Open access peer-reviewed chapter

Production of Plant Secondary Metabolites by Using Biotechnological Tools

Written By

Sandra Gonçalves and Anabela Romano

Submitted: 08 November 2017 Reviewed: 12 March 2018 Published: 05 September 2018

DOI: 10.5772/intechopen.76414

From the Edited Volume

Secondary Metabolites - Sources and Applications

Edited by Ramasamy Vijayakumar and Suresh S.S. Raja

Chapter metrics overview

5,388 Chapter Downloads

View Full Metrics

Abstract

Plants are a remarkable source of high-value secondary metabolites with applications in various domains. Plant cell and tissue culture techniques appear as environmentally friendly alternatives for the production of secondary metabolites when natural supply is limited or chemical synthesis is unviable. In this chapter, the main advantages of using plant cell and tissue culture techniques for the production of plant secondary metabolites are presented as well as the different biotechnological approaches available to improve their production. In addition, the production of anticancer compounds (camptothecin, podophyllotoxin, taxol, vinblastibe, and vincristine) and metabolites from Lamiaceae spp. (phenolics as rosmarinic acid) were selected as examples to be highlighted. The study reviewed shows that undifferentiated cells are the preferred culture system used for the production of high-value secondary metabolites in vitro although there are many examples reporting the production in differentiated tissues particularly in hairy roots. Efforts have been made to scale up the production, and several strategies have been successfully applied to increase the production yields at the laboratorial scale. Nevertheless, there are only few examples of plant secondary metabolites production at commercial level, and further in-depth studies are still required.

Keywords

  • alkaloids
  • anticancer compounds
  • cell suspension cultures
  • elicitation
  • Lamiaceae
  • metabolic engineering
  • phenolics

1. Introduction

Plant kingdom, comprising about 250,000 species, is a repository of probably hundreds of thousands of low-molecular-weight structurally complex chemical compounds known as secondary metabolites [1]. These high-value metabolites are biosynthesized through phenylpropanoid, mevalonate, 2-C-methyl-d-erythritol-4-phosphate, amino acid, glucose, acetate-malonate pathway, or combined pathways. Secondary metabolites have an important role in the interaction between plants and their environment (e.g., defense against herbivores and pathogens, protection against ultraviolet light, etc.) and, thus, are vital for their existence and subsistence. They are accumulated in specific tissues and structures (e.g., vacuoles, specialized glands, trichomes, etc.), and their production is affected by several factors, like genotype, plant physiology, climate, environmental conditions, and pathogens; in some cases, they are only produced during certain developmental stages [2, 3].

Over the past decades, efforts have been directed at the extraction, structure elucidation, and evaluation of biological activity of many plant secondary metabolites. Plants continue to be the main source for many important bioactive molecules/pharmacophores [4, 5]. About 25–28% of modern medicines are derived from higher plants [6], and over 60% of anticancer drugs are directly or indirectly derived from plants [7]. According to a recent report of the British Broadcasting Corporation (BBC), plant-derived drugs will grow from $29.3 billion in 2017 to around $39.2 billion by 2022 with an annual growth rate of 5.9% [8].

In the last decades, considerable progress has been made concerning the production of secondary metabolites by using plant tissue culture techniques owing to the advantages of this platform over other production systems as discussed in the next section of this chapter. The most studied classes of plant secondary metabolites using plant cell and tissue culture production systems are alkaloids and the landmark example is the anticancer-registered drug Taxol® [3]. Plant tissue culture techniques were even endorsed by Food and Agriculture Organization as safe for the production of compounds for food application [9]. This chapter aims to discuss the main advantages of using plant cell and tissue culture techniques for the production of plant secondary metabolites as well as the different biotechnological approaches available to improve their production. Important and representative examples produced through these methods, as is the case of plant anticancer compounds and metabolites from Lamiaceae spp., are addressed.

Advertisement

2. Advantages of plant tissue culture techniques for the production of secondary metabolites

In a context where consumers demand for safe natural products increases, because synthetic chemicals are perceived as potentially toxic, the interest in plant secondary metabolites from research and industry also increases [10]. Few important plant products with simple chemical structures can be produced via chemosynthesis; however, many compounds like alkaloids are difficult to be synthesized or the cost of their synthesis outweighs their commercial availability [11, 12]. Some compounds can be obtained from naturally grown plants, but sometimes there are regional and environmental restrictions, which can limit the commercial production [13]. Also, traditional cultivation of some plant species is difficult or takes several years. In this context, plant cell and tissue culture techniques appear as environmentally friendly alternative methods for the production of secondary metabolites when natural supply is limited and traditional methods are unfeasible. The mass propagation of plants in aseptic and environmental controlled conditions, and the large-scale production of secondary metabolites in a year-round system without seasonal constraints, are some of the advantages of plant tissue culture techniques [3]. Moreover, cultures can be established in any part of the world independently of the plant growth requisites and are free of microbes and insects avoiding the use of pesticides and herbicides [14, 15]. Plant tissue culture techniques provide a reliable and predictable method for isolating the secondary metabolites at a high efficiency within a short time when compared to the extraction from wild plant populations [16]. Also, the simplicity in the extraction of the metabolites from in vitro-produced tissues makes the method appellative for commercial application [17].

Apart from the abovementioned advantages, there are metabolites that are generally not found in the intact plant but can be produced by in vitro cultures [18]. Biotechnology opens the opportunity to apply traditional or metabolic engineering strategies to promote the accumulation of desired compounds by in vitro cultures. Products from in vitro cultures can still be used as models of whole plants, and cell cultures can be radiolabeled so that secondary products can be traced metabolically [19].

The massive and indiscriminate collection of plant material from plants producing important bioactive compounds has threatened the survival of some species. In vitro propagation through plant tissue culture techniques allows the large-scale multiplication of true-to-type plants within a short span of time and without a negative impact on the natural resources [16]. This method is particularly valuable for plants difficult to propagate by conventional techniques or with slow propagation rates. In this context, in the last years, there has been an increased interest on the use of these methodologies for the propagation and conservation of medicinal plants.

Advertisement

3. Culture systems

The production of secondary metabolites by in vitro cultures usually occurs in a two-step process, biomass accumulation and secondary metabolites synthesis, in which both steps need to be optimized independently [3, 14]. Production could be accomplished by using undifferentiated calli, cell suspension cultures, or organized structures like shoots, roots, or somatic embryos. In some cases, a certain degree of differentiation may be needed for the biosynthesis to occur [20]. The use of differentiated organ cultures is required, for instance, when the target metabolite is only produced in specialized plant tissues or glands as is the case of essential oils [20, 21].

