Next Article in Journal
Effects of Planting Pre-Germinated Buds on Stand Establishment in Sugarcane
Previous Article in Journal
Banana Pseudostem Visual Detection Method Based on Improved YOLOV7 Detection Algorithm
Previous Article in Special Issue
Genetic Diversity in Oilseed and Vegetable Mustard (Brassica juncea L.) Accessions Revealed by Nuclear and Mitochondrial Molecular Markers
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Transgene-Free Genome Editing for Biotic and Abiotic Stress Resistance in Sugarcane: Prospects and Challenges

by
Sakthivel Surya Krishna
1,
S R Harish Chandar
1,
Maruthachalam Ravi
2,
Ramanathan Valarmathi
1,
Kasirajan Lakshmi
1,
Perumal Thirugnanasambandam Prathima
1,
Ramaswamy Manimekalai
1,
Rasappa Viswanathan
3,
Govindkurup Hemaprabha
1 and
Chinnaswamy Appunu
1,*
1
Division of Crop Improvement, Indian Council of Agricultural Research-Sugarcane Breeding Institute, Coimbatore 641007, Tamil Nadu, India
2
Indian Institute of Science Education and Research (IISER), Thiruvananthapuram 695551, Kerala, India
3
ICAR—Indian Institute of Sugarcane Research, Lucknow 226002, Uttar Pradesh, India
*
Author to whom correspondence should be addressed.
Agronomy 2023, 13(4), 1000; https://doi.org/10.3390/agronomy13041000
Submission received: 3 March 2023 / Revised: 18 March 2023 / Accepted: 24 March 2023 / Published: 29 March 2023
(This article belongs to the Special Issue Application of Molecular Marker Technology in Crop Breeding)

Abstract

:
Sugarcane (Saccharum spp.) is one of the most valuable food and industrial crops. Its production is constrained due to major biotic (fungi, bacteria, viruses and insect pests) and abiotic (drought, salt, cold/heat, water logging and heavy metals) stresses. The ever-increasing demand for sugar and biofuel and the rise of new pest and disease variants call for the use of innovative technologies to speed up the sugarcane genetic improvement process. Developing new cultivars through conventional breeding techniques requires much time and resources. The advent of CRISPR/Cas genome editing technology enables the creation of new cultivars with improved resistance/tolerance to various biotic and abiotic stresses. The presence of genome editing cassette inside the genome of genome-edited plants hinders commercial exploitation due to regulatory issues. However, this limitation can be overcome by using transgene-free genome editing techniques. Transgene-free genome editing approaches, such as delivery of the RNPs through biolistics or protoplast fusion, virus-induced genome editing (VIGE), transient expression of CRISPR/Cas reagents through Agrobacterium-mediated transformation and other approaches, are discussed. A well-established PCR-based assay and advanced screening systems such as visual marker system and Transgene killer CRISPR system (TKC) rapidly identify transgene-free genome edits. These advancements in CRISPR/Cas technology speed up the creation of genome-edited climate-smart cultivars that combat various biotic and abiotic stresses and produce good yields under ever-changing conditions.

1. Introduction

Sugarcane (Saccharum spp.) is one of the most important food and industrial crops cultivated across the world [1]. Sugarcane contributes to 80% of global sugar production with India as the leading sugar producer followed by Brazil, European Union, Thailand and China [2]. In addition to this, sugarcane juice and the by-product, molasses, are the major raw materials used in distilleries for alcohol and bioethanol production [1]. The crushed sugarcane stalks are burned to generate electricity. Thus, sugarcane plays an indispensable role in supplying raw materials for the food and fuel industries. The human population reached approximately 7.95 billion in 2022 and it is projected that the global population will reach 9.7 billion in 2050 [3]. The pollution caused by industries, urban sewage and, heavy use of chemical inputs in agricultural fields causes severe damage to land and water resources, which in turn have serious impacts on all-life forms on Earth. The lethal impacts of climate change on agriculture are the epidemic outbreak of new pests and diseases and the periodic occurrence of drought and waterlogging. Sugarcane is adversely affected by various biotic and abiotic stresses. Biotic stresses, such as insect pests, diseases and noxious weeds, severely affect cane production and juice quality [4,5,6]. Severe drought due to monsoon failure, excessive rainfall, low temperature, salinity, alkalinity and heavy metal contamination in soil increase day-by-day and also have an adverse effect on cane production and juice quality [7,8,9,10].
Conventional breeding and molecular marker-assisted selection are carried out in sugarcane to develop new cultivars. Transgenic events were also developed in sugarcane for pest and disease resistance, but their commercialization has not been successful due to various regulatory issues [11]. Although conventional breeding strategies have led to commercial success, more time is required to develop cultivars. The polyploid nature of sugarcane makes crop improvement programs much more complicated than for diploid plants. The increasing human population and adverse impacts of climate change on plants and animals call for the use of sustainable modern techniques in crop production to meet global food demands and to ensure a better way of living. The advent of genome editing technologies, such as zinc finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENS), clustered regularly interspaced short palindromic repeats and CRISPR-associated protein 9 (CRISPR/Cas9), expedites the crop improvement process by creating site-specific mutations in the plant genome [12]. Among these, CRISPR/Cas9 is widely used because of its high editing efficiency, cost-effectiveness, and multiplexing [13].
CRISPR/cas9 consists of two main components: a guide RNA and Cas9 endonuclease. The single-stranded guide RNA consists of a 20 bp sequence complementary to the host target region, and Cas9 induces DSBs (Double-stranded breaks) in the 3 bp upstream of PAM sequence (5′NGG3′) in the DNA of the host, complementary to the guide RNA (gRNA) [14,15]. The host cell DNA repair machinery repairs the breaks by either NHEJ (Non-homologous end joining) or HDR (Homology directed repair). NHEJ is predominant and error-prone, thus creating mutations by base insertions, deletions and replacements [16,17,18]. Currently, many Cas variants other than Cas9 have been identified and used for genome editing purposes. The problem with using vector-based delivery of the CRISPR/Cas system is that the markers, promoters and Cas9 genes in the genome editing cassette integrate into the plant genome and are passed on to successive generations, which places the genome-edited (GE) plants developed through this technique under the category of genetically modified organisms (GMOs). In the case of seed propagated annuals, the transgenes can be eliminated by recombination and selection. Sugarcane is a highly heterozygous vegetatively propagated crop; eliminating the transgenes via meiotic recombination and producing GE lines with exact genome composition other than the difference in target region is highly difficult. However, the advent of transgene-free genome-editing techniques enables the creation of site specific mutations without integration of any residual transgenes in the plant genome and these techniques are very useful in creating GE sugarcane without any residual foreign genes. The objective of this review is to highlight strategies to develop and screen transgene-free GE sugarcane lines for rapid genetic improvement with special emphasis on imparting tolerance/resistance to major biotic and abiotic stresses.

2. Biotic Stresses Affecting Sugarcane

Diseases and insect pests adversely affect sugarcane and incur a huge yield loss, increasing the cost of cultivation [4]. As sugarcane is allowed to produce ratoons, the complete elimination of established diseases and insect pests is an arduous task to accomplish. However, the extent of damage caused by biotic agents can be reduced by adapting various agronomic practices.

2.1. Major Diseases

The major fungal diseases of sugarcane are red rot, wilt and smut [4]. Red rot caused by Colletotrichum falcatum is one of sugarcane’s most devastating diseases that severely affects juice quality by disrupting the sucrose metabolism [4]. The affected canes have a reddish discoloration in the stalk portion [4]. Wilt is caused by Fusarium sacchari, which causes yield loss of up to 15% and reduces sugar recovery during extraction [19]. The symptoms are yellowing and drying of canes with pinkish-brown discoloration in the internal cavities [19]. Smut is caused by Sporisorium scitamineum which causes yield losses of up to 50% and reduces sugar recovery. The symptoms of smut are affected cane shoots modified into sorus or a whip-like structures [20]. The other fungal diseases of sugarcane are pineapple disease, rust, pokkah boeng, stalk rot, brown spot, eye spot, ring spot, seedling rot and many others [4].
The major bacterial diseases affecting sugarcane are ratoon stunting and leaf scald. Ratoon stunting is caused by Leifsonia (Clavibacter) xyli subsp. xyli causes yield loss up to 30% [21]. The symptoms of the disease are stunted growth with thin stalks, reduction in tillering, shortened internodes and yellowing of the leaf. Leaf scald is caused by Xanthomonas albilineans; the pathogen shows acute and chronic expression [22]. During the chronic phase, the pathogen produces white lines along the leaf blade to the leaf sheath, followed by chlorosis, necrosis and death of young shoots and stalks. The acute phase is initially symptomless followed by the sudden outbreak and death of plants [22,23].
Sugarcane grassy shoot (SCGS) disease is a phytoplasma disease of sugarcane transmitted by leaf hoppers which causes huge yield loss in susceptible varieties. The symptoms of SCGS are excessive tillering with thin slender tillers and shortened internodes showing grass-like appearance with chlorosis. The affected canes fail to produce millable canes [4,24]. Yellow leaf is one of the most destructive diseases caused by the sugarcane yellow leaf virus (ScYLV), which accounts for 15–50% yield loss of sugarcane [25,26,27,28]. The symptoms of yellow leaf disease appear on 6–8-month-old crops and the symptoms are prominent midrib yellowing on the lower surface of the leaf followed by the lateral spread of yellowing and leaf tip necrosis, shortening of internodes and bunchy top appearance of leaves during severe infection [25,26,27,28]. Sugarcane mosaic disease is caused by three viruses, namely sugarcane mosaic virus (SCMV), sugarcane streak mosaic virus (SCSMV) and sorghum mosaic virus (SrMV), which cause severe yield losses of more than 50% in case of severe infection. The symptoms of the mosaic disease are yellow and green spots on the young leaves with interveinal chlorosis; upon severe infection, leaves turn yellow or yellow-white with green or necrotic patches [29,30,31].

2.2. Major Insect Pests

Pest infestation causes severe yield losses in sugarcane. The major insect pests affecting sugarcane are early shoot borer, top borer, internode borer, termites, root grub, aphids and white fly. The early shoot borer (Chilo infuscatellus) affects canes during the early crop stages from one to three months; the symptoms are bore holes in the shoot above ground level, dead heart and rotten portions that emit an offensive odour, and affected canes that can be easily pulled out [5,32,33]. The internode borer (Chilo sacchariphagus indicus) affects the crop soon after internodes emerge; the boreholes are plugged with excreta and frass is seen in the infected portions. The internodes are reduced in length and girth [5,33,34]. The top shoot borer (Scirpophaga excerptalis) affects the crop during all growth stages; its characteristic symptoms are bore holes in the apical portion of the stem, shot holes on opened leaves, dead heart in the fully grown canes and bunchy top formation [5,33,35]. The borers account for yield losses of 15–42% [5,33,36].
Whitefly (Aleurolobus barodensis and Neomaskellia andropogonis) is a serious sucking sugarcane pest that causes yield reduction up to 50% [37,38]. The symptoms are yellowing of leaves followed by drying and retarded growth [37,38]. The sugarcane woolly aphid (Ceratovacuna lanigera) produces symptoms similar to that of whitefly. Honeydew secretions from whitefly and aphids favour the growth of sooty mould fungus [33].
Subterranean pests, such as termites and white grubs, are difficult to control and cause severe yield loss. White grub (Holotrichia serrata) is another serious sugarcane pest that can cause yield losses up to 100% [33,39]. The larvae feed voraciously on the root system, leading to poor growth, lodging, yellowing and wilting of canes. The affected canes can be easily pulled out and the damage usually occurs in patches in the field [33,39]. Termites (Odontotermes obesus) mainly affect the germinating setts in the field, causing poor germination. The damaged portions are filled with soil, and damage in the later stages of the crop causes yellowing and drying of leaves [5,33].

3. Abiotic Stress in Sugarcane

Worldwide sugarcane production is affected by abiotic stresses, namely drought, salinity, cold and heat stress, and metal toxicity. These stresses incur huge economic loss to farmers and severely restrict crop cultivation in stress-prone areas.

3.1. Drought

Water deficit caused by meteorological and agricultural drought leads to drought stress [40]. Lack of water during key water requirement periods, such as germination, tillering, grand growth phase and maturation, reduces cane development and affects the sugar content [41]. Yield loss due to lack of water in sugarcane is estimated to be approximately 50–70% based on the degree of water stress [42]. Plants use various morphological and physiological strategies to mitigate drought stress, which varies from genotype to genotype [43]. The plant responds to water stress by exhibiting physiological changes such as leaf rolling, stomatal closure, inhibition of stalk and leaf growth, leaf senescence and reduced leaf area [44]. Water stress reduces biochemical activities in mesophyll and bundle sheath cells which in turn reduces the sugar accumulation in the plant [45]. The accumulation of ROS, such as superoxide radicals (O2), hydrogen peroxide (H2O2) and hydrogen radicals (OH∙), in the cells causes severe damage to cell components such as chlorophyll, lipids, proteins and nucleic acids [46,47]. The ROS also acts as a secondary messenger in response to stress conditions. Hormones play an important role as messengers that regulate plant’s responses to lack of soil water. Abscisic acid (ABA) synthesized by plant roots, which control the stomatal opening and closure, produces the first signal during plant stress and is transported through the xylem to the roots [48]. Controlled and regulated opening and closing of stomata is required to avoid loss of water through transpiration during water deficit conditions [49].

3.2. Salt

Since sugarcane is a glycophyte, it is highly sensitive to salinity [50]. Germination and early growth stages are more sensitive to salt stress compared to later stages [51]. High salt concentration leads to an increase in osmotic and ionic stresses. The majority of the ionic stress is caused by sodium toxicity which leads to ion imbalance, and hyperionic or hyper-osmotic stress disintegrates the overall metabolic activities leading the plant to death [51]. Salt stress leads to a reduction in leaf area, chlorophyll content and stomatal conductance which reduces productivity and juice quality. Plants have developed various self-defensive complex mechanisms, such as adaptation, accumulation and combinations of these, to protect themselves from the osmotic and ionic stresses caused by high salt concentration [52]. The adaptation mechanism mostly followed by plants to mitigate salt stress is a permanent genetic modification of the plant’s structure and function [10]. In addition to the adaptation mechanism, a majority of the plants respond to intermittent stresses by modifying physiological or morphological changes by a process called acclimation, a reversible process that does not involve any permanent genetic changes [10].

3.3. Cold and Heat

Sugarcane production and productivity are severely affected by high and low-temperature stresses [53]. Cold stress severely affects the plants’ metabolic processes and accumulates reactive oxygen species (ROS), which severely damages biomolecules such as DNA, proteins, carbohydrates, lipids, and pigments in the cell [54]. Cold stress causes shortening of internodes and leaves and stunting of canes, and also affects the ratooning ability [55]. Heat stress severely affects the germination and growth of seedlings despite having enough water. High-temperature stress leads to drying and necrosis of leaf tips and margins, and reduces photosynthesis and sugar accumulation [56].

