Next Article in Journal
Comparative Chloroplast Genomics of Fritillaria (Liliaceae), Inferences for Phylogenetic Relationships between Fritillaria and Lilium and Plastome Evolution
Next Article in Special Issue
Biosynthesis and Industrial Production of Androsteroids
Previous Article in Journal
Physiological, Biochemical and Reproductive Studies on Valeriana wallichii, a Critically Endangered Medicinal Plant of the Himalayan Region Grown under In-Situ and Ex-Situ Conditions
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Transgenesis as a Tool for the Efficient Production of Selected Secondary Metabolites from Plant in Vitro Cultures

1
Department of Molecular Biotechnology and Genetics, University of Lodz, Banacha 12/16, 90-237 Lodz, Poland
2
Department of Immunopathology, Medical University of Lodz, Żeligowskiego 7/9, 90-752 Lodz, Poland
3
Department of Biology and Pharmaceutical Botany, Medical University of Lodz, Muszynskiego 1, 90-151 Lodz, Poland
4
Laboratory of Medical Genetics, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland
*
Author to whom correspondence should be addressed.
Plants 2020, 9(2), 132; https://doi.org/10.3390/plants9020132
Submission received: 12 December 2019 / Revised: 18 January 2020 / Accepted: 19 January 2020 / Published: 21 January 2020
(This article belongs to the Special Issue Biotechnological Approaches for the Production of Bioactives)

Abstract

:
The plant kingdom abounds in countless species with potential medical uses. Many of them contain valuable secondary metabolites belonging to different classes and demonstrating anticancer, anti-inflammatory, antioxidant, antimicrobial or antidiabetic properties. Many of these metabolites, e.g., paclitaxel, vinblastine, betulinic acid, chlorogenic acid or ferrulic acid, have potential applications in medicine. Additionally, these compounds have many therapeutic and health-promoting properties. The growing demand for these plant secondary metabolites forces the use of new green biotechnology tools to create new, more productive in vitro transgenic plant cultures. These procedures have yielded many promising results, and transgenic cultures have been found to be safe, efficient and cost-effective sources of valuable secondary metabolites for medicine and industry. This review focuses on the use of various in vitro plant culture systems for the production of secondary metabolites.

Graphical Abstract

1. Introduction

Throughout history, in his struggle with disease, man has continually enlisted help from plants, one of the richer sources of biologically-active health-promoting substances. The oldest evidence of such use was found on a Sumerian clay slab from Nagpur, which is approximately 5000 years old [1]. However, the use of plants in treating ailments is also mentioned in the Pen T’Sao, written by the Emperor of China, Shen Nung, in 2500 BC, the Indian Vedas, the Ebers Papyrus written in 1550 BC, the Bible and the Talmud [2,3]. Almost all medical systems, be they Traditional Medicine, Kampo medicine, Ayurvedic medicine or European medicine, are based on plant-derived valuable medical compounds [4]. Currently, medicine is faced with a growing demand for a wide range of biologically-active compounds of natural origin that can demonstrate preventive or therapeutic effects against primary causes of death, such as cancer, cardiovascular disease, diabetes or respiratory disease. A large proportion of the phytochemicals used for this purpose are secondary metabolites. These compounds are structurally and functionally-diverse products synthesized in plant cells from various primary metabolites, either constitutively or in response to various stimuli. In nature, they have many different functions such as protecting plants from pathogens [5,6], ultraviolet light [7,8] or herbivores [9,10], and providing specific fragrances and colors to facilitate pollination and seed distribution by animals. They also play an important role as signaling and regulatory molecules for primary metabolic pathways.
In modern medicine, plant secondary metabolites play a vital role. Since their first isolation in 1803 and the introduction of the alkaloid morphine in 1827 [11], studies have expanded knowledge of their structure, biological function, biosynthesis pathway and possible modifications, thanks to which they can be used to protect human health. Many of their mechanisms of action on various types of normal and cancerous human cells have been described [12,13,14]. Secondary metabolites can affect cancer cells by interfering with their division, and by changing their metabolism and even the expression of selected genes. Many also have antioxidant [15,16], anti-inflammatory [17,18], antibacterial [19], antifungal [20,21], neurological [22] or hepatoprotective [23] effects. As plants represent such an important source of many secondary metabolites, there is great interest in increasing their biosynthetic rates as part of green biotechnology, which includes the use of transgenic plants or other photosynthetic organisms for industrial purposes. Such approaches allow the production of a wide range of products including secondary metabolites, recombinant proteins, biologically-active compounds, paper and biofuels. They can also be used to improve the nutritional quality of a plant and the development of environmentally-friendly farming solutions.
One of the key strategies enabling the overproduction of valuable plant secondary metabolites in in vitro cultures is the manipulation of existing metabolic pathways by overexpressing or silencing selected elements involved in their biosynthesis [24,25,26,27]. A wide range of plant vectors have been designed that allow the simple and quick introduction of synthetic expression cassettes, allowing easier and more effective creation of in vitro transgenic plant cultures [28,29,30]. Several studies confirm that such modulation of metabolic pathways increases the yield of naturally-occurring biologically-active compounds with potential use in medicine [31,32,33].
The aim of this work is to review the latest achievements in the transgenesis-based biosynthesis of selected secondary metabolites, particularly anticancer compounds, produced in various types of in vitro plant cultures.

2. In Vitro Plant Cells and Organ Cultures as an Alternative Source of Secondary Metabolites

Since the development of the concept of in vitro cell culture in 1902 by the German physiologist Gottlieb Haberlandt, it has been possible to cultivate cells, organs or whole plants of many species under strictly controlled conditions; such approaches employ various biotechnological methods to exploit the totipotency (natural ability to divide and produce differentiated cells) of plant tissue. At present, in vitro plant cultures have been used for agricultural purposes by clonal multiplication of plants [34] and the introduction of novel genetic variations and transgenic plants. They have also found use in pharmaceutical biotechnology, through the production of recombinant proteins, monoclonal antibodies and secondary metabolites, and environmental biotechnology, by developing new methods of eliminating ecological threats and phytoremediation. Plant cell and tissue culture offers many advantages, including the ability to maintain the cells under controlled conditions regardless of changes in environmental conditions, simple extraction of biologically-active compounds, efficient production of many valuable compounds, the ability to easily scale the production process, and faster reproduction of high-quality plant material, especially in the case of slow-growing plants or those producing a small number of seeds. Additionally, this strategy allows pathogen-free material with high levels of the desired biologically-active compounds to be produced. Typically, calli, cell suspension cultures, roots or shoots are most commonly used for obtaining secondary metabolites in vitro. In selected cases, it is necessary to induce cell differentiation in cultures because some metabolites can be synthesized only in specialized plant tissues or glands (e.g., essential oils).
Each of the aforementioned types of in vitro plant cultures has certain characteristic features making it suitable for a chosen group of compounds. All types of cultures have their advantages and disadvantages, and care should be taken to select an appropriate type of plant culture to obtain valuable secondary metabolites in the first stage.

3. From Natural Gene Transfer to Plant Metabolic Engineering

Over 100 years have passed since the first isolation of Agrobacterium tumefaciens, which later turned out to be a natural genetic engineer of the plant genome [35]. A. tumefaciens is a phytopathogen found in an environment causing neoplastic diseases (crown gall) on various plant species. The bacterium has a natural ability to infect wound sites in plants, resulting in the formation of characteristic tumors [36] by the transfer of T-DNA from the bacterial cell to the plant genome through a bacterial type IV secretion system (T4SS) [37]. The first use of A. tumefaciens infection to obtain transgenic plants was in 1983 [38], which predicted the advent of a new age in plant biotechnology. As the mechanism of transferring genetic material to the plant cell became better understood, such transfection by Agrobacterium has become the most popular method of genetic modification of plants. During transformation, T-DNA is exported from the bacterial cells to the nucleus of the plant cell, where it is integrated into the chromosomal DNA. Interestingly, when infecting plant tissue, Agrobacterium has the ability to inhibit the plant’s natural defense response [39].
The molecular mechanism of genetic transformation using Agrobacterium is well understood. It is known that the transfer of genes from a bacterial cell to a plant cell is based on the transfer of T-DNA, which is part of the Ti megaplasmid. This motile genetic element is responsible for tumor induction and biosynthesis of opines in plant tissue. It contains two regions associated with bacterial–plant interactions: vir regions, containing virulence genes (virA, virB, virC, virD, virE, virG, virF and virH) encoding proteins actively involved in the transfer and integration of the transgene with the plant genome, and a region containing genes coding the synthesis of inter alia opines, which are used as a source of carbon and nitrogen by bacteria [40]. This procedure allows the creation of transgenic plants by so called stable transformation, in which the new trait is inherited by further generations, or transient transformation, in which the genetic material persists in the cell nucleus without permanent integration with the genetic material. Since the first successful attempts at genetic transformation of plants by Agrobacterium in the early 1980s, the system has demonstrated great potential in transforming dicotyledonous and monocotyledonous plants.
However, other ways exist for changing the plant genome. Such chemical methods include treatment of protoplasts with polyethylene glycol (PEG), facilitating stable and transient transformation [41]. Transformation can also be made more effective by electroporation: the creation of transient micropores in the cell membrane by an electrical impulse to allow the passage of DNA into protoplasts [42]. Finally, microprojectile bombardment can be used, in which particles of gold or tungsten are coated with the appropriately prepared DNA molecules and pushed in the cell by high voltage or compressed gas [43].
Plant genetic transformation has a very wide application in expression of recombinant proteins [44]; these can have many therapeutic or diagnostic uses, increase plant resistance to biotic and abiotic stresses [45] or increase their nutritional or taste values [46]. Another approach is known as metabolic engineering: it can be used to increase the production of selected metabolites that are naturally synthesized in their tissues, or for the synthesis of completely new compounds [47,48].

4. Binary Vectors as a Basic Tool in Plant Genetic Transformation

New developments in genetic engineering have allowed much greater control over the transfer of foreign genes to plant cells. A number of vectors can now be used in transgenesis [28,49] that enable different marker and reporter genes and restriction sites to be chosen; they also allow high copy numbers of the binary vectors already in common use in biotechnological laboratories in bacterial cells. Agrobacterium- based transformation typically proceeds according to the following process: first, naturally-occurring oncogenes are removed to deprive the bacterium of pathogenicity; such a strain is often referred to as disarmed. The genes of interest are added to the T-DNA, together with appropriate selection or reporter genes. Due to its large size, the Ti (tumor inducing) or Ri (root inducing) plasmid does not replicate in Escherichia coli and hence only occurs in bacterial cells in low copy numbers, thus complicating its isolation; to allow easier manipulation, many works are carried out with a so-called binary vector system. The idea of this approach is to separate vir and T-DNA regions into two independent replicons. The T-DNA is located on a binary vector containing the origin of replication for E.coli and Agrobacterium. This vector also contains left and right borders designating the T-DNA region, selection genes for bacterial and plant cells, and a number of specific sites recognized by restriction enzymes allowing for easy cloning (Figure 1).