Among differentiated tissues, hairy roots culture offers new opportunities for the in vitro production of plant-valuable compounds [22]. Hairy roots are induced by the infection of plants with Agrobacterium rhizogenes, a Gram-negative soil bacterium. During the infection, a DNA segment (T-DNA) from the large root-inducing (Ri) plasmid of the bacterium is transferred into the genome of the infected plant. The higher level of cellular differentiation, rapid growth, genetic and biochemical stability, and maintenance facility are some of the advantages of hairy roots [22]. Also, they can accumulate metabolites in the aerial parts of the plant. However, the difficulties in cultivating hairy roots in an industrial system limit their commercial use to produce valuable plant secondary metabolites.

Although there are many studies reporting the production of secondary metabolites using callus cultures and differentiated tissues [3, 14, 23], in most cases, undifferentiated cells are the preferred culture system [13]. Cell suspension culture is a simple and cost-effective method that has been extensively used to overcome the problems of large-scale production. Plant cell is biosynthetically totipotent, which means that under suitable conditions, each cell has theoretically the capacity to produce compounds identical to those present in the parent plant [13]. Plant cell cultures have more immediate potential for commercial application than tissue or organ cultures [21, 24]. They are considered as a stable system for the continuous production of secondary metabolites of uniform quality and yield. Another great advantage of plant cell cultures is the possibility to synthesize novel products not usually produced by the native plant [25, 26]. This is the preferable biotechnological platform to produce high-value secondary metabolites, as taxol [27, 28], resveratrol [29], artemisinin [30], ginsenosides [31], and ajmalicine [32].

Advertisement

4. Strategies to improve the production of secondary metabolites

In the commercial exploitation of plant cell cultures for the production of high-value secondary metabolites, it is fundamental to achieve high yields and consistent productions. The production of secondary metabolites in plants is genotype-dependent; thus, the first step to initiate cell or organ cultures is the choice of the parent plant containing higher contents of the secondary product of interest for callus or organ induction, and the selection of high-producing cell/organ lines [14]. The selection is made by analyzing cell/organ growth and then by quantifying the desired product by chromatographic and spectroscopic techniques [14]. Nevertheless, even selecting a highly productive line, the production yields are not always adequate, and after long periods of cultivation they lose their production efficiency. Thus, many alternative strategies can be used to stimulate the production of secondary metabolites and obtain efficient yields including traditional and metabolic engineering strategies [3, 19].

4.1. Traditional strategies

There are several factors that can be optimized to improve the growth and metabolites productivity of the in vitro cultures. Among them, the following can be appointed: the culture medium composition, the medium pH, the inoculum density, the culture medium environment (e.g., temperature, light density and quality, etc.), the agitation and aeration, etc. [3, 14, 15]. The culture medium strongly affects the biomass and metabolites productivity, and thus the selection of the suitable culture medium formulation is an imperative step [3]. It must be selected according to the physiological requirements of the plant species, and there are several parameters that can be optimized, namely nutrients composition, salt strength, nitrate and phosphate levels, plant growth regulators type and concentration, carbon source, etc. For instance, carbon source plays significant roles in the signal transduction systems through regulating gene expression and developmental processes [3].

Secondary metabolites are produced by plant cells in response to environmental stimuli or as defensive mechanisms against invading pathogens. In this sense, the strategy available to improve the productivity of secondary metabolites, elicitation, aims to misguide the cells or tissues for a possible biotic/abiotic attack by using agents that trigger the defense response [33]. Elicitors have the ability to control an array of cellular activities at the biochemical and molecular level since they induce the upregulation of genes [33]. The elicitors can be biotic or abiotic and may comprise signaling molecules like methyl jasmonate, salicylic acid, microbial cell wall extracts (e.g., yeast extract, chitosan), inorganic salts, heavy metals, physical agents (e.g., UV radiation) among others [1, 34]. Methyl jasmonate and its related signal molecules, and salicylic acid are probably the most extensively used elicitors [5]. The combination of some elicitors with physical factors (e.g., UV light, temperature regime, and pulsed electric field) yielded good results for secondary metabolite production [35]. As reviewed by Giri and Zaheer [5], cell suspension culture is the most used culture system for elicitation treatment and secondary metabolites production. Due to its inherent characteristics of hormone autotrophy, uncontrolled growth, biosynthetic, and genetic stability distinctiveness, hairy root cultures have proved to be also a valuable culture system for elicitation experiments. In addition, there are some secondary metabolites that are synthesized only in the roots [14, 36, 37]. Multiple shoots culture is a less used culture system for elicitation treatments for the production of secondary metabolites which is particularly useful in the case of metabolites present in the leaves [5]. The elicitors can change the secondary metabolites production quantitatively and also qualitatively [5]. For extra information, consult the recent reviews on this subject [1, 5].

Nutrient and precursor feeding are also used to improve the yields of secondary metabolites production. Nutrient feeding involves the replenishment of nutrient medium, and in precursor feeding, plant cell cultures are used to convert precursors into products by utilizing preexisting enzyme systems [14]. Immobilization of plant cells is another strategy used to overcome problems of low shear resistance and cell aggregation. This procedure can be done by several methods, and the most widely used are surface immobilization or gel entrapment. In this technique, the cells are entrapped in a specific gel or a combination of gels. Examples of matrices used are calcium alginate (the most used), agarose, gelatin, carrageenan, or polyacrylamide [14]. This strategy has several advantages, such as the extension of cells’ viability in the stationary stage, the simplification of downstream processing, the high-cell density within small bioreactors reducing the costs and risk of contamination, an increased product accumulation, the minimization of fluid viscosity, among others [38].

The permeabilization of plant cell membranes with chemicals, the use of electric field stress, and ultrasound techniques are strategies used to facilitate the removal of secondary metabolites from vacuoles and membrane systems of the plant cell, facilitating the secretion of products into the culture medium and thus simplifying the purification process [14, 17].

The cultivated cells have the capacity for biotransformation of supplied compounds, which are not necessarily natural intermediaries of plant metabolism, into high-value compounds. This can occur through different reactions as hydroxylation, oxidation of hydroxyl group, reduction of carbonyl group, hydrogenation of carbon-carbon double bond, glycosyl conjugation, and hydrolysis, catalyzed by plant enzymes [14]. This is probably one of the most commercially realistic approaches; however, in some cases, the costly precursors may limit the economic viability [38].

