
[109] T. Ushimaru, T. Nakagawa, Y. Fujioka, K. Daicho, M. Naito, Y. Yamauchi, H. associated protein (SAP) gene family of rice confers tolerance to salt, drought
Nonaka, K. Amako, K. Yamawaki, N. Murata, Transgenic Arabidopsis plants and cold stress in transgenic tobacco and rice, Plant Mol. Biol. 66 (2008) 445–
expressing the rice dehydroascorbate reductase gene are resistant to salt 462.
stress, J. Plant Physiol. 163 (2006) 1179–1184. [134] Z. Yu, X. Duan, L. Luo, S. Dai, Z. Ding, G. Xia, How plant hormones mediate salt
[110] N.Q. Dong, Y. Sun, T. Guo, C.L. Shi, Y.M. Zhang, Y. Kan, Y.H. Xiang, H. Zhang, Y. stress responses, Trends Plant Sci. 25 (2020) 1117–1130.
B. Yang, Y.C. Li, H.Y. Zhao, H.X. Yu, Z.Q. Lu, Y. Wang, W.W. Ye, J.X. Shan, H.X. [135] I. Pottosin, O. Dobrovinskaya, Non-selective cation channels in plasma and
Lin, UDP-glucosyltransferase regulates grain size and abiotic stress tolerance vacuolar membranes and their contribution to K+ transport, J. Plant Physiol.
associated with metabolic flux redirection in rice, Nat. Commun. 11 (2020) 171 (2014) 732–742.
2629. [136] H.J. Kronzucker, D.T. Britto, Sodium transport in plants: a critical review, New
[111] T. Asano, N. Hayashi, M. Kobayashi, N. Aoki, A. Miyao, I. Mitsuhara, H. Phytol. 189 (2011) 54–81.
Ichikawa, S. Komatsu, H. Hirochika, S. Kikuchi, R. Ohsugi, A rice calcium- [137] M. Boudsocq, J. Sheen. Stress signaling II: calcium sensing and signaling, in: A.
dependent protein kinase OsCPK12 oppositely modulates salt-stress Pareek, S.K. Sopory, H.J. Bohnert, Govindjee (Eds), Abiotic Stress Adaptation in
tolerance and blast disease resistance, Plant J. 69 (2012) 26–36. Plants, Springer, Dordrecht, the Netherlands, 2009, pp. 75–90.
[112] S. Campo, P. Baldrich, J. Messeguer, E. Lalanne, M. Coca, B. San Segundo, [138] J.K. Zhu, Salt and drought stress signal transduction in plants, Annu. Rev.
Overexpression of a calcium-dependent protein kinase confers salt and Plant Biol. 53 (2002) 247–273.
drought tolerance in rice by preventing membrane lipid peroxidation, Plant [139] J.K. Zhu, Genetic analysis of plant salt tolerance using Arabidopsis, Plant
Physiol. 165 (2014) 688–704. Physiol. 124 (2000) 941–948.
[113] S.Q. Ouyang, Y.F. Liu, P. Liu, G. Lei, S.J. He, B. Ma, W.K. Zhang, J.S. Zhang, S.Y. [140] J. Liu, M. Ishitani, U. Halfter, C.S. Kim, J.K. Zhu, The Arabidopsis thaliana SOS2
Chen, Receptor-like kinase OsSIK1 improves drought and salt stress tolerance gene encodes a protein kinase that is required for salt tolerance, Proc. Natl.
in rice (Oryza sativa) plants, Plant J. 62 (2010) 316–329. Acad. Sci. U. S. A. 97 (2000) 3730–3734.
[114] Z. Zhang, H. Liu, C. Sun, Q. Ma, H. Bu, K. Chong, Y. Xu, A C2H2 zinc-finger [141] H. Shi, M. Ishitani, C. Kim, J.K. Zhu, The Arabidopsis thaliana salt tolerance gene
protein OsZFP213 interacts with OsMAPK3 to enhance salt tolerance in rice, J. SOS1 encodes a putative Na+/H+ antiporter, Proc. Natl. Acad. Sci. U. S. A. 97
Plant Physiol. 229 (2018) 100–110. (2000) 6896–6901.
[115] N. Lakra, K.K. Nutan, P. Das, K. Anwar, S.L. Singla-Pareek, A. Pareek, A nuclear- [142] G.Y. Xu, P.S. Rocha, M.L. Wang, M.L. Xu, Y.C. Cui, L.Y. Li, Y.X. Zhu, X. Xia, A
localized histone-gene binding protein from rice (OsHBP1b) functions in novel rice calmodulin-like gene, OsMSR2, enhances drought and salt
salinity and drought stress tolerance by maintaining chlorophyll content and tolerance and increases ABA sensitivity in Arabidopsis, Planta 234 (2011)
improving the antioxidant machinery, J. Plant Physiol. 176 (2015) 36–46. 47–59.
[116] N. Xu, Y. Chu, H. Chen, X. Li, Q. Wu, L. Jin, G. Wang, J. Huang, Rice transcription [143] B. Boonburapong, T. Buaboocha, Genome-wide identification and analyses of
factor OsMADS25 modulates root growth and confers salinity tolerance via the rice calmodulin and related potential calcium sensor proteins, BMC Plant
the ABA-mediated regulatory pathway and ROS scavenging, PLoS Genet. 14 Biol. 7 (2007) 4.
