Next Article in Journal
Ditching Phosphatic Fertilizers for Phosphate-Solubilizing Biofertilizers: A Step towards Sustainable Agriculture and Environmental Health
Next Article in Special Issue
Development of Halotolerant Microbial Consortia for Salt Stress Mitigation and Sustainable Tomato Production in Sodic Soils: An Enzyme Mechanism Approach
Previous Article in Journal
Impacts of Government Policies on the Adoption of Biomass Power: A System Dynamic Perspective
Previous Article in Special Issue
Biostimulants as Innovative Tools to Boost Date Palm (Phoenix dactylifera L.) Performance under Drought, Salinity, and Heavy Metal(Oid)s’ Stresses: A Concise Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Assessment of Benefits and Risk of Genetically Modified Plants and Products: Current Controversies and Perspective

1
Department of Crop Science, College of Sanghuh Life Science, Konkuk University, Seoul 05029, Republic of Korea
2
Bioherb Research Institute, Kangwon National University, Chuncheon 24341, Republic of Korea
*
Author to whom correspondence should be addressed.
Sustainability 2023, 15(2), 1722; https://doi.org/10.3390/su15021722
Submission received: 10 November 2022 / Revised: 12 January 2023 / Accepted: 12 January 2023 / Published: 16 January 2023
(This article belongs to the Special Issue Adaptive Response and Mechanism of Crops to Abiotic Stresses)

Abstract

:
Genetic transformation has emerged as an important tool for the genetic improvement of valuable plants by incorporating new genes with desirable traits. These strategies are useful especially in crops to increase yields, disease resistance, tolerance to environmental stress (cold, heat, drought, salinity, herbicides, and insects) and increase biomass and medicinal values of plants. The production of healthy plants with more desirable products and yields can contribute to sustainable development goals. The introduction of genetically modified food into the market has raised potential risks. A proper assessment of their impact on the environment and biosafety is an important step before their commercialization. In this paper, we summarize and discuss the risks and benefits of genetically modified plants and products, human health hazards by genetically transformed plants, environmental effects, Biosafety regulations of GMO foods and products, and improvement of medicinal values of plants by the genetic transformation process. The mechanisms of action of those products, their sources, and their applications to the healthcare challenges are presented. The present studies pointed out the existence of several controversies in the use of GMOs, mainly related to the human health, nutritions, environmental issues. Willingness to accept genetically modified (GM) products and the adoption of biosafety regulations varies from country to country. Knowledge about the gene engineering technology, debate between the government agencies, scientist, environmentalist and related NGOs on the GM products are the major factors for low adoptions of biosafety regulation. Therefore, the genetic transformation will help in the advancement of plant species in the future; however, more research and detailed studies are required.

1. Introduction

Present agriculture practices alone cannot solve food security, and eradicate malnutrition and hunger that exist globally [1]. Recent research reported that approximately 17.2% of the global population is lacked to the access of nutritious and sufficient food [2]. According to a survey, the present global rate of increase in crop yield is less than 1.7% and currently, the rate of increase in agricultural yield needs to be 2.4% to meet the world’s demands for grains and to improve the nutritional quality [3]. FAO predicted the loss of arable land available for crop production from the current 0.242 ha to 0.18 ha by 2050 [4]. Conventional breeding creates a new population by intercrossing several lines with another parental line, in hopes of expressing one or more desired traits [5]. The conventional breeding process process have certain limitations such as sexual incompatibility, gene linkage, and the time involved in obtaining cultivars [6].
Genetically modified organisms (GMOs) are usually referred to as living organisms whose genetic makeup has been artificially manipulated by inserting new genes through the process called the technology of recombinant DNA, or genetic engineering giving the plants new characteristics [7]. Genetically modified plants and products that emerged along with advanced biotechnology can contribute to the increase in agricultural production, and improved nutritional values that could relieve global food shortages [8]. Moreover, genetic transformation methods have generated the possibility of producing plants with desired traits in elite cultivars in considerably shorter time-frames with reduced gene linkage problems [9]. This technique not only provides desirable traits but also improves nutritional levels, transforming them into rich and healthy food items in both dicot and monocot plants [10]. Such technology is widely applicable in transferring genes from any organism to a great variety of plant species, from wild to cultivated species, thus preventing the natural barriers between species and reducing the time to obtain new varieties [11]. The use of these techniques in agriculture has also enabled the discovery of processes that involve the use of DNA techniques, which enable their propagation via cell and tissue culture in vitro, and in the production of transgenic plants to develop drug leads, and perform biosynthesis of functional compounds, enzymes, and hormones, blood substitutes, vaccines and antibodies [12], for the production of medicines, recombinants, and industrial products [13].
Agricultural products produced by genetic manipulation of crops such as soy, cotton, tomato, potato, canola, and corn, among others, have already been approved for marketing [14]. GMOs have great potential to solve the poverty of the global population, improve the nutritional value of crops, reduce environmental pollution, enhance medicinal values, and contribute to the sustainability of agriculture [15]. Despite the advantages of GMOs, there are widespread concerns about the biosafety of the products, causing the great concern regarding human health and environmental integrity, and political and regulatory issues [16]. The use of transgenics generates controversies related to possible risks to human health, such as; food allergies, antibiotic resistance, increase in toxic substances, more pesticides in food consumed, and also the lack of information on packaging labels [17]. Several arguments and intense debates have emerged from different forums analyzing potential benefits and possible risks associated with the cultivation and consumption of GMOs in terms of ethical, environmental, health, biodiversity, and religious issues [16]. It is, therefore, important to conduct the risk assessment of genetically modified plants and their products by making common regulatory methods before their release into nature and applications [18]. However, restrictions on transgenic plants exist and vary from country to country.
This study aims to analyze the possible harm and benefits of transgenic food according to the scientists working in the field, making people know what they are consuming. Moreover, the impact of transgenic plants on human health, the environment, and agriculture have been analyzed critically. This study will also take a look at the Gm biosafety and regulatory framework for GM foods in different countries. We will also take a look at the risks and controversies of GMOs

2. Literature Search Method

The literature presented in the study covers different social science field related to the GMOs. We conducted a mini survey of literature of GM plant and its products published in the journals (research articles and reviews paper) from 2000 to 2022. The search engines used in the collection of published papers (accessed on 1 October 2022) were Google scholar (http://www.google.co.kr), PubMed. The data base such as Scopus (https://ww.scopus.com, accessed on 1 October 2022) were used selection of identification of publications. All the downloaded papers were peer reviewed, English language, and related to GM products. Relevant published papers were searched in the Google scholar using the list of keywords (search terms). The search terms were organized in the following different groups: genetically modified organisms (GMOs), risk and advantages of GMOs, GMOs and biofortifications, phytoremediation, allergens, phytochemicals, GMOs and environmental and human health issues, and biosafety regulations, GMOs and controversies. The collected data were analyzed and illustrated to obtain the results based on the objectives of the present study.

3. Plant Genetic Transformation Methods

Hundreds of plants species have been successfully transformed by various genetic transformations and for numerous useful traits; however, these techniques have inherent problems and limitations, including the lack of an efficient plant regeneration system, low frequency of transformation, genotype specificity, low availability of genes of interest and biosafety, and time and labor-intensiveness [19]. The successful regeneration of transgenic plants requires two major factors: an efficient, rapid, reproducible regeneration system and an effective method for the integration of genes into the DNA of plant cells [20]. Foreign genes can be introduced into plant genomes by various methods, including biolistics, sonication, liposomes, viral vectors, transfer mediated by Agrobacterium, chemicals, silicon carbide fibers, floral dip method, microinjection, and microlaser treatment depending on the species to be transformed and types of explants used [21]. Among these, transformation mediated by Agrobacterium, electroporation, and biolistics, are the most commonly used methods for producing commercially released transgenic plants. Despite the limitations of transgenic plants, there has been a continuous increase in the production of such plants to improve the nutritional and medicinal value of crops.

3.1. Agrobacterium-Mediated Transformation of the Plant

Plasmids are the most commonly used vectors in the genetic transformation of plants (Figure 1). These vectors have an artificial T-DNA, into which different transgenes can be inserted and transferred to host plants [22]. Agrobacterium tumefaciens and A. rhizogenes have plasmid types Ti and Ri, respectively, both of which can be used for genetic transformation. The advantage of Agrobacterium-mediated transformation is that it possesses the natural ability to transfer and integrate transgenes into the host cell, transfer large segments of DNA with only minimal rearrangement, and possess the higher rates of genetic transformation efficiency, low copy number integration, and enable the transmission of integrated genes into progeny in a Mendelian manner [23]. This technique is applicable in both monocot and dicot plants, algae and fungi, human cells, and sea urchin embryos [24,25]. The limiting factor of this technique is the ability to regenerate the transformed tissues and the low transformant ratio; in addition, the size and complexity of the Ti and Tr plasmids also influence the rate of transformation [26].

3.2. Biolistics Method of Genetic Transformation of the Plant

This method is also called a gene gun, particle acceleration, or microparticle bombardment for the growth of transformants [27]. This method is useful for both dicot and monocot plants, consisting of bombarding cells or tissues with 0.5 mm gold or tungsten microparticles carrying exogenous DNA-coated projectiles using compressed helium incubated at 30 °C in a special chamber under vacuum conditions (Figure 2). The mechanism involves direct penetration of the cell wall and plasma membrane for direct DNA transfer [28]. Different systems are employed to accelerate the particles, including chemical explosion, higher pressure helium, electrical discharges, and vaporization of water drops [29]. This method readily transfers genes into intact plant tissues, including leaves, petals, and pollen endosperm, and has been successfully used to generate transgenic maize, soybeans, oats, rice, wheat, and barley [30]. The advantages of this method are that it consumes less time, allows the cells and tissues to undergo direct gene transformation, and can be applied to diverse groups of plant species with the higher stability of transformants [28].

3.3. Electroporation Method of Genetic Transformation of the Plant

The electroporation method was initially developed for the transformation of cereal genes and was later applied to other plant species. This method utilizes a high-voltage electric field to generate holes in the plasma membrane (Figure 3). The electrostatic forces formed in the process cause compression, which leads to the formation of holes in the membrane to integrate the transgene to be taken up by the cell [31]. The successful regeneration of transgenic plants using electroporation methods depends on various factors, including the diameter and source of host cells, electroporation medium (pH), electrical conductivity, membrane composition, size and shape of introduced DNA, and intensity and duration of electrical pulses used in the process [31]. The limitation of this method is the production of efficient protoplasm regeneration protocols and high cell mortality [32].

4. Benefits of Genetically Modified Plants and Products

4.1. Biofortification

Micronutrient deficiencies are posing a serious threat to the health of one–half of the global population [33]. Nutritionally enhanced food crops using modern biotechnology, conventional selective breeding, and agronomic practices to enhance nutritional values are considered an effective and alternative approach for mitigating in economically poor countries [34]. The production of foods using biotechnology offers both benefits and threats. The production of transgenic plants is not only helpful in developing new varieties with increased nutrition but also increased resistance against biotic and abiotic factors, thereby enhancing the quality and yield of plants [35]. In addition, plant production enables the production of materials of industrial interest, such as biodegradable plastics, vaccines (transgenic bananas that produce vaccines against hepatitis B, transgenic potatoes that are resistant to viruses, rice with increased iron and vitamin levels, with increased resistance to extreme weather, and drought, [12,36,37].
GMO consumption maintains a healthy balance by fortifying nutritional quantity in foodstuffs that may not normally occur in them. For example, the production of “golden rice” with elevated vitamin A levels, the development of herbicide- and insecticide-resistant crops, thereby reducing crop losses, and other therapeutic substances of specific interest [38]. Moreover, research reports have indicated that proteins produced by GMOs are non-toxic, easily digestible, and cause no allergies [39]. Genetically modified fish grow larger, and pigs are grown with less body fat [40]. Other studies have reported increased beneficial nutritional profiles, such as increased levels of antioxidant compounds in GMOs that may provide health benefits to humans [41], and provide useful medicines, such as insulin for treating diabetes, from genetically engineered bacteria [42].

