Next Article in Journal
Cholinergic Antagonists and Behavioral Disturbances in Neurodegenerative Diseases
Next Article in Special Issue
From Transgenesis to Genome Editing in Crop Improvement: Applications, Marketing, and Legal Issues
Previous Article in Journal
Structure-Based Design and Pharmacophore-Based Virtual Screening of Combinatorial Library of Triclosan Analogues Active against Enoyl-Acyl Carrier Protein Reductase of Plasmodium falciparum with Favourable ADME Profiles
Previous Article in Special Issue
Metabolic Composition and Quality Traits of Polygonatum cyrtonema Hua from Different Germplasms and Age Sections Based on Widely Targeted Metabolomics Analysis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Hairy Root Cultures as a Source of Phenolic Antioxidants: Simple Phenolics, Phenolic Acids, Phenylethanoids, and Hydroxycinnamates

1
Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna Street 12, 31-343 Kraków, Poland
2
Educational and Scientific Medical Institute, National Technical University “Kharkiv Polytechnic Institute”, Kyrpychova Street 2, 61002 Kharkiv, Ukraine
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(8), 6920; https://doi.org/10.3390/ijms24086920
Submission received: 1 March 2023 / Revised: 31 March 2023 / Accepted: 5 April 2023 / Published: 7 April 2023
(This article belongs to the Collection Feature Papers in Molecular Plant Sciences)

Abstract

:
Plant-derived antioxidants are intrinsic components of human diet and factors implicated in tolerance mechanisms against environmental stresses in both plants and humans. They are being used as food preservatives and additives or ingredients of cosmetics. For nearly forty years, Rhizobium rhizogenes-transformed roots (hairy roots) have been studied in respect to their usability as producers of plant specialized metabolites of different, primarily medical applications. Moreover, the hairy root cultures have proven their value as a tool in crop plant improvement and in plant secondary metabolism investigations. Though cultivated plants remain a major source of plant polyphenolics of economic importance, the decline in biodiversity caused by climate changes and overexploitation of natural resources may increase the interest in hairy roots as a productive and renewable source of biologically active compounds. The present review examines hairy roots as efficient producers of simple phenolics, phenylethanoids, and hydroxycinnamates of plant origin and summarizes efforts to maximize the product yield. Attempts to use Rhizobium rhizogenes-mediated genetic transformation for inducing enhanced production of the plant phenolics/polyphenolics in crop plants are also mentioned.

Graphical Abstract

1. Introduction

Hairy roots (HRs) emerge as a result of transfer and permanent incorporation of a foreign genetic material, delivered by the fragment (T-DNA) of the bacterial Ri plasmid into the plant genomic DNA. Rhizobacterium rhizogenes (formerly Agrobacterium rhizogenes), a plant growth-promoting Gram-negative bacterium that is a carrier of the Ri plasmid, has been known since the 1930s of the twentieth century as the etiological agent of the “hairy root disease”. Attempts to apply the HRs for the exploration of plant secondary metabolite biosynthesis started in the middle of 1980s [1,2]. Since then, HRs of numerous plant species have been described, and their biosynthetic potential has been thoroughly studied [3,4,5]. The advantages of the HR cultures are as follows: the rapid growth in nutrient media with reduced concentration of macronutrients and without plant growth regulators, the biosynthetic potential similar to that of the parent plant, and the ability to grow and synthesize natural products in the dark. The major drawbacks are unsatisfactory upscaling results (the need for large-volume bioreactors) and unsatisfactory costs/profit ratio. Along with the studies of plant metabolism, HRs have been found to have use in plant breeding, genetic engineering and research on gene functions, phytoremediation, and biotransformation [6]. The most promising area of their industrial application seems to be the production of recombinant proteins and production of specialized metabolites [3]. However, the process should be technically and economically feasible. Plant secondary metabolites of medical applications have dominated the research on natural products biosynthesized with HRs for decades. Recently, production of food ingredients and food by plant cell and tissue cultures has gained growing interest. The current regulations, however, do not allow the use of genetically transformed cultures for the production of commercial food products and ingredients [7].
Mechanism of T-DNA transfer from R. rhizogenes into the nucleus of a plant cell and subsequent HR formation, the bacterial rol and aux genes mechanisms of action, the HRs’ cultivation methods including their cultivation in bioreactors of different scale and design, methods applied to induce HRs’ growth and to enhance biosynthesis of secondary metabolites have been the subject of numerous review papers. Majority of the reviews presented lists of plant species transformed with R. rhizogenes together with the principal secondary metabolites synthesized by the obtained HRs. However, quantitative data that are of key importance for the assessment of HR prospects for application are usually missing.
Phenolics/polyphenolics and some terpenoids (e.g., carotenoids) are well-known plant secondary (specialized) metabolites of antioxidative and chemopreventive activity [8,9,10,11] that help combat lifestyle diseases [12,13,14,15]. Although the compounds are constituents of fruits and vegetables that are recommended components of human diet, their bioavailability often remains low and is strongly affected by food processing, food matrix, and gut microbiota [16,17,18]. Therefore, isolated plant polyphenols may be used for food supplementation and enrichment. Due to their antioxidative and antimicrobial properties, phenolics of plant origin may find application as food preservatives [19,20,21]. Antioxidative, antimelanogenic, and UV-B protecting activities of plant phenolic compounds also make them good candidates for antiaging skin care ingredients [22,23,24]. Apart from the application in the food and cosmetic industry, the compounds have garnered interest as potential natural herbicides and fungicides [25].
Although technological and economical limitations still hamper industrial use of HRs as a source of phytochemicals, a growing demand for high added-value molecules from plants and requirements for biodiversity protection may speed up the implementation of processes based on large-scale HR cultures. The objective of this review is to summarize the achievements in phenolic/polyphenolic production by HRs in order to show the current possibilities and limitations of the technique.
In our previous paper [26], we summarized the attempts to produce flavonoids, stilbenoids, and hydrolysable tannins in HRs. However, plant polyphenolics are a much larger group of natural products and encompass further subclasses of plant metabolites. The present review is based on the experimental results concerning the production of simple phenolics, hydroxycinnamates, and phenylethanoids in HRs that were published before February 2023 in journals covered by two databases: Web of Science and Scopus. The search terms used were the following: “hairy roots”, “Agrobacterium rhizogenes”, or “Rhizobium rhizogenes” in conjunction with “phenolics”, “phenolic acids”, “phenols”, “polyphenols”, “hydroxybenzoic”, “hydroxycinnamic”, “chlorogenic”, “caffeic”, “ferulic”, “coumaric”, “phenylethanoid”, “phenylpropanoid” “tyrosol”, “salidroside”, “acteoside”, “verbascoside”, “echinacoside”, “martynoside”, “chicoric acid”, “cichoric acid”, “caftaric acid”, “caffeoylquinic”, “rosmarinic acid”, “lithospermic acid”, and “salvianolic acid”. No date range was set in order to include more data. As a result, over two hundred research papers that contained quantitative data were analyzed in detail.

2. Phenolic and Polyphenolic Antioxidants in HRs

Phenolic compounds are present in all vascular plants. They occur in plant cells either in soluble form or bounded to cell walls. The natural products originate from the shikimate pathway that, via chorismate and prephenate, leads to aromatic amino acids tyrosine and phenylalanine. The latter compound is deaminated by phenylalanine ammonia lyase (PAL) to trans-cinnamic acid, and the reaction is the first committed step in the phenylpropanoid pathway that is involved in the biosynthesis of polyphenols [27,28].

