Open access peer-reviewed chapter

Application of Tissue Culture Techniques to Improve the Productivity of Medicinal Secondary Products from Medicinal Plants

Written By

Ahmed M. Hassanein

Submitted: 20 April 2022 Reviewed: 05 May 2022 Published: 31 May 2022

DOI: 10.5772/intechopen.105193

From the Edited Volume

Case Studies of Breeding Strategies in Major Plant Species

Edited by Haiping Wang

Chapter metrics overview

348 Chapter Downloads

View Full Metrics

Abstract

The plant kingdom is considered the most important source of medicinal chemicals. In vitro culture techniques are being considered a promising alternative to traditional agricultural processes to improve medicinal plants multiplication and their production of pharmaceutical compounds. In this chapter, several in vitro culture strategies are discussed to improve secondary metabolites production, including (1) plant kingdom as a source of medicinal chemicals, (2) in vitro culture of medicinal plants, (3) culture media optimization, (4) application of suspension cell culture for production of secondary metabolites, (5) elicitation to enhance the productivity of the culture, (6) precursor intermediates feeding, (7) selection of high-yielding cell lines, (8) overexpression of genes that control the production of bioactive compounds, and (9) scale-up production. Also, challenges that hinder the in vitro culture of medicinal plants using different techniques and the use of those techniques to produce pharmaceutical compounds are discussed in this chapter, including (a) secondary metabolites toxicity, (b) low growth rate, (c) culture browning, (d) limitation in the application of transformation, (e) somaclonal variation, and (f) vitrification. Therefore, the principal objective of the current chapter was to shed light on the studies on some medicinal plants and the used protocols to overcome some difficulties in terms of in vitro propagation that maximize their economic values.

Keywords

  • In vitro plant culture
  • medicinal plants
  • pharmaceutical compounds
  • tissue culture
  • medicinal chemicals

1. Introduction

Unlike animals, plants do not have the ability to move, making them vulnerable to attack by pests and sometimes animals. To overcome this problem, plant tissues synthesize enormous compounds, such as terpenes, polyphenols, cardenolides, steroids, alkaloids, and glycosides, and use them as defense strategies [1]. These defense compounds are called secondary metabolites and are not necessary for essential plant functions, such as growth, photosynthesis, and reproduction. These compounds are accumulated in the plant body to use by man as pharmaceutical, agrochemicals, aromatics, and food additives [1, 2]. Despite the progress in synthetic chemistry, plants are considered the most successful sources of drugs due to their bioactive compounds produced through secondary metabolism pathways [2].

In industrialized and developing countries, raw plant materials and plant-derived pharmaceuticals have naturally an essential component of present-day human healthcare systems. A known fact is that over 80% of the human beans use herbal medicines for healthy living [3]. In this respect, at present, more than 40% of the used pharmaceuticals by Western countries are derivatives of natural resources [4]. Worldwide, man uses about 35000–70000 plant species to prevent and cure diseases, most of them are reported in China (10,000–11,250), India (7500), Mexico (2237), and others [5]. Quality assurance and standardization of herbal medicines during the collection, handling, processing, and production of herbal medicine are essential prerequisites to ensure safety for the global herbal market. Wild plant materials are collected from gardens, open pasture, or forest land. In some cases, medicinal plants grow like weeds on agricultural land. While the bulk of the medicinal plant materials is still wild-harvested, a very small number of plant species are cultivated commercially [6]. However, increase populations and urban growth were associated with an over-exploitation of natural resources. Unfortunately, several medicinal plant species are disappeared due to the expansion of land for the purpose of growing crops, urban expansion, uncontrolled deforestation, and intensive collection [7]. Now, the increase in demand for these compounds encouraged the cultivation of large areas of medicinal plants and the application of new technologies, such as plant tissue culture (PTC) to preserve them from extinction and improve their productivity in quality and quantity.

Manufacturing of medicinal products from soil-grown plants faces some challenges, such as: (1) The wild-targeted plant does not exist in sufficient abundance in the local environment or is rare in general, (2) Cultivation of the target plant may need certain conditions, (3) Production of the target substance may require to grow plants for a long time, (4) The target substance may present at low concentration in cultivated or harvested plants, (5) Variations in environmental conditions may result in the production of bioactive compounds at a non-homogeneous quantity or quality, (6) Collection of plants for pharmaceuticals may be unsafe, (7) Harvest of propagated medicinal plants for drug industries is time- and money-consuming [8]. To overcome all the obstacles, PTC techniques express the great potential for bioproduction of phytoconstituents of high therapeutic value. By application of artificial techniques, regulation of the biosynthetic pathway of the certain plant to enhance the production of valuable compounds or avoidance of production of an unwanted substance become possible.

With the aid of gene technology and molecular techniques, in vitro culture procedures, such as cell, organ or tissue culture, somatic embryogenesis, somatic hybridization, genetic transformation, hairy roots, and induction of somaclonal variation, and others can be applied to the improvement of bioactive compounds yields. For example, recombinant DNA technology can be used to direct metabolic pathways and produce pharmaceuticals, such as antibodies and hormones. These in vitro culture techniques are better than others where they are carried out under precisely controlled physical and chemical conditions. PTC techniques are a resolution for the propagation of seedless medicinal plants and others with small or unviable seeds that not be able to germinate in soil [9, 10]. In addition, PTC techniques hold significant promise for true to type, disease-free, rapid and mass multiplication, and plant development [11].

Application of PTC technologies in the medicinal plant does not free from problems but avoiding their problems can be precisely controlled, which makes in vitro cultivation an ideal alternative to produce medicinal compounds from plants [12]. One of the obstacles is that the prices of the products resulting from biotechnology are higher than other products resulting from cultivated or wild plants. In this concern, the application of large-scale PTC techniques have been found to be an attractive alternative tools to the traditional plantations, where they offer a controlled supply of secondary metabolites independent of plant availability and a more consistent product in quantity and quality [13]. In the last decade, to meet pharmaceutical industry demand and conserve natural sources, researchers concentrated their efforts on optimizing culture conditions for maximizing the obtained yield of targeted secondary metabolites by application of several artificial-developed techniques [14].

Through PTC techniques, a whole plant can be regenerated from an organ, small tissue, or a plant cell but it should carry out on a suitable culture medium and under a controlled environment [15]. Under these conditions, the obtained plantlets are true to type and show characteristics identical to the mother plant. On the other hand, the culture conditions can be controlled to stimulate genetic variation for plant improvement, but it requires the construction of a selection procedure to select an elite mutant. For several decades, in vitro culture techniques are being used increasingly as a supplement to traditional breeding tools for the modification and improvement of plants. For example, Coryodalis yanhusuo, an important medicinal plant was improved through the application of the somatic embryogenesis technique to produce disease-free lines [16]. While PTC can be established from any part of a plant, meristematic tissues, such as shoot tip or nodal segments, are usually recommended [15, 17, 18]. In addition, the physiological state of the donor plant affects strongly on regeneration ability of the cultured plant materials [9, 18].

The application of PTC techniques in the medicinal and other plant species becomes an essential prerequisite for plant propagation and improvement [15, 17]. The application of plant tissue culture has several advantages: (1) It results in the production of thousands of plantlets in a short period from a small segment of the tested plant. (2) It is a main procedure to obtain pathogen-free plants. (3) It can be used to culture plants round the year, irrespective of weather or season. (4) It needs little space for the propagation of the southlands of plants. (5) It can be used as the main procedure to produce a new cultivar of a certain plant. (6) It can be used to understand the effect of a specific biotic or abiotic factor on a tested plant beyond the interaction of other factors. (7) It helps to understand the molecular biology of plant differentiation. (8) It is an essential prerequisite during the production of genetically engineered plants. (9) It is an effective procedure for the production of pharmaceutical compounds. (10) It is an essential procedure for the preservation of endangered plant species, genetic assets, and gene banks.

Advertisement

2. Plant kingdom as a source of medicinal chemicals

Phytotherapy becomes a complementary and important part of pharmacotherapy and modern medicine. It is a type of treatment based on natural medicinal resources (drugs) and herbal remedies for the purposes of prevention and treatment of illness. Herbal drugs mean using the whole plant or part of it, fresh or dry, to treat or prevent human disease. Any plant part (flower, leaf, root, bark, fruit, and seed), resins, balsams, rubber, plant exudates, algae, fungi, or lichen can be used as herbal drugs for its medicinal properties. Herbal drugs or herbal remedies contain active ingredients of herbal medicinal products. The aerial plant parts, such as leaves, seeds, and flowers, are often able to synthesize and accumulate secondary metabolites more than those obtained by underground parts, such as roots or rhizomes [19]. For example, in Scrophularia kakudensis, the total phenol and flavonoid, as well as free radical scavenging compounds, were higher in shoot than root extract [20]. The variable contents of bioactive compounds in different plant tissues may be due to the specialized ability of each tissue to synthesize the bioactive ingredients or their ability to store them considering the physiological condition and endogenous hormone levels [19].

Based on their biosynthetic origins, reports classify the bioactive secondary metabolites of the plant into major groups, including phenolic compounds, terpenoids, nitrogen-containing alkaloids, and sulfur-containing compounds [21]. Phenolic compounds were the most important group where they are largely used to enhance human health and they naturally occur in fruits, vegetables, cereals, and beverages. Phenols are classified into different groups, including phenolic acids, flavonoids, stilbenes, and lignans, and they include apigenin, diosmin, quercetin, kaempferol, eriodictyol, naringenin, hesperetin, baicalein, chrysin, catechin, morin, genistein, curcumin, colchicine, resveratrol, and emodin. For the production and extraction of hundreds of these secondary products, plant cell, tissue, or organ cultures were used [21].

As a part of complementary and alternative medicine, medicinal plant extracts are widely used in chronic diseases like diabetes, hypertension, cancer, etc. Melatonin and serotonin, as antioxidants, were detected in the field and greenhouse-grown Ocimum sanctum L. plants [22]. Extract of in vitro cultures of Hovenia dulcis has antitumor effects [23]. Aegle marmelos can be used as antibacterial, antifungal, antidiabetic, and antioxidant [24]; it is also useful to treat several symptoms, such as stomachalgia, diarrhea, dysentery, malaria, and fever [25]. In vitro propagated Artemisia japonica was used to obtain antioxidant, insecticidal, antimalarial, antisporulant, antimicrobial, cytotoxic, and osteoinductive activities [26]. Acacetin (5,7-dihydroxy-4-methoxyflavone) has several therapeutic effects, it is found in more than 200 plant species belonging to 60 plant families especially Asteraceae and Lamiaceae families [27]. Acacetin is used for antiplasmodial, anticancerous, antidiabetic, antiperoxidative, antipyretic, anti-inflammatory, and antiproliferative activities [27]. Several compounds with anti-uveal melanoma activity were extracted from Acacia nilotica, including gallocatechin 5-O-gallate, methyl gallate, gallic acid, catechin 5-O-gallate, catechin, 1-O-galloyl-β-D-glucose, digallic acid, and 1,6-di-O-galloyl-β-D-glucose [28]. Biotechnological systems can be used to obtain vaccines from many plant species to provide immune protection against diseases [29]. Production of plant-based edible vaccines is mainly manipulated by the integration of the transgene into in vitro cultured plant cells to produce the antigen protein for specific diseases [30].

Screening of 346 methanol extracts of 281 native and cultivated plant species in Egypt indicated that Agave americana, A. lophantha, Furcraea selloa, Calotropis procera, Pergularia tomentosa, Asclepias sinaica, Alkanna orientalis, Khaya grandifoliola, Swietenia mahogani, Pimenta racemosa, Pinus canariensis, Verbascum sinaiticum, Solanum elaeagnifolium, S. nigrum, and Brachychiton rupestris have strong antischistosomal activity [31]. In addition, the antioxidant activity of the extract of 90 plants was determined by 2, 2 diphenyl-1-picrylhydrazyl (DPPH) assay [32], and extracts of some plant species expressed high antioxidant and cytotoxic activities that inhibited the growth of cancer cells [33]. Leaves of A. marmelos contain several medicinal compounds including π-sitosterol, lupeol, aegelin, rutin, flavone, glycoside, marmesinine, oisopentenyl halfordiol, phenylethyl cinnamides, and marmeline [24].

Advertisement

3. Application of in vitro culture techniques on medicinal plants

Plant tissue culture is the most promising savior of medicinal plants that face problems of low yield and susceptibility to biotic or abiotic stress. Also, PTC can be used for in situ and ex situ conservation, propagation, polyploidy or aneuploidy induction, plant engineering, and bioreactor applications. In vitro multiplication was established in many threatened and endemic medicinal plants, such as Bacopa monnieri [34], Paedaria foetida [35], Picrorhiza kuroa [36], Salvadora persica [37], Potentilla fulgens [38], Eryngium foetidum [39], and H. dulcis [40].