5. Calli and Cell Suspension Cultures

Under natural conditions, callus tissue is formed in the plant at the site of its injury from surrounding cells to seal wound sites and prevent water loss [41]. In laboratory conditions, this cell mass is induced on artificial media by using plant growth regulators (mainly auxins and cytokinins). This type of plant culture can be successfully used for the regeneration of whole plants (embryogenic callus) or as a material for establishing plant suspension cultures. One strategy for obtaining selected secondary metabolites involves the use of medicinal plant cell suspension culture (MPCSC), where single cells or different sized aggregates of cells are cultured in agitated liquid medium.
Calli cultures are often used successfully to produce secondary metabolites of medical significance, many of which may be used in treating human diseases [50,51,52,53]. The synthesis of the desired compound can be increased by designing the genetic constructs and the way they are introduced. An example would be to induce overexpression of three stilbene synthase (STS) genes of Picea jezoensis (Siebold & Zucc.) Carr, viz. PjSTS1a, PjSTS2 and PjSTS3, in calli cultures of Vitis amurensis Rupr., resulting in an increase in the content of stilbene [54]. Another example is the overexpression of 1-deoxy-d-xylulose-5-phosphate synthase 1 (SrDXS1) and kaurenoic acid hydroxylase (SrKAH) in Stevia spp., which increases the production of steviol glycosides [55], or the overexpression of stilbene synthase (VaSTS7) to increase production of resveratrol in transgenic cell cultures of V. amurensis.
Another good source of secondary metabolites is by the use of transgenic cell suspension cultures. One example is the stable transformation of Silybum marianum (L.) Gaertn. cell suspension cultures with the Vitis vinifera L. stilbene synthase gene, allowing increased accumulation of t-resveratrol [56]. In addition, overexpression of the neutral / alkaline invertase (NINV) gene in Taxus chinensis (Rehder & E.H.Wilson) Rehder cell suspension significantly enhances the expression of the taxadiene synthase (TAS) gene, and the biosynthesis of seven individual taxanes [57].

6. Hairy Roots

Hairy root cultures arise as a result of infection of plant tissue by Agrobacterium rhizogenes, resulting in the unlimited growth of organized plant tissue. The ability of these bacteria to infect the plants is due to having large plasmids known as Ri plasmids (root inducing) that contain transfer rol (rolA, rolB, and rolC) genes responsible for the stable integration of genetic material into host cells DNA. For many reasons, this system of obtaining valuable biologically active compounds is very convenient. Hairy roots are fast growing, require no plant growth regulators, are highly genetically stable and are able to grow on a large scale [58]. Considering all the advantages of hairy roots over other types of in vitro plant culture systems, they seem to be the best transgenic alternative to medicinal plants occurring in the natural environment. Hairy roots produce relatively large amounts of biologically-active compounds without interference from the natural environment. In addition, the ability to change their metabolic pathways through genetic engineering puts them at the forefront of currently used in vitro plant cultures.
Hairy root cultures have an extremely wide range of applications in green biotechnology [59,60,61]. As well as production of recombinant proteins, biotransformation or phytoremediation, they are a rich source of many valuable secondary metabolites, especially those produced by medical plants. As a plant production system, hairy roots offer many advantages over bacterial or mammalian systems: they have the ability to synthesize many compounds that are difficult to synthesize chemically, while the ability to grow and develop on simple media makes them attractive for economic reasons. There is no risk of transmission of human or animal pathogens, which is very important for medicinal compounds. So far, many attempts have been made to induce hairy roots with specific traits, most of which are based on genetic engineering techniques that allow plant material with increased utility values to be obtained. A good example of this approach is the heterologous expression of Vitreoscilla hemoglobin in plastids using the pVHb-RecA construct, leading to increased production of hyoscyamine and scopolamine in Hyoscyamus niger L. in vitro transgenic plant cultures [62]. Another is the possibility of affecting the increase in the content of glycyrrhizic acid by overexpression of the β-amyrin synthase gene in hairy roots of Glycyrrhiza uralensis Fisch. ex DC. In this work, a genetic construct containing the tobacco root-specific promoter TobRB7 and GuBAS cDNA was used [63].

7. Selected Secondary Metabolites in Medical Use Obtained by in Vitro Transgenic Plant Culture

7.1. Anticancer Compounds

Cancer is a serious disease that causes the deaths of many people around the world every year [64] and new therapies are constantly being sought. In this context, compounds of natural origin, including many plant-derived chemicals, including paclitaxel, vinblastine, vincristine and camptothecin, play an extremely important role in prophylaxis and therapy [65,66,67]. These substances are often safer and less toxic than synthetic ones [68]. Great effort has been invested in increasing their production, and this has resulted in the design of a range of biotechnological methods to increase productivity.

7.2. Paclitaxel

Paclitaxel is an anti-cancer drug originally isolated from the bark of the Pacific yew tree, Taxus brevifolia Nutt. in 1971 [69]. It is known to act on microtubules, i.e., the structures responsible for the formation of the mitotic spindle during cell division and cytoplasmic movement in the cell; more specifically, the drug promotes the assembly of microtubules from tubulin dimers and stabilizes microtubules by preventing depolymerization. It thus blocks metaphase–anaphase transitions, inhibits mitosis, and induces apoptosis in a wide range of cancer cells [70].
The drug is used in the treatment of a range of conditions including lung, ovarian, breast, stomach, esophageal, cervical, and prostate cancer, as well as lymphoma and leukemia [70]. As this drug is widely used in cancer therapy in humans, there is great interest in increasing its productivity from natural sources. Although paclitaxel can be chemically synthesized [71], this process is not commercially profitable, and its best sources are in vitro and ex vitro plant cultures. A number of previous works have attempted to optimize the process of obtaining taxanes by genetic transformation. Some strategies have resulted in increased paclitaxel production: overexpression of 10-deacetylbaccatin III-10-O-acetyltransferase (DBAT) and taxadiene synthase (TXS) in transgenic Taxus marei (Lemee & H.Lev.) [72] or TXS in Taxus x media Rehder var. hicksii cell culture [73]. Other studies have found that enhancement of paclitaxel biosynthesis can be obtained by overexpression of the 9-cis-epoxycarotenoid dioxygenase gene in transgenic cell lines of T. chinensis [70]. In addition, genetic transformation of Nicotiana benthamiana Domin with a taxadiene synthase (TS) gene under control of the 35S Cauliflower Mosaic Virus (CaMV 35S) promoter was found to enable de novo production of taxadiene in N. benthamiana homozygous lines, yielding 11–27 μg taxadiene/g of dry weight; in addition, subsequent elicitor treatment (methyl jasmonate) increased taxadiene accumulation by a further 1.4 times [74]. A similar approach based on the in vitro transformation of T. x media hairy roots and subsequent elicitation allowed the production of paclitaxel; the vector was A. tumefaciens carrying the RiA4 plasmid and the binary vector pCAMBIA-TXS-His harboring the taxadiene synthase (txs) gene of Taxus baccata L. under the control of the 35S CaMV promoter [75].

7.3. Camptothecin

Camptothecin (CPT) is a monoterpene alkaloid and potent inhibitor of topoisomerase I (Top 1): a nuclear enzyme involved in DNA repair, recombination, transcription and replication [76]. This compound and several of its derivatives, such as irinothecan or topothecan, are in clinical use against a number of human cancers. In vitro plant cultures have long been used as a source of camptothecin, with the first works, in which Sakato et al. [77] presented the possibility of acquiring this compound from Camptotheca acuminate Decne cell suspension cultures, dating from the 1970s.
Since these initial studies, more focused attempts have been made to increase camptothecin production using modern biotechnology in plant cells, for example, overexpression of the ORCA3 gene in transgenic hairy root lines [78] and allene oxide cyclase from C. acuminate [79].
Cui et al. [80] obtained Ophiorrhiza pumila Champ. ex Benth hairy roots with separate or simultaneous overexpression of the transformed Catharanthus roseus (L.) G. Don genes for strictosidine synthase (STR) and geraniol 10-hydroxylase (G10H). Their findings clearly show greater accumulation of CPT in transformed O. pumila hairy roots. Co-overexpression of the G10H and STR genes resulted in a 56% increase in camptothecin accumulation compared to non-transgenic HR lines. In addition, a comparison of 25 hairy root lines of Ophiorrhiza mungos L., obtained via A. rhizogenes transformation, confirmed elevated camptothecin production in the hairy roots (0.32% CPT of DW) compared to non-transformed cultures (0.25% CPT of DW) [81]. Wang et al. also reported significantly greater total production of camptothecin in selected transgenic hairy roots of O. pumila. Transformation was performed using a genetic construct encoding tryptophan-arginine-lysine-tyrosine (WRKY), OpWRKY3 isolated from O. pumila, which has high homology with the VvWRKY30 factor [82]. These results further demonstrate the potential of transgenesis to modulate the productivity of plant cells.

7.4. Vincristine

Vincristine is one of the first plant alkaloids approved by the FDA for use in the treatment of cancer. In nature, it occurs in the leaves of Catharanthus roseus (L.) G.Don and has been used for a long time in various branches of medicine, including pediatric oncology, as an effective drug against lymphoblastic leukemia [83], rhabdomyosarcoma [84], neuroblastoma [85], Hodgkin lymphoma [86] and Wilms tumor lymphomas [87]. The mechanism of its anti-tumor activity is based on its prevention of microtubule formation by binding to tubulin. As a consequence, mitosis is halted in metaphase following disruption of mitotic spindle formation. The compound also has the ability to inhibit the synthesis of nucleic acids and proteins [88].
Canel et al. [89] overexpressed two genes coding for tryptophan decarboxylase and strictosidine synthase in callus and leaf tissue of C. roseus via A. tumefaciens LBA 1119 transformation. Pham et al. [1] also conducted an agrotransformation of periwinkle using a CrDAT transgene encoding deacetylvindoline 4-O-acetyltransferase (DAT): a key enzyme that catalyses the formation of vinblastine and vincristine. The authors demonstrated that selected C. roseus lines are capable of higher accumulation of vincristine through overexpression of the DAT protein, involved in the biosynthesis of terpenoid indole alkaloids (TIAs); in this case, the DAT gene itself was under the control of the CaMV 35S promoter. Transformation resulted in 1.63- to 2.48-fold greater production of vincristine compared to non-transgenic plants. These results show that the expression of DAT can significantly affect the accumulation of vincristine in C. roseus.

7.5. Vinblastine

Vinblastine has a similar mechanism of action to vincristine and is also a widely-used plant alkaloid in cancer therapy. Similar to other valuable plant metabolites, vinblastine is also of interest to modern plant biotechnology and many studies have attempted to increase its production in plant tissues. One study examined the effect of overexpression of the MYC1 transcription factor (CrMYC1) in C. roseus [90]. CrMYC1 has been characterized as one of the main components regulating the biosynthesis of terpenoid indole alkaloid metabolites in this plant. Briefly, the authors cloned the CrMYC1 coding sequence into a plant binary vector and then transiently expressed the gene in C. roseus by agroinfiltration. The resulting overexpression of this transcription factor increases the level of important terpenoid indole alkaloids such as vinblastine, vincristine or catharantine. The results showed a 2.5-fold increase in vinblastine production and a 3-fold increase in catharanthine relative to control plants. Another study employed transient overexpression of CrERF5 (AP2/ERF transcription factor) in C. roseus petals to increase the expression of key genes in the monoterpene indole alkaloid (MIA) biosynthesis pathways [91]. The transformation led to an increase in the content of the bisindole alkaloids anhydrovinblastine and vinblastine, and the monoindole alkaloids ajmalicine, vindoline and catharanthine.