4.2. Transgenic Approaches for Improving Phytochemicals and Biological Activities in Plants

Several authors have reported an improvement in the production of antioxidants, such as phenolic compounds, from transgenic plants transformed with the bacteria Agrobacterium tumefaciens and A. rhizogenes (Table 1). Increased concentrations of phenolic compounds have also been reported to improve antimicrobial activities in Cucumis melo [43]. Furthermore, scientists have produced transgenic lines by overexpressing genes in Lycopersicon esculentum Mill. cv. Per with increased phenolic compound content in plants that are involved in phytoremediation [44]. Moreover, an increase in metabolites such as triterpene and steroidal saponins, and phenolics [45], was reported in hairy root cultures of Trigonella foenum-graecum L., an elevated amount of phenolics acid, and flavonoids [46] was reported in Spagneticola calendulacea (L.) Pruski to increase food value. Increased resistance to Botrytis cinerea in transgenic Morus notabilis C.K. Schneid [47].
Genetic engineering has been successful in producing transgenic rice that contains 23 times higher concentrations of carotenoids than in previous transgenic golden rice [48]. Similarly, the genetic transformation of phytase in the transgenic soybean resulted in enhanced phytase activity by 2.5 fold compared to non-trangenic soybean [49]. Moreover, methyltransferase genes (VTE3 and VTE4) from Arabidopsis thaliana transformed into the soybean genome resulted in an enhanced α-tocopherol content by 95% more than in non-trangenic plants [50]. The transformation of lactoferrin in dehusked rice successfully enhanced the iron contents by 120% [51]. In another report, expression of soybean ferritin in rice resulted in an increase in the iron contents in Indica cv IRR68144 seeds, in wheat by 1.5–1.9 fold [52], lactoferrin genes enhanced the Fe content in Maize [53], potato, lettuce and tomato [54], Endogenous nicotianamine content was increased by 5–10 fold in transgenic rice over-expressed with HvNaSi [55]. Induces the proliferation of hairy roots, which increases the production of secondary metabolites. Many plant species have been transformed with A. rhizogenes for increased production of polyphenolic antioxidants such as phenolic acids and flavonoids (Table 1). Transformed plants of Codonopsis lanceolata and Perilla frutescens transformed with γ-tmt genes present higher concentrations of tocopherol and phenolic compounds, thereby enhancing the antioxidant properties of such plants [25,56]. Another approach to the recombinant production of foodstuffs is the genetic transformation of useful genes that enhance the production of beneficial compounds in plants and improve human health. Recently, researchers introduced genes into Lycopersicon esculentum Mill. cv Ailsa Craig, to increase the accumulation of antioxidants, such as phenolic compounds [57]. Similarly, increased amounts of phenolic compounds and resveratrol have been reported in transgenic Rehmannia glutinosa transformed by A. tumefaciens [58].
Table 1. Genetic transformation strategies and genes used for the biofortification in crops.
Table 1. Genetic transformation strategies and genes used for the biofortification in crops.
Scientific NameAgrobacterium Strains/VectorGenePhytochemicalsBiological ActivityReferences
Codonopsis lanceolataLBA4404/pYBI121,γ-tmtPhenolic compounds and tocopherolAntioxidant and antimicrobial activityGhimire et al. [25]
Perilla frutescensLBA4404/pYBI130γ-tmtPhenolic compounds and tocopherolAntioxidant and antimicrobial activityGhimire et al. [57]
Lycopersicon esculentum L.pBI101stilbene synthase (StSy)ResveratrolAntioxidant activityD’Introno et al. [59]
Cucumis meloMAFF 03–01724 (pRi1724)rolC geneAroma essential oils (Z)-3-hexenol, (E)-2-hexenal, 1-nonanol, and (Z)-6-nonenolAntimicrobial activityMatsuda et al. [43]
WheatpMDC32Nicotianamine synthase 2 (OsNAS2)Higher concentration of grain iron and zinc Beasley et al. [60]
CassavaLBA4404/p8023FER1 and IRT1Higher concentration of iron and zinc Narayanan et al. [61]
Rice pMDC3235S-OsGGPIncrease concentrations of ascorbate Broad et al. [62]
Soybean EHA105/pATPS1Overexpression of adenosine 5′-phosphosulfate sulfurylase 1Higher aamounts of sulfate, cysteine, and secondary metabolites in seeds Kim et al. [63]
Gynostemma pentaphyllumATCC 15834TL-DNA rolBTriterpene saponinsAntitumor, immunopotentiating, antioxidant, antidiabeticChang et al. [64]
Momordica charantiaATCC 15834rolC geneCharantinAntioxidant, antibacterial, antifungalThiruvengadam et al. [65]
Momordica dioicaKCTC 2703rolC genePhenolic compoundsAntioxidant, antibacterial. Thiruvengadam et al. [66]
Cucumis anguriaKCTC 2703rolC genePhenolic compoundsantioxidant, antibacterial Yoon et al. [67]
Lycopersicon esculentum Mill.pBBC200/pBBC3LC and C1.FlavonoidsAntioxidant activityLe Gall et al. [68]
Rehmannia glutinosaLBA4404/pMG-AhRS3Resveratrol Synthase Gene (RS3).Phenolic compounds and ResveratrolAntioxidant activityLim et al. [58]
Ipomoea batatas [L.] Lam.pCAMBIA1300IbCAD1lignin contents, monolignol levels, and syringyl (S)/guaiacyl (G)Stress toleranceLee et al. [69]
Miscanthus sinensisLBA4404/pMBP1antisense COMT gene.Lignin contentLignin biosynthesisYoo et al. [70]
Cucumis meloMAFF 03-01724rolCgeneVolatile compoundsAntimicrobial activityMatsuda et al. [43]
Trigonella foenum-graecum L.ARqua1 and LBA9402, nary vectorp35S::eGFP,Green fluorescent protein gene [eGFP S65T varianttriterpene and steroidal saponins, phenolics, and galactomanaHeterologous expressionGaragounis et al. [45]
Sphagneticola calendulacea (L.) PruskiLBA1334, pCAM:2 × 35S:grolA,rolB, rolC and gusAPhenolics acid and flavonoidsAnti-hepatotoxic activityKundua et al. [46]
Morus notabilisGV3101/pLGNLMnMET1Flavonoid contentInhibitory effect on Botrytis cinereaXin et al. [47]
Arabidopsis thaliana (L.)pCAMBIA1301-AtMyB12 AtMYB12Phenolic compoundsIncrease in the flavonoid contentsWang et al. [71]
Gynostemma pentaphyllumATCC 15834TL-DNA rolBTriterpene saponins (gypenosides)Antitumor, cholesterol lowering, immunopotentiating, antioxidant, hypoglycemic, antidiabetic activityChang et al. [64]
Aspergillus nigerANIp7-laeA LaeAflaviolin, orlandin and kotan Biosynthetic model for flaviolinWang et al. [72]
Nicotiana tabacumpCAMBIA1301-LlCCRPhenolic compounds,Wood propertiesPrashant et al. [73]
Brassica rapa ssp. rapaKCTC 2703rolC and virD2Phenolic compoundsAntioxidant activity, antimicrobial activityChung et al. [74]
Hypericum perforatum L.Ri plasmidrolBPhenolic compounds, hypericin, and pseudohypericinAntioxidant activityTusevski et al. [75]
Nicotiana tabacum L.pGANE7/pBAK61AK-6bPhenolic compoundsAuxin and cytokininGalis et al. [76]
Solanum tuberosumLB4404/pBinKan-TXTyrDC2Phenolic compounds, tyrosol glucosideIncreased resistance against pathogensLandtag et al. [77]
Salvia miltiorrhiza BungeGV3101/pHB-GFPRAS and CYP98A1Phenolic compoundsAntibacterial; Antioxidant activity;Fu et al. [78]
Nicotiana tabacum L.LB4404ipt-genePhenolic compoundsPeroxidase activitySchnablová et al. [79]
Artemisia carvifolia BuchGV3101 c/pPCV002rol GenesArtemisininIncreased production of artemisininDilshad et al. [80]
Cucumis anguria L.BA9402, A4, 15834, 13333, R1200, R1000rol A and rol BPhenolic compoundsAntioxidant and antimicrobial activitySahayarayan et al. [81]
Medicago sativaLBA4404 /pUC18-PALCOMT and CCoAOMTPhenolic compoundsLignin biosynthesisGuo et al. [82]
Nannochloropsis sp.BA4404/pCAMBIA130404 gus–mgfp5Phenolic compoundsTransient GUS expression inCha et al. [83]
Linum usitatissimumC58C1:pGV2260Chalcone synthase (CHS), chalcone isomerase (CHI), and dihydroflavonol reductase (DFR)Phenolic compounds, monounsaturated fatty acids, and lignans contentAntioxidant propertiesLorenc-Kukuła et al. [84]

4.3. Transgenic Approaches for Environmental Protection

The benefits of transgenics can be assessed from an environmental point of view (Table 2). Bacillus subtilis and Bacillus thuringiensis (Bt) strains can produce toxic proteins such as Cry or d-endotoxins [85], that are toxic to various kinds of pests, insects, and pathogens [86]. Bt toxins are also being used in generating trangenic crops effectively control crops pests such as CryIAc in rapeseed to control hairy bugs, diamondback moths, and cotton bollworms [87]. Cry2Aagene in transgenic pigeon beans to control pod borers [88], Cry3A gene in transgenic spruce to control bark beetles [89]. According to a recent report, a significant change in the amount of herbicides and pesticide application was observed in the USA with the adoption of herbicides tolerant GM plants [90], such as; transgenic soybean [91], summer corn and cotton. The reduction of herbicides and pesticides can reduce the environmental impacts on cultivated land. The reduction in the application of pesticides also minimizes the use of machinery for spraying them in the field, thus reducing fossil fuel consumption in the agriculture sector.

4.4. Transgenic Approaches for Removing Allergens

Genetic transformation technology successfully incorporated genes in the plants responsible for encoding non-allergic proteins, and hypoallergenic crops, thus improving food protein equality [103]. A significant reduction in peanut allergies was reported by silencing the gene encoding Arah2 using RNAi technology [104]. Similar technology was used by Le et al. [105], to silence the allergens Lyce 1.01 and Lyce 102 in tomato profiling. Similarly, allergic proteins such as Mal d from apple [106], and GlymBd 30K from soybean [107] were silenced using RNAi technology. In other studies, the hypoallergenic approach was effective to reduce allergenic protein in Rye gram pollen [108]. All these studies indicate that engineered plants can also be expected to improve food quality by reducing allergens.

4.5. Transgenic Approaches for Phytoremediation

Phytoremediation is a sustainable solution for solving environmental contaminants caused by pollutants including heavy metals sediments, and inorganic and organic pollutants. Recently, the application of transgenic plants for the removal of heavy metals or organic pollutants has gained more interest [109] (Table 3). It is possible to transfer genes responsible for the hyperaccumulation of traits into target plants having remediation potential. The introduction of such genes has been reported in several plants including A. thaliana, [110]. Metallothioneins (MTs) confer heavy metal tolerance and accumulation in yeast. For example, the overexpression of MT genes increased the Cd tolerance in tobacco and raper seed plants [111]. Overexpression of phytachelatin synthase (TaPCSi) in Nicotiana glauca significantly increased the tolerance to heavy metals such as Cd and Pb [112]. In another study, overexpression of AtPCS, increased the phytochelatins and high resistance to arsenic [113]. Arsenate (As), mercury (Hg), and selenium (Se) are important pollutants, and transfer approaches have been employed to remove them from the soil [114]. Expression of the mer B gene in transgenic Arabidopsis thaliana resulted in more tolerance to methylmercury [115]. Similarly, overexpression of ATP sulfurylase and CGS resulted in an increased phytovolatilization in Brassica sp. [116]. Enzymes such as peroxidases, laccases, peroxygenases, nitroreductases, and phosphatases play important roles in the phytodegradation of organic pollutants [117]. These plant enzymes shown to act on organic pollutants including atrazine, chloroacetanilide, and TNT (2,4,6trinitrotoluene) [118]. An increased rate of degradation of TNT and chloroacetanilide has been reported previously in poplar plants [119]. Other best example of phytoremediation includes the overexpression of ECS and GS genes in B. juncea resulted in increased tolerance to atrazine [120].

4.6. Transgenic Approaches for Vaccine Production

The expression of antigens using biotechnology in plants has opened up a new field for the production of plant-based vaccines. Advances in transgenic research have made use of plants to serve as a bioreactor for the production of certain vaccines for curing diseases [135]. Several plant-based vaccine antigens have been successfully expressed in plant tissues as a result of a stable expression or transient expression of genes [136] (Table 4). Plant-based vaccines are cost-effective, easy to carry, have less chance of contamination and degradation, require no medical professionals, high-tech machines, or preservation, and are less costlier than cell culture bioreactors [137]. By conceiving the idea of an edible vaccine, the antigens genes encoding Rabies Capsid proteins such as HBsAG, and HIVgag have been successfully expressed in transgenic tomatoes [138]. Exciting progress in achieving a high level of protein expression was achieved in transgenic carrots by Daniell et al. [139]. Later, Scotti and their research [140] team obtained chloroplast-based production of pharmaceuticals, vaccines, and antibodies. Transgenic N. benthamiana plants were successfully expressed with D antigen (PV3) to use a vaccine against polio diseases Marsian, et al. [141].

4.7. Transgenic Approach for Increased Biofuel Capacity in Plants

Lignocellulosic biomass from non-food crops has been considered a potential source of biofuel. Lignin, a major component of plant cell walls is considered a hindrance to cellulosic biofuel production. The application of biotechnology for biofuel production is gaining more interest, especially from the lignocellulosic biomass [154]. Recently, several studies have reported the successful cloning of genes responsible for increased biomass and sugar accumulation and higher production of biofuels in transgenic lines [155]. Several studies have reported the expression of genes in plants that are responsible for the degradation of the plant cell wall for more efficient biofuel production [156]. A low amount of lignin was reported by downregulating the lignin biosynthetic gene 4-hydroxycinnamoyl CoA ligase (4CL) [157]. In another study, the amount of lignin synthesized decreased to facilitate higher biofuel production in transgenic Miscanthus sinensis [70]. Overexpression of expansin genes which helps in loosening of cell walls [158] and successfully generated a transgenic plant with a suppressed debranching enzyme that produces soluble phytoglycogen. Vanden Wymelenberg et al. [156] reported the involvement of several genes in the breakdown of lignin from the Phanerochaete chrysosporium genome. Moreover, several other studies reported an alteration of lignin biosynthesis in the plant without affecting the vascular structure of plants [55]. They reported downregulating 4-hydroxy cinnamoyl CoA ligase (4cl) responsible for the reduction of the lignin composition and an increase in the biomass of plants. Ralph et al. [159] reported a drastic decrease in the lignin content and structure by decreasing the expression of 4-coumarate 3-hydroxylase (C3H) in alfalfa. A similar result was also observed by Chabannes et al. [160] in transgenic tobacco by deducting the expression of cinnamoyl CoA reductase (ccR). Furthermore, the suitability of biofuel production in transgenic lines of tobacco has been investigated by downregulating O-methyl-transferase (OMT) enzyme by Blaschke et al. [161]. They observed an increase in biomass and reduction in the lignin contents in the transgenic lines of tobacco. Other emphasizes the improvement of the fatty acid composition of plants to enhance biofuel production. Moreover, as compared to the WT plants, the transgenic line showed an increase in biofuel production in soybean by expressing diacylglycerol acyltransferase 2A (DGAT2A) from Umbelopsis sps fungus [162]. Furthermore, an increased caprylic acid and capric acid was observed in transgenic rapeseed by over-expressing a laurate-specific LPAAT gene from coconut [163]. Another approach for increasing biofuel is to increase the biomass production of plants by genetic transformation approach. Manipulation of ADP glucose pyrophosphorylase resulted in an increased starch content and biofuel yield [164]. They observed an increase in photosynthesis and biomass by overexpressing two enzymes from Cyanobacteria in the tobacco plant. Jing et al. [165] reported an increase in the plant height and biomass by expressing the glutamine synthase gene (GSi).

4.8. Increased Stress Resistance Capacity in Plants

The excessive use of herbicides and pesticides is causing serious hazards on croplands, which makes cultivating land unsuitable for farming in the future. Recently, the introduction of GMOs has not required the use of these products. Some genetically modified crops are highly tolerant to one herbicide, instead of the multiple types of herbicides used in the field to prevent environmental damage. For example, genetically modified Roundup Ready corn is not only a glyphosate-tolerant GM corn but also is as safe and nutritious as conventional corn grain [166]. Bt rice KND1 expressing Cry1Ab protein show high levels of resistance to insects and possess no toxic effects on human health [167]. Similarly, insect-resistant crops include wheat, potatoes, rice, and sugarcane [168]. Researchers have increased the level of lignin content, monolignol levels, and syringyl (S)/guaiacyl (G) in transgenic Ipomoea batatas [L.] Lam., cv. Xushu 29 to enhance stress tolerance [69]. The introduction of Bt corn effectively controls the application of chemical pesticides, thereby controlling the environmental pollution caused by pesticides and reducing the cost of growing crops in the field [169]. Plants that can tolerate high salinity and long periods of drought have been reported [170], which can help people to grow crops in cold and less irrigated areas.

5. Disadvantages of Genetically Modified Plants and Products

The introduction of genetically modified food in the market has raised some serious questions regarding human health, environmental economics, and legal issues. For instance, it has been reported that the transfer of genes poses serious genetic hazards and is associated with possible food toxicity [41]. Once GMOs are produced and released into the environment, they can be difficult to control [171] and any harmful products produced by these organisms will remain metabolically active as long as they survive and multiply [171].

5.1. Human Health Hazards

Despite the advantages of GMOs, there is increasing concern about food safety and health risks. The transgene may cause undesirable developmental and physiological effects on mammals, including humans. There is a likelihood that the transformed gene may produce toxic protein or allergens or causes allergenic reaction in the human body. Moreover, other potential concerns are incomplete digestion of GMO foodstuffs in the gastrointestinal tract, which could result in the horizontal transfer of genes to the microflora and somatic cells of the intestine [172]. Others have emphasized that the transfer of genes could cause infertility in animals, and result in allergic reactions [173].

5.2. Environmental Risks

The release of such products and their possible impacts on the environment regenerate high monitoring of environmental biosecurity to reduce or complete eradication of risk induced by them. Apart from direct effects on human health, GM plants have environmental effects on non-target organisms such as fish, worms, bees, and insects, biodiversity loss, and gene instability [174]. In other studies, Bt toxin produced by transgenic cotton killed many species of insect larvae, causing an imbalance in the ecosystem and food chain [175]. It has been argued that GM crops have a serious impact on farmers and their indigenous products because they compete with GMO products [176]. However, several previous studies reported the no-targeted impacts of novel genes transformed into the plant genome. For example, Bt maize showed potential hazards and toxic to monarch butterfly larvae that feed on milkweed leaves contaminated with pollens from Bt strains and caused delayed development, and increased maturity reported in Ostrinia nubilalis and Spodoptera littorals ingested with corn leaves expressing Bt CryIAS toxins [177].

5.3. Gene Flow

The most serious problem associated with gene flow is the loss of biodiversity and often cited as potential risk. Chances of accidental cross pollination between GM crops with its wild relatives are very high, making them super-weeds that resist diverse herbicides and become difficult to control. There are several examples where gene flow from crops to the relatives weeds such as in Beta vulgaris [178], in Avena strigose [179], in Brassica napus [180].

5.4. Increased Antibiotic Resistance

GM products enter the human body through food, vaccines, bacteria, or viruses. There is concern that the GM plants with bacterial resistance genes in their genome and might act as the source of drug resistance genes to the bacteria of clinical importance. Moreover, the possibility of developing antibiotic-resistant bacteria has been reported because of the frequent use of antibiotics in the genetic transformation process [181]. Most GM products contain marker genes and genes for certain useful traits. These marker genes can build resistance to particular antibiotics, and constant consumption of these foods could result in antibiotic resistance in the human body [182].

5.5. GMO Products Can Trigger Immune Reactions and Allergies

The introduction of new genes into plants can cause allergies by producing unexpected products (proteins and metabolites) in the plants [183]. For instance, the immune systems of rats respond more slowly to genetically modified potatoes than to normal plants [184]. In other studies, Bt bacteria can effectively control insects that attack crops. However, there is an equal chance of consuming Bt toxins and reacting to the mammals causing allergies [185]. Insects, birds, and other animals that feed on certain crops may not consume genetically modified crops due to allergic reactions or poisonous products. As a result, a great number of fauna can face starvation, affecting entire food chains and causing serious threats to ecosystems [186].