2.1. Simple Phenolics and Phenolic Acids

The most common types of the simple phenolics occurring in plants (for structures see Figure 1) are the hydroxybenzoic (C6-C1), phenylacetic (C6-C2), and hydroxycinnamic (C6-C3) acid derivatives as well as the phenolic alcohols (C6, C6-C1, C6-C2, and C6-C3). Some of them are the building blocks for the more complex molecules (polyphenolics), such as coumarins, lignans, depsides, and conjugates of phenols with sugars and organic acids. Hydroxybenzoic and hydroxycinnamic acid derivatives are common constituents of foods and beverages that, in some instances, are easily available from postproduction waste or side products in the food industry [29,30]. Their presence in the HRs is usually recorded together with polyphenolics of more complex structure.
Maximum contents of soluble phenolics in the HRs were shown to not usually exceed 0.1% calculated on a dry weight (DW) basis. Cell wall-bound phenolics were present in greater amounts. For example, the cell wall-bounded and soluble form of p-hydroxybenzoic acid (p-HBA, 4-HBA) constituted up to 0.44% and up to 0.04% DW, respectively, in HRs of Daucus carota L. (Apiaceae) elicited with chitosan [31]. The highest titers of p-HBA derivatives in the soluble phenolic fraction have been found in the Beta vulgaris L. (Amaranthaceae) and the Datura stramonium L. (Solanaceae) HRs expressing 4-hydroxycinnamoyl-CoA hydratase/lyase (HCHL) of bacterial origin [32,33]. The enzyme hydrates 4-hydroxycinnamoyl-CoA thioesters and then catalyzes a retro-aldol cleavage to produce the corresponding 4-hydroxybenzaldehydes and acetyl-CoA. The HRs with heterologous expression of HLCL demonstrate a diminished availability of feruloyl-CoA, an increased bounding of p-HBA to the cell wall, and a high content of the p-HBA derivatives (glucoside and glucose ester of p-HBA, p-hydroxybenzyl alcohol glucoside) in the soluble phenolic fraction. The most productive line of the HLCL-transgenic D. stramonium HRs accumulates up to 0.81% of the p-HBA-related compounds, calculated on the fresh weight (FW) basis [32]. One of the HLCL-transgenic B. vulgaris root clones was shown to accumulate nearly 14% DW of p-HBA glucose ester [32] while 4-Hydroxybenzoic acid-β-D-glucoside has been found to be a major compound in the HLCL-transgenic D. stramonium HRs.
The HRs demonstrate the potential to convert exogenously applied phenolic compounds into their glucosides. In one study, R. rhizogenes transformed roots of Lobelia sessilifolia Lamb. (Campanulaceae) glucosylated protocatechuic acid to its 3-O-β-D-glucopyranoside [34]. The HRs of Polygonum multiflorum Thunb. (Polygonaceae) glucosylate 4-hydroxybenzene derivatives to the corresponding 4-β-D-glucopyranosides [35]. The HR culture of Panax ginseng C.A. Mey. (Araliaceae) performed a biotransformation of two HBA isomers (m-HBA and p-HBA) into their corresponding β-D-glucopyranosides and β-D-glucopyranosyl esters whereas the products of o-HBA biotransformation were not detected [36].
Sircar et al. [37] found that the HRs of carrot (D. carota) that accumulated p-HBA may be a good experimental system to study the biosynthesis of this compound. The carrot HRs elicited with methyl jasmonate (MeJa, 100 μM) demonstrated higher content of cell wall-bounded p-HBA (0.42% DW) than did the control roots (0.18% DW). A detailed analysis of the enzymatic activities in cell-free extracts from the HRs elicited with MeJa revealed that the elevated activities of PAL and hydroxybenzoate dehydrogenase (HBD), that converts p-hydroxybenzaldehyde to p-HBA, preceded enhanced accumulation of p-HBA in the elicited roots. Moreover, the enhanced depletion of p-coumaric acid with subsequent formation of p-hydroxybenzaldehyde, catalyzed by the cell-free extract from the elicited roots, indicated the activity of p-hydroxybenzaldehyde synthase (HBS), an enzyme catalyzing phenylpropanoid side-chain cleavage of p-coumaric acid [38]. The inhibition of PAL and cinnamate-4-hydroxylase (C4H) in the D. carota HRs elicited with chitosan (200 mg/L) abolished the enhanced accumulation of p-HBA and p-coumaric acid by the elicited roots. The inhibition of 4-coumarate:CoA ligase (4CL) did not affect p-HBA or p-coumarate biosynthesis. The maximum content of the soluble (0.05% DW) and cell wall-bound (0.45% DW) p-HBA in the roots was achieved upon simultaneous elicitation with chitosan and precursor feeding at day 15 of the culture cycle [31]. The MeJa-elicited HRs of carrot, treated with inhibitors of alternative oxidase (AOX), i.e., salicylhydroxamic acid (SHAM) and propyl gallate (PG), demonstrated a reduced accumulation of phenolic acids, flavonoids and lignin, although transcript levels of PAL, DcAOX2a, and DcAOX1a significantly increased after the MeJa treatment. Thus, biosynthesis of phenylpropanoid derivatives in the MeJa-treated HRs was found to be positively associated with AOX activity [39]. The exposure of the D. carota HRs to continuous light (250 μmol m−2 s−1) induced a photooxidative stress response in the root tissue. The roots turned green and exhibited an enhanced superoxide dismutase (SOD) activity. The transfer of the carrot HRs from dark to light conditions changed the metabolism of the roots. HBD, shikimate dehydrogenase (SKDH), and PAL activities were suppressed, whereas activities of the 1-deoxy-D-xylulose 5-phosphate reductoisomerase (DXR) and pyruvate kinase (PK) were enhanced in the green HRs as compared to the normal ones. Gene expression analysis for DXR and PAL showed trends similar to those for the respective enzyme activities. The green roots accumulated 50% less of the cell wall-bound p-HBA than did the normal HRs cultured in dark but the production of volatile mono- and sesquiterpenoids was enhanced in the light-exposed roots [40,41].
Gentisic acid (0.17 mg/g DW) and ferulic acid (0.33 mg/g DW) were found to be the most abundant phenolic acids in the HRs of Brassica rapa L. ssp. rapa (Brassicaceae). The roots of in vitro grown B. rapa plantlets contained 0.18 mg/g DW and 0.27 mg/g DW of the acids, respectively [42]. Elicitation of the HRs with silver nanoparticles (100 mg/L; 48 h) caused an increase in the phenolic acid contents up to 0.20 mg/g DW (gentisic acid) and 0.39 mg/g DW (ferulic acid). The elicited HRs demonstrated an enhanced antimicrobial and radical scavenging activity and an enhanced expression of PAL [43]. Gentisic and ferulic acid were also major phenolic acids present in the HRs of B. rapa ssp. pekinensis. Their contents reached 0.68 mg/g DW and 0.56 mg/g DW, respectively, upon elicitation with copper oxide nanoparticles (100 mg/L) [44].
The HRs obtained by the inoculation of Momordica charantia L. (bitter melon, Cucurbitaceae) explants with R. rhizogenes KCTC 2703-synthesized gentisic (5.53 mg/g DW) and salicylic (SA; 2.52 mg/g DW) acid as major components of phenolic acid fraction. The spine gourd (Momordica dioica Roxb. ex Willd.) HRs produced much lower amounts of the phenolics (0.54 and 0.49 mg/g DW, respectively) [45,46]. Gentisic acid and SA were also the predominant hydroxybenzoic acids in the gherkin (Cucumis anguria L.) HRs (0.76 mg/g DW and 1.00 mg/g DW, respectively) and nontransformed roots of the plant (0.55 and 0.74 mg/g DW) [47].
The P. multiflorum HRs developed from leaf explants infected with R. rhizogenes KCTC 2703, except for hydroxycinnamic and hydroxybenzoic acids (gallic acid: 0.56 mg/g DW), were shown to accumulate pyrogallol (1.37 mg/g DW). The culture produced up to 10.95 g of dry biomass per 1 L of a nutrient medium in 20 days [48]. In the HRs of P. multiflorum obtained after the inoculation with R. rhizogenes KCCM 11879, flavonoids dominated the phenolic compound fraction [49]. The roots contained p-HBA and ferulic acid (3.46 mg/g and 2.20 mg/g DW, respectively) as well as several other phenolic acids in minor quantities. Five days after the addition of MeJa (50 μM) to the culture medium, a 5.48-fold increase in the p-HBA content and a 4.3-fold increase in the SA content was observed. Pyrogallol has not been mentioned as a metabolite of the culture.
The current status of research on the buckwheat (Fagopyrum esculentum Moench and Fagopyrum tataricum (L.) Gaertn.; Polygonaceae) HRs has been recently summarized by Tomasiak et al. [50]. The transformed buckwheat root cultures accumulate a number of phenolic acids, of which caffeic and ferulic acids were reported to be the most abundant ones (up to 1.0 mg/g DW).
The HRs of two Ficus carica L. (Moraceae) cultivars, cv. Sabz and cv. Siah, were obtained by the inoculation of leaf explants with R. rhizogenes line ATCC 15834 and A7, respectively. Upon the elicitation with MeJa (100 μM), the HRs of cv. Siah produced up to 0.71 mg/g DW of gallic acid and up to 6.5 mg/g of chlorogenic acid (5-CQA; IUPAC numbering, see Section 2.3.2). The HRs of cv. Sabz, after the elicitation with MeJa (200 μM), accumulated gallic (0.19 mg/g DW), caffeic (0.28 mg/g DW), chlorogenic (1.63 mg/g DW), and coumaric (0.23 mg/g DW) acids [51]. The same root culture elicited with the Piriformospora indica fungal cell extract or culture filtrate, at a concentration range of 2–6% (v/v), demonstrated an enhanced production of phenolic acids (up to 7.49 mg/g of gallic acid, up to 0.83 mg/g of caffeic acid, up to 1.58 mg/g of 5-CQA, and up to 0.94 mg/g of coumaric acid, as calculated on a DW basis). The most effective phenolic biosynthesis stimulant was the P. indica culture filtrate, at 2% (v/v) concentration, that was a potent inducer of PAL expression. The lowest concentration of both the fungal cell extract and the fungal culture filtrate strongly promoted the growth of the HRs [52].
Aster scaber Thunb. (Asteraceae) explants infected with R. rhizogenes KCTC 2703 were found to produce HRs that accumulated simple phenolics with SA (0.32 mg/g DW) as the major compound. The elicitation with MeJa (100 μM, 96 h treatment) enhanced the accumulation of SA up to 0.44 mg/g DW [53]. In the HRs of Ligularia fischeri Turcz. f. spiciformis (Nakai) (Asteraceae), SA (0.30 mg/g) and caffeic acid (0.18 mg/g DW) dominated the hydroxybenzoic and hydroxycinnamic acids fractions, respectively [54]. The main phenolic acid found in the HRs of Sphagneticola calendulacea (L.) Pruski (Asteraceae) was ferulic acid (0.21 mg/g DW). Feeding the roots with phenylalanine (0.5 mM added on day 10 of the culture), increased the ferulic acid content up to 1.19 mg/g DW [55].
Numerous wild-type and genetically modified HR cultures of Lamiaceae plants have been established for the studies on secondary metabolism. Expression of Arabidopsis PAP1 (Production of Anthocyanin Pigment 1 transcription factor from the MYB family) in the HRs of Leonurus sibiricus L. led to the enhanced production of hydroxycinnamic acids. Contents of the analyzed compounds were correlated with the relative expression levels of AtPAP1. The most productive of the AtPAP1-carrying HR clones accumulated up to 19.39 mg/g DW of chlorogenic acid, 11.38 mg/g DW of caffeic acid, and 1.17 mg/g DW of ferulic acid [56]. Mentha spicata L. roos transformed with R. rhizogenes A13, upon the elicitation with MeJa (100 μM), demonstrated an enhanced expression of PAL, 4CL, and C4H with a subsequent increase in cinnamic acid content. The contents of caffeic and chlorogenic acids were significantly higher than those in the control roots 3–6 h and 72–96 h after the addition of MeJa, respectively [57]. The HRs of Leonotis nepetifolia (L.) R. Br., obtained by infecting of the aseptically grown seedlings with R. rhizogenes strain A4, contained substantial amounts of p-coumaric acid (4.91 mg/g DW) and m-coumaric acid (2.05 mg/g DW) [58].
Kochan et al. [59] compared quantities of phenolic acids accumulated by the roots of three-year-old field-grown Panax quinquefolius L. (Araliaceae) plants with those in the wild-type HRs of the plant cultivated in shake flasks and a nutrient sprinkle bioreactor. In the bioreactor-cultivated HRs, a significant drop in p-HBA, syringic, and gentisic acid contents was observed. The HRs tended to accumulate higher contents of 5-CQA than did the roots of field-grown plants.
Ferulic acid was found to dominate a cell wall-bound fraction of phenolics in the HRs of tomato (Lycopersocum esculentum Mill. cv. Arka Suarabh; Solanaceae). Upon the elicitation with chitosan (1 mL of 1% solution in 60 mL of a nutrient medium) or fungal preparations from Fusarium oxysporum f. sp. lycopersici and Trichoderma viride, the content of ferulic acid in HRs rose from 0.3 (control HRs) to 1.0 mg/g DW (elicited HRs, 24 h after the induction) [60].
β-Cryptogein is a basic polypeptide of elicitin family, produced by the plant pathogenic fungus Phytophthora cryptogea. The polypeptide triggers a defense response in plants. In one study, leaves from axenic Nicotiana tabacum L. (Solanaceae) plants were infected with R. rhizogenes strain LBA 9402 carrying either the empty vector pBIN19 or pBIN19 harboring the synthetic β-cryptogein gene under the control of a CaMV promoter. The Ri-cryptogein-transformed plants were characterized by a diminished production of H2O2 and O2- radicals, diminished lipid peroxidation, and suppressed levels of transcripts for the isoprenoid biosynthetic pathway enzymes. Simultaneously, the plants exhibited enhanced activities of antioxidative enzymes (peroxidase, ascorbate peroxidase, catalase, SOD), higher lignin content, and higher cell wall-bound phenolics content [61].
To obtain the HRs of Pelargonium sidoides DC. (Geraniaceae), a medicinal plant that produces coumarins of unique structure, R. tumefaciens C58C1 carrying the pRiA4 plasmide was applied. The developed HRs were cultivated in the liquid media containing three different concentrations of MeJa (25, 50, and 100 μM). The elicitor, at the applied concentration range, enhanced the accumulation of hydroxybenzoic acids: gallic acid (maximum content: 2.43 mg/g DW; 50 μM MeJa) and vanillic acid (maximum content: 1.66 mg/g DW; 25 μM MeJa) [62].
The HR cultures that accumulated high contents of simple phenolics and phenolic acids are shown in Table 1.

