Next Article in Journal
Hybrid Nanogel Drug Delivery Systems: Transforming the Tumor Microenvironment through Tumor Tissue Editing
Previous Article in Journal
Inhibition of PDIs Downregulates Core LINC Complex Proteins, Promoting the Invasiveness of MDA-MB-231 Breast Cancer Cells in Confined Spaces In Vitro
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Uptake, Transfer, and Detoxification of Cadmium in Plants and Its Exogenous Effects

1
State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangdong Key Laboratory for Innovative Development and Utilization of Forest Plant Germplasm, College of Forestry and Landscape Architecture, South China Agricultural University, Guangzhou 510642, China
2
Root Biology Center, South China Agricultural University, Guangzhou 510642, China
3
College of Natural Resources and Environment, South China Agricultural University, Guangzhou 510642, China
4
Key Laboratory of Agricultural and Rural Pollution Control and Environmental Safety in Guangdong Province, Guangzhou 510642, China
*
Authors to whom correspondence should be addressed.
Cells 2024, 13(11), 907; https://doi.org/10.3390/cells13110907
Submission received: 21 April 2024 / Revised: 13 May 2024 / Accepted: 13 May 2024 / Published: 24 May 2024
(This article belongs to the Topic Metalloproteins and Metalloenzymes)

Abstract

:
Cadmium (Cd) exerts a toxic influence on numerous crucial growth and development processes in plants, notably affecting seed germination rate, transpiration rate, chlorophyll content, and biomass. While considerable advances in Cd uptake and detoxification of plants have been made, the mechanisms by which plants adapt to and tolerate Cd toxicity remain elusive. This review focuses on the relationship between Cd and plants and the prospects for phytoremediation of Cd pollution. We highlight the following issues: (1) the present state of Cd pollution and its associated hazards, encompassing the sources and distribution of Cd and the risks posed to human health; (2) the mechanisms underlying the uptake and transport of Cd, including the physiological processes associated with the uptake, translocation, and detoxification of Cd, as well as the pertinent gene families implicated in these processes; (3) the detrimental effects of Cd on plants and the mechanisms of detoxification, such as the activation of resistance genes, root chelation, vacuolar compartmentalization, the activation of antioxidant systems and the generation of non-enzymatic antioxidants; (4) the practical application of phytoremediation and the impact of incorporating exogenous substances on the Cd tolerance of plants.

1. Introduction

It is well known that the rapid evolution of modernized industrial and agricultural practices has led to various environmental challenges. One of the prominent problems is cadmium (Cd) pollution. Between 1950 and 1990, global cadmium production doubled, reaching approximately 20,000 tons annually. Anthropogenic sources deposited an estimated 900 to 3600 tons of Cd into aquatic environments [1], resulting in severe Cd pollution. This pollution poses a significant threat to both human health and environmental safety.
Cd pollution is a global concern, evident in various regions. In the suburbs of Dera Ismail Khan, Pakistan, vegetables cultivated using wastewater irrigation exhibited significantly higher levels of Cd accumulation compared to those grown with freshwater irrigation [2]. In southern China, tobacco–rice rotation causes soil pH to decrease, thereby enhancing the flow of cadmium to crops [3]. Notably, the transfer of cadmium from the soil to the human body through crops such as vegetables and rice can lead to a variety of health problems, such as central nervous system depression and kidney and liver damage [4]. In the last century, many people in Japan suffered immensely from Itai-itai disease due to cadmium contamination of farmland and water sources [4].
To cope with these formidable challenges, numerous scientists have dedicated their efforts to environmental science research, trying to explore strategies for mitigating Cd pollution through a variety of pathways, including physical, chemical, and phytoremediation. Based on previous studies, this review will focus on the mechanisms of Cd transport by plants, the forms of Cd toxicity suffered/coped by plants, and the effects of exogenous modes on phytoremediation.

2. The Pollution Status and Hazards of Cadmium

Cd is a rare dispersed element commonly found in soils and zinc (Zn) minerals in the form of divalent cations (Cd2+) and soluble complexes with a variety of toxic effects. Cd has an extremely long biological half-life, predominantly accumulating in the liver and kidneys of the human body where it is difficult to eliminate [5].

2.1. Sources and Distribution of Cadmium

Cd levels vary in different countries around the world due to geographic location, latitude and longitude, and environmental climate. Cd levels in soils are currently higher than the original environmental background values in all countries as a result of atmospheric deposition and overuse of phosphate fertilizers. For example, the average concentration of Cd in European soils is 0.33 mg kg−1 [6], while in agricultural land in China, it is 0.19 mg kg−1, with an environmental background value of 0.097 mg kg−1 [7]. In the United States agricultural soils, the average Cd concentration is 0.265 mg kg−1 [8].
Soil Cd comes from a wide range of sources and can be categorized into two main sources, including natural and anthropogenic sources. The presence of Cd in natural soils mostly originates from rock weathering and suspended soil particles transported through the air. Soil particle sources encompass various natural occurrences such as forest fires, volcanic emissions, and atmospheric dust [9]. In contrast, anthropogenic Cd emissions predominantly originate from activities like phosphate fertilizer application, tailings disposal, metal industry practices, mining operations, and fossil fuel combustion [10,11]. Since the Industrial Revolution, diverse industrial, mining, and agricultural activities worldwide have led to substantial heavy metal diffusion into soils and water bodies (Figure 1). By the beginning of the 21st century, global anthropogenic Cd production during the industrial era had accumulated to 1.1 million tons, with the global per capita burden estimated at 0.18 kg [12].

2.2. Hazards of Cadmium on the Human Body

With the escalation of mining activities and metal smelting, there has been a corresponding increase in Cd levels found in soil surfaces, air, and water sources. This raises a significant hazard to the health of animals, plants, and human beings. Cd in animals and humans mainly comes from drinking water, eating, and respiration, and a very small part is absorbed through the skin and hair. When Cd enters the body through the gastrointestinal or respiratory tract, it is transported into the bloodstream through erythrocytes and albumin, and then accumulates in the kidneys and liver [5]. Of note, the biological half-life of Cd in the kidney is 45 years [13].
To minimize the potential harm caused by exposure to or inhalation of Cd, individuals should consume foods that are rich in polyphenols [14], such as mint and strawberries. These foods possess antioxidant properties and can aid in chelating Cd2+. Additionally, incorporating more seafood, legume products, melon seeds, and other foods with high Zn content into one’s diet can help counteract the excessive accumulation of Cd in the body. Collectively, these dietary measures operate through distinct mechanisms to support overall physical well-being.
Figure 1. Sources of Cd and its transmission pathways in the environment. This figure shows the process of Cd deposition, accumulation, cycling in the environment, and enrichment in plants, animals, and humans through the food chain. Cd exists in different chemical forms in each pathway, such as in water bodies, where it is mainly in the form of chloride [15], in coal combustion and dust formed in non-ferrous metal production, where it mainly contains CdS and CdSO4 [16], and in phosphorus fertilizers in the form of Cd(H2PO4)2 and CdHPO4 [17].
Figure 1. Sources of Cd and its transmission pathways in the environment. This figure shows the process of Cd deposition, accumulation, cycling in the environment, and enrichment in plants, animals, and humans through the food chain. Cd exists in different chemical forms in each pathway, such as in water bodies, where it is mainly in the form of chloride [15], in coal combustion and dust formed in non-ferrous metal production, where it mainly contains CdS and CdSO4 [16], and in phosphorus fertilizers in the form of Cd(H2PO4)2 and CdHPO4 [17].
Cells 13 00907 g001

3. Mechanisms of Cadmium Uptake and Transport in Plants

3.1. Forms and Bioavailability of Cadmium in Soils

The chemical forms of Cd in different soils vary depending on conditions such as soil redox potential, moisture, texture, and inter-root environment. According to the five-step sequential extraction method proposed by Tessier et al. (1979), Cd in soils can be grouped into five forms: soluble and exchangeable state (Cd2+), carbonate-bound state (CdCO3), ferromanganese-oxidized state, organic-bound state, and residue [18], with soluble and exchangeable states dominating.
Not all Cd in soils can be absorbed by plants. Plants mainly take up the exchangeable and carbonate-bound states of Cd. However, the bioavailability of Cd can also be influenced by the soil’s properties. Among them, soil acidity and alkalinity will significantly affect the Cd solubility and morphological distribution in soils, and increasing soil pH is negatively correlated with the effectiveness of heavy metals in plants [19,20]. The main reason for the increased Cd pollution in recent years is soil acidification. In addition, soil properties are also a key factor influencing Cd morphology and utilization, and plants in sandy soils are more capable of Cd uptake compared with clay soils [21]. It is worth mentioning that the chelating agents such as ethylenediaminetetraacetic acid (EDTA) and nitrilotriacetic acid (NTA) can increase the effectiveness of Cd in soils, facilitate its movement from roots to aboveground tissues, and increase Cd accumulation in leaves [22]. In addition to Cd bioavailability, Cd uptake and accumulation in plants are also influenced by other factors, such as plant species, genotype, inter-root environment, and mineral nutrients such as silicon (Si), selenium (Se), and iron (Fe) [23,24,25,26].

3.2. Mechanism of Cadmium Uptake by Plants

Cd uptake and accumulation in plants mainly consist of the following physiological processes: uptake of Cd from the outside (soil or air), lateral and radial transport of Cd, and phloem-mediated aboveground redistribution (Figure 2).

3.2.1. Pathways of Cadmium Transport from Soil to Root Epidermis

The root is the first organ exposed to Cd in soils and the first barrier for plants to resist Cd toxicity. Plants absorb Cd from the soil through root hairs and epidermal cells in the mature zone of the root tip.
The entry of Cd into roots mainly includes the following pathways:
  • Cd2+ is exchanged with H+ produced during plant respiration and is thus adsorbed on the surface of root epidermal cells and then enters the cortex via the apoplast pathway [27].
  • As Cd is a non-essential metallic element lacking a specific transporter in plants, its entry into plant tissues typically occurs through the symplast pathway. This process involves competing for binding sites on metal transporter proteins, including IRT1 (a bivalent iron transporter protein) and NRAMP5 (a manganese transporter protein) [28]. Additionally, Cd can also enter plants through ion channels of divalent metals such as calcium (Ca).
  • To enhance ion utilization within the inter-root soil, certain low molecular compounds, such as erucic acid and oxalic acid, are secreted by plant roots. These compounds form metal–ligand complexes with Cd2+, thus facilitating Cd to enter the root epidermis as a chelate transported by yellow-stripe-Like (YSL) proteins [29].
Although Cd2+ binds to both organic ligands (such as dissolved organic substances like low-molecular-weight organic acids secreted by plant roots) and inorganic ligands (e.g., NO3−, Cl) to form soluble complexes, subsequently enhancing the mobility of Cd to the root surface, it is widely accepted that Cd2+ is the primary form of translocation through the plasma membrane into the root tip cells [30].

3.2.2. Lateral Transport of Cadmium from Root Epidermis to Xylem

Cd is synergistically transported through both the symplast and the apoplast pathways after entering the epidermal cells of the root hair zone (Figure 2).
In the symplast pathway, Cd is mainly transported by intracellular protoplasmic flow and plasmodesmata channels between cells, first across the endodermis into the stele, then through the pericycle sheaths into the parenchyma cell, and ultimately into the xylem conductance ducts [31]. In the apoplast pathway, Cd enters the xylem along the apoplast space such as the cell wall, cell interstitial space, intercellular layer, and conduit cavity. This pathway is impeded by casparian strips through the endodermis. Additionally, a fraction of the Cd2+ is sequestered into vacuoles through transporter proteins like OsHMA3 in rice [32], redirected to the symplast pathway for Cd transportation to the xylem of the stele [33].
Cd transport from the epidermis to the xylem completes within the root, and the entire mechanism is mediated by transporter proteins in the plasma membrane, which directs the ions to specific locations [34].

3.2.3. Radial Transport of Cadmium through the Xylem to Aboveground Parts

Upon Cd entering into roots, the xylem pathway subsequently plays a dominant role in the long-distance transport of Cd from roots to aboveground (Figure 2), with the main driving forces being transpiration pull and root pressure [35].
It is known that Cd accumulates mainly in the roots of plants and is partially translocated from roots to aboveground parts. The efficiency of translocation depends on root vacuole sequestration and xylem loading capacity [30]. The more vacuole sequestration in the root, the less Cd2+ is transferred to the aboveground part. As documented, the transporter proteins CAX2, CAX4, and HMA3 play key roles in chelating Cd to the vacuole [32,36,37]. In addition, the lignified casparian strips in the endodermis of plant roots serve as a barrier, impeding the entry of Cd into the shoots. This may account for the higher accumulation of Cd in the roots in comparison to the shoots [38].

3.2.4. Phloem Mediates Cadmium Redistribution

The phloem is mainly responsible for the redistribution of Cd in the aboveground part based on studies in Arabidopsis thaliana [39] and rice [40].
Cd absorbed by plant roots is mainly translocated to the aboveground part through the xylem and then to the rice grain through the phloem [41], suggesting that most of the Cd needs to be translocated from the xylem to the phloem before it can be re-translocated to the grain. In recent years, new insights into the process of Cd translocation from roots to stems have been gained, suggesting that past studies have overlooked the value of long-distance transport in the phloem [42]. It was found that the phloem is an important pathway for Cd long-distance transport from root to leaf in eggplant and oilseed rape [35,43], and that Cd-GS2 complexes contribute to Cd long-distance movement in the phloem [42].

3.3. Cadmium-Related Transporter Proteins in Plants

Cd is one of the non-essential metal ions; the uptake of Cd by plants from soils must be mediated by transporter proteins for essential cations [44,45]. As shown in Figure 3 and Table 1, the following protein families are commonly involved in Cd transport or detoxification.

3.3.1. The Natural Resistance-Associated Macrophage Protein Family

The natural resistance-associated macrophage protein (NRAMP) family has been identified in model plants such as Arabidopsis thaliana and rice. As revealed, NRAMP plays a key role in metal homeostasis.
A total of seven NRAMP family members in rice have been functionally characterized. NRAMP1, NRAMP2, and NRAMP5 are involved in Cd transport [46]. OsNRAMP2 is localized to the tonoplast and is primarily expressed in seeds, roots, and leaves [47]. Knockout of OsNRAMP2 resulted in a reduction in Cd translocation from vegetative tissues to rice grains; conversely, overexpression lines of OsNRAMP2 exhibited a significant increase in grain Cd concentrations [48,49]. OsNRAMP5 is localized in the plasma membrane and expressed in rice roots [50]. OsNRAMP5 serves as the primary transporter protein for manganese (Mn) and Cd, facilitating their translocation from soil solution to root cells. The knockout lines of OsNRAMP5 have a substantial reduction in Cd in rice, and overexpressing OsNRAMP5 increases Cd uptake in roots, but Cd levels in shoots remain at low levels [51,52]. OsNRAMP1 is localized in root and leaf cells and is a close homolog to OsNRAMP5 (72.78% amino acid sequence similarity) with similar but non-redundant functions [53,54,55]. Knockout of OsNRAMP1 or OsNRAMP5 results in reduced levels of Cd in rice. Moreover, the loss of function of OsNRAMP5 has a greater impact than OsNRAMP1. Further studies have revealed that double knockout mutants of these two genes have a significant reduction in Cd [53]. This indicates that gene editing of OsNRAMP1 or/and OsNRAMP5 can help in the breeding of low-Cd varieties of rice, especially by altering their expression levels via editing the promoter sequence in the future. It must be pointed out that the uptake of Mn is also impaired by the knockout of these two genes. As Mn is one of the essential elements for plant growth and development, how to balance the relationship between the two elements is an important goal for future investigations.

3.3.2. The Zinc/Iron-Regulated Transporter-like Protein Family

Lots of studies have demonstrated that the zinc–iron-regulated transport proteins (ZIPs) family plays an important role in metal uptake in roots and distribution in plants.
To date, 15 ZIP family members have been reported in Arabidopsis thaliana [56]. AtIRT1, the first identified ZIP family member in Arabidopsis thaliana, is mainly expressed in roots and plays a key role in the uptake of divalent iron from the soil [57,58]. Moreover, AtIRT1 is involved in the transport of divalent cations such as Zn, Mn, cobalt (Co), and Cd [58,59,60,61]. Knockout of AtIRT1 results in lower accumulation of the heavy metal Cd [60]. Although AtIRT2 shares phylogenetic similarity with AtIRT1, it is not directly responsible for Fe uptake from soils. Instead, AtIRT2 collaborates with AtIRT1 to maintain Fe homeostasis in plants [62,63]. Overexpression of AtIRT2 in Arabidopsis thaliana enhances the uptake of metals such as Fe, Cd, and Zn. However, the sensitivity of yeast cells to Cd remains unaffected when overexpressing AtIRT2 [62], possibly due to the indirect synergistic interaction between AtIRT2 and AtIRT1 in response to Cd stress. AtZIP1 is a vacuole transporter that transfers Mn [64]. AtZIP2 participates in the uptake of Mn and Zn [64]. Interestingly, AtZIP1, AtZIP3, and AtZIP4 respond to Zn deficiency [65]. In addition, AtZIP2 and AtZIP4 are also involved in copper (Cu) transport [66]. In short, ZIP family members have versatile roles in Mn, Zn, and Cd transportation.
There are 16 ZIP family members in rice [67]. OsIRT1 and OsIRT2 are mainly responsible for the uptake of Zn and Fe in the rice root system [68] and also play a role in Cd uptake because of the similar physicochemical properties of Zn2+, Fe2+, and Cd2+ [69,70]. OsZIP1, localized in the endoplasmic reticulum and plasma membrane, is a metal detoxification transporter. Overexpressing OsZIP1 can reduce the overaccumulation of Zn, Cu, and Cd in rice and promote rice growth [71,72]. OsZIP3 is preferred for Zn uptake over other divalent cations such as Cd. OsZIP3 co-regulates Cd transport and uptake together with OsHMA2 and OsLCT1 [73,74]. Both OsZIP5 and OsZIP9 are redundantly involved in Zn and Cd uptake. OsZIP9 is responsible for the broad regulation of Zn in roots and shoots, and OsZIP5 fine-tunes Zn uptake to maintain Zn homeostasis. Accordingly, rice with a single or both genes knocked out exhibits reduced uptake of Zn and Cd, whereas overexpression of OsZIP5 or OsZIP9 has the opposite effect [75]. OsZIP6 demonstrates transport activity for Fe2+, Cd2+, and Co2+, exhibiting the highest efficiency under acidic environmental conditions [76]. OsZIP7 is expressed in parenchyma cells of vascular bundles in rice roots and nodes and is involved in the transport of Zn and Cd. In line with this, the Cd levels in the roots and internode of knockout lines of OsZIP7 are higher [77].
Different species have different expression patterns in Cd stress, Arabidopsis thaliana up-regulates ZIP family genes in the roots, and rice mainly up-regulates aboveground ZIP family genes. It has been verified that AtIRT1, OsZIP1, and OsZIP3 play more important roles in Cd uptake [78].

3.3.3. The Heavy Metal ATPases

Known as P1B-ATPase, heavy metal ATPase (HMAs) plays an important role in the translocation or detoxification of heavy metals in plants [79], especially in hyperaccumulators. These transporters are reported to exhibit variations in various aspects, including subcellular localization and metal specificity. When plants are exposed to low concentrations of Cd stress, only a limited number of HMA genes are up-regulated. The majority of transporters are mobilized only in response to elevated concentrations [80].
The rice genome encodes nine heavy metal ATPases. OsHMA1 and OsHMA4 have higher expression levels when stressed with Cd. Generally, OsHMA1 and OsHMA4 play roles in maintaining homeostasis in plants under heavy metal stress [80], but their specific biological functions remain to be explored. OsHMA2 is localized in the plasma membrane of the root stele. OsHMA2 is mainly involved in mediating the xylem loading of Cd and Zn, and the translocation to the shoots [69,81]. OsHMA3 is expressed in the vesicular membrane of root cells and is responsible for chelating extra-membranous Cd and Zn to the vacuole to prevent their translocation to the aboveground organs [32,45]. OsHMA9 is expressed in root and mesophyll tissue and appears to be responsible for Cd, Cu, Zn, and Pb efflux [82].
Arabidopsis thaliana genome encodes eight HMAs. AtHMA1 is localized in the chloroplast periplasm and has been found to transport not only Cu and Zn [83,84], but also Cd and Ca after heterologous expression in yeast [85]. AtHMA2 and AtHMA4 are localized in the plasma membrane and mediate the translocation of Cd from roots to shoots [86,87,88,89]. AtHMA3 showed similar properties to OsHMA3 and mediated the vacuole sequestration of Cd in roots. Consistently, overexpression of AtHMA3 results in enhanced tolerance of Arabidopsis thaliana to Zn, Co, Cd, and Pb [90].
In summary, the heavy metal ATPase family can be divided into two groups based on the properties of the metal substrates; the first group is the Zn/Co/Cd/Pb subgroup as exemplified by rice OsHMA1–OsHMA3 and Arabidopsis thaliana AtHMA1–AtHMA4, and the second is the Cu/Ag subgroup, containing rice OsHMA4–OsHMA9, Arabidopsis thaliana AtHMA5–AtHMA8 [91]. As the second group of HMA is not related to Cd, we do not introduce them here.

