Next Article in Journal
Silibinin Suppresses the Hyperlipidemic Effects of the ALK-Tyrosine Kinase Inhibitor Lorlatinib in Hepatic Cells
Next Article in Special Issue
Transcriptome Analysis Reveals Critical Genes and Pathways in Carbon Metabolism and Ribosome Biogenesis in Poplar Fertilized with Glutamine
Previous Article in Journal
Nephritis-Associated Plasmin Receptor (NAPlr): An Essential Inducer of C3-Dominant Glomerular Injury and a Potential Key Diagnostic Biomarker of Infection-Related Glomerulonephritis (IRGN)
Previous Article in Special Issue
Knockdown of p-Coumaroyl Shikimate/Quinate 3′-Hydroxylase Delays the Occurrence of Post-Harvest Physiological Deterioration in Cassava Storage Roots
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Recent Advances in Molecular Improvement for Potato Tuber Traits

1
Institute of Nuclear Agricultural Sciences, College of Agriculture and Biotechnology, Zhejiang University, Xihu District, Hangzhou 310058, China
2
Yazhou Bay Seed Laboratory, Yazhou Bay Science and Technology City, Yazhou District, Sanya 572025, China
3
Hainan Institute, Zhejiang University, Yazhou Bay Science and Technology City, Yazhou District, Sanya 572025, China
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(17), 9982; https://doi.org/10.3390/ijms23179982
Submission received: 29 June 2022 / Revised: 24 August 2022 / Accepted: 25 August 2022 / Published: 1 September 2022
(This article belongs to the Special Issue Functional Genomics of Energy Crops)

Abstract

:
Potato is an important crop due to its nutritional value and high yield potential. Improving the quality and quantity of tubers remains one of the most important breeding objectives. Genetic mapping helps to identify suitable markers for use in the molecular breeding, and combined with transgenic approaches provides an efficient way for gaining desirable traits. The advanced plant breeding tools and molecular techniques, e.g., TALENS, CRISPR-Cas9, RNAi, and cisgenesis, have been successfully used to improve the yield and nutritional value of potatoes in an increasing world population scenario. The emerging methods like genome editing tools can avoid incorporating transgene to keep the food more secure. Multiple success cases have been documented in genome editing literature. Recent advances in potato breeding and transgenic approaches to improve tuber quality and quantity have been summarized in this review.

1. Introduction

Potato (Solanum tuberosum L.) is the most important non-grain food crop concerning its global consumption [1]. It was firstly domesticated in Perú around 10,000 years ago. The potato will become an important crop consumed as staple food in the future due to increasing food security issues, since the global population will rise to 9.7 billion by 2050 [2,3,4].
Potato has high nutritional values due to the presence of several essential phytochemicals with significant health benefits [5]. Various phytonutrients (carotenoids, anthocyanins, phenolics, and flavonoids), mineral elements (potassium, sodium, magnesium, copper, iron, and zinc), and vitamins (vitamin B1, B6, B9, C, E) are the major components of potatoes [6,7]. Due to their antioxidant properties, these phytonutrients are essential for maintaining human health [5,8].
The main motive for improving this crop is to improve the processing tuber quality with a higher yield. Yield of potatoes is determined by proper genotypes and growing environments, and is significantly correlated with the number of sprouts, plant height, shoot diameter, and leaves length [9]. The market values of table potatoes depend on the visual features of cultivars, i.e., depth of eyes, flesh color, tuber shape, and cooking suitability. The chemical composition of the tuber impacts the eating and appearance quality of the potato products, for example, starch contents impact cooked product texture, and sugar content directly affects fried product color [10].
Research on the genome sequencing of cultivars and their wild relatives have revealed more significant genetic variations and signs of selection of theinteresting traits via genes. Potato shows higher heterozygosity, tetrasomic polyploidity owing to its four homolog chromosomes, rendering challenges to alter its genome [11]. Scrutinizing genes with functional and molecular genetics approaches will also meet key hindrances [12]. Thus, combining the knowledge of functional genomics and basic plant breeding techniques with advanced molecular methods is crucial to improve agronomic traits. Though many review articles on potato breeding and transgenic potato have been written [10,11,12,13,14], there is a lack of a comprehensive review focusing on a bridge between potato breeding and advanced molecular techniques for improving the quality and yield of potato tubers. This review will provide a new direction for potato breeders to use a combined approach of advanced breeding and molecular techniques.

2. Quantitative Trait Loci (QTL) Mapping

Genetic modification does not rely merely on making crosses of donor species with the recipient cultivar of potato. Instead, molecular techniques offer an alternative approach, and reducing the chances of linkage drag associated with conventional methods. These methods can incorporate entire genes, gene groups, or even whole chromosomes [14]. Genotyping by next-generation sequencing tools can find the allelic variation in the genomics era (Figure 1). Various breeding tools such as QTL analysis, whole-genome sequencing, and marker-trait associations are crucial to understand the genetic differences between breeding clones, progeny crosses, and wild species (Figure 1). Wild germplasm shows a smaller genetic distance in self-incompatible germplasm than accessions of similar species in the germplasm [15].

2.1. QTL Mapping for Tuber Quality

Indirect manipulation and advances in QTL mapping have eased the way of crop tuber quantity and quality improvement. This article mainly describes QTL mapping and advanced molecular techniques based on the genetic improvement of potato tubers. Some major QTLs related to the potato tuber quality and quantity and their position regarding chromosomes are shown in Table 1.
Biosynthesis of “leaf starch” or “transient starch” usually occurs during the daytime [24]. In contrast, the breakdown of sucrose and its conversion to storage starch occur during the nighttime [24,25]. This conversion involves complexed procedures encoded by various gene sets located at 123 loci on I-XII chromosomes [26]. They also control tuber starch contents, sugar contents, and sugar-starch interconversion [26].
Starch granules in potato tubers are always more significant in size than the granules synthesized in the leaves [27,28]. For the size of starch granules, two QTLs viz. SGS02-8 and SGS03-8 have been documented on chromosome VIII [16]. Also, the structure and composition of storage starch are different from leaf starch [24,29]. Twelve QTLs related to tuber starch contents have been discovered (Table 1) [17]. Similarly, Li et al. [18] also reported other loci for starch contents (Table 1). Around 77 genomic loci encoding enzymes have been found associated with starch metabolism [30]. Various enzyme isoforms involved in starch metabolism also have been reported: 6 sub-units of ADP-glucose pyrophosphorylase large subunits; 5 different types of alpha-amylase; 4 different types of alpha-glucan phosphorylase; 2 different kinds of ATP-ADP antiporters; 10 different types of Beta-amylases; 4 different kinds of branching enzymes; disproportionating enzymes; glucan water dikinase (GWD); glucose transporters; glucose-6-phosphate translocators; granule bound starch synthases and many others [30]. The starch synthesis starts from bioconversion to adenosine diphosphate-glucose from sucrose, the primer substrate of starch biosynthesis, which are employed to synthesize the amylopectin and amylose. Five chromosomal regions on chromosome 2, 3, 5, 7, and 10 were linked with amylose content [16].
The distinctiveness of potato starch is that it has high level of starch-bound phosphate, conferring it new properties such as high peak viscosity and gel-forming capacity. A total of 17 loci was identified significantly for the phosphorus content in potato starch, 14 of which are assigned to 8 genomic regions on chromosomes 1, 4, 5, 7, 8, 10, and 11, and most of the SNPs identified belong to protein coding regions [31].
Allelic variation significantly affects the starch phosphorylation process, and the genes encoding GWD, starch branching enzymes I and II, and starch synthase III are associated with this process [32]. These allelic variants can be served as genetic markers for starch phosphorylation [32].
The important loci related to tuber starch contents were firstly documented by Li et al. [20]. QTLs related to starch contents were AmyZ (M79328), SssI (Y10416), StpL (X73684), GbssI (X52417), StpH, located on chromosomes IV, III, V, VIII and IX, respectively [20]. Another genetic mapping study has revealed 12 different loci regarding tuber starch contents viz. pPt-539763, pPt-535988, toPt-438845, pPt-533878, pPt-471789, pPt-538127, toPt-437014–pPt-538033, toPt-440651, and pPt-656237, on seven chromosomes: I, II, III, VIII, X, XI, and XII [17]. Average starch yield and chips quality are associated with Stp23-8b and Pain1-8c or Pain1prom-d/e [20,33].
Protein content is another importance quality trait of the potato industry. Genetic mapping analysis revealed potential QTLs on chromosomes 2, 3, 5 and 9, and further cofactor QTL analysis identified two masked QTLs on chromosomes 1 and 5 [34].