2.2. Phenylethanoids

Studies on the phenylethanoid production in the HRs mostly discuss verbascoside biosynthesis in the roots of plant species included into the Lamiales order or salidroside biosynthesis in Rhodiola spp. (Crassulaceae). Both compounds (for structures see Figure 2) demonstrate numerous activities of potential application in medicine. Verbascoside (synonyms: acteoside, kusaginin, orobanchin) protects human keratinocytes against the UV radiation, scavenges free radicals, and acts as an antioxidant and chemopreventive agent. The phenylethanoid glycoside, due to its interaction with estrogenic receptors, may exert anticancer and antimetastatic effects. Moreover, it has demonstrated antiproliferative activity against some cancer cell lines in vitro and may induce differentiation or apoptosis in some types of cancer cells. Anti-inflammatory and antimicrobial activities of the compound are also worth mentioning [63,64]. Salidroside, a major constituent of rosavin complex present in Rhodiola rosea L. roots, exerts antioxidant, cytoprotective, and antitumor activity [64,65]. A review on the pharmacological activity of salidroside with the focus on CNS functions has been published recently by Jin et al. [66]. Another phenylethanoid glycoside, echinacoside, shares similar pharmacological activity (neuro- and hepatoprotective, anticancer, anti-inflammatory) [64]. Its biosynthesis in HRs of Echinacea spp. (Asteraceae) is discussed in the Section 2.3.1.
In one study, the HR culture of Scutellaria baicalensis Georgi (Lamiaceae) was used to synthesize five known phenylethanoid glycosides: martynoside, leucosceptoside A, verbascoside, 2-(3-hydroxy-4-methoxyphenethyl) 1-O-α-L-rhamnopyranosyl-(1 -> 3)-β-D-(4-O-feruloyl) -glucopyranoside, and 4-hydroxy-β-phenethyl-β-D-glucopyranoside [67]. Acteoside (verbascoside) was the most abundant phenolic compound found in two clones of S. baicalensis HRs cultivated in the dark in a modified MS (Murashige and Skoog) [68] medium with reduced (50%) concentration of KNO3, NH4NO3, and CaCl2 and doubled concentrations of KH2PO4 and MgSO4. The phenylethanoid glycoside content in the HRs reached 3% DW. The compound was absent from roots of field-grown plants but was detected in leaves and roots of in-vitro-grown plantlets [69]. The HRs of S. lateriflora L., established by inoculation of leaf explants with R. rhizogenes strain A4, accumulated 18.5 mg/g DW of acteoside. The elicitation of the culture with yeast extract (YE; 50 and 200 μg/mL), significantly increased acteoside content after 7 and 14 days. An addition of the bacterial lysates (R. rhizogenes, Pseudomonas syringae, Pseudomonas carotovorum, Enterobacter sakazakii, Klebsiella pneumoniae) at two doses (7.5 and 15 mL/L) did not stimulate the biosynthesis of acteoside. On the contrary, in the HRs treated with Pseudomonas carotovorum lysate on the 26th day of culture, after the following 14 days, acteoside was not detected [70]. Marsh et al. [71] examined the effects of light, elicitation with MeJa, and the addition of β-cyclodextrin on the growth and production of phenolic compounds in the HRs of S. lateriflora obtained from the stem explants infected with R. rhizogenes ATCC 15834. The maximum content of verbascoside (c. 17 mg/g DW) was detected in the roots grown in the dark and treated (for 24 h) either with 15 mM of β-cyclodextrin or with 100 μM of MeJa.
In one study, the HRs of Paulownia tomentosa Steud. (Bignoniaceae) were obtained from the axenic stem explants infected with R. rhizogenes LBA 9402. The most productive root clone, cultivated in ½ B5 medium [72], accumulated up to 94.9 mg/g DW (1.21 g/L) of verbascoside [73]. The Catalpa ovata G. Don. (Bignoniaceae) HRs cultivated in similar conditions produced up to 41.2 mg/g DW of verbascoside and up to 5.8 mg/g DW of martynoside [74].
In another study, two types of root culture were initiated from axenic seedlings of Plantago lanceolata L. (Plantaginaceae): the HRs transformed with R. rhizogenes LBA 9402 and the untransformed roots. Both cultures produced similar yields of verbascoside (6–12 mg/g DW) and plantamoside (plantamajoside; 30–80 mg/g DW). Plantamoside content in the HRs matched that found in the roots of the whole plant. Cinnamic acid feeding had no effect on the phenylethanoid glycoside content. Instead, cinnamic acid glucose ester accumulated in the roots [75].
In other research, seedlings of Gmelina arborea Roxb. (Verbenaceae) were infected with a wild-type R. rhizogenes strain ATCC 15834 to initiate the HR culture. Verbascoside content in the developed HRs reached 0.91 mg/g DW versus 8.4 mg/g DW in the root of the mature tree [76].
Harpagophytum procumbens DC. (devil’s claw; Pedaliaceae), an African plant traditionally used to treat a variety of pain and inflammatory conditions, is known to produce iridoids and phenylethanoids. In one study, the HRs of the plant were cultivated either in flasks with shaking or in a bubble column bioreactor. The roots from the bioreactor contained more iridoids than did the roots cultivated in flasks. The phenylethanoid content was not analyzed [77]. The contents of four phenyletanoid glycosides (verbascoside, martynoside, β-OH-verbascoside, and leucosceptoside A) were compared for the roots grown in flasks and in a stirred tank bioreactor. Both cultures accumulated verbascoside (c. 1.1 mg/g DW) as a major phenyloethanoid. Roots grown in the bioreactor contained more leucosceptoside A but less β-OH-verbascoside and martynoside than did the roots from flasks [78]. Two wild-type R. rhizogenes strains, A4 and ATCC 15834, were used to obtain the H. procumbens HRs by Grąbkowska et al. [79]. Verbascoside content differed among the studied HR clones (5.34–8.12 mg/g DW) and was lower than that in the roots of the intact plant (c. 10 mg/g DW). In contrast, some of the HR clones accumulated more isoverbascoside (up to 16 mg/g DW) than did the plant roots. According to Gyurkovska et al. [80], cell suspension cultures of H. procumbens were a better source of phenylethanoids and demonstrated better anti-inflammatory activity than did the HRs of the plant. The maximum content of verbascoside in the examined HRs reached 0.94 mg/g DW. A phenylethanoid-containing fraction from the HRs inhibited both acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) at a concentration range 100–200 μg/mL, while a similar fraction from the cell suspension acted selectively against BChE. It is worth noting that the isolated phenyletanoids exerted a moderate anti-inflammatory effect [80,81].
Piątczak et al. [82] investigated the effects of the elicitation with MeJa and SA on phenyletanoide content in the HRs of Rehmannia glutinosa Libosch. (Orobanchaceae) transformed with R. rhizogenes strain A4. The 23-day-old HR cultures were treated either with MeJa or with SA (50–200 μM). A combination of the two elicitors was also tested. The highest verbascoside content (60.07 mg/g DW) was achieved with MeJa treatment (200 μM, 72 h). The elicitation with SA was used as a tool to detect genes and enzymes engaged in verbascoside biosynthesis in the HRs of R. glutinosa transformed with R. rhizogenes strain ACCC10060 [83]. The roots were treated with four elicitors (SA, MeJa, AgNO3, and putrescine), and SA was ultimately found to be the most effective in inducing verbascoside biosynthesis. The maximum yield of verbascoside (11.66 mg/g DW or 53.87 mg/L) was achieved by 12–24 h treatment with 25 μM/L of SA. The functional analysis of differentially expressed genes and the analysis of expression levels of the genes putatively involved in verbascoside biosynthesis (PAL; C4H; coumaroylquinate (coumaroylshikimate) 3′-monooxygenase, C3H; 4CL; tyrosine/DOPA decarboxylase, TyDC; polyphenol oxidase, PPO; copper-containing amine oxidase, CuAO; alcohol dehydrogenase, ALDH; UDP-glucose glucosyltransferase, UGT; shikimate O-hydroxycinnamoyltransferase, HCT) were performed to elucidate the biosynthetic pathway. The HR culture of R. elata N.E. Brown ex Prain synthesized iridoid glycosides and phenylethanoids. The most productive of the analyzed root clones accumulated 17.35 mg/g DW of verbascoside and c. 2 mg/g DW of isoverbascoside [84].
Plants from the genus Verbascum (Scrophulariaceae) have long been used in traditional medicine as remedies for respiratory tract infections and disorders. Iridoid glycosides, saponins, flavonoids, and phenylethanoids are regarded as the active constituents of the plants [85]. The HRs of V. xanthophoeniceum Griseb. were obtained to study their capability to produce active compounds. Attempts to achieve root proliferation with three different R. rhizogenes strains (TR 105, ATCC 15834 and LBA 9402) by direct infection and cocultivation method failed. Finally, the HRs were obtained with the ATCC 15834 strain with sonication-assisted transformation. In contrast to the leaves of the intact plant, verbascoside, not forsythoside B, was the major phenyloethanoid glycoside found in the HRs. Its content in one of the cultivated clones reached 23.31 mg/g DW. Leucosceptoside B and martynoside were present in smaller amounts [86]. V. nigrum L. was transformed with the R. rhizogenes strain ATCC 15834 using the sonication-assisted method. By means of NMR spectroscopy, metabolites from the leaves of parent plant and metabolites synthesized by the obtained HRs were compared, revealing that 5-CQA, harpagide, and harpagoside present in the leaves are absent from the HRs [87]. Similar analysis was performed for the HRs of V. eriophorum Godr. and leaves of the parent plant. The iridoids harpagide, aucubin, and aucubin glycosides were found only in the leaves, whereas the HRs were characterized by much higher contents of verbascoside and martynoside [88].
The current knowledge on the biosynthesis of salidroside and attempts to achieve biotechnological production of the compound from plant tissue cultures and genetically engineered microorganisms have been recently summarized by Grech-Baran et al. [89] and Liu et al. [90]. Cell suspension culture of Rhodiola crenulata (Hook.f. & Thomson) H.Ohba (Crassulaceae) that accumulates 26.48 mg/g DW of salidroside and compact callus of R. sachalinensis Boriss. containing 41.94 mg/g DW of the compound are among the best salidroside producers [89,90]. According to the WFO (The World Flora Online), R. sachalinensis Boriss. should be treated as a synonym of R. rosea L., the most reputed species of the Rhodiola genus. Botanical description of the plant as well as information on its distribution, ethnobotany, and medicinal properties is easy to find in the literature [65,91,92]. In one study, the HRs of R. sachalinensis were obtained via the infection of leaf explants with R. rhizogenes strain A4. The transformed roots cultivated in a liquid ½ MS medium, accumulated 4.2 mg/g FW of salidroside. Upon the elicitation with Aspergillus niger, Coriolus versicolor, or Ganoderma lucidum (0.05 mg/L), the content of salidroside increased to 7.1 mg/g FW, and a better growth of the roots was observed. A similar effect (6.6 mg/g DW salidroside and better growth) was achieved with the precursor feeding (tyrosol, tyrosine, phenylalanine; 1 mM/L) [93].
From roots and in-vitro-cultured MeJa-treated cells of R. sachalinensis, two putative UDP-glucosyltransferase (UGT) cDNAs were isolated: UGT72B14 and UGT74R1. Overexpression of the genes in the HRs of R. sachalinensis led to the enhanced production of salidroside. The roots overexpressing UGT72B14 contained 19.81 mg/g DW of the phenylethanoid glycoside. The content was 420% higher than that in the control HRs transformed with an “empty” vector [94].
The HRs of R. crenulata, when elicited with MeJa or SA, demonstrated an increase in salidrose content and a corresponding increase in the expression levels for the genes encoding tyrosine decarboxylase (TDC) and UGT. Overexpression of RcTDC in the genetically engineered HRs led to the enhanced accumulation of tyramine, tyrosol and salidroside [95]. Tyrosine aminotransferase (TAT) catalyzes the conversion of tyrosine into 4-hydroxyphenylpyruvate. Suppression of RcTAT in genetically engineered R. crenulata HRs caused increased salidroside production, suggesting that the enzyme may be engaged in a competing metabolic pathway [96].
Without the precursor (tyrosol) feeding, the HRs of R. kirilowii (Regel) Maxim. were found not to produce salidroside. Supplementation of the nutrient medium with tyrosol triggered the activity of tyrosol glucosyltransferase and salidroside biosynthesis. Measurements of tyrosol glucosyltransferase activity in the HRs revealed the maximum activity of the enzyme on day 18 of the culture. Tyrosol (2.5 mM) added on that day was converted into salidroside at the maximum rate [97]. Supplementation of the R. kirilowii HRs with cinnamic acid resulted in the rosavin (cinnamyl diglycosides) biosynthesis. In the optimum conditions (addition of 2.5 of mM cinnamic acid on the inoculation day and extra 1% sucrose on the 14th day of culture) the HRs accumulated 0.7 mg/g DW of rosarin and 14.0 mg/g DW of rosavin in the biomass, and released nearly 500 mg/L of rosavin to the culture medium. Exogenously applied cinnamic acid, however, hampered the growth of the HRs [98]. Salidroside content in the HRs of R. quadrifida was shown to be low (0.58–0.71 mg/g DW), but the rhizomes of the intact R. quadrifida plant accumulated much less salidroside than did those of R. rosea plants [99].
The HRs that accumulated high contents of phenylethanoids are listed in Table 2.