3.3.4. Others

In addition to the above family of transporter proteins, there are many other transporters also involved in the uptake, transport, and efflux of Cd (Table S1).
In rice, the common ones are Cd Accumulation in Leaves 1 (OsCAL1), Plant Cd Resistance 1 (OsPCR1), Low Cd (OsLCD), Low-affinity Cation Transporter genes 1 (OSLCT1), Cd transporter genes 1 (OsCd1) and Hypersensitive Induced Reaction Protein 1 (OsHIR1). OsCAL1 is responsible for chelating Cd and exports it from the cytoplasm to the outside of the cell, thus reducing Cd concentration in the cells [92,93]. OsPCR1 is involved in the transport of rice from roots to aerial parts [94]. OsLCD is mainly expressed in roots and leaves, and its absence reduces the accumulation of Cd in plants [95]. OsLCT1 is localized at the plasma membrane and exhibits Cd efflux activity in yeast [96,97]. As reported, knockout lines of OsLCT1 have lower Cd levels in phloem and grains [96,97]. OsCd1 belongs to the Major Facilitator Superfamily (MFS) family of transporter proteins. OsCd1 is localized in the plasma membrane of roots. OsCd1 is associated with Cd uptake in roots and contributes to Cd accumulation in rice grains [93,98]. Heterogeneous overexpressing OsHIR1 significantly reduces Cd and arsenic (As) accumulation, thus increasing plant tolerance to Cd and As [99].
Table 1. Gene family related to Cd uptake, transport and efflux.
Table 1. Gene family related to Cd uptake, transport and efflux.
Gene FamilyPlantGeneExpression SiteFunctionReference
The natural resistance-associated macrophage protein family
(NRAMP)
Oryza sativa L.OsNRAMP1Root cells and leaf mesophyll cellsCd uptake and transport[53,54,55]
OsNRAMP2Seeds, roots, leaf sheaths and leaf bladesCd efflux, translocation and distribution[47,48,49]
OsNRAMP5RootsCd uptake[28,50,51]
Arabidopsis thalianaAtNRAMP1Roots and aerial partsCd entry and transport[100,101]
AtNRAMP3Roots and aerial partsCd transport[100,102,103]
AtNRAMP4Roots and aerial partsCd transport[103,104]
AtNRAMP6Seeds and shootsCd transport and distribution[105]
Nicotiana tabacum L.NtNRAMP1RootsCd uptake and accumulation[106]
NtNRAMP3aLeavesCd transport, tolerance and accumulation[107]
NtNRAMP3bLeaves and rootsCd uptake, transport and maintain homeostasis[108]
NtNRAMP5RootsCd transport[109]
NtNRAMP6aRoots, stems, leaves and flowersCd transport[110]
NtNRAMP6bRoots, stems, leaves and flowersCd transport[110]
The natural resistance-associated macrophage protein family
(NRAMP)
Sedum alfredii HanceSaNRAMP1ShootsCd transport and accumulation[111]
SaNRAMP3vascular tissuesCd transport[112]
SaNRAMP6Leaves and rootsCd transport and accumulation[113]
Sedum plumbizincicolaSpNRAMP5-Cd transport[114]
Populus × canescensPcNRAMP1RootsCd uptake and transport[115]
Morus albaMaNRAMP1RootsCd transport[116]
Populus trichocarpaPtNRAMP1Leaves and rootsCd transport[117]
PtNRAMP2Leaves and rootsCd transport[117]
PtNRAMP4Leaves and rootsCd transport[117]
PtNRAMP9Leaves and rootsCd transport[117]
PtNRAMP10Leaves and rootsCd transport[117]
PtNRAMP11Leaves and rootsCd transport[117]
Malus hupehensisMhNRAMP1RootsCd uptake and accumulation[118]
Malus baccata (L.) BorkhMbNRAMP1RootsCd transport[119]
Noccaea caerulescens (Thlaspi caerulescens)NcNRAMP1RootsCd transport[120]
TcNRAMP3RootsCd accumulation and homeostasis[121,122]
TcNRAMP4RootsCd transport[122]
The natural resistance-associated macrophage protein family
(NRAMP)
Brassica rapa L. Chinensis.BcNRAMP1Whole plant bodyCd uptake and accumulation[123]
Brassica napusBnNRAMP1bSeedlings and vegetative tissueCd transport[124]
Triticum polonicum L.TpNRAMP5Roots and basal stemsCd transport[125]
Hordeum vulgareHvNRAMP5RootsCd uptake[126]
Spirodela polyrhizaSpNRAMP1Roots, fronds and joint between mother and daughter frondsCd uptake and accumulation[127,128]
Vigna radiataVrNRAMP5RootsCd uptake[129]
The zinc/iron-regulated transporter-like protein family
(ZIP)
Oryza sativa L.OsIRT1RootsCd uptake and transport[68,70,130]
OsIRT2RootsCd uptake[130,131]
OsZIP1RootsCd efflux[71,73]
OsZIP3NodesCd transport and distribution[73,74,132]
OsZIP5RootsCd uptake[75]
OsZIP6Shoots and rootsCd transport[76]
OsZIP7Roots and nodesCd transport[77]
OsZIP9Shoots and rootsCd uptake[75]
Arabidopsis thalianaAtIRT1RootsCd uptake and transport[58,59,60,133]
The zinc/iron-regulated transporter-like protein family
(ZIP)
Nicotiana tabacum L.NtIRT1RootsCd uptake and accumulation[134,135]
NtZIP1
(NtZIP5B)
RootsCd uptake[136,137]
NtZIP4BLeaves and rootsCd transport[137]
Arabidopsis halleriAhZIP6Leaves and rootsCd transport, tolerance[138]
Noccaea caerulescens (Thlaspi caerulescens)TcZNT1RootsCd uptake[139]
TcZNT5RootsCd transport[140]
TcZNT6Shoots and rootsCd transport[140]
Sedum alfredii HanceSaZIP4hShoots and rootsCd transport[141]
Morus albaMaIRT1LeavesCd transport[116]
MaZIP4RootsCd transport[116]
Thlaspi japonicumTjZNT1-Cd transport[142]
TjZNT2-Cd transport[142,143]
Brassica chinensis L.BcIRT1-Cd transport[144]
BcZIP2-Cd transport[144]
Avicennia marinaAmZIP1RootsCd transport[145]
AmIRT1Leaves, stems, and rootsCd transport[145]
Triticum polonicum L.TpIRT1A/BRoots, leaves, and reproductive organsCd uptake and transport[146]
The
heavy metal
ATPases
(The
P1B-type ATPases family)
Oryza sativa L.OsHMA9Roots and mesophyll tissuesCd efflux[82]
Arabidopsis thalianaAtHMA2Vascular tissues of roots, stems, and leavesCd transport and homeostasis[86,87]
Arabidopsis halleriAhHMA4Shoots and rootsCd transport[147]
Sedum alfredii HanceSaHMA3hShoots and rootsCd transport and sequestration within vacuoles[148]
SaHMA3nShoots and rootsCd transport and sequestration within vacuoles[148]
Sedum plumbizincicolaSpHMA6Leaves and rootsCd uptake, translocation and distribution[149]
Noccaea caerulescens (Thlaspi caerulescens)TcHMA4Shoots and rootsCd transport[150]
Morus albaMaHMA3RootsCd transport[116]
Capsicum sp. CaHMA1Pepper fruitsCd transport and accumulation[151]
Glycine Max (L.) Merr.GmHMA3wRootsCd transport and sequestration within endoplasmic reticulum[152]
Triticum aestivum L.TaHMA2-Cd translocation and transport[153]
Avicennia marinaAmHMA2Roots, leaves, stems, buds, and flowersCd transport[145]
The yellow-stripe-like transporter
(YSL)
Solanum nigrumSnYSL3Roots and stemsCd-NA compound transport[154]
Brassica junceaBjYSL7StemsCd transport and tolerance[155]
Zea mays L.ZmYS1-Cd-DMA compound transport[156]
Vaccinium ssp.VcYSL6-Cd-NA compound transport[157]
The ATP-binding cassette transporter family
(ABC)
Oryza sativa L.OsABCG36
(OsPDR9)
Roots and shootsefflux of Cd and Cd chelates[158]
OsABCG43
(OsPDR5)
Roots and shootsCd transport and tolerance[159]
OsPDR20
(OsABCG53)
Whole plant bodyefflux of Cd and Cd chelates[160]
Arabidopsis thalianaAtPDR8
(AtABCG36)
RootsCd efflux[161]
AtATM3
(AtABCB25)
RootsCd chelates transport[162,163]
Populus tomentosaPtoABCG36Leaves, stems and rootsCd efflux[164]
Sedum plumbizincicolaSpABCB28-Cd transport into organelles[114]
Rehmannia glutinosaRgABCC1RootsCd transport[165]
The placenta-specific 8-domain -containing family
(PLAC8)
Oryza sativa L.OsPCR1
(OsFWL5)
Grains, roots, stems and leavesCd accumulation and transport[166]
OsPCR3
(OsFWL2)
Grains, roots, stems and leavesCd accumulation and transport[166,167]
OsFWL3-Cd tolerance[168]
OsFWL4-Cd transport and translocation[168]
Populus euphraticaPePCR2RootsCd efflux[169]
PePCR10-Cd efflux[170]
Sedum alfredii HanceSaPCR2RootsCd uptake and accumulation[171]
Brassica napusBnPCR10.1Whole plant bodyCd transport[172]
Avicennia marinaAmPCR2Stems, pneumatophores and rootsCd efflux[145]
Salix linearistipularisSlPCR6RootsCd transport[173]
SlCNR8RootsCd uptake, efflux and accumulation[174]
Triticum aestivumTaCNR2Leaves and internodesCd transport and tolerance[175]
Triticum urartuTuCNR10Shoots and rootsCd transport[176]
Populus × canescensPcPLAC8-10RootsCd uptake[177]
The metal tolerance protein family
(MTPs)
Oryza sativa L.OsMTP1
(OZT1)
Roots, seeds and leavesCd transport[178,179,180]
Helianthus annuus L.HaMTP10-Cd efflux[181]
The defensin-like protein family
(DEFL)
Oryza sativa L.OsCAL1RootsCd chelation and transport[92,93]
Arabidopsis thalianaAtPDF2.5RootsCd chelation and efflux[182]
The cation/calcium superfamily (CaCA)Oryza sativa L.OsCAX1aRootsCd transport and tolerance[183]
OsCAX1cRoots and leavesCd transport and tolerance[183]
OsCAX4Roots and leavesCd transport and tolerance[183]
The cysteine-rich peptide family
(CYSTM)
Oryza sativa L.OsCCX2
(OsCDT1)
NodesCd efflux[184,185,186]
The low-affinity cation transporter family
(LCT)
Oryza sativa L.OsLCT1Leaves and nodesCd efflux and transport[97,187]
OsLCT2RootsCd transport[188]
The major facilitator superfamily
(MFS)
Oryza sativa L.OsCd1RootsCd uptake[98]
The Proliferation, Ion and Death superfamily (PID)Oryza sativa L.OsHIR1-Cd uptake and tolerance[99]
-Oryza sativa L.OsAAN4-Cd uptake[189]
-Oryza sativa L.OsGLR3.4-Cd uptake[189]
-Sorghum bicolor L.SbEXPA11-Cd uptake and transport[190]
“-” means unspecified.NA, nicotianamine; DMA,2’-deoxymugineic acid.

3.4. Cadmium-Related Transcription Factors in Plants

Apart from transporter proteins, transcription factors (TFs) also play crucial roles in mediating plants’ responses to Cd stress. It has been reported that TF families such as WRKY, MYB, and NAC play direct or indirect roles in regulating Cd tolerance in plants [191,192,193]. This regulation is achieved through the control of Cd-related genes, activation of specific signaling pathways, or interaction with other proteins.
The WRKY family is one of the largest families of TF in plants. A total of seven WRKYs (AtWRKY12, AtWRKY13, AtWRKY18, AtWRKY33, AtWRKY40, AtWRKY45, and AtWRKY60) related to Cd tolerance are currently reported in Arabidopsis thaliana [133,191,194,195,196,197]. In particular, AtWRKY45 facilitated the synthesis of phytochelatins by activating AtPCS1 and AtPCS2, thereby enhancing Cd tolerance in A. thaliana [191]. Moreover, there are many other families of transcription factors (Table S2) that play important roles in the alleviation of Cd stress. AtMYB49 regulates Cd accumulation through activation of the iron transport protein IRT1 and the abscisic acid (ABA) signaling pathway [192]. AtbHLH38/AtbHLH39 increased the expression of NAS1 and NAS2 and reduced Cd accumulation. Collectively, these TFs alter plant sensitivity and tolerance to Cd in a manner that regulates downstream genes or modulates signaling pathways.
TFs are widely involved in plant growth and development. However, the large number of TF and the complex regulatory networks have led to the fact that fewer Cd-related TFs have been identified. Deciphering the functions and regulatory networks of various types of Cd-related TFs is an important goal, which can provide the basis for solving Cd pollution at the molecular level.

4. Toxicity of Cadmium and Detoxification Mechanism of Cadmium in Plants

4.1. Toxicity of Cadmium to Plants

Excessive Cd usually negatively affects plant growth, development, and proliferative metabolism [198], such as plant biomass accumulation, germination rate, stomatal conductance and transpiration rate (Figure 4). Potential hazards include inhibition of photosynthetic pigment formation, reduction in photosynthetic efficiency, disruption of cellular homeostasis, chromosomal aberrations, damage to mitochondria, disruption of antioxidant mechanisms and metabolic pathways, and disruption of ATP synthesis [14,199]. Of note, the proton gradient generated by the electron transport chain and photochemical reactions is the main source of ATP synthase [200]. Cd stress inhibits electron transfer during the photoresponse by acting on different sites of the PSI and PSII electron transport chain (oxygen-evolving complex on the electron donor side of PSII and sites such as QA and QB on the electron acceptor side of PSII) [201,202], thereby affecting the synthesis of plant ATPase and other physiological processes. In addition, Cd induces the production of reactive oxygen species (ROS), leading to protein oxidation, DNA damage, malondialdehyde (MDA) accumulation, and even damage to cell membranes [203]. Apart from polluting the environment and poisoning plants and animals, Cd has been reported to act as a “hormone”, i.e., at low concentrations, it activates plant defense mechanisms without causing severe oxidative stress [29,204,205]. Currently, there are few studies in this area, which need to be verified in different environments and plants.
The toxic effects of Cd are manifested in physiological and ecological aspects, but the degree of its toxic effects is related to Cd concentration and treatment time, plant species, and cultivars [206]. For instance, a low Cd concentration of 100 nM affected the growth of sunflower [207], while Sesuvium portulacastrum showed significant cellular damage only after 300 μM CdCl2 treatment [208]. Furthermore, certain plants are classified as Cd hyperaccumulators for their ability to accumulate Cd in their aerial tissues in excess of 100 mg Kg−1, showing high tolerance and uptake capacity [209,210].
Hence, when exploring the mechanism of plants to cope with Cd toxicity, researchers use different concentrations of Cd in hydroponic or soil culture treatments to screen and explore Cd high-tolerant plant species based on physiological data such as phenotype, plant height, root parameters, and photosynthetic efficiency, and changes in antioxidant enzyme activities.

4.2. Mechanisms of Plant Response to Cadmium Stress

To cope with the stress of the heavy metal Cd, plants have evolved elegant defense mechanisms (Figure 4). Firstly, when confronting Cd in soils, the plant root system secretes substances such as malic acid and citric acid, which bind to Cd2+ to prevent their uptake by the root system [211]. Secondly, after entering into the roots, Cd binds to polygalacturonic acid and pectin in the cell wall [212,213], thus reducing the amount of Cd in the cytosol. On the other hand, the Casparian strips on the endodermis of the root also prevent Cd from entering the cell [214]. Thirdly, chelation and vacuole isolation of Cd is also one of the important detoxification pathways. On the one hand, free Cd2+ binds to glutathione (GSH), phytochelatins (PCs), and metallothioneins (MTs) to form non-toxic complexes such as Cd-GS2, PC-Cd, MT-Cd, and so on [29]. On the other hand, plants isolate Cd2+ and complexes by transporting them from the cytoplasm to the vacuole through transporter proteins, thereby reducing the toxic effects of Cd on plants and enhancing their tolerance to Cd. Finally, plants keep more Cd in their roots by decreasing the long-distance transport of Cd in the xylem from root to shoot, thus mitigating the negative effects of Cd on leaves or productive organs.
When Cd crosses the barrier of plant cells, it will trigger a burst of ROS. Then the activation of antioxidant enzymes such as superoxide dismutase (SOD), peroxidase, (POD) and increased production of non-enzymatic antioxidants tocopherol and flavonoids follow (Figure 3). They are widely present in various organelles and act to eliminate the excessive accumulation of O2-, H2O2 and malonaldehyde to maintain intracellular environmental homeostasis. In addition, sugars, amino acids, and polyols, as an osmotic pressure regulator in plants, can maintain intracellular balance and improve plant tolerance when plants are subjected to abiotic stresses [215,216], and they can also inhibit the production of oxyradicals, scavenge excess ROS, and mitigate oxidative damage in plants [217]. It is noteworthy that salicylic acid (SA), gibberellin (GA), and ABA have been demonstrated to play pivotal roles in mitigating Cd-induced oxidative stress [29,192,218,219]. While other hormones appear to exert a regulatory influence, their specific mechanisms remain to be thoroughly explored.
According to previous studies, the above-mentioned approaches can be classified into two different strategies: avoidance and tolerance of Cd. The former is to prevent Cd from entering the cells of plants to protect plants from Cd stress, and the latter is dependent on the plant’s own tolerance and mitigation mechanisms to alleviate the negative effects of Cd. The two strategies are complementary to each other and together constitute the plant’s defense mechanism against Cd toxicity.

5. Effect of Exogenous Substances on Phytoremediation of Soil Cadmium Pollution

It is obvious that high levels of heavy metals negatively affect plant growth and development. Under the strong selective pressures exerted by heavy metal in soils, many plants have evolved sophisticated biological mechanisms for resisting, tolerating, or thriving in metal-bearing soils, and are collectively referred to as heavy-metal plants [220]. Of these, those that can only survive in contaminated areas are known as obligate metallophytes [221].

5.1. Status of Phytoremediation and Its Application

Phytoremediation is a remediation technique via plants or soil microorganisms to reduce pollutants in the surrounding environment [222], which include heavy metals, radionuclides, or organic pollutants such as polynuclear aromatic hydrocarbons (PAHs), polychlorinated biphenyls (PCBs) and pesticides in soils or air [223]. Compared with traditional physical and chemical methods, phytoremediation is green, low-cost, and highly effective [224].
Currently, the most used plants for phytoremediation of Cd-excessive soils are hyper-enriched plants, which accumulate more than 0.01 percent of Cd in their aboveground dry weight [225]. Although more than 450 plant species have been identified as metal hyper-enriched plants, only a few plant species are recognized as Cd hyper-enriched, such as Solanum nigrum, Phytolacca acinosa, and Sedum plumbizincicola [226]. More interestingly, these Cd-hyper-enriched plants were also Zn-hyper-enriched plants and vice versa [227]. The Cd extraction capacity of plants was different in soils, with the Cd bioconcentration factor (BCF) of plant leaves being larger in acidic soils and smaller in alkaline soils [226].
In addition, woody plants are receiving increased attention as an alternative phytoremediation technique. Recently, it has been found that fast-growing woody plants can accumulate high levels of heavy metals. For instance, a one-year-old willow can extract 17% of the Cd in soils [228], and a four-year-old Averrhoa carambola can remove 0.3% to 51.8% of the total Cd from soils at the surface of 20 cm [229]. This suggests that woody plants have greater potential for absorbing and accumulating Cd.
However, plants are prone to toxicity during heavy metal uptake and growth is affected. This greatly limits the efficiency of phytoremediation. With the deterioration of the environment, staple food crops such as rice and maize are also at risk of exceeding the heavy metal content standards. Therefore, enhancing plant tolerance and improving the efficiency of phytoremediation through the addition of different exogenous substances, or converting soil Cd into a non-absorbable form is a valuable approach in agriculture.