3.4. Waterlogging

Waterlogging mainly caused by excessive rainfall and flood affects sugarcane yield and juice quality [53,57,58]. Waterlogging during the formative stage causes a yield reduction of up to 45% [58]. Water stagnation for a prolonged period of time severely affects the crop metabolism, nutrient and water uptake, decreases sucrose content and increases ROS, glucose, total colloids, non-proteinous nitrogen and gums [57,58]. The symptoms of waterlogging are aerial root formation, reduction in leaf size and number, stunted growth, yellowing and wilting of canes, ultimately leading to death of the plants [53,58]. The aerial roots with aerenchymatic cells help in gas exchange under submerged conditions. Ethylene accumulation in root systems increases under waterlogging which aids in the production of aerial roots [58].

3.5. Heavy Metals

Macro and micronutrients are required to the plant for their normal growth and development [59]. Excess accumulation of these macro and micronutrients such as manganese (Mn), copper (Cu), nickel (Ni) and others in the soil are harmful to plants [59,60]. Recent activities, such as mining, exploitation of groundwater, excessive use of fertilizers and industrialization, have greatly contributed to the repeated accumulation of toxic heavy metals such as lead, cadmium, arsenic, mercury, chromium and many others in the soil [61,62]. Accumulation of these heavy metals adversely affect soil health and is also harmful to all biological organisms, from microbes to humans [62,63,64]. Heavy metal toxicity affects all essential plant biological processes, such as germination, respiration, photosynthesis, metabolic reactions and reproduction. Symptoms of heavy metal toxicity in plants are chlorosis, necrosis, stunted growth, senescence, yield reduction and death of plants [59,60].

4. Transgene-Free Genome Editing: Approaches and Applications

Improvements in genome editing technique enable the creation of transgene-free GE plants in a single generation which is highly beneficial for producing transgene-free mutants in vegetatively propagated crops such as sugarcane. These techniques eliminate the need for meiotic recombination-based selection of transgene-free plants in successive generations. The possible techniques that may be applied to produce transgene-free GE lines in sugarcane are illustrated in Figure 1.

4.1. Virus-Induced Genome Editing

Virus-induced genome editing (VIGE) is one of the promising approaches to develop transgene-free GE plants. Both DNA and RNA viruses can be used to deliver the gRNA and Cas endonucleases into plant systems. This can be performed in two ways: by delivering the gRNA into a plant system that already has Cas9 integrated into its genome, or by delivering both gRNA and Cas9 into the plant genome simultaneously [65]. The VIGE has been successfully employed in both model plants and economically important crops, such as tobacco, Arabidopsis, wheat, rice, maize, soybean, potato and tomato [65]. DNA viruses such as bean yellow dwarf virus (BeYDV), wheat dwarfing virus (WDV), cotton leaf crumple virus (CLCrV), and cabbage leaf curl virus (CaLCuV), and RNA viruses such as tobacco rattle virus (TRV), potato virus X (PVX), pea early browning virus (PEBV), tobacco mosaic virus (TMV), apple latent spherical virus (ALSV), beet necrotic yellow vein virus (BNYVV), tobacco etch virus (TEV), and foxtail mosaic virus (FoMV), have been successfully used for gRNA delivery into the plant system [66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84]. However, the serious limitation of using these viruses is their limited cargo-carrying capacity which makes it highly difficult to carry both gRNA and Cas9 in the same virus. These viruses can be efficiently used for introducing gRNA into plants that already have Cas9 in their genome. The negative-strand RNA viruses, namely barley yellow striate mosaic virus (BYSMV) and sonchus yellow net virus (SYNV), have been used to produce targeted mutation in N. benthamiana by expressing both gRNA and Cas9 [85,86]. The large cargo-carrying capacity of these viruses has a potential application in delivering complete genome editing components into the host system. Since the size of Cas9 is large (~4.1 kb), it cannot be delivered together with gRNA in the plant system by most of the plant viruses due to their limited cargo-carrying capacity; this problem can be resolved by using small-size Cas variants, such as CasΦ and CasX, that enable them to deliver gRNA and Cas endonuclease together into the host plant [87,88]. The virus with genome editing components can be introduced into the host plant either by agroinfiltration or by mechanical method [65,82,89]. As the sugarcane is mainly propagated through setts, the following method can be effective for the VIGE approach in sugarcane. First, the viral vectors are agroinfiltrated into the growing canes and once the visible symptoms are expressed, the infected setts are propagated in the pot followed by meristem culture of the pot-grown setts to produce transgene and virus-free GE plants. Another approach is by subjecting canes to agroinfiltration, allowing them to set seeds and using the seeds as propagative material to develop transgene-free GE plants. Though the latter approach bypasses the need for tissue culture, it cannot be applied universally as sugarcane seed production is highly location and season-specific and the heterozygous nature produces more recombinants [90,91]. The main advantages of VIGE are the high gRNA expression due to the high proliferation rate of viruses, tissue culture-free development in the case of seed-propagated plants, less off-target effects and transgene-free plants [65,89]. Producing heritable mutations through the virus is difficult because the viruses cannot proliferate into the apical meristem or reproductive tissues. However, this problem can be overcome by fusing mobile RNA sequences, such as Flowering Locus T (FT), with gRNA to enable the virus to enter the shoot apical meristem portions to create heritable mutations without any residual transgenes [92].

4.2. Particle Bombardment/Biolistics

Particle bombardment/biolistics has been widely used for sugarcane genetic transformation along with Agrobacterium-mediated transformation [11,93]. Biolistics can deliver a variety of molecules, such as DNA, RNA, plasmid, and proteins, directly into the cells [94]. Delivering the plasmids containing genome editing cassette through biolistics randomly integrates the genome editing system into the plant genome, and the presence of foreign genes from plasmids into the plant genome leads the GE plants to fall under the category of GMOs. This problem can be circumvented by delivering ribonucleoproteins (RNPs) consisting of in vitro assembled gRNA and Cas9 through particle bombardment. This method uses Cas9 or cpf1 endonuclease that interacts with gRNA without any additional auxiliary factors [95]. The assembled genome editing reagents, such as Cas endonuclease and gRNA molecule, are active only for a short period of time and become degraded inside the cell. The RNP is the mixture of Cas9 protein and gRNA molecules in definite preparation along with other components such as a buffer [96]. The biolistics delivery of RNPs has been successfully used for genome editing in crops, such as rice, wheat and maize [96,97,98,99]. In sugarcane, embryogenic callus is widely used for particle bombardment and many successful transgenic events were developed using biolistics [11,93]. Biolistics was successfully used for delivering the CRISPR/Cas9 genome editing cassette into sugarcane callus by Oz, et al. [100] and Eid, et al. [101]. Oz, et al. [100] edited the alleles of the acetolactate synthase (ALS) gene to produce herbicide resistance. Upon spraying the herbicide nicosulfuron, the wild types died whereas the GE lines survived. Eid, et al. [101] edited the multiple alleles of the magnesium chelatase gene, which is essential for chlorophyll biosynthesis, and the edited plants were yellow in color whereas the wild-type plants showed normal green color. These studies highlighted the potential use of biolistics in CRISPR/Cas-mediated genome editing to produce targeted mutations in sugarcane.

4.3. Agrobacterium-Mediated Transformation

Agrobacterium-mediated transformation has been used for genetic transformation in sugarcane to produce transgenic events for various purposes [11,93]. Transient expression of gRNAs and Cas9 using Agrobacterium eliminates the integration of foreign genes into the plant system. This technique has been used to produce transgene-free GE plants in tobacco, tomato and potato [102,103,104,105,106]. Chen, et al. [103] targeted the PDS gene in tobacco by this method and generated transgene-free GE tobacco without using any selection media. Veillet, et al. [104] delivered Cytosine base editors by Agrobacterium-mediated transformation to edit the acetolactate synthase (ALS) gene to produce chlorsulfuron-resistant potato and tomato plants and obtained 10% and 12.9% of transgene-free potato and tomato plants, respectively [104]. The genome edited plants produced by this method contain a mixture of transgenic and non-transgenic individuals [103,104,105,106]. Leaf rolls and callus are the main explants used for Agrobacterium-mediated transformation which needs in vitro regeneration for its development. Mayavan, et al. [107], Mayavan, et al. [108] successfully demonstrated the in planta transformation using sugarcane seeds and setts through Agrobacterium-mediated transformation. This method bypasses the need for time-consuming and resource-intensive tissue culture facilities to produce transformed lines [107,108]. This opens a new window to exploit the possibilities of producing transgene-free GE plants using in planta transformation method.

4.4. Protoplast Transformation

The RNPs are delivered into protoplast through lipofection or PEG-Ca2+ transfection. The lipofection method has been used in crops such as cabbage, Chinese cabbage and tobacco [109,110]. PEG-Ca2+ transfection was the method mostly used for delivering the genome editing RPNs into the protoplasts and was used to produce transgene-free GE plants in model plant system and crop plants, such as Arabidopsis, tobacco, lettuce, maize, rice, banana, canola, wild tomato and potato [111,112,113,114,115,116]. In the case of sugarcane, electroporation and PEG-mediated protoplast transformation were successful in producing transgenic events, but transformation and regeneration efficiency was low [117,118,119,120]. Wang, et al. [121] optimized the protoplast isolation and transformation through PEG, which had a protoplast yield of approximately 1.26 × 107 per gram of leaf and achieved a transformation efficiency of 80.19% in Saccharum spontaneum [121]. Though efficient protoplast isolation and transformation methods have been developed, the regeneration of whole plants from the sugarcane protoplast is genotype specific and still a promising issue that needs to be addressed. By developing an efficient protoplast regeneration technique, the protoplast transformation could potentially be exploited for producing transgene-free GE sugarcane.

4.5. piggyBac-Mediated Transgenesis

Nishizawa-Yokoi and Toki [122] used piggyBac transposon to remove the randomly integrated CRISPR/Cas9 genome editing cassette from the rice genome. This system enables editing of the target site by temporary expression of CRISPR/Cas9 followed by the elimination of transgenes from the rice genome by piggybac transposase (PBase) [122]. This system successfully produced transgene-free genome-edits in rice and this technology opens a new avenue for eliminating the transgenes in vegetatively propagated crops such as sugarcane, where eliminating transgenes through recombination and selection is highly difficult and time-consuming [122,123].

4.6. Grafting and Mobile RNA-Mediated Genome Editing

A novel technique using grafting for transgene-free GE plants was demonstrated by Yang, et al. [124] in Arabidopsis thaliana and Brassica rapa. The fusion of tRNA-like sequences (TLS) has produced a mobile version of CRISPR/Cas9 RNA which enables the root-to-shoot movement from root stock to scion. Agrobacterium-mediated transformation was used to deliver Cas9-TLS mRNA and gRNA-TLS fusions into Arabidopsis thaliana which was used as root stock for both Arabidopsis scion and B. rapa scion [124]. The hypocotyl grafting was done between transformed A. thaliana root stock containing the genome editing cassette and wild type scions of Arabidopsis and B. rapa. The fusion product moved from root stock to scion with the aid of TLS and created edits in the genome of scions, while the edited scions produced seeds with transgene-free targeted mutation [124]. However, this method needs to be established in sugarcane because the monocots are devoid of vascular cambium and have scattered vascular bundles. Vascular cambium is required for successful graft establishment [125].

5. Screening of Transgene-Free GE Lines

Plasmid-based delivery of CRISPR/Cas genome editing cassette produces a mixture of transgenic and non-transgenic individuals. The techniques such as the transgene killer CRISPR system (TKC) and visual marker system help in the easy identification and selection of transgene-free GE lines generated by plasmid-based delivery methods.
TKC uses suicidal genes that kill the male and female gametophyte-containing transgenes and allows only the seed produced from the fusion of transgene-free pollen and embryo [126,127]. Two cassettes, one containing the barnase gene and the other with CMS2 were added into the CRISPR/Cas9 construct. The barnase gene under the control of rice REG2 promoter kills the embryo that contains transgenes and the CMS2 gene under the control of CaMV 35S promoter disrupts mitochondrial functions during male gametophyte development and causes male sterility [126,127]. Yang, et al. [128] demonstrated the potential of the TKC system along with multiplexing to edit multiple gene families in plants and produced multiple transgene-free null mutant combinations. This system can be potentially used for seed propagating plants, but for vegetatively propagated crops such as sugarcane, where flowering and seed set is highly difficult, this system is a big challenge to implement to produce transgene-free edits.
Transgene-free GE seeds/plants can be easily identified visually by utilizing fluorescent markers and pigments [123]. Introducing fluorescent marker proteins, such as mCherry, along with genome editing cassette produces bright color in transgenic seeds which enables easy differentiation of transgene-free GE seeds [129,130]. By using different marker proteins, such as DsRED, or green fluorescent Proteins (GFPs,) the same strategy was used to select transgene-free seeds in rice, Arabidopsis, tomato and maize [131,132,133]. The other approach to visually select transgene-free GE plants is by introducing genes such as the PAP1 gene of anthocyanin biosynthesis pathway or artificially synthesized RUBY reporter gene for betalain synthesis pathway in the genome editing cassette that produces purple and vivid red color, respectively in the transgenic calli and plants [134,135]. In sugarcane’s case, delivering the genome editing cassette through Agrobacterium-mediated transformation or biolistics, followed by selecting the transgene-free plants through a visual marker system, particularly pigment-based selection during callus stage, will eliminate the plants with integrated transgene at an early stage and reduce the cost and workload involved in analyzing GE lines for targeted mutation.
The transgene-free lines developed through RNP-based delivery systems can be analyzed for targeted mutation using a variety of PCR-based approaches, such as PCR-RFLP, T7 endonuclease 1(T7E1), semi-nested PCR, Droplet Digital PCR and Cas9/sgRNA-assisted reverse PCR (CARP), that can be used for preliminary screening of mutants [136,137,138,139,140]. Among these techniques, PCR-RFLP and T7E1 are mostly used techniques, and in PCR-RFLP, the PCR products are subjected to restriction digestion [139,140]. Restriction enzyme cleaves the target region in wild types whereas mutants remain unaffected. Site-specific mutation created by CRSIPR/Cas reagents leads to loss of restriction site in the genome-edited plants [139,140]. The other reliable technique to screen mutated lines is T7 endonuclease 1 (T7E1) assay [141,142,143,144]. During PCR amplification of mutants, the base changes at the target site cause heteroduplex formation; upon treating the amplified products with T7 endonuclease 1, the heteroduplex portion in mutants will be cleaved and the wild type remains unaffected [141,142,143,144]. These assays enable the preliminary screening of mutated lines and limits the number of individuals for further sequencing and analysis (Figure 2).