2.3. Hydroxycinnamates

Derivatives of hydroxycinnamic acids are ubiquitous in the plant kingdom. This subclass of polyphenolics includes acylquinic acids (with the most known chlorogenic acids) and conjugates of cinnamic acid derivatives with other organic acids (e.g., tartaric, shikimic, aldaric, dihydroxyphenyllactic) and with sugars. They are the common constituents of food plants (like blueberries, apples, sweet cherries, grapes, spinach, chicory, lettuce, and some culinary herbs). Coffee, ciders, and wines are also rich dietary sources of hydroxycinnamates. The importance of this group of polyphenols for human health is a subject of extensive research [16,30,100].

2.3.1. Conjugates with Tartaric Acid

Two compounds of this type have been described as metabolites of the HRs: caftaric (caffeoyltartaric acid, CTA) and chicoric (dicaffeoyltartaric, DCTA) acid (Figure 3). Recently, the chicoric acid biosynthetic pathway in Echinacea purpurea (L.) Moench (purple coneflower, Asteraceae) has been proposed by Fu and coworkers, with the HRs being one of the tools used in this research [101]. The final step in the biosynthesis takes place in the vacuole and is performed by a serine carboxypeptidase-like (SCPL) acyltransferase that uses chlorogenic acid as an acyl donor (instead of 1-O-caffeoyl-β-D-glucose) and transfers the caffeoyl group to CTA to form DCTA. CTA is synthesized in cytosol from caffeoyl-CoA and tartaric acid in a reaction catalyzed by an acyltransferase from the BAHD family.
The data on the pharmacological activity of CTA are sparse in comparison to those concerning DCTA. CTA counteracts oxidative damage of biological macromolecules and reduces inflammatory response. Moreover, it ameliorates indomethacin-induced gastric ulcer in rats [102,103]. DCTA enhances glucose uptake, diminishes insulin resistance and oxidative stress, demonstrates anti-inflammatory and antiviral activity, improves brain and liver functions, and may have beneficial effects in atherosclerosis and osteoarthritis [104,105,106,107]. In addition to ameliorating cognitive impairment via the promotion of the antioxidant defense system and preventing memory impairment and amyloidogenesis caused by systemic inflammation [108,109], DCTA improves neuron survival against inflammation [110].
Numerous attempts have been made to achieve the mass propagation of purple coneflower through in vitro methods and to start the biotechnological production of caffeoyltartaric acids from in vitro cultures of E. purpurea [111,112,113]. They have been lately summarized by Hussain et al. [5] in their comprehensive review on plant root cultures. The first HR cultures of E. purpurea were obtained by inoculation of hypocotyls of etiolated seedlings with R. rhizogenes strains ATCC 15834 and R1601. Hypocotyls infected with strains LMG63 and LMG150 produced only callus tissue. Alkamides [114] were the specialized metabolites analyzed in this study and no data on hydroxycinnamate content were provided [115]. The HRs initiated from leaf explants of purple coneflower with the R rhizogenes strain ATCC 43057 and cultivated in MS medium for 50 days produced up to 12.2 g/L of dry biomass, if extra sucrose was added on day 40. The roots contained DCTA (19.21 mg/g DW), CTA (3.56 mg/g DW), and 5-CQA (0.93 mg/g DW) [116]. The HRs transferred to continuous light (60 μmol/m2 s) did not show altered biomass accumulation but started to accumulate anthocyanins. Moreover, the contents of caffeoyltartaric acid rose markedly (DCTA up to 27 mg/g DW; CTA up to 6 mg/g DW). The increase in caffeic acid derivative content was correlated with a higher activity of PAL [117]. Addition of gibberellic acid (GA3; 0.025 μM) to the nutrient medium caused a further increase in PAL activity and caffeoyltartaric acid accumulation (DCTA: c. 36 mg/g DW; CTA: c. 7.5 mg/g DW). The content of 5-CQA was similar to that of CTA [118]. The roots grown in an airlift bioreactor with increased aeration (volume 17 L; 30 days growth cycle) yielded 9 g/L DW of biomass and a maximum 146.5 mg/L of DCTA (16.3 mg/g DW). An exposure to ultrasound (6 min at day 20) resulted in higher biomass accumulation (12.8 g/L) and higher DCTA content (178.2 mg/L) [119,120]. Sonication of the HRs grown in flasks (15-day-old roots sonicated for 6 min) caused a nearly 30% higher content of DCTA in the sonicated HRs compared to that in the control roots [121]. Alterations in macronutrient contents in ½ MS medium did not improve the productivity of the HRs obtained from leaf explants inoculated with the R. rhizogenes ATCC 15834 [122]. Demirci et al. [123] intiated the HR culture of purple coneflower from leaf explants inoculated with the R. rhizogenes strain ATCC 43057. The roots were maintained in the dark and subcultured every 21 days. After the initial 10 days of growth, the cultures were supplemented either with 24-epibrassinolide (0.5, 1.0, and 2.0 mg/L) or with L-phenylalanine (100, 500, and 1000 μM). DW, FW, and the metabolite contents in the roots were assessed every 10 days up to the 50th day of culture. Neither 24-epibrassinolide nor L-phenylalanine demonstrated detrimental effects on the HRs growth at the concentration range applied. The maximum DCTA content (20.2–24.1 mg/g DW) was achieved with 24-epibrassinolide treatment. Both treatments were advantageous for CTA and echinacoside production. This was the first study to describe the echinacoside production in E. purpurea HRs. Earlier, the compound was obtained from the adventitious root culture of E. angustifolia DC. [124].
Although DCTA is a major constituent of lettuces, neither the Lactuca virosa L. nor the L. indica L. (Asteraceae) HRs have been found to accumulate significant amounts of the compound [125,126].
Table 3 shows the HR cultures that accumulated high contents of CTA and DCTA.

2.3.2. Conjugates with Quinic Acid

Esters of trans-hydroxycinnamic acids with 1L-(-)-quinic acid synthesized and accumulated by plants are known as chlorogenic acids (CGAs). In the 1970s, new rules for describing the structure of 1L-(-)-quinic acid were recommended by the IUPAC, resulting in changes in the nomenclature of the compounds. According to IUPAC rules, one of the most common caffeoylquinic acids, chlorogenic acid, should be denoted as 5-O-caffeoylquinic acid (5-CQA). Formerly, the compound was known as 3-O-caffeoylquinic acid (3-CQA). Unfortunately, there is still no consensus regarding the nomenclature [16,127,128]. In this review, the IUPAC rules have been applied, but the numbering system used by the authors of particular papers cited herein is not always clear.
CGAs (Figure 4) are common constituents of food. Their daily intake in coffee drinkers is estimated to be 0.5–1.0 g (a cup of Espresso coffee contains about 200 mg of CGAs) [129]. Other than coffee, the main food contributors of CGAs are potatoes, apples, artichokes, plums, cherries, prunes, tomatoes, and carrots [130]. Questions concerning the bioavailability of CGAs have been summarized elsewhere [16,131]. One of the most recent studies on the subject examined the metabolic fate of artichoke polyphenols (mainly caffeoylquinic acids) after ingestion by healthy volunteers [132]. CGAs are the antioxidative and anti-inflammatory agents that demonstrate neuroprotective effects, prevent hypoxia-induced retinal degeneration, and counteract the formation of advanced glycation end products [16,128,133,134,135,136,137,138].
Although some of the CGAs can be easily obtained from crop plants, attempts have been made to achieve their production in plant tissue cultures [139].
Chlorogenic acid, an easy-to-detect and widely occurring plant metabolite, has been frequently described as a constituent of the HRs. Its content, however, is usually low. Some examples of 5-CQA occurrence in the HRs are given in Section 2.1. (Ficus carica, Leonotis nepetifolia, Leonurus sibiricus, Mentha spicata, Panax quinquefolius). The compound is engaged in plant defense against infections and insect infestation [140,141]. Yadav et al. [142] studied the interactions between R. tumefaciens and phenolic compounds exuded by cotyledonary nodal explants of Cicer arietinum L., the species that demonstrates recalcitrance to Rhizobium-mediated transformation. The inoculation with R. tumefaciens enhanced the production of some phenolics (including 5-CQA) in plant explants and the subsequent liberation of the compounds to the culture medium that in turn inhibited the growth of the bacterial vector and adversely affected the process of genetic transformation.
In another study, CGAs were detected in the HRs of Leontopodium alpinum Cass. (synonym of Leontopodium nivale subsp. alpinum (Cass.) Greuter; Asteraceae); however, they were not quantified [143].
The HRs of Lactuca virosa L. (Asteraceae), obtained by the infection of leaf explants with the R. rhizogenes strain LBA 9402 and cultivated in a modified MS medium in the dark were found to produce 3,5-DCQA (3,5-di-O-caffeoylquinic acid) as a major phenolic metabolite (25.8 mg/g DW). The content of 3,5-DCQA was much higher than that in the intact plant organs and callus tissue. The HRs also produced 5-CQA (11.3 mg/g DW) and DCTA (7.9 mg/g DW). Leaves of the intact L. virosa plant contained twice as much DCTA [125]. L. serriola L. transformed with the R rhizogenes strain AR15834 developed HRs that exhibited a higher total reducing capacity than did the untransformed roots. The CGA content in the roots was not analyzed [144]. L. indica was transformed using the R. rhizobacterium strain R1000. The developed roots accumulated 3,5-DCQA, DCTA, and 5-CQA, albeit in amounts lower than those found in L. virosa. The best results were achieved using MS medium and 100 μM of MeJa as an elicitor. The maximum contents of the CGAs and DCTA were detected 72 h after the addition of MeJa (3,5-CQA, c. 0.14 mg/g FW; DCTA, c. 0.14 mg/g FW; 5-CQA, c. 0.02 mg/g FW) [126].
Chicory (Cichorium intybus L.; Asteraceae) is closely taxonomically and chemically related to lettuces. In one study, the HRs of the plant were obtained by inoculation of leaf explants with the R. rhizogenes strain LBA 9402 and grown in a modified MS medium either under a photoperiod (cool white fluorescent tubes, 20 μE m−2 s−1, 16 h light) or in the dark. Exposure to light was advantageous for CGA production in the HRs. At the stationary phase of growth, the roots cultivated under the photoperiod contained 55.7 mg/g DW of 3,5-DCQA, 9.4 mg/g DW of 5-CQA, and 1.3 mg/g DW of DCTA [145].
In another study, leaves of C. intybus var Orchies were inoculated with the R. rhizogenes strain 2659 to obtain the HRs. The roots were grown in ½ MS medium under a 16 h light photoperiod. After the elicitation with MeJa (on day 12), production of CGAs was strongly promoted, and 12 days after the addition of MeJa (0.15 mM), the content of 3,5-DCQA in the HRs reached 12% DW (120 mg/g DW). From the elicited roots, 3,4,5-tri-O-caffeoylquinic acid was isolated, the compound formerly undetected in the HRs of chicory. Unelicited roots produced approximately 10 mg/g DW of 5-CQA and 50 mg/g DW of 3,5-DCQA [146].
Eclipta prostrata (L.) L. (Asteraceae) HRs, in other research, were obtained via the infection of aseptic seedlings with R. rhizogenes LBA 9796 and were elicited with jasmonic acid (JA) and MeJa to enhance production of wedelolactones and 3,5-DCQA. The roots, after 4 days of growth in the presence of either 100 μM of JA or 140 μM of MeJa, accumulated 44.71 mg/g DW and 41.62 mg/g DW of 3,5-DCQA, respectively. Control cultures contained 18.08 mg/g DW of the compound [147]. Lopes et al. [148] investigated the biosynthesis of demethylwedelolactone, wedelolactone, and 3,5-DCQA in the HRs of E. prostrata. Feeding with [3-13C]DL-phenylalanine proved that in contrast to wedelolactones, 3,5-DCQA molecule originates entirely from the shikimate pathway.
Young, aseptic leaves of Stevia rebaudiana Bertoni (Asteraceae) var. FengNong 3 were inoculated with R. rhizogenes strain C58C1 in one study. The developed HRs were cultivated in ½ MS medium under a 12/12 h (light/dark) photoperiod. The root clone that showed the best growth yielded 15.23 g/100 mL of FW and 1.54 g/100 mL DW of biomass, after 24 days of culture. Roots of the clone accumulated chlorogenic acid (39.41 mg/g DW) and 3,5-DCQA (48.10 mg/g DW). Some of the obtained root clones produced up to 4.29 mg/g of 4,5-DCQA. The optimum for both CGA production and root growth was the B5 medium [149].
A total of 11 CGAs produced by the HRs of Rhaponticum carthamoides (Willd.) Iljin (Asteraceae) transformed with R. rhizogenes strain A4 were tentatively identified using UPLC-PDA-ESI-MS3 analysis. 5-CQA (5.12 mg/g DW), 3,5-DCQA (3.08 mg/g DW), and one of the two tri-O-caffeoylquinic acid derivatives (5.97 mg/g DW) were the most abundant CGAs from the roots cultivated in the presence of light. The HRs cultivated in the dark produced less CGAs [150]. An extract from the HRs effectively induced apoptosis in human glioma primary cells [151].
CGAs produced by the HRs of Polyscias filicifolia L.H.Bailey (Araliaceae) transformed with R. rhizogenes strain ATCC 15834 were tentatively identified using the HPLC-PDA-ESI-MS method and nine p-coumaroylquinic, caffeoylquinic, and feruloylquinic acids were found in the culture. The HRs elicited for 1 day with 100 μM of MeJA and then transferred to control medium for 30-day cultivation produced up to 1.6 mg/g DW of chlorogenic acid [152].
The Hypericum perforatum L. (Hypericaceae) HRs transformed with R. rhizogenes strain A4M70GUS were found to accumulate the following monoacylquinic acids: chlorogenic acid (0–6.94 mg/g DW), p-coumaroylquinic acid (0.04–0.09 mg/g DW), and feruloylquinic acid (0.11–0.94 mg/g DW). The contents of CGAs depended on the analyzed root line and were much lower than those in the shoots regenerated from the HRs [153].
The HRs that accumulated high contents of CGAs are shown in Table 4.