6. Biosafety Regulatory of GMO Foods and Products

Considering the importance of GMOs, several countries have managed to develop biosafety regulatory systems for the safety of GM foods and products. The regulations surrounding GMOs are complex and the rate of consumer acceptance is crucial, which results in reduced usage of GMOs. GMOs and their products have been facing severe controversies and hurdles from the public sector, NGOs, and environmental organizations [187]. Different governments have different approaches to tackling the products of GMOs, which vary widely, and are country-specific [188]. Within the European Union (EU), Directive 2001/18/EL contains the biosafety regulation for the use of GMOs. It defines and control environmental release (case by case) evaluation of the environmental risk of GMOs [189]. Other directives such as 98/81/CE for the number of GM microorganisms, directive 1946/2003 for transboundary movement of GMOs, 1829/2003 for GM food and feed have been authorized [189]. GMO products have already been supplied to the EU market with appropriate labelling and identification methods under the title NOVWL-FOOD classification in May 1997 [190]. Currently, European Union-based legislation accepted the products of natural gene transfer methods, such as conjugation, auto-cloning, and gene transduction, and are considered non-genetically modified organisms [191]. However, EU has banned the application of clustered regularly interspaced short palindromic repeats genome (CRISPR-Cas9) editing technology, but the US has allowed the use of Cas9, which enables geneticists and medical researchers to edit parts of the genome [192]. Similarly, The Canadian Food Inspection Agency (CFIA) is responsible for regulating GM plants, a field trial of GM crops, their approval and commercial release in Canada. It also plays a major role in assessing impacts on biodiversity and environment, possible gene flow and impacts on non-targeted organisms [193]. In India, safety guidelines for GMOs such as research, field trails of GM foods and products assessment environmental risk assessment have been adopted from Rules 1989 [193]. Ministry of Environment Forest, Forest and Climate Change (MoEFCC) in association with the department of Biotechnology (DBT) recently adopted new guidelines for the environmental risk assessment of GE plants in India [194]. So far, Bt cotton (insect-resistant transgenic cotton) is the only GM plant to have been approved for commercial cultivation in India. Over 20 different GM plants with insect resistance, abiotic resistance, herbicidal resistance, enhance nutritional traits etc. have been under field trials [195].
The adoption of biosafety regulations is strongly impacted by the economical and political situation of countries. Despite their differences in approach and adoption of GMOs regulations framework, countries such as Brazil, Argentina, Chile, Mexico, Honduras, Costa Rica, and Uruguay were the first Latina America to approve GM crops [196,197]. Other Latin American nations such as Peru, Venezuela, and Ecuador implemented a complete ban on the application/test and import of GMOs [198,199]. To harmonize the regulations concerning GM products, Latin American countries such as Brazil, Argentina, Paraguay, Uruguay and Chile singed a declaration which legalizes the application of gene-edited products (case by case) amid strict regulation [200]. Countries such as Brazil and Argentina are major exporters of GM crops (cotton, soybean and Maize) and recently adopted legal provisions to allow the cultivation of GM crops [200], which not only play a bigger role in their economy but also play a key role to rapid adaption of biosafety law and regulations [200,201,202,203]. The Secretariat of Agriculture, livestock, fisheries and Food (SAGyO) is responsible for the regulation of GMOs, for conducting field tests, release and commercial application in Argentina [204]. While, national technological Biosafety committee (CTNBio), is responsible for scientific research on GMOs, field tests, risk assessment and assessing the safety of GMOs in Brazil [204]. Legal provisions of biosafety regulations are under discussion in the countries such as El Salvador, Mexico, Peru, Costa Rica, The Dominican Republic, and Ecuador. Other Latin American countries including Barbados, Dominica, Guyana, Haiti, The Bahamas, and Belize has no legal provision to deal with GMOs so far [205].
African nations can benefit from the adoption of the biosafety regulation to mitigate the food crisis, nutrition and economic livelihood [206,207,208]. The rapid adoption of GM crops regulations can address the existing food crises and ease hunger that exists in African countries. Some African countries welcomed GM technology and rapidly proceed for adopting GM crops to enhance agricultural production efficiency and increase the nutritional values of plants [209,210]. While, other African countries oppose GM technology stating its safety concerns, environmental and human health issues, intellectual property rights and ethical uncertainties [211,212,213]. However, several anti-GMO debates and controversies related to the safety of GMOs, and their impacts on human health and environmental issues are major hindrances in adopting biosafety regulations among African nations [214,215]. Despite hindrances, the majority of African nations (47 countries) currently allow the cultivation of GMO crops [216]. South Africa is the first African nation to enact the regulatory framework to allow the cultivation, export and import of GM crops [216], and other African countries are interested in collaborating and harmonising the regulation concerning GM crops (African Biosafety network of expertise ABNE, 2019 [217]. Successful confined field trials have been conducted for maize, sorghum, cassava and Bt cotton with a wide range of traits in Kanya [217,218]. It has been reported that early acceptance of biosafety regulation has been hindered by inadequate GM technology knowledge in Kenya, and less awareness and knowledge of GM technology in the countries like Ghana and Nigeria, [219]. Moreover, a slow and delayed GM adoption rate in Tanzania have been reported [220]. The restrictive regulations, lack of information and awareness of the GM crops regulations have played an important role to obstruct the commercialization of GM crops in African nations [221,222]. In addition, opposition to biosafety bills, laws and regulations from NGOs, media, political parties social and economic factors and multinational companies have further helped to restrict the adoption of GM crops regulations in these countries [223,224,225,226].
Similarly, China adopts strict safety evaluation of GM plants and products and promulgated a whole set of biosafety laws, regulations and management systems considering its national situation and international norms and regulations. For the implementation of biosafety regulation, the Ministry of Agriculture (MOA) played a pioneering role in the implementation of regulations, and administrative Measures for the Safety Assessment of Agricultural GMOs [227], and developed the guidelines for safety inspection of field trials, research, processing, import and exports of GM crops [228]. Recently, MOA has promulgated a set of new regulations to shorten the process involved commercialization of GM crops [229] and introduced biosafety guidelines to regulate gene-edited crops [230]. Similarly, Korea has released a set of laws and regulations guidelines for GMOs and GMO products. To ensure biosafety, proper assessment of GMOs is carried out according to the guidelines of the Korea Food and Drug Administration (KFDA) [231]. It is clear from the above data that there exists a diverse range of regulations and frameworks supporting the research and commercialization of GM crops. For the efficient and successful functioning of these regulations, there is a need for a collective and synergetic approach, and closer interaction among the different government, non-government agencies, and private sectors which may play a diverse role in coordinating and harmonizing biosafety issues. Moreover, for adopting unified biosafety regulations, regional and international agencies should focus on the proper dissemination of information on biosafety regulations and public awareness about biosafety measures.

7. Controversies of GM Foods and Products

GMOs have become a controversial topic from the beginning. The supporters of GMOs including GM technologists, GM distributors, scientists and related regulatory agencies emphasize that GM products are non-toxic and nutritious [232,233], and potential to mitigate the global food crisis with no human health and environmental impacts. [234]. Moreover, several independent studies found no significant biological differences when GM crops and products were fed to the animals [235,236]. Some studies reported the presence of remnants of fragmented GM DNA in some parts of the gastrointestinal tracts, which were not detected in the blood and tissues [237,238]. Moreover, the in vitro experiment showed no horizontal transfer of GM DNA/genes to the microbes so far [237,238]. On the other hand, environmentalist opposes and rejected such results citing that the results were unacceptable due to methodological issues [190]. At the same time, opponents of GM believe that their exist differences between genetically engineered crops and traditional breeding plants. Moreover, Breckling et al. in their report pointed out a wide range of potential risks from GMOs including vertical gene transfer, horizontal gene transfer, hybridization, and resistance [239].
The risks potentials of transgene escape is high due to contamination in gene pool of crop landraces or wild relatives due to pollination of surrounding GM crops fields. Unwanted and unintended gene flow from the transgenic lines to wild relatives may produce genetically modified organism with unwanted traits that compete and displace the native species causing loss of genetic information [240]. Critics claims that the application of GMO can provoked the emergence of super weeds and pests that compel the use of more herbicides and pesticides to eliminate them from the field [240,241]. Moreover, various gene escape have been reported from oil rape to weedy relatives with glyphosate resistant trait [242], in creeping bent grass, in turf grass [243]. The transgene escape have been reported from Mexico in Maize landraces and cotton, that could change gene pool of maize landraces permanently [244,245,246]. A similar controversy has been reported in eggplant and its wild types [247].
The huge concern about the GMO is the corporate control of agriculture. Social activities from different parts of world believed that GM is private property, not national property [248]. The biotechnology companies has huge control over biotechnology process, genes and chemicals involve in the GM production process. As a result, handful of companies started protecting GM products, genes and chemical products through patents and licensing. For instance, Delta and Land Pine Company of Scott, Mississippi acquired patent on GM seed terminator that restricts unauthorized use of second generations’ seeds, thus, consolidating its control over seed market for making huge profit. They claimed that seed terminator technique would solve the contamination of gene pool of relative wild plant species [249]. The sterile seed produced by the GM crops would not produce offspring. This will cause the non-availability crops seeds to the farmers and prevent farmers re-planting seeds. As a result, the farmer would be severely affected by patent rights, as they are required to sign contracts for replantation every year and timely seed supply and seed conservation [250]. Lured by high yields, farmer would quickly abundant traditional landraces causing huge loss in the biodiversity. Therefore, risks assessment of transgene escape and its possible consequence of recombination in plant genome, by monitoring the potential harmful effects on wild relatives is important steps in all the GM crops.
In a report from Chile, controversies regarding biosafety regulation have emerged from different sectors due to a lack of public access to regulatory information, and the location of GM fields or farm sites [251]. Anti GM campaign was initiated in 2011 and supported by coalition, organizations, farmers, “green” legislators, and anti-GM groups [251]. Similarly, GM policy debates in Ghana were initiated after enacting biosafety regulations related to GM (Biosafety Act 831, December 2011) [252]. Opponents of GMOs comprised of individuals, farmers, and civil society claimed that GM is discriminatory, with environmental and human health issues [252]. In Mexico, a social movement made up of indigenous, peasant, civil, cultural and scientific community organizations came together in an organized way in defence of Biosafety and Genetically Modified Organisms Law in the year 2005 [253]. Recently, the government has initiated a ban on GM maize and restricted the approval of new GM cotton seeds. Secretariat of Environment and Natural Resources (SEMARNAT) cited concerns about the possibility of genetically modified varieties being crossed with the native varieties of wild maize and cotton found in the country [253]. The rejection of GM cotton release permits has had a significant reduction on the cotton plantation (dropped by 30–35 per cent in 2020) and yield as growers can now only access poor yields of cotton with ineffective protection against pests on cotton varieties and impacted heavily on textile in Mexico. In some EU members such as Poland, the opposition to the distribution and cultivation of GM crops is as high as 60% [254]. EU ban on GM rice import from China was initiated after detecting GM rice in the tested sample. Illegal and large-scale planting and production of GM rice have been the practice before the certificate for GM rice issued by the Chinese government. As result, GM rice has been detected in China market without completing the proper experimental and biosafety test [255]. After detecting the GM contamination of rice, the EU blocked the import of GM rice (Bt Shanyo 63) to enter into its market and tightens its rules governing the imports of GM rice from China [256]. Other countries such as Russia, Israel, Norway and Netherland restricted the cultivation and commercialization of GM crops [256]. Other permissive countries such as South Korea, New Zealand, France and China have more restrictive regulations and permit the least number or no GM crops for commercial cultivation [257]. Similarly, a 2016 survey carried out in China showed about 47% of people held a negative view of GM crops [258].

8. Final Considerations and Future Prospects

Biotechnology is emerging with new opportunities for the production of food and energy, especially in countries where food production is still insufficient. Greater advantages of biotechnology will be established in the field of agriculture for the future demands of food security. Biotechnology can also help in generating plant species rich in cellulose for the production of biofuels, but also has many challenges. There are several advantages related to the genetic transformation of plant species and their application in improving medicinal value; plants resistant to abiotic and biotic stresses, plants with better nutritional value, and biomolecules important for industrial and therapeutic products. The introduction of biotechnology that introduces exogenous genes has made it possible for breeders to produce cultivars with improved genetic traits, which was not possible before. The growing global demand around these sectors is essential for the application of genetic transformation strategies for more plant species. The genetic transformation will help in the advancement of plant species in the future; however, more research and detailed studies are required. Despite the advantages of this technique, there is growing concern regarding the establishment of regulations for the efficient and safe use of GM plant products, and it is important to share knowledge concerning GM crops, including risks and benefits in terms of human health and the environment.

Author Contributions

B.K.G. contributed by writing the manuscript; C.Y.Y., W.-R.K., H.-S.M., J.L., S.H.K. and I.M.C. contributed by analyzing data and editing the revised manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the National Research Foundation of Korea (NRF) grant funded by the Korea government (MSIT) (NO. RS-2022-00165400).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