2.2. QTL Mapping for Tubers Storage Duration

Accumulating reducing sugars in cold-stored potatoes, called cold-induced sweetening (CIS), is a severe problem that causes unacceptable color changes and acrylamide formation in fried products. CIS has been found to be associated with various enzymes, such as starch synthases, adenosine diphosphate glucose pyrophosphorylase, branching enzymes, debranching enzymes, β-amylases, amylase inhibitors, and starch phosphorylases for degradation of starch contents [35]. The CIS has been related to the putative Kunitz-type tuber invertase inhibitor (KT-InvInh) position on the StKI locus at chromosome III [36].
The low sugar and fructose contents of tubers play pivotal roles in the processing quality of the potato. The organization of invertase genes (InvGE and InvGF) at the Inv ap -b locus has already been documented on chromosome IX [37]. Later, InvGE-A 85 (A 86), InvCD141-G 765, InvCD141_T 543 (A 280, T 288, T 339, A 630, C 1030, G 1031, T 1096) InvGE-G 95 (G 106), Pain1- A 718 (C 552) 2, and Pain1- A 1544, Pain1-T 741 on chromosomes III, X and IX were found to be associated with tuber starch contents and its processing quality [19]. The QTLs of CIS and reconditioning (REC; a process of reducing sugars into starch by storing the cold-stored potatoes) were identified on chromosomes V, VI, and VII of the CIS-susceptible and chromosomes V and XI in the CIS-resistant parents. Two functional genes, a starch hydrolysis gene GWD, were located with QTL as REC_B_05-1, and a starch synthesis gene AGPS2 has also been mapped in QTL CIS_E_07-1 [21].
The QTLs for tuber dormancy and sprouting traits were found on chromosomes 2, 3, and 7, which are good candidates for marker-assisted breeding [38].

2.3. QTL Mapping for Tuber Morphology

Tuber shape and color are very important quality traits regarding its market life or selling because consumers pay more attention to the shape of potato tubers. For the tuber shape, two loci, SNP Solcap_snp_c2_25510 (Alpha virus core protein family) and Solcap_snp_c2_25485 (ribosomal protein S6 kinase) on chromosome X were found significantly associated with the trait in 2016 [22], while another locus, SNP Solcap_snp_c2_34875 (serine/threonine-protein kinase) on chromosome number IV was found to be significant in 2017. For tuber skin color, a total of 12 QTLs, three on chromosome 1; one on chromosome 5; two on chromosome 7; three on chromosome 8; one on chromosome 10; one on chromosome 11, and one on chromosome 12 were detected (Table 1) [22]. Significant QTLs for tuber shape were detected on chromosomes 4, 7, and 10, with heritability estimates ranging from 0.09 to 0.36 [23]. For each grain shape trait (eye depth, tuber shape, regularity of tuber shape, mean tuber weight), four to seven QTLs were determined except for tuber flesh color, which was controlled by a major QTL on chromosome III, accounting for 76.8% of trait variance [39]. Moreover, a minor QTL was localized on chromosome II for flesh color, [39]. In a gynogenic dihaploid (2n = 2x = 24) population, three QTLs for tuber shape were identified on chromosomes 6, 10, and 11 [40].
Likewise, certain other QTLs linked with tuber morphology and pigmentation have been documented. For instance, the R2R3-MYB loci for tuber flesh color have been revealed on chromosome 10. Dihydroflavonol 4-reductase and flavonoid 3,5-hydroxylase are associated with color, whose loci have been reported on chromosomes 2 and 11 [41]. Similarly, the loci of Anthocyanin 1 associated with pigment flesh intensity was found on chromosomes 9 and 10, and that of β-carotene hydroxylase for yellow flesh was detected on chromosome 3 [41]. These mapping researches provide a better understanding of the genetics of various desirable characters and suitable diagnostic DNA markers for indirect marker aided selection (MAS) of target traits in potato breeding.

3. Genome Selection (GS)

In conventional breeding techniques, for several generations, recurrent phenotypic selection can be exploited [42]. Decreasing population size and increasing genotypes number under evaluation are the typical selection challenges in the conventional method. The breeder selects parents based on phenotypic characters for pair-wise crossing [43]. Generally, several high-yield and stress-resistant parents will be screened over several clonal generations in several years [44]. The environment significantly influences the expression of multiple traits in potatoes, such as yield, tuber size, and processing quality. Almost 40 traits are considered to be important for the cultivar development of potato, which are divided into yield, tuber quality, and stress tolerance attributes [45]. Knowledge of the gene × environment interactions will help the identification of superior parents, screening, and selection methods for superior genotypes [46]. Figure 1 shows a schematic flow of various steps involved in genome-assisted breeding.
MAS is a great tool for assisting plant breeding, but many agronomic traits are complicated and controlled by several loci. An improved and new breeding approach has been developed, which is genomic selection (GS). GS helps estimate plants’ genetic success based upon all molecular markers’ information simultaneously instead of a few ones like in MAS [47]. Increased accuracy of high-throughput genotyping techniques, decreased cost of sequencing techniques, and marker selection have reinforced the value of GS [48]. GS can predict the genetic value/breeding value. There are two populations: the training population that is genotyped and phenotyped, and the testing population that is genotyped but not phenotyped. GS is used to predict the phenotype of testing population for some selection cycles. Various statistical models are used to predict the traits of interest, such as Kinship GAUSS, PR-BLUP, Bayesian LASSO, Bayes B, and Bayes Cp [49]. For starch contents, the cross-prediction validation correlation of 0.56 in the training panel but the correlation was around 0.30 in the test panel derived from the tetraploid mapping population of a breeding program in Denmark [50]. The prediction accuracy of starch contents has improved by 8% in tetraploid German breeding clones [51]. Correlations of cross-prediction validation regarding tuber dry matter or specific gravity were 0.75–0.83 and 0.37–0.71 among different populations in Denmark [52,53]. In contrast, average cross-validation of the dry matter in European cultivars was 0.65 (ranging from 0.54 to 0.68) [54]. For chipping quality, prediction accuracy ranged from 0.4 to 0.45, as depicted from the pedigree depth of unselected US populations [52]. When tetraploid mapping populations were used in the Denmark breeding program, cross-prediction validation correlations ranged from 0.39 to 0.79, while the correlations declined across the populations, ranging from 0.28 to 0.48 [53]. The results gave a 0.73 cross-prediction validation correlation for the training panel, while in the test panel, 0.42 and 0.43 correlation values from mapping populations of tetraploid potato breeding programs in the rest of Europe [50]. For tuber yield and yield components, an additive genetic variance of SNPs captured 45% of the total genetic variance; the prediction accuracy was calculated in the range of 0.06 to 0.63 in tetraploid breeding unselected US F1 population [52]. In cultivated German clones of breeding potato, about an 8% increase was obtained in the prediction model accuracy using both additive and dominance effects [51]. Averages of 0.37, 0.32, and 0.17 for total yield, size, and number were revealed by cross-validation of several models, respectively [54]. Sallam et al. [55] indicated that if only the prediction accuracy exceeds 0.25, GS should surpass phenotypic selection in gain per unit time. Although the lowest average prediction correlation in some GS studies was around 0.30, GS still has potential to improve breeding efficiency in tetraploid potato for less cost per unit time.
Genome design breeding of hybrid potato has been carried out to exploit the heterosis in diploid potatoes with high homozygosity, enabling the transformation of potato breeding from a slow, non-accumulative mode into a fast iterative one [56]. The resultant hybrids showed strong heterosis [56]. For successful hybrid potato breeding, more inbred lines of high homozygosity are essential. Huang et al. [57] assembled 44 high-quality pan-genomes from 24 wild and 20 cultivated accessions that are representative of Solanum section Petota, the tuber-bearing clade, as well as 2 genomes from the neighbouring section, Etuberosum. The identification of 561,433 high-confidence structural variants and construction of a map of large inversions provides critical guidance for improving inbred lines and precluding potential linkage drag, which will accelerate hybrid potato breeding [57].

4. Transgenic Breeding

When gene identification tools combined with advanced biotechnological tools such as genome editing, incorporating the genes into the breeding program becomes fast [58,59,60,61,62]. Though there are many approaches to making transgenics, only four main approaches will be discussed here:
(1)
Conventional transgenic approaches of potato breeding include incorporation of a transgene via Agrobacterium-mediated transformation or any other vector for stable expression of a gene;
(2)
RNA interference (RNAi)-mediated transgenics is made to decrease the expression of undesirable traits by adding sense and antisense of the target gene with an intronic sequence. When this cassette is introduced into the plant genome, the target gene expression is significantly decreased;
(3)
Transcription Activator-Like Effector Nucleases (TALENs) mediated genome editing, which is utilized for generating non-GMO gene modification as well;
(4)
Clustered Regularly Interspaced Short Palindromic Nucleases (CRISPR)-associated (Cas) system-mediated genome-editing in potato plants. For this approach, a double-strand break is produced in the undesirable gene coding for an undesirable character. After a successful interruption, mutations are produced as required.