6. Candidate Genes for Genome Editing in Sugarcane for Biotic and Abiotic Stress—Lessons from Crop Plants

In sugarcane, a number of candidate genes were identified for biotic and abiotic stresses (Table 1) and targeting these genes through the CRISPR/Cas system may produce multiple stress-resistant/tolerant cultivars. Other than the genes listed in Table 1, many putative candidate genes and genomic loci have been identified in sugarcane and its wild species for various biotic and abiotic stresses [55,145,146,147,148,149].
Genome editing has been successfully used to create GE lines resistant/tolerant to various biotic and abiotic stresses in many crop plants [168,169,170]. Structural genes, regulatory genes, and cis-regulatory elements (CREs) of regulatory and structural genes are the potential targets for CRISPR/Cas system to produce abiotic stress resistant/tolerance plants [168]. In the case of biotic stresses, resistance can be induced either by manipulating the susceptible factors of the host that favors pathogen establishment and spread or by targeting the pathogen factors. Table 2 summarizes some of the recent reports on the use of CRISPR/Cas genome editing in plants for various abiotic and biotic stresses. Most of the published work on transgene-free genome editing was done in model plants, such as Arabidopsis thaliana and Nicotiana bethamiana, by targeting the genes that enable easy visual identification such as PDS. Only very few reports have been published on transgene-free genome editing for biotic and abiotic stresses in crop plants (Table 3).

7. Regulatory Issues in Commercializing GE Plants

The regulatory approvals for commercial utilization of GE products is location specific as it varies from country to country [216]. Generally, GE plants are regulated in two ways, namely product-based regulation and process-based regulation [217,218]. In the case of product-based regulation, the health and environmental risks posed by GE products are assessed by the final product of the technique and not with the methodology associated with it; whereas the process-based regulation, considers the techniques used for the development of GE crops for risk assessment. The process-based regulation restricts the commercialization of GE crops created with the aid of foreign genes even though the end products are devoid of any foreign genes [217,218]. This severely affects the commercial exploitation of GE crops and future developments in genome editing. Countries such as USA, Canada, Argentina, Australia, Russia, and Chile follow product-based regulation, whereas the European Union follows the process-based regulation [217,219]. Based on double-stranded breaks (DSBs) produced by site-directed nucleases (SDNs), the GE products are categorized into three classes, namely SDN1, SDN2 and SDN3 [219,220]. The SDN1 system repairs the DSBs through NHEJ without inserting any foreign genes and it is similar to that of natural mutations [219,221]. The SDN2 system repairs DSBs using a donor template with a few base changes and the resulting product is similar to that of SDN1 without any transgenes [219,222]. The SDN3 system inserts a gene or large DNA sequences at the DSBs, and the product is either transgenic or cisgenic based on the donor sequence [219,220,223]. SDN1 and SDN2 products are exempted from regulation in many countries, such as the USA, Canada, India, China, Russia, Switzerland, Japan, Philippines, Australia, Kenya, Nigeria, Argentina, Chile, Ecuador, Brazil, Columbia, Paraguay, Honduras and Guatemala [224]. In the European Union and New Zealand, GE crops are categorized under GMOs and in countries such as Indonesia, Bangladesh, Uruguay and Norway, commercialization of GE crops is under consideration [224]. Globally, unprecedented changes in climate and increasing demand for food highlight the need for use of potential technologies such as genome editing in the crop improvement process to meet all kinds of demands. In order to meet global food demands, the world nations that are restricting the commercialization of GE crops should consider the use of SDN1 and SDN2 products for commercial cultivation.

8. Challenges in Transgene-Free Genome Editing in Sugarcane

Transgene-free genome editing opens a new window for commercializing the GE lines of sugarcane, since eliminating transgenes, such as Cas endonuclease, promoters, marker genes and other components, from genome editing cassette in the host plant genome through genetic recombination is highly difficult. Although transgene-free genome editing techniques have numerous advantages over transgenics, many factors, such as long duration, complex genome with varying chromosome numbers ranging from 80–130, high polyploid nature with multiple alleles of a single gene, heterozygosity, and very high genome size, pose difficulties in establishing an efficient genome editing platform in sugarcane [225,226,227]. The availability of well-annotated whole genome sequence information enables the use of gRNA design tools, such as CHOP-CHOPv3, CRISPR-P 2.0 and many others, to select the gRNAs targeting the selective regions in the genome with good editing efficiency and less off-target site binding [228,229]. Unfortunately, such genome information is lacking in sugarcane and even a 1 or 2 bp mismatch between gRNA sequences and the host genome’s target region prevents the expression of the genome editing system inside the host plant. The release of the draft of the whole genome sequence of sugarcane hybrid cultivar SP80-3280 in 2017 [230], the monoploid reference genome of sugarcane cultivar R570 [231] and the allele-defined genome of a haploid Saccharum spontaneum AP85-441 [232] in 2018 provides the opportunities to exploit CRISPR/Cas technology in sugarcane for various traits. Even though the reference genome sequences are available, it should not be directly used as reference for gRNA designing. It is highly recommended to clone and analyze the sequence of the target gene from the cultivar chosen for genome editing prior to gRNA designing. This process helps avoid the nucleotide mismatches between gRNA and the target region and ensures the correct binding of gRNA in the target site. The lower transformation efficiency of sugarcane, when compared to other crops, calls for the development of efficient transformation techniques in sugarcane [226,227,233]. Despite there being many advantages in transgene-free techniques, the low editing efficiency and the lack of selection pressure for the identification of transgene-free GE plants at an early stage not only make the process more complicated, but also more expensive [95]. However, these limitations can be circumvented by using visual markers, such as pigments and fluorescent protein markers, for the identification and elimination of the transgene-containing sugarcane plants at early stages and limiting the number of plants for further sequencing and analysis [123]. The VIGE, protoplast transformation of RNPs and many other techniques show promising results in producing many transgene-free GE crops, but their use in sugarcane is yet to be explored [95,123,234].

9. Conclusions and Future Prospects

Crop improvement has seen a dramatic shift from traditional breeding methods, where the selection is purely based on the phenotype, to genome-based selection of an individual or the population. Phenotype-based methods are highly laborious and time-consuming, which requires approximately 12–15 years to develop a variety based on the duration and reproductive nature of the crop plants [225,235]. Though transgenic technology has great potential to create plants resistant/tolerant to various biotic and abiotic stresses, commercial success in food crops is very limited across the world due to the various regulatory concerns surrounding the cultivation of transgenic food crops. The advent of genome editing technologies, such as ZFNs, TALENS and CRISPR/Cas, helps to produce site-specific mutations in the plant genome within a very short span of time. Conventional plasmid-based delivery of CRISPR/Cas genome editing cassette has been successfully employed in various crop plants and has produced promising results in the creation of novel alleles, new traits/products. Nevertheless, the integration of transgenes from plasmids into the plant genome places the GE lines under GMOs [216,217]. Crop plants that reproduce through seeds such as rice, maize, tomato and many others, have the advantage of meiotic recombination to eliminate transgenes from their genomes. In the case of vegetatively propagated crops, especially sugarcane, the high heterozygosity along with the complex ploidy level pose difficulties in eliminating the transgenes through meiotic recombination. Moreover, exact genetic reconstitution is highly difficult due to the complex nature of the genome. PCR-based assays, visual marker system and TKC enable the preliminary screening of transgene-free genome edits. Employing transgene-free genome editing approaches, such as particle bombardment, Agrobacteruim-mediated transformation, VIGE, protoplast transformation and piggyBac-mediated transgenesis, along with advanced screening approaches, likely produce transgene-free GE lines resistant/tolerant to biotic and abiotic stresses.

Author Contributions

Conceptualization, S.S.K. and C.A.; methodology, S.S.K., S.R.H.C., C.A.; validation, R.V. (Ramanathan Valarmathi), K.L., M.R. and R.V. (Rasappa Viswanathan); writing—original draft preparation, S.S.K., S.R.H.C., M.R. and C.A.; writing—review and editing, R.M., M.R., S.S.K., R.V. (Ramanathan Valarmathi), G.H., P.T.P. and C.A. All authors have read and agreed to the published version of the manuscript.

Funding

Science and Engineering Research Board (SERB), Department of Science and Technology (DST)—Core Research Grant (F. No. CRG/2020/003761).