High multiplication using seedling tissues or shoot meristems was achieved in several plant species, such as Citrullus colocynthis [41], Zephyranthes bulbous [42], Plectranthus vetiveroids [43], Glossocardia bosvallea [44], Cannabis sativa [45], O. sanctum L. [22], Caralluma retrospeciens [46], Solanum nigrum [15], Moringa oliefera [47], Pulicaria incisa [48], Rosa damascena [49], A. marmelos [50], Artemisia judaica [51], and Hyoscyamus muticus [52].

For long-term storage of medicinal plant materials, cryopreservation is recommended where it is carried out in liquid nitrogen (−196°C). Different plant organs or parts, including seeds, corms, bulbs, rhizomes, roots, tubers, buds, and cuttings, can be stored for conservation purposes [11], especially in medicinal plants with recalcitrant seeds. The main applied techniques of cryopreservation of medicinal plants are vitrification, desiccation, and encapsulation–dehydration. Vitrification-cryopreservation of shoot tips of Dioscorea floribunda medicinal plant indicated that the genome of cryopreserved shoot tips was stable upon application of molecular, morphological, and biochemical procedures [53]. Vitrification–encapsulation–dehydration techniques of Dioscorea deltoidei medicinal plant shoot tips proved that the secondary metabolites of cryopreserved shoot tips were like control plants [54].

PTC is more efficient than naturally grown plant materials to assess the effect of different experimental conditions on the production of secondary metabolites of medicinal plants [55]. PTC opens the way for the production of engineered molecules and produces new forms of plant secondary metabolites [56]. These new forms of compounds may have a valuable effect on biological control, food, pharmaceutical, and other strategies. Transformation techniques are widely dependent on PTC for enhancing the in vitro production of valuable plant secondary metabolites [57].

Different types of PTC techniques are successfully exploited for in vitro propagation as well as synthesis and extraction of secondary metabolites [12]. Sometimes root culture is recommended because it provides valuable biomass in a short time and stable metabolite productivity. In addition, root cultures express genetic stability for long-term culture compared to other forms of in vitro cultures, such as cell aggregates and rhizoids. Roots are fully organized plant organ, ensures biochemical stability, and usually express the full biosynthetic capacity as same as soil-grown plant root. In vitro root cultures could be a better alternative for the accumulation of elevated contents of secondary metabolites. For example, root cultures of Hemidesmus indicus were used as a tool for in vitro production of 2-hydroxy 4-methoxy benzaldehyde [11, 58].

In vitro-produced hairy roots are formed without connection with any other plant organs. Then, the synthesized metabolites are not transported to other plant parts and are accumulated where they are synthesized. The produced secondary metabolites may be present in minor, undetectable quantities in vivo but they are present in higher levels in hairy roots due to the optimized culture conditions (14). Consequently, mass production of secondary compounds in the bioreactor was established using hairy root cultures [59].

Advertisement

4. Application of in vitro culture techniques for the production of pharmaceuticals

The synthetic capacity of secondary metabolites of the dedifferentiated tissue often differs substantially from that of differentiated one, both quantitatively and qualitatively. The differences in synthetic capacities are a direct response to differences in enzyme patterns between differentiated and undifferentiated tissues, they are mirrors for gene expression of these tissues. The culture of differentiated plant materials often shows biochemical and genetic stability, it offers a high-productivity system that does not need wide-ranging optimization. For example, the major alkaloid (vindoline) is scarcely produced by Catharanthus roseus suspension cultures but shoot cultures produce it in high quantity [60]. In addition, while the callus culture of Taraxacum officinale synthesizes and accumulates α and γ-amyrins, differentiated tissue synthesizes and accumulates taraxasterol and lupeol [61]. The previous studies indicate that different classes of secondary metabolites need different phases of cell or tissue differentiation.

Generally, in vitro conditions can be easily modulated to enhance the synthesis of secondary metabolites through modulation of the pathway of primary metabolism in plants. The in vitro obtained compounds are important as dyes, drugs, cosmetics, flavors, food additives, perfumes, agrochemicals, etc. Some of these compounds, such as flavors, fragrances, and colorants, cannot be produced by microbial cells or chemically synthesized but they can be synthesized by plant cell culture systems [62]. Several reports indicated that in vitro cultures were found to be more efficient than whole plants for the formation of bioactive secondary metabolites such as ajmalicine, ajmaline, anthraquinones, benzylisoquinoline alkaloids, berberine, bisoclaurine, coniferin, diosgenin, ginseng, ginsenoside, glutathione, nicotine, rosmarinic acid, raucaffricine, shikonin, taxol, terpentine, tripdiolide, and ubiquinone-10 [1, 2, 14, 62].

Under aseptic conditions, cultured plant materials can be used to generate bioactive or secondary metabolites, including flavonoids, alkaloids and other phenolics, terpenoids, saponins, steroids, tannins, glycosides, colorants, fragrances, and volatile oils [14]. Production of high-value active secondary metabolites at industrial levels, such as shikonin, berberine, and sanguinarine, was fulfilled from cell cultures of Lithospermum erythrorhizon, Coptis japonica, and Papaver somniferum, respectively [63]. Secondary bioactive metabolites in in vitro cultured Swertia chirayita were higher than in vivo plants [2]. The more antimicrobial property of the in vitro regenerated plant products was related to more bioactive metabolites. In addition, Manivannan et al. [20] reported that since the contents of phytochemicals in seed and in vitro derived plants were similar, the in vitro plantlets can be used as alternate for the seed grown plants for the production of bioactive metabolites. Also, acacetin (an individual flavonoid) was slightly increased in in vitro grown plantlets than that of in vivo grown plants due to the artificial conditions of the in vitro culture and modulation of endogenous hormone [20].

Pharmaceutical compounds that are obtained from in vitro cultured plant materials may be more easily extracted and purified due to the absence of significant amounts of pigments, thus resulting in lower manufacturing expenses [64]. Control of the production of secondary metabolites can be carried out using in vitro culture techniques. For example, low biomass and hypericin production of Hypericum perforatum shoots was improved by prolonging the time of culture for more than 30 days [65].

Advertisement

5. Strategies are used to improve secondary metabolites production

The biosynthesis of secondary metabolites using unorganized cultured cells or organized organs, such as roots, can be enhanced by altering the environmental conditions or selecting an elite variant clone [66]. There are many procedures that can be controlled to increase the productivity of in vitro cultured medicinal plants from the active substances with medicinal effects, and this is what will be discussed in this chapter.

5.1 Culture media optimization

To understand factors that control the biosynthesis of pharmaceutical compounds by cultured plant materials, studies on gene expression, enzyme activity, and signal transductions were carried out [12, 14]. The establishment of desired productivity of the PTC needs optimization of overall culture conditions to enhance both culture biomass and metabolites productivity. For example, while sulfate and ammonium nitrate ions increased the colchicine content of Gloriosa superba callus, a higher concentration of phosphate and calcium decreased alkaloid biosynthesis [67]. The differences in the composition of various PTC media formulations affect on water potential of the cultural environment [68]. Then, different media exerted different values of water potential. In vitro culture of certain medicinal plant materials on different media expresses different values of biomass and secondary metabolites. Medium selection is a major step in optimizing the culture conditions to produce an abundance of plant matter capable of producing an abundance of biological compounds [69]. Through media optimization of the in vitro cultured medicinal plants, the chemical composition is changed, the content of toxic compounds is reduced and novel chemical compounds may be formed [70]. In general, media optimization is an essential prerequisite to enhancing the production of antioxidants and other valuable secondary metabolites, it means that plant growth regulators and specific additives should be modulated to enhance in vitro production of biomass and secondary metabolites [71].

When nodal segments of Ocimum basilicum were cultured under the influence of different culture media including MS medium in different strengths and different combinations of PGRs, they expressed different values of methyl eugenol, linalool, and 1,8-cineole fractions [71]. Nodal segments are of Cunila menthoides medicinal plant cultured on MS medium containing different concentrations of PGRs resulting in biosynthesis of phenols, alkaloids, and terpenes in regenerated plants [71]. Media containing different types and concentrations of PGRs express different differentiation pathways and biomass values [72, 73], and it was associated with the expression of different types and concentrations of pharmaceuticals in cultured plant materials [74]. Contents of bioactive compounds in embryogenic callus and regenerated shoots of Rosa rugosa petal explants were influenced by PGRs type, concentration, and the nitrogen source [75]. Also, in Chonemorpha fragrance, the amount of synthesized camptothecin was influenced by the PGRs type and concentrations [76].

The effect of carbon source concentration and type on culture biomass and metabolites productivity should be investigated. To enhance the biomass and biosynthesize of secondary metabolites, sucrose is widely used as a carbon source and it was better than maltose, glucose, and others [77]. During in vitro propagation, the optimal concentration of sucrose depends on plant species [15, 47, 68, 73]. For example, feeding the culture medium with 60 mM nitrogen and rise sucrose concentration from 3% sucrose to 5% increase the biomass production and camptothecin accumulation by 2.4-fold in the cell suspension cultures of Nicotiana nimmoniana [77]. In Panax vietnamensis, 2–5% sucrose enhanced the biomass and ginsenoside production in the cell suspension but 6–7% sucrose inhibited ginsenoside accumulation [78]. Geraniol production in transgenic tobacco cell suspension cultures was influenced by several culture conditions including carbon source light, and inoculums size [14].

Physical culture conditions can also affect the ability of in vitro cultured plants for the production of secondary metabolites. For example, light as one of these physical conditions can affect strongly on the production of secondary metabolites in Abelmoschus esculentus [79]. Culture media pH is an essential factor for the production of valuable plant material mass and its content of secondary metabolites. The optimum pH for normal plant tissue cultures is 5.8 but it should be changed if the purpose of the culture is to produce bioactive compounds. In a comparative study by Hagendoom et al. [80], on different plant species, they detected a positive correlation between acidification of the cytoplasm and the accumulation of different secondary metabolites including coniferin and lignin. In cell suspension of C. roseus, an increase in the pH of culture media between 4.3 and 9.0 was associated with a sharp increase in alkaloid production [81]. In general, low and high pH of the medium retard biomass and withanolide production in Withania somnifera cell culture [82], but the optimal pH was 4.5 for enhancing biomass production and Bacoside A formation [83].

In a scale-up production system, modulation the composition of the culture media is an essential prerequisite to enhance the production efficiency of a selected cell line, but long-term cultivation may lead to the reduction of the yield [64] due to an increase in somaclonal variation [84]. Consequently, genetic stability of the cultured plant materials should be established using determined indicators, such as molecular markers, stability of growth parameter index over extended subculture cycles, and metabolite production.

5.2 Suspension and callus cultures

Callus culture is an undifferentiated-unorganized mass obtained by cell division on cultured plant material on an agar medium. Then, calli are subcultured either for in vitro propagation through organogenesis or embryogenesis or used to establish suspension culture [85]. When callus in suitable texture is obtained on solid or semisolid agar medium and suspended in a specific liquid growth medium, the cells disperse and divide more and more producing cell suspensions. Then, cells can have faster and uniform growth rates associated with secondary metabolite production. Suspension cultures are the most widely employed PTC techniques in the production of secondary metabolites. When cells are grown in aqueous media to produce cell suspensions, some cells do not disperse in the medium and form tissue clumps, which disrupts growth and weakens the production of targeted secondary compounds. Suspension cultures are also amenable for growth in small and giant fermenters, but these cultures may show genetic and biochemical variation. Under selected conditions, exploitation of cell cultures capable of producing medicinal compounds at a level similar or superior to that of intact plants.

Callus culture itself is exploited to produce and study secondary compounds in many medicinal plant species [66]. For induction of callus formation, specific culture conditions should be established, which means that cultured cells divide and proliferate rapidly as long as the cultural environment has sufficient nutrients and suitable growth regulators. Conditions for callus induction and proliferation are not favorable for the production of secondary metabolites. For induction of secondary metabolites, calli culture conditions should be changed or transferred to a new medium with a different composition [11]. High yields of proteolytic enzymes from the callus tissue culture of Allium sativum L. on MS medium containing NAA and BAP were obtained [86].