4.2. Metabolic engineering

Metabolic engineering offers a new perspective to understand the expression of genes involved in the biosynthesis of secondary metabolites through overexpression studies allowing the alteration of biosynthetic pathways [39, 40]. This involves the study of enzymatic reactions and biosynthetic processes at gene, transcriptomic, and proteomic levels, and the manipulation of the genes encoding the critical and rate-limiting enzymes in the biosynthetic pathways [41, 42]. Theoretically, the secondary metabolites productivity of plant cell cultures can be improved through the overexpression of genes encoding regulatory enzymes involved in their biosynthetic pathways [16]. However, the overexpression of certain genes may not always improve production [16].

Metabolic engineering approach also uses the inhibition of competitive pathways to increase metabolic flux of targeted biosynthetic pathway intermediates for a higher production through a variety of approaches. Certain steps in the biosynthetic pathway could be inhibited to induce the accumulation of preceding intermediates. The understanding of phenylpropanoyl biosynthetic pathway that is involved in the biosynthesis of several plant secondary metabolites is the most successful and recent application [43, 44].

The in-depth understanding of the biosynthetic pathways is still a barrier to the practical use of this strategy to enhance production [45, 46]. For the large-scale production of important secondary metabolites to meet industry demand, more studies are needed to identify rate-limiting steps and regulation along with bottlenecks on the lack of clarity of their biosynthetic pathways.

Advertisement

5. Scale-up production

Owing to the importance of some plant secondary metabolites, efforts have been made to study the feasibility of their production at the industrial scale. This is not always a simple process because plant cells have a relatively unstable productivity, a high shear sensitivity, a slow growth rate, and low oxygen requirements [14]. The scale-up involves the use of bioreactors of varying sizes and features, and cell suspension culture is the better culture system having several advantages in comparison with the other. The simplicity, predictability, and high efficiency at which the metabolites can be isolated from biomass or cultivation media are some of these advantages. Nevertheless, there are some examples of the use of differentiated tissues like shoots and somatic embryos [47].

Some important milestones in the production of secondary metabolites by plant cell cultures are the production of shikonin [48] and ginseng [49], and the most successful example of the scale-up process is probably the production of taxol by Phyton Biotech Company (Germany) to supply part of the demands of Bristol-Meyers Squibb Company during the year 2002 [50]. Phyton Biotech operates the largest cGMP plant cell culture facility in the world designed for large-scale production of Taxanes in 75,000 L-size bioreactors that run up to 880,000 L per year [51]. Berberine, ginsenosides, shikonin, scopolamine, and rosmarinic acid are also examples of plant secondary metabolites presently produced at the commercial scale (Figure 1) [3, 17].

Figure 1.

Structures of some relevant plant secondary metabolites produced on a commercial scale.

Several factors should be considered in scaling up the production of secondary metabolites using bioreactors, namely the optimization of culture conditions, biomass production measurement (especially with tissue and organ cultures), and so on [52, 53]. Several bioreactor designs have been tested and used for plant cell cultures. Some of them as is the case of stirred tank reactors, bubble column reactors, airlift reactors, and ebb and flood reactors are merely extension of microbial culture. For plant cells with a high shear sensitivity, Wang and Zhong [54] develop the centrifugal impeller bioreactors that are based on the principles of a centrifugal pump. Mechanically driven “wave reactors,” “slug bubble reactor,” and “undertow reactor” are also adequate for high shear-stress-sensitive cells [14]. On the other hand, airlift bioreactors are suitable for not highly shear-sensitive cells and for hairy and adventitious root cultures [14]. The interested reader can find more important details about the scale-up process in the works by Murphy et al. [14], Yue et al. [13], and Isah et al. [3].

Advertisement

6. Selected examples

There are several plant secondary metabolites including among others alkaloids, terpenes, flavonoids, and glycosides, which can be produced by plant tissue culture techniques using different strategies [3, 13, 14]. Two examples were selected to be described in this chapter: the production of important anticancer compounds and the production of metabolites from Lavandula spp.

6.1. Anticancer compounds

As mentioned before in this chapter, over 60% of anticancer drugs are directly or indirectly derived from plants [7]. The search for anticancer compounds from plants started in the 1950s when the alkaloids vinblastine and vincristine from Catharanthus roseus (L.) G. Don and podophyllotoxin from Podophyllum spp. were discovered. The United States National Cancer Institute initiated an extensive program in 1960 that led to the discovery of many novel chemotypes with cytotoxic activities [55], taxanes and camptothecins being some of the examples [7]. Camptothecin, podophyllotoxin, taxol, vinblastine, or vincristine are the most important plant-derived anticancer compounds [19, 56]. Most compounds with anticancer properties are alkaloids, and some of them have a complex structure, with multiple rings and chiral centers, and therefore the chemical synthesis is prohibitively expensive [17]. Plant cell and tissue culture techniques appear as environmentally friendly alternative methods for the production of these secondary metabolites [17, 19].

Taxanes from Taxus spp., terpenoid indole alkaloids from C. roseus, camptothecin from Camptotheca acuminata Decne among other species, and podophyllotoxin from Podophyllum and Linum spp. are the main compounds produced by using biotechnological approaches (Table 1). For the production of taxanes, cell suspension cultures are definitively the most adequate culture system. However, some studies demonstrated that differentiated tissues are more adequate than undifferentiated cells to produce other anticancer compounds. For instance, intact plants of C. acuminata contain around 0.2–5 mg/g dry weight (DW) of camptothecin while callus and suspension cultures produced only 0.002–0.004 mg/g DW or lesser [57]. Hairy root cultures have also proven to be a good option for in vitro production of secondary metabolites as indole alkaloids due to their higher level of cellular differentiation and improved genetic or biochemical stability. The hairy roots of Ophiorrhiza pumila Champ. ex Benth showed a high capacity to produce camptothecin (0.1% DW), although the callus culture failed to produce this compound [58].