This work was supported by funding from the KU Research Professor Program.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Shiferaw, B.; Smale, M.; Braun, H.-J.; Duveiller, E.; Reynolds, M.; Muricho, G. Crops that feed the world Past successes and future challenges to the role played by wheat in global food security. Food Secur. 2013, 5, 291–317. [Google Scholar] [CrossRef] [Green Version]
  2. UN News. Over 820 Million People Suffering from Hunger; New UN Report Reveals Stubborn Realities of ‘Immense’ Global Challenge. UN News 2019. [Google Scholar]
  3. Ray, D.K.; Mueller, N.D.; West, P.C.; Foley, J.A. Yield trends are insufficient to double global crop production by 2050. PLoS ONE 2013, 8, e66428. [Google Scholar] [CrossRef] [Green Version]
  4. Alexandratos, N.B.J. World Agriculture Towards 2030/2050. The 2012 Revision. Food and Agriculture Organization of the United Nations. ESA Working Paper no. 12-03. 2012. Available online: https://www.fao.org/3/ap106ee.pdf (accessed on 11 January 2023).
  5. Oliver, M.J. Why we need GMO crops in agriculture. MO Med. 2014, 111, 492–507. [Google Scholar]
  6. Moeller, L.; Wang, K. Engineering with Precision: Tools for the New Generation of Transgenic Crops. BioScience 2008, 58, 391. [Google Scholar] [CrossRef] [Green Version]
  7. Chandler, S.; Tanaka, Y. Genetic modifi cation in floriculture. Crit. Rev. Plant Sci. 2007, 26, 169–197. [Google Scholar] [CrossRef]
  8. Paiva, M.J.M.; Damasceno, I.A.M. O uso de dos alimentos geneticamente modificados: Principais desafios. Rev. Multidebates 2020, 4, 90–96. [Google Scholar]
  9. Torres, A.C.; Ferreira, A.T.; Sa, F.G.; Buso, J.A.; Caldas, L.S.; Nascimento, A.S.; Brigido, M.M.; Romano, E. Glossário De Biotecnologia Vegetal; Em Brapa-CNPH: Brasília, Brazil, 2000; p. 128. [Google Scholar]
  10. Welch, R.M. Biotechnology, biofortification and global health. Food Nutr Bull. 2005, 26, 4. [Google Scholar] [CrossRef]
  11. Nanjundan, J.; Singh, K.H.; Parmar, N.; Kumar, P.; Chauhan, D.K.; Khan, Y.J.; Thaku, A.K.; Sharma, D.; Singh, L. Genetic Engineering Strategies for Biotic and Abiotic Stress Tolerance and Quality Enhancement in Horticultural Crops: A Comprehensive Review. 3 Biotech 2017, 7, 1–35. [Google Scholar]
  12. Fischer, R.; Emans, N. Molecular farming of pharmaceutical proteins. Transgenic Res. 2000, 9, 279–299. [Google Scholar] [CrossRef]
  13. Lee, T.H.; Ho, H.K.; Leung, T.F. Genetically modified foods and allergy. Hong Kong Med. J. 2017, 23, 5–291. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Clive, J. Global Status of Commercialized Biotech/GM Crops; ISAAA Briefs 43; International Service for the Acquisition of Agri-Biotech Applications: Ithaca, NY, USA, 2011. [Google Scholar]
  15. Brookes, G.; Barfoot, P. Farm income and production impacts of using GM crop technology 1996–2016. GM. Crops Food 2018, 9, 59–89. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Gatew, H.; Mengistu, K. Genetically modified foods (GMOs); a review of genetic engineering. J. Life Sci. Biomed. 2019, 9, 157–163. [Google Scholar] [CrossRef]
  17. Ventura, M.V.A.; Batista, H.R.F.; Bessa, M.M.; Pereira, L.S.; Costa, E.M.; de Oliveira, M.H.R. Comparison of conventional and transgenic soybean production costs in different regions in Brazil. Res. Soc. Dev. 2020, 9, e154973977. [Google Scholar] [CrossRef]
  18. Kuiper, H.A.; Davies, H.V. The safe foods risk analysis framework suitable for GMOs? A case study. Food Control. 2010, 21, 1662–1676. [Google Scholar] [CrossRef]
  19. Twyman, R.M.; Kohli, A.; Stoger, E.; Christou, P. Foreign DNA: Integration and Expression in Transgenic Plants. In Genetic Engineering: Principles and Methods; Setlow, J.K., Ed.; Springer: Boston, MA, USA, 2002; Volume 24, pp. 107–136. [Google Scholar]
  20. Che1, P.; Chang, S.; Simon, M.K.; Zhang, Z.; Shaharyar, A.; Ourada, J.; O’Neill, D.; Torres-Mendoza, M.; Guo, Y.; Marasigan, K.M.; et al. Developing a rapid and highly efficient cowpea regeneration, transformation and genome editing system using embryonicaxis explants. Plant J. 2021, 106, 817–830. [Google Scholar] [CrossRef]
  21. Okpe, A.O.; Nka, F.A. Comparative Review of Plant Transformation Techniques. J. Adv. Biol. Biotechnol. 2021, 24, 1–18. [Google Scholar] [CrossRef]
  22. Matveeva, T.V.; Lutova, L.A. Horizontal gene transfer from Agrobacteriumto plants. Front. Plant Sci. 2014, 5, 1–11. [Google Scholar] [CrossRef] [Green Version]
  23. Xia, P.; Hu, W.; Liang, T.; Yang, D.; Liang, Z. An attempt toestablish an Agrobacterium-mediated transient expression system inmedicinal plants. Protoplasma 2020, 257, 1497–1505. [Google Scholar] [CrossRef]
  24. Ziemienowicz, A. Agrobacterium-mediated plant transformation: Factors, applications and recent advances. Bio Catal. Agric. Biotechnol. 2014, 3, 95–102. [Google Scholar] [CrossRef]
  25. Ghimire, B.K.; Seong, E.S.; Lim, J.D.; Heo, K.; Kim, M.J.; Chung, I.M.; Juvik, J.A.; Yu, C.Y. Agrobacterium-mediated transformation of Codonopsis lanceolata using the c-TMT gene. Plant Cell. Tiss. Organ. Cult. 2008, 95, 265–274. [Google Scholar] [CrossRef]
  26. Niazian, M.; Sadat Noori, S.A.; Galuszka, P.; Mortazavian, S.M.M. Tissue culture-based Agrobacterium-mediated and in planta transformation methods. Czech J. Genet. Plant Breed. 2017, 53, 133–143. [Google Scholar] [CrossRef] [Green Version]
  27. Christou, P. Genetic transformation of crops using microprojectile bombardment. Plant J. Oxf. 1992, 2, 275–281. [Google Scholar] [CrossRef]
  28. Lacroix, B.; Citovsky, V. Biolistic Approach for Transient Gene Expression Studies in Plants. Methods Mol. Biol. 2020, 2124, 125–139. [Google Scholar]
  29. Hernandez-Garcia, C.; Bouchard, R.; Rushton, P.; Jones, M.; Chen, X.; Timko, M.; Finer, J. High level transgenic expression of soybean (Glycine max) GmERF and Gmubi gene promoters isolated by a novel promoter analysis pipeline. BMC Plant Biol. 2010, 10, 237. [Google Scholar] [CrossRef]
  30. Taylor, N.J.; Fauquet, C.M. Microparticle bombardment as a tool in plant science and agricultural biotechnology. DNA Cell Biol. 2002, 21, 963–977. [Google Scholar] [CrossRef]
  31. Al-Dosari, M.S.; Gao, X. Non-viral Gene Delivery: Principle, limitations, and recent progress. AAPS J. 2009, 11, 671–681. [Google Scholar] [CrossRef] [Green Version]
  32. Rubinsky, B. Irreversible electroporation in medicine. Technol. Cancer Res. Treat. 2007, 6, 255–260. [Google Scholar] [CrossRef]
  33. United Naations. United Naations. United Nations System Standing Committee on Nutrition (SCN). In 5th Report on the World Nutrition Situation Nutrition for Improved Development Outcomes; SCN: Geneva, Switzerland, 2004. [Google Scholar]
  34. Szenkovics, D.; Tonk, M.; Balog, A. Can genetically modified (GM) crops act as possible alternatives to mitigate world political conflicts for food? Food Energy Secur. 2021, 10, e268. [Google Scholar] [CrossRef]
  35. Kamthan, A.; Chaudhuri, A.; Kamthan, M.; Datta, A. Genetically modified (GM) crops: Milestones and new advances in crop improvement. Theor. Appl. Genet. 2016, 129, 1639–1655. [Google Scholar] [CrossRef]
  36. Singh, O.V.; Ghai, S.; Paul, D.; Jain, R.K. Genetically modified crops: Success, safety assessment, and public concern. Appl. Microbiol. Biotechnol. 2006, 71, 598–607. [Google Scholar] [CrossRef] [PubMed]
  37. Taylor, S.L.; Goodman, R.E.; Hefle, S.L. The development of safety assessment for genetically modified foods. Asia Pac. Biotech. News 2006, 10, 614–616. [Google Scholar] [CrossRef]
  38. American Medical Association. Report 2 of the Council on Science and Public Health: Labeling of Bioengineered Foods; American Medical Association: Washington, DC, USA, 2012. [Google Scholar]
  39. Gilbert, P.R. Biotechnology Industries and Entrepreneurs; Darya Ganj: New Delhi, India, 2008; pp. 25–180. [Google Scholar]
  40. Houdebine, L.M. Impacts of genetically modified animals on the ecosystem and human activities. Glob. Bioeth. 2014, 25, 3–18. [Google Scholar] [CrossRef] [Green Version]
  41. Schubert, D. A different perspective on GM food. Nat. Biotechnol. 2002, 20, 969. [Google Scholar] [CrossRef] [PubMed]
  42. Wong, M.S.; Hawthorne, W.J.; Manolios, N. Gene therapy in diabetes. Self Nonself Pharm. 2010, 1, 165–175. [Google Scholar] [CrossRef]
  43. Matsuda, Y.; Toyoda, H.; Sawabe, A.; Maeda, K.; Shimizu, N.; Fujita, N.; Fujita, T.; Nonomura, T.; Ouchi, S. A hairy root culture of melon produces aroma compounds. J. Agric. Food Chem. 2000, 48, 1417–1420. [Google Scholar] [CrossRef]
  44. Oller, A.L.W.; Agostini, E.; Talano, M.A.; Capozucca, C.; Milrad, S.R.; Tigier, H.A.; Medina, M.I. Overexpression of a basic peroxidase in transgenic tomato (Lycopersicon esculentum Mill. cv. Pera) hairy roots increases phytoremediation of phenol. Plant Sci. 2005, 169, 1102–1111. [Google Scholar] [CrossRef]
  45. Garagounis, C.; Beritza, K.; Georgopoulou, M.E.; Sonawane, P.; Haralampidis, K.; Goossens, A.; Aharoni, A.; Papadopoulou, K.K. A hairy-root transformation protocol for Trigonella foenum-graecum L. as a tool for metabolic engineering and specialised metabolite pathway elucidation. Plant Physiol. Biochem. 2020, 154, 451–462. [Google Scholar] [CrossRef]
  46. Kundua, S.; Salma, U.; Ali, M.N.; Hazra, A.K.; Mandal, N. Development of transgenic hairy roots and augmentation of secondary metabolites by precursor feeding in Sphagneticola calendulacea (L.) Pruski. Ind. Crops Prod. 2018, 121, 206–215. [Google Scholar] [CrossRef]
  47. Xin, Y.; Ma, B.; Zeng, Q.; He, W.; Qin, M.; He, N. Dynamic changes in transposable element and gene methylation in mulberry (Morus notabilis) in response to Botrytis cinerea. Hortic. Res. 2021, 8, 154. [Google Scholar] [CrossRef]
  48. Paine, J.A.; Shipton, C.A.; Chaggar, S.; Howells, R.M.; Kennedy, M.J.; Vernon, G.; Wright, S.Y.; Hinchliffe, E.; Adams, J.L.; Silverstone, A.L.; et al. Improving the nutritional value of Golden Rice through increased pro-vitamin A content. Nat. Biotechnol. 2005, 23, 482–487. [Google Scholar] [CrossRef] [PubMed]
  49. Gao, Y.; Ma, Y.; Li, M.; Cheng, T.; Li, S.W.; Zhang, J.; Xia, N.S. Oral immunization of animals with transgenic cherry tomatillo expressing HbsAg. World J. Gastroenterol. 2003, 9, 996–1002. [Google Scholar] [CrossRef] [PubMed]
  50. Van Eenennaam, A.L.; Lincoln, K.; Durrett, T.P.; Valentin, H.E.; Shewmaker, C.K.; Thorne, G.M. Engineering vitamin E content: From Arabidopsis mutant to soil oil. Plant Cell. 2003, 5, 3007–3019. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Suzuki, Y.A.; Shin, K.; Lönnerdal, B. Molecular cloning and functional expression of a human intestinal lactoferrin receptor. Biochemistry 2001, 40, 15771–15779. [Google Scholar] [CrossRef]
  52. Xiaoyan, S.; Yan, Z.; Shubin, W. Improvement Fe content of wheat (Triticum aestivum) grain by soybean ferritin expression cassette without vector backbone sequence. J. Agric. Food Biochem. 2012, 20, 766–773. [Google Scholar]
  53. Drakakaki, G.; Marcel, S.; Glahn, R.P.; Lund, E.K.; Pariagh, S.; Fischer, R.; Christou, P.; Stoger, E. Endosperm-specific co-expression of recombinant soybean ferritin and Aspergillus phytase in maize results in significant increases in the levels of bioavailable iron. Plant Mol. Biol. 2005, 59, 869–880. [Google Scholar] [CrossRef]
  54. Goto, F.; Yoshihara, T. Improvement of micronutrient contents by genetic engineering development of high iron content crops. Plant Biotechnol. 2001, 18, 7–15. [Google Scholar] [CrossRef] [Green Version]
  55. Masuda, H.; Usuda, K.; Kobayashi, T.; Ishimaru, Y.; Kakei, Y.; Takahashi, M.; Higuchi, K.; Nakanishi, H.; Mori, S.; Nishizawa, N.K. Overexpression of the barley nicotianamine synthase gene HvNAS1 increases iron and zinc concentrations in rice grains. Rice 2009, 2, 155–166. [Google Scholar] [CrossRef] [Green Version]
  56. Ghimire, B.K.; Seong, E.S.; Lee, C.O.; Lim, J.D.; Lee, J.G.; Yoo, J.H.; Chung, I.M.; Kim, N.Y.; Yu, C.Y. Enhancement of α-tocopherol content in transgenic Perilla frutescens containing the γ-TMT gene. Afr. J. Biotechnol. 2011, 10, 2430–2439. [Google Scholar]
  57. Long, M.; Millar, D.J.; Kimura, Y.; Donovan, G.; Rees, J.; Fraser, P.D.; Bramley, P.M.; Bolwell, G.P. Metabolite profiling of carotenoid and phenolic pathways in mutant and transgenic lines of tomato: Identification of a high antioxidant fruit line. Phytochemistry 2006, 67, 1750–1757. [Google Scholar] [CrossRef]
  58. Lim, J.D.; Yang, D.C.; Yun, S.J.; Chung, I.M.; Sung, E.S.; Kim, M.J.; Heo, K.; Yu, C.Y. Isolation and Biological Activity of Resveratrol−3−O−β−D−GlucosideResveratrol−3−O−β−D−Glucoside in Transgenic Rehmannia glutinosa L. Transformed by Peanut Resveratrol Synthase Gene (RS3). Korean J. Med. Crop Sci. 2004, 12, 406–414. [Google Scholar]
  59. D’Introno, A.; Paradiso, A.; Scoditti, E.; D’Amico, L.; De Paolis, A.; Carluccio, M.A.; Nicoletti, I.; De Gara, L.; Santino, A.; Giovinazzo, G. Antioxidant and anti-inflammatory properties of tomato fruits synthesizing different amounts of stilbenes. Plant Biotechnol. J. 2000, 7, 422–429. [Google Scholar] [CrossRef] [PubMed]
  60. Beasley, J.T.; Bonneau, J.P.; Sánchez-Palacios, J.T.; Moreno-Moyano, L.T.; Callahan, D.L.; Tako, E.; Glahn, R.P.; Lombi, E.; Johnson, A.A.T. Metabolic engineering of bread wheat improves grain iron concentration and bioavailability. Plant Biotechnol. J. 2019, 17, 1514–1526. [Google Scholar] [CrossRef] [Green Version]
  61. Narayanan, N.; Beyene, G.; Chauhan, R.D.; Gaitán-Solís, E.; Gehan, J.; Butts, P.; Siritunga, D.; Okwuonu, I.; Woll, A.; Jiménez- Aguilar, D.M.; et al. Biofortification of field-grown cassava by engineering expression of an iron transporter and ferritin. Nat. Biotechnol. 2019, 37, 144–151. [Google Scholar] [CrossRef] [PubMed]