The advantages of the application of suspension-cell cultures are obvious including: (1) The biomass production is usually more rapid than that of other in vitro culture types as well as a whole plant, (2) Chemical and physical conditions can be easily controlled allowing the production of certain pharmaceuticals throughout the year if necessary, (3) Producers can provide their products in a sustainable manner that does not depend on large areas and leave the arable land areas to grow other crops, (4) The size and quality of the product can be controlled according to the market demand, (5) Producers can select plant cell line that ensures or improve product quality, and (6) Producers can combine more than one method, which leads to the development of new products.

Application of specific cell lines and selective culture of that cell lines lead to the production of secondary compounds more than those obtained from original tissues and normal culture conditions [87]. The addition of plant growth regulators enhances the production of target secondary metabolites in several medicinal plant species [88]. Cell immobilization [89] and genetic makeup [90] can be optimized to enhance the synthesis of secondary compounds under in vitro culture conditions. Cultured cells can be immobilized to form aggregates to enhance secondary metabolite production [91]. Cell immobilization is achieved through growing cells as aggregates or using substances such as alginate or polyurethane foam cubes [92].

Two-phase cell suspension cultures establish a growth medium for maximizing cell biomass and production of naphthoquinone pigment in the first phase, but the second phase was established at the dark condition and room temperature with alkaline pH. These two phases system enhanced biomass production six-fold and optimized metabolite production in Arnebia sp. [93]. In suspension cultures of C. roseus, cultures produced up to 20 g DW L − 1 of biomass. In addition, two phases culture technique increased active cell biomass with 10 times higher indole alkaloids production in comparison to that of the one-phase culture [94]. Under dark conditions at 25°C for 40 days, the two phases of co-culture of Panax ginseng and Echiancea purpurea adventitious root in bioreactors containing MS medium supplemented with IBA (25 μM), sucrose (50 g L − 1), and methyl jasmonate (200 μM) as elicitor for 30 days enhanced the production of ginsenosides and caffeic acid derivatives [95].

5.3 Elicitation as an effective strategy to enhance the productivity of in vitro cultures

In vitro or in vivo cultured plants show physiological and morphological responses to physical, chemical, or microbial agents which are called elicitors. Therefore, elicitation describes any processes that induce or enhance the synthesis of secondary metabolites to ensure plant survival and competitiveness [96, 97]. During in vivo growth, plant secondary metabolites are elicited in plant cells in response to environmental stresses as a defensive strategy against the abiotic agent or invading pathogen [2]. Elicitation effectiveness depends on several parameters, some of them are related to elicitor agents themselves, and others are related to the elicited in vitro cultured plant materials. The elicitor-related effects include elicitor type, concentration, and exposure duration. Cultures’ age, cultivated line, medium composition, type, and concentration of growth regulators are essential parameters during the application of elicitation strategies. Hence, the application of factors, such as biotic or abiotic agents, that trigger the defense response in in vitro-cultivated plant materials enhanced the productivity of bioactive compounds [98].

Most of the used biotic elicitors are either exogenous or endogenous microbial agents but abiotic is a wide range of materials, mainly heavy metals [14, 99]. Methyl jasmonate, salicylic acid, yeast extract, chitosan, inorganic salts, UV radiation, or others can be used as elicitors to improve secondary metabolites production of the cultured plant materials [97, 100]. Citric acid, L-ascorbic acid, and casein hydrolysate were also used as elicitors to enhance the total phenolic content in the callus of Rosa damascene [49].

In the suspension culture of Mentha pulegium, when media were supplemented with yeast extract and salicylic acid, a significant increase of limonene, menthone, menthol, and α-pinene was detected [101]. Fifty different substances were detected in an in vitro cultured Anemia tomentosa upon jasmonic acid application, whereas 20 substances were only detected in wild-type plants [102]. Secondary metabolites production in callus, cell suspension, or hairy roots of Ammi majus L. were elicited by autoclaved lysate of cell suspension of Enterobacter sakazaki bacteria [103]. Anthraquinone production in Rubia akane cell culture was elicited by chitosan [104]. Genetically stable in vitro regenerated plants of Capparis spinosa were confirmed by RAPD analysis with a two-fold increase in flavonoid content than those of the wild plants when plants were regenerated under the influence of methyl jasmonate elicitor [105]. Elicitation of Ambrosia artemisiifolia hairy root cultures to produce thiorubrine A was dependent on cultures’ age as well as elicitor concentration and exposure time. Maximum of eight-fold thiorubrine A production was achieved when 16-day-old cultures were elicited with 50 mg l-1 vanadyl sulfate elicitor for 72 h [106].

Abiotic stresses for a given period can be used as an elicitor. Temperature, light parameters (intensity, photoperiod, and wavelength), and water potential of the medium influence the fresh and dry biomass [15] as well as the concentration of active metabolites [107]. Any factor that affects the water stress of the media should affect growth and bioactive compound synthesis. The profound change in the culture water potential due to the addition of NaCl, mannitol, or polyethylene glycol can elicit the production of secondary metabolites [107]. The relationship between abiotic-nutritional deficiency stress and enhancement of the production of secondary metabolites was reported [108]. Deficiencies of nitrogen, phosphate, potassium, sulfur, or magnesium increase the production of phenolic compound accumulation in different plant species [109], which may be due to oxidative stress and modulation of the expression of some genes [110]. The combination between target gene overexpression and elicitors increased the yield of secondary metabolites. Across studied plant species, elicitors promoted the yield of secondary metabolites from 1.0 to a maximum of 2230-fold [100]. Abiotic elicitors were applied to enhance growth and ginseng saponin biosynthesis in P. ginseng hairy roots [111].

Specific microorganisms can be used for elicitor purposes [112]. It takes place through the co-cultivation of plant cells with microorganisms. Compared to non-elicited control tissues, coculture of Aspergillus flavus with C. roseus resulted in increases in vinblastine (7.88%) and vincristine (15.5%) concentrations [112]. Cocultivation between microorganisms and cultured plant tissue should avoid conditions that stimulate microorganism toxic components [12].

5.4 Precursor feeding

Under perfect and controlled conditions, in vitro cultured plants not only have a higher metabolic rate than differentiated or soil-grown plants but also compressed biosynthesis cycles in shorter periods of time. In addition, the addition of precursors and elicitors plays an important role in promoting the secondary metabolism of cells and tissues grown under well-controlled industrial conditions (PTC). Precursor feeding is a strategy that is based on the assumption that if intermediates of bioactive molecules are added at the beginning or during, the in vitro culture period, they can serve as a substrate to improve the production of secondary metabolites in cultured plant materials. Precursors refer to any compounds that can be converted by the in vitro cultured plant materials into secondary metabolites through biosynthetic pathways [14, 113], and they depend on the type and concentration of precursor, and addition timing [114]. According to the World Health Organization definition any plant that contains a substance that can be used for medicinal use or as a precursor to synthesize new or semi-synthetic pharmaceuticals as a medicinal plant. The addition of alanine precursor was used to stimulate the biosynthesis of plumbagin in Plumbago indick when it was added to the root cultures on the 14th day of cultivation along with sequential addition of Diaion HP-20 36 h after it was fed, this increased the target output 14 times [115]. Phenylalanine precursor was needed for the biosynthesis of silymarin in hairy roots of Silymarin marianum [116] or the biosynthesis of podophyllotoxin in the cell suspension cultures of Podophyllum hexandrium [117]. Combining elicitation with chitosan and precursor feeding with squalene was used to produce 27.49 mg/g DW withanolides [118].

Feeding the culture medium with organic compounds, such as vitamins or amino acids enhanced in vitro production of many secondary compounds. In callus and cell suspension cultures of Centella asiatica, amino acid feeding enhanced the production of triterpenes and asiaticoside [96]. Also, valine, threonine, and isoleucine enhanced adhyperforin production in shoot cultures of Hyraceum perforatum [119]. Feeding the suspension cultures of C. roseus with L- tryptophane or L-glutamine resulted in the production of the highest value of cell mass and indole alkaloids production [120]. Feeding the culture medium of Spilanthes acmella with casein hydrolysate and L-phenylalanine promoted biomass and scopoletin production [121]. Feeding squalene into culture medium of C. asiatica calli promoted production of madecassoside and asiaticoside [96]. In Solanum lyratum cell cultures, feeding with sterols such as cholesterol, stigmasterol or mixed sterols promoted the biosynthesis of solasodine, solasonidine, and solanine without effect on culture biomass [122].

The yield of salidroside was improved by feeding Rhodiola genus plants with an appropriate concentration of precursors and elicitors such as precursors, phenylalanine, tyrosol, and tyrosine [123]. Tyrosol feeding (0.5 mM) expressed the most obvious effect on salidroside content in the cell suspension cultures of R. sachalinensis [124]. When feeding the culture medium with precursors promoted the production of secondary metabolites without biomass accumulation, it needs a combination between precursors and elicitors to overcome the obstacle. This strategy was used to enhance the biosynthesis of sennoside A and B in callus cultures of Cassia augustifolia [125].

5.5 High-yielding cell lines selection

Genetic diversity within medicinal plants has great importance and can be used for plant improvement and the selection of an elite line. The selection of high biomass and metabolite(s) producing cell lines plays an important role in optimizing the productivity of in vitro cultivated plant materials. The yield of biomass and active metabolites may vary within varieties, genotypes, or populations of plant species [See 14]. The genotype has direct effects on the ability of the plant to produce valuable biomass and pharmaceutical compounds. To avoid high coast, the genotype with high yield and secondary metabolites contents should be carefully selected. For example, wright selection of Pilocarpus microphyllus resulted in the production of pilocarpine content ranging from 16.3 to 235.9 μg g-1 in dry weight [126], it was 15 times higher than the content found in wild plants.

To get a high yield of metabolites, Briskin [127] described the biotechnological methods for the selection of high-yielding cell lines in medicinal plants by addressing several topics, including media components, elicitation, immobilization, physical stress, and transformation. This means that the identification and establishment of high producing and fast-growing in vitro cultures are essential prerequisites, especially when the target secondary metabolite content of the selected cell line should be high. Selecting the higher-yielding cell lines was the essential step for optimizing the production of the anticancer drugs camptothecin [128].

Qualitative and quantitative estimation of active metabolites may show variability depending on the spatial and temporal changes that may happen during the process. Variation in secondary metabolites yield may be due to their repression or losses before or during the extraction processes. Consequently, the determined secondary metabolite value may not exactly indicate the actual content of secondary metabolite in a given tissue or plant species. Nevertheless, quantitative and qualitative methods can be applied to select high-yielding cell lines [14]. Selection of the high-yielding lines can be established by exposing the population of plant materials to toxic inhibitors, biosynthetic precursors, or stressful environments and followed by selecting cells that show higher production of targeted components [2]. Selection can be carried out using callus, cell suspension, or through any other in vitro culture procedure. In this regard, the answers to the following questions must be quite clear: Does diversity occur naturally or by using chemical, physical or biological substances that help in mutation to produce genetic diversity from which it can be selected? What are the methods used to identify and isolate the most qualitatively and quantitatively productive line?

5.6 Overexpression of genes that control the production of bioactive compounds

The production of secondary metabolites is a metabolic process that is influenced by several physicochemical factors. These factors can be controlled and optimized in large-scale production. Traditional mutagenesis programs have been used by the pharmaceutical industry for yield improvement of medicinal plants. Recently, the development of recombinant DNA technology has provided new and effective tools to obtain elite strains with high content of secondary metabolites through overexpression of specific enzymes involved in their biosynthetic pathways aiming to increase the production levels and speed the metabolic processes [67, 96]. Consequently, plant genetics, recombinant DNA technologies, and PTC have developed to improve the ability of several medicinal plants to biosynthesize secondary metabolites efficiently.

To control the synthesis of certain natural products, the enzymes involved in the synthesis of these reactions and how they are influenced by in vitro culture conditions should be carefully determined. Niggeweg et al. [129] identified the enzymes that control the pathway of synthesis of an important bioactive compound through controlling these pathways. This control can be investigated on a gene expression and genome level [1] but it is not enough because it does not always give clear and specific information on the nature of the encoded enzyme that controls the intended reaction. Consequently, genomic studies have been used in combination with physiological and biochemical aspects to understand the biosynthetic pathways of specific secondary metabolites [1]. In this concern, metabolic engineering strategies concentrate on the stimulation of certain pathways over others by overexpressing certain genes.