2.3.3. Rosmarinic Acid, Salvianolic Acids, and Miscellaneous Compounds

Plant polyphenolics of this group are typically associated with Lamiaceae plants. The seeds, leaves, and roots of many species included in the family are widely used either as spices (mint, basil, savory, rosemary, oregano, marjoram) or as medicines (lemon balm, lavender, thyme). Rosmarinic acid (RA) and salvianolic acids (see Figure 5), the compounds of potential use in medicine and cosmetics, have attracted much attention. Their biosynthesis, distribution in the plant kingdom, biotechnological production, and activity have been the subject of several review papers [154,155,156,157,158].
Ocimum basilicum L. (Lamiaceae), a medicinal and aromatic plant commonly used as a spice, is a source of RA and other valuable metabolites. Studies on the genus Ocimum have been recently summarized by Pandey et al. [159]. Research on HRs of Ocimum spp. began in the 1990s.
In one study, two agropine and three mikimopine clones of the HRs were obtained by inoculation of leaf discs from Ocimum basilicum L. with two different strains of R. rhizogenes: ATCC 15834 and MAFF 03-01724. The root clones were cultured in the dark in three different nutrient media: MS, B5, and WP (woody plant medium). The highest DW (450–700 mg), after 6-week culture, was achieved with the MS medium. The roots, depending on the clone, contained 2.5–7.0% DW of RA, except for one highly productive clone (c. 12% DW RA). The HRs grown in WP and B5 medium provided c. 400 mg DW of biomass at the end of the culture cycle. The RA content in the roots was 0–14% (B5 medium) and 6–12% (WP medium). Lithospermic acid (up to 1.5% DW) and lithospermic acid B (0–0.75% DW) were also found in the culture [160]. Bais et al. [161] infected axenic plantlets of O. basilicum with the R. rhizogenes strain ATCC 15834 to start the HR cultures of the plant. The established culture, grown in MS medium and subcultured every 4 weeks, produced 3% FW of RA. The elicitation attempts with SA (50, 100 and 200 μM), MeJa (100, 200 and 500 μM), and chitosan (0.10, 0.12, and 0.15%), added at the beginning of culture cycle, failed due to the detrimental effect of the elicitors on both the growth of roots and RA production. The HRs elicited with cell wall extracts from Phytophthora cinnamoni and P. drechsleri (1, 2, and 3%) exhibited enhanced accumulation of biomass (up to 105 g/L FW) and increased RA production (up to 8.1% FW). RA was not liberated to the culture medium. Moreover, antifungal activity of RA (50–250 μM) was proven against Pythium ultimum, Phytophthora aphanidermatum, Aspergillus niger, and Fusarium oxysporum. Four strains of R. rhizogenes were used to obtain HRs from three cultivars of O. basilicum (Subja, Holy Green, and Red Rubin). The highest biomass of roots (114-116 mg DW from one explant) was achieved after 60 days of growth, but the maximum RA content (66.2–71.0 mg/g DW) was found 40 days after the inoculation of the HRs into the fresh medium [162]. Coculture of the HRs with arbuscular mycorrhizal fungus Rhizophagus irregularis (earlier Glomus intraradices) resulted in a significantly higher biomass and RA accumulation [163]. Kwon et al. [164] investigated the HRs of purple and green basil. They found that although light favors both biomass and RA production in green basil, the purple cultivar used in the study demonstrated better productivity when grown in the dark. The cultures of red basil (O. basilicum cv. “Purpurascens’) accumulated less RA than did those of the green cultivar (O. basilicum cv. “Cinnamon”).
RA and nine phenolic acids were identified as constituents of the Hyssopus officinalis L. (Lamiaceae) HRs obtained by inoculation of leaf explants with R. rhizogenes LBA 9402 [160a]. The highest RA content, 6% DW (c. 0.9 g/L), was detected in the HRs grown in GB5 medium containing 10% sucrose [165].
The Dracocephalum kotschyi Boiss (Lamiaceae) HRs were induced by infecting with R. rhizogenes LBA 9402 in one study. The roots synthesized a number of polymethoxylated flavonoids and RA (up to 1.4 mg/g DW) [166]. Internodal explants of D. forrestii W.W. Smith, infected with the R. rhizogenes strain A4, developed HRs that produced up to 20 mg/g DW of RA and up to 6.9 mg/g DW of salvianolic acid B. The optimum for the polyphenols production was a WP medium containing 3% sucrose. The HRs were grown in the dark [167]. The maximum RA content (1.2 mg/g FW) in the HRs of D. kotschyi was achieved via elicitation with Fe3O4 nanoparticles (75 mg/L, 24 h) [168].
From the HRs of Nepeta teyeda Webb et Berth. (Lamiaceae) transformed with R. rhizogenes ATCC 15834, formerly unknown compounds, including the sesquiterpene (-)-cinalbicol, the diterpene teydeadione, a degraded C23-triterpene (teydealdehyde), and three fatty acid esters of lanosta-7,24-dien-3β-ol, were isolated together with two dehydroabietane diterpenes, eight triterpenes, and eighteen known phenolic compounds. Moreover, the propyl ester of rosmarinic acid was isolated for the first time from a natural source [169]. The HRs of N. cataria L. produced up to 13.6 mg/g DW of RA. Indole-3-butyric acid (IBA) added to the culture medium (0.5 mg/L) increased accumulation of RA in the roots (19.2 mg/g DW). A similar result was achieved via treatment with putrescine (50 mg/L) [170,171].
Undifferentiated cultures of Coleus spp and Plectranthus spp. (Lamiaceae) have been shown to be excellent producers of RA [154,172]. The C. forskohlii Briq. HRs, induced by R. rhizogenes MAFF 03-01724, accumulated up to 9.5 mg of RA per flask (c. 4g FW) one week after elicitation with 100 μM of Meja [173]. C. blumei Benth. (Plectranthus scutellarioides R.Br.) infected with R. rhizogenes A4 developed HRs that produced c. 50 mg/g DW of RA. Upon elicitation with MeJa (100 μM, 8 days), the RA content rose to c. 80 mg/g DW [174]. A synthetic gene encoding β-cryptogein (a peptide implicated in a response to pathogens) was introduced into the HRs of C. blumei with a R. rhizogenes A4-carrying binary vector. The roots with β-cryptogein expression liberated phenolics to the culture medium. The amount of liberated RA reached 0.25 mg per 1 L of the medium [175]. Effects of hydroxyphenylpyruvate reductase (HPPR) and rosmarinic acid synthase (RAS) overexpression and suppression were studied in C. blumei HRs transformed with R. rhizogenes ATCC 15834 carrying different genetic constructs. The HPPR-overexpressing line with increased HPPR mRNA levels accumulated more RA (3% DW) than did the control roots (c. 1.8%) [176].
The Prunella vulgaris L. (Lamiaceae) HRs transformed with R. rhizogenes ATCC 15834 were found to accumulate 15–30 times more RA than did the original P. vulgaris plant. The content of RA in the HRs (c. 40 mg/g DW) increased up to c. 60 mg/g DW when the roots were treated with either 0.2 mg/L of ethephon for 5 days or with 6.9 mg/L of SA for 2 days. The relative expression levels of the genes encoding PAL and HPPR, 2–8 days after exposure of the HRs to the elicitor, were significantly higher than were those in the control roots [177]. Tyrosine aminotransferase engaged in the RA biosynthesis in P. vulgaris (PvTAT) was isolated and described from root of the plant. Antisense- and sense PvTAT-expressing hairy root lines were developed, and PvTAT gene expression levels, TAT enzyme activity, and RA content were analyzed in the obtained root clones. Antisense-expressing HRs demonstrated a reduced TAT activity and a decrease in RA content. Overexpression of PvTAT led to the enhanced accumulation of RA (up to 70 mg/g DW) [178]. The HRs of P. vulgaris transformed with ATCC 15834 contained over 80 mg/g DW of RA (control roots c. 50 mg/g) after the elicitation with 100 μM of MeJa (5 days) [179].
Leaf and stem explants of Agastache rugosa Kuntze (Lamiaceae), after inoculation with R. rhizogenes R1000, were reported to develop HRs that produced 14.1 g DW/L of biomass and 116 mg/g DW of RA after two weeks of culture in MS medium containing 3% sucrose. The roots were maintained in a growth chamber with a 16 h photoperiod [180]. Lepechinia caulescens (Ortega) Epling (Lamiaceae) roots transformed with R. rhizogenes ATCC 15834 in optimum conditions (MS salts, B5 vitamins, 4.5% sucrose) produced 41.7 mg/g DW of RA. The content increased to c. 60 mg/g DW after the elicitation with 200 μM of MeJa (24 h) [181]. Numerous HRs of the Lamiaceae plants were established for the active metabolite production, including the formerly described HRs of Mentha spicata (70 μg/g DW of RA and 60 μg/g DW of lithospermic acid B) [57] and Leonotis nepetifolia (2.6 mg/g DW of RA) [58].
It is worth mentioning the biosynthesis of RA and lithospermic acid B (synonyms: monardic acid B and salvianolic acid B) in the HRs of Rindera graeca (A.DC.) Boiss. & Heldr. (Boraginaceae). It was found that the HRs contained up to 33.7 mg/g of RA and up to 106.1 mg/g DW of lithospermic acid B on the 30th day of the culture cycle. Cold stress increased concentration of the polyphenolics in the roots [182].
Numerous papers have reported on the HRs of different Salvia species: S. bulleyana Diels, S. nemorosa L., S. officinalis L., S. przewalskii Maxim., S. virgata Jacq., S. viridis L., and the most popular, S. miltiorrhiza Bunge, a renowned remedy of TCM. Several dozen of papers on the engineering of the secondary metabolism in S. miltiorrhiza have been published since 2015, and the volume of data clearly deserves separate review. This is why we have decided to exclude this species from the current paper.
The HRs of S. officinalis, obtained by infection of sterile shoots of the plant with R. rhizogenes ATCC 15834, were found to contain up to 45 mg/g DW of RA. The sage HRs that were cultivated in the dark tended to accumulate less RA than did the roots maintained under a photoperiod (16 h light) [183]. Methanolic extract from the HRs of S. officinalis contained 30.9 mg/g DW of RA and demonstrated high antioxidative and radical scavenging activity and in some assays, higher than those of roots and shoots of the in-vivo-grown plants [184]. After the transfer to laboratory-scale sprinkle nutrient bioreactor, the sage HRs produced 34.7 mg/g DW (477.1 mg/L) of RA [185]. The HRs of S. nemorosa, cultivated in ½ MS medium containing 3% sucrose accumulated over 50% less RA (15.9 mg/g DW) [186]. Ag+ ions, YE, and MeJa were applied to enhance the productivity of the S. virgata HRs. The best results were achieved with MeJa (72 h) at a final concentration of 22.4 ppm. The elicited roots produced up to 18.5 mg/g DW of RA and up to 2.1 mg/g DW of salvianolic acid A [187]. MeJa at a final concentration of 400 μM and SA (50 μM) were applied to stimulate the growth and production of polyphenolics in the HRs of S. przewalskii. The roots treated with MeJa, 3 days after addition of the elicitor, contained 67.1 mg/g DW of RA and 21.4 mg/g DW of salvianolic acid B. Moreover, the elicited roots produced increased amounts of tanshinones (diterpenoids). The accumulation of polyphenolics in the elicited HRs of S. przewalskii was accompanied by the elevated transcription levels of PAL, 4CL, TAT, HPPR, and RAS [188]. The HRs of S. viridis accumulated up to 35 mg/g DW of RA and minor quantities of other polyphenolics, tentatively identified by UPLC-PDA-ESI–MS2 as prolithospermic acid, salvianolic acid J, two hexosides of RA, salvianolic acid E, methyl rosmarinate, and two isomers of salvianolic acid F [189]. An attempt to optimize the culture conditions with the proper choice of a nutrient medium formulation and the appropriate light regime was also made [190]. Leaves and shoots of the axenic S. bulleyana plants were infected with R. rhizogenes A4 to obtain the HRs. The growth index of the HRs, measured after five weeks of growth in a liquid WP medium, was 8–14, depending on the clone. The RA content in the HRs reached 40 mg/g DW, and this compound dominated the polyphenolic fraction in the roots. Minor quantities of salvianolic acids E, K, and F and methyl rosmarinate were also detected [191]. The roots were elicited with MeJa, YE, trans-anethole, and CdCl2. Chromium salt had a detrimental effect on both biomass and polyphenol accumulation. The most effective method for the induction of the enhanced RA production (over 100 mg/g DW) was the application of MeJa (100 μM, 3 days). In the elicited roots, the increased accumulation of caffeic acid preceded the increased production of polyphenolics [192]. Optimization of the culture conditions for the S. bulleyana HRs revealed the advantageous effects of diminished sucrose concentration (2%), diminished contents of salts and vitamins, and the lack of illumination on polyphenol accumulation in the biomass. In the optimum conditions, the roots produced over 90 mg/g DW of RA and over 10 mg/g DW of salvianolic acid K [193].
The HR cultures that produced high yields of RA, lithospermic acid B, and salvianolic acid K are listed in Table 5.