5.2. Effects of Adding Exogenous Substances to Plants

It has been verified that exogenous supplementation of beneficial elements, plant growth regulators, or nanomaterials can alleviate the Cd stress, thus approaching normal levels of plant height, leaf photosynthesis, and respiration rate [93,230,231,232].
Ca is essential for plant growth and development [233]. When used as an exogenous substance, Ca can reduce Cd-induced physiological and biochemical disorders [234], and also down-regulate Cd accumulation by decreasing the negative charge of the cell membrane surface [235] in Salix matsudana [236] and Fagopyrum esculentum [237]. As one of the beneficial elements, Se plays an important role in improving plant stress tolerance. Low concentrations of Se can enhance antioxidant capacity and membrane stability and reduce the uptake of heavy metals and the accumulation of ROS [238]. Se has been reported to alleviate Cd stress in rice, oilseed rape, and sunflower by counteracting Cd-induced nutritional changes and reducing oxidative stress [239,240,241]. It is worth noting that Se is like a ‘‘rapier’’ in plants, with low concentrations producing beneficial effects and high concentrations causing stress instead [230,238].
In short, when plants are subjected to Cd stress, if the exogenous elements are metallic elements such as iron (Fe), Ca, and potassium (K), the mitigation mechanism may be because they regulate the biochemical and physiological aspects of the plants, mitigate toxicity, or compete with Cd to the transporter, thereby reducing the amount absorbed by the plant. If non-metallic elements such as boron (B), Se, and Si, are added as exogenous elements, the mitigation mechanism may be because these elements act as nutrients for plants and enhance the tolerance of plants, or they may form complexes with Cd and reduce the uptake of the plants.
Polyamines, along with hormones like 1-naphthaleneacetic acid (NAA) and ABA, serve as plant growth regulators for the modulation of plant development and the augmentation of plant tolerance to Cd [232,242,243]. Wherein, supplementation of exogenous NAA increases the content of Arabidopsis hemicellulose 1, which immobilizes more Cd in the roots [243]. Exogenous application of polyamines, on the other hand, mitigates the adverse effects of Cd contamination on wheat by activating antioxidant enzyme activities [242]. In addition, with the development of technology, nanomaterials and biochar materials have also become ideal candidates for solving Cd pollution. Biogenic hydroxyapatite nanoparticles effectively mitigate the toxicity of Cd to mung beans by adsorbing Cd2+ from the environment and forming a protective layer on plant roots [231]. Biochar materials reduce Cd availability and lignocellulosic biochar and herbaceous biochar have a broader range of remediation applications than manure biochar [244].
Of note, in addition to the exogenous substances mentioned above, the addition of melatonin, citric acid, and amino acids can also lead to higher Cd tolerance in plants [245,246,247]. Therefore, exploring more beneficial exogenous substances, forms, and proportions of additions are of practical significance to improve the efficiency of phytoremediation.

6. Conclusions and Perspectives

Cd initiates signal transduction cascades in plants. The exposure to Cd-induced stress in Arabidopsis reduced endogenous auxin content. Concurrently, exogenous supplementation with NAA enhanced the fixation of Cd to the cell wall through an elevation in hemicellulose 1 levels in A. thaliana [243]. ABA mitigates oxidative stress following exposure to Cd through the ABI5-MYB49-bHLH cascade, activation of the glutathione pathway, and the formation of an apoplastic barrier [232,248]. In contrast to ABA, ethylene amplifies the deleterious effects of Cd on plants in two distinct manners: by augmenting the generation of reactive oxygen species via RBOHC or by impeding the establishment of the apoplastic barrier through unidentified pathways [232,248]. Currently, the influences of auxin, ABA, ethylene, and other hormones on Cd tolerance in plants have been initially revealed [232], yet their molecular mechanisms and regulatory networks remain elusive. Given that distinct hormones may elicit contrasting responses under Cd-induced stress, how do they intricately interplay? How do hormone signals crosstalk when encountering ROS or other signals? In addition to the aforementioned circumstances, plant roots may experience heterogeneous Cd stress, characterized by intense pressure on one side and minimal or absent pressure on the other. At this point, plants remodel the root structure and avoid the side with the high level of Cd stress through the RBOH-ROS-growth hormone signaling cascade [249]. The exploration of whether other signaling cascades exist in this process and how they function necessitates further investigation.
A shows the screening of Cd-hyperaccumulator plants and fast-growing woody plants that can accumulate large amounts of Cd. B shows the application of different exogenous substances to plants or genetic modification to make plants have higher Cd tolerance.
Throughout plant evolution, certain species have developed robust resistance mechanisms as a result of prolonged adaptation to high Cd pollution. Therefore, these highly tolerant plants can be regarded as potential candidates for mitigating excessive Cd pollution in the environment. At present, Cd-hyperaccumulator plants, predominantly wild herbs, are frequently employed in phytoremediation. However, due to their limited aboveground biomass, the overall Cd absorption capacity remains relatively modest. Conversely, fast-growing woody plants, such as poplars and willows, boast larger size, greater biomass, and a swifter growth rate, thereby enabling them to absorb more Cd (Figure 5A). Phytoremediation technology has a broad application prospect in Cd-contaminated soil remediation, but many aspects have not yet been clarified, and for Cd-hyperaccumulator plants, it is worth exploring whether they can improve their growth rate, biomass and accumulated heavy metal content, and stress resistance. For fast-growing woody plants, it is necessary to continue to explore their absorption mechanisms at the molecular level and apply transgenic technology or gene editing to improve the uptake of Cd (Figure 5B).
To optimize the efficiency of Cd pollution phytoremediation, we propose the supplementation of plant regulators (exogenous hormones or polyamines) and beneficial elements to plant nutrition (Figure 5B). Studies have reported that employing this approach confers beneficial effects on plant metabolic pathways and enhances stress resilience [232,236,240,242]. At present, there are fewer studies on the compound addition of multiple substances, and it is worthwhile to explore in depth under what ratio different elements or hormones are added to produce better results. In addition, the application of nanomaterials, biochar materials, and microorganisms can effectively reduce the effectiveness of Cd in the soil, so the combination of nanomaterials/biochar, clumping rhizobial fungi, plant growth-promoting bacteria with Ca, K, B, Si may be an effective way to reduce the uptake of Cd by plants [250]. In conclusion, the discovery of environmentally friendly, cost-effective exogenous substances holds significance for crop and vegetable production.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/cells13110907/s1, Table S1: Gene family related to Cd Chelation, accumulation, and detoxification. Table S2: Transcription factors (TFs) families related to Cd uptake, transport, and tolerance. Refs. [251,252,253,254,255,256,257,258,259,260,261,262,263,264,265,266,267,268,269,270,271,272,273,274,275,276,277,278,279,280,281,282,283,284,285,286,287,288,289,290,291,292,293,294,295,296,297,298,299,300,301,302,303,304,305,306,307,308,309,310,311,312,313,314,315,316,317,318,319,320,321,322,323,324,325,326,327,328,329,330,331,332,333,334,335] are cited within.

Author Contributions

X.Z.: Conceptualization, Writing—original draft, Visualization, lnvestigation. M.Y.: Data curation, lnvestigation, Writing—review & editing. H.Y.: lnvestigation, Visualization, Writing—review & editing. R.P.: Conceptualization, Writing—review & editing, Supervision. J.W.: Funding acquisition, Writing—review & editing, Supervision. A.-M.W.: Conceptualization, Funding acquisition, Writing—review & editing. All authors commented on previous versions of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the National Key Research and Development Program of China (Grant No. 2022YFD2200102) and Guangzhou Science and Technology Plan Project (Grant No. 202206010163).

Institutional Review Board Statement

This article does not contain any studies with human participants or animals performed by any of the authors.

Data Availability Statement

The datasets generated during and/or analyzed during the current study are available from the corresponding author on reasonable request.

Acknowledgments

We thank all anonymous reviewers and editors for their constructive comments and suggestions. At the same time, we are also grateful to Freepik.com for providing materials and the visual drawing tools provided by the Figdraw (www.figdraw.com) platform, accessed on 30 January 2024.

Conflicts of Interest

The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: the figures in the review except Figure 1 and Figure 2 were drawn by Figdraw.