Using PTC, key gene overexpression that involved in the biosynthetic of valuable biologically active compounds can be controlled leading to produce compounds in high quantity and quantity. For example, the overexpression of geranyl diphosphate synthase and geraniol synthase genes in C. roseus led to a significant improvement in plant production from monoterpene indole alkaloids of vinblastine and vincristine [130]. In periwinkle cell lines, overexpression of the strictosidine synthase (Str) gene resulted in tenfold activity than wild type leading to the accumulation of high content of ajmalicine, strictosidine, serpentine, tabersonine, and catharanthine [131]. Overexpressing tryptophan decarboxylase (Tdc) gene resulted in accumulation of TIAs (serpentine, catharanthine, strictosidine) more than wild type in transgenic cell suspension culture of periwinkle [132]. In addition, overexpression of H6H (hyoscyamine 6β-hydroxylase) from Hyoscyamus niger in Atropa belladonna hairy roots enhanced scopolamine production [133]. In addition, suppression of the rosmarinic acid synthase gene led to an increase in the plant content of 3,4-dihydroxyphenyllactic acid which led to improving the quality of rosmarinic acid in Salvia miltiorrhiza [134].

Bioactive secondary metabolites are under coordinated control of the biosynthetic genes, and transcription factors (TFs) play an important role in this regulation [135]. Transcriptional regulation means the change in gene expression levels by modulation of transcription rates. Studies on the regulation of the production of secondary metabolite pathways are focused on the regulation of structural genes through TFs [135]. For example, the expression of genes involved in TIAs (terpenoid indole alkaloids, such as vincristine and vinblastine) metabolic pathway is elicited by jasmonates, it is regulated biosynthesis of terpenoid indole alkaloid (TIAs) and artemisinin [135]. Jasmonate was demonstrated as a regulator of deacetylvindoline 4-O-acetyltransferase (DAT) expression [136]. Expressed DAT is involved in the biosynthesis of TIAs member-vindoline through transferring an acetyl group to deacetylvindoline for vindoline production. It was clear that most of the genes codded for TIA pathway enzymes are tightly regulated by specific TFs under the regulation of JAs but it is carried out in coordination with developmental growth stage and environmental factors [135].

TFs of TIA genes respond to JAs and/or other elicitors. In C. roseus a few TFs (CrORCA2, CrORCA3, CrBPF1, CrWRKY1, CrMYC1, and CrMYC2) have been characterized, two of them (ORCA2 and ORCA3) are positively influenced by JAs [137]. ORCA2 plays a critical role in the regulation of TIA metabolism where it regulates gene expression of both feeder pathways as well as STR and SGD, genes that codded for enzymes catalyzing the first two steps in biosynthesis of TIA [138]. In addition, ORCA3 overexpression resulted in the increase of some genes such as TDC, STR, and desacetoxyvindoline- 4-hydroxylase (D4H) leading to the accumulation of vinblastine and other metabolites in the TIA pathway [139]. Other TF such WRKY family that is induced by JAs is involved in TIA biosynthesis [140]. In Catharanthus hairy roots, overexpression of CrWRKY1 results in up-regulation of TIA pathway genes, especially the TDC gene. TF-CrWRKY1 binds the TDC promoter resulting in and trans-activation of the TDC promoter in Catharanthus cells [141]. Preferential expression of CrWRKY1 and its interaction with other TFs (including CrORCAs and CrMYCs) play an essential role in the accumulation of vinblastine in C. roseus [135].

5.7 Transformation

The genetic transformation was used as a powerful tool to improve the productivity of secondary metabolites. In general, Agrobacterium rhizogenes was used to transfer genes in several dicotyledonous plants where roots are formed at the site of infection; what is called “hairy roots.” Agrobacterium-mediated transformation technology may be better than direct gene transfer techniques including particle bombardment and electroporation [129]. Transformed hairy roots mimic the biochemical machinery of normal roots and are used to produce secondary metabolites where they are stable and have high productivity under growth regulators free culture [88]. Hairy roots transformed systems have great potential for commercial production of viable secondary metabolites and become a good alternative for raw plant materials.

Gene transfer using Agrobacterium can possibly be used to transfer DNA fragments that contain the genes of interest at higher efficiencies and lower cost. In Raphanus sativus L., a medicinal plant, plants formed hairy roots using A. rhizogenes, it was associated with the production of higher content of phenolic flavonoid and quercetin content compared to non-transformed plants [142]. Hairy roots were used for the production of phenolic acid, flavonoid, and wedelolactone from Sphagneticola calendulacea [143], tropane alkaloids of hyoscyamine, anisodamine, and scopolamine from Scopolia lurida [144].

Bacopa monnieri was transformed using A. tumefaciens with tryptophan decarboxylase and strictosidine synthase genes, which were obtained from C. roseus. Transformed tissues showed an increase in the terpenoid indole alkaloid pathway which led to an increase of 25-fold in tryptophan content in comparison with nontransformants [145]. Sharma et al. [146] used A. tumefaciens to transfer tryptophan decarboxylase and strictosidine synthase genes to C. roseus, it increased the content of terpenoid indole alkaloid metabolite due to the transient overexpression of these genes. In addition, several medicinal plants were subjected to genetic transformation including Iphigenia indica [88], Artemisia annua [57], Aconitum heterophyllum [100], P. somniferum L. and Eschscholzia californica [147]. Solanum aviculare [148], Pueraria phaseoloides [149], Crataeva nurvala [150], Gymnema sylvestre [151] and Holostemma ada-kodien [152] and Araujia sericirfera and Ceropegia spp [153].

5.8 Scale-up production

The application of PTC in medicinal plants can be scaled up using “bioreactors,” which allow atomization and production of a high yield of medicinal secondary products [154]. Therefore, scale-up production is a bioreactor application for the cultivation of plant cells on large-scale aiming for the mass production of valuable bioactive compounds. Also, bioreactor-based micropropagation was found to increase shoot multiplication for the commercial propagation of B. monnieri plants and maximize the content of bacosides in shoot biomass using an airlift bioreactor system [154]. Production of secondary metabolites using in vitro culture techniques is recommended strategy, especially when studying morphological and physiological processes associated with metabolites biosynthesis is necessary [155].

Cell suspension offers the wright combination of physical and chemical environments that must be used in the large-scale production of secondary metabolites in the bioreactor process [156]. Consequently, scale-up production in the bioreactor was used to expand the production of secondary metabolites from research to the industrial level. Systems of various sizes and features of bioreactors were created and applied for the mass production of secondary metabolites [157]. The application of plant tissue culture techniques in bioreactors for scale-up production facilitates obtaining some expensive pharmaceuticals that are synthesized in low quantity during in vitro or in vivo cultures. Since scale-up production of skikonin substance was achieved using bioreactors by Tabata and Fujita [158], other successful scale-up productions were obtained such as ginseng [159] and taxol [160].

Bioreactor operating system should provide efficient oxygen and nutrient supply, homogenous distribution of cultivated plant materials, and other factors that ensure optimal biomass and metabolite production [161]. While most of these bioreactors rely on cell suspension cultures, few of which are rely on differentiated tissues such as somatic embryos and hairy roots [162]. Application of suspension culture facilitates metabolites isolation [157].

For scale-up production, automation becomes an essential prerequisite, where it controls the pH of the culture area, culture viscosity, osmolarity, temperature, redox potential, oxygen supply, production of carbon dioxide, nutrients, weight, and liquid levels, and follows the rate of cell density. This automation needs sensors and monitoring systems that ensure mass production of pharmaceuticals and monitoring of physical, chemical, and biological parameters [163].

Perfusion cultivation is a system where continuous feeding of fresh media into a bioreactor system and removal of cells-free media were carried out in a modified bioreactor. The aim of this type of bioreactor and perfusion cultivation is to scaling-up the production of pharmaceutical compounds using plant cell, tissue, and organ cultures. The perfusion system offers a great advantage where it overcomes nutrient depletion and accumulation of growth inhibitors within the cultivated system, and it resulted in the promotion of biomass and pharmaceutical compounds. Semi-continuous perfusion was established in Anchusa officinalis where it was carried out in the shake flasks with a manual exchange of media. It resulted in the promotion of more than two-fold cell density and rosmarinic acid production in comparison to batch cultures [164].

Advances in immobilization and scale-up production techniques increase the applications of plant cell cultures for the purpose of producing high added value secondary compounds such as compounds with chemotherapeutic or antioxidant properties. For example, cell cultures of Plumbago rosea were immobilized using an MS medium containing 10 mM CaCl2 and calcium alginate for the production of important medicinal compounds, such as plumbagin [165]. Their studies indicated the impact of immobilization on the increased accumulation of plumbagin where immobilization in calcium alginate resulted in enhancement of plumbagin production up to three folds compared with that of control [156].

Advertisement

6. limitations over secondary metabolite production in vitro

In general, there are many factors that may hinder the application of PTC for various purposes in the field of medicinal and other plant species. The production of medicinal compounds using PTC has two important aspects—the amount of plant materials should be sufficient for the production of the target substance, as well as the quantity and quality of the produced substance. Hence, it is necessary to identify and avoid the conditions and phenomena that may negatively affect the growth efficiency of the in vitro cultured plant tissue.

6.1 Avoidance of secondary metabolites toxicity

Obstacles facing the production of medicinal compounds from wild or cultivated plants can be avoided by using cell and tissue cultures, but these compounds may be toxic to the in vitro cultured cells or tissues and result in retardation of plant material growth and metabolite yield. Consequently, the toxicity of any secondary metabolite should be assessed and culture conditions should be modulated to avoid the production obstacles. On the other side, the toxic effect of a secondary metabolite can be beneficially used for the treatment of some illnesses, for example, cancer [166].

Long-term culture can be used for the accumulation of desirable metabolite(s), but it can be a problematic and limiting factor that should be avoided by the application of certain techniques, such as medium enrichment or substitution in bioreactors [167]. These strategies include accumulation of metabolites in vacuoles, and other subcellular compartments or the exudation of metabolites into the culture medium [168]. The last strategy needs the application of additional techniques to decrease the concentration of the accumulated metabolite leading to further biosynthesis. It is accomplished by changing the medium of the culture manually or mechanically. In this regard, hairy roots were recommended, but not all secondary compounds are synthesized and accumulated in the roots [66].

6.2 Avoidance of low growth rate of cultured plant materials

While successful production of a wide range of valuable secondary metabolites can be obtained using unorganized callus or suspension cultures, the differentiated organ can be used but each of them may face some problems. The most important problems are the slow growth rate and somaclonal variation [84]. Consequently, the production of secondary compounds through the application of PTC techniques becomes unstable at a specific period. Generally, the problems facing the production of secondary metabolites using PTC can be easily solved by changing the culture conditions to avoid growth retardation and somaclonal variation [11]. Also, the application of PTC techniques in combination with other approaches could be used to avoid growth retardation and genetic variation [11].

The appropriate conditions for increasing the growth of the cultured plant materials may be different from the conditions for increasing the concentration of the active substance. To overcome these dilemmas, a two-step protocol is used, one of which provides optimal conditions for growth and the other provides optimal conditions to produce the active substance [12]. For example, while growth stimulators should be used during the growth phase, elicitors should be used to stimulate the biosynthesis of active compounds [169].

Accumulation of secondary metabolites is obtained under the influence of biotic or abiotic stress, but it retards the biological mass. To ensure a high yield of secondary metabolites, producers hope to conserve conditions to stimulate high biomass and biosynthesis of the targeted metabolite. Consequently, optimization of culture conditions to increase growth parameters or application of elicitors become an essential prerequisite [169].

6.3 Avoidance of problems constrain the application of transformation in the production of active compounds

Despite Agrobacterium is an essential tool for gene transformation; sometimes some technical problems retard its application in some plants, it depends on genotype and/or transformation technique [170]. On the other hand, many factors can affect the efficiency of Agrobacterium-mediated transformation such as Agrobacterium’s optical density [171], antibiotic [118] or acetosyringone concentrations, and inoculation time [172]. All these difficulties should be avoided for the successful application of transformation techniques in the field of secondary metabolites production.

6.4 Avoidance culture browning

In vitro cultured explants release phenol compounds, which are oxidized by polyphenol oxidase and turned the media brown [173]. In woody plants, phenolic exudation appears early during the excision of plants causing browning of the cultured medium [173]. Browning closes the base of explants and retards the movement of nutrients from the medium into the cultured plant materials leading to retardation of plant growth. To overcome tissue browning, antioxidants or phenol absorbents, such as ascorbic acid, glutathione, activated charcoal, and polyvinylpyrrolidone were used. Also, transferring explants into new culture media at regular intervals can control the negative effects of the browning phenomenon [173]. To overcome the browning effect in the culture media in Glycyrrhiza inflata cell cultures, cultures were optimized in a bioreactor containing maximum cell concentration [174]. Dark conditions help to reduce the browning problem may be due to the reduction of the activity of the enzymes concerned with phenols synthesis and oxidation [175, 176].