8. Overproduction of Other Secondary Metabolites in Transgenic in Vitro Cell Culture

Each class of secondary metabolites is formed by a complex network of precursors, enzymes and co-factors, some of them leading to specific plant-derived medicinal compounds. In addition to the main anti-cancer alkaloids, which have been presented above, a number of other terpenes, or phenolic compounds, which are common in many other plant species of the Lamiaceae, Asteracea and Fabaceae families, among others, are of potential value. Many such compounds are known to possess various anti-inflammatory, anticancer, antioxidant, antidiabetic, hepatoprotective or antimicrobial properties [15,16,17,18,19,20,21,22,23]. Of these, the antibacterial properties have drawn significant interest due to the growing problem of bacterial infections around the world [92,93]. These compounds are known to disrupt membrane function and structures, interrupt DNA or RNA synthesis and function, and interfere with intermediary metabolism or intercellular communication via various mechanisms of action [92,94,95]. Importantly, the prevalence of multi antibiotic-resistant bacterial strains in the environment is growing [96,97]. A good example of the potential of transformation to improve the biosynthetic efficiency of antibacterial metabolites can be seen in transgenic Codonopsis lanceolata (Siebold & Zucc.) Beneth. and Hook.f. ex Trautv. over-expressing the γ-tocopherol methyl transferase (γ-tmt) gene, which leads to increased antimicrobial activity against gram-positive and gram-negative bacteria compared to controls. The plant was transformed with a genetic construct containing γ-tmt cDNAs from Arabidopsis thaliana (L.) Heynh. under the control of CaMV 35S promoter and NOS terminator [98]. In turn, Ghimire et al. [99] report that overexpression of the γ-tmt gene in Perilla frutescens L. increases the levels of phenolic compounds (gallic acid, pyrogallol, 5-sulfosalicylic acid, catechin, chlorogenic acid, vanillin, syringic acid, naringenin, salicylic acid, quercetin, o-coumaric, kaempferol, hesperetin and benzoic) in the transgenic plants, and that this elevated phenolic content was associated with stronger antimicrobial activity in comparison to wild plants.
Other broad-spectrum metabolites can be modified to increase the production of specific molecules in in vitro plant cultures. For example, over-expression of the AtPAP1 transcriptional factor was found to enhance phenolic acid (such as chlorogenic acid, caffeic acid, ferulic acid or p-coumaric acid) production in a transgenic root culture of Leonurus sibiricus L., and the transgenic roots with the AtPAP1 transcriptional factor demonstrated better antimicrobial potential and cytotoxic activity against grade IV glioma cells [100]. Other studies have found such transgenic roots, incorporating the AtPAP1 transcriptional factor, to demonstrate better anticancer effects vis DNA damage, PARP cleavage/increased H2A.X histone levels and UHRF-1/DNMT1 downregulation of mRNA levels compared to untransformed roots [101]. In addition, the extract derived from transgenic L. sibiricus roots overexpressing AtPAP1 demonstrated a stronger cytotoxic effect against melanoma cells and had a higher antioxidant potential in human blood plasma [102]. In turn, better cytotoxic and genotoxic effects were demonstrated against acute lymphoblastic leukemia (CCRF-CEM) and chronic myelogenous leukemia (K562) cell lines after treatment with extract from AtPAP1 transformed roots, possibly due to its higher phenolic acid content [103].
A representative terpene is betulinic acid, a pentacyclic triterpenoid, which is gaining considerable attention due to its unique anti-cancer activity, allowing selective inhibition of melanoma growth without damaging normal cells [104]. Its presence was initially reported in Betula spp., Ocimum spp., Senna spp. and Menynathes spp., among others. Suzuki et al. revealed that metabolic modification of Lotus japonicus L. by changes in CYP716A51 expression may increase betulinic acid biosynthesis [105]. These studies confirm that biotechnology techniques are legitimate tools for increasing the production of secondary metabolites in in vitro plant cultures. Other examples of metabolic pathway manipulation to increase secondary metabolite content in in vitro plant cultures are presented in Table 1 below.

9. In Vitro Transgenic Plant Cultures and Societal Implication

The intensive pace of recent scientific progress has been accompanied by a similar growth in public interest regarding issues related to health protection and the treatment of civilization diseases. In addition, with the growth in awareness of the dangers associated with a polluted environment, and the measurable economic benefits associated with productive agriculture, comes a greater willingness to accept research on genetic modification. With this in mind, the metabolic engineering of plant cells is an effective way to obtain valuable biologically-active compounds for pharmaceuticals. Today, thanks to modern biotechnology, it is possible to produce many therapeutics, even on an industrial scale.
Plant-based genetic manipulation is regarded as far more acceptable than such work with animals. Humans have modified plant genomes using conventional methods (crossing) for thousands of years. In addition, as the use of in vitro transgenic plant cultures is usually limited only to the laboratory, they do not have such negative connotations as the production of transgenic crops. In addition, given the appropriate cooperation of the scientific community, such research enjoys quite high social acceptance. Most importantly, green biotechnology can significantly contribute to achieving many of the sustainable development goals (SDGs) set in 2015 by the United Nations, these being intended to cover a broad range of global social and economic development targets by 2030 [149].

10. Conclusions

Many compounds currently used in medicine are of plant origin, and the literature suggests a growing tendency to return to biologically-active compounds of natural origin. In contrast to chemically synthesized compounds, many compounds from a natural origin show greater biological safety, generate fewer side effects and are often characterized by lower production costs. However, due to their limited adaptability, the diversity of medicinal plant species in their natural environment has been shrinking the face of intense and unfavorable climate change and growing anthropogenic environmental pollution. In response to this, and the constantly growing demand for compounds of plant origin, new and more efficient in vitro techniques for growing plants are being developed. The literature reviewed showed that in combination with the currently-available precision tools of molecular biology and genetic engineering, high-throughput in vitro plant cultures can be in some cases used to provide many natural secondary metabolites. Modern green biotechnology, which allows manipulation of cellular processes at many levels, can be an excellent alternative to traditional methods of obtaining biologically-active compounds. The ability to create various genetic constructs and introduce them into the plant genome can be an efficient production platform for a wide range of compounds used in medicine, diagnostics or industry. Currently, intensive work is underway on new biotechnological solutions and sustainable alternative methods of producing high-value plant metabolites.