References

  1. UNEP. Interim Review of Scientific Information on Cadmium. 2008. Available online: https://wedocs.unep.org/handle/20.500.11822/27636 (accessed on 30 January 2024).
  2. Ullah, H.; Khan, N.U.; Ali, F.; Shah, Z.A.; Ullah, Q. Health risk of heavy metals from vegetables irrigated with sewage water in peri-urban of dera ismail khan, Pakistan. Int. J. Environ. Sci. Technol. 2018, 15, 309–322. [Google Scholar] [CrossRef]
  3. Yang, Y.; Wang, T.; Li, Y.; Wang, M.; Chen, W.; Dai, Y. Mitigating cadmium contamination of rice soils supporting tobacco–rice rotation in southern China: Win–win or lose–lose? J. Hazard. Mater. 2022, 425, 128052. [Google Scholar] [CrossRef] [PubMed]
  4. Nogawa, K.; Kido, T. Itai-Itai Disease and Health Effects of Cadmium; CRC Press: Boca Raton, FL, USA, 2023; pp. 353–369. [Google Scholar]
  5. Satarug, S. Dietary cadmium intake and its effects on kidneys. Toxics 2018, 6, 15. [Google Scholar] [CrossRef] [PubMed]
  6. Smolders, E.; Mertens, J.; Cadmium; Alloway, B.J. Heavy Metals. In Soils: Trace Metals and Metalloids in Soils and Their Bioavailability; Springer: Dordrecht, The Netherlands, 2013; pp. 283–311. [Google Scholar]
  7. Yuan, X.; Xue, N.; Han, Z. A meta-analysis of heavy metals pollution in farmland and urban soils in China over the past 20 years. J. Environ. Sci. 2021, 101, 217–226. [Google Scholar] [CrossRef]
  8. Holmgren, G.; Meyer, M.W.; Chaney, R.L.; Daniels, R.B. Cadmium, lead, zinc, copper, and nickel in agricultural soils of the united states of america. J. Environ. Qual. 1993, 22, 335–348. [Google Scholar] [CrossRef]
  9. Crea, F.; Foti, C.; Milea, D.; Sammartano, S. Speciation of cadmium in the environment. In Cadmium: From Toxicity to Essentiality; Sigel, A., Sigel, H., Sigel, R., Eds.; Springer: Dordrecht, The Netherlands, 2013; pp. 63–83. [Google Scholar]
  10. Bigalke, M.; Ulrich, A.; Rehmus, A.; Keller, A. Accumulation of cadmium and uranium in arable soils in Switzerland. Environ. Pollut. 2017, 221, 85–93. [Google Scholar] [CrossRef] [PubMed]
  11. Sall, M.L.; Diaw, A.; Gningue-Sall, D.; Aaron, S.E.; Aaron, J.J. Toxic heavy metals: Impact on the environment and human health, and treatment with conducting organic polymers, a review. Environ. Sci. Pollut. Res. 2020, 27, 29927–29942. [Google Scholar] [CrossRef] [PubMed]
  12. Han, F.; Banin, A.; Su, Y.; Monts, D.L.; Plodinec, M.J.; Kingery, W.L.; Triplett, G.E. Industrial age anthropogenic inputs of heavy metals into the pedosphere. Sci. Nat. 2002, 89, 497–504. [Google Scholar] [CrossRef] [PubMed]
  13. Sigel, A.; Sigel, H.; Sigel, R.K. Cadmium: From Toxicity to Essentiality; Springer: Berlin/Heidelberg, Germany, 2013. [Google Scholar]
  14. Nortvedt, J.J.; Osborn, G. Studies on the chemical form of cadmium contaminants in phosphate fertilizers. Soil Sci. 1982, 134, 185–192. [Google Scholar] [CrossRef]
  15. Satarug, S.; Vesey, D.A.; Gobe, G.C. Current health risk assessment practice for dietary cadmium: Data from different countries. Food Chem. Toxicol. 2017, 106, 430–445. [Google Scholar] [CrossRef]
  16. Genchi, G.; Sinicropi, M.S.; Lauria, G.; Carocci, A.; Catalano, A. The effects of cadmium toxicity. Int. J. Environ. Res. Public Health 2020, 17, 3782. [Google Scholar] [CrossRef] [PubMed]
  17. Tessier, A.; Campbell, P.G.; Bisson, M. Sequential extraction procedure for the speciation of particulate trace metals. Anal. Chem. 1979, 51, 844–851. [Google Scholar] [CrossRef]
  18. Wei, B.; Peng, Y.; Jeyakumar, P.; Lin, L.; Zhang, D.; Yang, M.; Zhu, J.; Lin, C.S.K.; Wang, H.; Wang, Z.; et al. Soil ph restricts the ability of biochar to passivate cadmium: A meta-analysis. Environ. Res. 2023, 219, 115110. [Google Scholar] [CrossRef]
  19. Xian, X.; Shokohifard, G.I. Effect of ph on chemical forms and plant availability of cadmium, zinc, and lead in polluted soils. Water Air Soil Pollut. 1989, 45, 265–273. [Google Scholar] [CrossRef]
  20. Eriksson, J.E. The influence of ph, soil type and time on adsorbtion and uptake by plants of cd added to the soil. Water Air Soil Pollut. 1989, 48, 317–335. [Google Scholar] [CrossRef]
  21. Yin, Y.C.; Wang, Y.Q.; Liu, Y.G.; Zeng, G.M.; Hu, X.J.; Hu, X.; Zhou, L.; Guo, Y.M.; Li, J. Cadmium accumulation and apoplastic and symplastic transport in Boehmeria nivea (L.) Gaudich on cadmium-contaminated soil with the addition of edta or nta. RSC Adv. 2015, 5, 47584–47591. [Google Scholar] [CrossRef]
  22. Huang, H.; Li, M.; Rizwan, M.; Dai, Z.; Yuan, Y.; Hossain, M.M.; Cao, M.; Xiong, S.; Tu, S. Synergistic effect of silicon and selenium on the alleviation of cadmium toxicity in rice plants. J. Hazard. Mater. 2021, 401, 123393. [Google Scholar] [CrossRef] [PubMed]
  23. Kim, Y.; Khan, A.L.; Kim, D.; Lee, S.; Kim, K.; Waqas, M.; Jung, H.; Shin, J.; Kim, J.; Lee, I. Silicon mitigates heavy metal stress by regulating p-type heavy metal atpases, Oryza sativalow silicon genes, and endogenous phytohormones. BMC Plant Biol. 2014, 14, 13. [Google Scholar] [CrossRef] [PubMed]
  24. Shao, G.; Chen, M.; Wang, W.; Mou, R.; Zhang, G. Iron nutrition affects cadmium accumulation and toxicity in rice plants. Plant Growth Regul. 2007, 53, 33–42. [Google Scholar] [CrossRef]
  25. Volpe, M.G.; Nazzaro, M.; Di Stasio, M.; Siano, F.; Coppola, R.; De Marco, A. Content of micronutrients, mineral and trace elements in some mediterranean spontaneous edible herbs. Chem. Cent. J. 2015, 9, 57. [Google Scholar] [CrossRef]
  26. Yamaguchi, N.; Mori, S.; Baba, K.; Kaburagi-Yada, S.; Arao, T.; Kitajima, N.; Hokura, A.; Terada, Y. Cadmium distribution in the root tissues of solanaceous plants with contrasting root-to-shoot cd translocation efficiencies. Environ. Exp. Bot. 2011, 71, 198–206. [Google Scholar] [CrossRef]
  27. Sasaki, A.; Yamaji, N.; Yokosho, K.; Ma, J.F. Nramp5 is a major transporter responsible for manganese and cadmium uptake in rice. Plant Cell 2012, 24, 2155–2167. [Google Scholar] [CrossRef]
  28. Shahid, M.; Dumat, C.; Khalid, S.; Niazi, N.K.; Antunes, P.M.C. Cadmium bioavailability, uptake, toxicity and detoxification in soil-plant system. In Reviews of Environmental Contamination and Toxicology; de Voogt, P., Gunther, F.A., Eds.; Springer International Publishing: Cham, Switzerland, 2017; Volume 241, pp. 73–137. [Google Scholar]
  29. Zhao, F.; Wang, P. Arsenic and cadmium accumulation in rice and mitigation strategies. Plant Soil 2020, 446, 1–21. [Google Scholar] [CrossRef]
  30. Song, Y.; Jin, L.; Wang, X.J. Cadmium absorption and transportation pathways in plants. Int. J. Phytoremediat. 2017, 19, 133–141. [Google Scholar] [CrossRef] [PubMed]
  31. Cai, H.; Huang, S.; Che, J.; Yamaji, N.; Ma, J.F. The tonoplast-localized transporter oshma3 plays an important role in maintaining Zn homeostasis in rice. J. Exp. Bot. 2019, 70, 2717–2725. [Google Scholar] [CrossRef] [PubMed]
  32. Akhter, M.F.; Omelon, C.R.; Gordon, R.A.; Moser, D.; Macfie, S.M. Localization and chemical speciation of cadmium in the roots of barley and lettuce. Environ. Exp. Bot. 2014, 100, 10–19. [Google Scholar] [CrossRef]
  33. Ismael, M.A.; Elyamine, A.M.; Moussa, M.G.; Cai, M.; Zhao, X.; Hu, C. Cadmium in plants: Uptake; toxicity, and its interactions with selenium fertilizers. Metallomics 2019, 11, 255–277. [Google Scholar] [CrossRef] [PubMed]
  34. Qin, Q.; Li, X.; Zhuang, J.; Weng, L.; Liu, W.; Tai, P. Long-distance transport of cadmium from roots to leaves of solanum melongena. Ecotoxicology 2015, 24, 2224–2232. [Google Scholar] [CrossRef] [PubMed]
  35. Kov, V.K.; Park, S.; Cheng, N.H.; Sreevidya, C.; Lachmansingh, J.; Morris, J.; Hirschi, K.; Wagner, G.J. Enhanced Cd2+-selective root-tonoplast-transport in tobaccos expressing arabidopsis cation exchangers. Planta 2007, 225, 403–411. [Google Scholar]
  36. Korenkov, V.; Hirschi, K.; Crutchfield, J.D.; Wagner, G.J. Enhancing tonoplast cd/h antiport activity increases Cd, Zn, and Mn tolerance, and impacts root/shoot cd partitioning in Nicotiana tabacum L. Planta 2007, 226, 1379–1387. [Google Scholar] [CrossRef]
  37. Khanna, K.; Kohli, S.K.; Ohri, P.; Bhardwaj, R.; Ahmad, P. Agroecotoxicological aspect of Cd in soil-plant system: Uptake, translocation and amelioration strategies. Environ. Sci. Pollut. Res. 2022, 29, 30908–30934. [Google Scholar] [CrossRef] [PubMed]
  38. Van Belleghem, F.; Cuypers, A.; Semane, B.; Smeets, K.; Vangronsveld, J.; D’Haen, J.; Valcke, R. Subcellular localization of cadmium in roots and leaves of Arabidopsis thaliana. New Phytol. 2007, 173, 495–508. [Google Scholar] [CrossRef] [PubMed]
  39. Zhang, J.L.; Zhu, Y.C.; Yu, L.J.; Yang, M.; Zou, X.; Yin, C.X.; Lin, Y.J. Research advances in cadmium uptake, transport and resistance in rice (Oryza sativa L.). Cells 2022, 11, 569. [Google Scholar] [CrossRef] [PubMed]
  40. Tanaka, K.; Fujimaki, S.; Fujiwara, T.; Yoneyama, T.; Hayashi, H. Quantitative estimation of the contribution of the phloem in cadmium transport to grains in rice plants (Oryza sativa L.). Soil Sci. Plant Nutr. 2007, 53, 72–77. [Google Scholar] [CrossRef]
  41. Kutrowska, A.; Szelag, M. Low-molecular weight organic acids and peptides involved in the long-distance transport of trace metals. Acta Physiol. Plant. 2014, 36, 1957–1968. [Google Scholar] [CrossRef]
  42. Cózatl, D.G.M.; Butko, E.; Springer, F.; Torpey, J.W.; Komives, E.A.; Kehr, J.; Schroeder, J.I. Identification of high levels of phytochelatins, glutathione and cadmium in the phloem sap of Brassica napus. A role for thiol-peptides in the long-distance transport of cadmium and the effect of cadmium on iron translocation. Plant J. 2008, 54, 249–259. [Google Scholar] [CrossRef] [PubMed]
  43. Palmgren, M.G.; Clemens, S.; Williams, L.E.; Krämer, U.; Borg, S.; Schjørring, J.K.; Sanders, D. Zinc biofortification of cereals: Problems and solutions. Trends Plant Sci. 2008, 13, 464–473. [Google Scholar] [CrossRef] [PubMed]
  44. Ueno, D.; Yamaji, N.; Kono, I.; Huang, C.F.; Ando, T.; Yano, M.; Ma, J.F. Gene limiting cadmium accumulation in rice. Proc. Natl. Acad. Sci. USA 2010, 107, 16500–16505. [Google Scholar] [CrossRef]
  45. Tan, Z.; Li, J.; Guan, J.; Wang, C.; Zhang, Z.; Shi, G. Genome-wide identification and expression analysis reveals roles of the nramp gene family in iron/cadmium interactions in peanut. Int. J. Mol. Sci. 2023, 24, 1713. [Google Scholar] [CrossRef]
  46. Chang, J.; Xie, Y.; Zhang, H.; Zhang, S.; Zhao, F. The vacuolar transporter osnramp2 mediates fe remobilization during germination and affects cd distribution to rice grain. Plant Soil 2022, 476, 79–95. [Google Scholar] [CrossRef]
  47. Yang, W.; Chen, L.; Ma, Y.; Hu, R.; Wang, J.; Li, W.; Dong, J.; Yang, T.; Zhou, L.; Chen, J. Osnramp2 facilitates cd efflux from vacuoles and contributes to the difference in grain cd accumulation between japonica and indica rice. Crop J. 2023, 11, 417–426. [Google Scholar] [CrossRef]
  48. Li, Y.; Li, J.; Yu, Y.; Dai, X.; Gong, C.; Gu, D.; Xu, E.; Liu, Y.; Zou, Y.; Zhang, P. The tonoplast-localized transporter osnramp2 is involved in iron homeostasis and affects seed germination in rice. J. Exp. Bot. 2021, 72, 4839–4852. [Google Scholar] [CrossRef]
  49. Ishimaru, Y.; Takahashi, R.; Bashir, K.; Shimo, H.; Senoura, T.; Sugimoto, K.; Ono, K.; Yano, M.; Ishikawa, S.; Arao, T.; et al. Characterizing the role of rice nramp5 in manganese, iron and cadmium transport. Sci. Rep. 2012, 2, 286. [Google Scholar] [CrossRef]
  50. Chang, J.; Huang, S.; Konishi, N.; Wang, P.; Chen, J.; Huang, X.; Ma, J.F.; Zhao, F. Overexpression of the manganese/cadmium transporter osnramp5 reduces cadmium accumulation in rice grain. J. Exp. Bot. 2020, 71, 5705–5715. [Google Scholar] [CrossRef]
  51. Tang, L.; Mao, B.; Li, Y.; Lv, Q.; Zhang, L.; Chen, C.; He, H.; Wang, W.; Zeng, X.; Shao, Y.; et al. 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] [PubMed]
  52. 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] [PubMed]
  53. 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]
  54. Takahashi, R.; Ishimaru, Y.; Senoura, T.; Shimo, H.; Ishikawa, S.; Arao, T.; Nakanishi, H.; Nishizawa, N.K. The osnramp1 iron transporter is involved in cd accumulation in rice. J. Exp. Bot. 2011, 62, 4843–4850. [Google Scholar] [CrossRef]
  55. Mäser, P.; Thomine, S.; Schroeder, J.; Ward, J.; Hirschi, K.; Sze, H.; Talke, I.; Amtmann, A.; Maathuis, F.; Sanders, D.; et al. Phylogenetic relationships within cation transporter families of Arabidopsis. Plant Physiol. 2001, 126, 1646–1667. [Google Scholar] [CrossRef]
  56. Bughio, N.; Yamaguchi, H.; Nishizawa, N.K.; Nakanishi, H.; Mori, S. Cloning an iron-regulated metal transporter from rice. J. Exp. Bot. 2002, 53, 1677–1682. [Google Scholar] [CrossRef]
  57. Connolly, E.L.; Fett, J.P.; Guerinot, M.L. Expression of the irt1 metal transporter is controlled by metals at the levels of transcript and protein accumulation. Plant Cell 2002, 14, 1347–1357. [Google Scholar] [CrossRef] [PubMed]
  58. Rogers, E.E.; Eide, D.J.; Guerinot, M.L. Altered selectivity in an Arabidopsis metal transporter. Proc. Natl. Acad. Sci. USA 2000, 97, 12356–12360. [Google Scholar] [CrossRef] [PubMed]
  59. Vert, G.; Grotz, N.; De, F.; Gaymard, F.; Guerinot, M.L.; Briat, J.; Curie, C. IRT1, an Arabidopsis transporter essential for iron uptake from the soil and for plant growth. Plant Cell 2002, 14, 1223–1233. [Google Scholar] [CrossRef]
  60. Korshunova, Y.O.; Eide, D.; Clark, W.G.; Guerinot, M.L.; Pakrasi, H.B. The irt1 protein from Arabidopsis thaliana is a metal transporter with a broad substrate range. Plant Mol.Biol. 1999, 40, 37–44. [Google Scholar] [CrossRef] [PubMed]
  61. Vert, G.; Barberon, M.; Zelazny, E.; Séguéla, M.; Briat, J.; Curie, C. Arabidopsis irt2 cooperates with the high-affinity iron uptake system to maintain iron homeostasis in root epidermal cells. Planta 2009, 229, 1171–1179. [Google Scholar] [CrossRef] [PubMed]
  62. Vert, G.; Briat, J.F.; Curie, C. Arabidopsis irt2 gene encodes a root-periphery iron transporter. Plant J. 2001, 26, 181–189. [Google Scholar] [CrossRef] [PubMed]
  63. Milner, M.J.; Seamon, J.; Craft, E.; Kochian, L.V. Transport properties of members of the zip family in plants and their role in Zn and Mn homeostasis. J. Exp. Bot. 2013, 64, 369–381. [Google Scholar] [CrossRef] [PubMed]
  64. Grotz, N.; Guerinot, M.L. Molecular aspects of Cu, Fe and Zn homeostasis in plants. Biochim. Et Biophys. Acta (BBA)—Mol. Cell Res. 2006, 1763, 595–608. [Google Scholar] [CrossRef]
  65. Wintz, H.; Fox, T.; Wu, Y.; Feng, V.; Chen, W.; Chang, H.; Zhu, T.; Vulpe, C. Expression profiles of Arabidopsis thaliana in mineral deficiencies reveal novel transporters involved in metal homeostasis. J. Biol. Chem. 2003, 278, 47644–47653. [Google Scholar] [CrossRef]
  66. Tiong, J.; Mcdonald, G.; Genc, Y.; Shirley, N.; Langridge, P.; Huang, C.Y. Increased expression of six zip family genes by zinc (Zn) deficiency is associated with enhanced uptake and root-to-shoot translocation of Zn in barley (Hordeum vulgare). New Phytol. 2015, 207, 1097–1109. [Google Scholar] [CrossRef]
  67. Lee, S.; An, G. Over-expression of osirt1 leads to increased iron and zinc accumulations in rice. Plant Cell Environ. 2009, 32, 408–416. [Google Scholar] [CrossRef]
  68. Li, Z.; Liang, Y.; Hu, H.; Shaheen, S.M.; Zhong, H.; Tack, F.M.G.; Wu, M.; Li, Y.; Gao, Y.; Rinklebe, J.; et al. Speciation, transportation, and pathways of cadmium in soil-rice systems: A review on the environmental implications and remediation approaches for food safety. Environ. Int. 2021, 156, 106749. [Google Scholar] [CrossRef] [PubMed]
  69. Yang, Y.; Xiong, J.; Chen, R.; Fu, G.; Chen, T.; Tao, L. Excessive nitrate enhances cadmium (cd) uptake by up-regulating the expression of osirt1 in rice (Oryza sativa). Environ. Exp. Bot. 2016, 122, 141–149. [Google Scholar] [CrossRef]
  70. 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, 283. [Google Scholar] [CrossRef] [PubMed]
  71. Tao, J.; Lu, L. Advances in genes-encoding transporters for cadmium uptake, translocation, and accumulation in plants. Toxics 2022, 10, 411. [Google Scholar] [CrossRef] [PubMed]
  72. Ramesh, S.A.; Shin, R.; Eide, D.J.; Schachtman, D.P. Differential metal selectivity and gene expression of two zinc transporters from rice. Plant Physiol. 2003, 133, 126–134. [Google Scholar] [CrossRef] [PubMed]
  73. Tian, S.; Liang, S.; Qiao, K.; Wang, F.; Zhang, Y.; Chai, T. Co-expression of multiple heavy metal transporters changes the translocation, accumulation, and potential oxidative stress of cd and Zn in rice (Oryza sativa). J. Hazard. Mater. 2019, 380, 120853. [Google Scholar] [CrossRef] [PubMed]
  74. Tan, L.; Qu, M.; Zhu, Y.; Peng, C.; Wang, J.; Gao, D.; Chen, C. Zinc transporter5 and zinc transporter9 function synergistically in zinc/cadmium uptake. Plant Physiol. 2020, 183, 1235–1249. [Google Scholar] [CrossRef] [PubMed]
  75. Kavitha, P.G.; Kuruvilla, S.; Mathew, M.K. Functional characterization of a transition metal ion transporter, oszip6 from rice (Oryza sativa L.). Plant Physiol. Biochem. 2015, 97, 165–174. [Google Scholar]
  76. Tan, L.; Zhu, Y.; Fan, T.; Peng, C.; Wang, J.; Sun, L.; Chen, C. Oszip7 functions in xylem loading in roots and inter-vascular transfer in nodes to deliver zn/cd to grain in rice. Biochem. Biophys. Res. Commun. 2019, 512, 112–118. [Google Scholar] [CrossRef]
  77. Zheng, X.; Chen, L.; Li, X. Arabidopsis and rice showed a distinct pattern in zips genes expression profile in response to cd stress. Bot. Stud. 2018, 59, 22. [Google Scholar] [CrossRef] [PubMed]
  78. Argüello, J.M.; Eren, E.; González-Guerrero, M. The structure and function of heavy metal transport p1b-atpases. Biometals 2007, 20, 233–248. [Google Scholar] [CrossRef] [PubMed]
  79. Zhiguo, E.; Tingting, L.; Chen, C.; Lei, W. Genome-wide survey and expression analysis of p1b-atpases in rice, maize and sorghum. Rice Sci. 2018, 25, 208–217. [Google Scholar] [CrossRef]