  62. Broad, R.C.; Bonneau, J.P.; Beasley, J.T.; Roden, S.; Sadowski, P.; Jewell, N.; Brien, C.; Berger, B.; Tako, E.; Glahn, R.P.; et al. Effect of Rice GDP-L-Galactose Phosphorylase Constitutive Overexpression on Ascorbate Concentration, Stress Tolerance, and Iron Bioavailability in Rice. Front. Plant Sci. 2020, 11, 595439. [Google Scholar] [CrossRef]
  63. Kim, W.-S.; Sun-Hyung, J.; Oehrle, N.W.; Jez, J.M.; Krishnan, H.B. Overexpression of ATP sulfurylase improves the sulfur amino acid content, enhances the accumulation of Bowman-Birk protease inhibitor and suppresses the accumulation of the β-subunit of β-conglycinin in soybean seeds. Sci. Rep. 2020, 10, 14989. [Google Scholar] [CrossRef]
  64. Chang, C.K.; Chang, K.S.; Lin, Y.C.; Liu, S.Y.; Chen, C.Y. Hairy root cultures of Gynostemma pentaphyllum (Thunb.) Makino: A promising approach for the production of gypenosides as an alternative of ginseng saponins. Biotechnol Lett. 2005, 27, 1165–1169. [Google Scholar] [CrossRef]
  65. Thiruvengadam, M.; Praveen, N.; Kim, S.H.; Chung, I.M. Establishment of Momordica charantia hairy root cultures for the production of phenolic compounds and the determination of their biological activities. Plant Cell Tissue Organ Cult. 2014, 118, 545–557. [Google Scholar] [CrossRef]
  66. Thiruvengadam, M.; Rekha, K.; Chung, I.M. Induction of hairy roots by Agrobacterium rhizogenes-mediated transformation of spine gourd (Momordica dioica Roxb. ex. willd) for the assessment of phenolic compounds and biological activities. Sci. Hortic. 2016, 198, 132–141. [Google Scholar] [CrossRef]
  67. Yoon, J.Y.; Chung, I.M.; Thiruvengadam, M. Evaluation of phenolic compounds, antioxidant and antimicrobial activities from transgenic hairy root cultures of gherkin (Cucumis anguria L.). S. Afr. J. Bot. 2015, 100, 80–86. [Google Scholar] [CrossRef]
  68. Le Gall, G.; DuPont, M.S.; Mellon, F.A.; Davis, A.L.; Collins, G.J.; Verhoeyen, M.E.; Colquhoun, I.J. Characterization and Content of Flavonoid Glycosides in Genetically Modified Tomato (Lycopersicon esculentum) Fruits. J. Agric. Food Chem. 2003, 51, 2438–2446. [Google Scholar] [CrossRef] [PubMed]
  69. Lee, C.J.; Kim, S.E.; Park, S.U.; Lim, Y.H.; Choi, H.Y.; Kim, W.G.; Ji, C.Y.; Kim, H.S.; Kwak, S.S. Tuberous roots of transgenic sweet potato overexpressing IbCAD1 have enhanced low-temperature storage phenotypes. Plant Physiol. Biochem. 2021, 166, 549–557. [Google Scholar] [CrossRef] [PubMed]
  70. Yoo, J.H.; Seong, E.S.; Ghimire, B.K.; Heo, K.; Jin, X.; Yamada, T.; Clark, L.V.; Sacks, E.J.; Yu, C.Y. Establishment of Miscanthus sinensis with decreased lignin biosynthesis by Agrobacterium–mediated transformation using antisense COMT gene. Plant Cell Tissue Organ Cult. 2018, 133, 359–369. [Google Scholar] [CrossRef]
  71. Wang, F.; Kong, W.; Wong, G.; Fu, L.; Peng, R.; Li, Z.; Yao, Q. AtMYB12 regulates flavonoids accumulation and abiotic stress tolerance in transgenic Arabidopsis thaliana. Mol. Gen. Genom. 2016, 291, 1545–1559. [Google Scholar] [CrossRef] [PubMed]
  72. Wang, B.; Li, X.; Tabudravu, J.; Wang, S.; Deng, H.; Pan, L. The chemical profile of activated secondary metabolites by overexpressing LaeA in Aspergillus niger. Microbiol. Res. 2021, 248, 126735. [Google Scholar] [CrossRef] [PubMed]
  73. Prashant, S.; Srilakshmi Sunita, M.; Pramod, S.; Gupta, R.K.; Anil Kumar, S.; Karumanchi, S.R.; Rawal, S.K.; Kavi Kishor, P.B. Down-regulation of Leucaena leucocephala cinnamoyl CoA reductase (LlCCR) gene induces significant changes in phenotype, soluble phenolic pools and lignin in transgenic tobacco. Plant Cell Rep. 2011, 30, 2215–2231. [Google Scholar] [CrossRef]
  74. Chung, I.M.; Rekha, K.; Rajakumar, G.; Thiruvengadam, M. Production of glucosinolates, phenolic compounds and associated gene expression profiles of hairy root cultures in turnip (Brassica rapa ssp. rapa). 3 Biotech 2016, 6, 175. [Google Scholar] [CrossRef] [Green Version]
  75. Tusevski, O.; Petreska Stanoeva, J.; Stefova, M.; Pavokovic, D.; Gadzovska Simic, S. Identification and quantification of phenolic compounds in Hypericum perforatum L. transgenic shoots. Acta Physiol. Plant 2014, 36, 2555–2569. [Google Scholar] [CrossRef]
  76. Galis, I.; Kakiuchi, Y.; Simek, P.; Wabiko, H. Agrobacterium tumefaciens AK-6b gene modulates phenolic compound metabolism in tobacco. Phytochemistry 2004, 65, 169–179. [Google Scholar] [CrossRef]
  77. Landtag, J.; Baumert, A.; Degenkolb, T.; Schmidt, J.; Wray, V.; Scheel, D.; Strack, D.; Rosahl, S. Accumulation of tyrosol glucoside in transgenic potato plants expressing a parsley tyrosine decarboxylase. Phytochemistry 2002, 60, 683–689. [Google Scholar] [CrossRef]
  78. Fu, R.; Shi, M.; Deng, C.; Zhang, Y.; Zhang, X.; Wang, Y.; Kai, G. Improved phenolic acid content and bioactivities of Salvia miltiorrhiza hairy roots by genetic manipulation of RAS and CYP98A. Food Chem. 2020, 331, 1273652. [Google Scholar] [CrossRef] [PubMed]
  79. Schnablova, T.; Synkova, H.; Vicankova, A.; Burketova, L.; Eder, J.; Cvikrova, M. Transgenic ipt tobacco over producing cytokinins over accumulates phenolic compounds during in vitro growth. Physiol. Bio Chem. 2006, 44, 526–534. [Google Scholar]
  80. Dilshad, E.; Cusido, R.M.; Estrada, K.R.; Bonfill, M.; Mirza, B. Genetic Transformation of Artemisia carvifolia Buch with rol Genes Enhances Artemisinin Accumulation. PLoS ONE 2015, 10, e0140266. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Sahayarayan, J.; Udayakumar, R.; Arun, M.; Ganapathi, A.; Alwahibi, M.S.; Aldosari, N.S.; Morgan, A.M.A. Effect of different Agrobacterium rhizogenes strains for in-vitro hairy root induction, total phenolic, flavonoids contents, antibacterial and antioxidant activity of (Cucumis anguria L.). Saudi J. Biol. Sci. 2020, 27, 2972–2979. [Google Scholar] [CrossRef]
  82. Guo, D.; Chen, F.; Dixon, R.A. Monolignol biosynthesis in microsomal preparations from lignifying stems of alfalfa (Medicago sativa L.). Phytochemistry 2002, 61, 657–667. [Google Scholar] [CrossRef]
  83. Cha, T.S.; Chen, C.F.; Yee, W.; Aziz, A.; Loh, S.H. Cinnamic acid, coumarin and vanillin: Alternative phenolic compounds for efficient Agrobacterium-mediated transformation of the unicellular green alga, Nannochloropsis sp. J. Microbiol. Methods 2011, 84, 430–434. [Google Scholar] [CrossRef]
  84. Lorenc-Kukuła, K.; Amarowicz, R.; Oszmiański, J.; Doer_mann, P.; Starzycki, M.; Skała, J.; Zuk, M.; Kulma, A.; Szopa, J. Pleiotropic Effect of Phenolic Compounds Content Increases in Transgenic Flax Plant. J. Agric. Food Chem. 2005, 53, 3685–3692. [Google Scholar] [CrossRef]
  85. Ayaz, M.; Ali, Q.; Farzand, A.; Khan, A.R.; Ling, H.; Gao, X. Nematicidal volatiles from bacillus atrophaeus gbsc56 promote growth and stimulate induced systemic resistance in tomato against meloidogyne incognita. Int. J. Mol. Sci. 2021, 22, 5049. [Google Scholar] [CrossRef]
  86. Peng, D.; Wan, D.; Cheng, C.; Ye, X.; Sun, M. Nematode-specific cadherin CDH-8 acts as a receptor for Cry5B toxin in Caenorhabditis elegans. Appl. Microbiol. Biotechnol. 2018, 102, 3663–3673. [Google Scholar] [CrossRef]
  87. Stewart, C.N., Jr.; Adang, M.J.; All, J.N.; Raymer, P.L.; Ramachandran, S.; Parrott, W.A. Insect control and dosage effects in transgenic canola containing a synthetic Bacillus thuringiensis cryIAc gene. Plant Physiol. 1996, 112, 115–120. [Google Scholar] [CrossRef] [Green Version]
  88. Singh, S.; Kumar, N.R.; Maniraj, R.; Lakshmikanth, R.; Rao, K.Y.S.; Muralimohan, N.; Arulprakash, T.; Karthik, K.; Shashibhushan, N.B.; Vinutha, T. Expression of Cry2Aa, a Bacillus thuringiensis insecticidal protein in transgenic pigeon pea confers resistance to gram pod borer, Helicoverpa armigera. Sci. Rep. 2018, 8, 8820. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Bríza, J.; Pavingerová, D.; Vlasák, J.; Niedermeierová, H. Norway spruce (Picea abies) genetic transformation with modified Cry3A gene of Bacillus thuringiensis. Acta Biochim. Pol. 2013, 60, 395–400. [Google Scholar] [CrossRef] [Green Version]
  90. Heimlich, R.E.; Fernandez-Cornejo, J.F.; McBride, W.; Klotz-Ingram, C.; Jans, S.; Brooks, N. Adoption of Genetically Engineered Seed in US Agriculture: Implication for Pesticide Use; sld001; USDA Publication: Washington, DC, USA, 2000. [Google Scholar]
  91. United States Department of Agriculture. Genetically Engineered Crops: Has Adoption Reduced Pesticide Use? Agricultural Outlook August. 2000. Available online: www.ers.usda.gov/publications/agoutlook/aug2000/ao273f.pdf (accessed on 31 August 2000).
  92. Tohidfar, M.; Zare, N.; Jouzani, G.S.; Eftekhari, S.M. Agrobacterium mediated transformation of alfalfa (Medicago sativa) using a synthetic cry3a gene to enhance resistance against alfalfa weevil. Plant Cell Tissue Organ Cult. (PCTOC) 2013, 113, 227–235. [Google Scholar] [CrossRef]
  93. Alfonso-Rubí, J.; Ortego, F.; Castañera, P.; Carbonero, P.; Díaz, I. Transgenic expression of trypsin inhibitor CMe from barley in Indica and Japonica rice, confers resistance to the rice weevil Sitophilus oryzae. Transgenic Res. 2003, 12, 2331. [Google Scholar] [CrossRef] [PubMed]
  94. Tougou, M.; Furutani, N.; Yamagishi, N.; Shizukawa, Y.; Takahata, Y.; Hidaka, S. Development of resistant transgenic soybeans with inverted repeat-coat protein genes of soybean dwarf virus. Plant Cell Rep. 2006, 25, 1213–1218. [Google Scholar] [CrossRef] [PubMed]
  95. Franco, M.C.; Gomes, K.A.; de Carvalho Filho, M.M.; Ricardo Harakava, R.; Carels, N.; Siqueira, W.J.; Latado, R.R.; de Argollo Marques, D. Agrobacterium-mediated transformation of Jatropha curcas leaf explants with a fungal chitinase gene. Afr. J. Biotechnol. 2016, 15, 2006–2016. [Google Scholar]
  96. Macrae, T.C.; Baur, M.E.; Boethel, D.J.; Fitzpatrick, B.J.; Gao, A.G.; Gamundi, J.C.; Harrison, L.A.; Kabuye, V.T.; Mcpherson, R.M.; Miklos, J.A.; et al. Laboratory and field evaluations of transgenic soybean exhibiting highdose expression of a synthetic Bacillus thuringiensis cry1A gene for control of Lepidoptera. J. Econ. Entomol. 2005, 98, 577–587. [Google Scholar] [CrossRef]
  97. Tohidfar, M.; Ghareyazie, B.; Mosavi, M.; Yazdani, S.; Golabchian, R. Agrobacterium-mediated transformation of cotton (Gossypium hirsutum) using a synthetic cry1Ab gene for enhanced resistance against Heliothis armigera. Iran. J. Biotechnol. 2008, 6, 164–173. [Google Scholar]
  98. Papolu, P.K.; Dutta, T.K.; Tyagi, N.; Urwin, P.E.; Lilley, C.J.; Rao, U. Expression of a cystatin transgene in eggplant provides resistance to root-knot nematode, Meloidogyne incognita. Front Plant. Sci. 2016, 7, 1122. [Google Scholar] [CrossRef] [Green Version]
  99. Zhang, H.Y.; Liu, H.M.; Liu, X.Z. Production of transgenic kiwifruitplants harboring the SbtCry1Ac gene. Genet. Mol. Res. 2015, 14, 8483–8489. [Google Scholar] [CrossRef]
  100. Heydarian, Z.; Yu, M.; Gruber, M.; Glick, B.R.; Zhou, R.; Hegedus, D.D. Inoculation of soil with plant growth promoting bacteria producing 1-aminocyclopropane-1-carboxylate deaminase or expression of the corresponding acdS gene in transgenic plants increases salinity tolerance in Camelina sativa. Front. Microbiol. 2016, 7, 1–17. [Google Scholar] [CrossRef] [PubMed]
  101. Wang, L.; Gao, J.; Qin, X.; Shi, X.; Luo, L.; Zhang, G.; Yu, H.; Li, C.; Hu, M.; Liu, Q.; et al. JcCBF2 gene from Jatropha curcas improves freezing tolerance of Arabidopsis thaliana during the early stage of stress. Mol. Biol. Rep. 2015, 42, 937–945. [Google Scholar] [CrossRef] [PubMed]
  102. Park, W.; Feng, Y.; Ahn, S.J. Alteration of leaf shape, improved metal tolerance, and productivity of seed by overexpression of CsHMA3 in Camelina sativa. Biotechnol. Biofuels 2014, 7, 96. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Losada, O.A.; Fonseca, C.A.G. Alimentos transgenicosy alergenicidad. Rev. Fac. Med. 2007, 55, 251–269. [Google Scholar]
  104. Dodo, H.W.; Konan, N.K.; Chen, F.C.; Egnin, M.; Viquez, O.M. Alleviating peanut allergy using genetic engineering: The silencing of the immunodominant allergen Ara h 2 leads to its significant reduction and a decrease in peanut allergenicity. Plant Biotechnol. J. 2008, 6, 135–145. [Google Scholar] [CrossRef]
  105. Le, L.Q.; Mahler, V.; Lorenz, Y.; Scheurer, S.; Biemelt, S.; Vieths, S.; Sonnewald, U. Reduced allergenicity of tomato fruits harvested from Lyc e 1-silenced transgenic tomato plants. J. Allergy Clin. Immunol. 2006, 118, 1176–1183. [Google Scholar] [CrossRef]
  106. Gilissen, L.J.; Bolhaar, S.T.; Matos, C.I.; Rouwendal, G.J.; Boone, M.J.; Krens, F.A.; Zuidmeer, L.; Van Leeuwen, A.; Akkerdaas, J.; Hoffmann-Sommergruber, K.; et al. Silencing the major apple allergen Mal d 1 by using the RNA interference approach. J. Allergy Clin. Immunol. 2005, 115, 364–369. [Google Scholar] [CrossRef]
  107. Herman, E.M.; Helm, R.M.; Jung, R.; Kinney, A.J. Genetic modification removes an immunodominant allergen from soybean. Plant Physiol. 2003, 132, 36–43. [Google Scholar] [CrossRef] [Green Version]
  108. Bhalla, P.L.; Swoboda, I.; Singh, M.B. Reduction in allergenicity of grass pollen by genetic engineering. Int. Arch. Allergy Immunol. 2001, 124, 51–54. [Google Scholar] [CrossRef]
  109. Garbisu, C.; Hernandez, A.J.; Barrutia, O.; Alkorta, I.; Becerril, J.M. Phytoremediation: A technology using green plants to remove contaminants from polluted areas. Rev. Environ. Health 2002, 17, 173–188. [Google Scholar] [CrossRef]
  110. Sun, L.; Ma, Y.; Wang, H.; Huang, W.; Wang, X.; Han, L.; Sun, W.; Han, E.; Wang, B. Overexpression of PtABCC1 contributes to mercury tolerance and accumulation in Arabidopsis and poplar. Biochem. Biophys. Res. Commun. 2018, 497, 997–1002. [Google Scholar] [CrossRef] [PubMed]