Author Contributions

Conceptualization: T.K.; T.K. wrote the manuscript with support from P.S. and J.W., critical review: E.S. and T.Ś. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Pham, T.T.N.; Nguyen, T.N.L.; Bui, T.H.; Nguyen, H.Q.; Nguyen, T.T.; Le, V.S.; Chu, H.M. Agrobacterium-mediated transformation of the CrDAT gene and selection of transgenic periwinkle lines with a high vincristine accumulation. J. Hortic. Sci. Biotechnol. 2019, 94, 591–598. [Google Scholar] [CrossRef]
  2. Petrovska, B.B. Historical review of medicinal plants’ usage. Pharmacogn. Rev. 2012, 6, 1–5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Kumar, A.; Kumar, S.; Thomas, T.D.; Ramchiary, N.; Swamy, M.K.; Ahmad, I. Linking Omics Approaches to Medicinal Plants and Human Health. In Natural Bio-Active Compounds; Springer: Berlin/Heidelberg, Germany, 2019; pp. 31–57. [Google Scholar]
  4. Lu, X.; Tang, K.; Li, P. Plant metabolic engineering strategies for the production of pharmaceutical terpenoids. Front. Plant Sci. 2016, 7, 1–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Zaynab, M.; Fatima, M.; Abbas, S.; Sharif, Y.; Umair, M.; Zafar, M.H.; Bahadar, K. Role of secondary metabolites in plant defense against pathogens. Microb. Pathog. 2018, 124, 198–202. [Google Scholar] [CrossRef]
  6. Bednarek, P.; Schulze-Lefert, P. Role of Plant Secondary Metabolites at the Host-Pathogen Interface. In Molecular Aspects of Plant Disease Resistance 2018; Parker, J., Ed.; Wiley-Blackwell: Hoboken, NJ, USA, 2008; Volume 34, pp. 220–260. [Google Scholar]
  7. Köhler, H.; Contreras, R.A.; Pizarro, M.; Cortés-Antíquera, R.; Zúñiga, G.E. Antioxidant responses induced by UVB radiation in Deschampsia Antarctica Desv. Front. Plant Sci. 2017, 8, 1–10. [Google Scholar] [CrossRef] [Green Version]
  8. Takshak, S.; Agrawal, S.B. Defense potential of secondary metabolites in medicinal plants under UV-B stress. J. Photochem. Photobiol. B Biol. 2019, 193, 51–88. [Google Scholar] [CrossRef]
  9. Huang, W.; Gfeller, V.; Erb, M. Root volatiles in plant–plant interactions II: Root volatiles alter root chemistry and plant–herbivore interactions of neighbouring plants. Plant Cell Environ. 2019, 42, 1964–1973. [Google Scholar] [CrossRef] [Green Version]
  10. Huber, M.; Epping, J.; Schulze Gronover, C.; Fricke, J.; Aziz, Z.; Brillatz, T.; Swyers, M.; Köllner, T.G.; Vogel, H.; Hammerbacher, A.; et al. A Latex Metabolite Benefits Plant Fitness under Root Herbivore Attack. PLoS Biol. 2016, 14, 1–27. [Google Scholar] [CrossRef] [Green Version]
  11. Miró, Ò.; Gil, V.; Peacock, W.F. Morphine in acute heart failure: Good in relieving symptoms, bad in improving outcomes. J. Thorac. Dis. 2017, 9, E871–E874. [Google Scholar] [CrossRef] [Green Version]
  12. Holan, V.; Cechova, K.; Zajicova, A.; Kossl, J.; Hermankova, B.; Bohacova, P.; Hajkova, M.; Krulova, M.; Svoboda, P.; Javorkova, E. The Impact of Morphine on the Characteristics and Function Properties of Human Mesenchymal Stem Cells. Stem Cell Rev. Rep. 2018, 14, 801–811. [Google Scholar] [CrossRef]
  13. Tuerxun, H.; Cui, J. The dual effect of morphine on tumor development. Clin. Transl. Oncol. 2019, 21, 695–701. [Google Scholar] [CrossRef] [Green Version]
  14. Nishiwada, T.; Kawaraguchi, Y.; Uemura, K.; Kawaguchi, M. Morphine inhibits cell viability and growth via suppression of vascular endothelial growth factor in human oral cancer HSC-3 cells. J. Anesth. 2019, 33, 408–415. [Google Scholar] [CrossRef] [PubMed]
  15. Zlatić, N.; Jakovljević, D.; Stanković, M. Temporal, plant part, and interpopulation variability of secondary metabolites and antioxidant activity of Inula helenium L. Plants 2019, 8, 179. [Google Scholar] [CrossRef] [Green Version]
  16. Gonçalves, S.; Mansinhos, I.; Rodríguez-Solana, R.; Pérez-Santín, E.; Coelho, N.; Romano, A. Elicitation improves rosmarinic acid content and antioxidant activity in Thymus lotocephalus shoot cultures. Ind. Crops Prod. 2019, 137, 214–220. [Google Scholar] [CrossRef]
  17. Bernstein, N.; Akram, M.; Daniyal, M.; Koltai, H.; Fridlender, M.; Gorelick, J. Antiinflammatory Potential of Medicinal Plants: A Source for Therapeutic Secondary Metabolites, 1st ed.; Elsevier Inc.: Amsterdam, The Netherlands, 2018; Volume 150. [Google Scholar]
  18. Flores-Sánchez, K.; Cruz-Sosa, F.; Zamilpa-Alvarez, A.; Nicasio-Torres, P. Active compounds and anti-inflammatory activity of the methanolic extracts of the leaves and callus from Tilia americana var. mexicana propagated plants. Plant Cell Tissue Organ Cult. 2019, 137, 55–64. [Google Scholar] [CrossRef]
  19. Barbieri, R.; Coppo, E.; Marchese, A.; Daglia, M.; Sobarzo-Sánchez, E.; Nabavi, S.F.; Nabavi, S.M. Phytochemicals for human disease: An update on plant-derived compounds antibacterial activity. Microbiol. Res. 2017, 196, 44–68. [Google Scholar] [CrossRef]
  20. Reichling, J. Plant-Microbe Interactions and Secondary Metabolites with Antibacterial, Antifungal and Antiviral Properties, 2nd ed.; Functions and Biotechnology of Plant Secondary Metabolites, Chapter 4; Annual Plant Reviews; Wink, M., Ed.; Wiley: Oxford, UK, 2010; Volume 39. [Google Scholar]
  21. Lagrouh, F.; Dakka, N.; Bakri, Y. The antifungal activity of Moroccan plants and the mechanism of action of secondary metabolites from plants. Journal de Mycologie Medicale 2017, 27, 303–311. [Google Scholar] [CrossRef] [PubMed]
  22. Epifano, F.; Molinaro, G.; Genovese, S.; Ngomba, R.T.; Nicoletti, F.; Curini, M. Neuroprotective effect of prenyloxycoumarins from edible vegetables. Neurosci. Lett. 2008, 443, 57–60. [Google Scholar] [CrossRef] [PubMed]
  23. Pereira, C.; Barros, L.; Ferreira, I.C. Extraction, identification, fractionation and isolation of phenolic compounds in plants with hepatoprotective effects. J. Sci. Food Agric. 2016, 96, 1068–1084. [Google Scholar] [CrossRef]
  24. Zhou, M.; Memelink, J. Jasmonate-responsive transcription factors regulating plant secondary metabolism. Biotechnol. Adv. 2016, 34, 441–449. [Google Scholar] [CrossRef]
  25. Nielsen, E.; Temporiti, M.E.E.; Cella, R. Improvement of phytochemical production by plant cells and organ culture and by genetic engineering. Plant Cell Rep. 2019, 38, 1199–1215. [Google Scholar] [CrossRef] [PubMed]
  26. Sui, X.; Zhao, M.; Xu, Z.; Zhao, L.; Han, X. RrGT2, a key gene associated with anthocyanin biosynthesis in Rosa rugosa, was identified via virus-induced gene silencing and overexpression. Int. J. Mol. Sci. 2018, 19, 4057. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Catania, T.M.; Branigan, C.A.; Stawniak, N.; Hodson, J.; Harvey, D.; Larson, T.R.; Czechowski, T.; Graham, I.A. Silencing amorpha-4,11-diene synthase genes in Artemisia annua leads to FPP accumulation. Front. Plant Sci. 2018, 9, 1–12. [Google Scholar] [CrossRef]
  28. Bahramnejad, B.; Naji, M.; Bose, R.; Jha, S. A critical review on use of Agrobacterium rhizogenes and their associated binary vectors for plant transformation. Biotechnol. Adv. 2019, 1–14. [Google Scholar] [CrossRef] [PubMed]
  29. Lampropoulos, A.; Sutikovic, Z.; Wenzl, C.; Maegele, I.; Lohmann, J.U.; Forner, J. GreenGate—A novel, versatile, and efficient cloning system for plant transgenesis. PLoS ONE 2013, 8, e83043. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Dafny-Yelin, M.; Tzfira, T. Delivery of multiple transgenes to plant cells. Plant Physiol. 2007, 145, 1118–1128. [Google Scholar] [CrossRef] [Green Version]
  31. Ncube, B.; van Staden, J. Tilting plant metabolism for improved metabolite biosynthesis and enhanced human benefit. Molecules 2015, 20, 12698–12731. [Google Scholar] [CrossRef] [Green Version]
  32. Guerriero, G.; Berni, R.; Muñoz-Sanchez, J.A.; Apone, F.; Abdel-Salam, E.M.; Qahtan, A.A.; Alatar, A.A.; Cantini, C.; Cai, G.; Hausman, J.F.; et al. Production of plant secondary metabolites: Examples, tips and suggestions for biotechnologists. Genes 2018, 9, 309. [Google Scholar] [CrossRef] [Green Version]
  33. Shi, M.; Luo, X.; Ju, G.; Li, L.; Huang, S.; Zhang, T.; Wang, H.; Kai, G. Enhanced Diterpene Tanshinone Accumulation and Bioactivity of Transgenic Salvia miltiorrhiza Hairy Roots by Pathway Engineering. J. Agric. Food Chem. 2016, 64, 2523–2530. [Google Scholar] [CrossRef]
  34. Hemanthakumar, A.S.; Preetha, T.S.; Pillai, P.P.; Seeni, S. Embryogenesis followed by enhanced micro-multiplication and eco-restoration of Calamus thwaitesii Becc.: An economic non-wood forest produce for strengthening agroforestry system. Agrofor. Syst. 2019, 93, 1093–1105. [Google Scholar] [CrossRef]
  35. Kado, C.I. Historical account on gaining insights on the mechanism of crown gall tumorigenesis induced by Agrobacterium tumefaciens. Front. Microbiol. 2014, 5, 1–15. [Google Scholar] [CrossRef] [PubMed]
  36. Nester, E.W. Agrobacterium: Nature’s Genetic Engineer. Front. Plant Sci. 2015, 5, 1–16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Aguilar, J.; Zupan, J.; Cameron, T.A.; Zambryski, P.C. Agrobacterium type IV secretion system and its substrates form helical arrays around the circumference of virulence-induced cells. Proc. Natl. Acad. Sci. USA 2010, 107, 3758–3763. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Bevan, M.W.; Flavell, R.B.; Chilton, M.D. A chimaeric antibiotic resistance gene as a selectable marker for plant cell transformation. Nature 1983, 304, 184–187. [Google Scholar] [CrossRef]
  39. Pitzschke, A. Agrobacterium infection and plant defense-transformation success hangs by a thread. Front. Plant Sci. 2013, 4, 1–12. [Google Scholar] [CrossRef] [Green Version]