Compound(s)GroupSource (plant species)Culture systemReference(s)
CamptothecinMonoterpene indole alkaloidCamptotheca acuminata DecneHRC[75]
Camptotheca acuminata DecneCSC[76]
Nothapodytes foetida (Wight) SleumerCC[77]
Nothapodytes nimmoniana (J. Grah.)CSC[78]
Ophiorrhiza alata CraibHRC[79]
Ophiorrhiza mungos Linn.CSC[80]
Ophiorrhiza prostata D. DonARC[81]
Ophiorrhiza pumila Champ. ex Benth.HRC[58]
PodophyllotoxinAryltetralin lignanLinum spp.HRC[82, 83]
Linum album Kotschy ex Boiss.CSC[84]
Taxanes (taxol)Diterpene alkaloidsTaxus spp.CSC[27, 28]
Corylus avellana L.CSC[85]
Vinblastine and vincristineTerpene indole alkaloidsCatharanthus roseus (L.) G. DonHRC[86, 87]

Table 1.

Some examples of studies reporting the production of plant anticancer compounds using biotechnological approaches.

ARC: adventitious root culture; HRC: hairy root culture; CSC: cell suspension cultures; CC: callus culture.

Several researchers have focused on studies aiming for the optimization of biomass growth conditions and on the application of biotechnological strategies to increase production yields of anticancer compounds. By manipulating empirical factors related to plant cell and organ cultures, it has been possible to enhance production yields. Several factors have been optimized, such as nutrients, carbon source, plant growth regulators, or culture environmental conditions, and several biotic and abiotic elicitors have been tested. Studies have also been focused on the elucidation and regulation of biosynthetic pathways and on aiming the increase of production yields of anticancer compounds as taxanes [41] and indole alkaloids [59] by using elicitors to activate genes involved in metabolic pathways. In spite of all the advantages of producing anticancer compounds by using plant cell and tissue culture techniques and the significant advancements in the last years, the examples of the production of plant anticancer compounds on an industrial level are scarce. As previously mentioned in this chapter, the best success example is the production of taxanes by the Germany company Phyton Biotech [51].

Plant cell and tissue culture techniques have also been applied for the propagation of several anticancer plants. In vitro propagation allows the rapid mass multiplication of true-to-type plants within a short span of time which is particularly important in the case of endangered species. Some recently selected examples comprise plants producing the important anticancer compounds camptothecin [60, 61] and podophyllotoxin [62, 63].

6.2. Lamiaceae spp. metabolites

The mint family (Lamiaceae) contains about 236 genera and more than 7000 species with cosmopolitan distribution [64]. Some of the most important genera are Hyptis, Lavandula, Nepeta, Salvia, Scutellaria, Thymus, and Teucrium. Species from the family inhabit different natural ecosystems, and many are already cultivated. Most of the species belonging to this family are aromatic (possess essential oils) and are widely used in traditional medicine to cure various disorders. They also have great economic value due to their use in culinary or as ornamentals, and for cosmetic, flavoring, fragrance, perfumery, pesticide, and pharmaceutical applications [65]. Many Lamiaceae contain high levels of phenolics, which are probably the most relevant group of secondary metabolites synthesized by plants due to their health promotion effects [64]. Among phenolic compounds, rosmarinic acid is present in the tissues of many of these species being used as a chemical marker of the family [64, 66, 67]. In some species, this compound is accumulated as the main phenolic compound at a concentration above 0.5% dry weight [64]. Several species in the Lamiaceae family can also accumulate high levels of other phenolic acids, flavonoids, or phenolic terpenes [64]. There are some phenolic compounds as carnosic and clerodendranoic acids that are exclusive from this family [68, 69]. The interested reader can find an excellent overview on the phytochemical characterization and biological effects of Lamiaceae species in Trivellini et al. [64].

Phenolic compounds are generally produced as a defense mechanism or as a response to stressful environment conditions [9]. The activation of these protective mechanisms by applying stress stimulus can be used as a strategy to increase the production of phenolic compounds in plant cell and organ cultures [70]. Recently, several attempts were made regarding the production of secondary metabolites by several Lamiaceae species (mainly phenolics) using plant tissue cultures particularly applying elicitation as a strategy to achieve higher production yields [64]. These studies involve mainly the use of chemical elicitors like jasmonic acid (or methyl jasmonate), or physical elicitors as UV-B and ozone (O3), to increase the production of many compounds as essential oil constituents, phenylpropanoids, flavonoids, and phenolic acids. Overall, the results demonstrated that these elicitors had an immediate effect on enhancing the production of phenolics [64].

The revised study showed that a high number of studies reported an increase in the production of rosmarinic acid after elicitation of cultures of several Lamiaceae, such as Coleus, Lavandula, and Salvia genera [64, 66, 71]. Several studies reported the increase in rosmarinic acid production through the application of elicitors (Table 2). Elicitation with jasmonic acid induces a 4.6-fold increase of rosmarinic acid production in L. officinalis L. cell suspension cultures [72], and elicitation with methyl jasmonate induces a 3.4-fold increase in C. forskohlii (Willd.) Briq. hairy root cultures [73]. The production of this compound also increased (2.3-fold) in leaves of Rosmarinus officinalis L. after 14 days of UV-B exposure [74]. Recently, rosmarinic acid attracted the attention of the scientists due to its broad range of biological activities, such as anti-inflammatory, antioxidant, cognitive-enhancing, cancer chemoprotection effects, among others [71]. In the last years, there are many progresses in the biotechnological production of this compound but its large-scale production still requires further optimization. The molecular understanding of its biosynthesis and the application of metabolic engineering tools are crucial to improve the biotechnological production.

Plant speciesTissueElicitorReference
Coleus blumei Benth.HRCMethyl jasmonate[88]
Coleus forshohlii (Willd.) Briq.HRCMethyl jasmonate[73]
Lavandula officinalis ChaixCSCJasmonic acid[72]
Lavandula vera MMCSCVanadyl sulphate[89]
Lavandula vera MMCSCMethyl jasmonate[90]
Mentha × piperitaCSCMethyl jasmonate[91]
Rosmarinus officinalis L.LUV-B[74]

Table 2.

Selected examples of elicitation treatments applied in cultures of Lamiaceae species to increase the production of rosmarinic acid.

HRC: hairy root culture; CSC: cell suspension cultures; L: leaves.

Advertisement

7. Conclusions and prospects

Plant cell and tissue culture techniques are an attractive system for the cultivation of a broad range of secondary metabolites, including important alkaloids with anticancer properties and bioactive phenolics. This alternative provides a continuous, sustainable, economical, and viable production of secondary metabolites, independent of geographic and climatic conditions, which is particularly useful for the production of species at risk. Despite the great progresses in this area in the last decades, in some cases, production occurs at very low yields, and there are many difficulties in scaling up the production, and limited commercial success is achieved. Incomplete knowledge about the biosynthetic pathways of bioactive molecules limited the improvement of the production yields. Exploiting modern molecular biology techniques emerged as an alternative that needs to be harnessed to improve production efficiency by engineering biosynthetic pathway(s) of the molecules in plant cells. Also promising are new elicitors and permeabilizing agents such as coronatin or cyclodextrins. The production of bioactive molecules in endophytes also appears as an attractive alternative, although till date, there is no reported commercial exploitation.