  80. 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]
  81. Lee, S.; Kim, Y.; Lee, Y.; An, G. Rice p1b-type heavy-metal atpase; oshma9, is a metal efflux protein. Plant Physiol. 2007, 145, 831–842. [Google Scholar] [CrossRef] [PubMed]
  82. Kim, Y.Y.; Choi, H.; Segami, S.; Cho, H.T.; Martinoia, E.; Maeshima, M.; Lee, Y. Athma1 contributes to the detoxification of excess Zn (ii) in Arabidopsis. Plant J. 2009, 58, 737–753. [Google Scholar] [CrossRef] [PubMed]
  83. Seigneurin-Berny, D.; Gravot, A.; Auroy, P.; Mazard, C.; Kraut, A.; Finazzi, G.; Grunwald, D.; Rappaport, F.; Vavasseur, A.; Joyard, J. HMA1, a new cu-atpase of the chloroplast envelope, is essential for growth under adverse light conditions. J. Biol. Chem. 2006, 281, 2882–2892. [Google Scholar] [CrossRef]
  84. Moreno, I.; Norambuena, L.; Maturana, D.; Toro, M.; Vergara, C.; Orellana, A.; Zurita-Silva, A.; Ordenes, V.R. Athma1 is a thapsigargin-sensitive Ca2+/heavy metal pump. J. Biol. Chem. 2008, 283, 9633–9641. [Google Scholar] [CrossRef]
  85. Eren, E.; Argu, J.M. Arabidopsis hma2, a divalent heavy metal-transporting pib-type atpase, is involved in cytoplasmic Zn2+ homeostasis. Plant Physiol. 2004, 136, 3712–3723. [Google Scholar] [CrossRef]
  86. Hussain, D.; Haydon, M.J.; Wang, Y.; Wong, E.; Sherson, S.M.; Young, J.; Camakaris, J.; Harper, J.F.; Cobbett, C.S. P-type atpase heavy metal transporters with roles in essential zinc homeostasis in arabidopsis. Plant Cell 2004, 16, 1327–1339. [Google Scholar] [CrossRef]
  87. Mills, R.F.; Krijger, G.C.; Baccarini, P.J.; Hall, J.L.; Williams, L.E. Functional expression of athma4, a p1b-type atpase of the Zn/Co/Cd/Pb subclass. Plant J. 2003, 35, 164–176. [Google Scholar] [CrossRef] [PubMed]
  88. Verret, F.; Gravot, A.; Auroy, P.; Leonhardt, N.; David, P.; Nussaume, L.; Vavasseur, A.; Richaud, P. Overexpression of athma4 enhances root-to-shoot translocation of zinc and cadmium and plant metal tolerance. Febs Lett. 2004, 576, 306–312. [Google Scholar] [CrossRef] [PubMed]
  89. 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]
  90. Takahashi, R.; Bashir, K.; Ishimaru, Y.; Nishizawa, N.K.; Nakanishi, H. The role of heavy-metal atpases, hmas, in zinc and cadmium transport in rice. Plant Signal. Behav. 2012, 7, 1605–1607. [Google Scholar]
  91. Luo, J.; Huang, J.; Zeng, D.; Peng, J.; Zhang, G.; Ma, H.; Guan, Y.; Yi, H.; Fu, Y.; Han, B. A defensin-like protein drives cadmium efflux and allocation in rice. Nat. Commun. 2018, 9, 645. [Google Scholar] [CrossRef] [PubMed]
  92. Liu, Y.S.; Tao, Y.; Yang, X.Z.; Liu, Y.N.; Shen, R.F.; Zhu, X.F. Gibberellic acid alleviates cadmium toxicity in rice by regulating no accumulation and cell wall fixation capacity of cadmium. J. Hazard. Mater. 2022, 439, 129597. [Google Scholar] [CrossRef] [PubMed]
  93. Wang, F.; Wang, M.; Liu, Z.; Shi, Y.; Han, T.; Ye, Y.; Gong, N.; Sun, J.; Zhu, C. Different responses of low grain-cd-accumulating and high grain-cd-accumulating rice cultivars to cd stress. Plant Physiol. Biochem. 2015, 96, 261–269. [Google Scholar] [CrossRef] [PubMed]
  94. Shimo, H.; Ishimaru, Y.; An, G.; Yamakawa, T.; Nakanishi, H.; Nishizawa, N.K. Low cadmium (lcd), a novel gene related to cadmium tolerance and accumulation in rice. J. Exp. Bot. 2011, 62, 5727–5734. [Google Scholar] [CrossRef] [PubMed]
  95. 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]
  96. Uraguchi, S.; Kamiya, T.; Sakamoto, T.; Kasai, K.; Sato, Y.; Nagamura, Y.; Yoshida, A.; Kyozuka, J.; Ishikawa, S.; Fujiwara, T. Low-affinity cation transporter (oslct1 ) regulates cadmium transport into rice grains. Proc. Natl. Acad. Sci. USA 2011, 108, 20959–20964. [Google Scholar] [CrossRef]
  97. Yan, H.; Xu, W.; Xie, J.; Gao, Y.; Wu, L.; Sun, L.; Feng, L.; Chen, X.; Zhang, T.; Dai, C.; et al. Variation of a major facilitator superfamily gene contributes to differential cadmium accumulation between rice subspecies. Nat. Commun. 2019, 10, 2562. [Google Scholar] [CrossRef] [PubMed]
  98. Lim, S.D.; Hwang, J.G.; Han, A.R.; Park, Y.C.; Lee, C.; Ok, Y.S.; Jang, C.S. Positive regulation of rice ring e3 ligase oshir1 in arsenic and cadmium uptakes. Plant Mol. Biol. 2014, 85, 365–379. [Google Scholar] [CrossRef] [PubMed]
  99. Thomine, S.; Wang, R.; Ward, J.M.; Crawford, N.M.; Schroeder, J.I. Cadmium and iron transport by members of a plant metal transporter family in Arabidopsis with homology to nramp genes. Proc. Natl. Acad. Sci. USA 2000, 97, 4991–4996. [Google Scholar] [CrossRef] [PubMed]
  100. Cailliatte, R.; Schikora, A.; Briat, J.; Mari, S.; Curie, C. High-affinity manganese uptake by the metal transporter nramp1 is essential for Arabidopsis growth in low manganese conditions. Plant Cell 2010, 22, 904–917. [Google Scholar] [CrossRef] [PubMed]
  101. Thomine, S.; Lelièvre, F.; Debarbieux, E.; Schroeder, J.I.; Brygoo, H.B. Atnramp3, a multispecific vacuolar metal transporter involved in plant responses to iron deficiency. Plant J. 2003, 34, 685–695. [Google Scholar] [CrossRef] [PubMed]
  102. Thomine, S.; Schroeder, J.I. Plant Metal Transporters with Homology to Proteins of the NRAMP Family; Landes Bioscience: Austin, TX, USA, 2013. [Google Scholar]
  103. Lanquar, V.; Lelièvre, F.; Barbier-Brygoo, H.; Thomine, S. Regulation and function of atnramp4 metal transporter protein. Soil Sci. Plant Nutr. 2004, 50, 1141–1150. [Google Scholar] [CrossRef]
  104. Cailliatte, R.; Lapeyre, B.; Briat, J.; Mari, S.; Curie, C. The nramp6 metal transporter contributes to cadmium toxicity. Biochem. J. 2009, 422, 217–228. [Google Scholar] [CrossRef]
  105. Liu, W.; Huo, C.; He, L.; Ji, X.; Yu, T.; Yuan, J.; Zhou, Z.; Song, L.; Yu, Q.; Chen, J.; et al. The ntnramp1 transporter is involved in cadmium and iron transport in tobacco (Nicotiana tabacum). Plant Physiol. Biochem. 2022, 173, 59–67. [Google Scholar] [CrossRef] [PubMed]
  106. Jia, H.; Yin, Z.; Xuan, D.; Lian, W.; Han, D.; Zhu, Z.; Li, C.; Li, C.; Song, Z. Mutation of ntnramp3 improves cadmium tolerance and its accumulation in tobacco leaves by regulating the subcellular distribution of cadmium. J. Hazard. Mater. 2022, 432, 128701. [Google Scholar] [CrossRef]
  107. Kozak, K.; Papierniak-Wygladala, A.; Palusińska, M.; Barabasz, A.; Antosiewicz, D.M. Regulation and function of metal uptake transporter ntnramp3 in tobacco. Front. Plant Sci. 2022, 13, 867967. [Google Scholar] [CrossRef]
  108. Tang, Z.; Cai, H.; Li, J.; Lv, Y.; Zhang, W.; Zhao, F. Allelic variation of ntnramp5 associated with cultivar variation in cadmium accumulation in tobacco. Plant Cell Physiol. 2017, 58, 1583–1593. [Google Scholar] [CrossRef] [PubMed]
  109. Zhang, J.; Zhang, X.; Jia, M.; Fu, Q.; Guo, Y.; Wang, Z.; Kong, D.; Lin, Y.; Zhao, D. Two novel transporters ntnramp6a and ntnramp6b are involved in cadmium transport in tobacco (Nicotiana tabacum L.). Plant Physiol. Biochem. 2023, 202, 107953. [Google Scholar]
  110. Zhang, J.; Zhang, M.; Song, H.; Zhao, J.; Shabala, S.; Tian, S.; Yang, X. A novel plasma membrane-based nramp transporter contributes to Cd and Zn hyperaccumulation in Sedum alfredii hance. Environ. Exp. Bot. 2020, 176, 104121. [Google Scholar] [CrossRef]
  111. Feng, Y.; Wu, Y.; Zhang, J.; Meng, Q.; Wang, Q.; Ma, L.; Ma, X.; Yang, X. Ectopic expression of sanramp3 from Sedum alfredii enhanced cadmium root-to-shoot transport in Brassica juncea. Ecotox. Environ. Safe. 2018, 156, 279–286. [Google Scholar] [CrossRef] [PubMed]
  112. Chen, S.; Han, X.; Fang, J.; Lu, Z.; Qiu, W.; Liu, M.; Sang, J.; Jiang, J.; Zhuo, R. Sedum alfredii sanramp6 metal transporter contributes to cadmium accumulation in transgenic Arabidopsis thaliana. Sci. Rep. 2017, 7, 13318. [Google Scholar] [CrossRef]
  113. Zhu, Y.; Qiu, W.; Li, Y.; Tan, J.; Han, X.; Wu, L.; Jiang, Y.; Deng, Z.; Wu, C.; Zhuo, R. Quantitative proteome analysis reveals changes of membrane transport proteins in sedum plumbizincicola under cadmium stress. Chemosphere 2022, 287, 132302. [Google Scholar] [CrossRef] [PubMed]
  114. Yu, W.; Deng, S.; Chen, X.; Cheng, Y.; Li, Z.; Wu, J.; Zhu, D.; Zhou, J.; Cao, Y.; Fayyaz, P. Pcnramp1 enhances cadmium uptake and accumulation in Populus× canescens. Int. J. Mol. Sci. 2022, 23, 7593. [Google Scholar] [CrossRef]
  115. Fan, W.; Liu, C.; Cao, B.; Qin, M.; Long, D.; Xiang, Z.; Zhao, A. Genome-wide identification and characterization of four gene families putatively involved in cadmium uptake, translocation and sequestration in mulberry. Front. Plant Sci. 2018, 9, 879. [Google Scholar] [CrossRef]
  116. Ma, X.; Yang, H.; Bu, Y.; Zhang, Y.; Sun, N.; Wu, X.; Jing, Y. Genome-wide identification of the nramp gene family in populus trichocarpa and their function as heavy metal transporters. Ecotox. Environ. Safe. 2023, 261, 115110. [Google Scholar] [CrossRef]
  117. Zhang, W.; Yue, S.; Song, J.; Xun, M.; Han, M.; Yang, H. Mhnramp1 from Malus hupehensis exacerbates cell death by accelerating cd uptake in tobacco and apple calli. Front. Plant Sci. 2020, 11, 957. [Google Scholar] [CrossRef]
  118. Xiao, H.; Yin, L.; Xu, X.; Li, T.; Han, Z. The iron-regulated transporter; mbnramp1, isolated from malus baccata is involved in fe, mn and cd trafficking. Ann. Bot. 2008, 102, 881–889. [Google Scholar] [CrossRef] [PubMed]
  119. Milner, M.J.; Ueno, N.M.; Yamaji, N.; Yokosho, K.; Craft, E.; Fei, Z.; Ebbs, S.; Zambrano, M.C.; Ma, J.F.; Kochian, L.V. Root and shoot transcriptome analysis of two ecotypes of Noccaea caerulescens uncovers the role of ncnramp1 in cd hyperaccumulation. Plant J. 2014, 78, 398–410. [Google Scholar] [CrossRef] [PubMed]
  120. Wei, W.; Chai, T.; Zhang, Y.; Han, L.; Xu, J.; Guan, Z. The Thlaspi caerulescens nramp homologue tcnramp3 is capable of divalent cation transport. Mol. Biotechnol. 2009, 41, 15–21. [Google Scholar] [CrossRef] [PubMed]
  121. Oomen, R.J.; Wu, J.; Lelièvre, F.; Blanchet, S.; Richaud, P.; Brygoo, H.B.; Aarts, M.G.; Thomine, S. Functional characterization of nramp3 and nramp4 from the metal hyperaccumulator Thlaspi caerulescens. New Phytol. 2009, 181, 637–650. [Google Scholar] [CrossRef] [PubMed]
  122. Yue, X.; Song, J.; Fang, B.; Wang, L.; Zou, J.; Su, N.; Cui, J. Bcnramp1 promotes the absorption of cadmium and manganese in arabidopsis. Chemosphere 2021, 283, 131113. [Google Scholar] [CrossRef] [PubMed]
  123. 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 nramp 1 mediated by mir167 in Brassica napus. Biometals 2017, 30, 917–931. [Google Scholar] [CrossRef]
  124. Peng, F.; Wang, C.; Zhu, J.; Zeng, J.; Kang, H.; Fan, X.; Sha, L.; Zhang, H.; Zhou, Y.; Wang, Y. Expression of tpnramp5, a metal transporter from polish wheat (Triticum polonicum L.), Enhances the accumulation of cd, co and mn in transgenic Arabidopsis plants. Planta 2018, 247, 1395–1406. [Google Scholar] [CrossRef]
  125. Wu, D.; Yamaji, N.; Yamane, M.; Kashino-Fujii, M.; Sato, K.; Ma, J.F. The hvnramp5 transporter mediates uptake of cadmium and manganese, but not iron. Plant Physiol. 2016, 172, 1899–1910. [Google Scholar] [CrossRef]
  126. Chen, Y.; Li, G.; Yang, J.; Zhao, X.; Sun, Z.; Hou, H. Role of nramp transporter genes of Spirodela polyrhiza in cadmium accumulation. Ecotox. Environ. Safe. 2021, 227, 112907. [Google Scholar] [CrossRef]
  127. Chen, Y.; Zhao, X.; Li, G.; Kumar, S.; Sun, Z.; Li, Y.; Guo, W.; Yang, J.; Hou, H. Genome-wide identification of the nramp gene family in Spirodela polyrhiza and expression analysis under cadmium stress. Int. J. Mol. Sci. 2021, 22, 6414. [Google Scholar] [CrossRef]
  128. Qian, M.; Li, X.; Tang, L.; Peng, Y.; Huang, X.; Wu, T.; Liu, Y.; Liu, X.; Xia, Y.; Peng, K.; et al. Vrnramp5 is responsible for cadmium and manganese uptake in vigna radiata roots. Environ. Exp. Bot. 2022, 199, 104867. [Google Scholar] [CrossRef]
  129. Nakanishi, H.; Ogawa, I.; Ishimaru, Y.; Mori, S.; Nishizawa, N.K. Iron deficiency enhances cadmium uptake and translocation mediated by the Fe2+ transporters osirt1 and osirt2 in rice. Soil Sci. Plant Nutr. 2006, 52, 464–469. [Google Scholar] [CrossRef]
  130. Chang, J.; Huang, S.; Wiseno, I.; Sui, F.; Feng, F.; Zheng, L.; Ma, J.F.; Zhao, F. Dissecting the promotional effect of zinc on cadmium translocation from roots to shoots in rice. J. Exp. Bot. 2023, 74, 6790–6803. [Google Scholar] [CrossRef] [PubMed]
  131. Sasaki, A.; Yamaji, N.; Ueno, N.M.; Kashino, M.; Ma, J.F. A node-localized transporter oszip3 is responsible for the preferential distribution of zn to developing tissues in rice. Plant J. 2015, 84, 374–384. [Google Scholar] [CrossRef] [PubMed]
  132. Zhang, C.; Tong, C.; Cao, L.; Zheng, P.; Tang, X.; Wang, L.; Miao, M.; Liu, Y.; Cao, S. Regulatory module wrky33-atl31-irt1 mediates cadmium tolerance in arabidopsis. Plant Cell Environ. 2023, 46, 1653–1670. [Google Scholar] [CrossRef] [PubMed]
  133. Bovet, L.; Rossi, L.; Moulin, N.L. Cadmium partitioning and gene expression studies in Nicotiana tabacum and Nicotiana rustica. Physiol. Plant. 2006, 128, 466–475. [Google Scholar] [CrossRef]
  134. Wang, M.; Duan, S.; Zhou, Z.; Chen, S.; Wang, D. Foliar spraying of melatonin confers cadmium tolerance in Nicotiana tabacum L. Ecotox. Environ. Safe. 2019, 170, 68–76. [Google Scholar] [CrossRef] [PubMed]
  135. Palusińska, M.; Barabasz, A.; Kozak, K.; Papierniak, A.; Maślińska, K.; Antosiewicz, D.M. Zn/Cd status-dependent accumulation of Zn and Cd in root parts in tobacco is accompanied by specific expression of zip genes. BMC Plant Biol. 2020, 20, 37. [Google Scholar] [CrossRef] [PubMed]
  136. Barabasz, A.; Palusińska, M.; Papierniak, A.; Kendziorek, M.; Kozak, K.; Williams, L.E.; Antosiewicz, D.M. Functional analysis of ntzip4b and Zn status-dependent expression pattern of tobacco zip genes. Front. Plant Sci. 2019, 9, 1984. [Google Scholar] [CrossRef]
  137. Spielmann, J.; Ahmadi, H.; Scheepers, M.; Weber, M.; Nitsche, S.; Carnol, M.; Bosman, B.; Kroymann, J.; Motte, P.; Clemens, S. The two copies of the zinc and cadmium zip6 transporter of Arabidopsis halleri have distinct effects on cadmium tolerance. Plant Cell Environ. 2020, 43, 2143–2157. [Google Scholar] [CrossRef]
  138. Pence, N.S.; Larsen, P.B.; Ebbs, S.D.; Letham, D.L.; Lasat, M.M.; Garvin, D.F.; Eide, D.; Kochian, L.V. The molecular physiology of heavy metal transport in the Zn/Cd hyperaccumulator Thlaspi caerulescens. Proc. Natl. Acad. Sci. USA 2000, 97, 4956–4960. [Google Scholar] [CrossRef] [PubMed]
  139. Wu, J.; Zhao, F.; Ghandilyan, A.; Logoteta, B.; Guzman, M.O.; Schat, H.; Wang, X.; Aarts, M.G. Identification and functional analysis of two zip metal transporters of the hyperaccumulator Thlaspi caerulescens. Plant Soil 2009, 325, 79–95. [Google Scholar] [CrossRef]
  140. Yang, Q.; Ma, X.; Luo, S.; Gao, J.; Yang, X.; Feng, Y. Sazip4, an uptake transporter of Zn/Cd hyperaccumulator Sedum alfredii hance. Environ. Exp. Bot. 2018, 155, 107–117. [Google Scholar] [CrossRef]
  141. Mizuno, T.; Usui, K.; Horie, K.; Nosaka, S.; Mizuno, N.; Obata, H. Cloning of three zip/nramp transporter genes from a ni hyperaccumulator plant thlaspi japonicum and their Ni2+-transport abilities. Plant Physiol. Biochem. 2005, 43, 793–801. [Google Scholar] [CrossRef] [PubMed]
  142. Nishida, S.; Morinaga, Y.; Obata, H.; Mizuno, T. Identification of the n-terminal region of tjznt2, a zrt/irt-like protein family metal transporter, as a novel functional region involved in metal ion selectivity. FEBS J. 2011, 278, 851–858. [Google Scholar] [CrossRef] [PubMed]
  143. Wu, X.; Zhu, Z.B.; Chen, J.H.; Huang, Y.F.; Liu, Z.L.; Zou, J.W.; Chen, Y.H.; Su, N.N.; Cui, J. Transcriptome analysis revealed pivotal transporters involved in the reduction of cadmium accumulation in pak choi (Brassica chinensis L.) by exogenous hydrogen-rich water. Chemosphere 2019, 216, 684–697. [Google Scholar] [PubMed]
  144. Zhang, L.; Song, L.; Dai, M.; Liu, J.; Li, J.; Xu, C.; Guo, Z.; Song, S.; Liu, J.; Zhu, X.; et al. Inventory of cadmium-transporter genes in the root of mangrove plant avicennia marina under cadmium stress. J. Hazard. Mater. 2023, 459, 132321. [Google Scholar] [CrossRef] [PubMed]
  145. Jiang, Y.; Chen, X.; Chai, S.; Sheng, H.; Sha, L.; Fan, X.; Zeng, J.; Kang, H.; Zhang, H.; Xiao, X.; et al. Tpirt1 from polish wheat (Triticum polonicum L.) Enhances the accumulation of Fe, Mn, Co, and Cd in arabidopsis. Plant Sci. 2021, 312, 111058. [Google Scholar] [CrossRef] [PubMed]
  146. Courbot, M.; Willems, G.; Motte, P.; Arvidsson, S.; Roosens, N.; Saumitou-Laprade, P.; Verbruggen, N. A major quantitative trait locus for cadmium tolerance in Arabidopsis halleri colocalizes with hma4, a gene encoding a heavy metal atpase. Plant Physiol. 2007, 144, 1052–1065. [Google Scholar] [CrossRef]
  147. Zhang, J.; Zhang, M.; Shohag, M.J.I.; Tian, S.; Song, H.; Feng, Y.; Yang, X. Enhanced expression of sahma3 plays critical roles in Cd hyperaccumulation and hypertolerance in cd hyperaccumulator Sedum alfredii hance. Planta 2016, 243, 577–589. [Google Scholar] [CrossRef]
  148. Yang, Z.; Wu, H.; Yang, H.; Chen, W.; Liu, J.; Yang, F.; Tai, L.; Li, B.; Yuan, B.; Liu, W.; et al. Overexpression of sedum sphma2, sphma3 and spnramp6 in Brassica napus increases multiple heavy metals accumulation for phytoextraction. J. Hazard. Mater. 2023, 449, 130970. [Google Scholar] [CrossRef] [PubMed]
  149. Bernard, C.; Roosens, N.; Czernic, P.; Lebrun, M.; Verbruggen, N. A novel cpx-atpase from the cadmium hyperaccumulator thlaspi caerulescens. Febs Lett. 2004, 569, 140–148. [Google Scholar] [CrossRef] [PubMed]
  150. Xu, W.; Huang, H.; Li, X.; Yang, M.; Chi, S.; Pan, Y.; Li, N.; Paterson, A.H.; Chai, Y.; Lu, K. Cahma1 promotes cd accumulation in pepper fruit. J. Hazard. Mater. 2023, 460, 132480. [Google Scholar] [CrossRef] [PubMed]
