Next Article in Journal
Animal Nutrition and Productions
Previous Article in Journal
Method for Classifying Apple Leaf Diseases Based on Dual Attention and Multi-Scale Feature Extraction
Previous Article in Special Issue
Antigenotoxic and Antimutagenic Potentials of Proline in Allium cepa Exposed to the Toxicity of Cadmium
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Recent Developments in Rice Molecular Breeding for Tolerance to Heavy Metal Toxicity

1
Department of Crop Genetics and Breeding, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, China
2
College of Plant Science and Technology, Huazhong Agriculture University, Wuhan 430070, China
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Agriculture 2023, 13(5), 944; https://doi.org/10.3390/agriculture13050944
Submission received: 20 February 2023 / Revised: 26 March 2023 / Accepted: 21 April 2023 / Published: 25 April 2023
(This article belongs to the Special Issue Challenges and Side Effects of Heavy Metals in Agriculture)

Abstract

:
Heavy metal toxicity generally refers to the negative impact on the environment, humans, and other living organisms caused by exposure to heavy metals (HMs). Heavy metal poisoning is the accumulation of HMs in the soft tissues of organisms in a toxic amount. HMs bind to certain cells and prevent organs from functioning. Symptoms of HM poisoning can be life-threatening and not only cause irreversible damage to humans and other organisms; but also significantly reduce agricultural yield. Symptoms and physical examination findings associated with HM poisoning vary depending on the metal accumulated. Many HMs, such as zinc, copper, chromium, iron, and manganese, are present at extremely low levels but are essential for the functioning of plants. However, if these metals accumulate in the plants in sufficient concentrations to cause poisoning, serious damage can occur. Rice is consumed around the world as a staple food and incidents of HM pollution often occur in rice-growing areas. In many rice-producing countries, cadmium (Cd), arsenic (As), and lead (Pb) have been recognized as commonly widespread HMs contaminating rice fields worldwide. In addition to mining and irrigation activities, the use of fertilizers and pesticides has also contributed significantly to HM contamination of rice-growing soils around the world. A number of QTLs associated with HM stress signals from various intermediary molecules have been reported to activate various transcription factors (TFs). Some antioxidant enzymes have been studied which contribute to the scavenging of reactive oxygen species, ultimately leading to stress tolerance in rice. Genome engineering and advanced editing techniques have been successfully applied to rice to improve metal tolerance and reduce HM accumulation in grains. In this review article, recent developments and progress in the molecular science for the induction of HM stress tolerance, including reduced metal uptake, compartmentalized transportation, gene-regulated signaling, and reduced accumulation or diversion of HM particles to plant parts other than grains, are discussed in detail, with particular emphasis on rice.

1. Introduction

Heavy metal pollution is increasing globally, damaging the environment and posing serious health risks to humans and other living organisms [1]. Heavy metal (HM) toxicity is commonly referred to as pollution caused by some naturally occurring elements known as heavy metals [2]. They have high atomic masses and densities (higher than 4 g/cm3) that are cytotoxic above certain concentrations [3]. Toxic HMs are subdivided into essential HMs and non-essential HMs according to their requirements for normal functioning of life [4]. Arsenic (As), cadmium (Cd), lead (Pb), mercury (Hg), thallium (Tl), and chromium (Cr) are recognized as useless for the normal functioning of life and are classified as non-essential HM. On the other hand, copper (Cu), boron (B), zinc (Zn), iron (Fe), selenium (Se), molybdenum (Mo), and manganese (Mn) are essential for proper life functioning. These are mandatory HMs that are practically required for cell survival [5,6].
The root causes of HM pollution are often attributed to the rapid rise of urbanization, the reckless growth of industrialization, and its waste disposal mechanisms [6,7]. After the industrial revolution and economic globalization, there has been an exponential increase in the diversity of environmental pollutants from various anthropogenic sources [4,8]. Consequently, a series of emerging issues related to food security have become global issues, especially given their close relevance to agriculture and human health. Excessive HMs can affect soil microbiological balance and reduce its fertility [9,10]. All metals, even including essential HMs, inhibit plant growth and metabolism; and reduce yield when certain thresholds are exceeded (Table 1, Figure 1). Plants take up both essential and non-essential HMs through active or passive transport. Essential HMs (along with non-essential HMs) readily penetrate root cells and perform their major functions, but can also be transported to tissue/subcellular sites where they can damage cells and their functions [11]. Once part of the plant’s internal machinery, HMs are transported and accumulated in grains, leaves, and other plant parts, where they are consumed by animals, birds, and humans, causing serious health problems. In terms of human exposure, the risks of HM ingestion include permanent brain damage, cirrhosis of the liver, encephalopathy, dementia, hemorrhage, impaired renal function, alveolitis, bronchitis, emphysema, sperm motility, bone disorders, gastrointestinal malignancies, and cardiovascular illness [12,13]. In plants, HM interferes with many metabolic processes such as photosynthesis, water balance, and nutrient absorption, resulting in plant growth retardation, grain yield losses (Table 1), and even death [14].
Rice, an important food source in many parts of Asia, is currently being severely damaged by the repeated use of HM-rich pesticides, excess water, and sludge waste (as organic fertilizer), creating reservoirs of HMs in the topsoil [15,16]. HMs can accumulate in the soil through the leaching processes and dissolve in groundwater, contaminating groundwater as well as surface water [17]. Several investigative reports indicate that the consumption of rice grown in fields high in Potentially Toxic Elements (PTEs), i.e., contaminants exceeding maximum allowable concentrations, can endanger human health [18,19]. Many researchers have reported that toxic metals such as As, Cd, and Pb along with other essential metals, e.g., Fe, Zn, and Cu are transported from soil and stored in rice grains as well as other plant parts such as the husks, leaves, and stems that are directly or indirectly edible. The accumulation of As and Cd in rice grains has become a serious problem worldwide [20,21]. Under moderately dry soil conditions, excessive Zn and Cd uptake can negatively affect Fe uptake, which also affects photosynthesis and yield [22,23]. Bioaccumulation of HMs in rice leads to biomagnification in the food chain, which can be dangerous as HM causes severe disease in humans [22,23]. Low levels of zinc have been reported to be linked to various physiological functions in the human body, while high levels can potentially offer major health hazards [24]. Likewise, an excess Cu can cause diarrhea, nausea, and Wilson’s disease [19]. Therefore, understanding the underlying mechanisms of HM uptake by plants, HM-mediated signaling and distribution in plants, and further exploring various breeding techniques effective in minimizing metal buildup in rice grains is crucial from the perspective of human health. In light of this, this review offers a summary of the most recent research on efforts to reduce HM bioaccumulation in rice grains as well as efficient breeding techniques for developing metal-tolerant rice varieties.
Table 1. Rice yield loss due to different HM toxicities reported in different studies.
Table 1. Rice yield loss due to different HM toxicities reported in different studies.
Heavy MetalDose/Concentration
(mg/kg)
Yield Loss
(%)
Reference
Pb400–120046–69[25]
As24.540[26]
Cd50–15034–63[27]
Fe385–119716–78[28]
Cu100–100010–90[29]
Zn-20–40[30]
Ni40–10054–70[31]
Al-30–60[32]
Cr200–40030–48[32]
Hg4–550–70[33]

2. Natural and Anthropogenic Sources of HM Toxicity in Rice

Depending on the mode of origin, the bioaccumulation of HMs in the environment may be either anthropogenic (caused by humans) or natural. Anthropogenic sources can be subdivided into two main sources, agricultural waste and municipal or industrial waste (Figure 2). Rice grains actively accumulate HMs from contaminated soil. Soil characteristics are, therefore, regarded as the most significant influencing element [34]. Accumulation of HMs in agricultural soils tends to increase with weathering of metal-enriched rocks and other soil-forming processes [35]. HMs can be mobilized beyond the buffering capacity of soils due to saturation or changes in environmental conditions such as land exploitation, agricultural inputs, and climate change [36].
Some chemical fertilizers, such as phosphate fertilizers, can introduce HM into the soil. Trace amounts of HM are also present in chemical fertilizers because they occur as impurities in the raw materials and natural minerals used to manufacture these fertilizers [37]. Phosphogypsum from waste phosphate fertilizers can form a variety of toxic HMs in the soil, which can be transferred to plants. Later these soils become enriched mainly in toxic HMs (e.g., Cd, Cr, and Pb). Excessive application of cadmium-rich phosphorus fertilizers can also be a source of cadmium pollution in rice fields [38]. Consumption of rice from rice fields contaminated with HMs can be a potential source of exposure to toxic HMs, especially cadmium, lead, and arsenic [39]. Consumers may be exposed to health risks and illnesses if they ingest rice that has been farmed in contaminated fields over an extended period of time [40]. A recent study found that high concentrations of Cd, Hg, Pb, and Zn in soil were positively correlated with As in rice grains, and that As, Cr, Hg, Pb, and Zn increased the Hg content of rice, suggesting that the presence of these elements also promote the accumulation of toxic HMs such as As and Hg in rice [41]. The increasing use of large amounts of organic manure in paddy fields around the world has been identified as a source of HMs in the soil. It was also discovered that the amount of arsenic in rice grains increased when rice was cultivated in soils with high concentrations of As and FYM [42].
To determine the potential sources of HMs and their buildup in agricultural soils, several research investigations have been conducted in different countries. According to a survey report, phosphate fertilizers tested included traces (mg/kg) of 5.6–17.2 Pb, 19.4–273.0 As, 9.5–96.4 Cd, and 0.01–0.42 Hg, and ammonium phosphate, ammonium nitrate, and compound fertilizers tested positive for 50–60 mg/kg of As [43]. Another study found that phosphate fertilizers were a significant source of heavy metals (HMs) entering agricultural soils in England and Wales, particularly zinc, copper, and cadmium. In contrast, soils in Argentina were frequently fertilized with inorganic phosphate fertilizers, which led to the presence of cadmium, chromium, copper, zinc, nickel, and lead [44]. Today, organic fertilizers also contain HMs that find their way into drinking water or are added to bedding (such as straw) in paddy fields around the world. Therefore, the long-term excessive application of organic fertilizers containing HMs such as copper and zinc will also lead to their accumulation in agricultural soils.
An extensive study in eastern China investigated HM levels in urban soils and found that high concentrations of HMs in soil were mainly concentrated in central and western regions with relatively few spatial variations [45]. Municipal solid waste, automobile emissions, combustion of coal, and HM emissions from human activities including vehicle exhaust are all partially responsible for the rise of lead isotopes in urban soils. Whether for adults or children, the risk of HM contamination from soil when ingested is much higher than when it comes in contact with the skin. Another study conducted in Tangchang in 2022 to determine HM contamination (HM) in agricultural soil collected 788 samples of topsoil and analyzed their HM concentrations [46]. It was further reported that transport and deposition could be sources of Pb and Hg; Zn, Cr, and Ni could come from natural resources and industrial activities, while Cu and Cd came from agricultural activities. These studies additionally indicate that the location of mines is the primary factor influencing the environmental distribution of Zn, Ni, Pb, As, and Cd [3].