(2018) e1007662. [144] P. Jing, J. Zou, L. Kong, S. Hu, B. Wang, J. Yang, G. Xie, OsCCD1, a novel small
[117] A. Yang, X. Dai, W.H. Zhang, A R2R3-type MYB gene, OsMYB2, is involved in calcium-binding protein with one EF-hand motif, positively regulates
salt, cold, and dehydration tolerance in rice, J. Exp. Bot. 63 (2012) 2541–2556. osmotic and salt tolerance in rice, Plant Sci. 247 (2016) 104–114.
[118] Y. Tang, X. Bao, Y. Zhi, Q. Wu, Y. Guo, X. Yin, L. Zeng, J. Li, J. Zhang, W. He, W. [145] Z.M. Gu, H.J. Tang, X.F. Chen, L. Feng, H.S. ZHANG, Preliminary study on
Liu, Q. Wang, C. Jia, Z. Li, K. Liu, Overexpression of a MYB family gene, function of calcineurin B-like protein gene OsCBL8 in rice, Rice Sci. 17 (2010)
OsMYB6, increases drought and salinity stress tolerance in transgenic rice, 10–18.
Front. Plant Sci. 10 (2019) 168. [146] S. Saeng-ngam, W. Takpirom, T. Buaboocha, S. Chadchawan, The role of the
[119] L.G. Cui, J.X. Shan, M. Shi, J.P. Gao, H.X. Lin, DCA1 acts as a transcriptional Co- OsCam1-1 salt stress sensor in ABA accumulation and salt tolerance in rice, J.
activator of DST and contributes to drought and salt tolerance in rice, PLoS Plant Biol. 55 (2012) 198–208.
Genet. 11 (2015) e1005617. [147] Y. Saijo, S. Hata, J. Kyozuka, K. Shimamoto, K. Izui, Over-expression of a single
[120] X.Y. Huang, D.Y. Chao, J.P. Gao, M.Z. Zhu, M. Shi, H.X. Lin, A previously Ca2+-dependent protein kinase confers both cold and salt/drought tolerance
unknown zinc finger protein, DST, regulates drought and salt tolerance in rice on rice plants, Plant J. 23 (2000) 319–327.
via stomatal aperture control, Genes Dev. 23 (2009) 1805. [148] T. Asano, N. Hayashi, M. Kobayashi, N. Aoki, A. Miyao, I. Mitsuhara, H.
[121] G. Karimi, M. Ghorbanli, H. Heidari, R.A.K. Nejad, M.H. Assareh, The effects of Ichikawa, S. Komatsu, H. Hirochika, S. Kikuchi, R. Ohsugi, A rice calcium-
NaCl on growth, water relations, osmolytes and ion content in Kochia dependent protein kinase OsCPK12 oppositely modulates salt-stress
prostrata, Biol. Plant. 49 (2005) 301–304. tolerance and blast disease resistance, Plant J. 69 (2012) 26–36.
[122] Z.C. Chen, N. Yamaji, T. Horie, J. Che, J. Li, G. An, J.F. Ma, A magnesium [149] K. Kazan, Diverse roles of jasmonates and ethylene in abiotic stress tolerance,
transporter OsMGT1 plays a critical role in salt tolerance in rice, Plant Physiol. Trends Plant Sci. 20 (2015) 219–229.
174 (2017) 1837–1849. [150] B. van de Poel, D. Smet, D. van der Straeten, Ethylene and hormonal cross talk
[123] K.S. Kothari, P.K. Dansana, J. Giri, A.K. Tyagi, Rice stress associated protein 1 in vegetative growth and development, Plant Physiol. 169 (2015) 61–72.
(OsSAP1) interacts with aminotransferase (OsAMTR1) and pathogenesis- [151] S.K. Sah, K.R. Reddy, J. Li, Abscisic acid and abiotic stress tolerance in crop
related 1a protein (OsSCP) and regulates abiotic stress responses, Front. Plant plants, Front Plant Sci. 7 (2016) 571.
Sci. 7 (2016) 1057. [152] R. Finkelstein, Abscisic acid synthesis and response, Arabidopsis Book 11
[124] S. Gao, Y. Xiao, F. Xu, X. Gao, S. Cao, F. Zhang, G. Wang, D. Sanders, C. Chu, (2013) e0166.
Cytokinin-dependent regulatory module underlies the maintenance of zinc [153] K. Chen, G.J. Li, R.A. Bressan, C.P. Song, J.K. Zhu, Y. Zhao, Abscisic acid
nutrition in rice, New Phytol. 224 (2019) 202–215. dynamics, signaling, and functions in plants, J. Integr. Plant Biol. 62 (2020)
[125] A. Zhang, Y. Liu, F. Wang, T. Li, Z. Chen, D. Kong, J. Bi, F. Zhang, X. Luo, J. Wang, 25–54.