Acknowledgments

The authors thank the Director of the ICAR-Sugarcane Breeding Institute for providing facilities and support.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Solomon, S. Sugarcane by-products based industries in India. Sugar Tech 2011, 13, 408–416. [Google Scholar] [CrossRef]
  2. Statista. Available online: https://www.statista.com/statistics/495973/sugar-production-worldwide/ (accessed on 10 February 2023).
  3. UNFPA. Available online: https://pdp.unfpa.org/apps/0aeda6af00dd4544ba50452da2dda474/explore (accessed on 10 February 2023).
  4. Viswanathan, R.; Rao, G. Disease scenario and management of major sugarcane diseases in India. Sugar Tech 2011, 13, 336–353. [Google Scholar] [CrossRef]
  5. Geetha, M.; Kalyanasundaram, M.; Jayaraj, J.; Shanthi, M.; Vijayashanthi, V.; Hemalatha, D.; Karthickraja, K. Pests of sugarcane. In Pests and Their Management; Omkar, Ed.; Springer: Singapore, 2018; pp. 241–310. [Google Scholar]
  6. Suganthi, M.; Muthukrishnan, P.; Chinnusamy, C. Crop weed competition in sugarcane-a review. Agric. Rev. 2019, 40, 239–242. [Google Scholar] [CrossRef]
  7. Lingle, S.E.; Wiegand, C.L. Soil salinity and sugarcane juice quality. Field Crops Res. 1997, 54, 259–268. [Google Scholar] [CrossRef]
  8. Rietz, D.; Haynes, R. Effect of irrigation-induced salinity and sodicity on sugarcane yield. Proc. S. Afr. Sugar Technol. Assoc. 2002, 76, 173–185. [Google Scholar]
  9. Jain, R.; Shrivastava, A.; Solomon, S.; Yadav, R. Low temperature stress-induced biochemical changes affect stubble bud sprouting in sugarcane (Saccharum spp. hybrid). Plant Growth Regul. 2007, 53, 17–23. [Google Scholar] [CrossRef]
  10. Lakshmanan, P.; Robinson, N. Stress physiology: Abiotic stresses. In Sugarcane: Physiology, Biochemistry, and Functional Biology; Moore, P., Botha, F., Eds.; John Wiley & Sons Ltd.: Chichester, UK, 2013; pp. 411–434. [Google Scholar]
  11. Budeguer, F.; Enrique, R.; Perera, M.F.; Racedo, J.; Castagnaro, A.P.; Noguera, A.S.; Welin, B. Genetic transformation of sugarcane, current status and future prospects. Front. Plant Sci. 2021, 12, 2467. [Google Scholar] [CrossRef]
  12. Abdallah, N.A.; Prakash, C.S.; McHughen, A.G. Genome editing for crop improvement: Challenges and opportunities. GM Crops Food 2015, 6, 183–205. [Google Scholar] [CrossRef]
  13. Wang, C.; Shen, L.; Fu, Y.; Yan, C.; Wang, K. A simple CRISPR/Cas9 system for multiplex genome editing in rice. J. Genet. Genom. 2015, 42, 703–706. [Google Scholar] [CrossRef]
  14. Jinek, M.; Chylinski, K.; Fonfara, I.; Hauer, M.; Doudna, J.A.; Charpentier, E. A programmable dual-RNA–guided DNA endonuclease in adaptive bacterial immunity. Science 2012, 337, 816–821. [Google Scholar] [CrossRef]
  15. Jiang, W.; Yang, B.; Weeks, D.P. Efficient CRISPR/Cas9-mediated gene editing in Arabidopsis thaliana and inheritance of modified genes in the T2 and T3 generations. PLoS ONE 2014, 9, e99225. [Google Scholar] [CrossRef] [PubMed]
  16. Chapman, J.R.; Taylor, M.R.; Boulton, S.J. Playing the end game: DNA double-strand break repair pathway choice. Mol. Cell 2012, 47, 497–510. [Google Scholar] [CrossRef]
  17. Rodgers, K.; McVey, M. Error-prone repair of DNA double-strand breaks. J. Cell. Physiol. 2016, 231, 15–24. [Google Scholar] [CrossRef]
  18. Pannunzio, N.R.; Watanabe, G.; Lieber, M.R. Nonhomologous DNA end-joining for repair of DNA double-strand breaks. J. Biol. Chem. 2018, 293, 10512–10523. [Google Scholar] [CrossRef]
  19. Viswanathan, R. Fusarium diseases affecting sugarcane production in India. Indian Phytopathol. 2020, 73, 415–424. [Google Scholar] [CrossRef]
  20. Bhuiyan, S.A.; Magarey, R.C.; McNeil, M.D.; Aitken, K.S. Sugarcane smut, caused by Sporisorium scitamineum, a major disease of sugarcane: A contemporary review. Phytopathology 2021, 111, 1905–1917. [Google Scholar] [CrossRef]
  21. Comstock, J. Ratoon stunting disease. Sugar Tech 2002, 4, 1–6. [Google Scholar] [CrossRef]
  22. Hoy, J. Sugarcane leaf scald distribution, symptomatology. Plant Dis. 1994, 78, 1083–1087. [Google Scholar] [CrossRef]
  23. Lin, L.-H.; Ntambo, M.S.; Rott, P.C.; Wang, Q.-N.; Lin, Y.-H.; Fu, H.-Y.; Gao, S.-J. Molecular detection and prevalence of Xanthomonas albilineans, the causal agent of sugarcane leaf scald, in China. Crop Protect. 2018, 109, 17–23. [Google Scholar] [CrossRef]
  24. Viswanathan, R.; Chinnaraja, C.; Karuppaiah, R.; Ganesh Kumar, V.; Jenshi Rooba, J.; Malathi, P. Genetic diversity of sugarcane grassy shoot (SCGS)-phytoplasmas causing grassy shoot disease in India. Sugar Tech 2011, 13, 220–228. [Google Scholar] [CrossRef]
  25. Fitch, M.; Lehrer, A.T.; Komor, E.; Moore, P.H. Elimination of Sugarcane yellow leaf virus from infected sugarcane plants by meristem tip culture visualized by tissue blot immunoassay. Plant Pathol. 2001, 50, 676–680. [Google Scholar] [CrossRef]
  26. Viswanathan, R.; Chinnaraja, C.; Malathi, P.; Gomathi, R.; Rakkiyappan, P.; Neelamathi, D.; Ravichandran, V. Impact of Sugarcane yellow leaf virus (ScYLV) infection on physiological efficiency and growth parameters of sugarcane under tropical climatic conditions in India. Acta Physiol. Plant 2014, 36, 1805–1822. [Google Scholar] [CrossRef]
  27. Chinnaraja, C.; Viswanathan, R. Variability in yellow leaf symptom expression caused by the Sugarcane yellow leaf virus and its seasonal influence in sugarcane. Phytoparasitica 2015, 43, 339–353. [Google Scholar] [CrossRef]
  28. Comstock, J. Yellow leaf syndrome appears on the United States mainland. Sugar J. 1994, 56, 33–35. [Google Scholar]
  29. Viswanathan, R.; Balamuralikrishnan, M. Impact of mosaic infection on growth and yield of sugarcane. Sugar Tech 2005, 7, 61–65. [Google Scholar] [CrossRef]
  30. Deshumkh, G.; Sawant, D. Studies on Symptomatology of Sorghum and Sugarcane Mosaic Virus. J. Plant Dis. Sci. 2008, 3, 116–117. [Google Scholar]
  31. Lu, G.; Wang, Z.; Xu, F.; Pan, Y.-B.; Grisham, M.P.; Xu, L. Sugarcane mosaic disease: Characteristics, identification and control. Microorganisms 2021, 9, 1984. [Google Scholar] [CrossRef] [PubMed]
  32. Avasthy, P.; Tiwari, N. (Eds.) The Shoot Borer, Chilo Infuscatellus Snellen; Sugarcane Breeding Institute: Coimbatore, India, 1986; pp. 69–92.
  33. Srikanth, J.; Salin, K.; Jayanthi, R. Sugarcane Pests and Their Management; Sugarcane Breeding Institute: Coimbatore, India, 2012.
  34. Easwaramoorthy, S.; Nandagopal, V. Life tables of internode borer, Chilo sacchariphagus indicus (K.), on resistant and susceptible varieties of sugarcane. Int. J. Pest Manag. 1986, 32, 221–228. [Google Scholar] [CrossRef]
  35. Mukunthan, N. New aspects in the biology of sugarcane top borer, Scirpophaga (Nivella F.) excerptalis Wlk. Entomon 1985, 10, 235–238. [Google Scholar]
  36. Sankar, M.; Jaigeetha, S.; Rao, M.S. Exploitation of biocontrol agents, Trichogramma chilonis and Tetrastichus howardi on yield improvement in sugarcane at EID Parry (India) Ltd., Sugar Mill Command Areas. Int. J. Innov. Res. Dev. 2014, 3, 314–318. [Google Scholar]
  37. Bhavani, B.; Rao, C.V.N. Management of sugarcane white fly (Aleurolobus barodensis Mask.) in North coastal districts of Andhra Pradesh, India. Int. J. Social Sci. Interdis. Res. 2013, 2, 112–115. [Google Scholar]
  38. Askarianzadeh, A.; Minaeimoghadam, M. Biology, natural enemies and damage of the sugarcane whitefly (Neomaskellia andropogonis)(Homoptera: Aleyrodidae) in Iran. Int. J. Trop. Insect Sci. 2018, 38, 381–386. [Google Scholar] [CrossRef]
  39. Sosa Jr, O. Effect of white grub (Coleoptera: Scarabaeidae) infestation on sugarcane yields. J. Econ. Entomol. 1984, 77, 183–185. [Google Scholar] [CrossRef]
  40. Liu, X.; Zhu, X.; Pan, Y.; Li, S.; Liu, Y.; Ma, Y. Agricultural drought monitoring: Progress, challenges, and prospects. J. Geogr. Sci. 2016, 26, 750–767. [Google Scholar] [CrossRef]
  41. Mall, A.; Misra, V.; Pathak, A.; Srivastava, S. Breeding for Drought Tolerance in Sugarcane: Indian Perspective. Sugar Tech 2022, 24, 1625–1635. [Google Scholar] [CrossRef]
  42. Narwade, A.V.; Bhagat, K.; Patil, D.; Singh, Y.; Kumari, A.; Ban, Y.; Thakare, H.; Singh, C. Abiotic stress responses in sugarcane. In Challenges and Prospective of Plant Abiotic Stress; Pasala, R., Ed.; Today & Tomorrow’s Printers and Publishers: Delhi, India, 2016; pp. 419–446. [Google Scholar]
  43. Ferreira, T.H.; Tsunada, M.S.; Bassi, D.; Araújo, P.; Mattiello, L.; Guidelli, G.V.; Righetto, G.L.; Gonçalves, V.R.; Lakshmanan, P.; Menossi, M. Sugarcane water stress tolerance mechanisms and its implications on developing biotechnology solutions. Front. Plant Sci. 2017, 8, 1077. [Google Scholar] [CrossRef]
  44. Inman-Bamber, N.; Lakshmanan, P.; Park, S. Sugarcane for water-limited environments: Theoretical assessment of suitable traits. Field Crops Res. 2012, 134, 95–104. [Google Scholar] [CrossRef]
  45. Sage, R.F.; Peixoto, M.M.; Sage, T.L. Photosynthesis in sugarcane. In Sugarcane: Physiology, Biochemistry, and Functional Biology; Moore, P., Botha, F., Eds.; John Wiley & Sons Ltd.: Chichester, UK, 2013; pp. 121–154. [Google Scholar]
  46. Ogbaga, C.C.; Amir, M.; Bano, H.; Chater, C.C.; Jellason, N.P. Clarity on frequently asked questions about drought measurements in plant physiology. Sci. Afr. 2020, 8, e00405. [Google Scholar] [CrossRef]
  47. Rai, M.K.; Kalia, R.K.; Singh, R.; Gangola, M.P.; Dhawan, A. Developing stress tolerant plants through in vitro selection—an overview of the recent progress. Environ. Exp. Bot. 2011, 71, 89–98. [Google Scholar] [CrossRef]
  48. Huang, D.; Wu, W.; Abrams, S.R.; Cutler, A.J. The relationship of drought-related gene expression in Arabidopsis thaliana to hormonal and environmental factors. J. Exp. Bot. 2008, 59, 2991–3007. [Google Scholar] [CrossRef]
  49. Azevedo, R.A.; Carvalho, R.F.; Cia, M.C.; Gratão, P.L. Sugarcane under pressure: An overview of biochemical and physiological studies of abiotic stress. Trop. Plant Biol. 2011, 4, 42–51. [Google Scholar] [CrossRef]
  50. Patade, V.Y.; Suprasanna, P.; Bapat, V.A. Effects of salt stress in relation to osmotic adjustment on sugarcane (Saccharum officinarum L.) callus cultures. Plant Growth Regul. 2008, 55, 169–173. [Google Scholar] [CrossRef]
  51. Sengar, K.; Sengar, R.; Singh, A. Biotechnological and genomic analysis for salinity tolerance in sugarcane. Int. J. Biotechnol. Bioeng. Res. 2013, 4, 407–414. [Google Scholar]
  52. Polash, M.A.S.; Sakil, M.A.; Hossain, M.A. Plants responses and their physiological and biochemical defense mechanisms against salinity: A review. Trop. Plant Res. 2019, 6, 250–274. [Google Scholar] [CrossRef]
  53. Meena, M.R.; Kumar, R.; Chinnaswamy, A.; Karuppaiyan, R.; Kulshreshtha, N.; Ram, B. Current breeding and genomic approaches to enhance the cane and sugar productivity under abiotic stress conditions. 3 Biotech 2020, 10, 440. [Google Scholar] [CrossRef]
  54. Das, K.; Roychoudhury, A. Reactive oxygen species (ROS) and response of antioxidants as ROS-scavengers during environmental stress in plants. Front. Environ. Sci. 2014, 2, 53. [Google Scholar] [CrossRef]
  55. Rehman, S.; Muhammad, K.; Novaes, E.; Que, Y.; Din, A.; Islam, M.; Porto, A.; Sajid, M.; Ullah, N.; Iqsa, S. Expression analysis of transcription factors in sugarcane during cold stress. Braz. J. Biol. 2021, 83, e242603. [Google Scholar] [CrossRef]
  56. Hussain, S.; Khaliq, A.; Mehmood, U.; Qadir, T.; Saqib, M.; Iqbal, M.A.; Hussain, S. Sugarcane production under changing climate: Effects of environmental vulnerabilities on sugarcane diseases, insects and weeds. In Sugarcane Production-Agronomic, Scientific and Industrial Perspectives; IntechOpen: Rijeka, Croatia, 2018; pp. 1–17. [Google Scholar]
  57. Gomathi, R.; Chandran, K.; Gururaja Rao, P.; Rakkiyappan, P. Effect of Waterlogging in Sugarcane and Its Management; Sugarcane Breeding Institute: Coimbatore, India, 2010.
  58. Gomathi, R.; Gururaja Rao, P.; Chandran, K.; Selvi, A. Adaptive responses of sugarcane to waterlogging stress: An over view. Sugar Tech 2015, 17, 325–338. [Google Scholar] [CrossRef]
  59. Riyazuddin, R.; Nisha, N.; Ejaz, B.; Khan, M.I.R.; Kumar, M.; Ramteke, P.W.; Gupta, R. A comprehensive review on the heavy metal toxicity and sequestration in plants. Biomolecules 2022, 12, 43. [Google Scholar] [CrossRef]
  60. Kumar, A.; Aery, N. Impact, metabolism, and toxicity of heavy metals in plants. In Plant Responses to Xenobiotics; Singh, A., Prasad, S.M., Singh, R.P., Eds.; Springer: Singapore, 2016; pp. 141–176. [Google Scholar]
  61. Tiwari, S.; Lata, C. Heavy metal stress, signaling, and tolerance due to plant-associated microbes: An overview. Front. Plant Sci. 2018, 9, 452. [Google Scholar] [CrossRef] [PubMed]
  62. Baig, M.A.; Qamar, S.; Ali, A.A.; Ahmad, J.; Qureshi, M.I. Heavy metal toxicity and tolerance in crop plants. In Contaminants in Agriculture: Sources, Impacts and Management; Naeem, M., Ansari, A.A., Gill, S.S., Eds.; Springer: Cham, Switzerland, 2020; pp. 201–216. [Google Scholar]
  63. Hassan, T.U.; Bano, A.; Naz, I. Alleviation of heavy metals toxicity by the application of plant growth promoting rhizobacteria and effects on wheat grown in saline sodic field. Int. J. Phytoremediation 2017, 19, 522–529. [Google Scholar] [CrossRef]
  64. Panuccio, M.R.; Sorgonà, A.; Rizzo, M.; Cacco, G. Cadmium adsorption on vermiculite, zeolite and pumice: Batch experimental studies. J. Environ. Manag. 2009, 90, 364–374. [Google Scholar] [CrossRef]
  65. Zhang, C.; Liu, S.; Li, X.; Zhang, R.; Li, J. Virus-induced gene editing and its applications in plants. Int. J. Mol. Sci. 2022, 23, 10202. [Google Scholar] [CrossRef] [PubMed]
  66. Baltes, N.J.; Gil-Humanes, J.; Cermak, T.; Atkins, P.A.; Voytas, D.F. DNA replicons for plant genome engineering. Plant Cell 2014, 26, 151–163. [Google Scholar] [CrossRef]
  67. Čermák, T.; Baltes, N.J.; Čegan, R.; Zhang, Y.; Voytas, D.F. High-frequency, precise modification of the tomato genome. Genome Biol. 2015, 16, 232. [Google Scholar] [CrossRef]
  68. Butler, N.M.; Atkins, P.A.; Voytas, D.F.; Douches, D.S. Generation and inheritance of targeted mutations in potato (Solanum tuberosum L.) using the CRISPR/Cas system. PLoS ONE 2015, 10, e0144591. [Google Scholar] [CrossRef]
  69. Dahan-Meir, T.; Filler-Hayut, S.; Melamed-Bessudo, C.; Bocobza, S.; Czosnek, H.; Aharoni, A.; Levy, A.A. Efficient in planta gene targeting in tomato using geminiviral replicons and the CRISPR/Cas9 system. Plant J. 2018, 95, 5–16. [Google Scholar] [CrossRef] [PubMed]
  70. Wang, M.; Lu, Y.; Botella, J.R.; Mao, Y.; Hua, K.; Zhu, J.-k. Gene targeting by homology-directed repair in rice using a geminivirus-based CRISPR/Cas9 system. Mol Plant. 2017, 10, 1007–1010. [Google Scholar] [CrossRef]
  71. Gil-Humanes, J.; Wang, Y.; Liang, Z.; Shan, Q.; Ozuna, C.V.; Sánchez-León, S.; Baltes, N.J.; Starker, C.; Barro, F.; Gao, C. High-efficiency gene targeting in hexaploid wheat using DNA replicons and CRISPR/Cas9. Plant J. 2017, 89, 1251–1262. [Google Scholar] [CrossRef] [PubMed]