4.1. Conventional Potato Transgenics

4.1.1. Transgenics to Improve Tuber Quality and Yield

An overview of the development of transgenic potato through agrobacterium-mediated transformation is given in Figure 2. Few examples in the modification of genes in potatoes via different transgenic engineering techniques are given in Table 2.
Starch yield has been increased by modifying potato source and sink capacities. This purpose was achieved by increasing source capacity via overexpression of mesophyll-specific pyrophosphatases or by producing antisense expression of ADP-glucose pyrophosphorylase in potato leaves. Re-routing of photoassimilates was carried out to deploy both approaches, thus making use of sink organs by consuming the leaf starch. Instantaneous enhancement in sink capacity was carried out by increasing the expression of two plastidic metabolite translocators, i.e., an adenylate translocator in potato tubers and a glucose 6-phosphate/phosphate translocator. Using this ‘pull’ approach, an increase in potato starch contents and starch yield have also been documented when sink strength is increased. In the recent biotechnological approaches, source and sink capacities were successfully enhanced by combining the “push” and “pull” approaches using two different attempts. This method led to two-fold increase in the starch yield in tubers. This successful approach can also be applied to other crop plants in the future [63]. The foreign sucrose-phosphate synthase gene has been successfully introduced in potatoes to improve the supply of photosynthate from leaves (source) down to the tubers (sink), resulting in better quantity and quality of potato tubers [65]. The agrobacterium auxin biosynthesis gene has also been successfully inserted in potato lines to enhance indole acetic acid contents in tubers and tuber formation [66]. Insertion of purple acid phosphatase 2 of Arabidopsis (AtPAP2) in potato lines resulted in increased tuber yield and tuber starch content [64]. Tuber size has also been improved in potato lines by inserting Arabidopsis jasmonic acid carboxyl methyltransferase gene [93].
Production of amino acids using genetic modified potato crops is particularly important for improving human nourishment [67]. Functional genomics studies conducted on the biosynthesis of amino acids have revealed that sucrose supply directly affects the increase in amino acids at the cell level and is fully controlled at transcription level, thus mediating the biosynthesis of amino acids in potatoes [94]. The methionine contents have been enhanced by increasing the expression of CgSΔ90 in transgenic potato lines [95]. Cytidine base editor (CBE) was used to amino acid alterations, which led to a loss of function locus after substitutions of the KTGGL-encoding locus [96]. Scientists have successfully developed engineered potatoes inserted with the amaranth seed albumin (AmA1) gene or sunflower albumin gene increasing total methionine level 5–7 folds in tubers [67]. Different approaches have also been used to enhance methionine in potato plants by molecular breeders and biotechnologists [97,98,99]. Cysteine biosynthesis is associated with O-acetyl-L-serine produced by serine acetyltransferase (SAT). The SAT-encoding gene cysE was transformed in the white lady cultivar of potato, which significantly enhanced the transcript level of glutathione and cysteine in tubers to 1.5-fold higher on average than in the control plants [68].
Vitamins are pivotal in maintaining human health by mediating the metabolic system and bringing the metabolic processes associated with the energy in the tissues of living organisms obtained from food or other sources [100]. Many studies have been conducted on developing genetically modified potato tubers with increased vitamins. Vitamin A is functional in developing visual pigments, rod and cone cells inside the eye’s retina, synthesized from the precursor ß-carotene from potato. In potato, major carotenoids including antheraxanthin, lutein, xanthophyll, esters, and violaxanthin have been identified [101]. Researchers have successfully increased carotenoids to several folds in transgenic potatoes [70,102]. Research works have also revealed that the insertion of genes associated with the pathway of carotenoid biosynthesis along with the cauliflower orange (Or) gene in the potato genome increased the tuber astaxanthin contents successfully [69,71]. For successful commercial production of valuable ketocarotenoids in potato, the 3, 3′ β-hydroxylase (crtZ) and the 4, 4′ β-oxygenase (crtW) genes along with a suitable promoter were successfully inserted in the potato genome, which resulted in increased production of cellular carotenoids to form a wide array of ketolated and hydroxylated derivatives [103]. The crtB gene was successfully engineered in the potato genome using Agrobacterium to increase the carotenoid contents 4–7 times in Solanum tuberosum [104]. An algal bkt1 gene, encoding a β-ketolase, has also been successfully transformed into the potato genome to enhance the levels of ketocarotenoid in potato tubers [71]. Similarly, the potato has been transformed by adding the crtO gene to successfully produce ketocarotenoids like astaxanthin (3,3′-dihydroxy 4,4′-diketo-β-carotene) [70].
Researchers have significantly enhanced potatoes’ ascorbic acid (AsA, vitamin C) contents by increasing the production of recycling ascorbate by inserting the dehydroascorbate reductase (DHAR) gene [74].
Vitamin E (α-tocopherol), a potential antioxidant, is biosynthesized by photosynthetic organisms and is crucial for human health, but it should be consumed at the sub-optimal level. Scientists have successfully overexpressed vitamin E in potato tuber by inserting with two vitamin-E biosynthetic genes, homogentisate phytyl transferase (At-HPT) and p- hydroxyphenylpyruvate dioxygenase (At-HPPD) [75]. Vitamin B6, an essential metabolite, is mandatory for living organisms because it is a cofactor in various biochemical reactions in the body. It is documented as a potential antioxidant molecule that regulates the protein expression involved in the scavenging of cellular reactive oxygen species. The PDXII gene was successfully inserted in potato lines to enhance the production of vitamin B6 [76].

4.1.2. Transgenics to Improve Tuber Storage Duration

Successful engineering of novel RING finger gene SbRFP1 in potato plants inhibited invertase and β-amylase activity. The activity of these enzymes decreased the degradation of sucrose and starch and thus decelerated the accumulation of reducing sugars in tubers [77].
Since the activity of vacuolar invertase (StvacINV1) is importantly involved in the CIS process, the invertase inhibitors can play roles in the inhibition of StvacINV1 activity. Scientists have successfully decreased the activity of StvacINV1 by adding two putative inhibitors (StInvInh2A and StInvInh2B) to the potato genome, which resulted in reduced accumulation of reducing sugars and acrylamide in cold-stored tubers and thus the CIS [78,79]. Sugar metabolism was significantly modified by inserting bacterial genes encoding phosphofructokinase to decrease CIS, and this addition did not hinder any growth process of the plant [80].

4.1.3. Transgenics to Improve Tuber Morphology

Better tuber color and enhanced anthocyanin contents was successfully achieved by inserting a potato UDP-glucose: flavonoid-3-O-glucosyltransferase (3GT) gene in potato cultivar Désirée plants via agrobacterium-mediated transformation [105]. Similarly, the R2R3 MYB gene was added to the potato genome to increase the accumulation of pigments in the potato line [106].

4.2. RNAi for Potato Improvement

Since its discovery, RNAi technology has gained popularity in plant science [107]. Gene silencing at the post-transcription level has gained optimal results, which can be further applied in crop improvement.

4.2.1. RNAi Transgenics to Improve Tuber Quality

The RNAi plant expression vectors including sense and antisense fragments of the soluble starch synthase (SSIII) gene was constructed and transformed into potato cultivars using Agrobacterium-mediated transformation method [81]. The transgenic potatoes produced different starch granules. The amylose content of starch was increased by 2.68–29.05%, while the amylopectin to amylose ratio and the phosphorus contents in the starch was significantly reduced compared to the control plants. So, it has been established that RNAi-mediated plants can be used for starch improvement in potato plants [81].

4.2.2. RNAi Transgenics to Improve Tuber Storage Duration

The vacuolar invertase gene (VInv) was downregulated to more than 90%, reducing the sugars during cold storage by ~93% in the RNAi-mediated transgenic lines. Potato chips made with these transgenics were light in color because of the significantly low acrylamide content [84]. RNAi-mediated transgenics with the Désirée cultivar have also been produced to target the vacuolar invertase (VInv) gene, which converts starch into reducing sugars. The results were quite promising, like conventional transgenics, thus RNAi can be successfully used to reduce CIS in engineered potato tubers [83,84,85].
The activity of sucrose-phosphatase (SPP) was decreased in transgenic potatoes by RNAi, resulting in the decreased expression of vacuolar invertase upon cold treatment and thus decreased the CIS [86].
Acid invertases are key drivers among various enzymes, which playing an imperative role in sucrose to reducing sugar conversion. An average of 69.8% reduction in the expression of acid invertase in RNAi-mediated transgenic lines, decreased the CIS considerably. Compared to well-inhibited antisense invertase transgenic plants, RNAi mediated downregulation significant changed the invertase activity endogenously, showing its post-transcriptional gene silencing potential strategy in amelioration of cold sweetening in potato storage [108].

4.3. Genome Editing Technologies

4.3.1. TALENs Mediated Potato Modifications

TALENs have been tested and proven functional in several studies of potato genome editing [109]. The acetolactate synthesis (ALS) gene encodes acetohydroxy acid synthase which catalyzes the first step in the synthesis of branched amino acid and is sensitive to a number of herbicides such as imazamox [110]. By using the TALEN system, the mutated ALS gene was coupled for the targeted integration of foreign genes into the host potato [111]. TALENs technology successfully improved tuber potato by vacuolar invertase activity interruption, which reduced CIS and reduced sugars [89]. Starch alterations were made by Kusano et al. [87], who designed the “Emerald-Gateway TALEN system”, a unique delivery system, and targeted granule-bound starch synthase (GBSS) gene in the host for site-specific mutation. It has its role in starch biosynthesis in granulation, thus modifying starch quality.