  111. Pilon-Smits, E. Phytoremediation. Annu. Rev. Plant Biol. 2005, 56, 15–39. [Google Scholar] [CrossRef] [PubMed]
  112. Gisbert, C.; Ros, R.; Haro, A.D.; Walker, D.J.; Bernal, M.P.; Serrano, R.; Avino, J.N. A plant genetically modified that accumulates Pb is especially promising for phytoremediation. Biochem. Biophys. Res. Commun. 2003, 303, 440–445. [Google Scholar] [CrossRef] [PubMed]
  113. Li, Y.; Dhankher, O.P.; Carreira, L.; Lee, D.; Chen, A.; Schroeder, J.I.; Balish, R.S.; Meagher, R.B. Overexpression of phytochelatin synthase in Arabidopsis leads to enhanced arsenic tolerance and cadmium hypersensitivity. Plant Cell Physiol. 2004, 45, 1787–1797. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Moreno, F.N.; Anderson, C.W.; Stewart, R.B.; Robinson, B.H. Mercury volatilisation and phytoextraction from base-metal mine tailings. Environ. Pollut. 2005, 136, 341–352. [Google Scholar] [CrossRef] [PubMed]
  115. Bizily, S.P.; Rugh, C.L.; Meagher, R.B. Phytodetoxification of hazardous organomercurials by genetically engineered plants. Nat. Biotechnol. 2000, 18, 213–217. [Google Scholar] [CrossRef] [PubMed]
  116. Van Huysen, T.; Abdel-Ghany, S.; Hale, K.L.; LeDuc, D.; Terry, N.; Pilon-Smits, E.A. Overexpression of cystathionine-gamma- synthase enhances selenium volatilization in Brassica juncea. Planta 2003, 218, 71–78. [Google Scholar] [CrossRef]
  117. Wolfe, N.L.; Hoehamer, C.F. Enzymes used by plants and microorganisms to detoxify organic compounds. In Phytore-Mediation: Transformation and Control of Contaminants; McCutcheon, S.C., Schnoor, J.L., Eds.; New Wiley: New York, NY, USA, 2003; pp. 159–187. [Google Scholar]
  118. Collins, C.; Laturnus, F.; Nepovim, A. Remediation of BTEX and trichloroethene. Current knowledge with special emphasis on phytoremediation. Environ. Sci. Pollut. Res. Int. 2002, 9, 86–94. [Google Scholar] [CrossRef]
  119. Gullner, G.; Komives, T.; Rennenberg, H. Enhanced tolerance of transgenic poplar plants overexpressing g-glutamylcysteine synthetase towards chloroacetanilide herbicides. J. Exp. Bot. 2001, 52, 971–979. [Google Scholar] [CrossRef]
  120. Flocco, C.G.; Lindblom, S.D.; Smits, E.A. Overexpression of enzymes involved in glutathione synthesis enhances tolerance to organic pollutants in Brassica juncea. Int. J. Phytochem. 2004, 6, 289–304. [Google Scholar] [CrossRef]
  121. Migocka, M.; Papierniak, A.; Kosieradzka, A.; Posyniak, E.; Maciaszczyk-Dziubinska, E.; Biskup, R.; Garbiec, A.; Marchewka, T. Cucumber metal tolerance protein CsMTP9 is a plasma membrane H1-coupled antiporter involved in the Mn21 and Cd21 efflux from root cells. Plant J. 2015, 84, 10451058. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Das, N.; Bhattacharya, S.; Maiti, M.K. Enhanced cadmium accumulation and tolerance in transgenic tobacco overexpressing rice metal tolerance protein gene OsMTP1 is promising for phytoremediation. Plant. Physiol. Biochem. 2016, 105, 297309. [Google Scholar] [CrossRef] [PubMed]
  123. Yang, J.; Chen, Z.; Wu, S.; Cui, Y.; Zhang, L.; Dong, H. Overexpression of the Tamarix hispida ThMT3 gene increases copper tolerance and adventitious root induction in Salix matsudana Koidz. Plant Cell Tissue Organ Cult. (PCTOC) 2015, 121, 469479. [Google Scholar] [CrossRef]
  124. Zanella, L.; Fattorini, L.; Brunetti, P.; Roccotiello, E.; Cornara, L.; D’Angeli, S.; Della Rovere, F.; Cardarelli, M.; Barbieri, M.; Sanità di Toppi, L. Overexpression of AtPCS1 in tobacco increases arsenic and arsenic plus cadmium accumulation and detoxification. Planta 2016, 243, 605622. [Google Scholar] [CrossRef] [PubMed]
  125. Ai, T.N.; Naing, A.H.; Yun, B.W.; Kim, C.K. Overexpression of RsMYB1 enhances heavy metal stress tolerance in transgenic petunia by elevating the transcript levels of stress tolerant and antioxidant genes. bioRxiv 2018, 3028, 286849. [Google Scholar]
  126. Chen, J.; Yang, L.; Yan, X.; Liu, Y.; Wang, R.; Fan, T.; Ren, Y.; Tang, X.; Xiao, F.; Liu, Y.; et al. Zinc-finger transcription factor ZAT6 positively regulates cadmium tolerance through glutathionedependent pathway in Arabidopsis. Plant Physiol. 2016, 171, 01882.2015. [Google Scholar] [CrossRef]
  127. Liu, D.; An, Z.; Mao, Z.; Ma, L.; Lu, Z. Enhanced heavy metal tolerance and accumulation by transgenic sugar beets expressing Streptococcus thermophilus StGCSGS in the presence of Cd, Zn and Cu alone or in combination. PLoS ONE 2015, 10, e0128824. [Google Scholar]
  128. Wang, J.W.; Li, Y.; Zhang, Y.X.; Chai, T.Y. Molecular cloning and characterization of a Brassica juncea yellow stripe-like gene, BjYSL7, whose overexpression increases heavy metal tolerance of tobacco. Plant Cell Rep. 2013, 32, 651662. [Google Scholar] [CrossRef]
  129. Brunetti, P.; Zanella, L.; Paolis, A.D.; Litta, D.D.; Cecchetti, V.; Falasca, G.; Barbieri, M.; Altamura, M.M.; Costantino, P.; Cardarelli, M. Cadmium-inducible expression of the ABC-type transporter AtABCC3 increases phytochelatin-mediated cadmium tolerance in Arabidopsis. J. Exp. Bot. 2015, 66, 38153829. [Google Scholar] [CrossRef] [Green Version]
  130. Meng, J.G.; Zhang, X.D.; Tan, S.K.; Yang, Z.M. Genome-wide identification of Cd-responsive NRAMP transporter genes and analyzing expression of NRAMP 1 mediated by miR167 in Brassica napus. Biometals 2017, 30, 917931. [Google Scholar] [CrossRef]
  131. Banuelos, G.; Terry, N.; LeDuc, D.L.; Pilon-Smits, E.A.; Mackey, B. Field trial of transgenic Indian mustard plants shows enhanced phytoremediation of selenium-contaminated sediment. Environ. Sci. Technol. 2005, 39, 1771. [Google Scholar] [CrossRef]
  132. Dominguez-Solis, J.R.; Lopez-Martin, M.C.; Ager, F.J.; Ynsa, M.D.; Romero, L.C.; Gotor, C. Increased cysteine availability is essential for cadmium tolerance and accumulation in Arabidopsis thaliana. Plant Biotechnol. J. 2004, 2, 469–476. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Bai, J.; Wang, X.; Wang, R.; Wang, J.; Le, S.; Zhao, Y. Overexpression of Three Duplicated BnPCS Genes Enhanced Cd Accumulation and Translocation in Arabidopsis thaliana Mutant cad1. Bull. Environ. Contam. Toxicol. 2019, 102, 146–152. [Google Scholar] [CrossRef]
  134. Nehnevajova, E.; Mireddy, E.; Stolz, A.; Gerdemann-Knörck, M.; Novák, O.; Strnad, M.; Schmülling, T. Root enhancement in cytokinin-deficient oilseed rape causes leaf mineral enrichment, increases the chlorophyll concentration under nutrient limitation and enhances the phytoremediation capacity. BMC Plant Biol. 2019, 19, 83. [Google Scholar] [CrossRef] [PubMed]
  135. Yoshida, T.; Kimura, E.; Koike, S.; Nojima, J.; Futai, E.; Sasagawa, N.; Watanabe, Y.; Ishiura, S. Transgenic rice expressing amyloid βpeptide for oral immunization. Int. J. Biol. Sci. 2011, 7, 301. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Rybicki, E.P. Plant-made vaccines for humans and animals. Plant Biotechnol. J. 2010, 8, 620–637. [Google Scholar] [CrossRef] [PubMed]
  137. Xu, J.; Ge, X.; Dolan, M.C. Towards high-yield production of pharmaceutical proteins with plant cell suspension cultures. Biotechnol. Adv. 2011, 29, 278–299. [Google Scholar] [CrossRef]
  138. Sala, F.; Rigano, M.M.; Barbante, A.; Basso, B.; Walmsley, A.M.; Castiglione, S. Vaccine antigen production in transgenic plants: Strategies, gene constructs and perspectives. Vaccine 2003, 21, 803–808. [Google Scholar] [CrossRef]
  139. Daniell, H.; Kumar, S.; Dufourmantel, N. Breakthrough in chloroplast genetic engineering of agronomically important crops. Trend. Biotechnol. 2005, 23, 238–245. [Google Scholar] [CrossRef] [Green Version]
  140. Scotti, N.; Rigano, M.M.; Cardi, T. Production of foreign proteins using plastid transformation. Biotechnol. Adv. 2012, 30, 387–397. [Google Scholar] [CrossRef]
  141. Marsian, J.; Fox, H.; Bahar, M.W.; Kotecha, A.; Fry, E.E.; Stuart, D.I.; Macadam, A.J.; Rowlands, D.J.; Lomonossoff, G.P. Plant-made polio type 3 stabilized VLPs—A candidate synthetic polio vaccine. Nat. Commun. 2017, 8, 1–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Richter, L.J.; Thanavala, Y.; Arntzen, C.J.; Mason, H.S. Production of hepatitis B surface antigen in transgenic plants for oral immunization. Nat. Biotechnol. 2000, 18, 1796–1799. [Google Scholar] [CrossRef] [PubMed]
  143. Yusibov, V.; Hooper, D.C.; Spitsin, S.V.; Fleyish, N.; Kean, R.B.; Mikheeva, T.; Deka, D.; Karasev, A.; Cox, S.; Randall, J.; et al. Expression in plants and immunogenecity of plant virus based experimental rabies vaccine. Vaccine 2002, 20, 3155–3164. [Google Scholar] [CrossRef]
  144. Tacket, C.O. Plant-based vaccines against diarrheal diseases. Trans. Am. Clin. Climatol. Assoc. 2007, 118, 79–87. [Google Scholar]
  145. Mason, H.S.; Warzecha, H.; Tsafir, M.S.; Arntzen, C.J. Edible plant vaccines: Applications for prophylactic and therapeutic molecular medicine. Trends Mol. Med. 2002, 8, 324–329. [Google Scholar] [CrossRef] [PubMed]
  146. Makarkov, A.I.; Chierzi, S.; Pillet, S.; Murai, K.K.; Landry, N.; Ward, B.J. Plant-made virus-like particles bearing influenza hemagglutinin (HA) recapitulate early interactions of native influenza virions with human monocytes/macrophages. Vaccine 2017, 35, 4629–4636. [Google Scholar] [CrossRef] [PubMed]
  147. Abha, K.; Shaila, M.S.; Lakshmi, S.G. Transgenic plants as a source of “edible vaccines” for two morililiviral animal diseases. In Proceedings of the Abstracts of 10th IAPTC&B Congress 72-A, Orlando, FL, USA, 23–28 June 2002. [Google Scholar]
  148. Dennis, S.J.; O’Kennedy, M.M.; Rutkowska, D.; Tsekoa, T.; Lourens, C.W.; Hitzeroth, I.I.; Meyers, A.E.; Rybicki, E.P. Safety and immunogenicity of plant-produced African horse sickness virus-like particles in horses. Vet. Res. 2018, 49, 1–6. [Google Scholar] [CrossRef] [Green Version]
  149. Yusibov, V.; Streatfield, S.J.; Kushnir, N. Clinical development of plant-produced recombinant pharmaceuticals: Vaccines, antibodies and beyond. Hum. Vaccin. 2011, 7, 313–321. [Google Scholar] [CrossRef]
  150. Watson, J.; Koya, V.; Leppla, S.H.; Daniell, H. Expression of Bacillus anthracis protective antigen in transgenic chloroplasts of tobacco, a non-food/feed crop. Vaccine 2004, 22, 4374–4384. [Google Scholar] [CrossRef] [Green Version]
  151. Lamphear, B.J.; Jilka, J.M.; Kesl, L.; Welter, M.; Howard, J.A.; Streatfield, S.J. A corn-based delivery system for animal vaccines: An oral transmissible gastroenteritis virus vaccine boosts lactogenic immunity in swine. Vaccine 2004, 22, 2420–2424. [Google Scholar] [CrossRef]
  152. Kim, T.G.; Gruber, A.; Langridge, W.H.R. HIV-1 gp120 V3 cholera toxin B subunit fusion gene expression in transgenic potato. Protein Expres. Purif. 2004, 37, 196–202. [Google Scholar] [CrossRef] [PubMed]
  153. Maloney, B.J.; Takeda, N.; Suzaki, Y.; Ami, Y.; Li, T.C.; Miyamura, T.; Arntzen, C.J.; Mason, H.S. Challenges in creating a vaccine to prevent hepatitis E. Vaccine 2005, 23, 1870–1874. [Google Scholar] [CrossRef] [PubMed]
  154. Gressel, J. Transgenics are imperative for biofuel crops. Plant Sci. 2008, 174, 246–263. [Google Scholar] [CrossRef]
  155. Smith, A.M. Prospects for increasing starch and sucrose yields for bioethanol production. Plant J. 2008, 54, 546–558. [Google Scholar] [CrossRef]
  156. Vanden Wymelenberg, A.; Sabat, G.; Mozuch, M.; Kersten, P.J.; Cullen, D.; Blanchette, R.A. Structure, organization, and transcriptional regulation of a family of copper radical oxidase genes in the lignin-degrading basidiomycete Phanerochaete chrysosporium. Appl. Environ. Microbiol. 2006, 72, 4871–4877. [Google Scholar] [CrossRef]
  157. Li, L.; Zhou, Y.; Cheng, X.; Sun, J.; Marita, J.; Ralph, J.; Chiang, V. Combinatorial modification of multiple lignin traits in trees through multigene co-transformation. Proc. Natl. Acad. Sci. USA 2003, 100, 4939–4944. [Google Scholar] [CrossRef] [Green Version]
  158. Myers, A.M.; Morell, M.K.; James, M.G.; Ball, S.G. Recent progress toward understanding the biosynthesis of the amylopectin crystal. Plant Physiol. 2000, 122, 989–998. [Google Scholar] [CrossRef] [Green Version]
  159. Ralph, J.; Akiyama, T.; Kim, H.; Lu, F.; Schatz, P.F.; Marita, J.M.; Ralph, S.A.; Reddy, M.S.S.; Chen, F.; Dixon, R.A. Effects of coumarate 3-hydroxylase down-regulation on lignin structure. J. Biol. Chem. 2006, 281, 8843–8853. [Google Scholar] [CrossRef] [Green Version]
  160. Chabannes, M.; Barakate, A.; Capierre, C.; Marita, J.M.; Ralph, J.; Peem, M.; Danoun, S.; Halpin, C.; Grima-Pettenati, J.; Boudet, A.M. Strong decrease in lignin content without significant alteration of plant development is induced by simultaneous down-regulation of cinnamoyl CoA reductase (CCR) and cinnamyl alcohol dehydrogenase (CAD) in tobacco plants. Plant J. 2001, 28, 257–270. [Google Scholar] [CrossRef]
  161. Blaschke, L.; Legrand, M.; Mai, C.; Polle, A. Lignification and structural biomass production in tobacco with suppressed caffeic/5-hydroxy ferulic acid-O-methyl transferase activity under ambient and elevated CO2 concentrations. Physiol. Plant 2004, 121, 75–83. [Google Scholar] [CrossRef]
  162. Lardizabal, K.; Effertz, R.; Levering, C.; Mai, J.; Pedroso, M.; Jury, T.; Aasen, E.; Gruys, K.; Bennett, K. Expression of Umbelopsis ramanniana DGAT2A in seed increases oil in soybean. Plant Physiol. 2008, 148, 9–96. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Wiberg, E.; Edwards, P.; Byrne, J.; Stymne, S.; Dehesh, K. The distribution of caprylate, caprate and laurate in lipids from developing and mature seeds of transgenic Brassica napus L. Planta 2000, 212, 33–40. [Google Scholar] [CrossRef] [PubMed]
  164. Van Camp, W. Yield enhancement genes: Seeds for growth. Curr. Opin. Biotechnol. 2005, 16, 147–153. [Google Scholar] [CrossRef] [PubMed]