J. Tang, X. Yu, G. Liu, L. Luo, Enhanced rice salinity tolerance via CRISPR/Cas9- [154] R. Welsch, F. Wüst, C. Bär, S. Al-Babili, P. Beyer, A third phytoene synthase is
targeted mutagenesis of the OsRR22 gene, Mol. Breed. 39 (2019) 47. devoted to abiotic stress-induced abscisic acid formation in rice and defines
[126] H. Chen, N. Xu, Q. Wu, B. Yu, Y. Chu, X. Li, J. Huang, L. Jin, OsMADS27 regulates functional diversification of phytoene synthase genes, Plant Physiol. 147
the root development in a NO–3dependent manner and modulates the salt (2008) 367–380.
tolerance in rice (Oryza sativa L.), Plant Sci. 277 (2018) 20–32. [155] H.Y. Park, H.Y. Seok, B.K. Park, S.H. Kim, C.H. Goh, B.H. Lee, C.H. Lee, Y.H.
[127] P. Senguttuvel, C. Vijayalakshmi, K. Thiyagarajan, J. Kannanbapu, S. KOTA, G. Moon, Overexpression of Arabidopsis ZEP enhances tolerance to osmotic
Padmavathi, S. Geetha, N. Sritharan, B. Viraktamath, Changes in stress, Biochem. Biophys. Res. Commun. 375 (2008) 80–85.
photosynthesis, chlorophyll fluorescence, gas exchange parameters and [156] Z. Zhang, S. Ali, T. Zhang, W. Wang, L. Xie, Identification, evolutionary and
osmotic potential to salt stress during early seedling stage in rice (Oryza expression analysis of PYL-PP2C-SnRK2s gene families in soybean, Plants 9
sativa L.), SABRAO J. Breed. Genet. 46 (2014) 120–135. (2020) 1356.
[128] P. Das, K.K. Nutan, S.L. Singla-Pareek, A. Pareek, Understanding salinity [157] Y. Ma, I. Szostkiewicz, A. Korte, D. Moes, Y. Yang, A. Christmann, E. Grill,
responses and adopting ’omics-based’ approaches to generate salinity Regulators of PP2C phosphatase activity function as abscisic acid sensors,
tolerant cultivars of rice, Front. Plant Sci. 6 (2015) 712. Science 324 (2009) 1064–1068.
[129] M.M. Julkowska, H.C. Hoefsloot, S. Mol, R. Feron, G.J. de Boer, M.A. Haring, C. [158] S.Y. Park, P. Fung, N. Nishimura, D.R. Jensen, H. Fujii, Y. Zhao, S. Lumba, J.
Testerink, Capturing Arabidopsis root architecture dynamics with ROOT-FIT Santiago, A. Rodrigues, F.C. Tsz-fung, Abscisic acid inhibits type 2C protein
reveals diversity in responses to salinity, Plant Physiol. 166 (2014) 1387– phosphatases via the PYR/PYL family of START proteins, Science 324 (2009)
1402. 1068–1071.

23
C. Liu, B. Mao, D. Yuan et al. The Crop Journal 10 (2022) 13–25

[159] Y. Osakabe, K. Yamaguchi-Shinozaki, K. Shinozaki, L.S. Tran, ABA control of [184] W. Zong, N. Tang, J. Yang, L. Peng, S. Ma, Y. Xu, G. Li, L. Xiong, Feedback
plant macroelement membrane transport systems in response to water regulation of ABA Signaling and biosynthesis by a bZIP transcription factor
deficit and high salinity, New Phytol. 202 (2014) 35–49. targets drought-resistance-related genes, Plant Physiol. 171 (2016) 2810–
[160] K.E. Hubbard, N. Nishimura, K. Hitomi, E.D. Getzoff, J.I. Schroeder, Early 2825.
abscisic acid signal transduction mechanisms: newly discovered components [185] C. Yang, B. Ma, S.J. He, Q. Xiong, K.X. Duan, C.C. Yin, H. Chen, X. Lu, S.Y. Chen, J.
and newly emerging questions, Genes Dev. 24 (2010) 1695–1708. S. Zhang, MAOHUZI6/ETHYLENE INSENSITIVE3-LIKE1 and ETHYLENE
[161] B. BeldaPalazón, M. Adamo, C. Valerio, L.J. Ferreira, A. Confraria, D. ReisBarata, INSENSITIVE3-LIKE2 regulate ethylene response of roots and coleoptiles and
A. Rodrigues, C. Meyer, P.L. Rodriguez, E. BaenaGonzález, A dual function of negatively affect salt tolerance in rice, Plant Physiol. 169 (2015) 148–165.
SnRK2 kinases in the regulation of SnRK1 and plant growth, Nat. Plants 6 [186] C. Yang, X. Lu, B. Ma, S.Y. Chen, J.S. Zhang, Ethylene signaling in rice and
(2020) 1345–1353. Arabidopsis: conserved and diverged aspects, Mol. Plant 8 (2015) 495–505.
[162] S. Cai, G. Chen, Y. Wang, Y. Huang, D.B. Marchant, Y. Wang, Q. Yang, F. Dai, A. [187] C.H. Li, G. Wang, J.L. Zhao, L.Q. Zhang, L.F. Ai, Y.F. Han, D.Y. Sun, S.W. Zhang, Y.
Hills, P.J. Franks, E. Nevo, D.E. Soltis, P.S. Soltis, E. Sessa, P.G. Wolf, D. Xue, G. Sun, The receptor-like kinase SIT1 mediates salt sensitivity by activating
Zhang, B.J. Pogson, M.R. Blatt, Z.H. Chen, Evolutionary conservation of ABA MAPK3/6 and regulating ethylene homeostasis in rice, Plant Cell 26 (2014)
signaling for stomatal closure, Plant Physiol. 174 (2017) 732–747. 2538–2553.