3. Conclusions

Table 1, Table 2, Table 3, Table 4 and Table 5 summarize the most remarkable achievements of the research on highly productive hairy root cultures containing phenolic/polyphenolic antioxidants from the simple phenolics, phenylethanoid, and hydroxycinnamate group. Rosmarinic acid production by the HRs of the Lamiaceae plants has been shown to be one of the best examples of the biosynthetic potential of the HRs. The high yields of the phenolic/polyphenolic natural products in the HRs were obtained using procedures such as the optimization of culture conditions, selection of the optimum bacterial strain and plant material to initiate the HRs, elicitation, supplementation with phytohormones, precursor (feeding, and genetic engineering (overexpression, heterologous expression, or silencing of the genes encoding the regulatory transcription factors and biosynthetic enzymes). In the analyzed experimental works, the elicitation with methyl jasmonate was the most extensively used method for stimulation of polyphenolics production. Cell wall preparations from fungal or bacterial pathogens also showed some potential. However, the high contents of polyphenols in the HRs may be accompanied by poor root growth. This may be unfavorable for the potential applications of the cultures that accumulate products in the biomass.
The HR cultures proved to be a valuable tool for the characterization of regulatory mechanisms in plant phenol/polyphenol biosynthesis. They were used for elucidation of the functions of particular biosynthetic enzymes and transcription factors. At present, the HR cultures are primarily useful in the genetic engineering of plants. Despite the efforts to maximize production of valuable phenolics/polyphenolics using the HRs, the implementation of the industrial processes using HRs to produce antioxidant natural products seems to be limited not only by economic issues but also by the current regulations concerning genetically transformed cultures.

Author Contributions

Conceptualization, A.S.; methodology, A.S. and J.M.; resources, A.S., J.M. and Y.V.Y.; data curation, A.S.; writing—original draft preparation, A.S. and Y.V.Y.; writing—review and editing, A.S., J.M. and Y.V.Y.; visualization, J.M.; supervision, A.S.; project administration, A.S.; funding acquisition, A.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

3:5-DCQA3,5-Dicaffeoylquinic acid
4CL4-Coumarate:CoA ligase
5-CQA5-Caffeoylquinic acid
AChEAcetylcholinesterase
ALDHAlcohol dehydrogenase
AOXAlternative oxidase
BChEButyrylcholinesterase
C3HCoumaroylquinate (coumaroylshikimate) 3’-monooxygenase
C4HCinnamate-4-hydroxylase
CGAsChlorogenic acids
CTACaftaric acid
CuAOCopper-containing amine oxidase
DCTAChicoric acid
DWDry weight
DXR1-Deoxy-D-xylulose 5-phosphate reductoisomerase
FWFresh weight
GB5Gamborg’s B5
HBDHydroxybenzoate dehydrogenase
HBSp-Hydroxybenzaldehyde synthase
HCHL4-Hydroxycinnamoyl-CoA hydratase/lyase
HCTShikimate O-hydroxycinnamoyltransferase
HPLC-PDA-ESI-MSHigh performance liquid chromatography with photodiode detector electrospray ion source and mass spectrometry
HPPRHydroxyphenylpyruvate reductase
HRHairy root
MeJaMethyl jasmonate
MSMurashige and Skoog
PALPhenylalanine ammonia lyase
PAP1 Production of anthocyanin pigment 1
PGPropyl gallate
p-HBAp-Hydroxybenzoic acid (4-hydroxybenzoic acid)
PKPyruvate kinase
PPOPolyphenol oxidase
RARosmarinic acid
RASRosmarinic acid synthase
SASalicylic acid
SHAMSalicylhydroxamic acid
SKHDShikimate dehydrogenase
SODSuperoxide dismutase
TATTyrosine aminotransferase
TCMTraditional Chinese medicine
TDCTyrosine decarboxylase
TyDCTyrosine/DOPA decarboxylase
UGTUDP-Glucose glucosyltransferase
WPWoody plant
YEYeast extract