  151. Wang, Y.; Wang, C.; Liu, Y.; Yu, K.; Zhou, Y. Gmhma3 sequesters Cd to the root endoplasmic reticulum to limit translocation to the stems in soybean. Plant Sci. 2018, 270, 23–29. [Google Scholar] [CrossRef] [PubMed]
  152. Qiao, K.; Gong, L.; Tian, Y.; Wang, H.; Chai, T. The metal-binding domain of wheat heavy metal atpase 2 (tahma2) is involved in zinc/cadmium tolerance and translocation in arabidopsis. Plant Cell Rep. 2018, 37, 1343–1352. [Google Scholar] [CrossRef] [PubMed]
  153. Feng, S.; Tan, J.; Zhang, Y.; Liang, S.; Xiang, S.; Wang, H.; Chai, T. Isolation and characterization of a novel cadmium-regulated yellow stripe-like transporter (snysl3) in Solanum nigrum. Plant Cell Rep. 2017, 36, 281–296. [Google Scholar] [CrossRef] [PubMed]
  154. Wang, J.; Li, Y.; Zhang, Y.; Chai, T. Molecular cloning and characterization of a Brassica juncea yellow stripe-like gene, bjysl7, whose overexpression increases heavy metal tolerance of tobacco. Plant Cell Rep. 2013, 32, 651–662. [Google Scholar] [CrossRef] [PubMed]
  155. Schaaf, G.; Ludewig, U.; Erenoglu, B.E.; Mori, S.; Kitahara, T.; Von Wirén, N. Zmys1 functions as a proton-coupled symporter for phytosiderophore-and nicotianamine-chelated metals. J. Biol. Chem. 2004, 279, 9091–9096. [Google Scholar] [CrossRef] [PubMed]
  156. Chen, S.; Liu, Y.; Deng, Y.; Liu, Y.; Dong, M.; Tian, Y.; Sun, H.; Li, Y. Cloning and functional analysis of the vccxip4 and vcysl6 genes as cd-regulating genes in blueberry. Gene 2019, 686, 104–117. [Google Scholar] [CrossRef]
  157. Fu, S.; Lu, Y.; Zhang, X.; Yang, G.; Chao, D.; Wang, Z.; Shi, M.; Chen, J.; Chao, D.; Li, R.; et al. The abc transporter abcg36 is required for cadmium tolerance in rice. J. Exp. Bot. 2019, 70, 5909–5918. [Google Scholar] [CrossRef]
  158. Oda, K.; Otani, M.; Uraguchi, S.; Akihiro, T.; Fujiwara, T. Rice abcg43 is cd inducible and confers cd tolerance on yeast. Biosci. Biotechnol. Biochem. 2011, 75, 1211–1213. [Google Scholar] [CrossRef] [PubMed]
  159. Li, H.; Li, C.; Sun, D.; Yang, Z.M. Ospdr20 is an abcg metal transporter regulating cadmium accumulation in rice. J. Environ. Sci. 2024, 136, 21–34. [Google Scholar] [CrossRef] [PubMed]
  160. Kim, D.Y.; Bovet, L.; Maeshima, M.; Martinoia, E.; Lee, Y. The abc transporter atpdr8 is a cadmium extrusion pump conferring heavy metal resistance. Plant J. 2007, 50, 207–218. [Google Scholar] [CrossRef] [PubMed]
  161. Kim, D.; Bovet, L.; Kushnir, S.; Noh, E.W.; Martinoia, E.; Lee, Y. Atatm3 is involved in heavy metal resistance in arabidopsis. Plant Physiol. 2006, 140, 922–932. [Google Scholar] [CrossRef] [PubMed]
  162. Bhuiyan, M.S.U.; Min, S.R.; Jeong, W.J.; Sultana, S.; Choi, K.S.; Lee, Y.; Liu, J.R. Overexpression of atatm3 in Brassica juncea confers enhanced heavy metal tolerance and accumulation. Plant Cell Tissue Organ Cult (PCTOC) 2011, 107, 69–77. [Google Scholar]
  163. Wang, H.; Liu, Y.; Peng, Z.; Li, J.; Huang, W.; Liu, Y.; Wang, X.; Xie, S.; Sun, L.; Han, E. Ectopic expression of poplar abc transporter ptoabcg36 confers cd tolerance in Arabidopsis thaliana. Int. J. Mol. Sci. 2019, 20, 3293. [Google Scholar] [CrossRef] [PubMed]
  164. Yang, Y.H.; Wang, C.J.; Li, R.F.; Yi, Y.J.; Zeng, L.; Yang, H.; Zhang, C.F.; Song, K.Y.; Guo, S.J. Transcriptome-based identification and expression characterization of rgabcc transporters in Rehmannia glutinosa. PLoS ONE 2021, 16, e253188. [Google Scholar] [CrossRef] [PubMed]
  165. Wang, F.; Tan, H.; Han, J.; Zhang, Y.; He, X.; Ding, Y.; Chen, Z.; Zhu, C. A novel family of plac8 motif-containing/pcr genes mediates Cd tolerance and Cd accumulation in rice. Environ. Sci. Eur. 2019, 31, 82. [Google Scholar] [CrossRef]
  166. Ran, C.; Zhang, Y.; Chang, F.; Yang, X.; Liu, Y.; Wang, Q.; Zhu, W. Genome-wide analyses of slfwl family genes and their expression profiles under cold, heat, salt and drought stress in tomato. Int. J. Mol. Sci. 2023, 24, 11783. [Google Scholar] [CrossRef]
  167. Xiong, W.; Wang, P.; Yan, T.; Cao, B.; Xu, J.; Liu, D.; Luo, M. The rice “fruit-weight 2.2-like” gene family member osfwl4 is involved in the translocation of cadmium from roots to shoots. Planta 2018, 247, 1247–1260. [Google Scholar] [CrossRef]
  168. Lv, F.; Shan, Q.; Qiao, K.; Zhang, H.; Zhou, A. Populus euphratica plant cadmium resistance 2 mediates cd tolerance by root efflux of Cd ions in poplar. Plant Cell Rep. 2023, 42, 1777–1789. [Google Scholar] [CrossRef] [PubMed]
  169. Guan, J.; Yang, Y.; Shan, Q.; Zhang, H.; Zhou, A.; Gong, S.; Chai, T.; Qiao, K. Plant cadmium resistance 10 enhances tolerance to toxic heavy metals in poplar. Plant Physiol. Biochem. 2023, 203, 108043. [Google Scholar]
  170. Lin, J.; Gao, X.; Zhao, J.; Zhang, J.; Chen, S.; Lu, L. Plant cadmium resistance 2 (sapcr2) facilitates cadmium efflux in the roots of hyperaccumulator Sedum alfredii hance. Front. Plant Sci. 2020, 11, 568887. [Google Scholar] [CrossRef]
  171. Liu, Y.; Kong, L.; Gong, C.; Yang, G.; Xu, E.; Chen, W.; Zhang, W.; Chen, X. Identification of plant cadmium resistance gene family in brassica napus and functional analysis of bnpcr10.1 involved in cadmium and copper tolerance. Plant Physiol. Biochem. 2023, 202, 107989. [Google Scholar] [CrossRef]
  172. Hu, X.; Wang, S.; Zhang, H.; Zhang, H.; Feng, S.; Qiao, K.; Lv, F.; Gong, S.; Zhou, A. Plant cadmium resistance 6 from Salix linearistipularis (slpcr6) affects cadmium and copper uptake in roots of transgenic populus. Ecotox. Environ. Safe. 2022, 245, 114116. [Google Scholar] [CrossRef] [PubMed]
  173. Wang, D.; Zhang, H.; Hu, X.; Zhang, H.; Feng, S.; Zhou, A. Cell number regulator 8 from salix linearistipularis enhances cadmium tolerance in poplar by reducing cadmium uptake and accumulation. Plant Physiol. Biochem. 2024, 206, 108216. [Google Scholar]
  174. 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, 870. [Google Scholar]
  175. Qiao, K.; Tian, Y.; Hu, Z.; Chai, T. Wheat cell number regulator cnr10 enhances the tolerance, translocation, and accumulation of heavy metals in plants. Environ. Sci. Technol. 2019, 53, 860–867. [Google Scholar] [CrossRef]
  176. Cheng, Y.; Chen, X.; Liu, W.; Yang, L.; Wu, J.; Wang, Y.; Yu, W.; Zhou, J.; Fayyaz, P.; Luo, Z.; et al. Homolog of human placenta-specific gene 8, pcplac8-10, enhances cadmium uptake by Populus roots. J. Hazard. Mater. 2023, 460, 132349. [Google Scholar] [CrossRef]
  177. Das, N.; Bhattacharya, S.; Maiti, M.K. Enhanced cadmium accumulation and tolerance in transgenic tobacco overexpressing rice metal tolerance protein gene osmtp1 is promising for phytoremediation. Plant Physiol. Biochem. 2016, 105, 297–309. [Google Scholar]
  178. Yuan, L.; Yang, S.; Liu, B.; Zhang, M.; Wu, K. Molecular characterization of a rice metal tolerance protein, osmtp1. Plant Cell Rep. 2012, 31, 67–79. [Google Scholar] [CrossRef] [PubMed]
  179. Lan, H.; Wang, Z.; Wang, Q.; Wang, M.; Bao, Y.; Huang, J.; Zhang, H. Characterization of a vacuolar zinc transporter ozt1 in rice (Oryza sativa L.). Mol. Biol. Rep. 2013, 40, 1201–1210. [Google Scholar] [CrossRef] [PubMed]
  180. Li, J.; Abbas, M.; Desoky, E.M.; Zafar, S.; Soaud, S.A.; Hussain, S.S.; Abbas, S.; Hussain, A.; Ihtisham, M.; Ragauskas, A.J.; et al. Analysis of metal tolerance protein (mtp) family in sunflower (Helianthus annus L.) and role of hamtp10 as cadmium antiporter under moringa seed extract. Ind. Crop. Prod. 2023, 202, 117023. [Google Scholar] [CrossRef]
  181. Luo, J.S.; Yang, Y.; Gu, T.; Wu, Z.; Zhang, Z. The arabidopsis defensin gene atpdf2. 5 mediates cadmium tolerance and accumulation. Plant Cell Environ. 2019, 42, 2681–2695. [Google Scholar] [CrossRef] [PubMed]
  182. Zou, W.; Chen, J.; Meng, L.; Chen, D.; He, H.; Ye, G. The rice cation/H+ exchanger family involved in cd tolerance and transport. Int. J. Mol. Sci. 2021, 22, 8186. [Google Scholar] [CrossRef] [PubMed]
  183. Hao, X.; Zeng, M.; Wang, J.; Zeng, Z.; Dai, J.; Xie, Z.; Yang, Y.; Tian, L.; Chen, L.; Li, D. A node-expressed transporter osccx2 is involved in grain cadmium accumulation of rice. Front. Plant Sci. 2018, 9, 476. [Google Scholar] [CrossRef] [PubMed]
  184. Guo, J.; Zhang, X.; Ye, D.; Huang, H.; Wang, Y.; Zheng, Z.; Li, T.; Yu, H. Crucial roles of cadmium retention in nodeⅱ for restraining cadmium transport from straw to ear at reproductive period in a grain low-cadmium rice line (Oryza sativa L.). Ecotox. Environ. Safe. 2020, 205, 111323. [Google Scholar] [CrossRef] [PubMed]
  185. Kuramata, M.; Masuya, S.; Takahashi, Y.; Kitagawa, E.; Inoue, C.; Ishikawa, S.; Youssefian, S.; Kusano, T. Novel cysteine-rich peptides from Digitaria ciliaris and Oryza sativa enhance tolerance to cadmium by limiting its cellular accumulation. Plant Cell Physiol. 2009, 50, 106–117. [Google Scholar] [CrossRef] [PubMed]
  186. Uraguchi, S.; Kamiya, T.; Clemens, S.; Fujiwara, T. Characterization of oslct1, a cadmium transporter from indica rice (Oryza sativa). Physiol. Plant. 2014, 151, 339–347. [Google Scholar] [CrossRef]
  187. Tang, L.; Dong, J.; Tan, L.; Ji, Z.; Li, Y.; Sun, Y.; Chen, C.; Lv, Q.; Mao, B.; Hu, Y.; et al. Overexpression of oslct2, a low-affinity cation transporter gene, reduces cadmium accumulation in shoots and grains of rice. Rice 2021, 14, 89. [Google Scholar] [CrossRef]
  188. Chen, X.; Ouyang, Y.; Fan, Y.; Qiu, B.; Zhang, G.; Zeng, F. The pathway of transmembrane cadmium influx via calcium-permeable channels and its spatial characteristics along rice root. J. Exp. Bot. 2018, 69, 5279–5291. [Google Scholar] [CrossRef] [PubMed]
  189. Wang, H.; Yu, J.; Zhu, B.; Gu, L.; Wang, H.; Du, X.; Zeng, T.; Tang, H. The sbbhlh041–sbexpa11 module enhances cadmium accumulation and rescues biomass by increasing photosynthetic efficiency in sorghum. Int. J. Mol. Sci. 2023, 24, 13061. [Google Scholar] [CrossRef] [PubMed]
  190. Li, F.; Deng, Y.; Liu, Y.; Mai, C.; Xu, Y.; Wu, J.; Zheng, X.; Liang, C.; Wang, J. Arabidopsis transcription factor wrky45 confers cadmium tolerance via activating pcs1 and pcs2 expression. J. Hazard. Mater. 2023, 460, 132496. [Google Scholar] [CrossRef] [PubMed]
  191. Zhang, P.; Wang, R.; Ju, Q.; Li, W.; Tran, L.P.; Xu, J. The r2r3-myb transcription factor myb49 regulates cadmium accumulation. Plant Physiol. 2019, 180, 529–542. [Google Scholar] [CrossRef] [PubMed]
  192. Meng, Y.T.; Zhang, X.L.; Wu, Q.; Shen, R.F.; Zhu, X.F. Transcription factor anac004 enhances Cd tolerance in arabidopsis thaliana by regulating cell wall fixation, translocation and vacuolar detoxification of Cd, aba accumulation and antioxidant capacity. J. Hazard. Mater. 2022, 436, 129121. [Google Scholar] [CrossRef]
  193. Han, Y.; Fan, T.; Zhu, X.; Wu, X.; Ouyang, J.; Jiang, L.; Cao, S. Wrky12 represses gsh1 expression to negatively regulate cadmium tolerance in arabidopsis. Plant Mol. Biol. 2019, 99, 149–159. [Google Scholar]
  194. Sheng, Y.; Yan, X.; Huang, Y.; Han, Y.; Zhang, C.; Ren, Y.; Fan, T.; Xiao, F.; Liu, Y.; Cao, S. The wrky transcription factor, wrky13, activates pdr8 expression to positively regulate cadmium tolerance in arabidopsis. Plant Cell Environ. 2019, 42, 891–903. [Google Scholar] [CrossRef] [PubMed]
  195. Zhang, Q.; Cai, W.; Ji, T.; Ye, L.; Lu, Y.; Yuan, T. Wrky13 enhances cadmium tolerance by promoting d-cysteine desulfhydrase and hydrogen sulfide production1. Plant Physiol. 2020, 183, 345–357. [Google Scholar] [CrossRef] [PubMed]
  196. Liu, Z.; Fang, H.; Pei, Y.; Jin, Z.; Zhang, L.; Liu, D. Wrky transcription factors down-regulate the expression of h2s-generating genes, lcd and des in arabidopsis thaliana. Sci. Bull. 2015, 60, 995–1001. [Google Scholar] [CrossRef]
  197. El Rasafi, T.; Oukarroum, A.; Haddioui, A.; Song, H.; Kwon, E.E.; Bolan, N.; Tack, F.M.; Sebastian, A.; Prasad, M.; Rinklebe, J. Cadmium stress in plants: A critical review of the effects, mechanisms, and tolerance strategies. Crit. Rev. Environ. Sci. Technol. 2022, 52, 675–726. [Google Scholar] [CrossRef]
  198. Li, Y.; Rahman, S.U.; Qiu, Z.; Shahzad, S.M.; Nawaz, M.F.; Huang, J.; Naveed, S.; Li, L.; Wang, X.; Cheng, H. Toxic effects of cadmium on the physiological and biochemical attributes of plants, and phytoremediation strategies: A review. Environ. Pollut. 2023, 325, 121433. [Google Scholar] [CrossRef] [PubMed]
  199. Tikhonov, A.N. pH-dependent regulation of electron transport and ATP synthesis in chloroplasts. Photosynth. Res. 2013, 116, 511–534. [Google Scholar] [CrossRef] [PubMed]
  200. Naciri, R.; Lahrir, M.; Benadis, C.; Chtouki, M.; Oukarroum, A. Interactive effect of potassium and cadmium on growth, root morphology and chlorophyll a fluorescence in tomato plant. Sci. Rep. 2021, 11, 5384. [Google Scholar] [CrossRef] [PubMed]
  201. Faseela, P.; Sinisha, A.K.; Brestič, M.; Puthur, J.T. Chlorophyll a fluorescence parameters as indicators of a particular abiotic stress in rice. Photosynthetica 2020, 58, 293–300. [Google Scholar] [CrossRef]
  202. Zulfiqar, U.; Jiang, W.; Xiukang, W.; Hussain, S.; Ahmad, M.; Maqsood, M.F.; Ali, N.; Ishfaq, M.; Kaleem, M.; Haider, F.U. Cadmium phytotoxicity, tolerance, and advanced remediation approaches in agricultural soils: A comprehensive review. Front. Plant Sci. 2022, 13, 773815. [Google Scholar] [CrossRef] [PubMed]
  203. Carvalho, M.E.A.; Castro, P.R.C.; Azevedo, R.A. Hormesis in plants under cd exposure: From toxic to beneficial element? J. Hazard. Mater. 2020, 384, 121434. [Google Scholar] [CrossRef] [PubMed]
  204. Byrne, C.; Divekar, S.D.; Storchan, G.B.; Parodi, D.A.; Martin, M.B. Cadmium—A metallohormone? Toxicol. Appl. Pharmacol. 2009, 238, 266–271. [Google Scholar] [CrossRef] [PubMed]
  205. Sparks, D.L. Advances in Agronomy; Academic Press: Cambridge, MA, USA, 2012. [Google Scholar]
  206. Cornu, J.Y.; Bussière, S.; Coriou, C.; Robert, T.; Maucourt, M.; Deborde, C.; Moing, A.; Nguyen, C. Changes in plant growth, Cd partitioning and xylem sap composition in two sunflower cultivars exposed to low cd concentrations in hydroponics. Ecotox. Environ. Safe. 2020, 205, 111145. [Google Scholar] [CrossRef] [PubMed]
  207. Uddin, M.M.; Chen, Z.; Xu, F.; Huang, L. Physiological and cellular ultrastructural responses of Sesuvium portulacastrum under cd stress grown hydroponically. Plants 2023, 12, 3381. [Google Scholar] [CrossRef]
  208. Dou, X.; Dai, H.; Skuza, L.; Wei, S. Cadmium removal potential of hyperaccumulator Solanum nigrum L. Under two planting modes in three years continuous phytoremediation. Environ. Pollut. 2022, 307, 119493. [Google Scholar] [CrossRef]
  209. Gallego, S.M.; Pena, L.B.; Barcia, R.A.; Azpilicueta, C.E.; Iannone, M.F.; Rosales, E.P.; Zawoznik, M.S.; Groppa, M.D.; Benavides, M.P. Unravelling cadmium toxicity and tolerance in plants: Insight into regulatory mechanisms. Environ. Exp. Bot. 2012, 83, 33–46. [Google Scholar] [CrossRef]
  210. Kim, S.; Lim, H.; Lee, I. Enhanced heavy metal phytoextraction by echinochloa crus-galli using root exudates. J. Biosci. Bioeng. 2010, 109, 47–50. [Google Scholar] [CrossRef] [PubMed]
  211. Shaari, N.E.M.; Tajudin, M.T.F.M.; Khandaker, M.M.; Majrashi, A.; Alenazi, M.M.; Abdullahi, U.A.; Mohd, K.S. Cadmium toxicity symptoms and uptake mechanism in plants: A review. Braz. J. Biol. 2022, 84, e252143. [Google Scholar] [CrossRef] [PubMed]
  212. Han, G.H.; Huang, R.N.; Hong, L.H.; Xu, J.X.; Hong, Y.G.; Wu, Y.H.; Chen, W.W. The transcription factor nac102 confers cadmium tolerance by regulating wakl11 expression and cell wall pectin metabolism in arabidopsis. J. Integr. Plant Biol. 2023, 65, 2262–2278. [Google Scholar] [CrossRef]
  213. Wang, L.; Wu, K.; Liu, Z.; Li, Z.; Shen, J.; Wu, Z.; Liu, H.; You, L.; Yang, G.; Rensing, C. Selenite reduced uptake/translocation of cadmium via regulation of assembles and interactions of pectins, hemicelluloses, lignins, callose and casparian strips in rice roots. J. Hazard. Mater. 2023, 448, 130812. [Google Scholar] [CrossRef]
  214. Keunen, E.; Peshev, D.; Vangronsveld, J.; Van Den Ende, W.; Cuypers, A. Plant sugars are crucial players in the oxidative challenge during abiotic stress: Extending the traditional concept. Plant Cell Environ. 2013, 36, 1242–1255. [Google Scholar] [CrossRef]
  215. Slama, I.; Abdelly, C.; Bouchereau, A.; Flowers, T.; Savouré, A. Diversity, distribution and roles of osmoprotective compounds accumulated in halophytes under abiotic stress. Ann. Bot. 2015, 115, 433–447. [Google Scholar] [CrossRef] [PubMed]
  216. Rajasheker, G.; Jawahar, G.; Jalaja, N.; Kumar, S.A.; Kumari, P.H.; Punita, D.L.; Karumanchi, A.R.; Reddy, P.S.; Rathnagiri, P.; Sreenivasulu, N.; et al. Chapter 27-role and regulation of osmolytes and aba interaction in salt and drought stress tolerance. In Plant Signaling Molecules; Khan, M.I.R., Reddy, P.S., Ferrante, A., Khan, N.A., Eds.; Woodhead Publishing: Cambridge, UK, 2019; pp. 417–436. [Google Scholar]
  217. Liu, Z.; Ding, Y.; Wang, F.; Ye, Y.; Zhu, C. Role of salicylic acid in resistance to cadmium stress in plants. Plant Cell Rep. 2016, 35, 719–731. [Google Scholar] [CrossRef]
  218. Emamverdian, A.; Ding, Y.; Mokhberdoran, F. The role of salicylic acid and gibberellin signaling in plant responses to abiotic stress with an emphasis on heavy metals. Plant Signal. Behav. 2020, 15, 1777372. [Google Scholar] [CrossRef]
  219. Whiting, S.N.; Reeves, R.D.; Richards, D.; Johnson, M.S.; Cooke, J.A.; Malaisse, F.; Paton, A.; Smith, J.; Angle, J.S.; Chaney, R.L.; et al. Research priorities for conservation of metallophyte biodiversity and their potential for restoration and site remediation. Restor. Ecol. 2004, 12, 106–116. [Google Scholar] [CrossRef]
  220. Bothe, H.; Słomka, A. Divergent biology of facultative heavy metal plants. J. Plant Physiol. 2017, 219, 45–61. [Google Scholar] [CrossRef] [PubMed]
  221. Greipsson, S. Phytoremediation. Nat. Educ. Knowl. 2011, 3, 7. [Google Scholar]