Advertisement

Acknowledgments

This work is supported by National Funds—FCT (Fundação para a Ciência e a Tecnologia, I.P.), through the project UID/BIA/4325/2013. S. Gonçalves acknowledges a grant from the FCT (SFRH/BPD/84112/2012) financed by POPH-QREN and subsidized by the European Science Foundation.

References

  1. 1. Narayani M, Srivastava S. Elicitation: A stimulation of stress in in vitro plant cell/tissue cultures for enhancement of secondary metabolite production. Phytochemistry Reviews. 2017;16:1227-1252. DOI: 10.1007/s11101-017-9534-0
  2. 2. Shitan N. Secondary metabolites in plants: Transport and self-tolerance mechanisms. Bioscience, Biotechnology and Biochemistry. 2016;80:1283-1293. DOI: 10.1080/09168451.2016.1151344
  3. 3. Isah T, Umar S, Mujib A, Sharma MP, Rajasekharan PE, Zafar N, Frukh A. 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. DOI: 10.1007/s11240-017-1332-2
  4. 4. Dias DA, Urban S, Roessner U. A historical overview of natural products in drug discovery. Metabolites. 2012;2:303-336. DOI: 10.3390/metabo2020303
  5. 5. 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. 2016;126:1-18. DOI: 10.1007/s11240-016-0985-6
  6. 6. Samuelsson G. Drugs of Natural Origin: A Textbook of Pharmacognosy. 5th ed. Stock-holm: Swedish Pharmaceutical Press; 2004
  7. 7. Cragg GM, Newman DJ. Plants as a source of anticancer agents. Journal of Ethnopharma-cology. 2005;100:72-79. DOI: 10.1016/j.jep.2005.05.011
  8. 8. British Broadcasting Corporation (BBC) Research. Plant-derived Drugs: Global Markets. 2017. Available from: https://www.bccresearch.com/market-research/biotechnology/botanical-and-plant-derived-drugs-global-markets-bio022h.html [Accessed: 2018-02-08]
  9. 9. Dias MI, Sousa MJ, Alves RC, Ferreira ICFR. Exploring plant tissue culture to improve the production of phenolic compounds: A review. Industrial Crops and Products. 2016;82:9-22. DOI: 10.1016/j.indcrop.2015.12.016
  10. 10. Lucera A, Costa C, Conte A, Del Nobile MA. Food applications of natural antimicrobial compounds. Frontiers in Microbiology. 2012;3:287. DOI: 10.3389/fmicb.2012.00287
  11. 11. Stevenson DD, Szczeklik A. Clinical and pathologic perspectives on aspirin sensitivity and asthma. Journal of Allergy and Clinical Immunology. 2006;118:773-786. DOI: 10.1016/j.jaci.2006.07.024
  12. 12. Greger H. Phytocarbazoles: Alkaloids with great structural diversity and pronounced biological activities. Phytochemistry Reviews. 2017;16:1095-1153. DOI: 10.1007/s11101-017-9521-5
  13. 13. Yue W, Ming Q-L, Lin B, Rahman K, Zheng C-J, Han T, Qin L-P. Medicinal plant cell suspension cultures: Pharmaceutical applications and high-yielding strategies for the desired secondary metabolites. Critical Reviews in Biotechnology. 2016;36:215-232. DOI: 10.3109/07388551.2014.923986
  14. 14. Murthy HN, Lee EJ, Paek KY. Production of secondary metabolites from cell and organ cultures: Strategies and approaches for biomass improvement and metabolite accumulation. Plant Cell Tissue and Organ Culture. 2014;118:1-16. DOI: 10.1007/s11240-014-0467-7
  15. 15. Ochoa-Villarreal M, Howat S, Hong S, Jang MO, Jin Y-W, Lee E-K, Loake GJ. Plant cell culture strategies for the production of natural products. BMB Reports. 2016;49:149-158. DOI: 10.5483/BMBRep.2016.49.3.264
  16. 16. Verpoorte R, Contin A, Memelink J. Biotechnology for the production of plant secondary metabolites. Phytochemistry Reviews. 2002;1:13-25. DOI: 10.1023/A:1015871916833
  17. 17. Kolewe ME, Gaurav V, Roberts SC. Pharmaceutical active natural product synthesis and supply via plant cell culture technology. Molecular Pharmaceutics. 2008;5:243-256. DOI: 10.1021/mp7001494
  18. 18. Pavlov A, Popov S, Kovacheva E, Milen Georgiev M, Ilieva M. Volatile and polar compounds in Rosa damascena Mill 1803 cell suspension. Journal of Biotechnology. 2005;118:89-97. DOI: 10.1016/j.jbiotec.2005.03.005
  19. 19. Khani S, Barar J, Movafeghi A, Omidi Y. Production of anticancer secondary metabolites: Impacts of bioprocess engineering. In: Orhan IE, editor. Biotechnological Production of Secondary Metabolites. Benthan eBooks; 2012. pp. 215-240
  20. 20. Karuppusamy S. A review on trends in production of secondary metabolites from higher plants by in vitro tissue, organ and cell cultures. Journal of Medicinal Plants Research. 2009;3:1222-1239
  21. 21. Rao SR, Ravishankar GA. Plant cell cultures: Chemical factories of secondary metabolites. Biotechnology Advances. 2002;20:101-153. DOI: 10.1016/S0734-9750(02)00007-1
  22. 22. Chandra S, Chandra R. Engineering secondary metabolite production in hairy roots. Phytochemical Reviews. 2011;10:371-395. DOI: 10.1007/s11101-011-9210-8
  23. 23. Ali M, Isah T, Mujib A, Dipti. Climber plants: Medicinal importance and conservation strategies. In: Shahzad A, Sharma S, Siddiqui SA, editors. Biotechnological Strategies for the Conservation of Medicinal and Ornamental Climbers. New York: Springer; 2016. pp. 101-138
  24. 24. Xu J, Ge X, Dolan MC. Towards high-yield production of pharmaceutical proteins with plant cell suspension cultures. Biotechnology Advances. 2011;29:278-299. DOI: 10.1016/j.biotechadv.2011.01.002