References

  1. Young, J.M.; Kuykendall, L.D.; Martínez-Romero, E.; Kerr, A.; Sawada, H. A revision of Rhizobium Frank 1889, with an emended description of the genus, and the inclusion of all species of Agrobacterium Conn 1942 and Allorhizobium undicola de Lajudie et al. 1998 as new combinations: Rhizobium radiobacter, R. rhizogenes, R. rubi, R. undicola and R. vitis. Int. J. Syst. Evol. Microbiol. 2001, 51, 89–103. [Google Scholar] [CrossRef] [PubMed]
  2. Hamill, J.D.; Lidgett, A.J. Hairy Root Cultures—Opportunities and Key Protocols for Studies in Metabolic Engineering. In Hairy Roots. Culture and Applications, 1st ed.; Doran, P., Ed.; Harwood Academic Publishers: Amsterdam, The Netherlands, 1997; pp. 1–29. [Google Scholar]
  3. Gutierrez-Valdes, N.; Häkkinen, S.T.; Lemasson, C.; Guillet, M.; Oksman-Caldentey, K.M.; Ritala, A.; Cardon, F. Hairy root cultures—A versatile tool with multiple applications. Front. Plant Sci. 2020, 11, 33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Shi, M.; Liao, P.; Nile, S.H.; Georgiev, M.I.; Kai, G. Biotechnological exploration of transformed root culture for value-added products. Trends Biotechnol. 2021, 39, 137–149. [Google Scholar] [CrossRef] [PubMed]
  5. Hussain, M.J.; Abbas, Y.; Nazli, N.; Fatima, S.; Drouet, S.; Hano, C.; Abbasi, B.H. Root cultures, a boon for the production of valuable compounds: A comparative review. Plants 2022, 11, 439. [Google Scholar] [CrossRef]
  6. Gantait, S.; Mukherjee, E. Hairy root culture technology: Applications, constraints and prospect. Appl. Microbiol. Biotechnol. 2021, 105, 35–53. [Google Scholar] [CrossRef]
  7. Gubser, G.; Vollenweider, S.; Eibl, D.; Eibl, R. Food ingredients and food made with plant cell and tissue cultures: State-of-the art and future trends. Eng. Life Sci. 2021, 21, 87–98. [Google Scholar] [CrossRef]
  8. Brewer, M.S. Natural antioxidants: Sources, compounds, mechanisms of action, and potential applications. Compr. Rev. Food Sci. Food Saf. 2011, 10, 221–247. [Google Scholar] [CrossRef]
  9. Krishnajah, D.; Sarbatly, R.; Nithyanandam, R. A review of the antioxidant potential of medicinal plant species. Food Bioprod. Process. 2011, 89, 217–233. [Google Scholar] [CrossRef]
  10. Wang, H.; Khor, T.O.; Shu, L.; Su, Z.; Fuentes, F.; Lee, J.-H.; Kong, A.-N.T. Plants against cancer: A review on natural phytochemicals in preventing and treating cancers and their druggability. Anticancer Agents Med. Chem. 2012, 12, 1281–1305. [Google Scholar] [CrossRef]
  11. Shin, S.-A.; Moon, S.Y.; Kim, W.-Y.; Paek, S.-M.; Park, H.H.; Lee, C.S. Structure-based classification and anti-cancer effects of plant metabolites. Int. J. Mol. Sci. 2018, 19, 2651. [Google Scholar] [CrossRef] [Green Version]
  12. Olivero-David, R.; Ruiz-Roso, M.B.; Caporaso, N.; Perez-Olleros, L.; De las Heras, N.; Lahera, V.; Ruiz-Roso, B. In vivo bioavailability of polyphenols from grape by-product extracts, and effect on lipemia of normocholesterolemic Wistar rats. J. Sci. Food Agric. 2018, 98, 5581–5590. [Google Scholar] [CrossRef]
  13. Cui, X.; Lin, Q.; Liang, Y. Plant-derived antioxidants protect the nervous system from aging by inhibiting oxidative stress. Front. Aging Neurosci. 2020, 12, 209. [Google Scholar] [CrossRef] [PubMed]
  14. Toma, L.; Sanda, G.M.; Niculescu, L.S.; Deleanu, M.; Sima, A.V.; Stancu, C.S. Phenolic Compounds Exerting Lipid-Regulatory, Anti-Inflammatory and Epigenetic Effects as Complementary Treatments in Cardiovascular Diseases. Biomolecules 2020, 10, 641. [Google Scholar] [CrossRef] [Green Version]
  15. Sarkar, D.; Christopher, A.; Shetty, K. Phenolic bioactives from plant-based foods for glycemic control. Front. Endocrinol. 2022, 12, 727503. [Google Scholar] [CrossRef] [PubMed]
  16. Clifford, M.N. Chlorogenic acids and the acyl-quinic acids: Discovery, biosynthesis, bioavailability and bioactivity. Nat. Prod. Rep. 2017, 34, 1391–1421. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Edwards, C.A.; Havlik, J.; Cong, W.; Mullen, W.; Preston, T.; Morrison, D.J.; Combet, E. Polyphenols and health: Interactions between fibre, plant polyphenols and the gut microbiota. Nutr. Bull. 2017, 42, 356–360. [Google Scholar] [CrossRef] [PubMed]
  18. Pešić, M.B.; Milinčić, D.D.; Kostič, A.Ž.; Stanisavljević, N.S.; Vukotić, G.N.; Kojić, M.O.; Gašić, U.M.; Barać, M.B.; Stanojević, S.P.; Popović, D.A.; et al. In vitro digestion of meat- and cereal-based food matrix enriched with grape extracts: How are polyphenol composition, bioaccessibility and antioxidant activity affected? Food Chem. 2019, 284, 28–44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Ullah, H.; Hussain, Y.; Santarcangelo, C.; Baldi, A.; Di Minno, A.; Khan, H.; Xiao, J.; Daglia, M. Natural polyphenols for the preservation of meat and dairy products. Molecules 2022, 27, 1906. [Google Scholar] [CrossRef]
  20. Awad, A.M.; Kumar, P.; Ismail-Fitry, M.R.; Jusoh, S.; Ab Aziz, M.F.; Sazili, A.Q. Overview of plant extracts as natural preservatives in meat. J. Food Process. Preserv. 2022, 46, e16796. [Google Scholar] [CrossRef]
  21. Pateiro, M.; Domínguez, R.; Munekata, P.E.S.; Nieto, G.; Bangar, S.P.; Dhama, K.; Lorenzo, J.M. Bioactive compounds from leaf vegetables as preservatives. Foods 2023, 12, 637. [Google Scholar] [CrossRef]
  22. Kanlayavattanakul, M.; Lourith, N. Plants and natural products for the treatment of skin hyperpigmentation—A review. Planta Med. 2018, 84, 988–1006. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Shin, S.; Cho, S.H.; Park, D.; Jung, E. Anti-skin aging properties of protocatechuic acid in vitro and in vivo. J. Cosmet. Dermatol. 2020, 19, 977–984. [Google Scholar] [CrossRef] [PubMed]
  24. Zduńska, K.; Dana, A.; Kolodziejczak, A.; Rotsztejn, H. Antioxidant properties of ferulic acid and its possible application. Ski. Pharmacol. Physiol. 2018, 31, 332–336. [Google Scholar] [CrossRef]
  25. Cimmino, A.; Andolfi, A.; Abouzeid, M.; Evidente, A. Polyphenols as fungal phytotoxins, seed germination stimulants and phytoalexins. Phytochem. Rev. 2013, 12, 653–672. [Google Scholar] [CrossRef]
  26. Malarz, J.; Michalska, K.; Yudina, Y.V.; Stojakowska, A. Hairy Root Cultures as a Source of Polyphenolic Antioxidants: Flavonoids, Stilbenoids and Hydrolyzable Tannins. Plants 2022, 11, 1950. [Google Scholar] [CrossRef] [PubMed]
  27. Mustafa, N.R.; Verpoorte, R. Chorismate derived C6C1 compounds in plants. Planta 2005, 222, 1–5. [Google Scholar] [CrossRef]
  28. Dewick, P.M. Medicinal Natural Products: A Biosynthetic Approach, 3rd ed.; John Wiley & Sons Ltd.: Chichester, UK, 2009; pp. 137–184. [Google Scholar] [CrossRef]
  29. Tomás-Barberán, F.A.; Clifford, M.N. Dietary hydroxybenzoic acid derivatives—Nature, occurrence and dietary burden. J. Sci. Food Agric. 2000, 80, 1024–1032. [Google Scholar] [CrossRef]
  30. Coman, V.; Vodnar, D.C. Hydroxycinnamic acids and human health: Recent advances. J. Sci. Food Agric. 2020, 100, 483–499. [Google Scholar] [CrossRef]
  31. Sircar, D.; Mitra, A. Accumulation of p-hydroxybenzoic acid in hairy roots of Daucus carota 2: Confirming biosynthetic steps through feeding of inhibitors and precursors. J. Plant Physiol. 2009, 166, 1370–1380. [Google Scholar] [CrossRef]
  32. Mitra, A.; Mayer, M.J.; Mellon, F.A.; Michael, A.J.; Narbad, A.; Parr, A.J.; Waldron, K.W.; Walton, N.J. 4-Hydroxycinnamoyl-CoA hydratase/lyase, an enzyme of phenylpropanoid cleavage from Pseudomonas, causes formation of C6-C1 acid and alcohol glucose conjugates when expressed in hairy roots of Datura stramonium L. Planta 2002, 215, 79–89. [Google Scholar] [CrossRef]
  33. Ur Rahman, L.; Kouno, H.; Hashiguchi, Y.; Yamamoto, H.; Narbad, A.; Parr, A.; Walton, N.; Ikenaga, T.; Kitamura, Y. HCHL expression in hairy roots of Beta vulgaris yields a high accumulation of p-hydroxybenzoic acid (pHBA) glucose ester, and linkage of pHBA into cell walls. Bioresour. Technol. 2009, 100, 4836–4842. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Yamanaka, M.; Shimomura, K.; Sasaki, K.; Yoshihira, K.; Ishimaru, K. Glucosylation of phenolics by hairy root cultures of Lobelia sessilifolia. Phytochemistry 1995, 40, 1149–1150. [Google Scholar] [CrossRef]
  35. Yan, C.-Y.; Yu, R.-M.; Zhang, Z.; Kong, L.-Y. Biotransformation of 4-hydroxybenzen derivatives by hairy root cultures of Polygonum multiflorum Thunb. J. Integr. Plant Biol. 2007, 49, 207–212. [Google Scholar] [CrossRef]
  36. Chen, X.; Zhang, J.; Liu, J.-H.; Yu, B.-Y. Biotransformation of p-, m-, and o-hydroxybenzoic acids by Panax ginseng hairy root cultures. J. Mol. Catal. B Enzym. 2008, 54, 72–75. [Google Scholar] [CrossRef]
  37. Sircar, D.; Roychowdhury, A.; Mitra, A. Accumulation of p-hydroxybenzoic acid in hairy roots of Daucus carota. J. Plant Physiol. 2007, 164, 1358–1366. [Google Scholar] [CrossRef]
  38. Sircar, D.; Mitra, A. Evidence for p-hydroxybenzoate formation involving enzymatic phenylpropanoid side-chain cleavage in hairy roots of Daucus carota. J. Plant Physiol. 2008, 165, 407–414. [Google Scholar] [CrossRef]
  39. Sircar, D.; Cardosoa, H.G.; Mukherjee, C.; Mitra, A.; Arnholdt-Schmitt, B. Alternative oxidase (AOX) and phenolic metabolism in methyl jasmonate-treated hairy root cultures of Daucus carota L. J. Plant Physiol. 2012, 169, 657–663. [Google Scholar] [CrossRef]
  40. Mukherjee, C.; Sircar, D.; Chatterjee, M.; Das, S.; Mitra, A. Combating photooxidative stress in green hairy roots of Daucus carota cultivated under light irradiation. J. Plant Physiol. 2014, 171, 179–187. [Google Scholar] [CrossRef]
  41. Mukherjee, C.; Samanta, T.; Mitra, A. Redirection of metabolite biosynthesis from hydroxybenzoates to volatile terpenoids in green hairy roots of Daucus carota. Planta 2016, 243, 305–320. [Google Scholar] [CrossRef]
  42. Chung, I.-M.; Rekha, K.; Rajakumar, G.; Thiruvengadam, M. Production of glucosinolates, phenolic compounds and associated gene expression profiles of hairy root cultures in turnip (Brassica rapa ssp. rapa). 3 Biotech 2016, 6, 175. [Google Scholar] [CrossRef] [Green Version]
  43. Chung, I.-M.; Rekha, K.; Rajakumar, G.; Thiruvengadam, M. Influence of silver nanoparticles on the enhancement and transcriptional changes of glucosinolates and phenolic compounds in genetically transformed root cultures of Brassica rapa ssp. rapa. Bioprocess Biosyst. Eng. 2018, 41, 1665–1677. [Google Scholar] [CrossRef] [PubMed]
  44. Chung, I.-M.; Rekha, K.; Rajakumar, G.; Thiruvengadam, M. Production of bioactive compounds and gene expression alterations in hairy root cultures of chinese cabbage elicited by copper oxide nanoparticles. Plant Cell Tissue Org. Cult. 2018, 134, 95–106. [Google Scholar] [CrossRef]
  45. Thiruvengadam, M.; Praveen, N.; John, K.M.M.; Yang, Y.-S.; Kim, S.-H.; Chung, I.-M. Establishment of Momordica charantia hairy root cultures for the production of phenolic compounds and determination of their biological activities. Plant Cell Tissue Org. Cult. 2014, 118, 545–557. [Google Scholar] [CrossRef]
  46. Thiruvengadam, M.; Rekha, K.; Chung, I.-M. Induction of hairy roots by Agrobacterium rhizogenes-mediated transformation of spine gourd (Momordica dioica Roxb. ex. willd) for the assessment of phenolic compounds and biological activities. Sci. Hortic. 2016, 198, 132–141. [Google Scholar] [CrossRef] [PubMed]