[163] Y. Huang, Y. Guo, Y. Liu, F. Zhang, Z. Wang, H. Wang, F. Wang, D. Li, D. Mao, S. [188] T.S. Serra, D.D. Figueiredo, A.M. Cordeiro, D.M. Almeida, T. Lourenço, I.A.
Luan, M. Liang, L. Chen, 9-cis-epoxycarotenoid dioxygenase 3 regulates plant Abreu, A. Sebastián, L. Fernandes, B. Contreras-Moreira, M.M. Oliveira, N.J.
growth and enhances multi-abiotic stress tolerance in rice, Front. Plant Sci. 9 Saibo, OsRMC, a negative regulator of salt stress response in rice, is regulated
(2018) 162. by two AP2/ERF transcription factors, Plant Mol. Biol. 82 (2013) 439–455.
[164] Y. Huang, Y. Jiao, N. Xie, Y. Guo, F. Zhang, Z. Xiang, R. Wang, F. Wang, Q. Gao, L. [189] J. Zhuang, H.H. Jiang, F. Wang, R.H. Peng, Q.H. Yao, A.S. Xiong, A rice OsAP23,
Tian, D. Li, L. Chen, M. Liang, OsNCED5, a 9-cis-epoxycarotenoid dioxygenase functioning as an AP2/ERF transcription factor, reduces salt tolerance in
gene, regulates salt and water stress tolerance and leaf senescence in rice, transgenic Arabidopsis, Plant Mol. Biol. Rep. 31 (2013) 1336–1345.
Plant Sci. 287 (2019) 110188. [190] D. Liu, X. Chen, J. Liu, J. Ye, Z. Guo, The rice ERF transcription factor OsERF922
[165] H. Kim, K. Lee, H. Hwang, N. Bhatnagar, D.Y. Kim, I.S. Yoon, M.O. Byun, S.T. negatively regulates resistance to Magnaporthe oryzae and salt tolerance, J.
Kim, K.H. Jung, B.G. Kim, Overexpression of PYL5 in rice enhances drought Exp. Bot. 63 (2012) 3899–3911.
tolerance, inhibits growth, and modulates gene expression, J. Exp. Bot. 65 [191] V. Jisha, L. Dampanaboina, J. Vadassery, A. Mithöfer, S. Kappara, R. Ramanan,
(2014) 453–464. Overexpression of an AP2/ERF type transcription factor OsEREBP1 confers
[166] H. Kim, H. Hwang, J.W. Hong, Y.N. Lee, I.P. Ahn, I.S. Yoon, S.D. Yoo, S. Lee, S.C. biotic and abiotic stress tolerance in rice, PLoS ONE 10 (2015) e0127831.
Lee, B.G. Kim, A rice orthologue of the ABA receptor, OsPYL/RCAR5, is a [192] H. Qin, J. Wang, X. Chen, F. Wang, P. Peng, Y. Zhou, Y. Miao, Y. Zhang, Y. Gao, Y.
positive regulator of the ABA signal transduction pathway in seed Qi, J. Zhou, R. Huang, Rice OsDOF15 contributes to ethylene-inhibited primary
germination and early seedling growth, J. Exp. Bot. 63 (2012) 1013–1024. root elongation under salt stress, New Phytol. 223 (2019) 798–813.
[167] A. Singh, S.K. Jha, J. Bagri, G.K. Pandey, ABA inducible rice protein phosphatase [193] Y. Cao, Y. Wu, Z. Zheng, F.J.P. Song, M.P. Pathology, Overexpression of the rice
2C confers ABA insensitivity and abiotic stress tolerance in Arabidopsis, PLoS EREBP-like gene OsBIERF3 enhances disease resistance and salt tolerance in
ONE 10 (2015) e0125168. transgenic tobacco, Physiol. Mol. Plant Pathol. 67 (2005) 202–211.
[168] M. Amir Hossain, Y. Lee, J.I. Cho, C.H. Ahn, S.K. Lee, J.S. Jeon, H. Kang, C.H. Lee, [194] Y. Cao, F. Song, R.M. Goodman, Z. Zheng, Molecular characterization of four
G. An, P.B. Park, The bZIP transcription factor OsABF1 is an ABA responsive rice genes encoding ethylene-responsive transcriptional factors and their
element binding factor that enhances abiotic stress signaling in rice, Plant expressions in response to biotic and abiotic stress, J. Plant Physiol. 163
Mol, Biol. 72 (2010) 557–566. (2006) 1167–1178.
[169] M.A. Hossain, J. Cho, M. Han, C. Ahn, J. Jeon, G. An, P.B. Park, The ABRE-binding [195] C. Shan, Z. Mei, J. Duan, H. Chen, H. Feng, W. Cai, OsGA2ox5, a gibberellin
bZIP transcription factor OsABF2 is a positive regulator of abiotic stress and metabolism enzyme, is involved in plant growth, the root gravity response
ABA signaling in rice, J. Plant Physiol. 167 (2010) 1512–1520. and salt stress, PLoS ONE 9 (2014) e87110.