  25. 25. Zhang X, Ye M, Dong Y, Hu HB, Tao SJ, Yin J, Guo DA. Biotransformation of bufadienolides by cell suspension cultures of Saussurea involucrata. Phytochemistry. 2011;72:1779-1785. DOI: 10.1016/j.phytochem.2011.05.004
  26. 26. de Pádua RM, Meitinger N, Filho JDS, Waibel R, Gmeiner P, Braga FC, Kreis W. Biotransformation of 21-O-acetyl-deoxycorticosterone by cell suspension cultures of Digitalis lanata (strain W. 1.4). Steroids. 2012;77:1373-1380. DOI: 10.1016/j.steroids.2012.07.016
  27. 27. Patil RA, Lenka SK, Normanly J, Walker EL, Roberts SC. Methyl jasmonate represses growth and affects cell cycle progression in cultured Taxus cells. Plant Cell Reports. 2014;33:1479-1492. DOI: 10.1007/s00299-014-1632-5
  28. 28. Sharma K, Zafar R. Optimization of methyl jasmonate and β-cyclodextrin for enhanced production of taraxerol and taraxasterol in Taraxacum officinale Weber cultures. Plant Physiology and Biochemistry. 2016;103:24-30. DOI: 10.1016/j.plaphy.2016.02.029
  29. 29. Cai Z, Knorr D, Smetanska I. Enhanced anthocyanins & resveratrol production in Vitis vinifera cell suspension culture by indanoyl-isoleucine, N-linolenoyl-l-glutamine, and insect saliva. Enzyme and Microbial Technology. 2012;50:29-34. DOI: 10.1016/j.enzmictec.2011.09.001
  30. 30. Baldi A, Dixit VK. Yield enhancement strategies for artemisinin production by suspension cultures of Artemisia annua. Bioresource and Technology. 2008;99:4609-4614. DOI: 10.1016/j.biortech.2007.06.061
  31. 31. Jeong C, Murthy HN, Hahn E, Paek K. Improved production of ginsenosides in suspension cultures of ginseng by medium replenishment strategy. Journal of Bioscience and Bioengineering. 2008;105:288-291. DOI: 10.1263/jbb.105.288
  32. 32. Ten Hoopen HJG, Vinke JL, Moreno PRH, Verpoorte R, Heijnen JJ. Influence of temperature on growth and ajmalicine production by Catharantus roseus suspension cultures. Enzyme and Microbial Technology. 2002;30:56-65. DOI: 10.1016/S0141-0229(01)00456-2
  33. 33. Zhao J, Davis LC, Verpoorte R. Elicitor signal transduction leading to the production of the plant secondary metabolite. Biotechnology Advances. 2005;23:283-333. DOI: 10.1016/j. biotechadv.2005.01.003
  34. 34. Ramirez-Estrada K, Vidal-Limon H, Hidalgo D, Moyano E, Golenioswki M, Cusidó RM, Palazon J. Elicitation, an effective strategy for the biotechnological production of the bioactive high-added value compounds in plant cell factories. Molecules. 2016;21:182 https://doi.org/10.3390/molecules21020182
  35. 35. Saw NMMT, Riedel H, Cai Z, Kutuk O, Smetanska I. Stimulation of anthocyanin synthesis in grape (Vitis vinifera) cell cultures by pulsed electric fields and ethephon. Plant Cell, Tissue and Organ Culture. 2012;108:47-54. DOI: 10.1007/s11240-011-0010-z
  36. 36. Srivastava S, Srivastava AK. Hairy root culture for mass-production of high-value secondary metabolites. Critical Reviews in Biotechnology. 2007;27:29-43. DOI: 10.1080/07388550601173918
  37. 37. Zaheer M, Reddy VD, Giri CC. Enhanced daidzin production from jasmonic and acetyl salicylic acid elicited hairy root cultures of Psoralea corylifolia L. (Fabaceae). Natural Product Research. 2016;30:1542-1547. DOI: 10.1080/14786419.2015.1054823
  38. 38. Dicosmo F, Misawa M. Plant cell and tissue culture: Alternatives for metabolite production. Biotechnology Advances. 1995;13:425-453. DOI: 10.1016/0734-9750(95)02005-N
  39. 39. Verpoorte R, Alfermann AW. Metabolic Engineering of the Plant Secondary Metabolism. New York: Springer; 2000. pp. 1-29
  40. 40. O’Connor SE. Engineering of secondary metabolism. Annual Reviews in Genetics. 2015;49:71-94. DOI: 10.1146/annurev-genet-120213-092053
  41. 41. Cusido RM, Onrubia M, Sabater-Jara AB, Moyano E, Bonfill M, Goossens A, Pedreño MA, Palazon J. A rational approach to improving the biotechnological production of taxanes in plant cell cultures of Taxus spp. Biotechnology Advances. 2014;32:1157-1167. DOI: 10.1016/j.biotechadv.2014.03.002
  42. 42. Lu X, Tang K, Li P. Plant metabolic engineering strategies for the production of pharmaceutical terpenoids. Frontiers in Plant Science. 2016;7:1647. DOI: 10.3389/fpls.2016.01647
  43. 43. Dudareva N, Andersson S, Orlova I, Gatto N, Reichelt M, Rhodes D, Boland W, Gershenzon J. The nonmevalonate pathway supports both monoterpene and sesquiterpene formation in snapdragon flowers. Proceedings of the National Academy of Sciences USA. 2005;102:933-938. DOI: 10.1073/pnas.0407360102
  44. 44. Nanda S, Mohanty JN, Mishra R, Joshi RK. Metabolic engineering of phenyl propanoids in plants. In: Jha S, editor. Transgenesis and Secondary Metabolism: Part of the Series Reference Series in Phytochemistry. New York: Springer; 2016. pp. 1-26
  45. 45. Verpoorte R, Van der Heijden R, Ten Hoopen HJG, Memelink J. Metabolic engineering of plant secondary metabolite pathways for production of fine chemicals. Biotechnology Letters. 1999;21:467-479. DOI: 10.1023/A:1005502632053
  46. 46. Oksman-Caldentey KM, Arroo R. Regulation of tropane alkaloid metabolism in plants and plant cell cultures. In: Verpoorte R, Alfermann AW, editors. Metabolic Engineering of Plant Secondary Metabolism. New York: Springer; 2000. pp. 253-281