  222. Sarma, H. Metal hyperaccumulation in plants: A review focusing on phytoremediation technology. J. Environ. Sci. Technol. 2011, 4, 118–138. [Google Scholar] [CrossRef]
  223. Ali, H.; Khan, E.; Sajad, M.A. Phytoremediation of heavy metals—Concepts and applications. Chemosphere 2013, 91, 869–881. [Google Scholar] [CrossRef] [PubMed]
  224. Maestri, E.; Marmiroli, M.; Visioli, G.; Marmiroli, N. Metal tolerance and hyperaccumulation: Costs and trade-offs between traits and environment. Environ. Exp. Bot. 2010, 68, 1–13. [Google Scholar] [CrossRef]
  225. Huang, R.; Dong, M.; Mao, P.; Zhuang, P.; Paz-Ferreiro, J.; Li, Y.; Li, Y.; Hu, X.; Netherway, P.; Li, Z. Evaluation of phytoremediation potential of five Cd (hyper)accumulators in two Cd contaminated soils. Sci. Total Environ. 2020, 721, 137581. [Google Scholar] [CrossRef]
  226. Verbruggen, N.; Juraniec, M.; Baliardini, C.; Meyer, C. Tolerance to cadmium in plants: The special case of hyperaccumulators. Biometals 2013, 26, 633–638. [Google Scholar] [CrossRef]
  227. Mertens, J.; Vervaeke, P.; Meers, E.; Tack, F. Seasonal changes of metals in willow (salix sp.) Stands for phytoremediation on dredged sediment. Environ. Sci. Technol. 2006, 40, 1962–1968. [Google Scholar] [CrossRef]
  228. Li, J.T.; Liao, B.; Lan, C.Y.; Ye, Z.H.; Baker, A.; Shu, W.S. Cadmium tolerance and accumulation in cultivars of a high-biomass tropical tree (Averrhoa carambola) and its potential for phytoextraction. J. Environ. Qual. 2010, 39, 1262–1268. [Google Scholar] [CrossRef] [PubMed]
  229. Mozafariyan, M.; Shekari, L.; Hawrylak-Nowak, B.; Kamelmanesh, M.M. Protective role of selenium on pepper exposed to cadmium stress during reproductive stage. Biol. Trace Elem. Res. 2014, 160, 97–107. [Google Scholar] [CrossRef]
  230. Shen, Y.; Li, J.; Zhang, S.; Jiang, X.; Liang, J.; Li, T.; Guo, R.; Guan, W.; Yang, L. Cd stress alleviation in mung-bean seedlings with biogenic hydroxyapatite nanoparticles as ecofriendly remediation agents. Environ. Sci. Nano 2022, 9, 3844–3858. [Google Scholar] [CrossRef]
  231. Asgher, M.; Khan, M.I.R.; Anjum, N.A.; Khan, N.A. Minimising toxicity of cadmium in plants-role of plant growth regulators. Protoplasma 2015, 252, 399–413. [Google Scholar] [CrossRef] [PubMed]
  232. Mir, I.R.; Gautam, H.; Anjum, N.A.; Masood, A.; Khan, N.A. Calcium and nitric oxide signaling in plant cadmium stress tolerance: A cross talk. S. Afr. J. Bot. 2022, 150, 387–403. [Google Scholar] [CrossRef]
  233. Huang, D.; Gong, X.; Liu, Y.; Zeng, G.; Lai, C.; Bashir, H.; Zhou, L.; Wang, D.; Xu, P.; Cheng, M.; et al. Effects of calcium at toxic concentrations of cadmium in plants. Planta 2017, 245, 863–873. [Google Scholar] [CrossRef] [PubMed]
  234. Kinraide, T.B. Three mechanisms for the calcium alleviation of mineral toxicities. Plant Physiol. 1998, 118, 513–520. [Google Scholar] [CrossRef] [PubMed]
  235. Shang, X.S.; Xue, W.X.; Jiang, Y.; Zou, J. Effects of calcium on the alleviation of cadmium toxicity in Salix matsudana and its effects on other minerals. Pol. J. Environ. Stud. 2020, 29, 2001–2010. [Google Scholar] [CrossRef] [PubMed]
  236. Hakeem, K.R.; Alharby, H.F.; Pirzadah, T.B. Exogenously applied calcium regulates antioxidative system and reduces cadmium-uptake in Fagopyrum esculentum. Plant Physiol. Biochem. 2022, 180, 17–26. [Google Scholar] [CrossRef] [PubMed]
  237. Hasanuzzaman, M.; Bhuyan, M.H.M.B.; Raza, A.; Hawrylak-Nowak, B.; Matraszek-Gawron, R.; Mahmud, J.A.; Nahar, K.; Fujita, M. Selenium in plants: Boon or bane? Environ. Exp. Bot. 2020, 178, 104170. [Google Scholar] [CrossRef]
  238. Lin, L.; Zhou, W.; Dai, H.; Cao, F.; Zhang, G.; Wu, F. Selenium reduces cadmium uptake and mitigates cadmium toxicity in rice. J. Hazard. Mater. 2012, 235–236, 345–351. [Google Scholar] [CrossRef]
  239. Saidi, I.; Chtourou, Y.; Djebali, W. Selenium alleviates cadmium toxicity by preventing oxidative stress in sunflower (Helianthus annuus) seedlings. J. Plant Physiol. 2014, 171, 85–91. [Google Scholar] [CrossRef]
  240. Zembala, M.; Filek, M.; Walas, S.; Mrowiec, H.; Kornaś, A.; Miszalski, Z.; Hartikainen, H. Effect of selenium on macro-and microelement distribution and physiological parameters of rape and wheat seedlings exposed to cadmium stress. Plant Soil 2010, 329, 457–468. [Google Scholar] [CrossRef]
  241. Rady, M.M.; Hemida, K.A. Modulation of cadmium toxicity and enhancing cadmium-tolerance in wheat seedlings by exogenous application of polyamines. Ecotox. Environ. Safe. 2015, 119, 178–185. [Google Scholar] [CrossRef] [PubMed]
  242. Zhu, X.F.; Wang, Z.W.; Dong, F.; Lei, G.J.; Shi, Y.Z.; Li, G.X.; Zheng, S.J. Exogenous auxin alleviates cadmium toxicity in Arabidopsis thaliana by stimulating synthesis of hemicellulose 1 and increasing the cadmium fixation capacity of root cell walls. J. Hazard. Mater. 2013, 263, 398–403. [Google Scholar] [CrossRef] [PubMed]
  243. Duan, Z.; Chen, C.; Ni, C.; Xiong, J.; Wang, Z.; Cai, J.; Tan, W. How different is the remediation effect of biochar for cadmium contaminated soil in various cropping systems? A global meta-analysis. J. Hazard. Mater. 2023, 448, 130939. [Google Scholar] [CrossRef] [PubMed]
  244. Xue, W.; Zhang, X.; Zhang, C.; Wang, C.; Huang, Y.; Liu, Z. Mitigating the toxicity of reactive oxygen species induced by cadmium via restoring citrate valve and improving the stability of enzyme structure in rice. Chemosphere 2023, 327, 138511. [Google Scholar] [CrossRef] [PubMed]
  245. Huang, J.; Jing, H.K.; Zhang, Y.; Chen, S.Y.; Wang, H.Y.; Cao, Y.; Zhang, Z.; Lu, Y.H.; Zheng, Q.S.; Shen, R.F.; et al. Melatonin reduces cadmium accumulation via mediating the nitric oxide accumulation and increasing the cell wall fixation capacity of cadmium in rice. J. Hazard. Mater. 2023, 445, 130529. [Google Scholar] [CrossRef] [PubMed]
  246. Wang, W.; Cang, L.; Zhou, D.; Yu, Y. Exogenous amino acids increase antioxidant enzyme activities and tolerance of rice seedlings to cadmium stress. Environ. Prog. Sustain. Energy 2017, 36, 155–161. [Google Scholar] [CrossRef]
  247. Liu, J.; Yang, L.; Wen, Y.; Li, Y.; Shabala, S.; Zhang, J.; Song, W. The roles of abscisic acid and ethylene in cadmium accumulation and tolerance in plants. Plant Soil 2023, 1–12. [Google Scholar] [CrossRef]
  248. Wang, H.; Zhao, X.; Xuan, W.; Wang, P.; Zhao, F. Rice roots avoid asymmetric heavy metal and salinity stress via an rboh-ros-auxin signaling cascade. Mol. Plant. 2023, 16, 1678–1694. [Google Scholar] [CrossRef] [PubMed]
  249. Raza, A.; Habib, M.; Kakavand, S.N.; Zahid, Z.; Zahra, N.; Sharif, R.; Hasanuzzaman, M. Phytoremediation of Cadmium: Physiological, Biochemical, and Molecular Mechanisms. Biology 2020, 9, 177. [Google Scholar] [CrossRef]
  250. Luo, J.; Zhang, Z. Mechanisms of cadmium phytoremediation and detoxification in plants. Crop J. 2021, 9, 521–529. [Google Scholar] [CrossRef]
  251. Satoh-Nagasawa, N.; Mori, M.; Nakazawa, N.; Kawamoto, T.; Nagato, Y.; Sakurai, K.; Takahashi, H.; Watanabe, A.; Akagi, H. Mutations in rice (Oryza sativa) heavy metal atpase 2 (oshma2) restrict the translocation of zinc and cadmium. Plant Cell Physiol. 2012, 53, 213–224. [Google Scholar] [CrossRef] [PubMed]
  252. 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. N. Phytol. 2011, 189, 190–199. [Google Scholar] [CrossRef] [PubMed]
  253. Gravot, A.; Lieutaud, A.; Verret, F.; Auroy, P.; Vavasseur, A.; Richaud, P. Athma3, a plant p1b-atpase, functions as a cd/pb transporter in yeast. Febs Lett. 2004, 561, 22–28. [Google Scholar] [CrossRef] [PubMed]
  254. Zhao, H.; Wang, L.; Zhao, F.J.; Wu, L.; Liu, A.; Xu, W. Sphma1 is a chloroplast cadmium exporter protecting photochemical reactions in the cd hyperaccumulator Sedum plumbizincicola. Plant Cell Environ. 2019, 42, 1112–1124. [Google Scholar] [CrossRef] [PubMed]
  255. Liu, H.; Zhao, H.; Wu, L.; Liu, A.; Zhao, F.J.; Xu, W. Heavy metal atpase 3 (hma3) confers cadmium hypertolerance on the cadmium/zinc hyperaccumulator Sedum plumbizincicola. N. Phytol. 2017, 215, 687–698. [Google Scholar] [CrossRef] [PubMed]
  256. Ueno, D.; Milner, M.J.; Yamaji, N.; Yokosho, K.; Koyama, E.; Clemencia Zambrano, M.; Kaskie, M.; Ebbs, S.; Kochian, L.V.; Ma, J.F. Elevated expression of tchma3 plays a key role in the extreme cd tolerance in a cd-hyperaccumulating ecotype of Thlaspi caerulescens. Plant J. 2011, 66, 852–862. [Google Scholar] [CrossRef] [PubMed]
  257. Wang, X.; Zhi, J.; Liu, X.; Zhang, H.; Liu, H.; Xu, J. Transgenic tobacco plants expressing a p1b-atpase gene from Populus tomentosa carr. (Ptohma5) demonstrate improved cadmium transport. Int. J. Biol. Macromol. 2018, 113, 655–661. [Google Scholar] [CrossRef] [PubMed]
  258. Wang, J.; Liang, S.; Xiang, W.; Dai, H.; Duan, Y.; Kang, F.; Chai, T. A repeat region from the brassica juncea hma4 gene bjhma4r is specifically involved in cd2+ binding in the cytosol under low heavy metal concentrations. BMC Plant Biol. 2019, 19, 89. [Google Scholar] [CrossRef]
  259. Chen, H.; Zhang, C.; Guo, H.; Hu, Y.; He, Y.; Jiang, D. Overexpression of a miscanthus sacchariflorus yellow stripe-like transporter msysl1 enhances resistance of arabidopsis to cadmium by mediating metal ion reallocation. Plant Growth Regul. 2018, 85, 101–111. [Google Scholar] [CrossRef]
  260. Yang, G.; Fu, S.; Huang, J.; Li, L.; Long, Y.; Wei, Q.; Wang, Z.; Chen, Z.; Xia, J. The tonoplast-localized transporter osabcc9 is involved in cadmium tolerance and accumulation in rice. Plant Sci. 2021, 307, 110894. [Google Scholar] [CrossRef]
  261. Cai, X.; Wang, M.; Jiang, Y.; Wang, C.; Ow, D.W. Overexpression of osabcg48 lowers cadmium in rice (Oryza sativa L.). Agronomy 2021, 11, 918. [Google Scholar] [CrossRef]
  262. Park, J.; Song, W.Y.; Ko, D.; Eom, Y.; Hansen, T.H.; Schiller, M.; Lee, T.G.; Martinoia, E.; Lee, Y. The phytochelatin transporters atabcc1 and atabcc2 mediate tolerance to cadmium and mercury. Plant J. 2012, 69, 278–288. [Google Scholar] [CrossRef] [PubMed]
  263. Brunetti, P.; Zanella, L.; De Paolis, A.; Di Litta, D.; Cecchetti, V.; Falasca, G.; Barbieri, M.; Altamura, M.M.; Costantino, P.; Cardarelli, M. Cadmium-inducible expression of the abc-type transporter atabcc3 increases phytochelatin-mediated cadmium tolerance in Arabidopsis. J. Exp. Bot. 2015, 66, 3815–3829. [Google Scholar] [CrossRef] [PubMed]
  264. Gaillard, S.; Jacquet, H.; Vavasseur, A.; Leonhardt, N.; Forestier, C. Atmrp6/atabcc6, an atp-binding cassette transporter gene expressed during early steps of seedling development and up-regulated by cadmium in Arabidopsis thaliana. BMC Plant Biol. 2008, 8, 22. [Google Scholar] [CrossRef] [PubMed]
  265. Wojas, S.; Hennig, J.; Plaza, S.; Geisler, M.; Siemianowski, O.; Skłodowska, A.; Ruszczyńska, A.; Bulska, E.; Antosiewicz, D.M. Ectopic expression of arabidopsis abc transporter mrp7 modifies cadmium root-to-shoot transport and accumulation. Environ. Pollut. 2009, 157, 2781–2789. [Google Scholar] [CrossRef] [PubMed]
  266. Bhati, K.K.; Alok, A.; Kumar, A.; Kaur, J.; Tiwari, S.; Pandey, A.K. Silencing of abcc13 transporter in wheat reveals its involvement in grain development, phytic acid accumulation and lateral root formation. J. Exp. Bot. 2016, 67, 4379–4389. [Google Scholar] [CrossRef] [PubMed]
  267. Shi, M.; Wang, S.; Zhang, Y.; Wang, S.; Zhao, J.; Feng, H.; Sun, P.; Fang, C.; Xie, X. Genome-wide characterization and expression analysis of atp-binding cassette (abc) transporters in strawberry reveal the role of fvabcc11 in cadmium tolerance. Sci. Hortic. 2020, 271, 109464. [Google Scholar] [CrossRef]
  268. Gao, Q.; Liu, L.; Zhou, H.; Liu, X.; Li, W.; Min, Y.; Yan, Y.; Ji, J.; Zhang, H.; Zhao, X. Mutation in osfwl7 affects cadmium and micronutrient metal accumulation in rice. Int. J. Mol. Sci. 2021, 22, 12583. [Google Scholar] [CrossRef] [PubMed]
  269. Song, W.; Martinoia, E.; Lee, J.; Kim, D.; Kim, D.; Vogt, E.; Shim, D.; Choi, K.S.; Hwang, I.; Lee, Y. A novel family of cys-rich membrane proteins mediates cadmium resistance in Arabidopsis. Plant Physiol. 2004, 135, 1027–1039. [Google Scholar] [CrossRef]
  270. Feng, S.; Hou, K.; Zhang, H.; Chen, C.; Huang, J.; Wu, Q.; Zhang, Z.; Gao, Y.; Wu, X.; Wang, H.; et al. Investigation of the role of tmmyb16/123 and their targets (tmmtp1/11) in the tolerance of taxus media to cadmium. Tree Physiol. 2023, 43, 1009–1022. [Google Scholar] [CrossRef]
  271. Li, D.; He, G.; Tian, W.; Saleem, M.; Huang, Y.; Meng, L.; Wu, D.; He, T. Comparative and systematic omics revealed low cd accumulation of potato stmtp 9 in yeast: Suggesting a new mechanism for heavy metal detoxification. Int. J. Mol. Sci. 2021, 22, 10478. [Google Scholar] [CrossRef] [PubMed]
  272. Luo, J.; Xiao, Y.; Yao, J.; Wu, Z.; Yang, Y.; Ismail, A.M.; Zhang, Z. Overexpression of a defensin-like gene cal2 enhances cadmium accumulation in plants. Front. Plant Sci. 2020, 11, 217. [Google Scholar] [CrossRef] [PubMed]
  273. Kazachkova, Y. Seeds and heavy metal: Defensin-like protein def8 mediates cadmium accumulation in rice and phloem unloading. Plant Physiol. 2023, 191, 12–14. [Google Scholar] [CrossRef] [PubMed]
  274. Gu, T.; Qi, Z.; Chen, S.; Yan, J.; Fang, Z.; Wang, J.; Gong, J. Dual-function defensin 8 mediates phloem cadmium unloading and accumulation in rice grains. Plant Physiol. 2023, 191, 515–527. [Google Scholar] [CrossRef] [PubMed]
  275. Liu, X.; Gong, X.; Zhou, D.; Jiang, Q.; Liang, Y.; Ye, R.; Zhang, S.; Wang, Y.; Tang, X.; Li, F. Plant defensin-dissimilar thionin osthi9 alleviates cadmium toxicity in rice plants and reduces cadmium accumulation in rice grains. J. Agric. Food. Chem. 2023, 71, 8367–8380. [Google Scholar] [CrossRef] [PubMed]
  276. Luo, J.; Gu, T.; Yang, Y.; Zhang, Z. A non-secreted plant defensin atpdf2.6 conferred cadmium tolerance via its chelation in Arabidopsis. Plant Mol. Biol. 2019, 100, 561–569. [Google Scholar]
  277. Luo, J.; Zhang, Z. Proteomic changes in the xylem sap of brassica napus under cadmium stress and functional validation. BMC Plant Biol. 2019, 19, 280. [Google Scholar] [CrossRef] [PubMed]
  278. Uraguchi, S.; Tanaka, N.; Hofmann, C.; Abiko, K.; Ohkama-Ohtsu, N.; Weber, M.; Kamiya, T.; Sone, Y.; Nakamura, R.; Takanezawa, Y.; et al. Phytochelatin synthase has contrasting effects on cadmium and arsenic accumulation in rice grains. Plant Cell Physiol. 2017, 58, 1730–1742. [Google Scholar] [CrossRef]
  279. Yamazaki, S.; Ueda, Y.; Mukai, A.; Ochiai, K.; Matoh, T. Rice phytochelatin synthases os pcs 1 and os pcs 2 make different contributions to cadmium and arsenic tolerance. Plant Direct 2018, 2, e00034. [Google Scholar] [CrossRef]
  280. Park, H.C.; Hwang, J.E.; Jiang, Y.; Kim, Y.J.; Kim, S.H.; Nguyen, X.C.; Kim, C.Y.; Chung, W.S. Functional characterisation of two phytochelatin synthases in rice (Oryza sativa cv. Milyang 117) that respond to cadmium stress. Plant Biol. 2019, 21, 854–861. [Google Scholar]
  281. Brunetti, P.; Zanella, L.; Proia, A.; De Paolis, A.; Falasca, G.; Altamura, M.M.; Sanità Di Toppi, L.; Costantino, P.; Cardarelli, M. Cadmium tolerance and phytochelatin content of Arabidopsis seedlings over-expressing the phytochelatin synthase gene atpcs1. J. Exp. Bot. 2011, 62, 5509–5519. [Google Scholar] [CrossRef] [PubMed]
  282. Wojas, S.; Clemens, S.; Hennig, J.; Skłodowska, A.; Kopera, E.; Schat, H.; Bal, W.; Antosiewicz, D.M. Overexpression of phytochelatin synthase in tobacco: Distinctive effects of atpcs1 and cepcs genes on plant response to cadmium. J. Exp. Bot. 2008, 59, 2205–2219. [Google Scholar] [CrossRef] [PubMed]
  283. Wang, F.; Wang, Z.; Zhu, C. Heteroexpression of the wheat phytochelatin synthase gene (tapcs1) in rice enhances cadmium sensitivity. Acta Biochim. Biophys. Sin. 2012, 44, 886–893. [Google Scholar] [CrossRef] [PubMed]
  284. Ahmadi, H.; Corso, M.; Weber, M.; Verbruggen, N.; Clemens, S. Cax1 suppresses cd-induced generation of reactive oxygen species in Arabidopsis helleri. Plant Cell Environ. 2018, 41, 2435–2448. [Google Scholar] [CrossRef] [PubMed]
  285. Zhang, M.; Zhang, J.; Lu, L.L.; Zhu, Z.Q.; Yang, X.E. Functional analysis of cax2-like transporters isolated from two ecotypes of Sedum alfredii. Biol. Plant. 2016, 60, 37–47. [Google Scholar] [CrossRef]
  286. Liu, Y.; He, G.; He, Y.; Tang, Y.; Zhao, F.; He, T. Discovery of cadmium-tolerant biomacromolecule (stcax1/4 transportproteins) in potato and its potential regulatory relationship with wrky transcription factors. Int. J. Biol. Macromol. 2023, 228, 385–399. [Google Scholar] [CrossRef] [PubMed]
  287. Gu, C.; Liu, L.; Song, A.; Liu, Z.; Zhang, Y.; Huang, S. Iris lactea var. Chinensis (fisch.) Cysteine-rich gene llcdt1 enhances cadmium tolerance in yeast cells and Arabidopsis thaliana. Ecotoxicol. Environ. Saf. 2018, 157, 67–72. [Google Scholar] [CrossRef] [PubMed]
  288. Chen, H.; Ye, R.; Liang, Y.; Zhang, S.; Liu, X.; Sun, C.; Li, F.; Yi, J. Generation of low-cadmium rice germplasms via knockout of oslcd using crispr/cas9. J. Environ. Sci. 2023, 126, 138–152. [Google Scholar] [CrossRef]
  289. Gautam, N.; Tiwari, M.; Kidwai, M.; Dutta, P.; Chakrabarty, D. Functional characterization of rice metallothionein osmt-i-id: Insights into metal binding and heavy metal tolerance mechanisms. J. Hazard. Mater. 2023, 458, 131815. [Google Scholar] [CrossRef]
  290. Jin, S.; Xu, C.; Li, G.; Sun, D.; Li, Y.; Wang, X.; Liu, S. Functional characterization of a type 2 metallothionein gene, ssmt2, from alkaline-tolerant suaeda salsa. Sci. Rep. 2017, 7, 17914. [Google Scholar] [CrossRef]
  291. Peng, J.; Yi, H.; Gong, J. Isolation and characterization of cadmium tolerant gene spmt2 in the hyperaccumulator Sedum plumbizincicola. Sheng Wu Gong Cheng Xue Bao Chin. J. Biotechnol. 2020, 36, 541–548. [Google Scholar]
  292. Yu, C.; Sun, C.; Shen, C.; Wang, S.; Liu, F.; Liu, Y.; Chen, Y.; Li, C.; Qian, Q.; Aryal, B. The auxin transporter, os aux 1, is involved in primary root and root hair elongation and in cd stress responses in rice (Oryza sativa L.). Plant J. 2015, 83, 818–830. [Google Scholar] [CrossRef] [PubMed]