3. Progress in Molecular Breeding to Develop HMT Tolerance in Rice

The problem of HM toxicity has emerged as one of the global risks to sustainable agricultural output due to the rapid increase in the accumulation of HMs from various sources in agricultural soils during the past few decades. Agricultural experts have expressed interest in further investigating the underlying mechanisms that allow plants to resist HM toxicity [47]. Given a thorough understanding of HM transport and deposition in various plant organelles, researchers have proposed a number of experimental methods using innovative molecular approaches that can assist rice plants develop HM tolerance. In particular, detoxification, transport, and/or sequestration are the primary objectives of HM control techniques [48]. Fluid transport from roots to other plant parts involves water transpiration, root pressure, cation exchange in the cell walls of xylem vessels, formation of complexes with amino acids (Cu), histidine, peptides (Ni), and chelates with organic acids (Zn) [49]. Accordingly, the concentrations of most HMs gradually decreased with distance from the root [37]. These elements are transported inside the plant by cell wall charge interactions and the formation of soluble organic complexes in the sap [50].

3.1. Role of HM Transporters (HMTs) and Tolerant Proteins (HMPs)

Previously reported P1B subfamily of HM transporters P-type ATPases (HMAs) affect HM uptake and transport in different plants [51,52,53]. The researchers found upregulation of HMA genes and retrotransposons in rice after HM treatment and intergenerational inheritance of altered expressions [54]. This study demonstrated how rice plants respond to HM stress by altering locus-specific gene expression and trans-generational inheritance due to altered gene regulation, even after HM stress has been eliminated. Fu et al. linked a G-type ATP-binding cassette transporter OsABCG36 to Cd tolerance in rice. Cd-induced OsABCG36 was expressed localized to the plasma membrane in protoplast cells of the root tip and mature roots. Knockdown of OsABCG36 increased Cd accumulation in roots and enhanced sensitivity to Cd stress without affecting tolerance to other HMs (i.e., Al, Zn, Cu, and Pb) [55].
Metal tolerance proteins (MTPs), another type of membrane protein, have also been demonstrated to be involved in metal transport and to impart tolerance to certain HMs in plants. In a comprehensive molecular study of rice MTP genes, three thousand rice genotypes were investigated using available genome-wide resequencing information, highlighting the evolution and allelic diversity of MTP genes [56]. These findings indicated that MTP1, MTP8.1 and MTP9, MTP11 localize to QTL/m-QTL for Zn and Cd accumulation, respectively. MTP9 and MTP8.1 had specific root and shoot expression, respectively, at all stages of the rice plants. After getting exposed to toxic HMs, it was determined that some seed-specific MTPs regulated metal transport during the seed-filling stage downregulated the expression of the majority of MTP genes in the roots, and upregulated those in the shoots, indicating that MTP employs various mechanisms in different tissues.
Certain HM-associated proteins (HMPs) have been reported to be involved in HM detoxification. In earlier studies, the novel genes were found to regulate a protein known as cell number regulatory factor (CNR) in plants e.g., TaCNR2 in wheat and ZmCNR1 in maize [57]. This protein, like the cadmium-tolerant protein in plants, modulates cell number and HM transport. Overexpression of TaCNR2 in rice also increases stress resistance to Cd, Zn, and Mn, and their translocation from roots to shoots. TaCNR2 has been shown through experimental studies to transport HM ions, providing another source of genes that improve nutrient absorption and decrease the buildup of hazardous metals in rice plants [58]. Li et al. also identified 46 HMPs in rice, which they named OsHMPs 1–46 according to their chromosomal locations. Studies have shown that HMPs are governed by different TFs, and only 8 OsHMPs are assembled in rice tissues. Among them, OsHMP37 had a much higher expression level than the other seven HMPs, whereas OsHMP28 was solely expressed in the roots. Only OsHMP09, OsHMP18, and OsHMP22 showed increased expression levels in all tissues, despite the fact that most of the selected OsHMPs were variably expressed in different tissues under varying HM exposures [45]. Approximately 43 putative Fe–S cluster assembly genes have also been identified in the rice (Oryza sativa) genome and the expression of all genes has been validated [59,60]. Liang et al. investigated the role of Fe–S cluster assembly in leaf chloroplasts are particularly sensitive to HM treatment, and genes encoding Fe–S cluster assembly in roots are sensitive to iron toxicity, oxidative stress, and HM stress; and are shown to be upregulated in response [60]. OsHMA2 is a major Zn/Cd transporter identified in rice roots and has been experimentally shown to promote Zn/Cd transfer from roots to shoots. [61].

3.2. Role of microRNAs (miRNAs)

In several investigations, high-throughput sequencing and miRNA microarray data have indicated the involvement of several miRNAs in the responses of various plant species to toxic HMs [62]. Overexpression of miRNAs or the development of miRNA-resistant target genes have been widely utilized to demonstrate the role of metal-responsive miRNAs, and these miRNAs and their target genes are an essential component of a large-scale regulatory network controlling different metabolic processes in response to metal stress [63,64]. Several regulatory networks involving miRNAs and their TFs have been studied [35]. Ding et al., identified twelve (12) Cd-responsive miRNAs in rice using miRNA microarray assays. Expression of another Cd-responsive miRNA (miR166) was significantly suppressed under Cd exposure at the seedling stage and its overexpression reduced Cd-induced oxidative stress [65]. Overexpression of miR166 also reduced root-to-shoot Cd translocation and Cd accumulation in rice grains [61,66].

3.3. QTL and Fine Gene Mapping

Researchers have extensively used genetic markers in rice crops to identify genomic loci involved in various plant physiological functions under simulated stress conditions and associated tolerance mechanisms [67,68]. Breeders have used the same approach to identify QTLs associated with specific HM accumulation, translocation, and tolerance strategies in rice to reduce HM toxicity [69]. Arsenic is the most toxic of all HM and rice absorbs more arsenic than other crops [70]. Once the crop has absorbed HM through its roots, it is distributed throughout the plant and then transferred to the rice grain [71]. The International Agency for Research on Cancer (IARC) declared As, Cd, and Cr to be highly carcinogenic and can damage DNA by disrupting DNA synthesis/repair mechanisms and resulting in neuropsychiatric disorders [72]. Due to climate change, more arsenic is expected to be present in the environment as more rain releases arsenic and other HM previously trapped in mining areas [73]. Ingestion of 1 mg of arsenic per day can affect the skin for several years [74]. Therefore, As has now become the most priority research area among plant breeders. Numerous QTL mapping and genome-wide association studies (GWAS) have documented a number of QTLs/candidate genes that confer toxicity tolerance and HM accumulation in rice (Oryza sativa L.) [3]. In an investigation, an early backcross population from a cross between WTR1 (indica) and Haoannon (japonica) was grown hydroponically and exposed to As (10 ppm) for seven days and genotyped using 704 SNP markers. One QTL for relative chlorophyll content was identified on chromosome 1, two QTLs for arsenic content in roots were discovered on chromosome 8, and six QTLs for arsenic content in shoots were identified on chromosomes 2, 5, 6, and 9 [75].
Tyagi et al. [76] found that the tolerant genotypes accumulated less Al and showed a minimal reduction in root-shoot biomass under Al stress compared to susceptible genotypes. Transcriptomic data indicated that tolerant genotypes conserved energy by down-regulating key glycolytic pathway genes in roots, maintaining transcription levels of key energy-releasing enzymes, up-regulated signaling, and regulatory transcripts encoding zinc finger proteins and cell wall-associated transcripts under Al stress. Stein et al. [77] also designed a similar study to identify candidate genes for iron toxicity tolerance by investigating the effect of excess iron on two rice cultivars with apparent tolerance to iron toxicity. These results suggest that physiological and anatomical changes and HM permeability in various parts of rice may be related to toxic tolerance.
Shilin et al. [78] also investigated the genetic mechanism of Cd tolerance in rice, using a RIL population derived from crossing two parents (i.e., PA64s and 93–11) to analyze the associated QTLs to cadmium tolerance at the seedling stage. Two QTLs, i.e., qCDSL1.1 and qCDSL1.2 were identified in Hangzhou and Lingshui, respectively. The Chromosomal Segment Replacement Line, CSSLqCDSL1 was developed in the context of parent 93–11 which contained qCDSL1.1/qCDSL1 from parent PA64. Under Cd stress conditions, CSSLqCDSL1 had a longer shoot length compared to 93–11, demonstrating tolerance to Cd toxicity. In another investigation by Maghrebi et al. [79] two major rice cultivars (i.e., Capataz and Beirao) with different Cd tolerance were exposed to various Cd concentrations (0.01, 0.1, and 1 μM) and their potential capacities to sequester and transfer Cd. QTL mapping for toxic metal/metalloid stress tolerance in rice identified three QTLs associated with As, one associated with Cu and Hg, and two associated with Fe and Zn content.
In a genome-wide association study (GWAS), 188 different cultivated rice germplasms were examined at the seedling stage under normal and Cd stress conditions. By combining GWAS data, transcriptome analysis, database gene annotation intelligent Gene Ontology (GO), and homologous gene annotation and function, 148 candidate Cd-mediated growth response (CGR) genes were obtained [21]. In another GWA study, the correlation between arsenic and eight essential ions in the rice germplasm population was analyzed and it was shown that the association between arsenic and other essential ions was affected by growing conditions and various genetic factors. A cis-eQTL for AIR2 (arsenic-induced RING finger protein) was isolated by transcriptome-wide association studies, and the expression level of AIR2 was confirmed to be lower in indica than in japonica. By genome-wide analysis, arsenic-associated QTLs were discovered on chromosomes 5 and 6 under submerged and intermittent submerged conditions [70].
Another GWA study on low Cd accumulation in rice identified the OsABCB24 gene as the basis of a new QTL (qCd1-3) [80]. GWAS of trace iron and zinc in rice grains revealed novel associations of marker traits with 2.1–53.0% phenotypic variation, which may help identify candidate genes for increased iron tolerance and zinc [81]. Fe toxicity in rice crops was also investigated using a GWAS approach, and three linkage disequilibrium (LD) blocks were found to contribute primarily to Fe omission on chromosomes 1, 2, 3, 4, and 7 [82]. Other GWA studies found 22, 17, and 21 QTLs in rice associated with As, Cd, and Pb toxicity, respectively. To identify genomic areas regulating the covariance between mineral elements in the rice genome, Liu et al. used multivariate QTL analysis and principal component analysis (PCA). They sequenced the entire genome of rice RILs and identified potential candidate loci under the QTL clusters, including OsHMA4 and OsNRAMP5 [83].