6.5 Avoidance of somaclonal variation of the cultured plant materials

Production of the secondary metabolites using the cell culture technique is low during the early stage of growth where high carbon utilization exists and is associated with enhancement of primary metabolism. On the other hand, the production of secondary metabolites is high at the late stage when carbon is less needed for the production of primary metabolism [14]. Prolonged the age of the cultured plant materials is necessary but it may be associated with genetic variation [47, 84]. Therefore, the enhancement of growth criteria of the cultured plant materials is not sufficient to confirm the optimization of in vitro culture techniques for the production of secondary metabolites, but also genetic stability at the DNA level of the cultured plant materials is an essential parameter. For example, regenerates with high genetic fidelity and improved chemical profile of endangered C. spinosa L were reported, where the two-fold increase in flavonoids content than that of wild plants was obtained using methyl jasmonate and BAP [105]. Plant material with genetic fidelity after propagated in vitro culture was detected and used for the isolation of 20-hydroxyecdysone and polypodine B [177]. Production of true to type regenerants in Artemisia absinthium is very important in the commercial production of secondary metabolites [178].

Somaclonal variation results from chromosomal changes in number or structure, transposable elements, or possibly pre-existing genetic changes in the donor plant. To detect somaclonal variation, several molecular techniques such as Random Amplified Polymorphic DNA (RAPD), Inter Simple Sequence Repeat (ISSR), and Simple Sequence Repeat (SSR) were recommended [18, 47].

6.6 Avoidance of vitrification

Sometimes, the production of secondary metabolites through some techniques such as cell suspension is not always an adequate procedure. Then, other techniques such as organ culture can be used as a supernumerary method for the production of secondary metabolites [85]. Shoot cultures as same as hairy root cultures are recommended for production of pharmaceuticals where they are genetically stable [179]. Shooty teratomas were produced for the production of secondary metabolites, such as vincristine in C. roseus [180] and naphthoquinone in Drosera capensis var. alba [181]. In some plant species, shoot culture showed vitrification problems, such as in moringa [47].

Generally, tissue culture plant materials were incubated in vessels to prevent microbial contamination and retard culture desiccation but these conditions may cause restriction of gases exchange between cultures and their surrounds. Under insufficient ventilation stress, the growth of the cultured plant materials was retarded due to retardation of photosynthesis, transpiration, and uptake of water and nutrients leading to the accumulation of ethylene and the appearance of vitrification or hyperhydricity [182]. The symptoms of vitrification are slowing growth rate, necrosis of shoot tips, loss of apical dominance, disorganized cell wall, fragile leaves, reduction of shoot multiplication, poor acclimatization, impaired stomatal function, reduction of some metabolites, alteration of ion composition, inhibition of H2O2 detoxification enzymes [183, 184].

Vitrification in medicinal and other plant species can be avoided by reducing the relative humidity and improving the aeration within culture vessels [183, 184], decreasing the concentration of free water by increasing the concentration of agar [185], and using anti-ethylene compounds including CoCl2, AgNO3 or salicylic acid [47, 183]. To confirm which anti-ethylene compounds can be used to conserve the genetic fidelity of in vitro cultured moringa shoots, fingerprinting profiles of the long-term culture (14 subcultures) were assessed using RAPD, SSR, and ISSR. While the application of silver nitrate improved plant multiplication and reduced vitrification but it resulted in higher somaclonal variation in comparison to salicylic acid [47].

Advertisement

7. Conclusion

An increase in the world’s population imposed an important matter, which is the inevitability of leaving arable land for food production. Where modern agricultural techniques can be used to produce secondary metabolites and preserve the genetic assets of these plants, the most notable technique is PTC. In addition, different PTC techniques are used to propagate rare and endangered plant species. Changes in the physical and chemical conditions of in vitro culture are easy and under control in a way that cannot be provided at all under field conditions. The ease of controlling the conditions of PTC conditions made it possible to use certain conditions to obtain true-to-type clones and their products, but other conditions are used to establish somaclonal variation for noval line selection.

The use of plant tissue techniques has become dependent on it to produce pharmaceutical materials after laboratory and applied experiments have proven that in vitro cultured plant materials are able to produce pharmaceuticals with the same amount and quality that can be obtained from soil cultivated plants. Moreover, the application of elite physical and chemical conditions of in vitro cultured plant materials made their production of secondary metabolites superior in quantity and quality to that of wild or cultivated plants. Therefore, to produce pharmaceutical compounds in large quantities to suit the increase in the population and increase their demand for safe medical products, tissue and cell culture techniques have been improved under several names including culture media optimization, the establishment of suspension and callus cultures, elicitation to enhance the productivity of in vitro cultures, application of precursor feeding as a substrate to improve the production of secondary metabolites, high yielding cell lines selection, enhance the overexpression of genes that control the production of bioactive compounds, application of genetic transformation using A. rhizogenes and application of “bioreactors” for scale-up production.

The use of PTC techniques to produce pharmaceutical compounds depends on the availability of production of sufficient-viable plant biomass to produce pharmaceutical substances with the requested quality and quantity. Therefore, it is necessary to understand all the factors that limit the production of targeted mass to avoid them such as the toxicity of secondary metabolites, low growth rate of cultured plant materials, and problems that constrain the application of transformation on a wide spectrum of plant species, somaclonal variation during cell or tissue cloning and verification of the cultured plant organs.