  40. Guo, M.; Ye, J.; Gao, D.; Xu, N.; Yang, J. Agrobacterium-mediated horizontal gene transfer: Mechanism, biotechnological application, potential risk and forestalling strategy. Biotechnol. Adv. 2019, 37, 259–270. [Google Scholar] [CrossRef]
  41. Ikeuchi, M.; Iwase, A.; Rymen, B.; Lambolez, A.; Kojima, M.; Takebayashi, Y.; Heyman, J.; Watanabe, S.; Seo, M.; de Veylder, L.; et al. Wounding triggers callus formation via dynamic hormonal and transcriptional changes. Plant Physiol. 2017, 175, 1158–1174. [Google Scholar] [CrossRef] [Green Version]
  42. Chen, L.; Masuoka, S.; Nishimura, Y.; Sakai, T.; Takahata, Y.; Xu, C. Attempt to Establish Direct Gene Transformation System to Seeds of Sweet Potato (Ipomoea batatas) Using Electroporation Method. Biotechnol. J. Int. 2017, 19, 1–8. [Google Scholar] [CrossRef]
  43. Gao, S.; Yang, Y.; Xu, L.; Guo, J.; Su, Y.; Wu, Q.; Wang, C.; Que, Y. Particle bombardment of the cry2A gene cassette induces stem borer resistance in sugarcane. Int. J. Mol. Sci. 2018, 19, 1692. [Google Scholar] [CrossRef] [Green Version]
  44. Buyel, J.F. Plants as sources of natural and recombinant anti-cancer agents. Biotechnol. Adv. 2018, 36, 506–520. [Google Scholar] [CrossRef]
  45. Erpen, L.; Devi, H.S.; Grosser, J.W.; Dutt, M. Potential use of the DREB/ERF, MYB, NAC and WRKY transcription factors to improve abiotic and biotic stress in transgenic plants. Plant Cell Tissue Organ Cult. 2018, 132, 1–25. [Google Scholar] [CrossRef]
  46. Reddy, C.S.; Vijayalakshmi, M.; Kaul, T.; Islam, T.; Reddy, M.K. Improving Flavour and Quality of Tomatoes by Expression of Synthetic Gene Encoding Sweet Protein Monellin. Mol. Biotechnol. 2015, 57, 448–453. [Google Scholar] [CrossRef] [PubMed]
  47. Satish, L.; Rency, A.S.; Muthubharathi, B.C.; Shamili, S.; Rameshkumar, R.; Swamy, M.K.; Ramesh, M. Transgenic Plant Cell Cultures: A Promising Approach for Secondary Metabolite Production. In Natural Bio-Active Compounds; Akhtar, M., Swamy, M., Eds.; Springer: Singapore, 2019. [Google Scholar]
  48. Jensen, P.E.; Scharff, L.B. Engineering of plastids to optimize the production of high-value metabolites and proteins. Curr. Opin. Biotechnol. 2019, 59, 8–15. [Google Scholar] [CrossRef]
  49. Pasin, F.; Bedoya, L.C.; Bernabé-Orts, J.M.; Gallo, A.; Simón-Mateo, C.; Orzaez, D.; García, J.A. Multiple T-DNA Delivery to Plants Using Novel Mini Binary Vectors with Compatible Replication Origins. ACS Synth. Biol. 2017, 6, 1962–1968. [Google Scholar] [CrossRef]
  50. Deshpande, A.; Dhadi, S.R.; Hager, E.J.; Ramakrishna, W. Anticancer activity of rice callus suspension culture. Phytother. Res. 2012, 26, 1075–1081. [Google Scholar] [CrossRef]
  51. Mittal, J.; Sharma, M.M. Enhanced production of berberine in In vitro regenerated cell of Tinospora cordifolia and its analysis through LCMS QToF. 3 Biotech 2017, 7, 1–12. [Google Scholar] [CrossRef] [Green Version]
  52. Rady, M.R.; Aboul-Enein, A.M.; Ibrahim, M.M. Active compounds and biological activity of in vitro cultures of some Echinacea purpurea varieties. Bull. Nat. Res. Cent. 2018, 42, 1–12. [Google Scholar] [CrossRef]
  53. Rahpeyma, S.A.; Moieni, A.; Jalali Javaran, M. Paclitaxel production is enhanced in suspension-cultured hazel (Corylus avellana L.) cells by using a combination of sugar, precursor, and elicitor. Eng. Life Sci. 2015, 15, 234–242. [Google Scholar] [CrossRef]
  54. Suprun, A.R.; Ogneva, Z.V.; Dubrovina, A.S.; Kiselev, K.V. Effect of spruce PjSTS1a, PjSTS2, or PjSTS3 gene overexpression on stilbene biosynthesis in callus cultures of Vitis amurensis Rupr. Biotechnol. Appl. Biochem. 2019, 1–7. [Google Scholar] [CrossRef]
  55. Zheng, J.; Zhuang, Y.; Mao, H.Z.; Jang, I.C. Overexpression of SrDXS1 and SrKAH enhances steviol glycosides content in transgenic Stevia plants. BMC Plant Biol. 2019, 19, 1–16. [Google Scholar] [CrossRef]
  56. Hidalgo, D.; Martínez-Márquez, A.; Cusidó, R.; Bru-Martínez, R.; Palazón, J.; Corchete, P. Silybum marianum cell cultures stably transformed with Vitis vinifera stilbene synthase accumulate t-resveratrol in the extracellular medium after elicitation with methyl jasmonate or methylated β-cyclodextrins. Eng. Life Sci. 2017, 17, 686–694. [Google Scholar] [CrossRef] [Green Version]
  57. Dong, Y.; Duan, W.; He, H.; Su, P.; Zhang, M.; Song, G.; Fu, C.; Yu, L. Enhancing taxane biosynthesis in cell suspension culture of Taxus chinensis by overexpressing the neutral/alkaline invertase gene. Process Biochem. 2015, 50, 651–660. [Google Scholar] [CrossRef]
  58. Mehrotra, S.; Srivastava, V.; Rahman, L.U.; Kukreja, A.K. Hairy root biotechnology—Indicative timeline to understand missing links and future outlook. Protoplasma 2015, 252, 1189–1201. [Google Scholar] [CrossRef] [PubMed]
  59. Nigutová, K.; Kusari, S.; Sezgin, S.; Petijová, L.; Henzelyová, J.; Bálintová, M.; Spiteller, M.; Čellárová, E. Chemometric evaluation of hypericin and related phytochemicals in 17 in vitro cultured Hypericum species, hairy root cultures and hairy root-derived transgenic plants. J. Pharm. Pharmacol. 2019, 71, 46–57. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Mottaki, Z.; Rezayian, M.; Niknam, V.; Ebrahimzadeh, H.; Mirmasoumi, M. Using hairy roots for production of secondary metabolites in Artemisia. Plant Biotechnol. Rep. 2019, 13, 263–271. [Google Scholar] [CrossRef]
  61. Matvieieva, N.; Drobot, K.; Duplij, V.; Ratushniak, Y.; Shakhovsky, A.; Kyrpa-Nesmiian, T.; Mickevičius, S.; Brindza, J. Flavonoid content and antioxidant activity of Artemisia vulgaris L. “hairy” roots. Prep. Biochem. Biotechnol. 2019, 49, 82–87. [Google Scholar] [CrossRef]
  62. Guo, Z.; Tan, H.; Lv, Z.; Ji, Q.; Huang, Y.; Liu, J.; Chen, D.; Diao, Y.; Si, J.; Zhang, L. Targeted expression of Vitreoscilla hemoglobin improves the production of tropane alkaloids in Hyoscyamus niger hairy roots. Sci. Rep. 2018, 8, 1–12. [Google Scholar]
  63. Yin, Y.; Zhang, X.; Gao, Z.; Hu, T.; Yang, L.; Zhang, Z.; Li, W.; Liu, Y. Over-expressing root-specific β-amyrin synthase gene increases glycyrrhizic acid content in hairy roots of glycyrrhiza uralensis. Chin. Herb. Med. 2019, 11, 192–199. [Google Scholar] [CrossRef]
  64. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA A Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef] [Green Version]
  65. George, V.C.; Dellaire, G.; Rupasinghe, H.P.V. Plant flavonoids in cancer chemoprevention: Role in genome stability. J. Nutr. Biochem. 2017, 45, 1–14. [Google Scholar] [CrossRef]
  66. Bishayee, A.; Sethi, G. Bioactive natural products in cancer prevention and therapy: Progress and promise. Semin. Cancer Biol. 2016, 40–41, 1–3. [Google Scholar] [CrossRef] [PubMed]
  67. Chikara, S.; Nagaprashantha, L.D.; Singhal, J.; Horne, D.; Awasthi, S.; Singhal, S.S. Oxidative stress and dietary phytochemicals: Role in cancer chemoprevention and treatment. Cancer Lett. 2018, 413, 122–134. [Google Scholar] [CrossRef] [PubMed]
  68. Seca, A.M.L.; Pinto, D.C.G.A. Plant secondary metabolites as anticancer agents: Successes in clinical trials and therapeutic application. Int. J. Mol. Sci. 2018, 19, 263. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Wani, M.C.; Taylor, H.L.; Wall, M.E.; Coggon, P.; Mcphail, A.T. Plant Antitumor Agents.VI.The Isolation and Structure of Taxol, a Novel Antileukemic and Antitumor Agent from Taxus brevifolia. J. Am. Chem. Soc. 1971, 93, 2325–2327. [Google Scholar] [CrossRef]
  70. Stage, T.B.; Bergmann, T.K.; Kroetz, D.L. Clinical Pharmacokinetics of Paclitaxel Monotherapy: An Updated Literature Review. Clin. Pharmacokinet. 2018, 57, 7–19. [Google Scholar] [CrossRef] [Green Version]
  71. Fukaya, K.; Tanaka, Y.; Sato, A.C.; Kodama, K.; Yamazaki, H.; Ishimoto, T.; Nozaki, Y.; Iwaki, Y.M.; Yuki, Y.; Umei, K.; et al. Synthesis of paclitaxel. 1. synthesis of the abc ring of paclitaxel by SmI2-mediated cyclization. Org. Lett. 2015, 17, 2570–2573. [Google Scholar] [CrossRef]
  72. Ho, C.; Chang, S.; Lung, J. The Strategies to Increase Taxol Production by Using Taxus mairei Cells Transformed with TS and DBAT Genes. Int. J. 2005, 3, 179–185. [Google Scholar]
  73. Sykłowska-Baranek, K.; Pilarek, M.; Bonfill, M.; Kafel, K.; Pietrosiuk, A. Perfluorodecalin-supported system enhances taxane production in hairy root cultures of Taxus x media var. Hicksii carrying a taxadiene synthase transgene. Plant Cell Tissue Organ Cult. 2015, 120, 1051–1059. [Google Scholar] [CrossRef] [Green Version]
  74. Hasan, M.M.; Kim, H.S.; Jeon, J.H.; Kim, S.H.; Moon, B.K.; Song, J.Y.; Shim, S.H.; Baek, K.H. Metabolic engineering of Nicotiana benthamiana for the increased production of taxadiene. Plant Cell Rep. 2014, 33, 895–904. [Google Scholar] [CrossRef]
  75. Sykłowska-Baranek, K.; Grech-Baran, M.; Naliwajski, M.R.; Bonfill, M.; Pietrosiuk, A. Paclitaxel production and PAL activity in hairy root cultures of Taxus x media var. Hicksii carrying a taxadiene synthase transgene elicited with nitric oxide and methyl jasmonate. Acta Physiol. Plant. 2015, 37, 1–9. [Google Scholar] [CrossRef] [Green Version]
  76. Martino, E.; Della Volpe, S.; Terribile, E.; Benetti, E.; Sakaj, M.; Centamore, A.; Sala, A.; Collina, S. The long story of camptothecin: From traditional medicine to drugs. Bioorg. Med. Chem. Lett. 2017, 27, 701–707. [Google Scholar] [CrossRef] [PubMed]
  77. Sakato, K.; Tanaka, H.; Mukai, N.; Misawa, M. Isolation and Identification of Camptothecin from Cells of Camptotheca acuminata Suspension Cultures. Agric. Biol. Chem. 1974, 38, 217–218. [Google Scholar] [CrossRef]