  72. Yin, K.; Han, T.; Liu, G.; Chen, T.; Wang, Y.; Yu, A.Y.L.; Liu, Y. A geminivirus-based guide RNA delivery system for CRISPR/Cas9 mediated plant genome editing. Sci. Rep. 2015, 5, 14926. [Google Scholar] [CrossRef]
  73. Lei, J.; Dai, P.; Li, Y.; Zhang, W.; Zhou, G.; Liu, C.; Liu, X. Heritable gene editing using FT mobile guide RNAs and DNA viruses. Plant Methods 2021, 17, 20. [Google Scholar] [CrossRef]
  74. Cody, W.B.; Scholthof, H.B.; Mirkov, T.E. Multiplexed gene editing and protein overexpression using a tobacco mosaic virus viral vector. Plant Physiol. 2017, 175, 23–35. [Google Scholar] [CrossRef]
  75. Hu, J.; Li, S.; Li, Z.; Li, H.; Song, W.; Zhao, H.; Lai, J.; Xia, L.; Li, D.; Zhang, Y. A barley stripe mosaic virus-based guide RNA delivery system for targeted mutagenesis in wheat and maize. Mol. Plant Pathol. 2019, 20, 1463–1474. [Google Scholar] [CrossRef]
  76. Chiong, K.T.; Cody, W.B.; Scholthof, H.B. RNA silencing suppressor-influenced performance of a virus vector delivering both guide RNA and Cas9 for CRISPR gene editing. Sci. Rep. 2021, 11, 6769. [Google Scholar] [CrossRef]
  77. Ali, Z.; Eid, A.; Ali, S.; Mahfouz, M.M. Pea early-browning virus-mediated genome editing via the CRISPR/Cas9 system in Nicotiana benthamiana and Arabidopsis. Virus Res. 2018, 244, 333–337. [Google Scholar] [CrossRef]
  78. Jiang, N.; Zhang, C.; Liu, J.Y.; Guo, Z.H.; Zhang, Z.Y.; Han, C.G.; Wang, Y. Development of Beet necrotic yellow vein virus-based vectors for multiple-gene expression and guide RNA delivery in plant genome editing. Plant Biotechnol. J. 2019, 17, 1302–1315. [Google Scholar] [CrossRef]
  79. Mei, Y.; Beernink, B.M.; Ellison, E.E.; Konečná, E.; Neelakandan, A.K.; Voytas, D.F.; Whitham, S.A. Protein expression and gene editing in monocots using foxtail mosaic virus vectors. Plant Direct 2019, 3, e00181. [Google Scholar] [CrossRef]
  80. Zhang, X.; Kang, L.; Zhang, Q.; Meng, Q.; Pan, Y.; Yu, Z.; Shi, N.; Jackson, S.; Zhang, X.; Wang, H. An RNAi suppressor activates in planta virus–mediated gene editing. Funct. Integr. Genom. 2020, 20, 471–477. [Google Scholar] [CrossRef]
  81. Ariga, H.; Toki, S.; Ishibashi, K. Potato virus X vector-mediated DNA-free genome editing in plants. Plant Cell Physiol. 2020, 61, 1946–1953. [Google Scholar] [CrossRef]
  82. Uranga, M.; Aragonés, V.; Selma, S.; Vázquez-Vilar, M.; Orzáez, D.; Daròs, J.A. Efficient Cas9 multiplex editing using unspaced sgRNA arrays engineering in a Potato virus X vector. Plant J. 2021, 106, 555–565. [Google Scholar] [CrossRef]
  83. Uranga, M.; Vazquez-Vilar, M.; Orzáez, D.; Daròs, J.-A. CRISPR-Cas12a genome editing at the whole-plant level using two compatible RNA virus vectors. CRISPR J. 2021, 4, 761–769. [Google Scholar] [CrossRef]
  84. Calvache, C.; Vazquez-Vilar, M.; Selma, S.; Uranga, M.; Fernández-del-Carmen, A.; Daròs, J.A.; Orzáez, D. Strong and tunable anti-CRISPR/Cas activities in plants. Plant Biotechnol. J. 2022, 20, 399–408. [Google Scholar] [CrossRef]
  85. Gao, Q.; Xu, W.Y.; Yan, T.; Fang, X.D.; Cao, Q.; Zhang, Z.J.; Ding, Z.H.; Wang, Y.; Wang, X.B. Rescue of a plant cytorhabdovirus as versatile expression platforms for planthopper and cereal genomic studies. New Phytol. 2019, 223, 2120–2133. [Google Scholar] [CrossRef]
  86. Ma, X.; Zhang, X.; Liu, H.; Li, Z. Highly efficient DNA-free plant genome editing using virally delivered CRISPR–Cas9. Nat. Plants 2020, 6, 773–779. [Google Scholar] [CrossRef]
  87. Liu, J.-J.; Orlova, N.; Oakes, B.L.; Ma, E.; Spinner, H.B.; Baney, K.L.; Chuck, J.; Tan, D.; Knott, G.J.; Harrington, L.B. CasX enzymes comprise a distinct family of RNA-guided genome editors. Nature 2019, 566, 218–223. [Google Scholar] [CrossRef]
  88. Pausch, P.; Al-Shayeb, B.; Bisom-Rapp, E.; Tsuchida, C.A.; Li, Z.; Cress, B.F.; Knott, G.J.; Jacobsen, S.E.; Banfield, J.F.; Doudna, J.A. CRISPR-CasΦ from huge phages is a hypercompact genome editor. Science 2020, 369, 333–337. [Google Scholar] [CrossRef]
  89. Uranga, M.; Daròs, J.A. Tools and targets: The dual role of plant viruses in CRISPR–Cas genome editing. Plant Genome 2022, e20220. [Google Scholar] [CrossRef]
  90. Pereira, A.R.; Barbieri, V.; Nova, N.A.V. Climatic conditioning of flowering induction in sugarcane. Agric. Meteorol. 1983, 29, 103–110. [Google Scholar] [CrossRef]
  91. Moore, P.H.; Nuss, K. Flowering and flower synchronization. In Sugarcane Improvement Through Breeding; Heinz, D.J., Ed.; Elsevier Science: Amsterdam, The Netherlands, 1987; Volume 11, pp. 273–311. [Google Scholar]
  92. Ellison, E.E.; Nagalakshmi, U.; Gamo, M.E.; Huang, P.-j.; Dinesh-Kumar, S.; Voytas, D.F. Multiplexed heritable gene editing using RNA viruses and mobile single guide RNAs. Nat. Plants 2020, 6, 620–624. [Google Scholar] [CrossRef]
  93. Mohan, C.; Ashwin Narayan, J.; Esterling, M.; Yau, Y.-Y. Current transformation methods for genome–editing applications in energy crop sugarcane. In Climate Change, Photosynthesis and Advanced Biofuels: The Role of Biotechnology in the Production of Value-added Plant Bio-products; Kumar, A., Yau, Y., Ogita, S., Scheibe, R., Eds.; Springer: Singapore, 2020; pp. 369–388. [Google Scholar]
  94. Laforest, L.C.; Nadakuduti, S.S. Advances in delivery mechanisms of CRISPR gene-editing reagents in plants. Front. Genome Ed. 2022, 4, 1. [Google Scholar] [CrossRef]
  95. Tsanova, T.; Stefanova, L.; Topalova, L.; Atanasov, A.; Pantchev, I. DNA-free gene editing in plants: A brief overview. Biotechnol. Biotechnol. Equip. 2021, 35, 131–138. [Google Scholar] [CrossRef]
  96. Svitashev, S.; Schwartz, C.; Lenderts, B.; Young, J.; Cigan, M. Genome editing in maize directed by CRISPR-Cas9 ribonucleoprotein complexes. Nat. Commun. 2016, 7, 13274. [Google Scholar] [CrossRef]
  97. Banakar, R.; Eggenberger, A.L.; Lee, K.; Wright, D.A.; Murugan, K.; Zarecor, S.; Lawrence-Dill, C.J.; Sashital, D.G.; Wang, K. High-frequency random DNA insertions upon co-delivery of CRISPR-Cas9 ribonucleoprotein and selectable marker plasmid in rice. Sci. Rep. 2019, 9, 19902. [Google Scholar] [CrossRef] [PubMed]
  98. Banakar, R.; Schubert, M.; Collingwood, M.; Vakulskas, C.; Eggenberger, A.L.; Wang, K. Comparison of CRISPR-Cas9/Cas12a ribonucleoprotein complexes for genome editing efficiency in the rice phytoene desaturase (OsPDS) gene. Rice 2020, 13, 4. [Google Scholar] [CrossRef] [PubMed]
  99. Liang, Z.; Chen, K.; Zhang, Y.; Liu, J.; Yin, K.; Qiu, J.-L.; Gao, C. Genome editing of bread wheat using biolistic delivery of CRISPR/Cas9 in vitro transcripts or ribonucleoproteins. Nat. Protoc. 2018, 13, 413–430. [Google Scholar] [CrossRef] [PubMed]
  100. Oz, M.T.; Altpeter, A.; Karan, R.; Merotto, A.; Altpeter, F. CRISPR/Cas9-mediated multi-allelic gene targeting in sugarcane confers herbicide tolerance. Front. Genome Ed. 2021, 3, 673566. [Google Scholar] [CrossRef]
  101. Eid, A.; Mohan, C.; Sanchez, S.; Wang, D.; Altpeter, F. Multiallelic, targeted mutagenesis of magnesium chelatase with CRISPR/Cas9 provides a rapidly scorable phenotype in highly polyploid sugarcane. Front. Genome Ed. 2021, 3, 654996. [Google Scholar] [CrossRef] [PubMed]
  102. Zhang, J.; Xing, J.; Mi, Q.; Yang, W.; Xiang, H.; Xu, L.; Zeng, W.; Wang, J.; Deng, L.; Jiang, J. Highly efficient transgene-free genome editing in tobacco using an optimized CRISPR/Cas9 system, pOREU3TR. Plant Sci. 2023, 326, 111523. [Google Scholar] [CrossRef]
  103. Chen, L.; Li, W.; Katin-Grazzini, L.; Ding, J.; Gu, X.; Li, Y.; Gu, T.; Wang, R.; Lin, X.; Deng, Z. A method for the production and expedient screening of CRISPR/Cas9-mediated non-transgenic mutant plants. Hortic. Res. 2018, 5, 13. [Google Scholar] [CrossRef]
  104. Veillet, F.; Perrot, L.; Chauvin, L.; Kermarrec, M.-P.; Guyon-Debast, A.; Chauvin, J.-E.; Nogué, F.; Mazier, M. Transgene-free genome editing in tomato and potato plants using agrobacterium-mediated delivery of a CRISPR/Cas9 cytidine base editor. Int. J. Mol. Sci. 2019, 20, 402. [Google Scholar] [CrossRef] [PubMed]
  105. Danilo, B.; Perrot, L.; Mara, K.; Botton, E.; Nogué, F.; Mazier, M. Efficient and transgene-free gene targeting using Agrobacterium-mediated delivery of the CRISPR/Cas9 system in tomato. Plant Cell Rep. 2019, 38, 459–462. [Google Scholar] [CrossRef] [PubMed]
  106. Bánfalvi, Z.; Csákvári, E.; Villányi, V.; Kondrák, M. Generation of transgene-free PDS mutants in potato by Agrobacterium-mediated transformation. BMC Biotechnol. 2020, 20, 25. [Google Scholar] [CrossRef] [PubMed]
  107. Mayavan, S.; Subramanyam, K.; Arun, M.; Rajesh, M.; Kapil Dev, G.; Sivanandhan, G.; Jaganath, B.; Manickavasagam, M.; Selvaraj, N.; Ganapathi, A. Agrobacterium tumefaciens-mediated in planta seed transformation strategy in sugarcane. Plant Cell Rep. 2013, 32, 1557–1574. [Google Scholar] [CrossRef] [PubMed]
  108. Mayavan, S.; Subramanyam, K.; Jaganath, B.; Sathish, D.; Manickavasagam, M.; Ganapathi, A. Agrobacterium-mediated in planta genetic transformation of sugarcane setts. Plant Cell Rep. 2015, 34, 1835–1848. [Google Scholar] [CrossRef]
  109. Murovec, J.; Guček, K.; Bohanec, B.; Avbelj, M.; Jerala, R. DNA-free genome editing of Brassica oleracea and B. rapa protoplasts using CRISPR-Cas9 ribonucleoprotein complexes. Front. Plant Sci. 2018, 9, 1594. [Google Scholar] [CrossRef]
  110. Liu, W.; Rudis, M.R.; Cheplick, M.H.; Millwood, R.J.; Yang, J.-P.; Ondzighi-Assoume, C.A.; Montgomery, G.A.; Burris, K.P.; Mazarei, M.; Chesnut, J.D. Lipofection-mediated genome editing using DNA-free delivery of the Cas9/gRNA ribonucleoprotein into plant cells. Plant Cell Rep. 2020, 39, 245–257. [Google Scholar] [CrossRef]
  111. Sant’Ana, R.R.A.; Caprestano, C.A.; Nodari, R.O.; Agapito-Tenfen, S.Z. PEG-delivered CRISPR-Cas9 ribonucleoproteins system for gene-editing screening of maize protoplasts. Genes 2020, 11, 1029. [Google Scholar] [CrossRef]
  112. Toda, E.; Koiso, N.; Takebayashi, A.; Ichikawa, M.; Kiba, T.; Osakabe, K.; Osakabe, Y.; Sakakibara, H.; Kato, N.; Okamoto, T. An efficient DNA-and selectable-marker-free genome-editing system using zygotes in rice. Nat. Plants 2019, 5, 363–368. [Google Scholar] [CrossRef]
  113. Wu, S.; Zhu, H.; Liu, J.; Yang, Q.; Shao, X.; Bi, F.; Hu, C.; Huo, H.; Chen, K.; Yi, G. Establishment of a PEG-mediated protoplast transformation system based on DNA and CRISPR/Cas9 ribonucleoprotein complexes for banana. BMC Plant Biol. 2020, 20, 425. [Google Scholar] [CrossRef]
  114. Sidorov, V.; Wang, D.; Nagy, E.D.; Armstrong, C.; Beach, S.; Zhang, Y.; Groat, J.; Yang, S.; Yang, P.; Gilbertson, L. Heritable DNA-free genome editing of canola (Brassica napus L.) using PEG-mediated transfection of isolated protoplasts. In Vitro Cell. Dev. Biol. Plant 2022, 58, 447–456. [Google Scholar] [CrossRef]
  115. Lin, C.-S.; Hsu, C.-T.; Yuan, Y.-H.; Zheng, P.-X.; Wu, F.-H.; Cheng, Q.-W.; Wu, Y.-L.; Wu, T.-L.; Lin, S.; Yue, J.-J. DNA-free CRISPR-Cas9 gene editing of wild tetraploid tomato Solanum peruvianum using protoplast regeneration. Plant Physiol. 2022, 188, 1917–1930. [Google Scholar] [CrossRef] [PubMed]
  116. Andersson, M.; Turesson, H.; Olsson, N.; Fält, A.S.; Ohlsson, P.; Gonzalez, M.N.; Samuelsson, M.; Hofvander, P. Genome editing in potato via CRISPR-Cas9 ribonucleoprotein delivery. Physiol. Plant. 2018, 164, 378–384. [Google Scholar] [CrossRef] [PubMed]
  117. Chen, W.; Gartland, K.; Davey, M.; Sotak, R.; Gartland, J.; Mulligan, B.; Power, J.; Cocking, E. Transformation of sugarcane protoplasts by direct uptake of a selectable chimaeric gene. Plant Cell Rep. 1987, 6, 297–301. [Google Scholar] [CrossRef]
  118. Rathus, C.; Birch, R.G. Stable transformation of callus from electroporated sugarcane protoplasts. Plant Sci. 1992, 82, 81–89. [Google Scholar] [CrossRef]
  119. Chowdhury, M.; Vasil, I.K. Stably transformed herbicide resistant callus of sugarcane via microprojectile bombardment of cell suspension cultures and electroporation of protoplasts. Plant Cell Rep. 1992, 11, 494–498. [Google Scholar] [CrossRef]
  120. Arencibia, A.; Vázquez, R.I.; Prieto, D.; Téllez, P.; Carmona, E.R.; Coego, A.; Hernández, L.; De la Riva, G.A.; Selman-Housein, G. Transgenic sugarcane plants resistant to stem borer attack. Mol. Breed. 1997, 3, 247–255. [Google Scholar] [CrossRef]
  121. Wang, Q.; Yu, G.; Chen, Z.; Han, J.; Hu, Y.; Wang, K. Optimization of protoplast isolation, transformation and its application in sugarcane (Saccharum spontaneum L). Crop J. 2021, 9, 133–142. [Google Scholar] [CrossRef]
  122. Nishizawa-Yokoi, A.; Toki, S. A piggyBac-mediated transgenesis system for the temporary expression of CRISPR/Cas9 in rice. Plant Biotechnol. J. 2021, 19, 1386–1395. [Google Scholar] [CrossRef]
  123. He, Y.; Mudgett, M.; Zhao, Y. Advances in gene editing without residual transgenes in plants. Plant Physiol. 2022, 188, 1757–1768. [Google Scholar] [CrossRef]
  124. Yang, L.; Machin, F.; Wang, S.; Saplaoura, E.; Kragler, F. Heritable transgene-free genome editing in plants by grafting of wild-type shoots to transgenic donor rootstocks. Nat. Biotechnol. 2023, 1–10. [Google Scholar] [CrossRef]
  125. Melnyk, C.W.; Meyerowitz, E.M. Plant grafting. Curr. Biol. 2015, 25, R183–R188. [Google Scholar] [CrossRef] [PubMed]
  126. He, Y.; Zhu, M.; Wang, L.; Wu, J.; Wang, Q.; Wang, R.; Zhao, Y. Programmed self-elimination of the CRISPR/Cas9 construct greatly accelerates the isolation of edited and transgene-free rice plants. Mol Plant. 2018, 11, 1210–1213. [Google Scholar] [CrossRef]
  127. Yubing, H.; Min, Z.; Lihao, W.; Junhua, W.; Qiaoyan, W.; Rongchen, W.; Yunde, Z. Improvements of TKC technology accelerate isolation of transgene-free CRISPR/Cas9-edited rice plants. Rice Sci. 2019, 26, 109–117. [Google Scholar] [CrossRef]
  128. Yang, N.; Yan, L.; Zheng, Z.; Zhang, Y.; Zhan, H.; Tian, Y.; Zhang, T.; Li, R.; Gong, X.; Xu, M. Editing gene families by CRISPR/Cas9: Accelerating the isolation of multiple transgene-free null mutant combinations with much reduced labor-intensive analysis. Plant Biotechnol. J. 2022, 20, 241. [Google Scholar] [CrossRef] [PubMed]
  129. Gao, X.; Chen, J.; Dai, X.; Zhang, D.; Zhao, Y. An effective strategy for reliably isolating heritable and Cas9-free Arabidopsis mutants generated by CRISPR/Cas9-mediated genome editing. Plant Physiol. 2016, 171, 1794–1800. [Google Scholar] [CrossRef]
  130. He, Y.; Wang, R.; Dai, X.; Zhao, Y. On improving CRISPR for editing plant genes: Ribozyme-mediated guide RNA production and fluorescence-based technology for isolating transgene-free mutants generated by CRISPR. Prog. Mol. Biol. Transl. Sci. 2017, 149, 151–166. [Google Scholar]
  131. Aliaga-Franco, N.; Zhang, C.; Presa, S.; Srivastava, A.K.; Granell, A.; Alabadí, D.; Sadanandom, A.; Blázquez, M.A.; Minguet, E.G. Identification of transgene-free CRISPR-edited plants of rice, tomato, and Arabidopsis by monitoring DsRED fluorescence in dry seeds. Front. Plant Sci. 2019, 10, 1150. [Google Scholar] [CrossRef]
  132. Yan, Y.; Zhu, J.; Qi, X.; Cheng, B.; Liu, C.; Xie, C. Establishment of an efficient seed fluorescence reporter-assisted CRISPR/Cas9 gene editing in maize. J. Integr. Plant Biol. 2021, 63, 1671–1680. [Google Scholar] [CrossRef]