  47. 47. 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
  48. 48. 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
  49. 49. Hibino K, Ushiyama K. Commercial production of ginseng by the plant tissue culture technology. In: Fu TJ, Singh G, Curtis WR, editors. Plant Cell & Tissue Culture for Production of Food Ingredients. New York: Springer; 1999. pp. 215-224
  50. 50. Wink M, Alfermann AW, Franke R, Wetterauer B, Distl M, Windhövel J, Krohn O, Fuss E, Garden H, Mohagheghzadeh A, Wildi E, Ripplinger P. Sustainable bioproduction of phytochemicals by plant in vitro cultures: Anticancer agents. Plant Genetic Resources: Characterisation and Utilisation. 2005;3:90-100
  51. 51. Yesil-Celiktas O, Gurel A, Vardar-Sukan F. Large Scale Cultivation of Plant Cell and Tissue Culture in Bioreactors. Trivandrum: Transworld Research Network; 2010. pp. 1-54
  52. 52. Ruffoni B, Pistelli L, Bertoli A, Pistelli L. Plant cell cultures: Bioreactors for industrial production. In: Giardi MT, Rea G, Berra B, editors. Bio-Farms for Nutraceuticals. New York: Springer; 2010. pp. 203-221
  53. 53. Steingroewer J, Bley T, Georgiev V, Ivanov I, Lenk F, Marchev A, Pavlov A. Bioprocessing of differentiated plant in vitro systems. Engineering in Life Sciences. 2013;13:26-38. DOI: 10.1002/elsc.201100226
  54. 54. Wang SJ, Zhong JJ. A novel centrifugal impeller bioreactor. I. Fluid circulation, mixing, and liquid velocity profiles. Biotechnology and Bioengineering. 1996;51:511-519. DOI: 10.1002/(SICI)1097-0290(19960905)51:5<511::AID-BIT2>3.0.CO;2-F
  55. 55. Cassady JM, Douros JD. Anticancer Agents Based on Natural Product Models. New York: Academic Press; 1980
  56. 56. Fridlender M, Kapulnik Y, Koltai H. Plant derived substances with anti-cancer activity: From folklore to practice. Frontiers in Plant Science. 2015:6-799. DOI: 10.3389/fpls.2015.00799
  57. 57. Lopez-Meyer M, Nessler CL, McKnight TD. Sites of accumulation of the antitumor alkaloid camptothecin in Camptotheca acuminata. Planta Medica. 1994;60:558-560. DOI: 10.1055/s-2006-959571
  58. 58. Saito K, Sudo H, Yamazaki M, Koseki-Nakamura M, Kitajima M, Takayama H, Aimi N. Feasible production of camptothecin by hairy root cultures of Ophiorrhiza pumila. Plant Cell Reports. 2001;20:267-271. DOI: 10.1007/s002990100320
  59. 59. Almagro L, Fernández-Pérez F, Pedreño MA. Indole alkaloids from Catharanthus roseus: Bioproduction and their effect on human health. Molecules. 2015;20:2973-3000. DOI: 10.3390/molecules20022973
  60. 60. Roja G. Micropropagation and production of Camptothecin from in vitro plants of Ophiorrhiza rugosa var. decumbens. Natual Product Research. 2008;22:1017-1023. DOI: 10.1080/14786410802006165
  61. 61. Dandin VS, Murthy HN. Enhanced in vitro multiplication of Nothapodytes nimmoniana Graham using semisolid and liquid cultures and estimation of camptothecin in the regenerated plants. Acta Physiologiae Plantarum. 2012;34:1381-1386. DOI: 10.1007/s11738-012-0934-x
  62. 62. Kim YS, Lim S, Choi YE, Anbazhagan VR. High frequency plant regeneration via somatic embryogenesis in Podophyllum peltatum L., an important source of anticancer drug. Current Science. 2017;92:662-666
  63. 63. Rajesh M, Sivanandhan G, Jeyaraj M, Chackravarthy R, Manickavasagam M, Selvaraj N, Ganapathi A. An efficient in vitro system for somatic embryogenesis and podophyllotoxin production in Podophyllum hexandrum Royle. Protoplasma. 2014;251:1231-1243. DOI: 10.1007/s00709-014-0632-1
  64. 64. Trivellini A, Lucchesini M, Maggini R, Mosadegh H, Villamarin TSS, Vernieri P, Mensuali-Sodi A, Pardossi A. Lamiaceae phenols as multifaceted compounds: Bioactivity, industrialprospects and role of “positive-stress”. Industrial Crops and Products. 2016;83:241-254. DOI: 10.1016/j.indcrop.2015.12.039
  65. 65. Ozkan M. Glandular and eglandular hairs of Salvia recognita Fisch. & Mey. (Lamiaceae) in Turkey. Bangladesh Journal of Botany. 2008;37:93-95. DOI: 10.3329/bjb.v37i1.1571
  66. 66. Gonçalves S, Romano A. In vitro culture of lavenders (Lavandula spp.) and the production of secondary metabolites. Biotechnology Advances. 2013;31:166-174. DOI: 10.1016/j.biotechadv.2012.09.006
  67. 67. Gonçalves S, Moreira E, Grosso C, Andrade PB, Valentão P, Romano A. Phenolic profile, antioxidant activity and enzyme inhibitory activities of extracts from aromatic plants used in Mediterranean diet. Journal of Food Science and Technology. 2017;54:219-227. DOI: 10.1007/s13197-016-2453-z
  68. 68. Zheng Q, Sun Z, Zhang X, Yuan J, Wu H, Yang J, Xu X. Clerodendranoic acid, a new phenolic acid from Clerodendranthus spicatus. Molecules. 2012;17:13656-13661. DOI: 10.3390/molecules171113656
  69. 69. Birtić S, Dussort P, Pierre FX, Bily AC, Roller M. Carnosic acid. Phytochemistry. 2015;115:9-19. DOI: 10.1016/j.phytochem.2014.12.026
  70. 70. Matkowski A. Plant in vitro culture for the production of antioxidants—A review. Biotechnology Advances. 2008;26:548-560. DOI: 10.1016/j.biotechadv.2008.07.001
  71. 71. Khojasteh A, Mirjalili MH, Hidalgo D, Corchete P, Palazon J. New trends in biotechnological production of rosmarinic acid. Biotechnology Letters. 2014;36:2393-2406. DOI: 10.1007/s10529-014-1640-0