  293. Chen, J.; Huang, X.Y.; Salt, D.E.; Zhao, F.J. Mutation in oscadt1 enhances cadmium tolerance and enriches selenium in rice grain. N. Phytol. 2020, 226, 838–850. [Google Scholar] [CrossRef] [PubMed]
  294. Jin, S.; Cheng, Y.; Guan, Q.; Liu, D.; Takano, T.; Liu, S. A metallothionein-like protein of rice (rgmt) functions in E. coli and its gene expression is induced by abiotic stresses. Biotechnol. Lett. 2006, 28, 1749–1753. [Google Scholar] [CrossRef] [PubMed]
  295. Yang, J.; Gao, M.X.; Hu, H.; Ding, X.M.; Lin, H.W.; Wang, L.; Xu, J.M.; Mao, C.Z.; Zhao, F.J.; Wu, Z.C. Osclt1, a crt-like transporter 1, is required for glutathione homeostasis and arsenic tolerance in rice. N. Phytol. 2016, 211, 658–670. [Google Scholar] [CrossRef] [PubMed]
  296. Zhang, Y.; Sa, G.; Zhang, Y.; Hou, S.; Wu, X.; Zhao, N.; Zhang, Y.; Deng, S.; Deng, C.; Deng, J.; et al. Populus euphratica annexin1 facilitates cadmium enrichment in transgenic Arabidopsis. J. Hazard. Mater. 2021, 405, 124063. [Google Scholar] [CrossRef] [PubMed]
  297. Chen, S.; Zhang, M.; Feng, Y.; Sahito, Z.A.; Tian, S.; Yang, X. Nicotianamine synthase gene 1 from the hyperaccumulator Sedum alfredii hance is associated with cd/zn tolerance and accumulation in plants. Plant Soil 2019, 443, 413–427. [Google Scholar] [CrossRef]
  298. Han, Y.; Sa, G.; Sun, J.; Shen, Z.; Zhao, R.; Ding, M.; Deng, S.; Lu, Y.; Zhang, Y.; Shen, X.; et al. Overexpression of populus euphratica xyloglucan endotransglucosylase/hydrolase gene confers enhanced cadmium tolerance by the restriction of root cadmium uptake in transgenic tobacco. Environ. Exp. Bot. 2014, 100, 74–83. [Google Scholar] [CrossRef]
  299. Liu, X.; Wang, H.; He, F.; Du, X.; Ren, M.; Bao, Y. The tawrky22-tacopt3d pathway governs cadmium uptake in wheat. Int. J. Mol. Sci. 2022, 23, 10379. [Google Scholar] [CrossRef]
  300. Song, J.; Feng, S.J.; Chen, J.; Zhao, W.T.; Yang, Z.M. A cadmium stress-responsive gene atfc1 confers plant tolerance to cadmium toxicity. BMC Plant Biol. 2017, 17, 187. [Google Scholar] [CrossRef]
  301. Wu, X.; Chen, L.; Lin, X.; Chen, X.; Han, C.; Tian, F.; Wan, X.; Liu, Q.; He, F.; Chen, L.; et al. Integrating physiological and transcriptome analyses clarified the molecular regulation mechanism of pywrky48 in poplar under cadmium stress. Int. J. Biol. Macromol. 2023, 238, 124072. [Google Scholar] [CrossRef]
  302. Chen, X.; Wu, X.; Han, C.; Jia, Y.; Wan, X.; Liu, Q.; He, F.; Zhang, F. A wrky transcription factor, pywrky71, increased the activities of antioxidant enzymes and promoted the accumulation of cadmium in poplar. Plant Physiol. Biochem. 2023, 205, 108163. [Google Scholar]
  303. Wu, X.; Chen, Q.; Chen, L.; Tian, F.; Chen, X.; Han, C.; Mi, J.; Lin, X.; Wan, X.; Jiang, B.; et al. A wrky transcription factor, pywrky75, enhanced cadmium accumulation and tolerance in poplar. Ecotoxicol. Environ. Saf. 2022, 239, 113630. [Google Scholar] [CrossRef] [PubMed]
  304. Yang, G.; Wang, C.; Wang, Y.; Guo, Y.; Zhao, Y.; Yang, C.; Gao, C. Overexpression of thvhac1 and its potential upstream regulator, thwrky7, improved plant tolerance of cadmium stress. Sci. Rep. 2016, 6, 18752. [Google Scholar] [CrossRef]
  305. Wang, Y.; Qiu, W.; Li, H.; He, X.; Liu, M.; Han, X.; Zhuo, R. Research on the response of sawrky7 gene to cadmium stress in Sedum alfredii hance. J. Nanjing For. Univ. 2019, 62, 59–66. [Google Scholar]
  306. Jia, Z.; Li, M.; Wang, H.; Zhu, B.; Gu, L.; Du, X.; Ren, M. Tawrky70 positively regulates tacat5 enhanced cd tolerance in transgenic Arabidopsis. Environ. Exp. Bot. 2021, 190, 104591. [Google Scholar] [CrossRef]
  307. Li, G.; Zheng, Y.; Liu, H.; Liu, J.; Kang, G. Wrky74 regulates cadmium tolerance through glutathione-dependent pathway in wheat. Environ. Sci. Pollut. Res. 2022, 29, 68191–68201. [Google Scholar] [CrossRef]
  308. Cai, Z.; Xian, P.; Wang, H.; Lin, R.; Lian, T.; Cheng, Y.; Ma, Q.; Nian, H. Transcription factor gmwrky142 confers cadmium resistance by up-regulating the cadmium tolerance 1-like genes. Front. Plant Sci. 2020, 11, 534347. [Google Scholar] [CrossRef] [PubMed]
  309. Xian, P.; Yang, Y.; Xiong, C.; Guo, Z.; Alam, I.; He, Z.; Zhang, Y.; Cai, Z.; Nian, H. Overexpression of gmwrky172 enhances cadmium tolerance in plants and reduces cadmium accumulation in soybean seeds. Front. Plant Sci. 2023, 14, 1133892. [Google Scholar] [CrossRef]
  310. He, G.; Saleem, M.; Deng, T.; Zhong, Z.; He, T.; Wu, J. Unraveling the mechanism of stwrky6 in potato (Solanum tuberosum) & rsquo;s cadmium tolerance for ensuring food safety. Foods 2023, 12, 2303. [Google Scholar]
  311. Dang, F.; Lin, J.; Chen, Y.; Li, G.X.; Guan, D.; Zheng, S.J.; He, S. A feedback loop between cawrky41 and h2o2 coordinates the response to ralstonia solanacearum and excess cadmium in pepper. J. Exp. Bot. 2019, 70, 1581–1595. [Google Scholar] [CrossRef] [PubMed]
  312. Chiab, N.; Charfeddine, S.; Ayadi, M.; Abdelkafi, Y.; Mzid, R.; Gargouri-Bouzid, R.; Nouri-Ellouz, O. Enhancing cadmium stress tolerance in potato plants through overexpression of the vvwrky2 transcription factor. Potato Res. 2024, 1–19. [Google Scholar] [CrossRef]
  313. Hong, C.; Cheng, D.; Zhang, G.; Zhu, D.; Chen, Y.; Tan, M. The role of zmwrky4 in regulating maize antioxidant defense under cadmium stress. Biochem. Biophys. Res. Commun. 2017, 482, 1504–1510. [Google Scholar] [CrossRef] [PubMed]
  314. Gu, L.; Hou, Y.; Sun, Y.; Chen, X.; Wang, G.; Wang, H.; Zhu, B.; Du, X. The maize wrky transcription factor zmwrky64 confers cadmium tolerance in arabidopsis and maize (Zea mays L.). Plant Cell Rep. 2024, 43, 44. [Google Scholar] [CrossRef] [PubMed]
  315. Hu, S.; Yu, Y.; Chen, Q.; Mu, G.; Shen, Z.; Zheng, L. Osmyb45 plays an important role in rice resistance to cadmium stress. Plant Sci. 2017, 264, 1–8. [Google Scholar] [CrossRef] [PubMed]
  316. Agarwal, P.; Mitra, M.; Banerjee, S.; Roy, S. Myb4 transcription factor, a member of r2r3-subfamily of myb domain protein, regulates cadmium tolerance via enhanced protection against oxidative damage and increases expression of pcs1 and mt1c in Arabidopsis. Plant Sci. 2020, 297, 110501. [Google Scholar] [CrossRef] [PubMed]
  317. Zheng, P.; Cao, L.; Zhang, C.; Pan, W.; Wang, W.; Yu, X.; Li, Y.; Fan, T.; Miao, M.; Tang, X.; et al. Myb43 as a novel substrate for crl4prl1 e3 ligases negatively regulates cadmium tolerance through transcriptional inhibition of hmas in Arabidopsis. N. Phytol. 2022, 234, 884–901. [Google Scholar] [CrossRef] [PubMed]
  318. Zheng, T.; Lu, X.; Yang, F.; Zhang, D. Synergetic modulation of plant cadmium tolerance via myb75-mediated ros homeostasis and transcriptional regulation. Plant Cell Rep. 2022, 41, 1515–1530. [Google Scholar] [CrossRef] [PubMed]
  319. Zhu, S.; Shi, W.; Jie, Y.; Zhou, Q.; Song, C. A myb transcription factor, bnmyb2, cloned from ramie (Boehmeria nivea) is involved in cadmium tolerance and accumulation. PLoS ONE 2020, 15, e0233375. [Google Scholar] [CrossRef]
  320. Xu, Z.; Wang, T.; Hou, S.; Ma, J.; Li, D.; Chen, S.; Gao, X.; Zhao, Y.; He, Y.; Yang, G. A r2r3-myb, bpmyb1, from paper mulberry interacts with della protein bpgai1 in soil cadmium phytoremediation. J. Hazard. Mater. 2024, 463, 132871. [Google Scholar] [CrossRef]
  321. Gao, W.; Liu, B.; Phetmany, S.; Li, J.; Wang, D.; Liu, Z.; Gao, C. Thdiv2, an r-r-type myb transcription factor of tamarix hispida, negatively regulates cadmium stress by modulating ros homeostasis. Environ. Exp. Bot. 2023, 214, 105453. [Google Scholar] [CrossRef]
  322. Sapara, K.K.; Khedia, J.; Agarwal, P.; Gangapur, D.R.; Agarwal, P.K. Sbmyb15 transcription factor mitigates cadmium and nickel stress in transgenic tobacco by limiting uptake and modulating antioxidative defence system. Funct. Plant Biol. 2019, 46, 702–714. [Google Scholar] [CrossRef] [PubMed]
  323. Wu, H.; Chen, C.; Du, J.; Liu, H.; Cui, Y.; Zhang, Y.; He, Y.; Wang, Y.; Chu, C.; Feng, Z.; et al. Co-overexpression fit with atbhlh38 or atbhlh39 in arabidopsis-enhanced cadmium tolerance via increased cadmium sequestration in roots and improved iron homeostasis of shoots. Plant Physiol. 2012, 158, 790–800. [Google Scholar] [CrossRef] [PubMed]
  324. Yao, X.; Cai, Y.; Yu, D.; Liang, G. Bhlh104 confers tolerance to cadmium stress in Arabidopsis thaliana. J. Integr. Plant Biol. 2018, 60, 691–702. [Google Scholar] [CrossRef] [PubMed]
  325. Du, X.; Fang, L.; Li, J.; Chen, T.; Cheng, Z.; Zhu, B.; Gu, L.; Wang, H. The tabhlh094–tamyc8 complex mediates the cadmium response in wheat. Mol. Breed. 2023, 43, 57. [Google Scholar] [CrossRef] [PubMed]
  326. Wang, H.; Zuo, D.; Zhu, B.; Du, X.; Gu, L. Tamyc8 regulates taerf6 and inhibits ethylene synthesis to confer cd tolerance in wheat. Environ. Exp. Bot. 2022, 198, 104854. [Google Scholar] [CrossRef]
  327. Xu, Z.; Liu, X.; He, X.; Xu, L.; Huang, Y.; Shao, H.; Zhang, D.; Tang, B.; Ma, H. The soybean basic helix-loop-helix transcription factor org3-like enhances cadmium tolerance via increased iron and reduced cadmium uptake and transport from roots to shoots. Front. Plant Sci. 2017, 8, 1098. [Google Scholar] [CrossRef] [PubMed]
  328. Zhan, J.; Zou, W.; Li, S.; Tang, J.; Lu, X.; Meng, L.; Ye, G. Osnac15 regulates tolerance to zinc deficiency and cadmium by binding to oszip7 and oszip10 in rice. Int. J. Mol. Sci. 2022, 23, 11771. [Google Scholar] [CrossRef] [PubMed]
  329. Hu, S.; Shinwari, K.I.; Song, Y.; Xia, J.; Xu, H.; Du, B.; Luo, L.; Zheng, L. Osnac300 positively regulates cadmium stress responses and tolerance in rice roots. Agronomy 2021, 11, 95. [Google Scholar] [CrossRef]
  330. Yu, Y.; Zhang, L. The wheat nac transcription factor tanac22 enhances cadmium stress tolerance in wheat. Cereal Res. Commun. 2023, 51, 867–877. [Google Scholar]
  331. Du, X.; He, F.; Zhu, B.; Ren, M.; Tang, H. Nac transcription factors from Aegilops markgrafii reduce cadmium concentration in transgenic wheat. Plant Soil 2020, 449, 39–50. [Google Scholar] [CrossRef]
  332. Shim, D.; Hwang, J.; Lee, J.; Lee, S.; Choi, Y.; An, G.; Martinoia, E.; Lee, Y. Orthologs of the class a4 heat shock transcription factor hsfa4a confer cadmium tolerance in wheat and rice. Plant Cell 2009, 21, 4031–4043. [Google Scholar] [CrossRef] [PubMed]
  333. Cai, S.; Zhang, Y.; Xu, Y.; Qi, Z.; Li, M.; Ahammed, G.J.; Xia, X.; Shi, K.; Zhou, Y.; Reiter, R.J.; et al. Hsfa1a upregulates melatonin biosynthesis to confer cadmium tolerance in tomato plants. J. Pineal Res. 2017, 62, e12387. [Google Scholar] [CrossRef] [PubMed]
  334. Chen, S.; Yu, M.; Li, H.; Wang, Y.; Lu, Z.; Zhang, Y.; Liu, M.; Qiao, G.; Wu, L.; Han, X.; et al. Sahsfa4c from Sedum alfredii hance enhances cadmium tolerance by regulating ros-scavenger activities and heat shock proteins expression. Front. Plant Sci. 2020, 11. [Google Scholar] [CrossRef]
  335. Lin, T.; Yang, W.; Lu, W.; Wang, Y.; Qi, X. Transcription factors pverf15 and pvmtf-1 form a cadmium stress transcriptional pathway. Plant Physiol. 2017, 173, 1565–1573. [Google Scholar] [CrossRef]
Figure 2. Pathways of Cd uptake and transfer by plants through roots, stems, and leaves. (A) The pathway of Cd uptake in plant roots from root hair cells in the maturation zone of the root tip, through the exodermis and endodermis to the stele (Xylem). (B) The translocation process of Cd to the aboveground parts through the xylem and phloem after reaching the stele. (C) Schematic diagram of Cd uptake and transport by plant leaves under Cd stress.
Figure 2. Pathways of Cd uptake and transfer by plants through roots, stems, and leaves. (A) The pathway of Cd uptake in plant roots from root hair cells in the maturation zone of the root tip, through the exodermis and endodermis to the stele (Xylem). (B) The translocation process of Cd to the aboveground parts through the xylem and phloem after reaching the stele. (C) Schematic diagram of Cd uptake and transport by plant leaves under Cd stress.
Cells 13 00907 g002
Figure 3. Schematic model of the major proteins/enzymes that are absorbed, transported, sequestered, and detoxified in plants. Red circles represent Cd2+ and [number] represents the serial number. Plants take up Cd and Cd chelates through NRAMP, YSL, ZIP families, and Ca2+ channels; ABC and PLAC8 families have been shown to function in effluxing Cd out of the plant; CDT1 and XTH can avoid Cd entry by binding Cd or by reducing the Cd-binding site; the DEFL family can bind to Cd and convert Cd ions into stable compounds; HMA, CaCA, and ABC families can transport Cd and chelates into vacuoles to alleviate the toxic effects; YSL and HMA can transport some Cd to xylem and transfer it to the aboveground part. Refs. [1,2,3,4,5] are proteins that have been reported to be related to Cd transport. Ref. [1] SpHMA6, SaPCR2, SlCNR8, PcPLAC8-10, OsCd1, OsHIR1, OsAAN4, and OsGLR3.4, respectively; Ref. [2] SlCNR8, OsZIP1, OsHMA9, HaMTP10, AtPDF2.5, OsCCX2 (OsCDT1), OsLCT1; Ref. [3] OsNRAMP2, AtNRAMP3, AtNRAMP4; Ref. [4] TmMTP1, TmMTP11, AtCAX2; Ref. [5] OsZIP7, OsCAL1. On the right is the Cd-induced ROS scavenging cycle. Cd enters the cytoplasm and stimulates the synthesis of osmoprotectants, antioxidants, glutathione and phytochelatin, and metallothionein. MT, GSH, and PC can bind to Cd to generate Cd-GS2, Cd-MT, and Cd-PC to alleviate the toxicity of Cd caused to the cells. ROS, reactive oxygen species; NRAMP, natural resistance-associated macrophage protein; YSL, yellow-stripe-1-like; ABC, ATP-binding cassette family; PLAC8, the placenta-specific 8-domain-containing family; ZIP, ZRT-IRT-like protein family; zinc-regulated; HMA, heavy metal ATPase; CaCA, cation/calcium superfamily; DEFL, defensin-like protein family; PCS, phytochelatin synthetase; Gly, Glycine; Glu, Glutamate; Cys, Cysteine; MT, metallothioneins; GSH, glutathione; PC, phytochelatin; NA, nicotianamine; DMA,2’-deoxymugineic acid.
Figure 3. Schematic model of the major proteins/enzymes that are absorbed, transported, sequestered, and detoxified in plants. Red circles represent Cd2+ and [number] represents the serial number. Plants take up Cd and Cd chelates through NRAMP, YSL, ZIP families, and Ca2+ channels; ABC and PLAC8 families have been shown to function in effluxing Cd out of the plant; CDT1 and XTH can avoid Cd entry by binding Cd or by reducing the Cd-binding site; the DEFL family can bind to Cd and convert Cd ions into stable compounds; HMA, CaCA, and ABC families can transport Cd and chelates into vacuoles to alleviate the toxic effects; YSL and HMA can transport some Cd to xylem and transfer it to the aboveground part. Refs. [1,2,3,4,5] are proteins that have been reported to be related to Cd transport. Ref. [1] SpHMA6, SaPCR2, SlCNR8, PcPLAC8-10, OsCd1, OsHIR1, OsAAN4, and OsGLR3.4, respectively; Ref. [2] SlCNR8, OsZIP1, OsHMA9, HaMTP10, AtPDF2.5, OsCCX2 (OsCDT1), OsLCT1; Ref. [3] OsNRAMP2, AtNRAMP3, AtNRAMP4; Ref. [4] TmMTP1, TmMTP11, AtCAX2; Ref. [5] OsZIP7, OsCAL1. On the right is the Cd-induced ROS scavenging cycle. Cd enters the cytoplasm and stimulates the synthesis of osmoprotectants, antioxidants, glutathione and phytochelatin, and metallothionein. MT, GSH, and PC can bind to Cd to generate Cd-GS2, Cd-MT, and Cd-PC to alleviate the toxicity of Cd caused to the cells. ROS, reactive oxygen species; NRAMP, natural resistance-associated macrophage protein; YSL, yellow-stripe-1-like; ABC, ATP-binding cassette family; PLAC8, the placenta-specific 8-domain-containing family; ZIP, ZRT-IRT-like protein family; zinc-regulated; HMA, heavy metal ATPase; CaCA, cation/calcium superfamily; DEFL, defensin-like protein family; PCS, phytochelatin synthetase; Gly, Glycine; Glu, Glutamate; Cys, Cysteine; MT, metallothioneins; GSH, glutathione; PC, phytochelatin; NA, nicotianamine; DMA,2’-deoxymugineic acid.
Cells 13 00907 g003
Figure 4. Toxic effects of Cd on plants and plant coping mechanisms. The blue part on the left shows the negative effects of Cd stress on plants, and the pink color on the right shows the mitigation mechanisms that plants have evolved over millions of years to cope with Cd stress.
Figure 4. Toxic effects of Cd on plants and plant coping mechanisms. The blue part on the left shows the negative effects of Cd stress on plants, and the pink color on the right shows the mitigation mechanisms that plants have evolved over millions of years to cope with Cd stress.
Cells 13 00907 g004
Figure 5. Hot spots for future phytoremediation of Cd pollution. (A) Screening of Cd hyperaccumulation-tolerant plants and woody plants that can take up more Cd; (B) Enhancement of Cd tolerance in plants by exogenous means (chemical regulators or genetic engineering).
Figure 5. Hot spots for future phytoremediation of Cd pollution. (A) Screening of Cd hyperaccumulation-tolerant plants and woody plants that can take up more Cd; (B) Enhancement of Cd tolerance in plants by exogenous means (chemical regulators or genetic engineering).
Cells 13 00907 g005
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

Zhang, X.; Yang, M.; Yang, H.; Pian, R.; Wang, J.; Wu, A.-M. The Uptake, Transfer, and Detoxification of Cadmium in Plants and Its Exogenous Effects. Cells 2024, 13, 907. https://doi.org/10.3390/cells13110907

AMA Style

Zhang X, Yang M, Yang H, Pian R, Wang J, Wu A-M. The Uptake, Transfer, and Detoxification of Cadmium in Plants and Its Exogenous Effects. Cells. 2024; 13(11):907. https://doi.org/10.3390/cells13110907

Chicago/Turabian Style

Zhang, Xintong, Man Yang, Hui Yang, Ruiqi Pian, Jinxiang Wang, and Ai-Min Wu. 2024. "The Uptake, Transfer, and Detoxification of Cadmium in Plants and Its Exogenous Effects" Cells 13, no. 11: 907. https://doi.org/10.3390/cells13110907

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