  133. Wang, J.; Chen, H. A novel CRISPR/Cas9 system for efficiently generating Cas9-free multiplex mutants in Arabidopsis. Abiotech 2020, 1, 6–14. [Google Scholar] [CrossRef]
  134. Liu, Y.; Zeng, J.; Yuan, C.; Guo, Y.; Yu, H.; Li, Y.; Huang, C. Cas9-PF, an early flowering and visual selection marker system, enhances the frequency of editing event occurrence and expedites the isolation of genome-edited and transgene-free plants. Plant Biotechnol. J. 2019, 17, 1191. [Google Scholar] [CrossRef]
  135. He, Y.; Zhu, M.; Wu, J.; Ouyang, L.; Wang, R.; Sun, H.; Yan, L.; Wang, L.; Xu, M.; Zhan, H. Repurposing of anthocyanin biosynthesis for plant transformation and genome editing. Front. Genome Ed. 2020, 2, 607982. [Google Scholar] [CrossRef] [PubMed]
  136. Peng, C.; Zheng, M.; Ding, L.; Chen, X.; Wang, X.; Feng, X.; Wang, J.; Xu, J. Accurate detection and evaluation of the gene-editing frequency in plants using droplet digital PCR. Front. Plant Sci. 2020, 11, 610790. [Google Scholar] [CrossRef] [PubMed]
  137. Zhang, Y.; Chi, X.; Feng, L.; Wu, X.; Qi, X. Improvement of multiplex semi-nested PCR system for screening of rare mutations by high-throughput sequencing. BioTechniques 2019, 67, 294–298. [Google Scholar] [CrossRef]
  138. Zhang, B.; Zhou, J.; Li, M.; Wei, Y.; Wang, J.; Wang, Y.; Shi, P.; Li, X.; Huang, Z.; Tang, H. Evaluation of CRISPR/Cas9 site-specific function and validation of sgRNA sequence by a Cas9/sgRNA-assisted reverse PCR technique. Anal. Bioanal. Chem. 2021, 413, 2447–2456. [Google Scholar] [CrossRef]
  139. Kamiya, Y.; Abe, F.; Mikami, M.; Endo, M.; Kawaura, K. A rapid method for detection of mutations induced by CRISPR/Cas9-based genome editing in common wheat. Plant Biotechnol. 2020, 37, 247–251. [Google Scholar] [CrossRef]
  140. Ueta, R.; Abe, C.; Watanabe, T.; Sugano, S.S.; Ishihara, R.; Ezura, H.; Osakabe, Y.; Osakabe, K. Rapid breeding of parthenocarpic tomato plants using CRISPR/Cas9. Sci. Rep. 2017, 7, 507. [Google Scholar] [CrossRef]
  141. Kim, D.; Alptekin, B.; Budak, H. CRISPR/Cas9 genome editing in wheat. Funct. Integr. Genom. 2018, 18, 31–41. [Google Scholar] [CrossRef] [PubMed]
  142. Sentmanat, M.F.; Peters, S.T.; Florian, C.P.; Connelly, J.P.; Pruett-Miller, S.M. A survey of validation strategies for CRISPR-Cas9 editing. Sci. Rep. 2018, 8, 888. [Google Scholar] [CrossRef] [PubMed]
  143. Zong, Y.; Wang, Y.; Li, C.; Zhang, R.; Chen, K.; Ran, Y.; Qiu, J.-L.; Wang, D.; Gao, C. Precise base editing in rice, wheat and maize with a Cas9-cytidine deaminase fusion. Nat. Biotechnol. 2017, 35, 438–440. [Google Scholar] [CrossRef]
  144. Pan, C.; Ye, L.; Qin, L.; Liu, X.; He, Y.; Wang, J.; Chen, L.; Lu, G. CRISPR/Cas9-mediated efficient and heritable targeted mutagenesis in tomato plants in the first and later generations. Sci. Rep. 2016, 6, 24765. [Google Scholar] [CrossRef]
  145. Li, M.; Liang, Z.; Zeng, Y.; Jing, Y.; Wu, K.; Liang, J.; He, S.; Wang, G.; Mo, Z.; Tan, F. De novo analysis of transcriptome reveals genes associated with leaf abscission in sugarcane (Saccharum officinarum L.). BMC Genom. 2016, 17, 195. [Google Scholar] [CrossRef] [PubMed]
  146. Wanderley-Nogueira, A.; Soares-Cavalcanti, N.; Morais, D.; Belarmino, L.; Barbosa Da Silva, A.; Benko-Iseppon, A. Abundance and diversity of resistance genes in the sugarcane transcriptome revealed by in silico analysis. Gen. Mol. Res. 2007, 6, 866–889. [Google Scholar]
  147. Sharma, R.; Tamta, S. Red rot resistant gene characterization using RGAP markers among sugarcane cultivars resistant and susceptible to the red rot disease. 3 Biotech 2017, 7, 306. [Google Scholar] [CrossRef]
  148. Raju, G.; Shanmugam, K.; Kasirajan, L. High-throughput sequencing reveals genes associated with high-temperature stress tolerance in sugarcane. 3 Biotech 2020, 10, 198. [Google Scholar] [CrossRef] [PubMed]
  149. Sun, T.; Chen, Y.; Feng, A.; Zou, W.; Wang, D.; Lin, P.; Chen, Y.; You, C.; Que, Y.; Su, Y. The allene oxide synthase gene family in sugarcane and its involvement in disease resistance. Ind. Crops Prod. 2023, 192, 116136. [Google Scholar] [CrossRef]
  150. Zhu, K.; Huang, C.; Phan, T.-T.; Yang, L.-T.; Zhang, B.-Q.; Xing, Y.-X.; Li, Y.-R. Overexpression of SoACLA-1 gene confers drought tolerance improvement in sugarcane. Plant Mol. Biol. Rep. 2021, 39, 489–500. [Google Scholar] [CrossRef]
  151. Li, J.; Phan, T.-T.; Li, Y.-R.; Xing, Y.-X.; Yang, L.-T. Isolation, transformation and overexpression of sugarcane SoP5CS gene for drought tolerance improvement. Sugar Tech 2018, 20, 464–473. [Google Scholar] [CrossRef]
  152. Ferreira, T.H.; Gentile, A.; Vilela, R.D.; Costa, G.G.L.; Dias, L.I.; Endres, L.; Menossi, M. microRNAs associated with drought response in the bioenergy crop sugarcane (Saccharum spp.). PLoS ONE 2012, 7, e46703. [Google Scholar] [CrossRef]
  153. Lin, S.; Chen, T.; Qin, X.; Wu, H.; Khan, M.A.; Lin, W. Identification of microrna families expressed in sugarcane leaves subjected to drought stress and the targets thereof. Pak. J. Agric. Sci. 2014, 51, 925–934. [Google Scholar]
  154. Andrade, L.M.; Benatti, T.R.; Nobile, P.M.; Goldman, M.H.; Figueira, A.; Marin, A.L.A.; dos Santos Brito, M.; da Silva, J.; Creste, S. Characterization, isolation and cloning of sugarcane genes related to drought stress. BMC Proceedings BioMed Cent. 2014, 8, 110. [Google Scholar]
  155. Carnavale Bottino, M.; Rosario, S.; Grativol, C.; Thiebaut, F.; Rojas, C.A.; Farrineli, L.; Hemerly, A.S.; Ferreira, P.C.G. High-throughput sequencing of small RNA transcriptome reveals salt stress regulated microRNAs in sugarcane. PLoS ONE 2013, 8, e59423. [Google Scholar] [CrossRef]
  156. Murugan, N.; Palanisamy, V.; Channappa, M.; Ramanathan, V.; Ramaswamy, M.; Govindakurup, H.; Chinnaswamy, A. Genome-Wide In Silico Identification, Structural Analysis, Promoter Analysis, and Expression Profiling of PHT Gene Family in Sugarcane Root under Salinity Stress. Sustainability 2022, 14, 15893. [Google Scholar] [CrossRef]
  157. Trujillo, L.E.; Menéndez, C.; Ochogavía, M.E.; Hernández, I.; Borrás, O.; Rodríguez, R.; Coll, Y.; Arrieta, J.G.; Banguela, A.; Ramírez, R. Engineering drought and salt tolerance in plants using SodERF3, a novel sugarcane ethylene responsive factor. Biotecnol. Apl. 2009, 26, 168–171. [Google Scholar]
  158. Shingote, P.R.; Kawar, P.G.; Pagariya, M.C.; Kuhikar, R.S.; Thorat, A.S.; Babu, K. SoMYB18, a sugarcane MYB transcription factor improves salt and dehydration tolerance in tobacco. Acta Physiol. Plant 2015, 37, 217. [Google Scholar] [CrossRef]
  159. Begcy, K.; Mariano, E.D.; Lembke, C.G.; Zingaretti, S.M.; Souza, G.M.; Araújo, P.; Menossi, M. Overexpression of an evolutionarily conserved drought-responsive sugarcane gene enhances salinity and drought resilience. Ann. Bot. 2019, 124, 691–700. [Google Scholar] [CrossRef]
  160. Begcy, K.; Mariano, E.D.; Gentile, A.; Lembke, C.G.; Zingaretti, S.M.; Souza, G.M.; Menossi, M. A novel stress-induced sugarcane gene confers tolerance to drought, salt and oxidative stress in transgenic tobacco plants. PLoS ONE 2012, 7, e44697. [Google Scholar] [CrossRef]
  161. Jin-Long, G.; Li-Ping, X.; Jing-Ping, F.; Ya-Chun, S.; Hua-Ying, F.; You-Xiong, Q.; Jing-Sheng, X. A novel dirigent protein gene with highly stem-specific expression from sugarcane, response to drought, salt and oxidative stresses. Plant Cell Rep. 2012, 31, 1801–1812. [Google Scholar] [CrossRef] [PubMed]
  162. Li, X.; Liu, Z.; Zhao, H.; Deng, X.; Su, Y.; Li, R.; Chen, B. Overexpression of sugarcane ScDIR genes enhances drought tolerance in Nicotiana benthamiana. Int. J. Mol. Sci. 2022, 23, 5340. [Google Scholar] [CrossRef]
  163. Liu, F.; Huang, N.; Wang, L.; Ling, H.; Sun, T.; Ahmad, W.; Muhammad, K.; Guo, J.; Xu, L.; Gao, S. A novel L-ascorbate peroxidase 6 gene, ScAPX6, plays an important role in the regulation of response to biotic and abiotic stresses in sugarcane. Front. Plant Sci. 2018, 8, 2262. [Google Scholar] [CrossRef]
  164. Park, J.-W.; Benatti, T.R.; Marconi, T.; Yu, Q.; Solis-Gracia, N.; Mora, V.; Da Silva, J.A. Cold responsive gene expression profiling of sugarcane and Saccharum spontaneum with functional analysis of a cold inducible Saccharum homolog of NOD26-like intrinsic protein to salt and water stress. PLoS ONE 2015, 10, e0125810. [Google Scholar] [CrossRef]
  165. Ramasamy, M.; Damaj, M.B.; Vargas-Bautista, C.; Mora, V.; Liu, J.; Padilla, C.S.; Irigoyen, S.; Saini, T.; Sahoo, N.; DaSilva, J.A. A sugarcane G-protein-coupled receptor, ShGPCR1, confers tolerance to multiple abiotic stresses. Front. Plant Sci. 2021, 12, 745891. [Google Scholar] [CrossRef]
  166. Wu, Q.; Chen, Y.; Zou, W.; Pan, Y.-B.; Lin, P.; Xu, L.; Grisham, M.P.; Ding, Q.; Su, Y.; Que, Y. Genome-wide characterization of sugarcane catalase gene family identifies a ScCAT1 gene associated disease resistance. Int. J. Biol. Macromol. 2023, 232, 123398. [Google Scholar] [CrossRef] [PubMed]
  167. Asnaghi, C.; Roques, D.; Ruffel, S.; Kaye, C.; Hoarau, J.-Y.; Telismart, H.; Girard, J.-C.; Raboin, L.-M.; Risterucci, A.-M.; Grivet, L. Targeted mapping of a sugarcane rust resistance gene (Bru 1) using bulked segregant analysis and AFLP markers. Theor. Appl. Genet. 2004, 108, 759–764. [Google Scholar] [CrossRef] [PubMed]
  168. Zafar, S.A.; Zaidi, S.S.E.A.; Gaba, Y.; Singla-Pareek, S.L.; Dhankher, O.P.; Li, X.; Mansoor, S.; Pareek, A. Engineering abiotic stress tolerance via CRISPR/Cas-mediated genome editing. J. Exp. Bot. 2020, 71, 470–479. [Google Scholar] [CrossRef]
  169. Li, Y.; Wu, X.; Zhang, Y.; Zhang, Q. CRISPR/Cas genome editing improves abiotic and biotic stress tolerance of crops. Front. Genome Ed. 2022, 4, 987817. [Google Scholar] [CrossRef]
  170. Wang, Y.; Zafar, N.; Ali, Q.; Manghwar, H.; Wang, G.; Yu, L.; Ding, X.; Ding, F.; Hong, N.; Wang, G. CRISPR/Cas Genome Editing Technologies for Plant Improvement against Biotic and Abiotic Stresses: Advances, Limitations, and Future Perspectives. Cells 2022, 11, 3928. [Google Scholar] [CrossRef] [PubMed]
  171. Ogata, T.; Ishizaki, T.; Fujita, M.; Fujita, Y. CRISPR/Cas9-targeted mutagenesis of OsERA1 confers enhanced responses to abscisic acid and drought stress and increased primary root growth under nonstressed conditions in rice. PLoS ONE 2020, 15, e0243376. [Google Scholar] [CrossRef] [PubMed]
  172. Santosh Kumar, V.; Verma, R.K.; Yadav, S.K.; Yadav, P.; Watts, A.; Rao, M.; Chinnusamy, V. CRISPR-Cas9 mediated genome editing of drought and salt tolerance (OsDST) gene in indica mega rice cultivar MTU1010. Physiol. Mol. Biol. Plants 2020, 26, 1099–1110. [Google Scholar] [CrossRef]
  173. Usman, B.; Nawaz, G.; Zhao, N.; Liao, S.; Liu, Y.; Li, R. Precise editing of the OsPYL9 gene by RNA-guided Cas9 nuclease confers enhanced drought tolerance and grain yield in rice (Oryza sativa L.) by regulating circadian rhythm and abiotic stress responsive proteins. Int. J. Mol. Sci. 2020, 21, 7854. [Google Scholar] [CrossRef] [PubMed]
  174. Lim, C.; Kang, K.; Shim, Y.; Yoo, S.-C.; Paek, N.-C. Inactivating transcription factor OsWRKY5 enhances drought tolerance through abscisic acid signaling pathways. Plant Physiol. 2022, 188, 1900–1916. [Google Scholar] [CrossRef]
  175. Illouz-Eliaz, N.; Nissan, I.; Nir, I.; Ramon, U.; Shohat, H.; Weiss, D. Mutations in the tomato gibberellin receptors suppress xylem proliferation and reduce water loss under water-deficit conditions. J. Exp. Bot. 2020, 71, 3603–3612. [Google Scholar] [CrossRef]
  176. Wang, L.; Chen, L.; Li, R.; Zhao, R.; Yang, M.; Sheng, J.; Shen, L. Reduced drought tolerance by CRISPR/Cas9-mediated SlMAPK3 mutagenesis in tomato plants. J. Agric. Food Chem. 2017, 65, 8674–8682. [Google Scholar] [CrossRef] [PubMed]
  177. Abdallah, N.A.; Elsharawy, H.; Abulela, H.A.; Thilmony, R.; Abdelhadi, A.A.; Elarabi, N.I. Multiplex CRISPR/Cas9-mediated genome editing to address drought tolerance in wheat. GM Crops Food 2022, 1–17. [Google Scholar] [CrossRef]
  178. Mohr, T.; Horstman, J.; Gu, Y.Q.; Elarabi, N.I.; Abdallah, N.A.; Thilmony, R. CRISPR-Cas9 Gene Editing of the Sal1 Gene Family in Wheat. Plants 2022, 11, 2259. [Google Scholar] [CrossRef] [PubMed]
  179. Zhong, X.; Hong, W.; Shu, Y.; Li, J.; Liu, L.; Chen, X.; Islam, F.; Zhou, W.; Tang, G. CRISPR/Cas9 mediated gene-editing of GmHdz4 transcription factor enhances drought tolerance in soybean (Glycine max [L.] Merr.). Front. Plant Sci. 2022, 13, 988505. [Google Scholar] [CrossRef]
  180. Badhan, S.; Ball, A.S.; Mantri, N. First report of CRISPR/Cas9 mediated DNA-free editing of 4CL and RVE7 genes in chickpea protoplasts. Int. J. Mol. Sci. 2021, 22, 396. [Google Scholar] [CrossRef] [PubMed]
  181. Alam, M.S.; Kong, J.; Tao, R.; Ahmed, T.; Alamin, M.; Alotaibi, S.S.; Abdelsalam, N.R.; Xu, J.-H. CRISPR/Cas9 mediated knockout of the OsbHLH024 transcription factor improves salt stress resistance in rice (Oryza sativa L.). Plants 2022, 11, 1184. [Google Scholar] [CrossRef] [PubMed]
  182. Han, X.; Chen, Z.; Li, P.; Xu, H.; Liu, K.; Zha, W.; Li, S.; Chen, J.; Yang, G.; Huang, J. Development of novel rice germplasm for salt-tolerance at seedling stage using CRISPR-Cas9. Sustainability 2022, 14, 2621. [Google Scholar] [CrossRef]
  183. Yue, E.; Cao, H.; Liu, B. OsmiR535, a potential genetic editing target for drought and salinity stress tolerance in Oryza sativa. Plants 2020, 9, 1337. [Google Scholar] [CrossRef]
  184. Bouzroud, S.; Gasparini, K.; Hu, G.; Barbosa, M.A.M.; Rosa, B.L.; Fahr, M.; Bendaou, N.; Bouzayen, M.; Zsögön, A.; Smouni, A. Down regulation and loss of auxin response factor 4 function using CRISPR/Cas9 alters plant growth, stomatal function and improves tomato tolerance to salinity and osmotic stress. Genes 2020, 11, 272. [Google Scholar] [CrossRef]
  185. Tran, M.T.; Doan, D.T.H.; Kim, J.; Song, Y.J.; Sung, Y.W.; Das, S.; Kim, E.J.; Son, G.H.; Kim, S.H.; Van Vu, T. CRISPR/Cas9-based precise excision of SlHyPRP1 domain (s) to obtain salt stress-tolerant tomato. Plant Cell Rep. 2021, 40, 999–1011. [Google Scholar] [CrossRef]
  186. Wang, T.; Xun, H.; Wang, W.; Ding, X.; Tian, H.; Hussain, S.; Dong, Q.; Li, Y.; Cheng, Y.; Wang, C. Mutation of GmAITR genes by CRISPR/Cas9 genome editing results in enhanced salinity stress tolerance in soybean. Front. Plant Sci. 2021, 2752. [Google Scholar] [CrossRef]
  187. Chu, C.; Huang, R.; Liu, L.; Tang, G.; Xiao, J.; Yoo, H.; Yuan, M. The rice heavy-metal transporter OsNRAMP1 regulates disease resistance by modulating ROS homoeostasis. Plant, Cell Environ. 2022, 45, 1109–1126. [Google Scholar] [CrossRef] [PubMed]
  188. 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]
  189. Wang, F.-Z.; Chen, M.-X.; Yu, L.-J.; Xie, L.-J.; Yuan, L.-B.; Qi, H.; Xiao, M.; Guo, W.; Chen, Z.; Yi, K. OsARM1, an R2R3 MYB transcription factor, is involved in regulation of the response to arsenic stress in rice. Front. Plant Sci. 2017, 8, 1868. [Google Scholar] [CrossRef] [PubMed]