  165. Jing, Z.P.; Gallardo, F.; Pascual, M.B.; Sampalo, R.; Romero, J.; Torres de Navarra, A.; Ca´novas, F.M. Improved growth in a field trial of transgenic hybrid poplar overexpressing glutamine synthetase. New Phytol. 2004, 164, 137–145. [Google Scholar] [CrossRef]
  166. Hammond, B.; Dudek, R.; Lemen, L.; Nemeth, M. Results of a 13 week safety assurance study with rats fed grain from glyphosate tolerant corn. Food Chem Toxicol. 2004, 42, 1003–1014. [Google Scholar] [CrossRef]
  167. Schroder, M.; Poulsen, M.; Wilcks, A.; Kroghsbo, S.; Miller, A.; Frenzel, T.; Danier, J.; Rychlik, M.; Emami, K.; Gatehouse, A. A 90-day safety study of genetically modified rice expressing Cry1Ab protein (Bacillus thuringiensis toxin) in Wistar rats. Food Chem. Toxicol. 2007, 45, 339–349. [Google Scholar] [CrossRef]
  168. Setamou, M.; Berna, J.S.; Legaspi, J.C.; Mirkov, T.E. Parasitism and location of sugarcane borer (Lepidoptera: Pyralidae) by Cotesia flavipes (Hymenoptera: Braconidae) on transgenic and conventional sugarcane. Environ. Entomol. 2002, 31, 1219–1225. [Google Scholar] [CrossRef] [Green Version]
  169. Moellenbeck, D.J.; Peters, M.L.; Bing, J.W.; Rouse, J.R.; Higgins, L.S.; Sims, L.; Nevshemal, T.; Marshall, L.; Ellis, R.T. Insecticidal proteins from Bacillus thuringiensis protect corn from corn rootworms. Nat. Biotechnol. 2001, 19, 668–672. [Google Scholar] [CrossRef]
  170. Hu, L.; Zhou, K.; Liu, Y.; Yang, S.; Zhang, J.; Gong, X.; Ma, F. Overexpression of MdMIPS1 enhances salt tolerance by improving osmosis, ion balance, and antioxidant activity in transgenic apple. Plant Sci. 2020, 301, 110654. [Google Scholar] [CrossRef]
  171. Prakash, D.; Verma, S.; Bhatia, R.; Tiwary, B.N. Risks and Precautions of Genetically Modified Organisms. ISRN Ecol. 2011, 2011, 369573. [Google Scholar] [CrossRef] [Green Version]
  172. Deepa, A. Genetically Modified Foods: Benefits and Risks; Massachusetts Medical Society: Boston, MA, USA, 2015. [Google Scholar]
  173. Pelletier, D.L. Science, law, and politics in FDA’s genetically engineered foods policy: Scientific concerns and uncertainties. Nutr. Rev. 2005, 63, 210–223. [Google Scholar] [CrossRef] [PubMed]
  174. USDA. Biotechnology Frequently Asked Questions; USDA: Washington, DC, USA, 1998.
  175. Yul, R.J.; Choi, J.Y.; Li, M.S.; Jin, B.R.; Je, Y.H. Bacillus thuringiensis as a Specific, Safe, and Effective Tool for Insect Pest Control. J. Microbiol. Biotechnol. 2007, 17, 547–559. [Google Scholar]
  176. Hall, J.; Matos, M.; Langford, C.H. Social exclusion and transgenic technology: The case of Brazilian agriculture. J. Bus. Ethics. 2008, 77, 45–63. [Google Scholar] [CrossRef]
  177. Hilbeck, A.; Baumgartner, M.; Fried, P.M.; Bigler, F. Effects of transgenic Bacillus thuringiensis corn-fed prey on mor- tality and development time of immature Chrysoperla carnea (Neuroptera: Chrysopidae). Environ. Entomol. 1998, 27, 480–487. [Google Scholar] [CrossRef]
  178. Arnaud, J.F.; Viard, F.; Delescluse, M.; Cuguen, J. Evidence for gene flow via seed dispersal from crop to wild relatives in Beta vulgaris (Chenopodiaceae): Consequences for the release of genetically modified crop species with weedy lineages. Proc. R. Soc. B Biol. Sci. 2003, 270, 1565–1575. [Google Scholar] [CrossRef] [Green Version]
  179. Ellstrand, N.C.; Scierenbeck, K.A. Hybridization as a stimulus for the evolution of invasiveness in plants? Proc. Natl. Acad. Sci. USA 2000, 97, 7043–7050. [Google Scholar] [CrossRef] [PubMed]
  180. Gressel, J. Molecular biology of weed control. Transgenic Res. 2000, 9, 355–382. [Google Scholar] [CrossRef] [PubMed]
  181. Ricroch, A.E.; Berge, J.E.; Kuntz, M. Evaluation of genetically engineered crops using transcriptomic, proteomic, and metabolomic profiling techniques. Plant Physiol. 2011, 155, 1752–1761. [Google Scholar] [CrossRef] [Green Version]
  182. Keese, P. Risks from GMOs due to Horizontal Gene Transfer. Environ. Biosaf. Res. 2008, 7, 123–149. [Google Scholar] [CrossRef] [Green Version]
  183. Dutton, A.; Klein, H.; Romeis, J.; Bigler, F. Uptake of Bt-toxin by herbivores feeding on transgenic maize and consequences for the predator Chrysoperia carnea. Ecol. Entomol. 2002, 27, 441–447. [Google Scholar] [CrossRef]
  184. Tudisco, R.; Lombardi, P.; Bovera, F.; Cutrignelli, M.I.; Mastellone, V.; Terzi, V.; Avallone, L.; Infascelli, F. Genetically Modified Soya Bean in Rabbit Feeding: Detection of DNA fragments and evaluation of metabolic effects by enzymatic analysis. J. Anim. Sci. 2006, 82, 193–199. [Google Scholar] [CrossRef] [Green Version]
  185. Romeis, J.; Dutton Bigler, F. Bacillus thuringiences toxin (Cry 1 Ab) has no direct on larvae of the green lacewing Chrysoperla carnea (stephens) (Neuroptera: Chrysopidae). J. Insect. Physiol. 2004, 50, 175–183. [Google Scholar] [CrossRef] [PubMed]
  186. Bob, B.M.W.; Candolin, U. Behavioral responses to changing environments. Behav. Ecol. 2015, 26, 665–673. [Google Scholar]
  187. Chekol, C. The Health Effects of Genetically Modified Foods: A Brief Review. Int. J. Nutr. Sci. 2021, 6, 1047. [Google Scholar] [CrossRef]
  188. Paparini, A.; Romano-Spica, V. Public health issues related with the consumption of food obtained from genetically modified organisms. Biotechnol. Annu. Rev. 2004, 10, 85–122. [Google Scholar] [PubMed]
  189. Alexandrova, N.; Georgieva, K.; Atanassov, A. Biosafety regulations of GMOs: National and International aspects and regional cooperation. Biotechnol. Biotechnol. Equip. 2005, 19, 153–172. [Google Scholar] [CrossRef]
  190. de Vendômois, J.S.; Roullier, F.; Cellier, D.; Séralini, G.E. A Comparison of the effects of three GM corn varieties on mammalian health. Int. J. Biol. Sci. 2009, 5, 706–726. [Google Scholar] [CrossRef]
  191. Wozniak, E.; Tyczewska, A.; Twardowski, T. Bioeconomy development factors in the European Union and Poland. New Biotechnol. 2021, 60, 2–8. [Google Scholar] [CrossRef]
  192. Cribbs, A.P.; Perera, S.M. Focus: Genome editing: Science and bioethics of CRISPR-Cas9 gene editing: An analysis towards separating facts and fiction. Yale J. Biol. Med. 2017, 90, 625. [Google Scholar]
  193. Vibha Ahuja Regulation of emerging gene technologies in India. BMC Proc. 2018, 12, 14.
  194. MoEFCC; DBT. Guidelines, User’s Guide and Risk Analysis Framework. In Environmental Risk Assessment of GE Plants; MoEFCC: New Delhi, India, 2016. [Google Scholar]
  195. MoEFCC; BCIL. Frequently Asked Questions About GE Plants. In Phase II Capacity Building Project on Biosafety; MoEFCC: New Delhi, India, 2015. [Google Scholar]
  196. Ishii, T.; Araki, M. A future scenario of the global regulatory landscape regarding genome-edited crops. GM Crop. Food 2017, 8, 44–56. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  197. Rosado, A.; Craig, W. Biosafety regulatory systems overseeing the use of genetically modified organisms in the latin america and caribbean region. AgBioForum 2017, 20, 120–132. [Google Scholar]
  198. Global Agriculture. Venezuela passes new seed law banning genetically modified crops. 2016. Available online: https://www.globalagriculture.org/whatsnew/news/en/31519.html. (accessed on 11 January 2023).
  199. APBREBES. The New Seed Law of Venezuela; APBREBES: Geneva, Switzerland, 2016. [Google Scholar]
  200. Gatica-Arias, A. The regulatory current status of plant breeding technologies in some Latin American and the Caribbean countries. Plant Cell Tissue Organ Cult. 2020, 141, 229–242. [Google Scholar] [CrossRef]
  201. Smith, P.; Katovich, E. Are GMO policies “trade related”? Empirical analysis of Latin America. Appl. Econ. Perspect. Policy 2017, 39, 286–312. [Google Scholar] [CrossRef]
  202. ISAAA. Brief 55–2019. Executive Summary, Biotech Crops Drive Socio-Economic Development and Sustainable Development in the New Frontier; ISAAA: Nairobi, Kenya, 2019. [Google Scholar]
  203. Turnbull, C.; Lillemo, M.; Hvoslef-Eide, T. Global regulation of genetically modified crops amid the gene edited crop boom—A review. Front. Plant Sci. Plant Biotechnol. 2021, 12, 1–19. [Google Scholar] [CrossRef]
  204. Zarrilli, S. International trade in gmos and gm products: National and multilateral legal frameworks. In Policy Issues in International Trade and Commodities Study Series no. 29; United Nations: New York, NY, USA, 2005; pp. 1–16. [Google Scholar]
  205. Rosado, A.; Eriksson, D. Biosafety legislation and the regulatory status of the products of precision breeding in the Latin America and the Caribbean region. Plants People Planet 2021, 4, 214–231. [Google Scholar] [CrossRef]
  206. Akaakohol, M.A.; Aye, G.C. Diversification and farm household welfare in Makurdi, Benue state, Nigeria. Dev. Stud. Res. 2014, 1, 168–175. [Google Scholar] [CrossRef]
  207. ASSAf (Academies of Science of South Africa). GMOs for African agriculture: Challenges and opportunities. Workshop Proceedings Report. ASSAf: 2010. Available online: www.assaf.org.za (accessed on 11 January 2023).
  208. EFSA (European Food Safety Authority). Guidance on the environmental risk assessment of genetically modified plants. EFSA J. 2010, 8, 1879. [Google Scholar] [CrossRef]
  209. Bawa, A.S.; Anilakumar, K.R. Genetically modified foods: Safety, risks and public concerns-a review. J. Food Sci. Technol. 2013, 50, 1035–1046. [Google Scholar] [CrossRef] [Green Version]
  210. Tung, O.J.L. A comparative analysis of the South African and burkinabe experiences with genetically modified crop regulation. In Verfassung und Recht in Übersee/Law and Politics in Africa, Asia, Latin America; Nomos Verlagsgesellschaft: Baden-Baden, Germany, 2017; pp. 3–29. [Google Scholar]
  211. Anderson, K.; Nielsen, C. Golden Rice and The Looming Gmo Trade Debate: Implications for the Poor; CEPR. Discussion Papers 4195; CEPR: London, UK, 2004. [Google Scholar]
  212. Deffor, E.W. Consumer acceptance of genetically modified foods in the greater accra region of Ghana. J. Biosaf. Health Educ. 2014, 2, 116. [Google Scholar] [CrossRef]
  213. Newell-McGloughlin, M.; Burke, J. Biotechnology crop adoption: Potential and challenges of genetically improved crops. Encycl. Agric. Food Syst. 2014, 2014, 69–93. [Google Scholar]
  214. Akinbo, O.; Obukosia, S.; Ouedraogo, J.; Sinebo, W.; Savadogo, M.; Timpo, S.; Mbabazi, R.; Maredia, K.; Diran Makinde, D.; Ambali, A. Commercial release of genetically modified crops in Africa: Interface between biosafety regulatory systems and varietal release systems. Front. Plant Sci. 2021, 12, 605937. [Google Scholar] [CrossRef] [PubMed]
  215. Cooke, J.G.; Downie, R. African Perspectives on Genetically Modified Crops: Assessing the Debate in Zambia, Kenya and South Africa. A Report of the Csis Global Food Security Project; Centre for Strategic & International Studies: Washington, DC, USA, 2010. [Google Scholar]
  216. ISAAA. Global Status of Commercialised Biotech/GM Crops in ISAAA Brief Ithaca; ISAAA: New York, NY, USA, 2019. [Google Scholar]
  217. ABNE. African biosafety network of expertise (ABNE) african union development agency NEPAD (AUDA-NEPAD); ABNE: Midrand, South Africa, 2019. [Google Scholar]
  218. ISAAA. Global Status of Commercialized Biotech/GM Crops in 2018: Executive Brief; ISAAA: Ithaca, NY, USA, 2018. [Google Scholar]
  219. Kimenju, S.C.; De Groote, H.; Karugia, J.; Mbogoh, S.; Poland, D. Consumer awareness and attitudes toward GM foods in Kenya. Afr. J. Biotechnol. 2005, 4, 1–10. [Google Scholar]
  220. Nyinondi, P.S.; Dulle, F.W.; Nawe, J. Perception of agricultural biotechnology among farmers. Journalists and Scientists in Tanzania. Univ. Dares Salaam Libr. J. 2017, 12, 106–120. [Google Scholar]
  221. ISAAA. Global Status of Commercialised Biotech/Gm Crops in 2017: Biotech Crop Adoption Surges as Economic Benefits Accumulate in 22 Years; ISAAA Brief: New York, NY, USA, 2017; Volume 53, pp. 25–26. [Google Scholar]
  222. ISAAA. Global Status of Commercialised Biotech/GM Crops in ISAAA Brief no. 54; ISAAA: Ithaca, NY, USA, 2018. [Google Scholar]
  223. Aerni, P. Stakeholder attitudes towards the risks and benefits of genetically modified crops in South Africa. Environ. Sci. Policy 2005, 8, 464–476. [Google Scholar] [CrossRef]
  224. Aerni, P.; Bernauer, T. Stakeholder attitudes toward GMOs in the Philippines, Mexico, and South Africa: The issue of public trust. World Dev. 2006, 34, 557–575. [Google Scholar] [CrossRef]
  225. Marques, M.D.; Critchley, C.R.; Walshe, J. Attitudes to genetically modified food over time: How trust is, and the media cycle predict support. Public Underst. Sci. 2015, 24, 601–618. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  226. Rock, J.; Schurman, R. The complex choreography of agricultural biotechnology in Africa. Afr. Affairs 2020, 119, 499–525. [Google Scholar] [CrossRef]
  227. MOA (Ministry of Agriculture). Measures of Labelling Administration of Agricultural GMO–Decree 10 (in Chinese), 2002. Available online: http://www.moa.gov.cn/fwllm/zxbs/xzxk/bszl/201405/t20140527_3917464.htm (accessed on 11 January 2023).
  228. MOA (Ministry of Agriculture of China). Guideline for Safety Inspection of Field Trials of GM Crops, 2012. Available online: http://www.moa.gov.cn/zwllm/zcfg/qtbmgz/200606/t20060612_627852.htm (accessed on 22 August 2015).
  229. Xiao, Z.; Kerr, W.A. Biotechnology in China–regulation, investment, and delayed commercialization. GM CROPS FOOD 2022, 13, 86–96. [Google Scholar] [CrossRef] [PubMed]
  230. Loppacher, L.J.; Kerr, W.A. Biotechnology in China: Food policy and international trade issues. In Food Policy Control and Research; Riley, A.P., Ed.; Nova Science Publishers: New York, NY, USA, 2005; pp. 1–15. [Google Scholar]
  231. Kim, H.Y.; Kim, J.H.; Mi-Hwa Oh, M.H. Regulation and detection methods for genetically modified foods in Korea. Pure Appl. Chem. 2010, 82, 129–137. [Google Scholar] [CrossRef]