References

  1. 1. Oksman-Caldentey KM, Inzé D. Plant cell factories in the postgenomic era: New ways to produce designer secondary metabolites. Trends in Plant Science. 2004;9:433-440
  2. 2. Rao SR, Ravishankar GA. Plant cell cultures: Chemical factories of secondary metabolites. Biotechnology Advances. 2002;20(2):101-153
  3. 3. Folashade OK, Omoregie EH, Peter OA. Standardization of herbal medicines—A review. International Journal of Biodiversity and Conservation. 2012;4(3):101-112
  4. 4. Gantait S, Debnath S, Ali MN. Genomic profile of the plants with pharmaceutical value. 3 Biotech. 2014;4:563-578
  5. 5. Toledo VM. New paradigms for a new ethnobotany: Reflections on the case of Mexico. In: Schultes RE, Von Reis S, editors. Ethnobotany: Evolution of a Discipline. London, UK: Chapman and Hall; 1995. pp. 75-88
  6. 6. FAO. Trade in Medicinal Plants. Raw Materials, Tropical and Horticultural Products Service Commodities and Trade Division, Economic and Social Department, Food and Agriculture Organization of the United Nations. 2004. Rome. Available from: http://www.fao.org/3/af285e/af285e00.pdf
  7. 7. Nalawade SM, Tsay H. In vitro propagation of some important Chinese medicinal plants and their sustainable usage. In vitro cellular & developmental biology. Plant. 2004;40:143-154
  8. 8. An C. In Vitro propagation of commonly used medicinal trees in Korea. Journal of Environmental Sciences. 2019;35:272-280
  9. 9. Hassanein AM, Mazen AMA. Adventitious bud formation in Alhagi graecorum. Cell, Tissue and Organ Culture. 2001;65:31-35
  10. 10. Hassanein AM, Galal DM, Soltan DM, Saad GK. Effect of medium strength and activated charcoal on in vitro shoot multiplication and growth of jojoba. Journal of Environmental Studies. 2015;14:81-90
  11. 11. Thangavel K, Ebbie MG, Ravichandran P. Biotechnology and In Vitro conservation of Medicinal Plants. Annals of Plant Sciences. 2014;3(06)
  12. 12. Cardoso JC, de Oliveira ME, de Cl Cardoso F. Advances and challenges on the in vitro production of secondary metabolites from medicinal plants. Horticultura Brasileira. 2019;37:124-132
  13. 13. Sajc L, Grubisic D, Vunjak-Novakovic G. Bioreactors for plant engineering: An outlook for further research. Biochemical Engineering Journal. 2000;4:89-99
  14. 14. Isah T, Umar S, Mujib A, Sharma MP, Rajasekharan PE, Zafar N, et al. Secondary metabolism of pharmaceuticals in the plant in vitro cultures: Strategies, approaches, and limitations to achieving higher yield. Plant Cell, Tissue and Organ Culture. 2018;132:239-265
  15. 15. Hassanein AM, Soltan DM. Solanum nigrum is a model plant in tissue and protoplast culture. Biologia Plantarum. 2000;43:501-509
  16. 16. Sagare AP, Lee YL, Lin TC, Chen CC, Tsay HS. Cytokinin-induced somatic embryogenesis and plant regeneration in Corydalis yanhusuo (Fumariaceae)—A medicinal plant. Plant Science. 2000;160(1):139-147
  17. 17. Hassanein AM, Faheed FA, Salem JM, El-nagish AA. Some important aspects in moringa micropropagation. Acta Agriculturae Slovenica. 2019;113(1):13-27. DOI: 10.14720/aas.2019.113.1.02
  18. 18. Hassanein AM, Salem JM, Hassan RA, Soltan DM. Morpho-molecular characterization and differential totipotency of three Solanum taxa. Phyton (Horn, Austria). 2020;60:145-159
  19. 19. Amoo SO, Aremu AO, van Staden J. In vitro plant regeneration, secondary metabolite production and antioxidant activity of micropropagated Aloe arborescens Mill. Plant Cell, Tissue and Organ Culture. 2012;111(3):345-358
  20. 20. Manivannan A, Soundararajan P, Park YG, Jeong PR. In vitro propagation, phytochemical analysis, and evaluation of free radical scavenging property of Scrophularia kakudensis Franch tissue extracts. BioMed Research International. 2015;2015:11
  21. 21. Kabera JN, Semana E, Mussa AR, He X. Plant secondary metabolites: Biosynthesis, classification, function and pharmacological properties. The Journal of Pharmacy and Pharmacology. 2014;2:377-392
  22. 22. Shukla MR, Kibler A, Turi CE, Erland LAE, Sullivan JA, Murch SJ, et al. Selection and micropropagation of an Elite Melatonin Rich Tulsi (Ocimum sanctum L.) Germplasm Line. Agronomy. 2021;11:207
  23. 23. Castro TC, Pelliccione VLB, Figueiredo MR, Soares RODA, Bozza MT, Viana VRC, et al. Atividade antineoplsica e tripanocida de Hovenia dulcis Thunb. Cultivada in vivo e in vitro. Brazilian Journal of Pharmacogenetics. 2002;12:96-99
  24. 24. Mandal S, Parsai A, Tiwari PK, Nataraj M. The effect of additional additives on the axillary shoot micropropagation of medicinal plant Aegle marmelos (L.) Corrêa. World News of Natural Sciences. 2021;34:54-71
  25. 25. Kothari S, Mishra V, Bharat S, Tonpay SD. Antimicrobial activity and phytochemical screening of serial extracts from leaves of Aegle marmelos (Linn.). Acta Polymer Pharmaceutical Drug Research. 2011;68(5):687-692
  26. 26. Shinde S, Pooja R, Katewal PR, Shanbhag DD, Joseph KS, Murthy HN. In Vitro Propagation of Artemisia japonica. Journal of Herbs, Species & Medicinal Plants. 2017;23(1):36-43. DOI: 10.1080/10496475.2016.1256010
  27. 27. Yao L, Zhu S, Liu W, Manzoor Z, Nisar M F, Li M. A comprehensive review of pharmacological and Analytical Aspects of Acacetin-Natural Resources for Human Health. Open access article—CC BY-NC-ND license 2021. Available from: http://creativecommons.org/licenses/by-nc-nd/4.0/
  28. 28. Salem MM, Davidorf FH, Abdel-Rahman MH. In vitro anti-uveal melanoma activity of phenolic compounds from the Egyptian medicinal plant Acacia nilotica. Fitoterapia. 2011;82:1279-1284
  29. 29. Sander VA, Corigliano MG, Clemente M. Promising plant-derived adjuvants in the development of coccidial vaccines. Frontier in Veterinary Science. 2019;6:20. DOI: 10.3389/fvets.2019.00020
  30. 30. Gunasekaran B, Gothandam KM. A review on edible vaccines and their prospects. Brazilian Journal of Medical and Biological Research. 2020;53(2). DOI: 10.1590/1414-431X20198749
  31. 31. Yousif F, Hifnawy MS, Soliman G, Boulos L, Labib T, Mahmoud S, et al. Large-scale in Vitro. Screening of Egyptian Native and Cultivated Plants for Schistosomicidal Activity. Pharmaceutical Biology. 2007;45(6):501-510. DOI: 10.1080/13880200701389425
  32. 32. El-Hallouty S, Fayad W, Meky N, Hasabo AA, editors. In vitro anticancer activity of some Egyptian plant extracts against different human cancer cell lines. International Journal of Pharm Tech Research. 2015, 2015;8(2):267-327
  33. 33. Aboul-Enein AM, Abu El-Ela F, Shalaby EA, El-Shemy HA. Traditional medicinal plants research in Egypt: Studies of antioxidant and anticancer activities. Journal of Medicinal Plant Research. 2012;6(5):689-703
  34. 34. Mathur S, Sharma S, Gupta MM, Kumar S. Evalution of an Indian germplasm collection of the medicinal plant Bacopa monnieri (L.) Pennell by use of multivariate approaches. Euphytica. 2003;133(3):255-265
  35. 35. Srivastava SK, Srivastava N. In Vitro multiplication of Paedaria foetida L—A rare medicinal plant. Journal of Plant Biochemistry and Biotechnology. 2004;13:89-91
  36. 36. Sood H, Chauhan HS. Development of a low cost micropropagation technology for an endangered medicinal herb (Picorhiza kurroa) of North-Western Himalayas. Journal of Plant Science. 2009;4(2):21-31
  37. 37. Mathur S, Shekhawat GS, Batra A. Micropropagation of Salvadora persica Linn. via cotyledonary nodes. Indian Journal of Biotechnology. 2002;1(2):197-200
  38. 38. Sambyal M, Dogra A, Koul S, Ahuja A. Rapid in vitro propagation of Potentilla fulgens Wall—A Himalayan alpine herb of medicinal value. Journal of Plant Biochemistry and Biotechnology. 2006;15:143-145
  39. 39. Arockiasamy S, Prakash S, Ignacimuthu S. Direct organogenesis from mature leaves and petioles of Eryngium foetidum L. Biologia Plantarum. 2002;45:129-132
  40. 40. Saavedra AM, Castrol T, Cordeiro L, Athayde T, Albarello N, Simoes C. In vitro propagation and cryopreservation of the medicinal species Hovenia dulcis Thunb. (Rhamnaceae). Plant Cell, Tissue and Organ Culture. 2021;144:577-591
  41. 41. Meena MC, Patni V. In vitro clonal propagation of Citrullus colocynthis (Linn.) Scharad: Threatened medicinal plant. Plant Cell Biotechnology and Molecular Biology. 2007;8(3&4):147-152
  42. 42. Gayathri D, Ramagopal G. In vitro propagation of Amaryllis and Zephyranthes bulbous species using household tissue culture medium. Plant Cell Biotechnology and Molecular Biology. 2007;8(3&4):131-136
  43. 43. Sivasubramanian S, Vallinayagam S, Patric RD, Manickam VS. Microproagation of Plectranthus vetiveroides (Jacob) Sing & Sharma—A medicinal plant. Phytomorphology. 2002;52:55-59
  44. 44. Geetha R, Gopal GV. Direct regeneration in Glossocardia bosvallea: A medicinal plant. Plant Cell Biotechnology and Molecular Biology. 2007;8:119-124
  45. 45. Monthony AS, Page SR, Hesami M, Jones AMP. The past, present and future of Cannabis sativa tissue culture. Planta. 2021;10:185. DOI: 10.3390/plants
  46. 46. Shawky R, Mussa EA. In vitro propagation of the rare Caralluma retrospeciens plant in Egypt. International Journal of Advanced Research. 2020;8(8):812-818
  47. 47. Hassanein AM, Salem JM, Faheed FA, El-nagish AA. Effect of anti-ethylene compounds on isoenzyme patterns and genome stability during long term culture of Moringa oliefera. Plant Cell, Tissue and Organ Culture. 2018;132:201-212
  48. 48. Ghareb HE. In vitro propagation of the rare Pulicaria incisa. Journal of Basic Applied Science and Research. 2018;8(7):26-33
  49. 49. Darwish HY, Ahmed SM. Elicitors enhancing phenolics content and related gene expression variation in petal—Derived calli of Rosa damascena Mill. Egyptian Journal of Bottomline. 2020;60(1):71-79
  50. 50. Puhan P, Rath SP. In vitro propagation of Aegle marmelos (L.) corr., a medicinal plant through axillary bud multiplication. Advances in Bioscience and Biotechnology. 2012;3:121-125
  51. 51. Liu CZ, Murch SJ, El-Demerdash M, Saxena PK. Regeneration of the Egyptian medicinal plant Artemisia judaica L. Plant Cell Reports. 2003;21:525-530
  52. 52. Abed Elmaksood WM, Ebad FA, Bosila HA. In vitro propagation of the endangered medicinal plant Hyoscyamus muticus L. (Egyptian Henbane). Journal of Applied Environmental Biological Science. 2016;6(4):25-34
  53. 53. Ahuja S, Mandal BB, Dixit S, Srivastava PS. Molecular, phenotypic and biosynthetic stability in Dioscorea floribunda plants derived from cryopreserved shoot tips. Plant Science. 2002;163:971-977
  54. 54. Dixit-Sharma S. Cryopreservation of somatic embryos. Indian Journal of Biotechnology. 2005;4:47-55
  55. 55. Lalaleo L, Testillano P, Risueňo MC, Cusidó RM, Palazon J, Alcazar R, et al. Effect of in vitro morphogenesis on the production of podophyllotoxin derivatives in callus cultures of Linum album. Journal of Plant Physiology. 2018;228:47-58
  56. 56. Espinosa-Leal CA, Puente-Garza A, García-Lara CA. In vitro plant tissue culture: Means for production of biological active compounds. Planta. 2018;248:1-18
  57. 57. Cai G, Li G, Ye H, Li G. Hairy root culture of Artemisia annua L. by Ri plasmid transformation and biosynthesis of artemisinin. Chinese Journal of Biotechnology. 1995;11:227-235
  58. 58. Sreekumar S, Seeni S, Pushpangadan P. Production of 2 – hydroxy 4- methoxy benzaldehyde using root cultures of Hemidesmus indicus. Biotechnology Letters. 1998;20(7):631
  59. 59. Vanisree M, Tsay HS. Studies on the production of some important secondary metabolites from medicinal plants by plant tissue cultures. Botanical Bulletin of Academia Science. 2004;45:1-22
  60. 60. Hirata K, Yamanaka A, Kurano N, Miyamota K, Miura Y. Production of indole alkaloids in multiple shoot culture of (Catharanthus roseus 9L.) G. Don. Agricultural and Biological Chemistry. 1987;5:11311-11317
  61. 61. Akashi T, Furuno T, Takahashi T, Ayabe S. Biosynthesis of triterpenoids in cultured cells regenerated and wild plant organs of Taraxacum officinale. Phytochemistry. 1994;36:303-308
  62. 62. Alamgir ANM. Therapeutic medicinal plants and their extracts. Pharmacognosy. 2017;73:403-426
  63. 63. Sathuluri RR, Gokare R. Plant cell cultures: Chemical factories of secondary metabolites. Biotechnology Advances. 2002;20(2):101-153. DOI: 10.1016/S0734-9750(02)00007-1
  64. 64. Chang WD, Huang WW, Chen CC, Chang YS, Tsay HS. The production of secondary metabolites from Chinese medicinal herbs by suspension cell and tissue culture. In: Proc. 7th Int. Congr of SABRAO and WASS. Taipei, Taiwan; 1994. pp. 535-540
  65. 65. Figueiro ADA, Correa CM, Astarita LV, Santarem ER. Long-term maintenance of the in vitro cultures affects growth & secondary metabolism of St. John’s Wort. Ciencia Rural. 2010;40(10):2115-2121
  66. 66. Kim W, Pamela J. Weathers Secondary metabolism of hairy root cultures in bioreactors. Plant. 2002;38:1-10
  67. 67. Bharathi P, Philomina D. Effect of nutritional factors and precursors on the formation of colchicine in Gloriosa superba in vitro. Research Biotechnology. 2010;1:29-37