3.4. HM Tolerant Transgenic Rice

Over the past decade, genetically modified (GM) crops have been officially adopted in many countries around the world, with their coverage increasing rapidly every year. Therefore, extensive research has been conducted on genetically modified (GM) rice in particular, focusing on the development of rice species tolerant to various biotic and abiotic factors [84]. Researchers are now also focusing on developing genetically modified rice by inserting new genes, which could reduce the buildup of HM in rice grains [85,86]. For instance, glutathione peroxidase (PgGPx) was discovered to impart salt and drought resistance to transgenic rice plants in a previous investigation [85]. The peroxidase gene family plays an essential role in maintaining ion homeostasis because it transports metal ions, including Cd [87,88]. Elaborated the role of PgGPx on Cd stress in rice and found that PgGPx transcript level was strongly up-regulated in response to exogenous Cd level. Under Cd stress, overexpression of PgGPx in transgenic rice improves control of ROS scavenging pathways and cellular ion homeostasis maintenance.
Another multigene family of plant-specific peroxidases (Class III) is involved in various physiological and developmental processes and tolerates abiotic stresses and HM such as aluminum, zinc, cadmium, and copper by removing ROS and RNS [89]. In a study, Kidwai et al. [90] identified a class III peroxidase (OsPRX38) from rice that was consistently increased in response to arsenate (As) and arsenite stress. Overexpression of OsPRX38 in transgenic rice significantly increased arsenic (As) tolerance by regulating lignin biosynthesis which acts as an apoplast barrier for As entry into root cells, resulting in reduced As accumulation in transgenics. Another protein (MTH1745) from the thermophilic archaea Methanothermobacter thermoautotrophicum has already been shown to prevent citrate synthase aggregation after heat denaturation and to play an important role in heat stress resistance [35]. Overexpression of MTH1745 in transgenic rice plants also increased mercury tolerance and increased photosynthesis rate compared to non-transgenic rice. Chen et al. [91] & Ding et al. [92] down-regulated the expression of miR5144-3p and overexpressed OsPDIL1-1, respectively, in transgenic rice plants which significantly improved mercury stress tolerance.
Researchers improved rice varieties low in As, Cd, and Cs by eliminating the genes (such as OsNRAMP5, OsNRAMP1, OsARM1, and OsHAK1) [93,94]. Other transporters such as OsNRAMP1 have been identified as a mediator of Cd and Mn uptake [95]. Overexpression of another gene, LmMTP1, metal resistance protein 1 (MTP1), isolated from ryegrass, resulted in increased resistance to Zn, Co, and Cd in transgenic lines [1]. Overexpression of microRNA (miR166) enhanced tolerance to Cd toxicity in transgenic rice plants thereby reducing Cd accumulation in grains [65]. Plants overexpressing OsHB4 have been shown to have increased sensitivity to cadmium and accumulate cadmium in their leaves. Conversely, OsHB4 silencing increased the tolerance of transgenic plants to Cd.
In a study, to limit arsenic in the roots of rice plants as a detoxification mechanism, the phytochelatin synthase CdPCS1 from cornflower, an arsenic-accumulating aquatic plant, was transgenically expressed [86]. Increased arsenic accumulation in roots and shoots was observed in transgenic rice lines. However, compared to non-transgenic plants, all transgenic lines accumulated much less arsenic in grain and hulls. Huang et al. [96] identified OsHMA4 in yeast as a possible causative gene of a QTL controlling Cu accumulation in rice grains and provided evidence that OsHMA4 functions as a Cu chelate in root vacuoles, limiting the accumulation of Cu in grains. The resulting transgenic lines were more resistant to HM stress than the nontransgenic or wild type.
Gu et al. isolated DEFENSIN 8 (DEF8), a defensin-like gene expressed in rice grains, and suggested that DEF8 promotes Cd loading into the xylem and mediates Cd translocation from root to shoot and subsequent distribution in the grain [97]. DEF8 is also a modulator of Cd unloading to the phloem without affecting the accumulation of essential mineral nutrients and key crop traits [98]. Li [45] reported that low cadmium accumulation rice lines could be constructed by overexpressing a truncated OsO3L2 gene. Researchers have successfully developed rice plants that reduce cadmium accumulation by overexpressing the rice genes OsO3L2, OsO3L3, or their truncated versions [62]. Since the tissue localization of both these genes showed high expression in roots, it seems possible that roots are involved in low Cd accumulation. According to another study, transgenic rice plants overexpressing V-PPase accumulated more cadmium in the roots than in the shoots [99]. Similarly, another mutant line overexpressing the OsHIPP16 (OE) gene significantly improved rice growth under Cd toxicity stress [100].

3.5. CRISPR/Cas Genome Editing

In the past decade, advanced genome engineering technology, CRISPR-Cas9 has also been used to improve detoxification efficiency in rice by targeting HM-specific genes [101,102] (Table 2). Gene function and the way in which it affects other biochemical processes are changed by indel mutations and targeted substitutions generated through the CRISPR/Cas9 system. The OsNRAMP1 gene that controls the uptake of various HMs (including Cd, Fe, As, and Mn) in various crops was successfully deleted by the gene editing approach, which also greatly reduced the uptake and storage of Cd and As in the grains of rice [103]. Cd accumulation in rice was studied by deleting a segment of the OsABCG36 gene by CRISPR/Cas9 technology. Cd tolerance developed in knockout mutants as a result of Cd accumulation in root cells and excretion of the Cd content from the cytosol to detoxify its effects [55]. The OsLsi gene family has been reported to be involved in Si xylem loading in rice roots, which is required for efficient Si transport from roots to stems. CRISPR-Cas-based inactivation of this gene (OsLsi) resulted in reduced Si uptake in xylem sap and also reduced As accumulation in rice [33]. Similarly, CRISPR/Cas9 has been used to knock down OsNramp5 and OsLCT1 to reduce cadmium (Cd) accumulation [104] and inactivate the low cesium (Cs) K+ transporter OsHAK1 in rice plants [94]. Overexpression of gene OsLCT2 (a low-affinity cation transporter) using CRISPR-Cas9 also reduced Cd accumulating in rice grains [105]. More recently other genome-editing technologies, such as CRISPR-Cas12a, CRISPR-directed evolution, and base-editors, have also succeeded in more efficiently multiplex genome-editing in rice [60].

4. Breeding Tools for Improving HM Toxicity Tolerance in Rice

4.1. Physiological Screening

The study of natural variation in related traits is of great importance for the breeding and development of new crop varieties. Previous studies have identified wide genetic diversity for various HMs (e.g., Zn, Cu, Cd, Pb, Ni, As, and Sn) based on physiological criteria (e.g., relative shoot weight, root weight, and chlorophyll) [106]. One of the most effective screening strategies for developing HM-tolerant rice genotypes is to test for HM tolerance using valid screening systems (e.g., a hydroponic system) to estimate the tolerance level. The copper tolerance of 16 hydroponic rice cultivars was studied and four rice genotypes (Xiushui 123, Xiushui 134, Jiahe 991, and Xianghu 301) showed improvements in root length, root dehydrogenase activity, and root dry weight under copper stress, and the screened genotypes were used for local cultivation [107]. Another study found that under arsenic stress, the rice variety BRRI-29 from the Bangladesh Rice Research Institute (BRRI) had the best germination rate, biomass, and longest roots [108]. Genotypes with higher proline content, leaf chlorophyll content, and flag leaf area are also considered effective physiological screening indicators for lead tolerance in rice. Natural rice populations grown around mine sites have been reported to be more tolerant to HM [109], and their use in breeding programs to increase HM tolerance is being studied.

4.2. Mutation Breeding

One of the main approaches to understanding plant responses to HM is to use mutants with altered sensitivity or tolerance to HMs. Mutation breeding is an important tool for generating genetic diversity for desired traits, including the development of new varieties with increased tolerance to HM [110]. In several mutagenesis studies, functionally deficient mutants of some HM tolerance genes were successfully obtained, which significantly reduced the accumulation of the associated HM in different parts of the rice plants [111]. Common methods of mutagenesis include EMS mutagenesis, space mutagenesis, and irradiation mutagenesis [112]. The expression of these genes has been silenced or modified by knocking out their alleles or promoters to study their effect on HM accumulation or translocation efficiency in plants, which can be used to develop genotypes with reduced HM accumulation, making them tolerant to HM toxicity. Several transporter genes have also been identified to mediate HM uptake, transport, or distribution in rice plants [93,95,113].

4.3. Molecular Gene Mapping

Different molecular studies such as genome-wide association studies (GWAS) have been widely used as effective tools to exploit elite alleles controlling important agronomic traits in rice germplasm [38,39] (Table 3). Researchers have also used GWAS in several molecular screening studies to detect a large number of QTLs for HM accumulation in rice grains collected from rice germplasm [41,42]. Several molecular studies have investigated the response of rice plants to high HM stress [114]. Based on GWAS and transcriptome analysis, tolerance germplasm resources were investigated, and superior natural variants associated with HM resistance were identified [113,115]. Collectively, the identified QTLs serve as a resource to potentially improve tolerance to HM toxicity in today’s major cultivars and provide useful information for the use of these QTLs/genes in future breeding programs.

4.4. Gene Silencing

Several investigation reports have been published regarding the development of new rice lines with low HM accumulation in grains by knocking out the HM transporter genes using different gene silencing techniques such as RNA interference (RNAi) [124], CRISPR/Cas9 system [93,125] and transgenics. Li et al. [124] successfully reduced cadmium (Cd) accumulation in rice seeds by inhibiting the expression of the phytochelatin synthase (PCS) gene OsPCS1 using RNA interference (RNAi). Two mutant lines (pmei12-J3 and pmei12-J8) were obtained by editing the OsPMEI12 gene using CRISPR/Cas9 editing technology, and the cadmium stress resistance of the pmei12 line was found to be significantly improved [30]. Similarly, silencing of genes such as OsNramp5 and OsLCT1 has been reported to reduce Cd accumulation [104] and OsHAK1 (low Cs K+ transporter) [94] to attenuate Cs accumulation in rice. Related studies on genome editing via CRISPR-Cas technology are shown in Table 2 above.