  232. Godfray, H.C.J.; Beddington, J.R.; Crute, I.R.; Haddad, L.; Lawrence, D.; Muir, J.F.; Pretty, J.; Robinson, S.; Thomas, S.M.; Toulmin, C. Food security: The challenge of feeding 9 billion people. Science 2010, 327, 812–818. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  233. ILSI. Nutritional and Safety Assessments of Foods And Feeds Nutritionally Improved through Biotechnology: Case Studies. Available online: http://www.ilsi.org/foodbiotech/publications/10_ilsi2008_casestudies_crfsfs.pdf (accessed on 11 January 2023).
  234. Shahzadi, F.; Malik, M.F.; Ali, R. Genetically Modified Food Controversies: A Review. Int. J. Sci. Eng. Res. 2015, 6, 2072–2089. [Google Scholar]
  235. De Vos, C.J.; Swanenburg, M. Health effects of feeding genetically modified (GM) crops to livestock animals: A review. Food Chem. Toxicol. 2018, 117, 3–12. [Google Scholar] [CrossRef] [PubMed]
  236. Clark, J.H.; Ipharraguerre, I.R. Livestock performance: Feeding biotech crops. J. Dairy Sci. 2001, 1, E9–E18. [Google Scholar] [CrossRef]
  237. Rizzi, A.; Raddadi, N.; Sorlini, C.; Nordgrd, L.; Nielsen, K.M.; Daffonchio, D. The stability and degradation of dietary DNA in the gastrointestinal tract of mammals: Implications for horizontal gene transfer and the biosafety of GMOs. Crit. Rev. Food Sci. Nutr. 2012, 52, 142–161. [Google Scholar] [CrossRef]
  238. Nielsen, K.M.; Bones, A.M.; Smalla, K.; van Elsas, J.D. Horizontal gene transfer from transgenic plants to terrestrial bacteria − a rare event? FEMS Microbiol. Rev. 1998, 22, 79–103. [Google Scholar] [CrossRef]
  239. Breckling, B.; Reuter, H.; Middelhoff, U.; Glemnitz, M.; Wurbs, A.; Schmidt, G.; Windhorst, W. Risk indication of genetically modified organisms (GMO): Modelling environmental exposure and dispersal across different scales: Oilseed rape in Northern Germany as an integrated case study. Ecol. Indic. 2011, 11, 936–941. [Google Scholar] [CrossRef]
  240. Beckwith, M.; Hadlock, T.; Suffron, H. Public perceptions of plant biotechnology—A focus group study. New Genet Soc. 2003, 22, 93–109. [Google Scholar] [CrossRef]
  241. Lundquist, K.A. Unapproved genetically modified corn: It’s what’s for dinner. Iowa Law Rev. 2015, 100, 825–851. [Google Scholar]
  242. Mikkelsen, T.R.; Andersen, B.; Jorgensen, R.B. The risk of crop transgene spread. Nature 1996, 380, 31. [Google Scholar] [CrossRef]
  243. Snow, A.A. Illegal gene flow from transgenic creeping bentgrass: The saga continues. Mol. Ecol. 2012, 21, 4663–4664. [Google Scholar] [CrossRef] [PubMed]
  244. Pineyro-Nelson, A.; Van Heerwaarden, J.; Perales, H.; Serratos-Hernández, J.A.; Rangel, A.; Hufford, M.B.; Gepts, P.; Garay-Arroyo, A.; Rivera-Bustamante, R.; Alvarez-Buylla, E.R. Resolution of the Mexican transgene detection controversy: Error sources and scientific practice in commercial and ecological contexts. Mol. Ecol. 2009, 18, 4145–4150. [Google Scholar] [CrossRef]
  245. Wegier, A.; Pineyro-Nelson, A.; Alarcon, J.; Galvez-Mariscal, A.; Varez-Buylla, E.R.; Pinero, D. Recent long-distance transgene flow into wild populations conforms to historical patterns of gene flow in cotton (Gossypium hirsutum) at its centre of origin. Mol. Ecol. 2011, 20, 4182–4194. [Google Scholar] [CrossRef]
  246. Acevedo, F.; Huerta., E.; Burgeff, C.; Koleff, P.; Sarukhan, J. Is transgenic maize what Mexico really needs? Nat. Biotechnol. 2011, 29, 23–24. [Google Scholar] [CrossRef] [PubMed]
  247. Samuels, J. Transgene flow from Bt brinjal: A real risk? Trends Biotechnol 2013, 31, 332–334. [Google Scholar] [CrossRef] [PubMed]
  248. Qaim, M. Benefits of genetically modified crops for the poor: Household income, nutrition, and health. New Biotechnol. 2010, 27, 552–557. [Google Scholar] [CrossRef] [PubMed]
  249. Fraser, L. The terminator. Altern. J. 2006, 32, 24. [Google Scholar]
  250. Maghari, M.B.; Ardekani, M.A. Genetically Modified Foods and Social Concerns. Avicenna J. Med. Biotech. 2011, 3, 109–117. [Google Scholar]
  251. Tironi, M.; Salazar, M.; Valenzuela, D. Resisting and accepting: Farmers’ hybrid epistemologies in the GMO controversy in Chile. Technol. Soc. 2013, 35, 93–104. [Google Scholar] [CrossRef]
  252. Kangmennaanga, J.; Oseia, L.; Frederick, A.; Armah, F.A. Genetically modified organisms and the age of (Un) reason? A critical examination of the rhetoric in the GMO public policy debates in Ghana. Futures 2016, 83, 37–49. [Google Scholar] [CrossRef]
  253. Soleri, D.; Cleveland, D.; Cuevas, F. Transgenic crops and crop varietal diversity: The case of maize in Mexico. BioScience 2006, 56, 503–513. [Google Scholar] [CrossRef]
  254. Rzymski, P.; Królczyk, A. Attitudes toward genetically modified organisms in Poland: To GMO or not to GMO? Food Sec. 2016, 8, 689–697. [Google Scholar] [CrossRef] [Green Version]
  255. Liu, L.; Cao, C. Who owns the intellectual property rights to Chinese genetically modified rice? Evidence from patent portfolio analysis. Biotechnol. Law Rep. 2014, 33, 181–192. [Google Scholar] [CrossRef] [Green Version]
  256. Wong, A.Y.T.; Chan, A.W.K. Genetically modified foods in China and the United States: A primer of regulation and intellectual property protect. Food Sci. Hum. Wellness 2016, 5, 124–140. [Google Scholar] [CrossRef] [Green Version]
  257. Rodriguez-Ferrand, G. Restrictions on Genetically Modified Organisms, The Law Library of Congress, Global Legal Research Center. 2014. Available online: http://www.loc.gov/law/help/restrictions-on-gmos/ (accessed on 11 January 2023).
  258. Cui, K.; Shoemaker, S.P. Public perception of genetically-modified (GM) food: A Nationwide Chinese Consumer Study. Npj Sci. Food. 2018, 2, 1–8. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Agrobacterium-mediated genetic transformation of the plant. The schematic diagram shows the steps associated with the cloning of the gene of interest in the Ti-plasmid of Agrobacterium tumefaciens and its transfer to plant cells in culture to regenerate the transgenic plants with desirable traits.
Figure 1. Agrobacterium-mediated genetic transformation of the plant. The schematic diagram shows the steps associated with the cloning of the gene of interest in the Ti-plasmid of Agrobacterium tumefaciens and its transfer to plant cells in culture to regenerate the transgenic plants with desirable traits.
Sustainability 15 01722 g001
Figure 2. Biolistics method of genetic transformation of the plant. The schematic diagram shows the bombardment of gold particles containing the gene of interest onto the plant cells in culture to regenerate the transgenic plants with desirable traits.
Figure 2. Biolistics method of genetic transformation of the plant. The schematic diagram shows the bombardment of gold particles containing the gene of interest onto the plant cells in culture to regenerate the transgenic plants with desirable traits.
Sustainability 15 01722 g002
Figure 3. Electroporation method of genetic transformation of the plant. The schematic diagram shows the steps involved during the electroporation that lead to the insertion of the exogenous gene of interest into the plant cell.
Figure 3. Electroporation method of genetic transformation of the plant. The schematic diagram shows the steps involved during the electroporation that lead to the insertion of the exogenous gene of interest into the plant cell.
Sustainability 15 01722 g003
Table 2. Genetic transformation strategies and genes used for the improvement of biotic and abiotic stress resistance in crops.
Table 2. Genetic transformation strategies and genes used for the improvement of biotic and abiotic stress resistance in crops.
Scientific Name Plant PartsA. tumefaciens Strains/VectorGeneBiotic and Abiotic ResistanceReferences
Medicago sativaLeaves and petioleAgrobacterium. tumefaciens LBA4404/ AGL01/s GV101cry3a (Bt Toxin) Insect resistanceTohidfar et al. [92]
Oryza sativa L.SeedParticle bombardmentItr1geneInsect resistanceAlfanso-Rubi et al. [93]
Glycine max L.Somatic embryoMicro projectile bombardmentViral coat proteinSoybean dwarf virus resistanceTougou et al. [94]
Jatropha curcas L.LeavesAgrobacterium tumefaciens EHA 105 strainChitinaseDisease resistanceFranco et al. [95]
Glycine max L.LeavesAgrobacterium tumefacienscry1A gene (tic107) Insect resistanceMacrae et al. [96]
Gossypium hirsutum var CokerSeedAgrobacterium tumefaciens (LBA 4404)/pBI121cry1Ab geneInsect resistance Tohidfar et al. [97]
BrinjalLeavesAgrobacterium tumefaciens LBA4404/pBI121cystatin geneHigher rate of inhibition of root-knot nematode in transgenic plantPapolu et al. [98]
Kiwi fruitsLeavesAgrobacterium tumefaciens LBA4404/pBin513sbtCryIAcgeneResistance against Oraesia excavateZhang et al. [99]
Camelina sativa L.Floral partsAgrobacterium rhizogenes (pB172)/plasmid pKYLX71.1acdS: ACC deaminaseSalinity tolerance Heydarian et al. [100]
Arabidopsis thaliana L.SeedlingsAgrobacterium tumefaciens GV3101/pBI121 expression vectorTranscription factor JcCBF2Freezing tolerance Wang et al. [101]
Camelina sativa L.Flower, stem, leaf, and rootAgrobacterium tumefaciens/pCB302-3 vectorsCsHMA3Heavy metals tolerancePark et al. [102]
Table 3. Genetic transformation strategies and genes used for increasing phytoremediation efficiency in crops.
Table 3. Genetic transformation strategies and genes used for increasing phytoremediation efficiency in crops.
PlantGeneA. tumefaciens Strains/VectorProductActivityReferences
Arabidopsis thaliana L. and PoplarPtABCC1A. tumefaciens GV3101/pCX-SNABC transporterHg toleranceSun et al. [110]
Arabidopsis thaliana L.TpNRAMP5pMD19-T, HBT95-GFP, pCAMBIA1305.1,Numerous natural resistance-associated macrophage proteinsIncreased accumulation of Cd, Co, and MnPeng et al. [86]
Arabidopsis thaliana L.CsMTP9pENTR/D-TOPO vector into pMDC43 or pMDC83Metal transport protein 9Increased accumulation of Mn and CdMigocka et al. [121]
TobaccoOsMTP1E. coli, DH10B (GIBCO BRLp/UC18)Metal transport protein 1Cd hyperaccumulationDas et al. [122]
Salix matsudanaThMT3A. tumefaciens LBA4404/PROKII-ThMT3MetallothioneinIncreased Cu tolerance and root growthYang et al. [123]
TobaccoAtPCS1A. tumefaciens LBA4404/pBI121 and pCAMBIAPhytochelatin synthaseCd and As accumulationZanella et al. [124]
PetuniaRsMYB1A. tumefaciens C58C1/pB7WG2DTranscription factorEnhanced tolerant to Cd,, Cu, ZnAi et al. [125]
Arabidopsis thaliana L. ZAT6A. tumefaciens GV3101/pXB93Zinc-finger transcription factorEnhanced Cd toleranceChen et al. [126]
Beta vulgarisSt GCS-GSA. tumefaciens EHA105/pGWB2StGCS-GSIncreased Cd, Zn, Cu toleranceLiu et al. [127]
RiceTaPCS1A. tumefaciens EHA105/pBI121Phytochelatin synthase, non-protein thiolsCd hypersensitivityWang et al. [128]
Arabidopsis thaliana L.AtABCC3A. tumefaciens GV3101/pER8PhytochelatinIncreased Cd toleranceBrunetti et al. [129]
Brassica napusBnNRAMP1bycf1 (Y04069), zrc1 (Y00829), smf1 (Y06272), BY4741/pYES2Transport functionsEnhanced uptake of Cd, Zn, MnMeng et al. [130]
Indian mustardgshI, gshII and APS1pFF19γ-Glu-Cys synthetase, glutathione Synthetase, and ATP sulfurylaseEnhanced Se,Banuelos et al. [131]
Arabidopsis thaliana L.OASTdA. tumefaciens CV50/pBI121Cysteine synthaseTolerance to CdDominguez-Solis et al. [132]
Arabidopsis thalianaBnPCSA. tumefaciens CV50/pBI121PhytochelatinTolerance to CdBai et al. [133]
Brassica napusCKX2A. tumefaciens GV3101Cytokinin contentTolerance to Cd, ZnNehnevajova et al. [134]
Table 4. Representative transgenic plant-based vaccines.
Table 4. Representative transgenic plant-based vaccines.
PlantsAntigen/VirusDiseasesMethod of AdministrationReference
Transgenic potatoesHepatitis B surface antigen (HBsAg)Hepatitis BOralRichter et al. [142]
N. tabacum cv. SamsunVirus glycoprotein and nucleoprotein fused with A1Mvcoat proteinRabiesParenteralYusibov et al. [143]
Potato, Maize kernels
Potato
E. coli LT-BDiarrheaOralTacket et al. [144]
PotatoNorwalk virus like particles (rNV)Diarrhea, nauseaOralMason et al. [145]
N. benthamianaD antigen (PV3)/PolioviruspolioIntraperitoneal injectionsMarsian, et al. [141]
N. benthamianaH1, H5/Influenza virusInfluenzaNAMakarkov et al. [146]
Peanut and tobaccoGlycoproteins hemaglutinin (H), Hemaglutinin neuraminidase (HS)“cattle plague” and “Goat plague”NAAbha Khandelwal et al. [147]
N. benthamianaVP2,VP3,VP5,VP7/African horse sickness virus (AHSV)African horseIntramuscular Dennis et al. [148]
N. benthamianainfluenza HAC1H1N1 “swine” influenzaIntramuscularYusibov et al. [149]
N. benthamianaProtective antigen (PA)AnthraxSubcutaneousWatson et al. [150]
MaizeSpike proteinSwine transmissible gastroenteritis virusOralLamphear et al. [151]
PotatoCTB-gpl20 (HIV-1 gp 120V3 cholera toxin B subunit fusion gene)Cholera Kim et al. [152]
PotatoHEV CP (HEV capsid proteins)Hepatitis EOralMaloney et al. [153]
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

Ghimire, B.K.; Yu, C.Y.; Kim, W.-R.; Moon, H.-S.; Lee, J.; Kim, S.H.; Chung, I.M. Assessment of Benefits and Risk of Genetically Modified Plants and Products: Current Controversies and Perspective. Sustainability 2023, 15, 1722. https://doi.org/10.3390/su15021722

AMA Style

Ghimire BK, Yu CY, Kim W-R, Moon H-S, Lee J, Kim SH, Chung IM. Assessment of Benefits and Risk of Genetically Modified Plants and Products: Current Controversies and Perspective. Sustainability. 2023; 15(2):1722. https://doi.org/10.3390/su15021722

Chicago/Turabian Style

Ghimire, Bimal Kumar, Chang Yeon Yu, Won-Ryeol Kim, Hee-Sung Moon, Joohyun Lee, Seung Hyun Kim, and Ill Min Chung. 2023. "Assessment of Benefits and Risk of Genetically Modified Plants and Products: Current Controversies and Perspective" Sustainability 15, no. 2: 1722. https://doi.org/10.3390/su15021722

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