  78. Ni, X.; Wen, S.; Wang, X.; Wang, W.; Xu, H.; Kai, G. Enhancement of camptothecin production in Camptotheca acuminata hairy roots by overexpressing ORCA3 gene. J. Appl. Pharm. Sci. 2011, 1, 85–88. [Google Scholar]
  79. Yan Pi Effects of over-expression of allene oxide cyclase on camptothecin production by cell cultures of Camptotheca acuminata. Afr. J. Biotechnol. 2012, 11, 6535–6541.
  80. Cui, L.; Ni, X.; Ji, Q.; Teng, X.; Yang, Y.; Wu, C.; Zekria, D.; Zhang, D.; Kai, G. Co-overexpression of geraniol-10-hydroxylase and strictosidine synthase improves anti-cancer drug camptothecin accumulation in Ophiorrhiza pumila. Sci. Rep. 2015, 5, 1–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Wetterauer, B.; Wildi, E.; Wink, M. Production of the anticancer compound camptothecin in root and hairy root cultures of Ophiorrhiza mungos L. In Biotechnological Approaches for Medicinal and Aromatic Plants—Conservation, Genetic Improvement and Utilization; Nitish, K., Ed.; Springer: Singapore, 2018; pp. S303–S341. [Google Scholar]
  82. Wang, C.; Wu, C.; Wang, Y.; Xie, C.; Shi, M.; Nile, S.; Zhou, Z.; Kai, G. Transcription factor OpWRKY3 is involved in the development and biosynthesis of camptothecin and its precursors in Ophiorrhiza pumila hairy roots. Int. J. Mol. Sci. 2019, 20, 3996. [Google Scholar] [CrossRef] [Green Version]
  83. Gutierrez-Camino, Á.; Umerez, M.; Martin-Guerrero, I.; García de Andoin, N.; Santos, B.; Sastre, A.; Echebarria-Barona, A.; Astigarraga, I.; Navajas, A.; Garcia-Orad, A. Mir-pharmacogenetics of Vincristine and peripheral neurotoxicity in childhood B-cell acute lymphoblastic leukemia. Pharm. J. 2018, 18, 704–712. [Google Scholar] [CrossRef]
  84. Setty, B.A.; Stanek, J.R.; Mascarenhas, L.; Miller, A.; Bagatell, R.; Okcu, F.; Nicholls, L.; Lysecki, D.; Gupta, A.A. VIncristine, irinotecan, and temozolomide in children and adolescents with relapsed rhabdomyosarcoma. Pediatr. Blood Cancer 2018, 65, e26728. [Google Scholar] [CrossRef]
  85. Büyükkapu Bay, S.; Kebudi, R.; Görgün, O.; Zülfikar, B.; Darendeliler, E.; Çakır, F.B. Vincristine, irinotecan, and temozolomide treatment for refractory/relapsed pediatric solid tumors: A single center experience. J. Oncol. Pharm. Pract. 2019, 25, 1343–1348. [Google Scholar] [CrossRef]
  86. Schönning, A.; Karlén, J.; Frisk, T.; Heyman, M.; Svahn, J.E.; Øra, I.; Kawan, L.; Holmqvist, B.M.; Björklund, C.; Harila-Saari, A.; et al. Venous thrombosis in children and adolescents with Hodgkin lymphoma in Sweden. Thromb. Res. 2017, 152, 64–68. [Google Scholar] [CrossRef]
  87. Batra, S.; Wistinghausen, B. Pediatric Cancer: Solid Tumors and Lymphoma. In Mount Sinai Expert Guides; John Wiley & Sons, Inc.: Hoboken, NJ, USA, 2019; pp. 543–555. [Google Scholar]
  88. Schiller, G.J.; Damon, L.E.; Coutre, S.E.; Hsu, P.; Bhat, G.; Douer, D. High-dose vincristine sulfate liposome injection, for advanced, relapsed, or refractory philadelphia chromosome-negative acute lymphoblastic leukemia in an adolescent and young adult subgroup of a phase 2 clinical trial. J. Adolesc. Young Adult Oncol. 2018, 7, 546–552. [Google Scholar] [CrossRef] [PubMed]
  89. Tomioka, K.; Nishikawa, J.; Moriwaki, J.; Sato, T. Effects of over-expression of strictosidine synthase and tryptophan decarboxylase on alkaloid production by cell cultures of Catharanthus roseus. J. Gen. Plant Pathol. 2013, 79, 414–419. [Google Scholar]
  90. Sazegari, S.; Niazi, A.; Shahriari-Ahmadi, F.; Moshtaghi, N.; Ghasemi, Y. CrMYC1 transcription factor overexpression promotes the production of low abundance terpenoid indole alkaloids in Catharanthus roseus. Plant OMICS 2018, 11, 30–36. [Google Scholar] [CrossRef]
  91. Pan, Q.; Wang, C.; Xiong, Z.; Wang, H.; Fu, X.; Shen, Q.; Peng, B.; Ma, Y.; Sun, X.; Tang, K. CrERF5, an AP2/ERF transcription factor, positively regulates the biosynthesis of bisindole alkaloids and their precursors in Catharanthus roseus. Front. Plant Sci. 2019, 10, 1–13. [Google Scholar] [CrossRef] [Green Version]
  92. Othman, L.; Sleiman, A.; Abdel-Massih, R.M. Antimicrobial activity of polyphenols and alkaloids in middle eastern plants. Front. Microbiol. 2019, 10, 911. [Google Scholar] [CrossRef]
  93. Lelario, F.; Scrano, L.; de Franchi, S.; Bonomo, M.G.; Salzano, G.; Milan, S.; Milella, L.; Bufo, S.A. Identification and antimicrobial activity of most representative secondary metabolites from different plant species. Chem. Biol. Technol. Agric. 2018, 5, 1–12. [Google Scholar] [CrossRef]
  94. Radulovic, N.S.; Blagojevic, P.D.; Stojanovic-Radic, Z.Z.; Stojanovic, N.M. Antimicrobial Plant Metabolites: Structural Diversity and Mechanism of Action. Curr. Med. Chem. 2013, 20, 932–952. [Google Scholar]
  95. Khameneh, B.; Iranshahy, M.; Soheili, V.; Fazly Bazzaz, B.S. Review on plant antimicrobials: A mechanistic viewpoint. Antimicrob. Resist. Infect. Control 2019, 8, 118. [Google Scholar] [CrossRef] [Green Version]
  96. Çetinkaya, R.A.; Yenilmez, E.; Petrone, P.; Yılmaz, S.; Bektöre, B.; Şimsek, B.; Kula Atik, T.; Özyurt, M.; Ünlü, A. Platelet-rich plasma as an additional therapeutic option for infected wounds with multi-drug resistant bacteria: In vitro antibacterial activity study. Eur. J. Trauma Emerg. Surg. 2019, 45, 555–565. [Google Scholar] [CrossRef]
  97. Mahmoud, M.; Askora, A.; Barakat, A.B.; Rabie, O.E.F.; Hassan, S.E. Isolation and characterization of polyvalent bacteriophages infecting multi drug resistant Salmonella serovars isolated from broilers in Egypt. Int. J. Food Microbiol. 2018, 266, 8–13. [Google Scholar] [CrossRef]
  98. Ghimire, B.K.; Seong, E.S.; Yu, C.Y.; Kim, S.H.; Chung, I.M. Evaluation of phenolic compounds and antimicrobial activities in transgenic Codonopsis lanceolata plants via overexpression of the γ-tocopherol methyltransferase (γ-tmt) gene. S. Afr. J. Bot. 2017, 109, 25–33. [Google Scholar] [CrossRef]
  99. Ghimire, B.K.; Yu, C.Y.; Chung, I.M. Assessment of the phenolic profile, antimicrobial activity and oxidative stability of transgenic Perilla frutescens L.overexpressing tocopherol methyltransferase (γ-tmt) gene. Plant Physiol. Biochem. 2017, 118, 77–87. [Google Scholar] [CrossRef] [PubMed]
  100. Sitarek, P.; Kowalczyk, T.; Rijo, P.; Białas, A.J.; Wielanek, M.; Wysokińska, H.; Garcia, C.; Toma, M.; Śliwiński, T.; Skała, E. Over-Expression of AtPAP1 Transcriptional Factor Enhances Phenolic Acid Production in Transgenic Roots of Leonurus sibiricus L. and Their Biological Activities. Mol. Biotechnol. 2018, 60, 74–82. [Google Scholar] [CrossRef] [PubMed]
  101. Sitarek, P.; Kowalczyk, T.; Santangelo, S.; Białas, A.J.; Toma, M.; Wieczfinska, J.; Śliwiński, T.; Skała, E. The Extract of Leonurus sibiricus Transgenic Roots with AtPAP1 Transcriptional Factor Induces Apoptosis via DNA Damage and Down Regulation of Selected Epigenetic Factors in Human Cancer Cells. Neurochem. Res. 2018, 43, 1363–1370. [Google Scholar] [CrossRef] [Green Version]
  102. Sitarek, P.; Kowalczyk, T.; Picot, L.; Michalska-Hejduk, D.; Bijak, M.; Białas, A.J.; Wielanek, M.; Śliwiński, T.; Skała, E. Growth of Leonurus sibiricus L. roots with over-expression of AtPAP1 transcriptional factor in closed bioreactor, production of bioactive phenolic compounds and evaluation of their biological activity. Ind. Crops Prod. 2018, 122, 732–739. [Google Scholar] [CrossRef]
  103. Sitarek, P.; Synowiec, E.; Kowalczyk, T.; Śliwiński, T.; Skała, E. An in vitro estimation of the cytotoxicity and genotoxicity of root extract from Leonurus sibiricus L. overexpressing AtPAP1 against different cancer cell lines. Molecules 2018, 23, 2049. [Google Scholar] [CrossRef] [Green Version]
  104. Zalesińska, M.D.; Drąg, M.; Poręba, M.; Borska, S.; Kulbacka, J. Anticancer properties of ester derivatives of betulin in human metastatic melanoma cells (Me-45). Cancer Cell Int. 2017, 17, 1–7. [Google Scholar]
  105. Suzuki, H.; Fukushima, E.O.; Shimizu, Y.; Seki, H.; Fujisawa, Y.; Ishimoto, M.; Osakabe, K.; Osakabe, Y.; Muranaka, T. Lotus japonicus Triterpenoid Profile and Characterization of the CYP716A51 and LjCYP93E1 Genes Involved in Their Biosynthesis in Planta. Plant Cell Physiol. 2019, 60, 2496–2509. [Google Scholar] [CrossRef]
  106. Xia, K.; Liu, X.; Zhang, Q.; Qiang, W.; Guo, J.; Lan, X.; Chen, M.; Liao, Z. Promoting scopolamine biosynthesis in transgenic Atropa belladonna plants with pmt and h6h overexpression under field conditions. Plant Physiol. Biochem. 2016, 106, 46–53. [Google Scholar] [CrossRef]
  107. Sohrabi, S.M.; Ismaili, A.; Nazarian-Firouzabadi, F. Simultaneous over-expression and silencing of some benzylisoquinoline alkaloid biosynthetic genes in opium poppy. Ind. Crops Prod. 2018, 123, 581–590. [Google Scholar] [CrossRef]
  108. Li, Q.; Yu, H.M.; Meng, X.F.; Lin, J.S.; Li, Y.J.; Hou, B.K. Ectopic expression of glycosyltransferase UGT76E11 increases flavonoid accumulation and enhances abiotic stress tolerance in Arabidopsis. Plant Biol. 2018, 20, 10–19. [Google Scholar] [CrossRef]
  109. Lee, W.J.; Kim, J.; Lee, D.; Hong, S.W.; Lee, H. Arabidopsis UDP-glycosyltransferase 78D1-overexpressing plants accumulate higher levels of kaempferol 3-O-β-d-glucopyranoside than wild-type plants. Appl. Biol. Chem. 2017, 60, 647–652. [Google Scholar] [CrossRef]
  110. 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]