5. Challenges and Limitations in Breeding for HM-Tolerant Rice

Compared to other abiotic stresses, HM toxicity tolerance is genetically more complex and involves many components of signal transduction and translocation pathways that are polygenic in nature. Therefore, it is relatively more difficult to control and reproduce [126]. Moreover, genetic manipulation of HM-responsive genes, metabolites, and proteins has yielded unexpected results, but its potential has not been fully exploited due to complex genetics and insufficient understanding of their epigenetic behavior [127]. Several other factors, such as environmental influences and the significant interrelationships among various HMs, have been reported to negatively affect the outcome of effective breeding for the development of HM-tolerant plants [128]. Several metal-uptake and transporter genes are associated with more than one HM. For example, OsNRAMP1 and OsNRAMP5 are involved in the uptake/transport of cadmium, iron, and manganese [95], and gene editing of HM tolerance targeting a specific HM can also lead to nutritional deficiencies due to insufficient absorption of other essential metals. To counter this, some weak alleles of HM transporter genes (such as OsNRAMP5) have also been identified that confer low Cd uptake without causing Mn deficiency in rice [105]. Field screening of HM for tolerance to specific HMs is also very difficult due to other factors such as the large breeding population size, associated field management, and labor costs [129]. In recent decades, considerable work has been done on the detoxification of HMs in soils through phytoremediation, chemicals, bio-chemicals, and microorganisms [124]. However, these strategies are not cost-effective due to the large area on which rice and other crops are grown. Although many measures have been taken to reduce the release of HMs into the environment, the accumulation of HMs in the environment continues to increase rapidly [130].

6. Conclusions and Future Prospects

As the population grew, the demand for rice cultivation, which required fertile land and large amounts of water, increased dramatically. Unfortunately, a lot of toxic metals such as, arsenic, lead, and cadmium, some of which are carcinogenic pollutants with a long half-life and are not biodegradable, are contaminating rice fields. Phytoremediation (also known as detoxification of polluted environments) has been widely studied by different researchers to show its potential against HM stress. However, developing natural plant tolerance to HM is more sustainable, less expensive, and more environmentally friendly. Therefore, there is an urgent need to search for new genes to develop rice varieties that are more tolerant to HM toxicity to avoid drastic yield decline due to HM accumulation in the soil and fatal diseases caused by exposure to humans and other organisms.
This review summarizes recent developments and advances in molecular science to increase our understanding of the genetic mechanisms of adaptive resistance in the context of HM toxicity and to accelerate the development of rice genotypes more tolerant to HM toxicity. To date, tolerance to some HM has been shown to be moderate and insufficient to fully mitigate the future impact of HM on food security and human health. Additionally, efforts have been made to increase the tolerance of rice to HM stress to reduce the uptake and accumulation of HM in the grains. However, the complete identification of the master transcriptional regulators that activate the entire signal transduction cascade is still pending. Using molecular approaches to select climate-tolerant plants with excess metal accumulation may open up new possibilities to combat HM toxicity.
In recent years, various advanced technologies and molecular breeding tools have been widely used to better understand the absorption, transport, and tolerance mechanisms of HMs in rice, in order to breed new rice varieties with better tolerance to HM toxicity. Recently, more advanced genome engineering technologies such as CRISPR-Cas9 CRISPR-Cas12, Base Editing (BE), etc. have also been used to improve the efficiency of HM detoxification by modifying various biochemical pathways. Similar to this, other transcriptome approaches have been employed to examine and quantify the abundance of important transcripts in cells from various rice parts. Studies have demonstrated that changes in local gene expression caused by stress in various tissues can result in the generation of novel stress proteins, stress-mediated metabolic compounds, or genes encoding TFs, enhancing the efficiency of plants in regulating HM-stress responses.
An emerging area of research with great potential for commercial application is the creation of genetically modified plants with an excellent ability to chelate certain metals and prevent their deleterious effects. Further research is needed to find specific transporters for specific HMs to facilitate the development of rice varieties tolerant to specific HMs. In the future, bioremediation of contaminated sites could benefit from the coordinated use of traditional breeding techniques combined with molecular biology. Additionally, the use of genome sequencing to identify genes associated with metal tolerance may pave the way for the construction of transgenes with desired properties that can be used in plant extraction techniques. These findings, coupled with in-depth evolutionary studies of putative genes associated with HM tolerance, could yield encouraging results.

Author Contributions

Conceptualization, Y.G.; Investigation, Z.H., I.A. and S.Z.; Resources, Y.G., Z.H. and I.A.; Data Curation, Z.H. and I.A.; Writing—Original Draft Preparation, Z.H., I.A. and S.Z.; Writing—Review & Editing, Y.G., Z.H. and I.A.; Supervision, Y.G.; Funding Acquisition, Y.G. All authors have read and agreed to the published version of the manuscript.

Funding

The research was funded by Zhejiang Provincial Natural Science Foundation of China (Grant No. LZ22C130002); National Key R & D Program of China (2021YFF1000400; 2022YFD1200403); and National Natural Science Foundation of China (Grant No. U2202204).

Institutional Review Board Statement

Not applicable.

Data Availability Statement

Data sharing is not applicable to this article.