  190. Wang, F.; Wang, C.; Liu, P.; Lei, C.; Hao, W.; Gao, Y.; Liu, Y.-G.; Zhao, K. Enhanced rice blast resistance by CRISPR/Cas9-targeted mutagenesis of the ERF transcription factor gene OsERF922. PLoS ONE 2016, 11, e0154027. [Google Scholar] [CrossRef]
  191. Ma, J.; Chen, J.; Wang, M.; Ren, Y.; Wang, S.; Lei, C.; Cheng, Z. Disruption of OsSEC3A increases the content of salicylic acid and induces plant defense responses in rice. J. Exp. Bot. 2018, 69, 1051–1064. [Google Scholar] [CrossRef]
  192. Zhou, Y.; Xu, S.; Jiang, N.; Zhao, X.; Bai, Z.; Liu, J.; Yao, W.; Tang, Q.; Xiao, G.; Lv, C. Engineering of rice varieties with enhanced resistances to both blast and bacterial blight diseases via CRISPR/Cas9. Plant Biotechnol. J. 2022, 20, 876–885. [Google Scholar] [CrossRef]
  193. Nekrasov, V.; Wang, C.; Win, J.; Lanz, C.; Weigel, D.; Kamoun, S. Rapid generation of a transgene-free powdery mildew resistant tomato by genome deletion. Sci. Rep. 2017, 7, 482. [Google Scholar] [CrossRef]
  194. Santillán Martínez, M.I.; Bracuto, V.; Koseoglou, E.; Appiano, M.; Jacobsen, E.; Visser, R.G.; Wolters, A.-M.A.; Bai, Y. CRISPR/Cas9-targeted mutagenesis of the tomato susceptibility gene PMR4 for resistance against powdery mildew. BMC Plant Biol. 2020, 20, 284. [Google Scholar] [CrossRef]
  195. Wang, Y.; Cheng, X.; Shan, Q.; Zhang, Y.; Liu, J.; Gao, C.; Qiu, J.-L. Simultaneous editing of three homoeoalleles in hexaploid bread wheat confers heritable resistance to powdery mildew. Nat. Biotechnol. 2014, 32, 947–951. [Google Scholar] [CrossRef]
  196. Wan, D.-Y.; Guo, Y.; Cheng, Y.; Hu, Y.; Xiao, S.; Wang, Y.; Wen, Y.-Q. CRISPR/Cas9-mediated mutagenesis of VvMLO3 results in enhanced resistance to powdery mildew in grapevine (Vitis vinifera). Hortic. Res. 2020, 7, 116. [Google Scholar] [CrossRef]
  197. Sun, Q.; Lin, L.; Liu, D.; Wu, D.; Fang, Y.; Wu, J.; Wang, Y. CRISPR/Cas9-mediated multiplex genome editing of the BnWRKY11 and BnWRKY70 genes in Brassica napus L. Int. J. Mol. Sci. 2018, 19, 2716. [Google Scholar] [CrossRef]
  198. Fister, A.S.; Landherr, L.; Maximova, S.N.; Guiltinan, M.J. Transient expression of CRISPR/Cas9 machinery targeting TcNPR3 enhances defense response in Theobroma cacao. Front. Plant Sci. 2018, 9, 268. [Google Scholar] [CrossRef] [PubMed]
  199. Králová, M.; Bergougnoux, V.; Frébort, I. CRISPR/Cas9 genome editing in ergot fungus Claviceps purpurea. J. Biotechnol. 2021, 325, 341–354. [Google Scholar] [CrossRef] [PubMed]
  200. Arulganesh, T.; Kumam, Y.; Kumar, K.; Arul, L.; Kokiladevi, E.; Nakeeran, S.; Varanavasiappan, S.; Manonmani, S.; Sudhakar, D. Genome editing of elite rice cultivar CO51 for bacterial leaf blight resistance. Electron. J. Plant Breed. 2021, 12, 1060–1068. [Google Scholar]
  201. Ortigosa, A.; Gimenez-Ibanez, S.; Leonhardt, N.; Solano, R. Design of a bacterial speck resistant tomato by CRISPR/Cas9-mediated editing of Sl JAZ 2. Plant Biotechnol. J. 2019, 17, 665–673. [Google Scholar] [CrossRef]
  202. Thomazella, D.P.D.T.; Seong, K.; Mackelprang, R.; Dahlbeck, D.; Geng, Y.; Gill, U.S.; Qi, T.; Pham, J.; Giuseppe, P.; Lee, C.Y. Loss of function of a DMR6 ortholog in tomato confers broad-spectrum disease resistance. Proc. Natl. Acad. Sci. USA 2021, 118, e2026152118. [Google Scholar] [CrossRef]
  203. Tripathi, J.N.; Ntui, V.O.; Ron, M.; Muiruri, S.K.; Britt, A.; Tripathi, L. CRISPR/Cas9 editing of endogenous banana streak virus in the B genome of Musa spp. overcomes a major challenge in banana breeding. Commun. Biol. 2019, 2, 46. [Google Scholar] [CrossRef] [PubMed]
  204. Tripathi, J.N.; Ntui, V.O.; Shah, T.; Tripathi, L. CRISPR/Cas9-mediated editing of DMR6 orthologue in banana (Musa spp.) confers enhanced resistance to bacterial disease. Plant Biotechnol. J. 2021, 19, 1291. [Google Scholar] [CrossRef]
  205. Kumam, Y.; Rajadurai, G.; Kumar, K.; Varanavasiappan, S.; Reddy, M.K.; Krishnaveni, D.; Mangrauthia, S.K.; Raveendran, M.; Arul, L.; Kokiladevi, E. Genome editing of indica rice ASD16 for imparting resistance against rice tungro disease. J. Plant Biochem. Biotechnol. 2022, 31, 880–893. [Google Scholar] [CrossRef]
  206. Macovei, A.; Sevilla, N.R.; Cantos, C.; Jonson, G.B.; Slamet-Loedin, I.; Čermák, T.; Voytas, D.F.; Choi, I.R.; Chadha-Mohanty, P. Novel alleles of rice eIF4G generated by CRISPR/Cas9-targeted mutagenesis confer resistance to Rice tungro spherical virus. Plant Biotechnol. J. 2018, 16, 1918–1927. [Google Scholar] [CrossRef] [PubMed]
  207. Wang, W.; Ma, S.; Hu, P.; Ji, Y.; Sun, F. Genome editing of rice eIF4G loci confers partial resistance to rice black-streaked dwarf virus. Viruses 2021, 13, 2100. [Google Scholar] [CrossRef] [PubMed]
  208. Liu, C.; Kong, M.; Yang, F.; Zhu, J.; Qi, X.; Weng, J.; Di, D.; Xie, C. Targeted generation of Null Mutants in ZmGDIα confers resistance against maize rough dwarf disease without agronomic penalty. Plant Biotechnol. J. 2022, 20, 803. [Google Scholar] [CrossRef]
  209. Lucioli, A.; Tavazza, R.; Baima, S.; Fatyol, K.; Burgyan, J.; Tavazza, M. CRISPR-Cas9 Targeting of the eIF4E1 Gene Extends the Potato Virus Y Resistance Spectrum of the Solanum tuberosum L. cv. Desirée. Front. Microbiol. 2022, 13, 873930. [Google Scholar] [CrossRef] [PubMed]
  210. Noureen, A.; Khan, M.Z.; Amin, I.; Zainab, T.; Mansoor, S. CRISPR/Cas9-mediated targeting of susceptibility factor eIF4E-enhanced resistance against potato virus Y. Front. Genet. 2022, 13, 922019. [Google Scholar]
  211. Ishikawa, M.; Yoshida, T.; Matsuyama, M.; Kouzai, Y.; Kano, A.; Ishibashi, K. Tomato brown rugose fruit virus resistance generated by quadruple knockout of homologs of TOBAMOVIRUS MULTIPLICATION1 in tomato. Plant Physiol. 2022, 189, 679–686. [Google Scholar] [CrossRef]
  212. Ghorbani Faal, P.; Farsi, M.; Seifi, A.; Mirshamsi Kakhki, A. Virus-induced CRISPR-Cas9 system improved resistance against tomato yellow leaf curl virus. Mol. Biol. Rep. 2020, 47, 3369–3376. [Google Scholar] [CrossRef]
  213. Zhang, P.; Du, H.; Wang, J.; Pu, Y.; Yang, C.; Yan, R.; Yang, H.; Cheng, H.; Yu, D. Multiplex CRISPR/Cas9-mediated metabolic engineering increases soya bean isoflavone content and resistance to soya bean mosaic virus. Plant Biotechnol. J. 2020, 18, 1384–1395. [Google Scholar] [CrossRef]
  214. Chandrasekaran, J.; Brumin, M.; Wolf, D.; Leibman, D.; Klap, C.; Pearlsman, M.; Sherman, A.; Arazi, T.; Gal-On, A. Development of broad virus resistance in non-transgenic cucumber using CRISPR/Cas9 technology. Mol. Plant Pathol. 2016, 17, 1140–1153. [Google Scholar] [CrossRef]
  215. Yu, K.; Liu, Z.; Gui, H.; Geng, L.; Wei, J.; Liang, D.; Lv, J.; Xu, J.; Chen, X. Highly efficient generation of bacterial leaf blight-resistant and transgene-free rice using a genome editing and multiplexed selection system. BMC Plant Biol. 2021, 21, 197. [Google Scholar] [CrossRef]
  216. Ahmad, A.; Ghouri, M.Z.; Munawar, N.; Ismail, M.; Ashraf, S.; Aftab, S.O. Regulatory, ethical, and social aspects of CRISPR crops. In CRISPR Crops: The Future of Food Security; Ahmad, A., Khan, S.H., Khan, Z., Eds.; Springer: Singapore, 2021; pp. 261–287. [Google Scholar]
  217. Gupta, S.; Kumar, A.; Patel, R.; Kumar, V. Genetically modified crop regulations: Scope and opportunity using the CRISPR-Cas9 genome editing approach. Mol. Biol. Rep. 2021, 48, 4851–4863. [Google Scholar] [CrossRef] [PubMed]
  218. Sprink, T.; Eriksson, D.; Schiemann, J.; Hartung, F. Regulatory hurdles for genome editing: Process-vs. product-based approaches in different regulatory contexts. Plant Cell Rep. 2016, 35, 1493–1506. [Google Scholar] [CrossRef]
  219. Ahmad, A.; Munawar, N.; Khan, Z.; Qusmani, A.T.; Khan, S.H.; Jamil, A.; Ashraf, S.; Ghouri, M.Z.; Aslam, S.; Mubarik, M.S. An outlook on global regulatory landscape for genome-edited crops. Int. J. Mol. Sci. 2021, 22, 11753. [Google Scholar] [CrossRef] [PubMed]
  220. Podevin, N.; Davies, H.V.; Hartung, F.; Nogué, F.; Casacuberta, J.M. Site-directed nucleases: A paradigm shift in predictable, knowledge-based plant breeding. Trends Biotechnol. 2013, 31, 375–383. [Google Scholar] [CrossRef] [PubMed]
  221. Zannoni, L. Evolving regulatory landscape for genome-edited plants. CRISPR J. 2019, 2, 3–8. [Google Scholar] [CrossRef] [PubMed]
  222. Verma, P.; Tandon, R.; Yadav, G.; Gaur, V. Structural aspects of DNA repair and recombination in crop improvement. Front. Genet. 2020, 1103. [Google Scholar] [CrossRef] [PubMed]
  223. Van de Wiel, C.; Schaart, J.; Lotz, L.; Smulders, M. New traits in crops produced by genome editing techniques based on deletions. Plant Biotechnol. Rep. 2017, 11, 1–8. [Google Scholar] [CrossRef]
  224. Buchholzer, M.; Frommer, W.B. An increasing number of countries regulate genome editing in crops. New Phytol. 2022, 237, 12–15. [Google Scholar] [CrossRef]
  225. Meena, D.M.R.; Reddy, G.; Kumar, R.; Pandey, S.; Hemaprabha, G. Recent Advances in Sugarcane Genomics, Physiology, and Phenomics for Superior Agronomic Traits. Front. Genet. 2022, 1523. [Google Scholar] [CrossRef]
  226. Mohan, C.; Easterling, M.; Yau, Y.-Y. Gene editing technologies for sugarcane improvement: Opportunities and limitations. Sugar Tech 2022, 1–17. [Google Scholar] [CrossRef]
  227. Hussin, S.H.; Liu, X.; Li, C.; Diaby, M.; Jatoi, G.H.; Ahmed, R.; Imran, M.; Iqbal, M.A. An Updated Overview on Insights into Sugarcane Genome Editing via CRISPR/Cas9 for Sustainable Production. Sustainability 2022, 14, 12285. [Google Scholar] [CrossRef]
  228. Labun, K.; Montague, T.G.; Krause, M.; Torres Cleuren, Y.N.; Tjeldnes, H.; Valen, E. CHOPCHOP v3: Expanding the CRISPR web toolbox beyond genome editing. Nucleic Acids Res. 2019, 47, W171–W174. [Google Scholar] [CrossRef] [PubMed]
  229. Liu, H.; Ding, Y.; Zhou, Y.; Jin, W.; Xie, K.; Chen, L.-L. CRISPR-P 2.0: An improved CRISPR-Cas9 tool for genome editing in plants. Mol Plant. 2017, 10, 530–532. [Google Scholar] [CrossRef] [PubMed]
  230. Riaño-Pachón, D.M.; Mattiello, L. Draft genome sequencing of the sugarcane hybrid SP80-3280. F1000Research 2017, 6. [Google Scholar] [CrossRef]
  231. Garsmeur, O.; Droc, G.; Antonise, R.; Grimwood, J.; Potier, B.; Aitken, K.; Jenkins, J.; Martin, G.; Charron, C.; Hervouet, C. A mosaic monoploid reference sequence for the highly complex genome of sugarcane. Nat. Commun. 2018, 9, 2638. [Google Scholar] [CrossRef] [PubMed]
  232. Zhang, J.; Zhang, X.; Tang, H.; Zhang, Q.; Hua, X.; Ma, X.; Zhu, F.; Jones, T.; Zhu, X.; Bowers, J. Allele-defined genome of the autopolyploid sugarcane Saccharum spontaneum L. Nat. Genet. 2018, 50, 1565–1573. [Google Scholar] [CrossRef]
  233. Joyce, P.; Kuwahata, M.; Turner, N.; Lakshmanan, P. Selection system and co-cultivation medium are important determinants of Agrobacterium-mediated transformation of sugarcane. Plant Cell Rep. 2010, 29, 173–183. [Google Scholar] [CrossRef]
  234. Metje-Sprink, J.; Menz, J.; Modrzejewski, D.; Sprink, T. DNA-free genome editing: Past, present and future. Front. Plant Sci. 2019, 9, 1957. [Google Scholar] [CrossRef]
  235. Gazaffi, R.; Oliveira, K.; Souza, A.; Garcia, A. The importance of the germplasm in developing agro-energetic profile sugarcane cultivars. In Sugar Cane Bioethanol: R&D for Productivity and Sustainability; Cortez, L.A.B., Ed.; Blucher: Sao Paulo, Brazil, 2010; pp. 333–343. [Google Scholar]
Figure 1. Approaches for producing transgene-free GE sugarcane lines.
Figure 1. Approaches for producing transgene-free GE sugarcane lines.
Agronomy 13 01000 g001
Figure 2. Strategies for screening and analyzing the transgene-free GE lines. TKC and visual marker system are used for selecting transgene-free GE plants/seeds developed through plasmid-based delivery methods. The plants developed through RNP-based delivery systems are subjected to PCR-RFLP and T7E1 assays to identify mutated individuals. Finally, mutants identified in these methods are further sequenced and analyzed for type of mutation in the targeted region.
Figure 2. Strategies for screening and analyzing the transgene-free GE lines. TKC and visual marker system are used for selecting transgene-free GE plants/seeds developed through plasmid-based delivery methods. The plants developed through RNP-based delivery systems are subjected to PCR-RFLP and T7E1 assays to identify mutated individuals. Finally, mutants identified in these methods are further sequenced and analyzed for type of mutation in the targeted region.
Agronomy 13 01000 g002
Table 1. Candidate genes for biotic and abiotic stresses in Sugarcane.
Table 1. Candidate genes for biotic and abiotic stresses in Sugarcane.
StressCandidate GenesReferences
DroughtSoACLA-1[150]
DroughtSoP5CS[151]
DroughtmiRNAs [152,153]
DroughtScLoX, Dehydrin. Dirigent-jacalin[154]
SalinitymiRNAs[155]
SalinityShPHT[156]
Drought and salinitySodERF3[157]
Drought and salinitySoMYB18[158]
Drought and salinityScdr2[159]
Drought, salinity and oxidativeScdr1[160]
Drought, salinity and oxidativeScDir[161,162]
Drought, salinity and oxidativeScAPX6[163]
Salinity and low temperatureSspNIP2[164]
Drought, salinity and coldShGPCR1[165]
Whip smutScCAT1[166]
RustBru1[167]
Table 2. CRISPR/Cas mediated genome editing for various biotic and abiotic stresses in crops.
Table 2. CRISPR/Cas mediated genome editing for various biotic and abiotic stresses in crops.
StressCropTarget GeneReferences
DroughtRiceOsERA1[171]
OsDST[172]
OsPYL9[173]
OsWRKY5[174]
TomatoSlGID1[175]
SlMAPK3[176]
WheatTaSal1[177]
Sal1[178]
SoybeanGmHdz4[179]
Chickpea4CL and RVE7 [180]
SalinityRiceOsbHLH024[181]
OsRR22[182]
OsmiR535[183]
TomatoSlARF4[184]
SlHyPRP1[185]
SoybeanGmAITR[186]
Heavy metalsRiceOsNRAMP5[187]
OsNRAMP1[188]
OsARM1[189]
Fungus
Rice BlastRiceOsERF922[190]
RiceOsSEC3A[191]
RiceBsr-d1, Pi21 and ERF922[192]
Powdery mildewTomatoSlML[193]
TomatoSlPMR4[194]
WheatTaMLO-A1[195]
GrapevineVvMOL3[196]
Stem rotRapeseedBnWRKY70[197]
Black podCacaoTcNPR3[198]
ErgotRyepyr4 and TrpE[199]
Bacteria
Bacterial leaf blightRicePi21 or ERF922[192]
OsSWEET14[200]
Bacterial leaf spotTomatoSlJAZ2[201]
Sldmr6-1[202]
Banana
Xanthomonas wilt
Musa balbisianaLRR, WAK2, WAK5, Vicilin, RPM1, PR1, NPR1[203]
BananaMusaDMR6[204]
Bacterial speckTomatoSLDMR6-1[202]
Virus
Rice tungro spherical virusRiceeIF4G[205,206]
Rice black streaked dwarf virusRiceeIF4G[207]
Rice black-streaked dwarf virusMaizeZmGDIα[208]
Potato virus Y Potato eIF4E1[209]
eIF4E[210]
Tomato brown rugose fruit virusTomatoTOM1[211]
Tomato yellow leaf curl virusTomatoIR, CP of virus[212]
Soybean mosaic virusSoybeanGmF3H1, GmF3H2 and GmFNSII-1[213]
Cucumber vein yellowing virus, Zucchini yellow mosaic virus; Papaya ring spot mosaic virus-CucumbereIF4G[214]
Banana streak virusBananaORF1, ORF2,ORF3 of virus[203]
Table 3. Transgene-free genome editing in crops.
Table 3. Transgene-free genome editing in crops.
MethodStressCrop Target GeneReferences
Protoplast transformation DroughtChickpea4CL and RVE7 [180]
Agrobacterium-mediated transformation Bacterial leaf blightRiceXa13[215]
Agrobacterium-mediated transformationPowdery mildewTomatoSlMlo1[193]
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