[170] A. RoyChoudhury, B. Gupta, D.N. Sengupta, Trans-acting factor designated [196] W. Mo, W. Tang, Y. Du, Y. Jing, Q. Bu, R. Lin, Phytochrome-interacting factor-
OSBZ8 interacts with both typical abscisic acid responsive elements as well like14 and slender rice1 interaction controls seedling growth under salt
as abscisic acid responsive element-like sequences in the vegetative tissues stress, Plant Physiol. 184 (2020) 506–517.
of indica rice cultivars, Plant Cell Rep. 27 (2008) 779–794. [197] S.F. Lo, M.L. Cheng, Y.C. Hsing, Y.S. Chen, K.W. Lee, Y.F. Hong, Y. Hsiao, A.S.
[171] K. Mukherjee, A.R. Choudhury, B. Gupta, S. Gupta, D.N. Sengupta, An ABRE- Hsiao, P.J. Chen, L.I. Wong, N.C. Chen, C. Reuzeau, T.D. Ho, S.M. Yu, Rice Big
binding factor, OSBZ8, is highly expressed in salt tolerant cultivars than in Grain 1 promotes cell division to enhance organ development, stress
salt sensitive cultivars of indica rice, BMC Plant Biol. 6 (2006) 18. tolerance and grain yield, Plant Biotechnol. J. 18 (2020) 1969–1983.
[172] H. Nakagawa, K. Ohmiya, T. Hattori, A rice bZIP protein, designated OSBZ8, is [198] K. Xia, R. Wang, X. Ou, Z. Fang, C. Tian, J. Duan, Y. Wang, M. Zhang, OsTIR1 and
rapidly induced by abscisic acid, Plant J. 9 (1996) 217–227. OsAFB2 downregulation via OsmiR393 overexpression leads to more tillers,
[173] Z. Tao, Y. Kou, H. Liu, X. Li, J. Xiao, S. Wang, OsWRKY45 alleles play different early flowering and less tolerance to salt and drought in rice, PLoS ONE 7
roles in abscisic acid signalling and salt stress tolerance but similar roles in (2012) e30039.
drought and cold tolerance in rice, J. Exp. Bot. 62 (2011) 4863–4874. [199] C. Mao, J. He, L. Liu, Q. Deng, X. Yao, C. Liu, Y. Qiao, P. Li, F. Ming, OsNAC2
[174] H. Hu, M. Dai, J. Yao, B. Xiao, X. Li, Q. Zhang, L. Xiong, Overexpressing a NAM, integrates auxin and cytokinin pathways to modulate rice root development,
ATAF, and CUC (NAC) transcription factor enhances drought resistance and Plant Biotechnol. J. 18 (2020) 429–442.
salt tolerance in rice, Proc. Natl. Acad. Sci. U. S. A. 103 (2006) 12987–12992. [200] R. Joshi, K.K. Sahoo, A.K. Tripathi, R. Kumar, B.K. Gupta, A. Pareek, S.L. Singla-
[175] J. Shen, B. Lv, L. Luo, J. He, C. Mao, D. Xi, F. Ming, The NAC-type transcription Pareek, Knockdown of an inflorescence meristem-specific cytokinin oxidase -
factor OsNAC2 regulates ABA-dependent genes and abiotic stress tolerance in OsCKX2 in rice reduces yield penalty under salinity stress condition, Plant
rice, Sci. Rep. 7 (2017) 40641. Cell Environ. 41 (2018) 936–946.
[176] Y. Hong, H. Zhang, L. Huang, D. Li, F. Song, Overexpression of a stress- [201] W.C. Wang, T.C. Lin, J. Kieber, Y.C. Tsai, Response regulators 9 and 10
responsive nac transcription factor gene ONAC022 improves drought and salt negatively regulate salinity tolerance in rice, Plant Cell Physiol. 60 (2019)
tolerance in rice, Front. Plant Sci. 7 (2016) 4. 2549–2563.
[177] N. Zhu, S. Cheng, X. Liu, H. Du, M. Dai, D.X. Zhou, W. Yang, Y. Zhao, The R2R3- [202] W. Yin, Y. Xiao, M. Niu, W. Meng, L. Li, X. Zhang, D. Liu, G. Zhang, Y. Qian, Z.
type MYB gene OsMYB91 has a function in coordinating plant growth and salt Sun, R. Huang, S. Wang, C.M. Liu, C. Chu, H. Tong, ARGONAUTE2 enhances
stress tolerance in rice, Plant Sci. 236 (2015) 146–156. grain length and salt tolerance by activating BIG GRAIN3 to modulate
[178] H. Xiong, J. Li, P. Liu, J. Duan, Y. Zhao, X. Guo, Y. Li, H. Zhang, J. Ali, Z. Li, cytokinin distribution in rice, Plant Cell 32 (2020) 2292–2306.