4.3.2. CRISPR-Cas Mediated Potato Modifications

The overview of CRISPR-Cas9 mediated gene editing in an organism is depicted in Figure 3. The phytoene desaturase (PDS) has an imperative role in the carotenoid biosynthesis pathway, and caused the depigmentation due to the knockdown of PDS in the edited plants. Different sgRNAs have been utilized to decrease the activity of phytoene desaturase (PDS) by targeting its different sites. However, the unpaired nucleotides of target DNA can stimulate or decrease the activity of the Cas9-sgRNA complex in vitro according to the mismatch position [112].
The GBSS gene is considered to have a higher efficiency at the protoplast level. Replacing the Arabidopsis U6 promoter with endogenous potato U6 promoter, resulting in 35% editing of the allelic genes in the ex-plants. However, a significant reduction in laborious cell culturing, regeneration of ex-plants, and tedious screening procedures of potatoes can be avoided with this tool [113]. Potato protoplasts were transfected with Ribonucleoprotein complexes (RNPs), consisting of sgRNAs and Cas9 nuclease. It resulted in mutations happened in around 68% of regenerated plants with at least one allele of the target gene. Successful mutations in the StPPO2 gene resulted in a 69% reduction in the PPO activity in tubers and a 73% reduction in enzymatic browning compared to the control. Concludingly, a significant decrease in CIS can be made by CRISPR/Cas9 mediated genome editing [91].
The StGBSS gene was targeted via CRISPR-associated nuclease 9/guide-RNA (Cas9/gRNA), resulting in 25% mutagenesis in edited plants for GBSS [104]. Consequently, no enzyme activity related to carotenoids was observed due to the successful silencing of the StGBSS gene [112]. CRISPR-Cas9 has successfully altered the amylose-amylopectin ratio of potato starch. Gene encoding for GBSS was targeted from different sites, resulting in 67% mutant alleles in the edited plants. Full knockout of GBSS resulted in four allele-mutated lines of potato. Knockout of amylose alleles led to an enhanced amount of amylopectin contents [90].
The steroidal glycoalkaloids (SGAs) add a bitter taste to the potato tubers and have shown toxicity to various organisms. Fortunately, scientists have successfully decreased the SGA contents in the potato tubers. Nine candidate guide RNAs (gRNA) were prepared to target St16DOX in order to manipulate the characteristics of potato tubers according to the customer preference [92]. The gene StSSR2 was targeted, which decreased the level of SGAs in edited plants by 44% in leaves and 66% in tubers [114].

4.4. Production of Non-GMOs Using Advanced Biotechnological Techniques

In this modern age of technology, there are several ethical, scientific, and public concerns about releasing genetically modified organisms (GMOs) as cultivars. The organism genetic components are better undisturbed for food security purpose except the acceptable genome modifications. TALENs and CRISPR-Cas9 have the potential to realize the objectives that genetically new but transgene-free plants can be obtained in T2 mutant lines after segregating generations from T0 [115].

5. Conclusions

QTL mapping has provided the molecular markers for marker assisted selection for potato yield and quality improvement. Genomic selection uses genome wide markers and facilitates rapid selection of potato breeding lines with superior yield and quality performance and increase the accuracy of selections. Many improvements in potato yield and nutrition levels regarding starch contents, protein contents, and vitamins, among other, have been achieved by conventional transgenic engineering and current genome editing technologies, e.g., TALENS and CRISPR-Cas9. Future researches can be focused on production of various vaccines in potatoes, which can easily be used to fight global pandemics like coronavirus, hepatitis, ebolavirus. Since population rise and arable land decline have always threatened our world, these new technologies are expected to increase yield and nutrition in potatoes that provide insights to fighting potential hunger in the growing world.

Author Contributions

Conceptualization, J.B. and X.X.; writing—original draft preparation, D.A.; H.R. and J.B.; writing—review and editing, Z.Z. and J.B.; funding acquisition, J.B. and X.X. All authors have read and agreed to the published version of the manuscript.