  72. 72. Stehfest K, Boese M, Kerns G, Piry A, Wilhelm C. Fourier transform infrared spectroscopy as a new tool to determine rosmarinic acid in situ. Journal of Plant Physiology. 2004;161:151-156. DOI: 10.1078/0176-1617-01099
  73. 73. Li W, Koike K, Asada Y, Yoshikawa T, Nikaido T. Rosmarinic acid production by Coleus forskohlii hairy root cultures. Plant Cell Tissue and Organ Culture. 2005;80:151-155. DOI: 10.1007/s11240-004-9541-x
  74. 74. Luis JC, Martın Perez R, Valdes Gonzalez F. UV-B radiation effects on foliar concentrations of rosmarinic and carnosic acids in rosemary plants. Food Chemistry. 2007;101:1211-1215. DOI: 10.1016/j.foodchem.2006.03.023
  75. 75. Lorence A, Medina-Bolivar F, Nessler CL. Camptothecin and 10-hydroxycamptothecin from Camptotheca acuminata hairy roots. Plant Cell Reports. 2004;22:437-441. DOI: 10.1007/s00299-003-0708-4
  76. 76. Pi Y, Jiang K, Hou R, Gong Y, Lin J, Sun X, Tang K. Examination of camptothecin and 10-hydroxycamptothecin in Camptotheca acuminata plant and cell culture, and the affected yields under several cell culture treatments. Biocell. 2010;34:139-143
  77. 77. Fulzele DP, Satdive R, Kamble S, Singh S, Singh S. Improvement of anticancer drug camptothecin production by gamma irradiation on callus cultures of Nothapodytes foetida. International Journal of Pharmaceutical Research and Allied Sciences. 2015;4:19-27
  78. 78. Karwasara MVS, Dixit VK. Culture medium optimization for camptothecin production in cell suspension cultures of Nothapodytes nimmoniana (J. Grah.). Plant Biotechnology Reports. 2013;7:357-369
  79. 79. Ya-ut P, Chareonsap P, Sukrong S. Micropropagation and hairy root culture of Ophiorrhiza alata Craib for camptothecin production. Biotechnology Letters. 2011;33:2519-2526. DOI: 10.1007/s10529-011-0717-2
  80. 80. Deepthi S, Satheeshkumar K. Enhanced camptothecin production induced by elicitors in the cell suspension cultures of Ophiorrhiza mungos Linn. Plant Cell Tissue and Organ Culture. 2016;124:483-493. DOI: 10.1007/s11240-015-0908-y
  81. 81. Martin KP, Zhang C-L, Hembrom ME, Slater A, Madassery J. Adventitious root induction in Ophiorrhiza prostrata: A tool for the production of camptothecin (an anticancer drug) and rapid propagation. Plant Biotechnology Reports. 2008;2:163-169. DOI: 10.1007/s11816-008-0057-4
  82. 82. Bahabadi SE, Sharifi M, Chashmi NA, Murata J, Satake H. Significant enhancement of lignan accumulation in hairy root cultures of Linum album using biotic elicitors. Acta Physiolgiae Plantarum. 2014;36:3325-3331. DOI: 10.1007/s11738-014-1700-z
  83. 83. Cong LH, Dauwe R, Lequart M, Vinchon S, Renouard S, Fliniaux O, Colas C, Corbin C, Doussot J, Hano C, Lamblin F, Molinié R, Pilard S, Jullian N, Boitel M, Gontier E, Mesnard F, Laberche J-C. Kinetics of glucosylated and non-glucosylated aryltetralin lignans in Linum hairy root cultures. Phytochemistry. 2015;115:70-78. DOI: 10.1016/j.phytochem.2015.01.001
  84. 84. Yousefzadi M, Sharifi M, Behmanesh M, Ghasempour A, Moyano E, Palazon J. Salicylic acid improves podophyllotoxin production in cell cultures of Linum album by increasing the expression of genes related with its biosynthesis. Biotechnology Letters. 2010;32:1739-1743. DOI: 10.1007/s10529-010-0343-4
  85. 85. Jamshidi M, Ghanati F. Taxanes content and cytotoxicity of hazel cells extract after elicitation with silver nanoparticles. Plant Physiology and Biochemistry. 2017;110:178-184. DOI: 10.1016/j.plaphy.2016.04.026
  86. 86. Li M, Peebles CAM, Shanks JV, San K-Y. Effect of sodium nitroprusside on growth and terpenoid indole alkaloid production in Catharanthus roseus hairy root cultures. Biotechnology Progress. 2011;27:625-630. DOI: 10.1002/btpr.605
  87. 87. Rizvi NF, Cornejo M, Stein K, Weaver J, Cram EJ, Lee-Parsons CWT. An efficient transformation method for estrogen-inducible transgene expression in Catharanthus roseus hairy roots. Plant Cell Tissue and Organ Culture. 2015;120:475-487. DOI: 10.1007/s11240-014-0614-1
  88. 88. Bauer N, Kiseljak D, Jelaska S. The effect of yeast extract and methyljasmonate on rosmarinic acid accumulation in Coleus blumei hairy roots. Biologia Plantarum. 2009;53:650-656. DOI: 10.1007/s10535-009-0118-8
  89. 89. Georgiev M, Kuzeva S, Pavlov A, Kovacheva E, Ilieva M. Enhanced rosmarinic acid production by Lavandula vera MM cell suspension culture through elicitation with vanadyl sulfate. Zeitschrift für Naturforschung C. 2006;61:241-244. DOI: 10.1515/znc-2006-3-414
  90. 90. Georgiev MI, Kuzeva SL, Pavlov AI, Kovacheva EG, Ilieva MP. Elicitation of rosmarinic acid by Lavandula vera MM cell suspension culture with abiotic elicitors. World Journal of Microbiology and Biotechnology. 2007;23:301-304. DOI: 10.1007/s11274-006-9214-5
  91. 91. Krzyzanowska J, Czubacka A, Pecio L, Przybys M, Doroszewska T, Stochmal A, Oleszek W. The effects of jasmonic acid and methyl jasmonate on rosmarinic acid production in Mentha × piperita cell suspension cultures. Plant Cell Tissue and Organ Culture. 2012;108:73-81. DOI: 10.1007/s11240-011-0014-8

Written By

Sandra Gonçalves and Anabela Romano

Submitted: 08 November 2017 Reviewed: 12 March 2018 Published: 05 September 2018