Overexpression of OsMYB48-1, a novel MYB-related transcription factor, [203] Y. Xiao, D. Liu, G. Zhang, S. Gao, L. Liu, F. Xu, R. Che, Y. Wang, H. Tong, C. Chu,
enhances drought and salinity tolerance in rice, PLoS ONE 9 (2014) e92913. Big Grain3, encoding a purine permease, regulates grain size via modulating
[179] D.E. Zeng, P. Hou, F. Xiao, Y. Liu, Overexpressing a novel RING-H2 finger cytokinin transport in rice, J. Integr. Plant Biol. 61 (2019) 581–597.
protein gene, OsRHP1, enhances drought and salt tolerance in rice (Oryza [204] L. Sun, Q. Zhang, J. Wu, L. Zhang, X. Jiao, S. Zhang, Z. Zhang, D. Sun, T. Lu, Y.
sativa L.), J. Plant Biol. 57 (2014) 357–365. Sun, Two rice authentic histidine phosphotransfer proteins, OsAHP1 and
[180] M. Zou, Y. Guan, H. Ren, F. Zhang, F. Chen, A bZIP transcription factor, OsABI5, OsAHP2, mediate cytokinin signaling and stress responses in rice, Plant
is involved in rice fertility and stress tolerance, Plant Mol. Biol. 66 (2008) Physiol. 165 (2014) 335–345.
675–683. [205] P. Tian, J. Liu, C. Mou, C. Shi, H. Zhang, Z. Zhao, Q. Lin, J. Wang, J. Wang, X.
[181] Y. Zhang, H. Lan, Q. Shao, R. Wang, H. Chen, H. Tang, H. Zhang, J. Huang, An Zhang, X. Guo, Z. Cheng, S. Zhu, Y. Ren, C. Lei, H. Wang, J. Wan, GW5-Like, a
A20/AN1-type zinc finger protein modulates gibberellins and abscisic acid homolog of GW5, negatively regulates grain width, weight and salt resistance
contents and increases sensitivity to abiotic stress in rice (Oryza sativa), J. in rice, J. Integr. Plant Biol. 61 (2019) 1171–1185.
Exp. Bot. 67 (2016) 315–326. [206] K. Xia, X. Ou, H. Tang, R. Wang, P. Wu, Y. Jia, X. Wei, X. Xu, S.H. Kang, S.K. Kim,
[182] A. Bhattacharjee, J.P. Khurana, M. Jain, Characterization of rice homeobox M. Zhang, Rice microRNA osa-miR1848 targets the obtusifoliol 14a-
genes, OsHOX22 and OsHOX24, and over-expression of OsHOX24 in transgenic demethylase gene OsCYP51G3 and mediates the biosynthesis of
arabidopsis suggest their role in abiotic stress response, Front. Plant Sci. 7 phytosterols and brassinosteroids during development and in response to
(2016) 627. stress, New Phytol. 208 (2015) 790–802.
[183] S. Zhang, I. Haider, W. Kohlen, L. Jiang, H. Bouwmeester, A.H. Meijer, H. [207] S. Koh, S.C. Lee, M.K. Kim, J.H. Koh, S. Lee, G. An, S. Choe, S.R. Kim, T-DNA
Schluepmann, C.M. Liu, P.B. Ouwerkerk, Function of the HD-Zip I gene tagged knockout mutation of rice OsGSK1, an orthologue of Arabidopsis BIN2,
Oshox22 in ABA-mediated drought and salt tolerances in rice, Plant Mol. Biol. with enhanced tolerance to various abiotic stresses, Plant Mol. Biol. 65 (2007)
80 (2012) 571–585. 453–466.

24
C. Liu, B. Mao, D. Yuan et al. The Crop Journal 10 (2022) 13–25

[208] K. Kurotani, K. Hayashi, S. Hatanaka, Y. Toda, D. Ogawa, H. Ichikawa, Y. Takeda, RICE SALT SENSITIVE3 forms a ternary complex with JAZ and class-C
Ishimaru, R. Tashita, T. Suzuki, M. Ueda, T. Hattori, S. Takeda, Elevated levels bHLH factors and regulates jasmonate-induced gene expression and root cell
of CYP94 family gene expression alleviate the jasmonate response and elongation, Plant Cell 25 (2013) 1709–1725.
enhance salt tolerance in rice, Plant Cell Physiol. 56 (2015) 779–789. [228] Z. Qiu, J. Guo, A. Zhu, L. Zhang, M. Zhang, Exogenous jasmonic acid can
[209] H. Wu, H. Ye, R. Yao, T. Zhang, L. Xiong, OsJAZ9 acts as a transcriptional enhance tolerance of wheat seedlings to salt stress, Ecotoxicol. Environ. Saf.
regulator in jasmonate signaling and modulates salt stress tolerance in rice, 104 (2014) 202–208.
Plant Sci. 232 (2015) 1–12. [229] D.J. Kang, Y.J. Seo, J.D. Lee, R. Ishii, K. Kim, D. Shin, S. Park, S. Jang, I.J. Lee,
[210] P.K. Peethambaran, R. Glenz, S. Höninger, S.M. Shahinul Islam, S. Hummel, K. Jasmonic acid differentially affects growth, ion uptake and abscisic acid
Harter, Ü. Kolukisaoglu, D. Meynard, E. Guiderdoni, P. Nick, M. Riemann, Salt- concentration in salt-tolerant and salt-sensitive rice cultivars, J. Agron. Crop
inducible expression of OsJAZ8 improves resilience against salt-stress, BMC Sci. 191 (2005) 273–282.