Funding

This research is supported by the Starry Night Science Fund of the Zhejiang University Shanghai Institute for Advanced Study (grant No. SN-ZJU-SIAS-0012).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Devaux, A.; Goffart, J.-P.; Petsakos, A.; Kromann, P.; Gatto, M.; Okello, J.; Suarez, V.; Hareau, G. Global Food Security, Contributions from Sustainable Potato Agri-Food Systems. In The Potato Crop: Its Agricultural, Nutritional and Social Contribution to Humankind; Campos, H., Ortiz, O., Eds.; Springer International Publishing: Cham, Switzerland, 2020; pp. 3–35. [Google Scholar] [CrossRef]
  2. Alexandratos, N.; Bruinsma, J. World Agriculture towards 2030/2050: The 2012 Revision. 2012. Available online: https://www.fao.org/3/ap106e/ap106e.pdf (accessed on 28 June 2022).
  3. Hameed, A.; Zaidi, S.S.E.A.; Shakir, S.; Mansoor, S. Applications of new breeding technologies for potato improvement. Front. Plant Sci. 2018, 9, 925. [Google Scholar] [CrossRef] [PubMed]
  4. FAO. FAOSTAT; Food and Agriculture Organization of the United Nations: Rome, Italy, 2019. [Google Scholar]
  5. Rasheed, H.; Ahmad, D.; Bao, J. Genetic diversity and health properties of polyphenols in potato. Antioxidants 2022, 11, 603. [Google Scholar] [CrossRef] [PubMed]
  6. White, P.J.; Broadley, M.R.; Hammond, J.P.; Ramsay, G.; Subramanian, N.K.; Thompson, J.; Wright, G. Bio-fortification of potato tubers using foliar zinc-fertiliser. J. Hortic. Sci. Biotechnol. 2012, 87, 123–129. [Google Scholar] [CrossRef]
  7. Ngobese, N.Z.; Workneh, T.S.; Alimi, B.A.; Tesfay, S. Nutrient composition and starch characteristics of eight European potato cultivars cultivated in South Africa. J. Food Compos. Anal. 2017, 55, 1–11. [Google Scholar] [CrossRef]
  8. Al-Saikhan, M.; Howard, L.; Miller, J., Jr. Antioxidant activity and total phenolics in different genotypes of potato (Solanum tuberosum, L.). J. Food Sci. 1995, 60, 341–343. [Google Scholar] [CrossRef]
  9. Tiwari, A.; Mishra, A.; Saxena, V.; Saxena, S.; Raghuvanshi, A.P. Association of characters and selection parameters for high tuber yield in Potato (Solanum tuberosum L.). Trop. Plant Res. 2020, 7, 205–208. [Google Scholar] [CrossRef]
  10. Andrivon, D. Potato facing global challenges: How, how much, how well? Potato Res. 2017, 60, 389–400. [Google Scholar] [CrossRef]
  11. Ortiz, R.; Mihovilovich, E. Genetics and Cytogenetics of the Potato. In The Potato Crop; Springer: Cham, Switzerland, 2020; pp. 219–247. [Google Scholar] [CrossRef]
  12. Das Dangol, S.; Barakate, A.; Stephens, J.; Çalıskan, M.E.; Bakhsh, A. Genome editing of potato using CRISPR technologies: Current development and future prospective. Plant Cell Tissue Organ Cult. (PCTOC) 2019, 139, 403–416. [Google Scholar] [CrossRef]
  13. Thiele, G.; Dufour, D.; Vernier, P.; Mwanga, R.O.M.; Parker, M.L.; Schulte Geldermann, E.; Teeken, B.; Wossen, T.; Gotor, E.; Kikulwe, E.; et al. A review of varietal change in roots, tubers and bananas: Consumer preferences and other drivers of adoption and implications for breeding. Int. J. Food Sci. Technol. 2020, 56, 1076–1092. [Google Scholar] [CrossRef]
  14. Bethke, P.C.; Halterman, D.A.; Jansky, S.H. Potato germplasm enhancement enters the genomics era. Agronomy 2019, 9, 575. [Google Scholar] [CrossRef]
  15. Hardigan, M.A.; Bamberg, J.; Buell, C.R.; Douches, D.S. Taxonomy and genetic differentiation among wild and cultivated germplasm of Solanum sect. Petota. Plant Genome 2015, 8, plantgenome2014.06.0025. [Google Scholar] [CrossRef] [PubMed]
  16. Werij, J.S.; Furrer, H.; van Eck, H.J.; Visser, R.G.F.; Bachem, C.W.B. A limited set of starch related genes explain several interrelated traits in potato. Euphytica 2012, 186, 501–516. [Google Scholar] [CrossRef]
  17. Śliwka, J.; Sołtys-Kalina, D.; Szajko, K.; Wasilewicz-Flis, I.; Strzelczyk-Żyta, D.; Zimnoch-Guzowska, E.; Jakuczun, H.; Marczewski, W. Mapping of quantitative trait loci for tuber starch and leaf sucrose contents in diploid potato. Theor. Appl. Genet. 2016, 129, 131–140. [Google Scholar] [CrossRef]
  18. Li, J.; Wang, Y.; Wen, G.; Li, G.; Li, Z.; Zhang, R.; Ma, S.; Zhou, J.; Xie, C. Mapping QTL underlying tuber starch content and plant maturity in tetraploid potato. Crop J. 2019, 7, 261–272. [Google Scholar] [CrossRef]
  19. Draffehn, A.M.; Meller, S.; Li, L.; Gebhardt, C. Natural diversity of potato (Solanum tuberosum) invertases. BMC Plant Biol. 2010, 10, 271. [Google Scholar] [CrossRef]
  20. Li, L.; Paulo, M.-J.; Strahwald, J.; Lübeck, J.; Hofferbert, H.-R.; Tacke, E.; Junghans, H.; Wunder, J.; Draffehn, A.; van Eeuwijk, F.; et al. Natural DNA variation at candidate loci is associated with potato chip color, tuber starch content, yield and starch yield. Theor. Appl. Genet. 2008, 116, 1167–1181. [Google Scholar] [CrossRef]
  21. Xiao, G.; Huang, W.; Cao, H.; Tu, W.; Wang, H.; Zheng, X.; Liu, J.; Song, B.; Xie, C. Genetic loci conferring reducing sugar accumulation and conversion of cold-stored potato tubers revealed by QTL analysis in a diploid population. Front. Plant Sci. 2018, 9, 315. [Google Scholar] [CrossRef]
  22. Zia, M.A.B.; Demirel, U.; Nadeem, M.A.; Çaliskan, M.E. Genome-wide association study identifies various loci underlying agronomic and morphological traits in diversified potato panel. Physiol. Mol. Biol. Plants 2020, 26, 1003–1020. [Google Scholar] [CrossRef]
  23. Park, J.; Massa, A.N.; Douches, D.; Coombs, J.; Akdemir, D.; Yencho, G.C.; Whitworth, J.L.; Novy, R.G. Linkage and QTL mapping for tuber shape and specific gravity in a tetraploid mapping population of potato representing the russet market class. BMC Plant Biology 2021, 21, 507. [Google Scholar] [CrossRef] [PubMed]
  24. Santacruz, S.; Koch, K.; Andersson, R.; Åman, P. Characterization of potato leaf starch. J. Agric. Food Chem. 2004, 52, 1985–1989. [Google Scholar] [CrossRef] [PubMed]
  25. Buleon, A.; Colonna, P.; Planchot, V.; Ball, S. Starch granules: Structure and biosynthesis. Int. J. Biol. Macromol. 1998, 23, 85–112. [Google Scholar] [CrossRef]
  26. Schreiber, L.; Nader-Nieto, A.C.; Schönhals, E.M.; Walkemeier, B.; Gebhardt, C. SNPs in genes functional in starch-sugar interconversion associate with natural variation of tuber starch and sugar content of potato (Solanum tuberosum L.). G3 Genes|Genomes|Genet. 2014, 4, 1797–1811. [Google Scholar] [CrossRef] [PubMed]
  27. Ahmed, S.; Zhou, X.; Pang, Y.; Jin, L.; Bao, J. Improving starch-related traits in potato crops: Achievements and future challenges. Starch—Stärke 2018, 70, 1700113. [Google Scholar] [CrossRef]
  28. Xu, Y.; Ding, J.; Gong, S.; Li, M.; Yang, T.; Zhang, J. Physicochemical properties of potato starch fermented by amylolytic Lactobacillus plantarum. Int. J. Biol. Macromol. 2020, 158, 656–661. [Google Scholar] [CrossRef] [PubMed]
  29. Slattery, C.J.; Kavakli, I.H.; Okita, T.W. Engineering starch for increased quantity and quality. Trends Plant Sci. 2000, 5, 291–298. [Google Scholar] [CrossRef]
  30. Van Harsselaar, J.K.; Lorenz, J.; Senning, M.; Sonnewald, U.; Sonnewald, S. Genome-wide analysis of starch metabolism genes in potato (Solanum tuberosum L.). BMC Genom. 2017, 18, 37. [Google Scholar] [CrossRef]
  31. Khlestkin, V.K.; Rozanova, I.V.; Efimov, V.M.; Khlestkina, E.K. Starch phosphorylation associated SNPs found by genome-wide association studies in the potato (Solanum tuberosum L.). BMC Genet. 2019, 20, 45–53. [Google Scholar] [CrossRef]
  32. Carpenter, M.A.; Joyce, N.I.; Genet, R.A.; Cooper, R.D.; Murray, S.R.; Noble, A.D.; Butler, R.C.; Timmerman-Vaughan, G.M. Starch phosphorylation in potato tubers is influenced by allelic variation in the genes encoding glucan water dikinase, starch branching enzymes I and II, and starch synthase III. Front. Plant Sci. 2015, 6, 143. [Google Scholar] [CrossRef] [Green Version]
  33. Li, L.; Tacke, E.; Hofferbert, H.-R.; Lübeck, J.; Strahwald, J.; Draffehn, A.M.; Walkemeier, B.; Gebhardt, C. Validation of candidate gene markers for marker-assisted selection of potato cultivars with improved tuber quality. Theor. Appl. Genet. 2013, 126, 1039–1052. [Google Scholar] [CrossRef]
  34. Klaassen, M.T.; Bourke, P.M.; Maliepaard, C.; Trindade, L.M. Multi-allelic QTL analysis of protein content in a bi-parental population of cultivated tetraploid potato. Euphytica 2019, 215, 14. [Google Scholar] [CrossRef]
  35. Zhang, H.; Hou, J.; Liu, J.; Zhang, J.; Song, B.; Xie, C. The roles of starch metabolic pathways in the cold-induced sweetening process in potatoes. Starch—Stärke 2017, 69, 1600194. [Google Scholar] [CrossRef]