  68. 68. Hassanein AM, Galal A, Saad G, Soltan DM. Impact of slight changes in water potential of culture media on in vitro shoot multiplication, esterase, and protein patterns of Simmondsia chinensis L. Egyptian Journal of Botany. 2022;62(1):97-111. DOI: 10.21608/ejbo.2021.46723.1571
  69. 69. Trejo-Tapia G, Arias-Castro C, Rodrguez-Mendiola M. Influence of the culture medium constituents & inoculum size on the accumulation of blue pigment & cell growth of Lavandula spica. Plant Cell, Tissue and Organ Culture. 2003;72:7-12
  70. 70. Gandhi SG, Mahajan V, Bedi YS. Changing trends in biotechnology of secondary metabolism in medicinal and aromatic plants. Planta. 2015;241:303-317
  71. 71. Monfort LEF, Bertolucci SKV, Lima AF, de Carvalho AA, Mohammed A, Blank AF, et al. Effects of plant growth regulators, different culture media and strength MS on production of volatile fraction composition in shoot cultures of Ocimum basilicum. Industrial Crops and Products. 2018;116:231-239
  72. 72. Oliveira JPS, Hakimi O, Murgu M, Koblitz MGB, Ferreira MSL, Cameron LC, et al. Tissue culture and metabolome investigation of a wild endangered medicinal plant using high definition mass spectrometry. Plant Cell Tissue Organ Culture. 2018;134:153-162
  73. 73. Hassanein AM. Hormonal requirements trigger different regeneration pathways in Alhagi Graecorum. Journal of Plant Biotech. 2004;6:171-179
  74. 74. Raj D, Kokotkiewicz A, Drys A, Luczkiewicz M. Effect of plant growth regulators on the accumulation of indolizidine alkaloids in Securinega suffruticosa callus cultures. Plant Cell, Tissue and Organ Culture. 2015;123(1):39-45. DOI: 10.1007/s11240-015-0811-6
  75. 75. Jang HR, Lee HJ, Park BJ, et al. Establishment of embryogenic cultures and determination of their bioactive properties in Rosa rugosa. Horticulture, Environment and Biotechnology. 2016;57(3):291-298
  76. 76. Isah T, Umar S. Influencing in vitro clonal propagation of Chonemorpha fragrans (moon) Alston by culture media strength, plant growth regulators, carbon source and photoperiodic incubation. Journal of Forest Research. 2020;31(1):27-43
  77. 77. Karwasara VS, Dixit VK. Culture medium optimization for camptothecin production in cell suspension cultures of Nothapodytes nimmoniana (J. Grah.) Mabberley. Plant Biotechnology Reports. 2013;7(3):357-369
  78. 78. Thanh NT, Van KN, Paek KY. Effecting of medium composition on biomass and ginsenoside production in cell suspension culture of Panax vietnamensis Haet Grushv. VNUJ Science Natural Technology. 2007;23:269-274
  79. 79. Irshad M, Debnath B, Mitra S, Arafat Y, Li M, Sun Y, et al. Accumulation of anthocyanin in callus cultures of red-pod okra (Abelmoschus esculentus (L) Hongjiao) in response to light and nitrogen levels. Plant Cell Tissue and Organ Culture. 2018;134:29-39
  80. 80. Hagendoom JM, Wagner AM, Segers C, van der Plas LHW, André Oostdam A, van Walraven HS. Cytoplasmic acidification and secondary metabolite production in different plant cell suspensions. Plant Physiology. 1994;106:723-730
  81. 81. Majerus F, Pareilleus A. Production of indole alkaloids by gelentrapped cells of Catharanthus roseus cells in a continuous flow reactor. Biotechnology Letters. 1986;8:863-866. DOI: 10.1007/BF01078646
  82. 82. Nagella P, Murthy HN. Establishment of cell suspension cultures of Withania somnifera for production of withanolide A. Bioresource Technology. 2010;101(17):6735-6739. DOI: 10.1016/j
  83. 83. Naik PM, Manohar SH, Praveen N, Murthy HN. Effects of sucrose & pH levels on the in vitro shoot regeneration from the leaf explants of Bacopa monnieri and accumulation of bacoside A in regenerated shoots. Plant Cell, Tissue and Organ Culture. 2010;100(2):235-239. DOI: 10.1007/s11240-009-9639-2
  84. 84. Hassanein AM, Ahmed AM, Soltan DM. Study of somaclonal variation and gene expression as influenced by long term culture in sorghum. Current Opinion in Biotechnology. 2008;4:13-20
  85. 85. Fazili MF, Bashir I, Ahmad M, Yaqoob U, Geelani SN. In vitro strategies for the enhancement of secondary metabolite production in plants: A review. Bulletin of the National Research Centre. 2022;46:35
  86. 86. Pan ZG, Liu CZ, Murch SJ, El-Demerdash M, Saxena PK. Plant regeneration from mesophyll protoplasts of the Egyptian medicinal plants, Artemisia judiica L. and Echinops spinosissimum (Turra). Plant Science. 2003;165:681-687
  87. 87. Kurz WGW, Constabel F. Production of secondary metabolites. In: Altman A, editor. Agricultural Biotechnology. New York: Marcel Dekker Inc; 1998. pp. 183-224
  88. 88. Mukhopadhyay MJ, Mukhopadhyay S. Biotechnological Approach for enhancement of Colchicine Accumulation in Iphigenia indica Kunth. Journal of Plant Biochemistry and Biotechnology. 2008;17(2):185-188
  89. 89. Feher A, Pasternak T, Otvos K, Miskolczi P, Dudits D. Induction of embryogenic competence in somatic plant cells: A review. Biologia (Bratislava). 2002;57:5-12
  90. 90. Raghavan V, Srivastava PS. Embryo culture. In: Jhori JM, editor. Experimental Embryology of Vascular Plants. Berlin, Heidelberg, New York: Springer Verlag; 1982. pp. 195-230
  91. 91. Roberts SC, Shule ML. Large-scale plant cell culture. Current Opinion in Biotechnology. 1997;8(2):154-159. DOI: 10.1016/s0958-1669(97)80094-8
  92. 92. Gontier E, Sangwan BS, Barbotin JN. Effects of calcium, alginate, and calcium-alginate immobilization on growth and tropane alkaloid levels of a stable suspension cell line of Datura innoxia Mill. Plant Cell Reports. 1994;13:533-536
  93. 93. Gupta K, Garg S, Singh J, Kumar M. Enhanced production of naphthoquinone metabolite (shikonin) from the cell suspension culture of Arnebia sp. & its up-scaling through bioreactor. Biotech. 2014;4(3):263-273. DOI: 10.1007/s13205-013-0149-x
  94. 94. Tom R, Jardin B, Chavarie C, Rho D, Archambault J. Effect of culture process on alkaloid production by Catharanthus roseus cells: II. Immobilized cultures. Journal of Biotechnology. 1991;21(1-2):21-42
  95. 95. Wu CH, Murthy HN, Hahn EJ, Paek KY. Establishment of adventitious root co-culture of Ginseng & Echinacea for the production of secondary metabolites. Acta Physiologiae Plantarum. 2008;30(6):891-896. DOI: 10.1007/s11738-008-0181-3
  96. 96. Kiong AL, Mahmood M, Fodzillan NM, Daud SK. Effects of precursor supplementation on the production of triterpenes by Centella asiatica callus culture. Pakistan Journal of Biological Sciences. 2005;8:1160-1169. DOI: 10.3923/pjbs.2005.1160.1169
  97. 97. Giri CC, Zaheer M. Chemical elicitors versus secondary metabolite production in vitro using plant cell, tissue and organ cultures: Recent trends and a sky eye view appraisal. Plant Cell, Tissue and Organ Culture (PCTOC). 2016;126:1-18
  98. 98. Zhao J, Liu W, Wang J. Recent advances regarding constituents and bioactivities of plants from the genus Hypericum. Chemistry & Biodiversity. 2015;12:309-349
  99. 99. Garcia-Perez I, Posma JM, Chambers ES, Mathers JC, Draper J, Beckmann M, et al. Dietary metabotype modelling predicts individual responses to dietary interventions. Nature Food. 2020;1:355-364. DOI: 10.1038/s43016-020-0092-z
  100. 100. Giri A, Banerjee S, Ahuja PS, Giri CC. Production of hairy roots in Aconitum heterophyllum Wall. using Agrobacterium rhizogenes. In vitro Cell Development Biology. 1997;33:293-306
  101. 101. Darvishi E, Kahrizi D, Bahraminejad S, Mansouri M. In vitro induction of α-pinene, pulegone, menthol, menthone and limonene in cell suspension culture of pennyroyal (Mentha pulegium). Cellular and Molecular Biology. 2016;62:7-9
  102. 102. Castilho CVV, Neto JFF, Leitمo SG, Barreto CS, Pinto SC, da Silva NCB. Anemia tomentosa var. anthriscifolia in vitro culture: Sporophyte development and volatile compound profile of an aromatic fern. Plant Cell Tissue and Organ Culture. 2018;133:311-323
  103. 103. Staniszewska I, Królicka A, Maliński E, Łojkowska E, Szafranek J. Elicitation of secondary metabolites in in vitro cultures of Ammi majus L. Enzyme and Microbial Technology. 2003;33:565-568
  104. 104. Jin JH, Shin JH, Kim JH, Chung IS, Lee HJ. Effect of chitosan elicitation and media components on the production of anthraquinone colorants in madder (Rubia akane Nakai) cell culture. Biotechnology and Bioprocess Engineering. 1999;4:300-304
  105. 105. Goda SM, Ahmed SA, El Sherif F, Hassanean HA, Ibrahim AK. Genetically stable plants with boosted flavonoids content after in vitro regeneration of the endangered Capparis spinosa L. Global Drugs Therapy. 2017;2(4):1-7
  106. 106. Bhagwath SG, Hjortso MA. Statistical analysis of elicitation strategies for thiarubrine A production in hairy root cultures of Ambrosia artemisiifolia. Journal of Biotechnology. 2000;80(2):159-167. DOI: 10.1016/S0168-1656(00)00256-X
  107. 107. Isah T. Stress and defense responses in plant secondary metabolites production. Biological Research. 2019;52:39
  108. 108. García-Pérez P, Lozano-Milo E, Landín M, Gallego PP. Combining medicinal plant in vitro culture with machine learning technologies for maximizing the production of phenolic compounds. Antioxidants. 2020;9:210
  109. 109. Blasco B, Navarro-León E, Ruiz JM. Oxidative Stress in Relation with Micronutrient Deficiency or Toxicity. Amsterdam, The Netherlands: Elsevier Inc; 2018
  110. 110. Sharma A, Shahzad B, Rehman A, Bhardwaj R, Landi M, Zheng B. Response of phenylpropanoid pathway and the role of polyphenols in plants under abiotic stress. Molecules. 2019;24:2452
  111. 111. Jeong GA, Park DH. Enhanced secondary metabolite biosynthesis by elicitation in transformed plant root system: Effect of abiotic elicitors. Applied Biochemistry and Biotechnology. 2006;129:436-446
  112. 112. Tonk D, Mujib A, Maqsood M, Ali M, Zafar N. Aspergillus flavus fungus elicitation improves vincristine and vinblastine yield by augmenting callus biomass growth in Catharanthus roseus. Plant Cell, Tissue and Organ Culture. 2016;126:291-303
  113. 113. Kowalczyk T, Merecz-Sadowska A, Picot L, Brċiċ Karaċonji I, Wieczfinska J, Sliwiński T, et al. Genetic manipulation and bioreactor culture of plants as a tool for industry and its applications. Molecules. 2022;27:795. DOI: 10.3390/molecules27030795
  114. 114. Jackson P, Attalla MI. N-Nitrosopiperazines form at high pH in post-combustion capture solutions containing piperazine: A low-energy collisional behavior study. Rapid Communications in Mass Spectrometry. 2010;24(24):3567-3577. DOI: 10.1002/rcm
  115. 115. Jaisi A, Panichayupakaranant P. Enhanced plumbagin production in Plumbago indica root cultures by l-alanine feeding and in situ adsorption. Plant Cell, Tissue and Organ Culture. 2016;38(2):351-355. DOI: 10.1007/s11240-016-1155-6
  116. 116. Rahimi S, Hasanloo T, Najafi F, Khavari-Nejad RA. Enhancement of silymarin accumulation using precursor feeding in Silybum marianum hairy root cultures. Plant Omics Journal. 2011;4(1):34-39
  117. 117. Tumova L, Rimakova J, Tuma J, Dusck J. Silybum marianum in vitro-flavolignan production. Plant, Cell & Environment. 2006;52:454-458
  118. 118. Sivanandhan G, Selvaraj N, Ganapathi A, Manickavasagam M. Enhanced biosynthesis of withanolides by elicitation and precursor feeding in cell suspension culture of Withania somnifera (L.) Dunal in Shake-Flask Culture and Bioreactor. PLoS One. 2014;9:e104005
  119. 119. Karppinen K, Hokkanen J, Tolonen A, Maltila S, Hohtola A. Biosynthesis of hyperforin & adhyperforin from amino acid precursors in the shoot cultures of Hypericum perforatum. Phytochemistry. 2007;68:1038-1045. DOI: 10.1016/j.phytochem.2007.01.001
  120. 120. Taha H, El-Bahr MK, Seif-Nasr MM. In vitro studies on Egyptian Catharanthus roseus (L.) G.Don. IV: Manipulation of Some Amino Acids as Precursors for Enhanced of Indole Alkaloids Production in Suspension Cultures. Australian Journal of Basic and Applied Sciences. 2009;3(4):3137-3144
  121. 121. Abyari M, Nasr N, Soorni J, Sadhu D. Enhanced accumulation of scopoletin in cell suspension culture of Spilanthes acmella Murr. using precursor feeding. Brazilian Archives of Biology and Technology. 2016;59:e16150533
  122. 122. Lee MH, Cheng JJ, Lin CY, Chen YJ, Lu MK. Precursor-feeding strategy for production of solanine, solanidine, & solasodine by a cell culture of Solanum lyratum. Process Biochemistry. 2007;42(5):899-903
  123. 123. Liu Y, Wang J, Wang L, Zhang C, Bai Q , Lyu X, et al. Biosynthesis and biotechnological production of salidroside from Rhodiola genus plants. Phytochemistry Reviews. 2022. DOI: 10.1007/s11101-021-09800-1
  124. 124. Xu JF, Liu CB, Han AM, Feng PS, Su ZG. Strategies for the improvement of salidroside production in cell suspension cultures of Rhodiola sachalinensis. Plant Cell Reports. 1998;17:288-293. DOI: 10.1007/s002990050394
  125. 125. Chetri SK, Kapoor H, Agrawal V. Marked enhancement of sennoside bioactive compounds through precursor feeding in Cassia angustifolia Vahl & cloning of isochorismate synthase gene involved in its biosynthesis. Plant Cell, Tissue and Organ Culture. 2016;124(2):431-446. DOI: 10.1007/s11240-015-0905-1