Plant Biol. 18 (2018) 311. [230] C.C. Filgueiras, A.D. Martins, R.V. Pereira, D.S. Willett, The ecology of salicylic
[211] T. Yamamoto, Y. Yoshida, K. Nakajima, M. Tominaga, A. Gyohda, H. Suzuki, T. acid signaling: primary, secondary and tertiary effects with applications in
Okamoto, T. Nishimura, N. Yokotani, E. Minami, Y. Nishizawa, K. Miyamoto, agriculture, Int. J. Mol. Sci. 20 (2019) 5851.
H. Yamane, K. Okada, T. Koshiba, Expression of RSOsPR10 in rice roots is [231] M.A. Ahanger, U. Aziz, A.A. Alsahli, M.N. Alyemeni, P. Ahmad, Influence of
antagonistically regulated by jasmonate/ethylene and salicylic acid via the exogenous salicylic acid and nitric oxide on growth, photosynthesis, and
activator OsERF87 and the repressor OsWRKY76, respectively, Plant Direct 2 ascorbate-glutathione cycle in salt stressed Vigna angularis, Biomolecules 10
(2018) e00049. (2019) 42.
[212] K. Takeuchi, H. Hasegawa, A. Gyohda, S. Komatsu, T. Okamoto, K. Okada, T. [232] F. Palma, M. López-Gómez, N.A. Tejera, C. Lluch, Salicylic acid improves the
Terakawa, T. Koshiba, Overexpression of RSOsPR10, a root-specific rice PR10 salinity tolerance of Medicago sativa in symbiosis with Sinorhizobium meliloti
gene, confers tolerance against drought stress in rice and drought and salt by preventing nitrogen fixation inhibition, Plant Sci. 208 (2013) 75–82.
stresses in bentgrass, Plant Cell Tissue Organ. Cult. 127 (2016) 35–46. [233] M. Keisham, S. Mukherjee, S.C. Bhatla, Mechanisms of sodium transport in
[213] B. Ma, S.J. He, K.X. Duan, C.C. Yin, H. Chen, C. Yang, Q. Xiong, Q.X. Song, X. Lu, plants-progresses and challenges, Int. J. Mol. Sci. 19 (2018) 647.
H.W. Chen, Identification of rice ethylene-response mutants and [234] J.Z. Zhang, R.A. Creelman, J.K. Zhu, From laboratory to fieldUsing information
characterization of MHZ7/OsEIN2 in distinct ethylene response and yield from Arabidopsis to engineer salt, cold, and drought tolerance in crops, Plant
trait regulation, Mol. Plant 6 (2013) 1830–1848. Physiol. 135 (2004) 615–621.
[214] H. Ellouzi, K. Ben Hamed, I. Hernández, J. Cela, M. Müller, C. Magné, C. [235] S. Negrão, M.C. Almadanim, I.S. Pires, I.A. Abreu, J. Maroco, B. Courtois, G.B.
Abdelly, S. Munné-Bosch, A comparative study of the early osmotic, ionic, Gregorio, K.L. McNally, M.M. Oliveira, New allelic variants found in key rice
redox and hormonal signaling response in leaves and roots of two halophytes salt-tolerance genes: an association study, Plant Biotechnol. J. 11 (2013) 87–
and a glycophyte to salinity, Planta 240 (2014) 1299–1317. 100.
[215] H. Ma, L. Song, Y. Shu, S. Wang, J. Niu, Z. Wang, T. Yu, W. Gu, H. Ma, [236] M. Al Nayef, C. Solis, L. Shabala, T. Ogura, Z. Chen, J. Bose, F.J.M. Maathuis, G.
Comparative proteomic analysis of seedling leaves of different salt tolerant Venkataraman, K. Tanoi, M. Yu, M. Zhou, T. Horie, S. Shabala, Changes in
soybean genotypes, J. Proteomics 75 (2012) 1529–1546. expression level of OsHKT1;5 alters activity of membrane transporters
[216] M.E. Ghanem, A. Albacete, C. Martínez-Andújar, M. Acosta, R. Romero- involved in K+ and Ca2+ acquisition and homeostasis in salinized rice roots,
Aranda, I.C. Dodd, S. Lutts, F. Pérez-Alfocea, Hormonal changes during Int. J. Mol. Sci. 21 (2020) 4882.
salinity-induced leaf senescence in tomato (Solanum lycopersicum L.), J. Exp. [237] C. Theerawitaya, T. Samphumphuang, R. Tisarum, M. Siangliw, S. Cha-Um, T.
Bot. 59 (2008) 3039–3050. Takabe, T. Toojinda, Expression level of Na+ homeostasis-related genes and
[217] A. Albacete, C. Martínez-Andújar, M.E. Ghanem, M. Acosta, J. Sánchez-Bravo, salt-tolerant abilities in backcross introgression lines of rice crop under salt
M.J. Asins, J. Cuartero, S. Lutts, I.C. Dodd, F. Pérez-Alfocea, Rootstock- stress at reproductive stage, Protoplasma 257 (2020) 1595–1606.
mediated changes in xylem ionic and hormonal status are correlated with [238] S. Negrão, B. Courtois, N. Ahmadi, I. Abreu, N. Saibo, M. Oliveira, Recent
delayed leaf senescence, and increased leaf area and crop productivity in updates on salinity stress in rice: from physiological to molecular responses,
salinized tomato, Plant Cell Environ. 32 (2009) 928–938. Crit. Rev. Plant Sci. 30 (2011) 329–377.