Acknowledgments

The authors acknowledge Zhejiang University and funding sources for the financial support in publishing this paper.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Tong, M.; Liu, X.; Guan, J.; Lin, Y.; Zhou, A.; Qiao, K. Novel biofortification candidate: MTP1 increases microelement contents and decreases toxic heavy metal accumulation in grains. Chemosphere 2023, 318, 137967. [Google Scholar] [CrossRef]
  2. Singh, A.; Sharma, A.; Verma, R.K.; Chopade, R.L.; Pandit, P.P.; Nagar, V.; Aseri, V.; Choudhary, S.K.; Awasthi, G.; Awasthi, K.K. Heavy metal contamination of water and their toxic effect on living organisms. In The Toxicity of Environmental Pollutants; Dorta, D., De Oliveira, D.P., Eds.; IntechOpen: London, UK, 2022. [Google Scholar]
  3. Zhang, W.; Liu, M.; Li, C. Soil heavy metal contamination assessment in the Hun-Taizi River watershed, China. Sci. Rep. 2020, 10, 8730. [Google Scholar] [CrossRef]
  4. Shen, E.; Wang, X.; Lu, Z.; Zhou, F.; Ma, W.; Cui, Z.; Li, Z.; Li, C.; Lin, Y. Overexpression of a beta-1, 6-glucanase gene GluM in transgenic rice confers high resistance to rice blast, sheath blight and false smut. Pest Manag. Sci. 2023, 79, 7394. [Google Scholar] [CrossRef] [PubMed]
  5. Sodhi, K.K.; Mishra, L.C.; Singh, C.K.; Kumar, M. Perspective on the heavy metal pollution and recent remediation strategies. Curr. Res. Microb. Sci. 2022, 3, 100166. [Google Scholar] [CrossRef] [PubMed]
  6. Malinowska, E.; Jankowski, K. The effect of different doses of sewage sludge and liming on total cobalt content and its speciation in soil. Agronomy 2020, 10, 1550. [Google Scholar] [CrossRef]
  7. Bing, H.; Qiu, S.; Tian, X.; Li, J.; Zhu, H.; Wu, Y.; Zhang, G. Trace metal contamination in soils from mountain regions across China: Spatial distribution, sources, and potential drivers. Soil Ecol. Lett. 2021, 3, 189–206. [Google Scholar] [CrossRef]
  8. Tóth, G.; Hermann, T.; Da Silva, M.; Montanarella, L. Heavy metals in agricultural soils of the European Union with implications for food safety. Environ. Int. 2016, 88, 299–309. [Google Scholar] [CrossRef] [PubMed]
  9. Rai, P.K. Phytoremediation of Emerging Contaminants in Wetlands; CRC Press: Boca Raton, FL, USA, 2018. [Google Scholar]
  10. Giri, S.; Singh, A.K.; Mahato, M.K. Monte Carlo simulation-based probabilistic health risk assessment of metals in groundwater via ingestion pathway in the mining areas of Singhbhum copper belt, India. Int. J. Environ. Health Res. 2020, 30, 447–460. [Google Scholar] [CrossRef]
  11. Giri, S.; Mahato, M.K.; Bhattacharjee, S.; Singh, A.K. Development of a new noncarcinogenic heavy metal pollution index for quality ranking of vegetable, rice, and milk. Ecol. Indic. 2020, 113, 106214. [Google Scholar] [CrossRef]
  12. Ugulu, I.; Akhter, P.; Khan, Z.I.; Akhtar, M.; Ahmad, K. Trace metal accumulation in pepper (Capsicum annuum L.) grown using organic fertilizers and health risk assessment from consumption. Food Res. Int. 2021, 140, 109992. [Google Scholar] [CrossRef]
  13. Jin, T.; Nordberg, M.; Frech, W.; Dumont, X.; Bernard, A.; Ye, T.-T.; Kong, Q.; Wang, Z.; Li, P.; Lundström, N.-G. Cadmium biomonitoring and renal dysfunction among a population environmentally exposed to cadmium from smelting in China (ChinaCad). Biometals 2002, 15, 397–410. [Google Scholar] [CrossRef] [PubMed]
  14. Mayor, D.J.; Gray, N.B.; Elver-Evans, J.; Midwood, A.J.; Thornton, B. Metal-macrofauna interactions determine microbial community structure and function in copper contaminated sediments. PLoS ONE 2013, 8, e64940. [Google Scholar] [CrossRef] [PubMed]
  15. Kazberuk, W.; Szulc, W.; Rutkowska, B. Use bottom sediment to agriculture—Effect on plant and heavy metal content in soil. Agronomy 2021, 11, 1077. [Google Scholar] [CrossRef]
  16. Slaveykova, V.I.; Cheloni, G. Preface: Special issue on environmental toxicology of trace metals. Environments 2018, 5, 138. [Google Scholar] [CrossRef]
  17. Ayejoto, D.A.; Agbasi, J.C.; Egbueri, J.C.; Echefu, K.I. Assessment of oral and dermal health risk exposures associated with contaminated water resources: An update in Ojoto area, southeast Nigeria. Int. J. Environ. Anal. Chem. 2022, 1–21. [Google Scholar] [CrossRef]
  18. Mu, T.; Zhou, T.; Li, Z.; Hu, P.; Luo, Y.; Christie, P.; Wu, L. Prediction models for rice cadmium accumulation in Chinese paddy fields and the implications in deducing soil thresholds based on food safety standards. Environ. Pollut. 2020, 258, 113879. [Google Scholar] [CrossRef] [PubMed]
  19. Hussain, B.; Ashraf, M.N.; Abbas, A.; Li, J.; Farooq, M. Cadmium stress in paddy fields: Effects of soil conditions and remediation strategies. Sci. Total Environ. 2021, 754, 142188. [Google Scholar] [CrossRef]
  20. Sharma, L.K.; McCray, J.M.; Morgan, K. Plant Essential Nutrients and Their Role: SS-AGR-463/AG462, 5/2022. EDIS 2022, 2022, 3. [Google Scholar] [CrossRef]
  21. Yu, H.-Y.; Liu, C.; Zhu, J.; Li, F.; Deng, D.-M.; Wang, Q.; Liu, C. Cadmium availability in rice paddy fields from a mining area: The effects of soil properties highlighting iron fractions and pH value. Environ. Pollut. 2016, 209, 38–45. [Google Scholar] [CrossRef]
  22. Özyiğit, İ.İ.; Abakirova, A.; Hocaoğlu-Özyiğit, A.; Kurmanbekova, G.; Chekirov, K.; Yalcin, B.; Yalçin, İ.E. Cadmium stress in barley seedlings: Accumulation, growth, anatomy and physiology. Int. J. Life Sci. Biotechnol. 2021, 4, 204–223. [Google Scholar] [CrossRef]
  23. Slamet-Loedin, I.H.; Johnson-Beebout, S.E.; Impa, S.; Tsakirpaloglou, N. Enriching rice with Zn and Fe while minimizing Cd risk. Front. Plant Sci. 2015, 6, 121. [Google Scholar] [CrossRef]
  24. Sageena, G.; Khatana, K.; Nagar, J.K. Biomonitoring of heavy metals contamination in soil ecosystem. In Hazardous and Trace Materials in Soil and Plants; Elsevier: Amsterdam, The Netherlands, 2022; pp. 313–325. [Google Scholar]
  25. Ashraf, U.; Kanu, A.S.; Deng, Q.; Mo, Z.; Pan, S.; Tian, H.; Tang, X. Lead (Pb) Toxicity; Physio-Biochemical Mechanisms, Grain Yield, Quality, and Pb Distribution Proportions in Scented Rice. Front. Plant Sci. 2017, 8, 259. [Google Scholar] [CrossRef] [PubMed]
  26. Muehe, E.M.; Wang, T.; Kerl, C.F.; Planer-Friedrich, B.; Fendorf, S. Rice production threatened by coupled stresses of climate and soil arsenic. Nat. Commun. 2019, 10, 4985. [Google Scholar] [CrossRef] [PubMed]
  27. Kanu, A.S.; Ashraf, U.; Mo, Z.; Fuseini, I.; Mansaray, L.R.; Duan, M.; Pan, S.; Tang, X. Cadmium Uptake and Distribution in Fragrant Rice Genotypes and Related Consequences on Yield and Grain Quality Traits. J. Chem. 2017, 2017, 1405878. [Google Scholar] [CrossRef]
  28. Audebert, A.; Fofana, M. Rice Yield Gap due to Iron Toxicity in West Africa. J. Agron. Crop Sci. 2009, 195, 66–76. [Google Scholar] [CrossRef]
  29. Xu, J.; Yang, L.; Wang, Z.; Dong, G.; Huang, J.; Wang, Y. Toxicity of copper on rice growth and accumulation of copper in rice grain in copper contaminated soil. Chemosphere 2007, 62, 602–607. [Google Scholar] [CrossRef]
  30. Panhwar, Q.A.; Naher, U.A.; Radziah, O.; Shamshuddin, J.; Razi, I.M. Eliminating aluminum toxicity in an acid sulfate soil for rice cultivation using plant growth promoting bacteria. Molecules 2015, 20, 3628–3646. [Google Scholar] [CrossRef] [PubMed]
  31. Aziz, H.; Sabir, M.; Ahmad, H.R.; Aziz, T.; Zia-ur-Rehman, M.; Hakeem, K.R.; Ozturk, M. Alleviating Effect of Calcium on Nickel Toxicity in Rice. Clean Soil Air Water 2015, 43, 901–909. [Google Scholar] [CrossRef]
  32. AbdElgawad, H.; Mohammed, A.E.; van Dijk, J.R.; Beemster, G.T.S.; Alotaibi, M.O.; Saleh, A.M. The impact of chromium toxicity on the yield and quality of rice grains produced under ambient and elevated levels of CO2. Front. Plant Sci. 2023, 14, 1019859. [Google Scholar] [CrossRef]
  33. Hillary, V.E.; Ceasar, S.A. Prime editing in plants and mammalian cells: Mechanism, achievements, limitations, and future prospects. BioEssays 2022, 44, e2200032. [Google Scholar] [CrossRef]
  34. Xu, D.; Shen, Z.; Dou, C.; Dou, Z.; Li, Y.; Gao, Y.; Sun, Q. Effects of soil properties on heavy metal bioavailability and accumulation in crop grains under different farmland use patterns. Sci. Rep. 2022, 12, 9211. [Google Scholar] [CrossRef] [PubMed]
  35. Ding, Y.; Ding, L.; Xia, Y.; Wang, F.; Zhu, C. Emerging roles of microRNAs in plant heavy metal tolerance and homeostasis. J. Agric. Food Chem. 2020, 68, 1958–1965. [Google Scholar] [CrossRef] [PubMed]
  36. Kaur, R.; Das, S.; Bansal, S.; Singh, G.; Sardar, S.; Dhar, H.; Ram, H. Heavy metal stress in rice: Uptake, transport, signaling, and tolerance mechanisms. Physiol. Plant. 2021, 173, 430–448. [Google Scholar] [CrossRef]
  37. Wan, Y.; Camara, A.Y.; Yu, Y.; Wang, Q.; Guo, T.; Zhu, L.; Li, H. Cadmium dynamics in soil pore water and uptake by rice: Influences of soil-applied selenite with different water managements. Environ. Pollut. 2018, 240, 523–533. [Google Scholar] [CrossRef]
  38. Sebastian, A.; Prasad, M.N.V. Cadmium minimization in rice. A review. Agron. Sustain. Dev. 2014, 34, 155–173. [Google Scholar] [CrossRef]