  36. 36 Fischer, M.; Schreiber, L.; Colby, T.; Kuckenberg, M.; Tacke, E.; Hofferbert, H.-R.; Schmidt, J.; Gebhardt, C. Novel candidate genes influencing natural variation in potato tuber cold sweetening identified by comparative proteomics and association mapping. BMC Plant Biol. 2013, 13, 113. [Google Scholar] [CrossRef] [PubMed]
  37. Maddison, A.L.; Hedley, P.E.; Meyer, R.C.; Aziz, N.; Davidson, D.; Machray, G.C. Expression of tandem invertase genes associated with sexual and vegetative growth cycles in potato. Plant Mol. Biol. 1999, 41, 741–752. [Google Scholar] [CrossRef] [PubMed]
  38. Bisognin, D.; Manrique-Carpintero, N.; Douches, D. QTL analysis of tuber dormancy and sprouting in potato. Am. J. Potato Res. 2018, 95, 374–382. [Google Scholar] [CrossRef]
  39. Hara-Skrzypiec, A.; Śliwka, J.; Jakuczun, H.; Zimnoch-Guzowska, E. QTL for tuber morphology traits in diploid potato. J. Appl. Genet. 2018, 59, 123–132. [Google Scholar] [CrossRef]
  40. Manrique-Carpintero, N.C.; Coombs, J.J.; Pham, G.M.; Laimbeer, F.P.E.; Braz, G.T.; Jiang, J.; Veilleux, R.E.; Buell, C.R.; Douches, D.S. Genome reduction in tetraploid potato reveals genetic load, haplotype variation, and loci associated with agronomic traits. Front. Plant Sci. 2018, 9, 944. [Google Scholar] [CrossRef]
  41. Endelman, J.B.; Jansky, S.H. Genetic mapping with an inbred line-derived F2 population in potato. Theor. Appl. Genet. 2016, 129, 935–943. [Google Scholar] [CrossRef]
  42. Bradshaw, J.; Mackay, G. Breeding strategies for clonally propagated potatoes. In Potato Genetics; Bradshaw, J.E., Mackay, G.R., Eds.; CAB International: Wallingford, UK, 1994; pp. 467–497. [Google Scholar]
  43. Bradshaw, J.E. Potato-breeding strategy. In Potato Biology and Biotechnology; Elsevier: Amsterdam, The Netherlands, 2007; pp. 157–177. [Google Scholar]
  44. Jansky, S.H.; Jin, L.P.; Xie, K.Y.; Xie, C.H.; Spooner, D.M. Potato production and breeding in China. Potato Res. 2009, 52, 57–65. [Google Scholar] [CrossRef]
  45. Gebhardt, C. Bridging the gap between genome analysis and precision breeding in potato. Trends Genet. 2013, 29, 248–256. [Google Scholar] [CrossRef]
  46. Slater, A.T.; Cogan, N.O.I.; Hayes, B.J.; Schultz, L.; Dale, M.F.B.; Bryan, G.J.; Forster, J.W. Improving breeding efficiency in potato using molecular and quantitative genetics. Theor. Appl. Genet. 2014, 127, 2279–2292. [Google Scholar] [CrossRef]
  47. Meuwissen, T.H.E.; Hayes, B.J.; Goddard, M.E. Prediction of total genetic value using genome-wide dense marker maps. Genetics 2001, 157, 1819–1829. [Google Scholar] [CrossRef] [PubMed]
  48. Jonas, E.; de Koning, D.-J. Does genomic selection have a future in plant breeding? Trends Biotechnol. 2013, 31, 497–504. [Google Scholar] [CrossRef] [PubMed]
  49. Crossa, J.; Pérez-Rodríguez, P.; Cuevas, J.; Montesinos-López, O.; Jarquín, D.; De Los Campos, G.; Burgueño, J.; González-Camacho, J.M.; Pérez-Elizalde, S.; Beyene, Y. Genomic selection in plant breeding: Methods, models, and perspectives. Trends Plant Sci. 2017, 22, 961–975. [Google Scholar] [CrossRef] [PubMed]
  50. Sverrisdóttir, E.; Byrne, S.; Sundmark, E.H.R.; Johnsen, H.Ø.; Kirk, H.G.; Asp, T.; Janss, L.; Nielsen, K.L. Genomic prediction of starch content and chipping quality in tetraploid potato using genotyping-by-sequencing. Theor. Appl. Genet. 2017, 130, 2091–2108. [Google Scholar] [CrossRef]
  51. Stich, B.; Van Inghelandt, D. Prospects and potential uses of genomic prediction of key performance traits in tetraploid potato. Front. Plant Sci. 2018, 9, 159. [Google Scholar] [CrossRef] [PubMed]
  52. Endelman, J.B.; Carley, C.A.S.; Bethke, P.C.; Coombs, J.J.; Clough, M.E.; da Silva, W.L.; De Jong, W.S.; Douches, D.S.; Frederick, C.M.; Haynes, K.G.; et al. Genetic variance partitioning and genome-wide prediction with allele dosage information in autotetraploid potato. Genetics 2018, 209, 77–87. [Google Scholar] [CrossRef] [PubMed]
  53. Sverrisdóttir, E.; Sundmark, E.H.R.; Johnsen, H.Ø.; Kirk, H.G.; Asp, T.; Janss, L.; Bryan, G.; Nielsen, K.L. The value of expanding the training population to improve genomic selection models in tetraploid potato. Front. Plant Sci. 2018, 9, 1118. [Google Scholar] [CrossRef]
  54. Habyarimana, E.; Parisi, B.; Mandolino, G.; Wehling, P. Genomic prediction for yields, processing and nutritional quality traits in cultivated potato (Solanum tuberosum L.). Plant Breed. 2017, 136, 245–252. [Google Scholar] [CrossRef]
  55. Sallam, A.H.; Endelman, J.B.; Jannink, J.-L.; Smith, K.P. Assessing genomic selection prediction accuracy in a dynamic barley breeding Population. Plant Genome 2015, 8, plantgenome2014-05. [Google Scholar] [CrossRef]
  56. Zhang, C.; Yang, Z.; Tang, D.; Zhu, Y.; Wang, P.; Li, D.; Zhu, G.; Xiong, X.; Shang, Y.; Li, C.; et al. Genome design of hybrid potato. Cell 2021, 184, 3873–3883. [Google Scholar] [CrossRef]
  57. Tang, D.; Jia, Y.; Zhang, J.; Li, H.; Cheng, L.; Wang, P.; Bao, Z.; Liu, Z.; Feng, S.; Zhu, X.; et al. Genome evolution and diversity of wild and cultivated potatoes. Nature 2022, 606, 535–541. [Google Scholar] [CrossRef] [PubMed]
  58. Naeem, M.; Demirel, U.; Yousaf, M.F.; Caliskan, S.; Caliskan, M.E. Overview on domestication, breeding, genetic gain and improvement of tuber quality traits of potato using fast forwarding technique (GWAS): A review. Plant Breed. 2021, 140, 519–542. [Google Scholar] [CrossRef]
  59. Gaj, T.; Gersbach, C.A.; Barbas, C.F. ZFN, TALEN, and CRISPR/Cas-based methods for genome engineering. Trends Biotechnol. 2013, 31, 397–405. [Google Scholar] [CrossRef] [PubMed]
  60. Fichtner, F.; Urrea Castellanos, R.; Ülker, B. Precision genetic modifications: A new era in molecular biology and crop improvement. Planta 2014, 239, 921–939. [Google Scholar] [CrossRef] [PubMed]
  61. Christian, M.; Cermak, T.; Doyle, E.L.; Schmidt, C.; Zhang, F.; Hummel, A.; Bogdanove, A.J.; Voytas, D.F. Targeting DNA double-strand breaks with TAL effector nucleases. Genetics 2010, 186, 757–761. [Google Scholar] [CrossRef]
  62. Zhu, X.; Richael, C.; Chamberlain, P.; Busse, J.S.; Bussan, A.J.; Jiang, J.; Bethke, P.C. Vacuolar invertase gene silencing in potato (Solanum tuberosum L.) improves processing quality by decreasing the frequency of sugar-end defects. PLoS ONE 2014, 9, e93381. [Google Scholar] [CrossRef]
  63. Jonik, C.; Sonnewald, U.; Hajirezaei, M.-R.; Flügge, U.-I.; Ludewig, F. Simultaneous boosting of source and sink capacities doubles tuber starch yield of potato plants. Plant Biotechnol. J. 2012, 10, 1088–1098. [Google Scholar] [CrossRef]
  64. Zhang, Y.; Sun, F.; Fettke, J.; Schöttler, M.A.; Ramsden, L.; Fernie, A.R.; Lim, B.L. Heterologous expression of AtPAP2 in transgenic potato influences carbon metabolism and tuber development. FEBS Lett. 2014, 588, 3726–3731. [Google Scholar] [CrossRef] [Green Version]
  65. Ishimaru, K.; Hirotsu, N.; Kashiwagi, T.; Madoka, Y.; Nagasuga, K.; Ono, K.; Ohsugi, R. Overexpression of a maize SPS gene improves yield characters of potato under field conditions. Plant Prod. Sci. 2008, 11, 104–107. [Google Scholar] [CrossRef]
  66. Kolachevskaya, O.O.; Alekseeva, V.V.; Sergeeva, L.I.; Rukavtsova, E.B.; Getman, I.A.; Vreugdenhil, D.; Buryanov, Y.I.; Romanov, G.A. Expression of auxin synthesis gene tms1 under control of tuber-specific promoter enhances potato tuberization in vitro. J. Integr. Plant Biol. 2015, 57, 734–744. [Google Scholar] [CrossRef]
  67. Chakraborty, S.; Chakraborty, N.; Datta, A. Increased nutritive value of transgenic potato by expressing a nonallergenic seed albumin gene from Amaranthus hypochondriacus. Proc. Natl. Acad. Sci. USA 2000, 97, 3724–3729. [Google Scholar] [CrossRef]
  68. Stiller, I.; Dancs, G.; Hesse, H.; Hoefgen, R.; Banfalvi, Z. Improving the nutritive value of tubers: Elevation of cysteine and glutathione contents in the potato cultivar White Lady by marker-free transformation. J. Biotechnol. 2007, 128, 335–343. [Google Scholar] [CrossRef] [PubMed]
  69. Lopez, A.B.; Van Eck, J.; Conlin, B.J.; Paolillo, D.J.; O’Neill, J.; Li, L. Effect of the cauliflower Or transgene on carotenoid accumulation and chromoplast formation in transgenic potato tubers. J. Exp. Bot. 2008, 59, 213–223. [Google Scholar] [CrossRef] [PubMed]
  70. Gerjets, T.; Sandmann, G. Ketocarotenoid formation in transgenic potato. J. Exp. Bot. 2006, 57, 3639–3645. [Google Scholar] [CrossRef]
  71. Campbell, R.; Morris, W.L.; Mortimer, C.L.; Misawa, N.; Ducreux, L.J.M.; Morris, J.A.; Hedley, P.E.; Fraser, P.D.; Taylor, M.A. Optimising ketocarotenoid production in potato tubers: Effect of genetic background, transgene combinations and environment. Plant Sci. 2015, 234, 27–37. [Google Scholar] [CrossRef] [PubMed]
  72. Morris, W.L.; Ducreux, L.J.; Fraser, P.D.; Millam, S.; Taylor, M.A. Engineering ketocarotenoid biosynthesis in potato tubers. Metab. Eng. 2006, 8, 253–263. [Google Scholar] [CrossRef] [PubMed]