[218] M. Zhang, J.A. Smith, N.P. Harberd, C. Jiang, The regulatory roles of ethylene [239] S.L. Krishnamurthy, P. Pundir, A.S. Warraich, S. Rathor, B.M. Lokeshkumar, N.
and reactive oxygen species (ROS) in plant salt stress responses, Plant Mol. K. Singh, P.C. Sharma, Introgressed saltol QTL lines improves the salinity
Biol. 91 (2016) 651–659. tolerance in rice at seedling stage, Front. Plant Sci. 11 (2020) 833.
[219] J.J. Tao, H.W. Chen, B. Ma, W.K. Zhang, S.Y. Chen, J.S. Zhang, The role of [240] V.K. Singh, B.D. Singh, A. Kumar, S. Maurya, S.G. Krishnan, K.K. Vinod, M.P.
ethylene in plants under salinity stress, Front. Plant Sci. 6 (2015) 1059. Singh, R.K. Ellur, P.K. Bhowmick, A.K. Singh, Marker-assisted introgression of
[220] E.H. Colebrook, S.G. Thomas, A.L. Phillips, P. Hedden, The role of gibberellin saltol QTL enhances seedling stage salt tolerance in the rice variety ‘‘Pusa
signalling in plant responses to abiotic stress, J. Exp. Biol. 217 (2014) 67–75. Basmati 1”, Int. J. Genomics 2018 (2018) 8319879.
[221] J. Zhou, Z. Li, G. Xiao, M. Zhai, X. Pan, R. Huang, H. Zhang, CYP71D8L is a key [241] S.A. Ganie, M.J. Borgohain, K. Kritika, A. Talukdar, D.R. Pani, T.K. Mondal,
regulator involved in growth and stress responses by mediating gibberellin Assessment of genetic diversity of Saltol QTL among the rice (Oryza sativa L.)
homeostasis in rice, J. Exp. Bot. 71 (2020) 1160–1170. genotypes, Physiol. Mol. Biol. Plants 22 (2016) 107–114.
[222] B. Lv, Z. Yan, H. Tian, X. Zhang, Z. Ding, Local auxin biosynthesis mediates [242] H. Walia, C. Wilson, P. Condamine, X. Liu, A.M. Ismail, L. Zeng, S.I.
plant growth and development, Trends Plant Sci. 24 (2019) 6–9. Wanamaker, J. Mandal, J. Xu, X. Cui, T.J. Close, Comparative transcriptional
[223] R. Nishiyama, Y. Watanabe, Y. Fujita, D.T. Le, M. Kojima, T. Werner, R. profiling of two contrasting rice genotypes under salinity stress during the
Vankova, K. Yamaguchi-Shinozaki, K. Shinozaki, T. Kakimoto, H. Sakakibara, vegetative growth stage, Plant Physiol. 139 (2005) 822–835.
T. Schmülling, L.S. Tran, Analysis of cytokinin mutants and regulation of [243] A. Younis, F. Ramzan, Y. Ramzan, F. Zulfiqar, M. Ahsan, K.B. Lim, Molecular
cytokinin metabolic genes reveals important regulatory roles of cytokinins in markers improve abiotic stress tolerance in crops: a review, Plants 9 (2020)
drought, salt and abscisic acid responses, and abscisic acid biosynthesis, Plant 1374.
Cell 23 (2011) 2169–2183. [244] H. Li, Z. Bao, X. Sun, Genomic selection and its application, Hereditas 33
[224] X. Wu, J. He, J. Chen, S. Yang, D. Zha, Alleviation of exogenous 6- (2011) 1308–1316 (in Chinese with English abstract).
benzyladenine on two genotypes of eggplant (Solanum melongena Mill.) [245] G. Charmet, L.G. Tran, J. Auzanneau, R. Rincent, S.J.P.o. Bouchet, BWGS, AR
growth under salt stress, Protoplasma 251 (2014) 169–176. package for genomic selection and its application to a wheat breeding
[225] U.K. Divi, T. Rahman, P. Krishna, Brassinosteroid-mediated stress tolerance in programme, PLoS ONE 15 (2020) e0222733.
Arabidopsis shows interactions with abscisic acid, ethylene and salicylic acid [246] S. Tyagi, R. Kumar, V. Kumar, S.Y. Won, P. Shukla, Engineering disease
pathways, BMC Plant Biol. 10 (2010) 151. resistant plants through CRISPR-Cas9 technology, GM Crops Food 12 (2021)
[226] I. Sharma, E. Ching, S. Saini, R. Bhardwaj, P.K. Pati, Exogenous application of 125–144.
brassinosteroid offers tolerance to salinity by altering stress responses in rice [247] A. Dey, M.K. Samanta, S. Gayen, S.K. Sen, M.K. Maiti, Enhanced gene
variety Pusa Basmati-1, Plant Physiol. Biochem. 69 (2013) 17–26. expression rather than natural polymorphism in coding sequence of the
[227] Y. Toda, M. Tanaka, D. Ogawa, K. Kurata, K. Kurotani, Y. Habu, T. Ando, K. OsbZIP23 determines drought tolerance and yield improvement in rice
Sugimoto, N. Mitsuda, E. Katoh, K. Abe, A. Miyao, H. Hirochika, T. Hattori, S. genotypes, PLoS ONE 11 (2016) e0150763.

25

You might also like