  39. Jallad, K.N. Heavy metal exposure from ingesting rice and its related potential hazardous health risks to humans. Environ. Sci. Pollut. Res. 2015, 22, 15449–15458. [Google Scholar] [CrossRef]
  40. Ali, H.; Khan, E.; Ilahi, I. Environmental chemistry and ecotoxicology of hazardous heavy metals: Environmental persistence, toxicity, and bioaccumulation. J. Chem. 2019, 2019, 6730305. [Google Scholar] [CrossRef]
  41. Zhang, C.; Zou, X.; Yang, H.; Liang, J.; Zhu, T. Bioaccumulation and risk assessment of potentially toxic elements in soil-rice system in karst area, southwest china. Front. Environ. Sci. 2022, 10, 232. [Google Scholar] [CrossRef]
  42. Norton, G.J.; Adomako, E.E.; Deacon, C.M.; Carey, A.-M.; Price, A.H.; Meharg, A.A. Effect of organic matter amendment, arsenic amendment and water management regime on rice grain arsenic species. Environ. Pollut. 2013, 177, 38–47. [Google Scholar] [CrossRef]
  43. Lin, H.-J.; Sung, T.-I.; Chen, C.-Y.; Guo, H.-R. Arsenic levels in drinking water and mortality of liver cancer in Taiwan. J. Hazard. Mater. 2013, 262, 1132–1138. [Google Scholar] [CrossRef]
  44. Si, L.; Xie, Y.; Ma, Q.; Wu, L. The short-term effects of rice straw biochar, nitrogen and phosphorus fertilizer on rice yield and soil properties in a cold waterlogged paddy field. Sustainability 2018, 10, 537. [Google Scholar] [CrossRef]
  45. Li, Y.; Dong, Z.; Feng, D.; Zhang, X.; Jia, Z.; Fan, Q.; Liu, K. Study on the risk of soil heavy metal pollution in typical developed cities in eastern China. Sci. Rep. 2022, 12, 3855. [Google Scholar] [CrossRef]
  46. Gong, C.; Wang, S.; Wang, D.; Lu, H.; Dong, H.; Liu, J.; Yan, B.; Wang, L. Ecological and human health risk assessment of heavy metal (loid) s in agricultural soil in hotbed chives hometown of Tangchang, Southwest China. Sci. Rep. 2022, 12, 8563. [Google Scholar] [CrossRef]
  47. Shao, J.F.; Xia, J.; Yamaji, N.; Shen, R.F.; Ma, J.F. Effective reduction of cadmium accumulation in rice grain by expressing OsHMA3 under the control of the OsHMA2 promoter. J. Exp. Bot. 2018, 69, 2743–2752. [Google Scholar] [CrossRef]
  48. Colangelo, E.P.; Guerinot, M.L. Put the metal to the petal: Metal uptake and transport throughout plants. Curr. Opin. Plant Biol. 2006, 9, 322–330. [Google Scholar] [CrossRef] [PubMed]
  49. Greger, M. Metal availability, uptake, transport and accumulation in plants. Heavy Met. Stress Plants Biomol. Ecosyst. 2004, 1–27. [Google Scholar] [CrossRef]
  50. Thi, K.V.; Lan, P.D.T.; Hang, N.N.T.; Thanh, H.N. Cadmium Immobilization in the Rice–Paddy Soil with Biochar Additive. J. Ecol. Eng. 2022, 23. [Google Scholar]
  51. Krämer, U.; Talke, I.N.; Hanikenne, M. Transition metal transport. FEBS Lett. 2007, 581, 2263–2272. [Google Scholar] [CrossRef]
  52. Morel, M.; Crouzet, J.; Gravot, A.; Auroy, P.; Leonhardt, N.; Vavasseur, A.; Richaud, P. AtHMA3, a P1B-ATPase allowing Cd/Zn/co/Pb vacuolar storage in Arabidopsis. Plant Physiol. 2009, 149, 894–904. [Google Scholar] [CrossRef] [PubMed]
  53. Miyadate, H.; Adachi, S.; Hiraizumi, A.; Tezuka, K.; Nakazawa, N.; Kawamoto, T.; Katou, K.; Kodama, I.; Sakurai, K.; Takahashi, H. OsHMA3, a P1B-type of ATPase affects root-to-shoot cadmium translocation in rice by mediating efflux into vacuoles. New Phytol. 2011, 189, 190–199. [Google Scholar] [CrossRef]
  54. Cong, W.; Miao, Y.; Xu, L.; Zhang, Y.; Yuan, C.; Wang, J.; Zhuang, T.; Lin, X.; Jiang, L.; Wang, N. Transgenerational memory of gene expression changes induced by heavy metal stress in rice (Oryza sativa L.). BMC Plant Biol. 2019, 19, 1–14. [Google Scholar] [CrossRef] [PubMed]
  55. Fu, S.; Lu, Y.; Zhang, X.; Yang, G.; Chao, D.; Wang, Z.; Shi, M.; Chen, J.; Chao, D.-Y.; Li, R. The ABC transporter ABCG36 is required for cadmium tolerance in rice. J. Exp. Bot. 2019, 70, 5909–5918. [Google Scholar] [CrossRef] [PubMed]
  56. Ram, H.; Kaur, A.; Gandass, N.; Singh, S.; Deshmukh, R.; Sonah, H.; Sharma, T.R. Molecular characterization and expression dynamics of MTP genes under various spatio-temporal stages and metal stress conditions in rice. PLoS ONE 2019, 14, e0217360. [Google Scholar] [CrossRef] [PubMed]
  57. Guo, M.; Rupe, M.A.; Dieter, J.A.; Zou, J.; Spielbauer, D.; Duncan, K.E.; Howard, R.J.; Hou, Z.; Simmons, C.R. Cell Number Regulator1 affects plant and organ size in maize: Implications for crop yield enhancement and heterosis. Plant Cell 2010, 22, 1057–1073. [Google Scholar] [CrossRef]
  58. Qiao, K.; Wang, F.; Liang, S.; Wang, H.; Hu, Z.; Chai, T. Improved Cd, Zn and Mn tolerance and reduced Cd accumulation in grains with wheat-based cell number regulator TaCNR2. Sci. Rep. 2019, 9, 1–10. [Google Scholar] [CrossRef] [PubMed]
  59. Balk, J.; Pilon, M. Ancient and essential: The assembly of iron–sulfur clusters in plants. Trends Plant Sci. 2011, 16, 218–226. [Google Scholar] [CrossRef] [PubMed]
  60. Kar, S.; Panda, S.K. Iron homeostasis in rice: Deficit and excess. Proc. Natl. Acad. Sci. India Sect. B Biol. Sci. 2020, 90, 227–235. [Google Scholar] [CrossRef]
  61. Takahashi, R.; Ishimaru, Y.; Shimo, H.; Ogo, Y.; Senoura, T.; Nishizawa, N.K.; Nakanishi, H. The OsHMA2 transporter is involved in root-to-shoot translocation of Zn and Cd in rice. Plant Cell Environ. 2012, 35, 1948–1957. [Google Scholar] [CrossRef] [PubMed]
  62. Wang, B.; Cheng, D.; Chen, Z.; Zhang, M.; Zhang, G.; Jiang, M.; Tan, M. Bioinformatic exploration of the targets of xylem sap miRNAs in maize under cadmium stress. Int. J. Mol. Sci. 2019, 20, 1474. [Google Scholar] [CrossRef]
  63. Kumar, K.; Mandal, S.N.; Neelam, K.; Reyes, B.G.D.L. MicroRNA-mediated host defense mechanisms against pathogens and herbivores in rice: Balancing gains from genetic resistance with trade-offs to productivity potential. BMC Plant Biol. 2022, 22, 351. [Google Scholar] [CrossRef]
  64. Sasi, J.M.; Vijaya, K.C.; Kukreja, B.; Budhwar, R.; Shukla, R.N.; Agarwal, M.; Katiyar, A.S. Integrated transcriptomics and miRNAomics provide insights into the complex multi-tiered regulatory networks associated with coleoptile senescence in rice. Front. Plant Sci. 2022, 13, 985402. [Google Scholar] [CrossRef] [PubMed]
  65. Ding, Y.; Gong, S.; Wang, Y.; Wang, F.; Bao, H.; Sun, J.; Cai, C.; Yi, K.; Chen, Z.; Zhu, C. MicroRNA166 modulates cadmium tolerance and accumulation in rice. Plant Physiol. 2018, 177, 1691–1703. [Google Scholar] [CrossRef] [PubMed]
  66. Meng, J.G.; Zhang, X.D.; Tan, S.K.; Zhao, K.X.; Yang, Z.M. Genome-wide identification of Cd-responsive NRAMP transporter genes and analyzing expression of NRAMP1 mediated by miR167 in Brassica napus. Biometals 2017, 30, 917–931. [Google Scholar] [CrossRef] [PubMed]
  67. Wei, L.; Zhang, J.; Wang, C.; Liao, W. Recent progress in the knowledge on the alleviating effect of nitric oxide on heavy metal stress in plants. Plant Physiol. Biochem. 2020, 147, 161–171. [Google Scholar] [CrossRef] [PubMed]
  68. Anwar, A.; Kim, J.-K. Transgenic breeding approaches for improving abiotic stress tolerance: Recent progress and future perspectives. Int. J. Mol. Sci. 2020, 21, 2695. [Google Scholar] [CrossRef]
  69. Jahan, N.; Javed, M.A.; Anwar, K.; Tabassum, B.; Parveen, S.; Muzaffar, N. Multiple interval mapping of QTLs and epistasis for iron toxicity tolerance in segregating population of Indica rice. Not. Bot. Horti Agrobot. Cluj-Napoca 2022, 50, 12773. [Google Scholar] [CrossRef]
  70. Lee, S.-B.; Kim, G.-J.; Shin, J.-D.; Chung, W.; Moon, J.-K.; Choi, G.-H.; Park, Y.-J.; Park, S.-W. Genome-scale profiling and high-throughput analyses unravel the genetic basis of arsenic content variation in rice. Front. Plant Sci. 2022, 13, 2333. [Google Scholar] [CrossRef] [PubMed]
  71. Adeva, C.; Yun, Y.-T.; Shim, K.-C.; Luong, N.H.; Lee, H.-S.; Kang, J.-W.; Kim, H.-J.; Ahn, S.-N. QTL Mapping of Mineral Element Contents in Rice Using Introgression Lines Derived from an Interspecific Cross. Agronomy 2023, 13, 76. [Google Scholar] [CrossRef]
  72. Ginting, E.E.; Silalahi, J.; Putra, E.D.L. Analysis of Arsenic in Rice in Medan, North Sumatera Indonesia by Atomic Absorption Spectrophotometer. Orient. J. Chem. 2018, 34, 2651. [Google Scholar] [CrossRef]
  73. Change, F.C. Unpacking the Burden on Food Safety; FAO—Food and Agriculture Organization of the United Nations: Rome, Italy, 2020. [Google Scholar]
  74. Jansen, S.; Dera, R.T.S.; MargarethRuth, M.; Nanda, S.I.; Yosy, C.S. Analysis of arsenic in raw and cooked rice by atomic absorption spectrophotometer. In Proceedings of the IOP Conference Series: Earth and Environmental Science, Medan, Indonesia, 19–21 September 2018; p. 012040. [Google Scholar]
  75. Murugaiyan, V.; Ali, J.; Mahender, A.; Aslam, U.M.; Jewel, Z.A.; Pang, Y.; Marfori-Nazarea, C.M.; Wu, L.-B.; Frei, M.; Li, Z. Mapping of genomic regions associated with arsenic toxicity stress in a backcross breeding populations of rice (Oryza sativa L.). Rice 2019, 12, 1–14. [Google Scholar] [CrossRef]
  76. Tyagi, W.; Yumnam, J.S.; Sen, D.; Rai, M. Root transcriptome reveals efficient cell signaling and energy conservation key to aluminum toxicity tolerance in acidic soil adapted rice genotype. Sci. Rep. 2020, 10, 4580. [Google Scholar] [CrossRef] [PubMed]