  111. Wang, F.; Ren, G.; Li, F.; Qi, S.; Xu, Y.; Wang, B.; Yang, Y.; Ye, Y.; Zhou, Q.; Chen, X. A chalcone synthase gene AeCHS from Abelmoschus esculentus regulates flavonoid accumulation and abiotic stress tolerance in transgenic Arabidopsis. Acta Physiol. Plant. 2018, 40, 1–13. [Google Scholar] [CrossRef]
  112. Wang, F.; Zhu, H.; Kong, W.; Peng, R.; Liu, Q.; Yao, Q. The Antirrhinum AmDEL gene enhances flavonoids accumulation and salt and drought tolerance in transgenic Arabidopsis. Planta 2016, 244, 59–73. [Google Scholar] [CrossRef]
  113. Park, S.C.; Kim, Y.H.; Kim, S.H.; Jeong, Y.J.; Kim, C.Y.; Lee, J.S.; Bae, J.Y.; Ahn, M.J.; Jeong, J.C.; Lee, H.S.; et al. Overexpression of the IbMYB1 gene in an orange-fleshed sweet potato cultivar produces a dual-pigmented transgenic sweet potato with improved antioxidant activity. Physiol. Plant. 2015, 153, 525–537. [Google Scholar] [CrossRef]
  114. Park, S.C.; Kim, S.H.; Park, S.; Lee, H.U.; Lee, J.S.; Park, W.S.; Ahn, M.J.; Kim, Y.H.; Jeong, J.C.; Lee, H.S.; et al. Enhanced accumulation of carotenoids in sweetpotato plants overexpressing IbOr-Ins gene in purple-fleshed sweetpotato cultivar. Plant Physiol. Biochem. 2015, 86, 82–90. [Google Scholar] [CrossRef]
  115. Mierziak, J.; Wojtasik, W.; Kostyn, K.; Czuj, T.; Szopa, J.; Kulma, A. Crossbreeding of transgenic flax plants overproducing flavonoids and glucosyltransferase results in progeny with improved antifungal and antioxidative properties. Mol. Breed. 2014, 34, 1917–1932. [Google Scholar] [CrossRef] [Green Version]
  116. Huang, W.; Khaldun, A.B.M.; Chen, J.; Zhang, C.; Lv, H.; Yuan, L.; Wang, Y. A R2R3-MYB transcription factor regulates the flavonol biosynthetic pathway in a traditional Chinese medicinal plant, Epimedium sagittatum. Front. Plant Sci. 2016, 7, 1–12. [Google Scholar] [CrossRef] [Green Version]
  117. Jeong, Y.J.; An, C.H.; Woo, S.G.; Park, J.H.; Lee, K.W.; Lee, S.H.; Rim, Y.; Jeong, H.J.; Ryu, Y.B.; Kim, C.Y. Enhanced production of resveratrol derivatives in tobacco plants by improving the metabolic flux of intermediates in the phenylpropanoid pathway. Plant Mol. Biol. 2016, 92, 117–129. [Google Scholar] [CrossRef]
  118. Shi, J.; Li, W.; Gao, Y.; Wang, B.; Li, Y.; Song, Z. Enhanced rutin accumulation in tobacco leaves by overexpressing the NtFLS2 gene. Biosci. Biotechnol. Biochem. 2017, 81, 1721–1725. [Google Scholar] [CrossRef] [Green Version]
  119. Yuan, Y.; Qi, L.; Yang, J.; Wu, C.; Liu, Y.; Huang, L. A Scutellaria baicalensis R2R3-MYB gene, SbMYB8, regulates flavonoid biosynthesis and improves drought stress tolerance in transgenic tobacco. Plant Cell Tissue Organ Cult. 2015, 120, 961–972. [Google Scholar] [CrossRef]
  120. Gwak, Y.S.; Han, J.Y.; Adhikari, P.B.; Ahn, C.H.; Choi, Y.E. Heterologous production of a ginsenoside saponin (compound K) and its precursors in transgenic tobacco impairs the vegetative and reproductive growth. Planta 2017, 245, 1105–1119. [Google Scholar] [CrossRef]
  121. Liu, Y.; Wang, L.; Zhang, J.; Yu, B.; Wang, J.; Wang, D. The MYB transcription factor StMYBA1 from potato requires light to activate anthocyanin biosynthesis in transgenic tobacco. J. Plant Biol. 2017, 60, 93–101. [Google Scholar] [CrossRef]
  122. Sun, W.; Meng, X.; Liang, L.; Li, Y.; Zhou, T.; Cai, X.; Wang, L.; Gao, X. Overexpression of a Freesia hybrida flavonoid 3-O-glycosyltransferase gene, Fh3GT1, enhances transcription of key anthocyanin genes and accumulation of anthocyanin and flavonol in transgenic petunia (Petunia hybrida). Vitro Cell. Dev. Biol. Plant 2017, 53, 478–488. [Google Scholar] [CrossRef]
  123. Ai, T.N.; Naing, A.H.; Yun, B.W.; Lim, S.H.; Kim, C.K. Overexpression of rsmyb1 enhances anthocyanin accumulation and heavy metal stress tolerance in transgenic petunia. Front. Plant Sci. 2018, 9, 1–15. [Google Scholar] [CrossRef] [Green Version]
  124. Li, H.; Liu, J.; Pei, T.; Bai, Z.; Han, R.; Liang, Z. Overexpression of SmANS enhances anthocyanin accumulation and alters phenolic acids content in Salvia miltiorrhiza and Salvia miltiorrhiza bge f. Alba plantlets. Int. J. Mol. Sci. 2019, 20, 2225. [Google Scholar] [CrossRef] [Green Version]
  125. Liu, Y.; Sun, G.; Zhong, Z.; Ji, L.; Zhang, Y.; Zhou, J.; Zheng, X.; Deng, K. Overexpression of AtEDT1 promotes root elongation and affects medicinal secondary metabolite biosynthesis in roots of transgenic Salvia miltiorrhiza. Protoplasma 2017, 254, 1617–1625. [Google Scholar] [CrossRef]
  126. Yang, N.; Zhou, W.; Su, J.; Wang, X.; Li, L.; Wang, L.; Cao, X.; Wang, Z. Overexpression of smMYC2 increases the production of phenolic acids in Salvia miltiorrhiza. Front. Plant Sci. 2017, 8, 1–12. [Google Scholar] [CrossRef] [Green Version]
  127. Wang, B.; Niu, J.; Li, B.; Huang, Y.; Han, L.; Liu, Y.; Zhou, W.; Hu, S.; Li, L.; Wang, D.; et al. Molecular characterization and overexpression of SmJMT increases the production of phenolic acids in Salvia miltiorrhiza. Int. J. Mol. Sci. 2018, 19, 3788. [Google Scholar] [CrossRef] [Green Version]
  128. Lim, W.; Li, J. Co-expression of onion chalcone isomerase in Del/Ros1-expressing tomato enhances anthocyanin and flavonol production. Plant Cell Tissue Organ Cult. 2017, 128, 113–124. [Google Scholar] [CrossRef]
  129. Jian, W.; Cao, H.; Yuan, S.; Liu, Y.; Lu, J.; Lu, W.; Li, N.; Wang, J.; Zou, J.; Tang, N.; et al. SlMYB75, an MYB-type transcription factor, promotes anthocyanin accumulation and enhances volatile aroma production in tomato fruits. Hortic. Res. 2019, 6, 22. [Google Scholar] [CrossRef] [Green Version]
  130. Kaushik, P. Standardisation of an Agroinfiltration Protocol for Eggplant Fruits and Proving its Usefulness by Over-expressing the SmHQT Gene. Preprints 2019. [Google Scholar] [CrossRef] [Green Version]
  131. Qiu, J.; Sun, S.; Luo, S.; Zhang, J.; Xiao, X.; Zhang, L.; Wang, F.; Liu, S. Arabidopsis AtPAP1 transcription factor induces anthocyanin production in transgenic Taraxacum brevicorniculatum. Plant Cell Rep. 2014, 33, 669–680. [Google Scholar] [CrossRef]
  132. Nomani, M.; Sadat Noori, S.A.; Tohidfar, M.; Ramshini, H. Overexpression of TPS2 gene to increase thymol content using Agrobacterium tumefaciens-mediated transformation in Trachyspermum ammi (Qom ecotype). Ind. Crops Prod. 2019, 130, 63–70. [Google Scholar] [CrossRef]
  133. Pandey, V.; Srivastava, R.; Akhtar, N.; Mishra, J.; Mishra, P.; Verma, P.C. Expression of Withania somnifera Steroidal Glucosyltransferase gene Enhances Withanolide Content in Hairy Roots. Plant Mol. Biol. Rep. 2016, 34, 681–689. [Google Scholar] [CrossRef]
  134. Yuan, Y.; Liu, W.; Zhang, Q.; Xiang, L.; Liu, X.; Chen, M.; Lin, Z.; Wang, Q.; Liao, Z. Overexpression of artemisinic aldehyde Δ11 (13) reductase gene-enhanced artemisinin and its relative metabolite biosynthesis in transgenic Artemisia annua L. Biotechnol. Appl. Biochem. 2015, 62, 17–23. [Google Scholar] [CrossRef]
  135. Inthima, P.; Nakano, M.; Otani, M.; Niki, T.; Nishijima, T.; Koshioka, M.; Supaibulwatana, K. Overexpression of the gibberellin 20-oxidase gene from Torenia fournieri resulted in modified trichome formation and terpenoid metabolities of Artemisia annua. Plant Cell Tissue Organ Cult. 2017, 129, 223–236. [Google Scholar] [CrossRef]
  136. Fan, G.; Nie, T.; Huang, Y.; Zhan, Y. GSNOR deficiency enhances betulin production in Betula platyphylla. Trees Struct. Funct. 2018, 32, 847–853. [Google Scholar] [CrossRef]
  137. Lu, S.; Zhang, Y.; Zhu, K.; Yang, W.; Ye, J.; Chai, L.; Xu, Q.; Deng, X. The citrus transcription factor CsMADS6 modulates carotenoid metabolism by directly regulating carotenogenic genes. Plant Physiol. 2018, 176, 2657–2676. [Google Scholar] [CrossRef] [Green Version]
  138. Mendoza-Poudereux, I.; Muñoz-Bertomeu, J.; Navarro, A.; Arrillaga, I.; Segura, J. Enhanced levels of S-linalool by metabolic engineering of the terpenoid pathway in spike lavender leaves. Metab. Eng. 2014, 23, 136–144. [Google Scholar] [CrossRef] [PubMed]
  139. Wang, Q.; Reddy, V.A.; Panicker, D.; Mao, H.Z.; Kumar, N.; Rajan, C.; Venkatesh, P.N.; Chua, N.H.; Sarojam, R. Metabolic engineering of terpene biosynthesis in plants using a trichome-specific transcription factor MsYABBY5 from spearmint (Mentha spicata). Plant Biotechnol. J. 2016, 14, 1619–1632. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  140. Kang, Y.M.; Park, D.J.; Lee, D.G.; Song, H.J.; Kang, S.M.; Min, J.Y.; Moon, B.C.; Lee, C.K.; Jeon, K.S.; Shivappakarigar, C.; et al. Over expression of IPP isomerase and limonene synthase enzymes in Mentha spicata and their influence on the terpenoid metabolism. Rom. Biotechnol. Lett. 2015, 20, 10358–10368. [Google Scholar]
  141. Hamachi, A.; Nisihara, M.; Saito, S.; Rim, H.; Takahashi, H.; Islam, M.; Uemura, T.; Ohnishi, T.; Ozawa, R.; Maffei, M.E.; et al. Overexpression of geraniol synthase induces heat stress susceptibility in Nicotiana tabacum. Planta 2019, 249, 235–249. [Google Scholar] [CrossRef] [PubMed]
  142. Rahimi, S.; Kim, Y.J.; Sukweenadhi, J.; Zhang, D.; Yang, D.C. PgLOX6 encoding a lipoxygenase contributes to jasmonic acid biosynthesis and ginsenoside production in Panax ginseng. J. Exp. Bot. 2016, 67, 6007–6019. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Jadaun, J.S.; Sangwan, N.S.; Narnoliya, L.K.; Singh, N.; Bansal, S.; Mishra, B.; Sangwan, R.S. Over-expression of DXS gene enhances terpenoidal secondary metabolite accumulation in rose-scented geranium and Withania somnifera: Active involvement of plastid isoprenogenic pathway in their biosynthesis. Physiol. Plant. 2017, 159, 381–400. [Google Scholar] [CrossRef]