  126. 126. Sandhu SS, Abreu IN, Colombo CA, Mazaferra P. Pilocarpine content and molecular diversity in jaborandi. Scientia Agrária. 2006;63:478-482
  127. 127. Briskin DP. Biotechnological methods for selection of high-yielding cell lines and production of secondary metabolites in medicinal plants. In: Kayser O, Quax WJ, editors. Medicinal Plant Biotechnology. Weinheim: Wiley; 2007. pp. 187-201
  128. 128. Isah T. Production of camptothecin in the elicited callus cultures of Nothapodytes nimmoniana (J.Graham) mabberly. Chemical Papers. 2017;71(6):1091-1106. DOI: 10.1007/s11696-016-0056-9
  129. 129. Niggeweg R, Michael AJ, Martin C. Engineering plants with increased levels of the antioxidant chlorogenic acid. Nature Biotechnology. 2004;22(6):746-754
  130. 130. Kumar S, Singh A, Kumar B, Singh B, Bahadur L, Lal M. Simultaneous quantitative determination of bioactive terpene indole alkaloids in ethanolic extracts of Catharanthus roseus (L.) G. Don by ultrahigh performance liquid chromatography-tandem mass spectrometry. Journal of Pharmaceutical and Biomedical Analysis. 2018;151:32-41
  131. 131. Canel C, Lopes-Cardoso MI, Whitmer S, van der Fits L, Pasquali G, van der Heijden R, et al. Effects of over-expression of strictosidine synthase and tryptophan decarboxylase on alkaloid production by cell cultures of Catharanthus roseus. Planta. 1998;205:414-419
  132. 132. Whitmer S, van der Heijden R, Verpoorte R. Effect of precursor feeding on alkaloid accumulation by a tryptophan decarboxylase over-expressing transgenic cell line T22 of Catharanthus roseus. Journal of Biotechnology. 2002;96:193-203
  133. 133. Hashimoto T, Yun DJ, Yamada Y. Production of tropane alkaloids in genetically engineered root cultures. Phytochemistry. 1993;32:713-718
  134. 134. Zhou Z, Tan H, Li Q , Chen J, Gao S, Wang Y, et al. CRISPR/Cas9-mediated efficient targeted mutagenesis of RAS in Salvia miltiorrhiza. Phytochemistry. 2018;148:63-70
  135. 135. Afrin S, Huang J, Luo Z. JA-mediated transcriptional regulation of secondary metabolism in medicinal plants. Scientific Bulletin. 2015;60(12):1062-1072
  136. 136. Goldhaber-Pasillas GD, Mustafa NR, Verpoorte R, et al. Jasmonic acid effect on the fatty acid and terpenoid indole alkaloid accumulation in cell suspension cultures of Catharanthus roseus. Molecules. 2014;19:10242-10260
  137. 137. Tang KX, Liu DH, Wang YL, et al. Overexpression of transcriptional factor ORCA3 increases the accumulation of catharanthine and vindoline in Catharanthus roseus hairy roots. Russian Journal of Plant Physiology. 2011;58:415-422
  138. 138. Li CY, Leopold AL, Sander GW, Shanks JV, Zhao L, Gibson S. The ORCA2 transcription factor plays a key role in regulation of the terpenoid indole alkaloid pathway. BMC Plant Biology. 2013;13:155
  139. 139. Pan Q , Wang Q , Yuan F, et al. Overexpression of ORCA3 and G10H in Catharanthus roseus plants regulated alkaloid biosynthesis and metabolism revealed by NMR-metabolomics. PLoS One. 2012;7:e43038
  140. 140. Suttipanta N, Pattanaik S, Kulshrestha M, et al. The transcription factor CrWRKY1 positively regulates the terpenoid indole alkaloid biosynthesis in Catharanthus roseus. Plant Physiology. 2011;157:2081-2093
  141. 141. Schluttenhofer C, Pattanaik S, Patra B, et al. Analyses of Catharanthus roseus and Arabidopsis thaliana WRKY transcription factors reveal involvement in jasmonate signaling. BMC Genomics. 2014;15:502-522
  142. 142. Balasubramanian M, Anbumegala M, Surendran R, Arun M, Shanmugam G. Elite hairy roots of Raphanus sativus (L.) as a source of antioxidants and flavonoids. 3 Biotech. 2018;8:128
  143. 143. Kundu S, Salma U, Ali MN, Hazra AK, Mandal N. Development of transgenic hairy roots and augmentation of secondary metabolites by precursor feeding in Sphagneticola calendulacea (L.) Pruski. Industrial Crops and Products. 2018;121:206-215
  144. 144. Lan X, Zeng J, Liu K, Zhang F, Bai G, Chen M, et al. Comparison of two hyoscyamine 6β-hydroxylases in engineering scopolamine biosynthesis in root cultures of Scopolia lurida. Biochemical and Biophysical Research Communications. 2018;497:25-31
  145. 145. Sharma A, Verma N, Verma P, Verma RK, Mathur A, Mathur AK. Optimization of a Bacopa monnieri-based genetic transformation model for testing the expression efficiency of pathway gene constructs of medicinal crops. In Vitro Cell Development and Biological Plant. 2017;53:22-32
  146. 146. Sharma A, Verma P, Mathur A, Mathur AK. Genetic engineering approach using early Vinca alkaloid biosynthesis genes led to increased tryptamine and terpenoid indole alkaloids biosynthesis in differentiating cultures of Catharanthus roseus. Protoplasma. 2018;255:425-435
  147. 147. Park SU, Facchini PJ. Agrobacterium rhizogenes mediated transformation of opium poppy, Papaver somniferum and California poppy, Eschscholzia californica cham root cultures. Journal of Experimental Botany. 2000;51:1005-1016
  148. 148. Argolo AC, Charlwood BV, Pletsch M. The regulation of solasodine production by Agrobacterium rhizogenes transformed roots of Solanum aviculare. Planta Medica. 2000;66:448-451
  149. 149. Shi HP, Kintzios S. Genetic transformation of Pueraria phaseoloides with Agrobacterium rhizogenes and puerarin production in hairy roots. Plant Cell Reports. 2003;21:1103-1107
  150. 150. Inamdar JA, Natraj M, Mohan JSS, Subramanian RB. Somatic embryogenesis from callus cultures of Crataeva nurvala Buch. Ham. Phytomorphology. 1990;40(3):280-284
  151. 151. Kumar HGA, Murthy HN, Paek KY. Somatic embryogenesis and plant regeneration in Gymnema sylvestre. Plan Cell Tissues Organs. 2002;71:85-88
  152. 152. Martin KP. Plant regeneration through somatic embryogenesis on Holostemma ada-Kodian, a rare medicinal plant. Plant Cell Tissue Organs. 2003;72:79-82
  153. 153. Patil MV. Micropropagation studies in Ceropegia spp. In Vitro Cell Development. 1998;34:240-243
  154. 154. Saha PS, Sarkar S, Jeyasri R, Muthuramalingam P, Ramesh M, Jha S. In Vitro Propagation, Phytochemical and Neuropharmacological Profiles of Bacopa monnieri (L.) Wettst.: A review. Plants. 2020;9:411. DOI: 10.3390/plants9040411
  155. 155. Tejavathi DH, Rajanna MD, Sowmya R, Gayathramma K. Induction of somatic embryos from cultures of Agave Veracruz Mill. In Vitro Cellular & Developmental Biology. 2007;43:423-428. DOI: 10.1007/s11627-007-9088-8
  156. 156. Hussain MS, Fareed S, Ansari S, Rahman A, Ahmad IZ, Saeed M. Current approaches toward production of secondary plant metabolites. Journal of Pharmacy & Bioallied Sciences. 2012;4(1):10-20. DOI: 10.4103/0975-7406.92725
  157. 157. Park SY, Paek KY. Bioreactor culture of shoots and somatic embryos of medicinal plants for production of bioactive compounds. In: Paek KY, Murthy HN, Zhong JJ, editors. Production of Biomass and Bioactive Compounds Using Bioreactor Technology. New York: Springer; 2014. pp. 337-368
  158. 158. Tabata M, Fujita Y. Production of shikonin by the plant cell cultures. In: Zatlin M, Day P, Hollaender A, editors. Biotechnology in the Plant Science. Cambridge: Academic Press; 1985. pp. 207-218
  159. 159. Hibino K, Ushiyama K. Commercial production of ginseng by the plant tissue culture technology. In: Fu TJ, Singh G, Curtis WR, editors. Plant Cell and Tissue Culture for Production of Food Ingredients. New York: Springer; 1999. pp. 215-224
  160. 160. Phyton Biotech. Capacity, reliability and quality in taxane API supply. 2014. Available from: https://phytonbiotech.com/apis/docetaxel/2014
  161. 161. Steingroewer J, Bley T, Georgiev V, et al. Bioprocessing of differentiated plant in vitro systems. Engineering in Life Sciences. 2013;13(1):26-38. DOI: 10.1002/elsc.201100226
  162. 162. Ferrie AM. Protocols for in vitro cultures and secondary metabolite analysis of aromatic and medicinal plants. Annals of Botany. 2010;105(4):vii-viii
  163. 163. Pohlscheidt M, Charaniya S, Bork C, Jenzsch M, Noetzel TL, Luebbert A. Bioprocess and Fermentation Monitoring. Hoboken: John Wiley & Sons; 2013
  164. 164. Su WW, Humphrey AE. Production of rosmarinic acid in highdensity perfusion cultures of Anchusa officinalis using a high sugar medium. Biotechnology Letters. 1990;12(11):793-798. DOI: 10.1007/BF01022597
  165. 165. Vanisree M, Chen YL, Shu-Fung L, Satish MN, Chien YL, HsinSheng T. Studies on the production of some important secondary metabolites from medicinal plants by plant tissue cultures. Botanical Bulletin of Academia Sinica. 2004;45:1-22
  166. 166. Madariaga-Mazón A, Hernández-Alvarado RB, Noriega-Colima KO, Osnaya-Hernández A, Martinez-Mayorga K. Toxicity of secondary metabolites. De Gruyter. 2019. DOI: 10.1515/psr-2018-0116
  167. 167. Sharma AK, Sharma MK. Plants as bioreactors: Recent developments and emerging opportunities. Biotechnology Advances. 2009;27(6):811-832
  168. 168. Shitan N. Secondary metabolites in plants: Transport and self-tolerance mechanisms. Bioscience, Biotechnology, and Biochemistry. 2016;80(7):1-11. DOI: 10.1080/09168451.2016.1151344
  169. 169. Siddiqui MS, Thodey K, Trenchard I, Smolke CD. Advancing secondary metabolite biosynthesis in yeast with synthetic biology tools. FEMS Yeast Research. 2012;12(2):144-170. DOI: 10.1111/j.1567-1364.2011.00774.x
  170. 170. Niazian M, Sadat-Noori SA, Galuszka P, Mortazavian M. Tissue culture-based Agrobacterium-mediated and in plant transformation methods. Czech Journal of Genetics and Plant Breeding. 2017;53(4):133-143. DOI: 10.17221/177/2016-CJGPB
  171. 171. Pandey S, Mishra A, Patel MK, Jha B. An Efficient method for Agrobacterium-mediated genetic transformation and plant regeneration in cumin (Cuminum cyminum L.). Applied Biochemistry and Biotechnology. 2013;171(1):1-9. DOI: 10.1007/s12010-013-0349-1
  172. 172. Fernando WGD, Zhang X, Amarasinghe CC. Detection of Leptosphaeria maculans and Leptosphaeria biglobosa causing blackleg disease in canola from Canadian canola seed lots and dockage. Plants. 2016;5:12. DOI: 10.3390/plants5010012
  173. 173. Rout GR, Samantaray S, Das P. In vitro manipulation and propagation of medicinal plants. Biotechnology Advances. 2000;18:91-120
  174. 174. Wang G, Qi NM. Perfusion culture of Glycyrrhiza inflata suspension cells in a stirtank bioreactor. Australian Journal of Botany. 2009;57(3):240-246. DOI: 10.1071/BT08187
  175. 175. Hassanein AM, Ahmed AM, Abdel-Hafez AII, Soltan DM. Phenol oxidizing isoenzymes and malate dehyderogenases patterns, and organogenesis of Solanum nigrum L. as affected by light treatments. Acta Agronomica Hungarica. 1999;47:127-136
  176. 176. Jones AMP, Saxena PK. Inhibition of Phenylpropanoid Biosynthesis in Artemisia annua L.: A Novel Approach to Reduce Oxidative Browning in Plant Tissue Culture. PLoS One. 2013;8(10):e76802. DOI: 10.1371/journal.pone.0076802
  177. 177. Malinski MP, Budzianowsk J, Kikowska M, Derda M, Jaworska MM, Mlynarczyk DT, et al. Two ecdysteroids isolated from micropropagated Lychnis floscuculi and the biological activity of plant material. Molecules. 2021;26:904. DOI: 10.3390/molecules26040904
  178. 178. Kour B, Kour G, Kaul S, Dhar MK. In vitro mass multiplication and assessment of genetic stability of in vitro raised Artemisia absinthium L. plants using ISSR and SSAP molecular markers. Advances in Botany. 2014;2014:7
  179. 179. Massot B, Milesi S, Gontier E, Bourgaud F, Guckert A. Optimized culture conditions for the production of furanocoumarins by micropropagated shoots of Ruta graveolens. Plant Cell, Tissue and Organ Culture. 2000;62:11-19
  180. 180. Begum F. Augmented production of vincristine in induced tetraploids of Agrobacterium transformed shooty teratomas of Catharanthus roseus. Medicinal Plants. 2011;3:59-64
  181. 181. Krolicka A, Szpitter A, Stawujak K, Baranski R, Gwizdek-Wisniewska A, Skrzypczak A, et al. Teratomas of Drosera capensis var. alba as a source of naphthoquinone: Ramentaceone. Plant Cell, Tissue and Organ Culture. 2010;103:285-292
  182. 182. Chen J, Ziv M. The effect of ancymidol on hyperhydricity, regeneration, starch and antioxidant enzymatic activities in liquid-cultured Narcissus. Plant Cell Reports. 2001;20:22-27
  183. 183. Isah T, Mujib A. In vitro propagation & camptothecin production in Nothapodytes nimmoniana. Plant Cell, Tissue and Organ Culture. 2015;121:1-10. DOI: 10.1007/s11240-014-0683-1
  184. 184. Salem JM. Effects of anti-ethylene compounds on vitrification and genome fidelity of Stevia rebaudiana Bertoni. Egyptian Journal of Botany. 2020;60:519-535
  185. 185. Debergh PC, Harbaoui Y, Lemeur R. Mass propagation of globe artichoke (Cynara scolymus): Evaluation of different hypotheses to overcome vitrification with special reference to water potential. Physiologia Plantarum. 1981;53:181-187

Written By

Ahmed M. Hassanein

Submitted: 20 April 2022 Reviewed: 05 May 2022 Published: 31 May 2022