  77. Stein, R.J.; Duarte, G.L.; Scheunemann, L.; Spohr, M.G.; de Araújo Júnior, A.T.; Ricachenevsky, F.K.; Rosa, L.M.G.; Zanchin, N.I.T.; dos Santos, R.P.; Fett, J.P. Genotype variation in rice (Oryza sativa L.) tolerance to Fe toxicity might be linked to root cell wall lignification. Front. Plant Sci. 2019, 10, 746. [Google Scholar] [CrossRef] [PubMed]
  78. Shilin, D.; Chaolei, L.; Lianguang, S.; Shenglong, Y.; Anpeng, Z.; Hongzhen, J.; Banpu, R.; Guonan, F.; Biao, T.; Guoyou, Y. Identification of QTLs for cadmium tolerance during seedling stage and validation of qCDSL1 in rice. Rice Sci. 2021, 28, 81–88. [Google Scholar] [CrossRef]
  79. Maghrebi, M.; Baldoni, E.; Lucchini, G.; Vigani, G.; Valè, G.; Sacchi, G.A.; Nocito, F.F. Analysis of cadmium root retention for two contrasting rice accessions suggests an important role for OsHMA2. Plants 2021, 10, 806. [Google Scholar] [CrossRef]
  80. Pan, X.; Li, Y.; Liu, W.; Liu, S.; Min, J.; Xiong, H.; Dong, Z.; Duan, Y.; Yu, Y.; Li, X. QTL mapping and candidate gene analysis of cadmium accumulation in polished rice by genome-wide association study. Sci. Rep. 2020, 10, 1–11. [Google Scholar] [CrossRef]
  81. Bollinedi, H.; Yadav, A.K.; Vinod, K.; Gopala Krishnan, S.; Bhowmick, P.K.; Nagarajan, M.; Neeraja, C.; Ellur, R.K.; Singh, A.K. Genome-wide association study reveals novel marker-trait associations (MTAs) governing the localization of Fe and Zn in the rice grain. Front. Genet. 2020, 11, 213. [Google Scholar] [CrossRef]
  82. Utami, D.D.; Rosdianti, I.; Chrisnawati, L.; Subardi, S.; Nurani, S.; Suwarno, S. Identification of iron tolerant candidate loci in rice determined through genome-wide association study. Indones. J. Agric. Sci. 2020, 21, 17–29. [Google Scholar] [CrossRef]
  83. Liu, H.; Long, S.-X.; Pinson, S.R.; Tang, Z.; Guerinot, M.L.; Salt, D.E.; Zhao, F.-J.; Huang, X.-Y. Univariate and multivariate QTL analyses reveal covariance among mineral elements in the rice ionome. Front. Genet. 2021, 12, 638555. [Google Scholar] [CrossRef]
  84. Sim, J.-E.; Oh, S.-D.; Kang, K.; Shin, Y.-M.; Yun, D.-W.; Baek, S.-H.; Choi, Y.-E.; Park, S.-U.; Kim, J.-K. Metabolite Profiling to Evaluate Metabolic Changes in Genetically Modified Protopanaxadiol-Enriched Rice. Plants 2023, 12, 758. [Google Scholar] [CrossRef]
  85. Islam, T.; Manna, M.; Reddy, M.K. Glutathione peroxidase of Pennisetum glaucum (PgGPx) is a functional Cd2+ dependent peroxiredoxin that enhances tolerance against salinity and drought stress. PLoS ONE 2015, 10, e0143344. [Google Scholar] [CrossRef]
  86. Shri, M.; Dave, R.; Diwedi, S.; Shukla, D.; Kesari, R.; Tripathi, R.D.; Trivedi, P.K.; Chakrabarty, D. Heterologous expression of Ceratophyllum demersum phytochelatin synthase, CdPCS1, in rice leads to lower arsenic accumulation in grain. Sci. Rep. 2014, 4, 1–10. [Google Scholar] [CrossRef] [PubMed]
  87. Ghosh, A.; Mustafiz, A.; Pareek, A.; Sopory, S.K.; Singla-Pareek, S.L. Glyoxalase III enhances salinity tolerance through reactive oxygen species scavenging and reduced glycation. Physiol. Plant. 2022, 174, e13693. [Google Scholar] [CrossRef] [PubMed]
  88. Islam, T.; Reddy, M. Evaluation of Cd2+ stress tolerance in transgenic rice overexpressing PgGPx gene that maintains cellular ion and reactive oxygen species homeostasis. PLoS ONE 2022, 17, e0273974. [Google Scholar] [CrossRef]
  89. Wu, Y.; Yang, Z.; How, J.; Xu, H.; Chen, L.; Li, K. Overexpression of a peroxidase gene (AtPrx64) of Arabidopsis thaliana in tobacco improves plant’s tolerance to aluminum stress. Plant Mol. Biol. 2017, 95, 157–168. [Google Scholar] [CrossRef] [PubMed]
  90. Kidwai, M.; Dhar, Y.V.; Gautam, N.; Tiwari, M.; Ahmad, I.Z.; Asif, M.H.; Chakrabarty, D. Oryza sativa class III peroxidase (OsPRX38) overexpression in Arabidopsis thaliana reduces arsenic accumulation due to apoplastic lignification. J. Hazard. Mater. 2019, 362, 383–393. [Google Scholar] [CrossRef]
  91. Chen, Z.; Pan, Y.; Wang, S.; Ding, Y.; Yang, W.; Zhu, C. Overexpression of a protein disulfide isomerase-like protein from Methanothermobacter thermoautotrophicum enhances mercury tolerance in transgenic rice. Plant Sci. 2012, 197, 10–20. [Google Scholar] [CrossRef]
  92. Ding, Y.; Qu, A.; Gong, S.; Huang, S.; Lv, B.; Zhu, C. Molecular identification and analysis of Cd-responsive microRNAs in rice. J. Agric. Food Chem. 2013, 61, 11668–11675. [Google Scholar] [CrossRef]
  93. Tang, L.; Mao, B.; Li, Y.; Lv, Q.; Zhang, L.; Chen, C.; He, H.; Wang, W.; Zeng, X.; Shao, Y. Knockout of OsNramp5 using the CRISPR/Cas9 system produces low Cd-accumulating indica rice without compromising yield. Sci. Rep. 2017, 7, 14438. [Google Scholar] [CrossRef]
  94. Nieves-Cordones, M.; Mohamed, S.; Tanoi, K.; Kobayashi, N.I.; Takagi, K.; Vernet, A.; Guiderdoni, E.; Périn, C.; Sentenac, H.; Véry, A.A. Production of low-Cs+ rice plants by inactivation of the K+ transporter OsHAK1 with the CRISPR-Cas system. Plant J. 2017, 92, 43–56. [Google Scholar] [CrossRef]
  95. Takahashi, R.; Ishimaru, Y.; Nakanishi, H.; Nishizawa, N.K. Role of the iron transporter OsNRAMP1 in cadmium uptake and accumulation in rice. Plant Signal Behav. 2011, 6, 1813–1816. [Google Scholar] [CrossRef]
  96. Huang, X.-Y.; Deng, F.; Yamaji, N.; Pinson, S.R.; Fujii-Kashino, M.; Danku, J.; Douglas, A.; Guerinot, M.L.; Salt, D.E.; Ma, J.F. A heavy metal P-type ATPase OsHMA4 prevents copper accumulation in rice grain. Nat. Commun. 2016, 7, 12138. [Google Scholar] [CrossRef] [PubMed]
  97. Tian-Yu, G.; Zi-Ai, Q.; Si-Ying, C.; Jing, Y.; Zi-Jun, F.; Jun-Min, W.; Ji-Ming, G. Dual-function DEFENSIN8 mediates phloem cadmium unloading and accumulation in rice grains. Plant Physiol. 2023, 191, 515–527. [Google Scholar] [CrossRef]
  98. Mehmood, S.S.; Lu, G.; Luo, D.; Hussain, M.A.; Raza, A.; Zafar, Z.; Zhang, X.; Cheng, Y.; Zou, X.; Lv, Y. Integrated analysis of transcriptomics and proteomics provides insights into the molecular regulation of cold response in Brassica napus. Environ. Exp. Bot. 2021, 187, 104480. [Google Scholar] [CrossRef]
  99. Cao, F.; Dai, H.; Hao, P.-F.; Wu, F. Silicon regulates the expression of vacuolar H+-pyrophosphatase 1 and decreases cadmium accumulation in rice (Oryza sativa L.). Chemosphere 2020, 240, 124907. [Google Scholar] [CrossRef] [PubMed]
  100. Cao, H.W.; Zhao, Y.N.; Liu, X.S.; Rono, J.K.; Yang, Z.M. A metal chaperone OsHIPP16 detoxifies cadmium by repressing its accumulation in rice crops. Environ. Pollut. 2022, 311, 120058. [Google Scholar] [CrossRef]
  101. Gostimskaya, I. CRISPR-Cas9: A History of Its Discovery and Ethical Considerations of Its Use in Genome Editing. Biochemistry 2022, 87, 777–788. [Google Scholar] [CrossRef]
  102. Raza, A.; Su, W.; Hussain, M.A.; Mehmood, S.S.; Zhang, X.; Cheng, Y.; Zou, X.; Lv, Y. Integrated analysis of metabolome and transcriptome reveals insights for cold tolerance in rapeseed (Brassica napus L.). Front. Plant Sci. 2021, 12, 1796. [Google Scholar] [CrossRef] [PubMed]
  103. Chang, J.D.; Huang, S.; Yamaji, N.; Zhang, W.; Ma, J.F.; Zhao, F.J. OsNRAMP1 transporter contributes to cadmium and manganese uptake in rice. Plant Cell Environ. 2020, 43, 2476–2491. [Google Scholar] [CrossRef]
  104. Songmei, L.; Jie, J.; Yang, L.; Jun, M.; Shouling, X.; Yuanyuan, T.; Youfa, L.; Qingyao, S.; Jianzhong, H. Characterization and evaluation of OsLCT1 and OsNramp5 mutants generated through CRISPR/Cas9-mediated mutagenesis for breeding low Cd rice. Rice Sci. 2019, 26, 88–97. [Google Scholar] [CrossRef]
  105. Tang, L.; Dong, J.; Qu, M.; Lv, Q.; Zhang, L.; Peng, C.; Hu, Y.; Li, Y.; Ji, Z.; Mao, B. Knockout of OsNRAMP5 enhances rice tolerance to cadmium toxicity in response to varying external cadmium concentrations via distinct mechanisms. Sci. Total Environ. 2022, 832, 155006. [Google Scholar] [CrossRef]
  106. Tang, L.; Dong, J.; Tan, L.; Ji, Z.; Li, Y.; Sun, Y.; Chen, C.; Lv, Q.; Mao, B.; Hu, Y. Overexpression of OsLCT2, a low-affinity cation transporter gene, reduces cadmium accumulation in shoots and grains of rice. Rice 2021, 14, 1–15. [Google Scholar] [CrossRef]
  107. Mishra, N.; Srivastava, A.P.; Esmaeili, N.; Hu, W.; Shen, G. Overexpression of the rice gene OsSIZ1 in Arabidopsis improves drought-, heat-, and salt-tolerance simultaneously. PLoS ONE 2018, 13, e0201716. [Google Scholar] [CrossRef] [PubMed]
  108. Liu, X.S.; Feng, S.J.; Zhang, B.Q.; Wang, M.Q.; Cao, H.W.; Rono, J.K.; Chen, X.; Yang, Z.M. OsZIP1 functions as a metal efflux transporter limiting excess zinc, copper and cadmium accumulation in rice. BMC Plant Biol. 2019, 19, 1–16. [Google Scholar] [CrossRef] [PubMed]
  109. Jeong, J.S.; Kim, Y.S.; Redillas, M.C.; Jang, G.; Jung, H.; Bang, S.W.; Choi, Y.D.; Ha, S.H.; Reuzeau, C.; Kim, J.K. OsNAC5 overexpression enlarges root diameter in rice plants leading to enhanced drought tolerance and increased grain yield in the field. Plant Biotechnol. J. 2013, 11, 101–114. [Google Scholar] [CrossRef] [PubMed]
  110. Kumagai, S.; Suzuki, T.; Tezuka, K.; Satoh-Nagasawa, N.; Takahashi, H.; Sakurai, K.; Watanabe, A.; Fujimura, T.; Akagi, H. Functional analysis of the C-terminal region of the vacuolar cadmium-transporting rice OsHMA3. FEBS Lett. 2014, 588, 789–794. [Google Scholar] [CrossRef]