Surya Krishna, S.; Harish Chandar, S.R.; Ravi, M.; Valarmathi, R.; Lakshmi, K.; Prathima, P.T.; Manimekalai, R.; Viswanathan, R.; Hemaprabha, G.; Appunu, C. Transgene-Free Genome Editing for Biotic and Abiotic Stress Resistance in Sugarcane: Prospects and Challenges. Agronomy 2023, 13, 1000. https://doi.org/10.3390/agronomy13041000

AMA Style

Surya Krishna S, Harish Chandar SR, Ravi M, Valarmathi R, Lakshmi K, Prathima PT, Manimekalai R, Viswanathan R, Hemaprabha G, Appunu C. Transgene-Free Genome Editing for Biotic and Abiotic Stress Resistance in Sugarcane: Prospects and Challenges. Agronomy. 2023; 13(4):1000. https://doi.org/10.3390/agronomy13041000

Chicago/Turabian Style

Surya Krishna, Sakthivel, S R Harish Chandar, Maruthachalam Ravi, Ramanathan Valarmathi, Kasirajan Lakshmi, Perumal Thirugnanasambandam Prathima, Ramaswamy Manimekalai, Rasappa Viswanathan, Govindkurup Hemaprabha, and Chinnaswamy Appunu. 2023. "Transgene-Free Genome Editing for Biotic and Abiotic Stress Resistance in Sugarcane: Prospects and Challenges" Agronomy 13, no. 4: 1000. https://doi.org/10.3390/agronomy13041000

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