  144. Wei, T.; Gao, Y.; Deng, K.; Zhang, L.; Yang, M.; Liu, X.; Qi, C.; Wang, C.; Song, W.; Zhang, Y.; et al. Enhancement of tanshinone production in Salvia miltiorrhiza hairy root cultures by metabolic engineering. Plant Methods 2019, 15, 1–11. [Google Scholar] [CrossRef]
  145. Deng, C.; Hao, X.; Shi, M.; Fu, R.; Wang, Y.; Zhang, Y.; Zhou, W.; Feng, Y.; Makunga, N.P.; Kai, G. Tanshinone production could be increased by the expression of SmWRKY2 in Salvia miltiorrhiza hairy roots. Plant Sci. 2019, 284, 1–8. [Google Scholar] [CrossRef]
  146. Vaccaro, M.; Malafronte, N.; Alfieri, M.; de Tommasi, N.; Leone, A. Enhanced biosynthesis of bioactive abietane diterpenes by overexpressing AtDXS or AtDXR genes in Salvia sclarea hairy roots. Plant Cell Tissue Organ Cult. 2014, 119, 65–77. [Google Scholar] [CrossRef]
  147. Tian, N.; Liu, F.; Wang, P.; Yan, X.; Gao, H.; Zeng, X.; Wu, G. Overexpression of BraLTP2, a lipid transfer protein of Brassica napus, results in increased trichome density and altered concentration of secondary metabolites. Int. J. Mol. Sci. 2018, 19, 1733. [Google Scholar] [CrossRef] [Green Version]
  148. Chahel, A.A.; Zeng, S.; Yousaf, Z.; Liao, Y.; Yang, Z.; Wei, X.; Ying, W. Plant-specific transcription factor LrTCP4 enhances secondary metabolite biosynthesis in Lycium ruthenicum hairy roots. Plant Cell Tissue Organ Cult. 2019, 136, 323–337. [Google Scholar] [CrossRef]
  149. Yamazaki, M.; Rai, A.; Yoshimoto, N.; Saito, K. Perspective: Functional genomics towards new biotechnology in medicinal plants. Plant Biotechnol. Rep. 2018, 12, 69–75. [Google Scholar] [CrossRef]
Figure 1. Schematic representation of binary and helper vectors used in plant genetic transformation.
Figure 1. Schematic representation of binary and helper vectors used in plant genetic transformation.
Plants 09 00132 g001
Table 1. Secondary metabolites derived from genetically-modified plant in vitro cultures with their biological properties.
Table 1. Secondary metabolites derived from genetically-modified plant in vitro cultures with their biological properties.
Plant SpeciesVector/Genetic ConstructPlant MaterialExtraction SolventClass of CompoundsEffectReferences
Atropa belladonna L.pXI vector containing NtPMT and HnH6HWhole plantmethanol and acetate acetate (methanol:50mM ammonium acetate = 58:42)AlkaloidsEnhanced biosynthesis of scopolamine[106]
Papaver somniferum L.pTRV2-BBE, pTRV2-COM, pTRV2-BBECOMLeavesMethanolAlkaloidsChanges in the different alkaloids content[107]
Arabidopsis thaliana (L.) Heynh.pBI121 vector containing UGT76E11SeedlingsMethanolPolyphenolsincreased content of flavonoid glycosides (kaempferol 3-O-[6″-O-(rhamnosyl)
glucoside] 7-O-rhamnoside kaempferol 3-O-glucoside 7-
O-rhamnoside, kaempferol 3-O-rhamnoside 7-O-rhamnoside
quercetin 3-O-rhamnoside 7-O-rhamnoside and
quercetin 3-O-glucoside 7-O-rhamnoside)
[108]
Arabidopsis thaliana (L.) Heynh.35Spro: AtUGT78D1SeedlingsMethanolPolyphenolsIncreased accumulation of flavonoids[109]
Arabidopsis thaliana (L.) Heynh.pCAMBIA1301-AtMYB12SeedlingsHCl-methanolPolyphenolsIncreased content of flavonoids[110]
Arabidopsis thaliana (L.) Heynh.pCAMBIA1301- AeCHSSeedlingsHCl-methanolPolyphenolsIncreased level of flavonoids[111]
Arabidopsis thaliana (L.) Heynh.pCAMBIA1301-AmDELSeedlingsHCl-methanolPolyphenolsIncreased level of flavonoids[112]
Ipomoea batatas
(L. Poir.)
pCam-SPO-IbMYB1aStorage rootMethanolPolyphenolsIncreased anthocyanin content[113]
Ipomoea batatas
(L. Poir.)
pGWB11 vector containing IbOrStorage rootHCl-methanolPolyphenolsEnhanced accumulation of zeaxanthin and β-carotene[114]
Leonurus sibiricus L.pCAMBIA1305.1-AtPAP1Hairy rootsMethanol-waterPolyphenolsHigher phenolic acid content. In addition, tested extracts with higher amounts of phenolic acids showed better antimicrobial and cytotoxic effect.[102]
Linum usitatissimum L.pBinARWhole plantsHCl-methanolPolyphenolsIncreased level of flavonoids[115]
Nicotiana benthamiana DominpMV-EsMYBF1FlowersHCl-methanolPolyphenolsIncreased production of flavonol content[116]
Nicotiana tabacum L.pGR-STS and pGR-ROSTWhole plant80% metanolPolyphenolsIncreased production of resveratrol derivatives (piceid, piceid methyl ether, resveratrol
methyl ether O-hexoside, and 5-methyl resveratrol-3,4-O-β-d-diglucopyranoside)
[117]
Nicotiana tabacum L.pK2GW7 vector containing NtFLS2LeavesMethanol-water-chloroform (5:2:2)PolyphenolsIncreased accumulation of rutin[118]
Nicotiana tabacum L.pCambia1305 containing SbMYB8LeavesEthyl alcoholPolyphenolshigher caffeoylquinic acid contents[119]
Nicotiana tabacum L.pZIP-Bar containing PgDDS, CYP716A47 and UGT71A28Leaves100% methanolPolyphenolsEnhanced production of ginsenoside saponin[120]
Nicotiana tabacum L.pSAK277 vector containing 35S:StMYBA1-1 constructLeavesHCl-methanolPolyphenols Enhanced anthocyanin accumulation[121]
Petunia x hybrida hort. ex E.VilmpBI-121 containing Fh3GT1Blooming flowersHCl-methanolPolyphenolsIncreased production of cyaniding, peonidin derivatives, kaempferol derivatives and quercetin derivatives[122]
Petunia x hybrida hort. ex E.VilmpB7WG2D vector containing RsMYB1LeavesHCl-methanolPolyphenolsEnhanced accumulation of flavonoids[123]
Salvia miltiorrhiza BungepCAMBIA2300 vector containing SmANSPlantletsHCl-methanolPolyphenolsIncreased anthocyanin accumulation, flavonols and proanthocyanidins biosynthesis[124]
Salvia miltiorrhiza BungepCB2006-EDT1Roots80% methanolPolyphenolsIncreased accumulation of salvianolic acids[125]
Salvia miltiorrhiza Bunge pEarleyGate201–SmMYC2Roots75% methanolPolyphenolsEnhanced production of hydrophilic phenolic acids[126]
Salvia miltiorrhiza BungepEarleyGate202-SmJMTRootsMethanol-acetone (7:3)PolyphenolsIncreased production of salvianolic and rosmarinic acids[127]
Solanum lycopersicum L.pE1775-CHI
pDEL.ROS
Flesh and peelHCl-methanolPolyphenolsEnhanced anthocyanins and flavonols[128]
Solanum lycopersicum L.K303 vector containing SlMYB75Fruits80% methanolPolyphenolsIncreased accumulation of anthocyanin, phenolics and flavonoids[129]
Solanum melongena L.pBIN19+SmHQT
pBIN19+p19
FruitsMethanol-water (80:20)PolyphenolsIncreased level of phenolic compounds[130]
Taraxacum brevicorniculatum Korol.pBI-AtPAP1LeavesMethanol and formic acidPolyphenolsIncreased production of anthocyanins, phenolic acids and flavonoids[131]
Trachyspermum ammi L. SpraguepBI121-TPLeaves80% ethanolPolyphenolsIncreased production of thymol[132]
Withania somnifera L.pYL436 vector containing Ws-Sgtl4Hairy rootsMethanolSteroidsIncreased withanolide and withanolide-A contents[133]
Artemisia annua L.pCAMBIA1305–DBR2LeavesMethanolTerpenoidsIncreased level of artemisinin[134]
Artemisia annua L.pIG-TfGA20ox2LeavesMethanolTerpenoidsIncreased production of artemisinin, sesquiterpenes. Eucalyptol, borneol, α-caryophyllene, β-guaiene, δ-cadinene and β-cubebene and isomultiflorenone were detected only in transgenic extract[135]
Betula platyphyllapSGRNAi-GSNORCell suspension or plantlet stemsEthanolTriterpenoidsIncreased betulin content[136]
Citrus grandis L.pK2- CsMADS6Calli and fruit-Terpenoidsincreased carotenoid contents[137]
Lavandula latifolia Medik.pBILISLeavesHexaneTriterpenoidsIncreased production of terpenes (S-linalool)[138]
Mentha spicata L.pK7WG2D- MsYABBY5LeavesEthyl acetateTriterpenoidsIncreased production of terpenes by gene silencing[139]
Mentha spicata L.pB1121 vector containing IPPWhole plants-TerpenoidsIncreased production of terpenoids[140]
Nicotiana tabacum L.pSKAN35SGESLeavesMethanolTriterpenoidsIncreased production of terpenes[141]
Panax ginseng CA MeyerpCAMBIA1390 vector containing PgLOX6Roots80% methanolTerpenoidsIncreased production of ginsenosides [142]
Pelargonium graveolens L’Her and Withania somnifera (L.) DunalpBI121 vector containing
GrDXS
Whole plants-TerpenoidsIncreased production of essential oil and withanolides [143]
Salvia miltiorrhiza BungepBI121 vector containing SmMDSHairy roots80% methanolTerpenoidsincreased accumulation of tanshinones (dihydrotanshinone I, cryptotanshinone, tanshinone I andtanshinone IIA)[144]
Salvia miltiorrhiza Bunge pCAMBIA2300sm-SmWRKY2Hairy rootsMethanol/di- chloromethane (3:1)TerpenoidsIncreased accumulation of tanshinones[145]
Salvia sclarea L.PKYLX71:35S vector containing DXS or DXRHairy rootsAcetoneTerpenoidsEnhanced biosynthesis of abietane diterpenes[146]
Brassica rapa L.pBI121S vector containing BraLTP2LeavesMethanol-waterDifferent metabolitesUpregulation of 43 different secondary metabolites.[147]
Lycium ruthenicum Murr.pCAMBIA1307-TCP4-OEHairy rootsMethanolDifferent metaboliteshigher relative abundances of different secondary metabolites[148]

Share and Cite

MDPI and ACS Style

Kowalczyk, T.; Wieczfinska, J.; Skała, E.; Śliwiński, T.; Sitarek, P. Transgenesis as a Tool for the Efficient Production of Selected Secondary Metabolites from Plant in Vitro Cultures. Plants 2020, 9, 132. https://doi.org/10.3390/plants9020132

AMA Style

Kowalczyk T, Wieczfinska J, Skała E, Śliwiński T, Sitarek P. Transgenesis as a Tool for the Efficient Production of Selected Secondary Metabolites from Plant in Vitro Cultures. Plants. 2020; 9(2):132. https://doi.org/10.3390/plants9020132

Chicago/Turabian Style

Kowalczyk, Tomasz, Joanna Wieczfinska, Ewa Skała, Tomasz Śliwiński, and Przemysław Sitarek. 2020. "Transgenesis as a Tool for the Efficient Production of Selected Secondary Metabolites from Plant in Vitro Cultures" Plants 9, no. 2: 132. https://doi.org/10.3390/plants9020132

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