  111. Shen, J.; Lv, B.; Luo, L.; He, J.; Mao, C.; Xi, D.; Ming, F. The NAC-type transcription factor OsNAC2 regulates ABA-dependent genes and abiotic stress tolerance in rice. Sci. Rep. 2017, 7, 40641. [Google Scholar] [CrossRef]
  112. Qi, D.-L.; Lee, J.-R.; Yang, C.-G.; Lee, M.-C.; Cao, G.-L.; Zhang, J.-G.; Zhou, Q.-Y.; Suh, S.-C.; Zhang, S.-Y.; Han, L.-Z. Detection of QTL for alkali tolerance at the germination stage in japonica rice. Chin. J. OF Rice Sci. 2009, 23, 589. [Google Scholar]
  113. Chen, J.; Zou, W.; Meng, L.; Fan, X.; Xu, G.; Ye, G. Advances in the uptake and transport mechanisms and QTLs mapping of cadmium in rice. Int. J. Mol. Sci. 2019, 20, 3417. [Google Scholar] [CrossRef] [PubMed]
  114. Wu, F.; Luo, X.; Wang, L.; Wei, Y.; Li, J.; Xie, H.; Zhang, J.; Xie, G. Genome-wide association study reveals the QTLs for seed storability in world rice core collections. Plants 2021, 10, 812. [Google Scholar] [CrossRef]
  115. Yamazaki, S.; Ueda, Y.; Mukai, A.; Ochiai, K.; Matoh, T. Rice phytochelatin synthases OsPCS1 and OsPCS2 make different contributions to cadmium and arsenic tolerance. Plant Direct 2018, 2, e00034. [Google Scholar] [CrossRef]
  116. Lu, C.; Zhang, L.; Tang, Z.; Huang, X.-Y.; Ma, J.F.; Zhao, F.-J. Producing cadmium-free Indica rice by overexpressing OsHMA3. Environ. Int. 2019, 126, 619–626. [Google Scholar] [CrossRef]
  117. Arya, G.C.; Sarkar, S.; Manasherova, E.; Aharoni, A.; Cohen, H. The Plant Cuticle: An Ancient Guardian Barrier Set Against Long-Standing Rivals. Front. Plant Sci. 2021, 12, 663165. [Google Scholar] [CrossRef] [PubMed]
  118. Rono, J.K.; Le Wang, L.; Wu, X.C.; Cao, H.W.; Zhao, Y.N.; Khan, I.U.; Yang, Z.M. Identification of a new function of metallothionein-like gene OsMT1e for cadmium detoxification and potential phytoremediation. Chemosphere 2021, 265, 129136. [Google Scholar] [CrossRef]
  119. Zhao, D.-D.; Park, J.-R.; Jang, Y.-H.; Kim, E.-G.; Du, X.-X.; Farooq, M.; Yun, B.-J.; Kim, K.-M. Identification of one major QTL and a novel gene OsIAA17q5 associated with tiller number in rice using QTl analysis. Plants 2022, 11, 538. [Google Scholar] [CrossRef]
  120. Ogo, Y.; Itai, R.N.; Kobayashi, T.; Aung, M.S.; Nakanishi, H.; Nishizawa, N.K. OsIRO2 is responsible for iron utilization in rice and improves growth and yield in calcareous soil. Plant Mol. Biol. 2011, 75, 593–605. [Google Scholar] [CrossRef] [PubMed]
  121. Yamaji, N.; Huang, C.F.; Nagao, S.; Yano, M.; Sato, Y.; Nagamura, Y.; Ma, J.F. A zinc finger transcription factor ART1 regulates multiple genes implicated in aluminum tolerance in rice. Plant Cell 2009, 21, 3339–3349. [Google Scholar] [CrossRef]
  122. Sasaki, A.; Yamaji, N.; Ma, J.F. Overexpression of OsHMA3 enhances Cd tolerance and expression of Zn transporter genes in rice. J. Exp. Bot. 2014, 65, 6013–6021. [Google Scholar] [CrossRef] [PubMed]
  123. Tao, J.; Lu, L. Advances in genes-encoding transporters for cadmium uptake, translocation, and accumulation in plants. Toxics 2022, 10, 411. [Google Scholar] [CrossRef]
  124. Li, J.-C.; Guo, J.-B.; Xu, W.-Z.; Ma, M. RNA Interference-mediated Silencing of Phytochelatin Synthase Gene Reduce Cadmium Accumulation in Rice Seeds. J. Integr. Plant Biol. 2007, 49, 1032–1037. [Google Scholar] [CrossRef]
  125. Zhao, J.; Yang, W.; Zhang, S.; Yang, T.; Liu, Q.; Dong, J.; Fu, H.; Mao, X.; Liu, B. Genome-wide association study and candidate gene analysis of rice cadmium accumulation in grain in a diverse rice collection. Rice 2018, 11, 1–15. [Google Scholar] [CrossRef]
  126. Vinocur, B.; Altman, A. Recent advances in engineering plant tolerance to abiotic stress: Achievements and limitations. Curr. Opin. Biotechnol. 2005, 16, 123–132. [Google Scholar] [CrossRef] [PubMed]
  127. Singh, S.; Parihar, P.; Singh, R.; Singh, V.P.; Prasad, S.M. Heavy Metal Tolerance in Plants: Role of Transcriptomics, Proteomics, Metabolomics, and Ionomics. Front Plant Sci. 2016, 6, 1143. [Google Scholar] [CrossRef] [PubMed]
  128. Shephard, A.M.; Zambre, A.M.; Snell-Rood, E.C. Evaluating costs of heavy metal tolerance in a widely distributed, invasive butterfly. Evol. Appl. 2021, 14, 1390–1402. [Google Scholar] [CrossRef] [PubMed]
  129. Lam, P.K.S. Interpopulation differences in acute response of Brotia hainanensis (Gastropoda, Prosobranchia) to cadmium: Genetic or environmental variance? Environ. Pollut. 1996, 94, 1–7. [Google Scholar] [CrossRef]
  130. Sahrawat, K.Á. Iron toxicity in wetland rice and the role of other nutrients. J. Plant Nutr. 2005, 27, 1471–1504. [Google Scholar] [CrossRef]
Figure 1. Schematic representation of the various negative effects of excess heavy metals on different parts of rice plants.
Figure 1. Schematic representation of the various negative effects of excess heavy metals on different parts of rice plants.
Agriculture 13 00944 g001
Figure 2. Anthropogenic and natural sources of heavy metal pollution in the environment.
Figure 2. Anthropogenic and natural sources of heavy metal pollution in the environment.
Agriculture 13 00944 g002
Table 2. List of targeted genes modified for HM tolerance in rice using CRISPR-Cas9 genome editing technology.
Table 2. List of targeted genes modified for HM tolerance in rice using CRISPR-Cas9 genome editing technology.
MetalloidTargeted Gene(s)Molecular FunctionsRole in HM Toxicity ToleranceReferences
As, SiOsLsiSilicon (Si) transporterLow arsenic uptake[33]
CdOsABCG36G-type ATP-binding cassette (ABC) transporterCadmium sequestration and toxicity tolerance[55]
CsOsHAK1High-Affinity Potassium (K+) TransporterLow cesium accumulation in root and shoots[94]
As, CdOsNRAMP1Cadmium, Iron, and manganese uptake/transporterLow arsenic and cadmium content in grains[103]
Cd, Pb, Mn, FeOsNRAMP5Major transporter for metal uptakeLow cadmium content in grains[104]
CdOsLCT1Low-Affinity Cation TransporterLow cadmium uptake[104]
CdOsLCT2Low-Affinity Cation TransporterLow cadmium accumulation in grains[105]
Table 3. Identified genes/QTLs associated with heavy metal tolerance in rice.
Table 3. Identified genes/QTLs associated with heavy metal tolerance in rice.
SRGenes/QTLMetalloidsMechanism InvolvedRef
1OsNRAMP5Fe and Mn, as well as Cd and ZnOverexpression of OsNRAMP5 to increase tolerance to Cd toxicity[93]
2OsHMA3CdOverexpression of OsHMA3 to increase tolerance to Cd toxicity[116]
3OsABCG31Cd and PbOverexpression of OsABCG31 enhanced their tolerance to Cd and Pd toxicity[117]
4OsLCT1AlOverexpression of OsLCT1 to increase tolerance to Al toxicity[106]
5OsSIZCdOverexpression of OsSIZ1 enhanced tolerance to Cd toxicity.[107]
6OsZIP1ZnOverexpression of OsZIP1 can enhance tolerance to Zn toxicity[108]
7OsNAC5Cd and PbOverexpression of OsNAC5 enhanced their tolerance to Cd and Pb toxicity[109]
8OsMT1eCd and ZnOsMT1e encodes a metallothionein protein involved in metal detoxification[118]
9OsNAC2CdOsNAC2 is a transcription factor that is involved in regulating the expression of genes involved in stress responses in rice[111]
10qNRG2-1NiqNRG2-1, a QTL on chromosome 2 is associated with Ni tolerance in rice associated with the expression of the OsNramp5 gene[119]
11qCdt1CdqCdt1-2, a QTL on chromosome 1 is associated with Cd tolerance with overexpression of OsHMA3[110]
12RM219Asaccumulation of As[112]
13qPC1CdqPC1 QTL was associated with lower Cd accumulation in rice grains[113]
14OsIRO2CdOsIRO2 is a transcription factor regulating the expression of genes involved in Fe homeostasis in rice[120]
15qGCD7CdThis QTL has been found to be important for rice tolerance to Cd toxicity[121]
16qHLR1Cd and PbThis QTL has been found to be important for rice tolerance to both Cd and Pb toxicity[122]
17qCST11PbThis QTL is associated with Cd and Pb tolerance in rice identified through a GWAS[80]
18OsIRT1FeThis gene encodes an Fe transporter involved in Cd uptake in rice. It was found to be upregulated under Cd stress[123]
19qMRS6.1MnThis QTL is associated with manganese (Mn) tolerance in rice. It was identified through a GWAS[114]
20OsPCS1CdThis gene encodes phytochelatin synthase involved in the synthesis of phytochelatins, a group of peptides that bind to HMs and detoxify them in plants. It was found to be upregulated under Cd stress in rice.[115]
21qHTSF4.1AsThe QTL is associated with Cd and As tolerance in rice[116]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Haider, Z.; Ahmad, I.; Zia, S.; Gan, Y. Recent Developments in Rice Molecular Breeding for Tolerance to Heavy Metal Toxicity. Agriculture 2023, 13, 944. https://doi.org/10.3390/agriculture13050944

AMA Style

Haider Z, Ahmad I, Zia S, Gan Y. Recent Developments in Rice Molecular Breeding for Tolerance to Heavy Metal Toxicity. Agriculture. 2023; 13(5):944. https://doi.org/10.3390/agriculture13050944

Chicago/Turabian Style

Haider, Zulqarnain, Irshan Ahmad, Samta Zia, and Yinbo Gan. 2023. "Recent Developments in Rice Molecular Breeding for Tolerance to Heavy Metal Toxicity" Agriculture 13, no. 5: 944. https://doi.org/10.3390/agriculture13050944

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

Article Metrics

Back to TopTop