  73. Song, X.-Y.; Zhu, W.-J.; Tang, R.-M.; Cai, J.-H.; Chen, M.; Yang, Q. Over-expression of StLCYb increases β-carotene accumulation in potato tubers. Plant Biotechnol. Rep. 2016, 10, 95–104. [Google Scholar] [CrossRef]
  74. Qin, A.; Shi, Q.; Yu, X. Ascorbic acid contents in transgenic potato plants overexpressing two dehydroascorbate reductase genes. Mol. Biol. Rep. 2010, 38, 1557–1566. [Google Scholar] [CrossRef]
  75. Crowell, E.F.; McGrath, J.M.; Douches, D.S. Accumulation of vitamin E in potato (Solanum tuberosum) tubers. Transgenic Res. 2007, 17, 205–217. [Google Scholar] [CrossRef]
  76. Bagri, D.S.; Upadhyaya, D.C.; Kumar, A.; Upadhyaya, C.P. Overexpression of PDX-II gene in potato (Solanum tuberosum L.) leads to the enhanced accumulation of vitamin B6 in tuber tissues and tolerance to abiotic stresses. Plant Sci. 2018, 272, 267–275. [Google Scholar] [CrossRef]
  77. Zhang, H.; Liu, X.; Liu, J.; Ou, Y.; Lin, Y.; Li, M.; Song, B.; Xie, C. A novel RING finger gene, SbRFP1, increases resistance to cold-induced sweetening of potato tubers. FEBS Lett. 2013, 587, 749–755. [Google Scholar] [CrossRef]
  78. Liu, X.; Lin, Y.; Liu, J.; Song, B.; Ou, Y.; Zhang, H.; Li, M.; Xie, C. StInvInh2 as an inhibitor of Stvac INV 1 regulates the cold-induced sweetening of potato tubers by specifically capping vacuolar invertase activity. Plant Biotechnol. J. 2013, 11, 640–647. [Google Scholar] [CrossRef] [PubMed]
  79. Liu, X.; Cheng, S.; Liu, J.; Ou, Y.; Song, B.; Zhang, C.; Lin, Y.; Li, X.-Q.; Xie, C. The potato protease inhibitor gene, St-Inh, plays roles in the cold-induced sweetening of potato tubers by modulating invertase activity. Postharvest Biol. Technol. 2013, 86, 265–271. [Google Scholar] [CrossRef]
  80. Navrátil, O.; Fischer, L.; Čmejlová, J.; Linhart, M.; Vacek, J. Decreased amount of reducing sugars in transgenic potato tubers and its influence on yield characteristics. Biol. Plant. 2007, 51, 56–60. [Google Scholar] [CrossRef]
  81. Du, H.-H.; Yang, T.; Ma, C.-Y.; Feng, D.; Zhang, N.; Si, H.-J.; Wang, D. Effects of RNAi silencing of SSIII gene on phosphorus content and characteristics of starch in potato tubers. J. Integr. Agric. 2012, 11, 1985–1992. [Google Scholar] [CrossRef]
  82. Visser, R.G.F.; Somhorst, I.; Kuipers, G.J.; Ruys, N.J.; Feenstra, W.J.; Jacobsen, E. Inhibition of the expression of the gene for granulebound starch synthase in potato by antisense constructs. Mol. Gen. Genet. 1991, 225, 289–296. [Google Scholar] [CrossRef] [PubMed]
  83. Hameed, A.; Bilal, R.; Latif, F.; Van Eck, J.; Jander, G.; Mansoor, S. RNAi-mediated silencing of endogenous Vlnv gene confers stable reduction of cold-induced sweetening in potato (Solanum tuberosum L. cv. Désirée). Plant Biotechnol. Rep. 2018, 12, 175–185. [Google Scholar] [CrossRef]
  84. Wu, L.; Bhaskar, P.B.; Busse, J.S.; Zhang, R.; Bethke, P.C.; Jiang, J. Developing Cold-Chipping Potato Varieties by Silencing the Vacuolar Invertase Gene. Crop Sci. 2011, 51, 981–990. [Google Scholar] [CrossRef] [Green Version]
  85. Bhaskar, P.B.; Wu, L.; Busse, J.S.; Whitty, B.R.; Hamernik, A.J.; Jansky, S.H.; Buell, C.R.; Bethke, P.C.; Jiang, J. Suppression of the vacuolar invertase gene prevents cold-induced sweetening in potato. Plant Physiol. 2010, 154, 939–948. [Google Scholar] [CrossRef]
  86. Chen, S.; Hajirezaei, M.R.; Zanor, M.I.; Hornyik, C.; Debast, S.; Lacomme, C.; Fernie, A.R.; Sonnewald, U.; Boernke, F. RNA interference-mediated repression of sucrose-phosphatase in transgenic potato tubers (Solanum tuberosum) strongly affects the hexose-to-sucrose ratio upon cold storage with only minor effects on total soluble carbohydrate accumulation. Plant Cell Environ. 2008, 31, 165–176. [Google Scholar] [CrossRef]
  87. Kusano, H.; Onodera, H.; Kihira, M.; Aoki, H.; Matsuzaki, H.; Shimada, H. A simple Gateway-assisted construction system of TALEN genes for plant genome editing. Sci. Rep. 2016, 6, 30234. [Google Scholar] [CrossRef]
  88. Ma, J.; Xiang, H.; Donnelly, D.J.; Meng, F.-R.; Xu, H.; Durnford, D.; Li, X.-Q. Genome editing in potato plants by agrobacterium-mediated transient expression of transcription activator-like effector nucleases. Plant Biotechnol. Rep. 2017, 11, 249–258. [Google Scholar] [CrossRef]
  89. Clasen, B.M.; Stoddard, T.J.; Luo, S.; Demorest, Z.L.; Li, J.; Cedrone, F.; Tibebu, R.; Davison, S.; Ray, E.E.; Daulhac, A. Improving cold storage and processing traits in potato through targeted gene knockout. Plant Biotechnol. J. 2016, 14, 169–176. [Google Scholar] [CrossRef] [PubMed]
  90. Andersson, M.; Turesson, H.; Nicolia, A.; Fält, A.-S.; Samuelsson, M.; Hofvander, P. Efficient targeted multiallelic mutagenesis in tetraploid potato (Solanum tuberosum) by transient CRISPR-Cas9 expression in protoplasts. Plant Cell Rep. 2017, 36, 117–128. [Google Scholar] [CrossRef] [PubMed]
  91. González, M.N.; Massa, G.A.; Andersson, M.; Turesson, H.; Olsson, N.; Fält, A.-S.; Storani, L.; Décima Oneto, C.A.; Hofvander, P.; Feingold, S.E. Reduced Enzymatic Browning in Potato Tubers by Specific Editing of a Polyphenol Oxidase Gene via Ribonucleoprotein Complexes Delivery of the CRISPR/Cas9 System. Front. Plant Sci. 2020, 10, 1649. [Google Scholar] [CrossRef] [PubMed]
  92. Nakayasu, M.; Akiyama, R.; Lee, H.J.; Osakabe, K.; Osakabe, Y.; Watanabe, B.; Sugimoto, Y.; Umemoto, N.; Saito, K.; Muranaka, T.; et al. Generation of α-solanine-free hairy roots of potato by CRISPR/Cas9 mediated genome editing of the St16DOX gene. Plant Physiol. Biochem. 2018, 131, 70–77. [Google Scholar] [CrossRef]
  93. Sohn, H.B.; Lee, H.Y.; Seo, J.S.; Jung, C.; Jeon, J.H.; Kim, J.-H.; Lee, Y.W.; Lee, J.S.; Cheong, J.-J.; Choi, Y.D. Overexpression of jasmonic acid carboxyl methyltransferase increases tuber yield and size in transgenic potato. Plant Biotechnol. Rep. 2010, 5, 27–34. [Google Scholar] [CrossRef]
  94. Roessner-Tunali, U.; Urbanczyk-Wochniak, E.; Czechowski, T.; Kolbe, A.; Willmitzer, L.; Fernie, A.R. De Novo amino acid biosynthesis in potato tubers is regulated by sucrose levels. Plant Physiol. 2003, 133, 683–692. [Google Scholar] [CrossRef] [Green Version]
  95. Dancs, G.; Kondrák, M.; Bánfalvi, Z. The effects of enhanced methionine synthesis on amino acid and anthocyanin content of potato tubers. BMC Plant Biol. 2008, 8, 65. [Google Scholar] [CrossRef]
  96. Nikiforova, V.; Kempa, S.; Zeh, M.; Maimann, S.; Kreft, O.; Casazza, A.P.; Riedel, K.; Tauberger, E.; Hoefgen, R.; Hesse, H. Engineering of cysteine and methionine biosynthesis in potato. Amino Acids 2002, 22, 259–278. [Google Scholar] [CrossRef]
  97. Goo, Y.-M.; Kim, T.-W.; Lee, M.-K.; Lee, S.-W. Accumulation of PrLeg, a perilla legumin protein in potato tuber results in enhanced level of sulphur-containing amino acids. Comptes Rendus Biol. 2013, 336, 433–439. [Google Scholar] [CrossRef]
  98. Chong, D.K.X.; Chong, D.K.X.; Langridge, W.H.R. Expression of full-length bioactive antimicrobial human lactoferrin in potato plants. Transgenic Res. 2000, 9, 71–78. [Google Scholar] [CrossRef]
  99. Huang, T.; Joshi, V.; Jander, G. The catabolic enzyme methionine gamma-lyase limits methionine accumulation in potato tubers. Plant Biotechnol. J. 2014, 12, 883–893. [Google Scholar] [CrossRef] [PubMed]
  100. Jube, S.; Borthakur, D. Recent Advances in Food Biotechnology Research. In Food Biochemistry and Food Processing; Hui, Y.H., Ed.; Blackwell Publishing: Hoboken, NJ, USA, 2006; pp. 35–70. [Google Scholar] [CrossRef]
  101. Fernandez-Orozco, R.; Gallardo-Guerrero, L.; Hornero-Méndez, D. Carotenoid profiling in tubers of different potato (Solanum sp.) cultivars: Accumulation of carotenoids mediated by xanthophyll esterification. Food Chem. 2013, 141, 2864–2872. [Google Scholar] [CrossRef]
  102. Pasare, S.; Wright, K.; Campbell, R.; Morris, W.; Ducreux, L.; Chapman, S.; Bramley, P.; Fraser, P.; Roberts, A.; Taylor, M. The sub-cellular localisation of the potato (Solanum tuberosum L.) carotenoid biosynthetic enzymes, CrtRb2 and PSY2. Protoplasma 2013, 250, 1381–1392. [Google Scholar] [CrossRef]
  103. Mortimer, C.L.; Dugdale, B.; Dale, J.L. Updates in inducible transgene expression using viral vectors: From transient to stable expression. Curr. Opin. Biotechnol. 2015, 32, 85–92. [Google Scholar] [CrossRef]
  104. Ducreux, L.J.M. Metabolic engineering of high carotenoid potato tubers containing enhanced levels of beta-carotene and lutein. J. Exp. Bot. 2004, 56, 81–89. [Google Scholar] [CrossRef] [Green Version]
  105. Wei, Q.; Wang, Q.-Y.; Feng, Z.-H.; Wang, B.; Zhang, Y.-F.; Yang, Q. Increased accumulation of anthocyanins in transgenic potato tubers by overexpressing the 3GT gene. Plant Biotechnol. Rep. 2011, 6, 69–75. [Google Scholar] [CrossRef]
  106. Jung, C.S.; Griffiths, H.M.; De Jong, D.M.; Cheng, S.; Bodis, M.; Kim, T.S.; De Jong, W.S. The potato developer (D) locus encodes an R2R3 MYB transcription factor that regulates expression of multiple anthocyanin structural genes in tuber skin. Theor. Appl. Genet. 2009, 120, 45–57. [Google Scholar] [CrossRef]
  107. Fire, A.; Xu, S.; Montgomery, M.K.; Kostas, S.A.; Driver, S.E.; Mello, C.C. Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature 1998, 391, 806–811. [Google Scholar] [CrossRef]
  108. Chi, Z.; Cong-Hua, X.; Bo-Tao, S.; Xun, L.; Jun, L. RNAi effects on regulation of endogenous acid invertase activity in potato (Solanum tuberosum L.) tubers. Chin. J. Agric. Biotechnol. 2008, 5, 107–112. [Google Scholar] [CrossRef]
  109. Chen, K.; Gao, C. TALENs: Customizable molecular DNA scissors for genome engineering of plants. J. Genet. Genom. 2013, 40, 271–279. [Google Scholar] [CrossRef]
  110. Nicolia, A.; Proux-Wéra, E.; Åhman, I.; Onkokesung, N.; Andersson, M.; Andreasson, E.; Zhu, L.-H. Targeted gene mutation in tetraploid potato through transient TALEN expression in protoplasts. J. Biotechnol. 2015, 204, 17–24. [Google Scholar] [CrossRef] [PubMed]
  111. Forsyth, A.; Weeks, T.; Richael, C.; Duan, H. Transcription activator-like effector nucleases (TALEN)-mediated targeted DNA insertion in potato plants. Front. Plant Sci. 2016, 7, 1572. [Google Scholar] [CrossRef]
  112. Khromov, A.V.; Gushchin, V.A.; Timerbaev, V.I.; Kalinina, N.O.; Taliansky, M.E.; Makarov, V.V. Guide RNA Design for CRISPR/Cas9-Mediated Potato Genome Editing. Dokl. Biochem. Biophys. 2018, 479, 90–94. [Google Scholar] [CrossRef]
  113. Johansen, I.E.; Liu, Y.; Jørgensen, B.; Bennett, E.P.; Andreasson, E.; Nielsen, K.L.; Blennow, A.; Petersen, B.L. High efficacy full allelic CRISPR/Cas9 gene editing in tetraploid potato. Sci. Rep. 2019, 9, 17715. [Google Scholar] [CrossRef]
  114. Zheng, Z.; Ye, G.; Zhou, Y.; Pu, X.; Su, W.; Wang, J. Editing sterol side chain reductase 2 gene (StSSR2) via CRISPR/Cas9 reduces the total steroidal glycoalkaloids in potato. All Life 2021, 14, 401–413. [Google Scholar] [CrossRef]
  115. Ricroch, A.; Clairand, P.; Harwood, W. Use of CRISPR systems in plant genome editing: Toward new opportunities in agriculture. Emerg. Top. Life Sci. 2017, 1, 169–182. [Google Scholar] [CrossRef] [Green Version]
Figure 1. A schematic flow diagram of genome-assisted breeding. ① A core collection of S. tuberosum germplasm exhibits the trait of interest and wide genetic diversity. ②/③ After genetic resource selection, a trait-based phenotyping assessment is performed, followed by genotyping of selected populations. ④ Genotype-phenotype linkage is detected through various techniques. ⑤ The breeding program results in the development of improved potato varieties.
Figure 1. A schematic flow diagram of genome-assisted breeding. ① A core collection of S. tuberosum germplasm exhibits the trait of interest and wide genetic diversity. ②/③ After genetic resource selection, a trait-based phenotyping assessment is performed, followed by genotyping of selected populations. ④ Genotype-phenotype linkage is detected through various techniques. ⑤ The breeding program results in the development of improved potato varieties.
Ijms 23 09982 g001
Figure 2. An overview of agrobacterium-mediated gene transformation in plants. ① Ti-Plasmid isolation from parent cell (Agrobacterium tumefaciens). ② Digestion of the plasmid with specific endonucleases enzymes. ③ Ligation of foreign DNA into the plasmid. ④ Insertion of recombinant-plasmid into the bacterium and its incorporation into plant’s cell nuclear DNA after infection. ⑤ Growth of transgenic plant tissues artificially. ⑥ Development of transgenic potato grown in a field.
Figure 2. An overview of agrobacterium-mediated gene transformation in plants. ① Ti-Plasmid isolation from parent cell (Agrobacterium tumefaciens). ② Digestion of the plasmid with specific endonucleases enzymes. ③ Ligation of foreign DNA into the plasmid. ④ Insertion of recombinant-plasmid into the bacterium and its incorporation into plant’s cell nuclear DNA after infection. ⑤ Growth of transgenic plant tissues artificially. ⑥ Development of transgenic potato grown in a field.
Ijms 23 09982 g002
Figure 3. Overview of CRISPR-Cas9 mediated gene editing. ① A complex of CRISPR-Cas9 genetic scissors and artificially constructed single guide RNA (sgRNA) scans DNA and traces code where a cut has to be made. ② Formation of non-homologous end-joining (NHEJ) and homology direct repair (HDR) strands after DNA double-strand break (DSB). ③ Ligation of the DNA DSB by nucleotides addition on the right and deletion on the left result in gene disruption. ④ Repairing the DSB in HDR by employing an externally provided homologous DNA template for copying. The donor template’s DNA sequence is duplicated at the targeted site, which results in a guided repair.
Figure 3. Overview of CRISPR-Cas9 mediated gene editing. ① A complex of CRISPR-Cas9 genetic scissors and artificially constructed single guide RNA (sgRNA) scans DNA and traces code where a cut has to be made. ② Formation of non-homologous end-joining (NHEJ) and homology direct repair (HDR) strands after DNA double-strand break (DSB). ③ Ligation of the DNA DSB by nucleotides addition on the right and deletion on the left result in gene disruption. ④ Repairing the DSB in HDR by employing an externally provided homologous DNA template for copying. The donor template’s DNA sequence is duplicated at the targeted site, which results in a guided repair.
Ijms 23 09982 g003
Table 1. QTL for potato tuber quality and quantity traits.
Table 1. QTL for potato tuber quality and quantity traits.
TraitQTLChromosome NumberReferences
Starch granule sizeSGS02-8 and SGS03-8VIII[16]
Starch contentspPt-535988–pPt-538127, toPt-440651I[17]
pPt-539763 II
toPt-437014–pPt-538033III
toPt-438845VIII
pPt-533878X
pPt-471789XI
pPt-656237XII
STM1049-1, STWIN12G, STM1049-3 I [18]
EAAT_MCGA_381III
EACG_MCAA_191 DS, STM1002-1, STM1002-2IV
EACG_MCAT_925, StI022-3, StI022-5VIII
EATC_MCCG_182X
EACG_MCAA_191 DS, StI017-2DSXI
EACC_MCGA_114, EACG_MCAA_119 DSXII
PCT_MACT_86, StI022-2 DS, EACG_MCAA_119 DSUnlinked
Pain1-A 718 (C 552) 2, Pain1- A 1544 and Pain1-T 741, Pain1-8c III[19]
InvGE-A 85 (A 86), InvGE-G 95 (G 106)IX
InvCD141_T 543 (A 280, T 288, T 339, A 630, C 1030, G 1031, T 1096), InvCD141-G 765X
Stp23-8bIII[20]
CISREC_B_05-1V [21]
CIS_E_07-1VII
Tuber shapeSolcap_snp_c2_34875IV[22]
Solcap_snp_c2_25485, Solcap_snp_c2_25510X
Solcap_snp_c1_1847I[23]
Solcap_snp_c2_54790IV
Solcap_snp_c2_26012, VII
Solcap_snp_c1_15594, Solcap_snp_c1_11535X
Tuber skin colorSolcap_snp_c2_31852, Solcap_snp_c2_25759, Solcap_snp_c2_21178I[22]
solcap_snp_c1_12440 V
solcap_snp_c2_4342, solcap_snp_c2_45215 VII
solcap_snp_c2_50702, solcap_snp_c2_53902, solcap_snp_c2_15803VIII
solcap_snp_c2_22697X
solcap_snp_c2_39889 XI
and solcap_snp_c2_5385XII
Table 2. Comparison of three different kinds of transgenics to improve potato tubers’ quality and quantity.
Table 2. Comparison of three different kinds of transgenics to improve potato tubers’ quality and quantity.
TransgenicsTrait Introduced/ModifiedGene Added/SilencedReferences
ConventionalStarch yieldPsGPT[63]
Tuber yieldAtPAP2[64]
SPS[65]
tms1[66]
Amino acid (methionine)AmA1[67]
Amino acid (cysteine)cysE[68]
AstaxanthinOr[69]
KetocarotenoidscrtO[70]
CarotenoidscrtZ,[71]
bkt1[72]
StLCYb[73]
Ascorbic acidDHAR[74]
Vitamin-EAt-HPPD & At-HPT[75]
Vitamin B6PDXII[76]
CISSbRFP1[77]
NtInvInh2, StInvInh2A and StInvInh2B[78,79]
LbPFK[80]
RNAiStarch qualitySSIII[81]
GBSS[82]
CISVInv, [83,84,85]
SPP[86]
TALENSStarch qualityGBSS[87]
CISStvacINV2[88]
VInv[89]
CRISPRStarch qualityGBSS[90]
Anti-browningStPPO2[91]
Reducing steroidal alkaloidsSt16DOX[92]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Ahmad, D.; Zhang, Z.; Rasheed, H.; Xu, X.; Bao, J. Recent Advances in Molecular Improvement for Potato Tuber Traits. Int. J. Mol. Sci. 2022, 23, 9982. https://doi.org/10.3390/ijms23179982

AMA Style

Ahmad D, Zhang Z, Rasheed H, Xu X, Bao J. Recent Advances in Molecular Improvement for Potato Tuber Traits. International Journal of Molecular Sciences. 2022; 23(17):9982. https://doi.org/10.3390/ijms23179982

Chicago/Turabian Style

Ahmad, Daraz, Zhongwei Zhang, Haroon Rasheed, Xiaoyong Xu, and Jinsong Bao. 2022. "Recent Advances in Molecular Improvement for Potato Tuber Traits" International Journal of Molecular Sciences 23, no. 17: 9982. https://doi.org/10.3390/ijms23179982

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