  47. Yoon, J.-Y.; Chung, I.-M.; Thiruvengadam, M. Evaluation of phenolic compounds, antioxidant and antimicrobial activities from transgenic hairy root cultures of gherkin (Cucumis anguria L.). S. Afr. J. Bot. 2015, 100, 80–86. [Google Scholar] [CrossRef]
  48. Thiruvengadam, M.; Praveen, N.; Kim, E.-H.; Kim, S.-H.; Chung, I.-M. Production of anthraquinones, phenolic compounds and biological activities from hairy root cultures of Polygonum multiflorum Thunb. Protoplasma 2014, 251, 555–566. [Google Scholar] [CrossRef] [PubMed]
  49. Ho, T.-T.; Lee, J.-D.; Ahn, M.-S.; Kim, S.-W.; Park, S.-Y. Enhanced production of phenolic compounds in hairy root cultures of Polygonum multiflorum and its metabolite discrimination using HPLC and FT-IR methods. Appl. Microbiol. Biotechnol. 2018, 102, 9563–9575. [Google Scholar] [CrossRef] [PubMed]
  50. Tomasiak, A.; Zhou, M.; Betekhtin, A. Buckwheat in Tissue Culture Research: Current Status and Future Perspectives. Int. J. Mol. Sci. 2022, 23, 2298. [Google Scholar] [CrossRef]
  51. Amani, S.; Mohebodini, M.; Khademvatan, S.; Jafari, M. Agrobacterium rhizogenes mediated transformation of Ficus carica L. for the efficient production of secondary metabolites. J. Sci. Food Agric. 2020, 100, 2185–2197. [Google Scholar] [CrossRef]
  52. Amani, S.; Mohebodini, M.; Khademvatan, S.; Jafari, M.; Kumar, V. Piriformospora indica based elicitation for overproduction of phenolic compounds by hairy root cultures of Ficus carica. J. Biotechnol. 2021, 327, 43–53. [Google Scholar] [CrossRef]
  53. Ghimire, B.K.; Thiruvengadam, M.; Chung, I.-M. Identification of elicitors enhances the polyphenolic compounds and pharmacological potential in hairy root cultures of Aster scaber. S. Afr. J. Bot. 2019, 125, 92–101. [Google Scholar] [CrossRef]
  54. Ansari, M.A.; Chung, I.-M.; Rajakumar, G.; Alzohairy, M.A.; Almatroudi, A.; Khanna, V.G.; Thiruvengadam, M. Evaluation of polyphenolic compounds and pharmacological activities in hairy root cultures of Ligularia fischeri Turcz. f. spiciformis (Nakai). Molecules 2019, 24, 1586. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Kundu, S.; Salma, U.; Ali, M.N.; Hazra, A.K.; Mandal, N. Development of transgenic hairy roots and augmentation of secondary metabolites by precursor feeding in Sphagneticola calendulacea (L.) Pruski. Ind. Crops Prod. 2018, 121, 206–215. [Google Scholar] [CrossRef]
  56. Sitarek, P.; Kowalczyk, T.; Rijo, P.; Białas, A.J.; Wielanek, M.; Wysokińska, H.; Garcia, C.; Toma, M.; Śliwiński, T.; Skała, E. Over-expression of AtPAP1 transcriptional factor enhances phenolic acid production in transgenic roots of Leonurus sibiricus L. and their biological activities. Mol. Biotechnol. 2018, 60, 74–82. [Google Scholar] [CrossRef] [PubMed]
  57. Yousefian, S.; Lohrasebi, T.; Farhadpour, M.; Haghbeen, K. Effect of methyl jasmonate on phenolic acids accumulation and the expression profile of their biosynthesis-related genes in Mentha spicata hairy root cultures. Plant Cell Tissue Org. Cult. 2020, 142, 285–297. [Google Scholar] [CrossRef]
  58. Kowalczyk, T.; Merecz-Sadowska, A.; Rijo, P.; Isca, V.M.S.; Picot, L.; Wielanek, M.; Śliwiński, T.; Sitarek, P. Preliminary phytochemical analysis and evaluation of the biological activity of Leonotis nepetifolia (L.) R. Br transformed roots extracts obtained through Rhizobium rhizogenes-mediated transformation. Cells 2021, 10, 1242. [Google Scholar] [CrossRef]
  59. Kochan, E.; Szymańska, G.; Wielanek, M.; Wiktorowska-Owczarek, A.; Jóźwiak-Bębenista, M.; Grzegorczyk-Karolak, I. The content of triterpene saponins and phenolic compounds in American ginseng hairy root extracts and their antioxidant and cytotoxic properties. Plant Cell Tissue Organ Cult. 2019, 138, 353–362. [Google Scholar] [CrossRef] [Green Version]
  60. Mandal, S.; Mitra, A. Accumulation of cell wall-bound phenolic metabolites and their upliftment in hairy root cultures of tomato (Lycopersicon esculentum Mill.). Biotechnol. Lett. 2008, 30, 1253–1258. [Google Scholar] [CrossRef]
  61. Kumar, M.; Basu, A.; Kumari, P.; Jha, S.; Mitra, A. Tobacco plantlets ameliorate oxidative stress upon expression of a cryptogein gene. Plant Cell Tissue Organ Cult. 2016, 125, 553–570. [Google Scholar] [CrossRef]
  62. Yousefian, Z.; Golkar, P.; Mirjalili, M.H. Production enhancement of medicinally active coumarin and phenolic compounds in hairy root cultures of Pelargonium sidoides: The effect of elicitation and sucrose. J. Plant Growth Regul. 2021, 40, 628–641. [Google Scholar] [CrossRef]
  63. Alipieva, K.; Korkina, L.; Orhan, I.E.; Georgiev, M.I. Verbascoside—A review of its occurrence, (bio)synthesis and pharmacological significance. Biotechnol. Adv. 2014, 32, 1065–1076. [Google Scholar] [CrossRef] [PubMed]
  64. Wu, L.; Georgiev, M.I.; Cao, H.; Nahar, L.; El-Seedi, H.R.; Sarker, S.D.; Xiao, J.; Lu, B. Therapeutic potential of phenylethanoid glycosides: A systematic review. Med. Res. Rev. 2020, 40, 2605–2649. [Google Scholar] [CrossRef] [PubMed]
  65. Sęczyk, Ł.; Sugier, D.; Dervişoğlu, G.; Özdemir, F.A.; Kołodziej, B. Phytochemical profile, in vitro bioaccessibility, and anticancer potential of golden root (Rhodiola rosea L.) extracts. Food Chem. 2023, 404, 134779. [Google Scholar] [CrossRef]
  66. Jin, M.; Wang, C.; Xu, Y.; Zhang, Z.; Wu, X.; Ye, R.; Zhang, Q.; Han, D. Pharmacological effects of salidroside on central nervous system diseases. Biomed. Pharmacother. 2022, 156, 113746. [Google Scholar] [CrossRef]
  67. Zhou, Y.; Hirotani, M.; Yoshikawa, T.; Furuya, T. Flavonoids and phenylethanoids from hairy root cultures of Scutellaria baicalensis. Phytochemistry 1997, 44, 83–87. [Google Scholar] [CrossRef]
  68. Murashige, T.; Skoog, F. A revised medium for rapid growth and bioassays with tobacco tissue cultures. Physiol. Plant. 1962, 15, 473–497. [Google Scholar] [CrossRef]
  69. Nishikawa, K.; Furukawa, H.; Fujioka, T.; Fujii, H.; Mihashi, K.; Shimomura, K.; Ishimaru, K. Flavone production in transformed root cultures of Scutellaria baicalensis Georgi. Phytochemistry 1999, 52, 885–890. [Google Scholar] [CrossRef]
  70. Wilczańska-Barska, A.; Królicka, A.; Głód, D.; Majdan, M.; Kawiak, A.; Krauze-Baranowska, M. Enhanced accumulation of secondary metabolites in hairy root cultures of Scutellaria lateriflora following elicitation. Biotechnol. Lett. 2012, 34, 1757–1763. [Google Scholar] [CrossRef]
  71. Marsh, Z.; Yang, T.; Nopo-Olazabal, L.; Wu, S.; Ingle, T.; Joshee, N.; Medina-Bolivar, F. Effect of light, methyl jasmonate and cyclodextrin on production of phenolic compounds in hairy root cultures of Scutellaria lateriflora. Phytochemistry 2014, 107, 50–60. [Google Scholar] [CrossRef]
  72. Gamborg, O.L.; Miller, R.A.; Ojima, K. Nutrient requirements of suspension cultures of soybean root cells. Exp. Cell Res. 1968, 50, 151–158. [Google Scholar] [CrossRef]
  73. Wysokińska, H.; Rózga, M. Establishment of transformed root cultures of Paulownia tomentosa for verbascoside production. J. Plant Physiol. 1998, 152, 78–83. [Google Scholar] [CrossRef]
  74. Wysokińska, H.; Lisowska, K.; Floryanowicz-Czekalska, K. Transformation of Catalpa ovata by Agrobacterium rhizogenes and phenylethanoid glycosides production in transformed root cultures. Z. Naturforsch. 2001, 56, 375–381. [Google Scholar] [CrossRef] [PubMed]
  75. Fons, F.; Tousch, D.; Rapior, S.; Gueiffier, A.; Roussel, J.L.; Gargadennec, A.; Andary, C. Phenolic profiles of untransformed and hairy root cultures of Plantago lanceolata. Plant Physiol. Biochem. 1999, 37, 291–296. [Google Scholar] [CrossRef]
  76. Dhakulkar, S.; Ganapathi, T.R.; Bhargava, S.; Bapat, V.A. Induction of hairy roots in Gmelina arborea Roxb. and production of verbascoside in hairy roots. Plant Sci. 2005, 169, 812–818. [Google Scholar] [CrossRef]
  77. Ludwig-Müller, J.; Georgiev, M.; Bley, T. Metabolite and hormonal status of hairy root cultures of Devil’s claw (Harpagophytum procumbens) in flasks and in a bubble column bioreactor. Process. Biochem. 2008, 43, 15–23. [Google Scholar] [CrossRef]
  78. Homova, V.; Weber, J.; Schulze, J.; Alipieva, K.; Bley, T.; Georgiev, M. Devil’s claw hairy root culture in flasks and in a 3-L bioreactor: Bioactive metabolite accumulation and flow cytometry. Z. Naturforsch. 2010, 65c, 472–478. [Google Scholar] [CrossRef]
  79. Grąbkowska, R.; Królicka, A.; Mielicki, W.; Wielanek, M.; Wysokińska, H. Genetic transformation of Harpagophytum procumbens by Agrobacterium rhizogenes: Iridoid and phenylethanoid glycoside ccumulation in hairy root cultures. Acta Physiol. Plant. 2010, 32, 665–673. [Google Scholar] [CrossRef]
  80. Gyurkovska, V.; Alipieva, K.; Maciuk, A.; Dimitrova, P.; Ivanovska, N.; Haas, C.; Bley, T.; Georgiev, M. Anti-inflammatory activity of Devil’s claw in vitro systems and their active constituents. Food Chem. 2011, 125, 171–178. [Google Scholar] [CrossRef]
  81. Georgiev, M.I.; Alipieva, K.; Orhan, I.E. Cholinesterases inhibitory and antioxidant activities of Harpagophytum procumbens from in vitro systems. Phytother. Res. 2012, 26, 313–316. [Google Scholar] [CrossRef]
  82. Piątczak, E.; Kuźma, Ł.; Wysokińska, H. The influence of methyl jasmonate and salicylic acid on secondary metabolite production in Rehmannia glutinosa Libosch. hairy root culture. Acta Biol. Crac. Ser. Bot. 2016, 58, 57–65. [Google Scholar] [CrossRef] [Green Version]
  83. Wang, F.; Zhi, J.; Zhang, Z.; Wang, L.; Suo, Y.; Xie, C.; Li, M.; Zhang, B.; Du, J.; Gu, L.; et al. Transcriptome analysis of salicylic acid treatment in Rehmannia glutinosa hairy roots using RNA-seq technique for identification of genes involved in acteoside biosynthesis. Front. Plant Sci. 2017, 8, 787. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Piątczak, E.; Jeleń, A.; Makówczyńska, J.; Zielińska, S.; Kuźma, Ł.; Balcerczak, E. Establishment of hairy root cultures of Rehmannia elata N.E. Brown ex Prain and production of iridoid and phenylethanoid glycosides. Ind. Crops Prod. 2019, 137, 308–314. [Google Scholar] [CrossRef]
  85. Alipieva, K.I.; Orhan, I.E.; Cankaya, I.I.T.; Kostadinova, E.P.; Georgiev, M.I. Treasure from garden: Chemical profiling, pharmacology and biotechnology of mulleins. Phytochem. Rev. 2014, 13, 417–444. [Google Scholar] [CrossRef]
  86. Georgiev, M.I.; Ludwig-Müller, J.; Alipieva, K.; Lippert, A. Sonication-assisted Agrobacterium rhizogenes-mediated transformation of Verbascum xanthophoeniceum Griseb. for bioactive metabolite accumulation. Plant Cell Rep. 2011, 30, 859–866. [Google Scholar] [CrossRef]
  87. Georgiev, M.I.; Radziszewska, A.; Neumann, M.; Marchev, A.; Alipieva, K.; Ludwig-Müller, J. Metabolic alterations of Verbascum nigrum L. plants and SAArT transformed roots as revealed by NMR-based metabolomics. Plant Cell Tissue Organ Cult. 2015, 123, 349–356. [Google Scholar] [CrossRef]
  88. Marchev, A.; Yordanova, Z.; Alipieva, K.; Zahmanov, G.; Rusinova-Videva, S.; Kapchina-Toteva, V.; Simova, S.; Popova, M.; Georgiev, M.I. Genetic transformation of rare Verbascum eriophorum Godr. plants and metabolic alterations revealed by NMR-based metabolomics. Biotechnol. Lett. 2016, 38, 1621–1629. [Google Scholar] [CrossRef]
  89. Grech-Baran, M.; Sykłowska-Baranek, K.; Pietrosiuk, A. Biotechnological approaches to enhance salidroside, rosin and its derivatives production in selected Rhodiola spp. in vitro cultures. Phytochem. Rev. 2015, 14, 657–674. [Google Scholar] [CrossRef] [Green Version]
  90. Liu, Y.; Wang, J.; Wang, L.; Zhang, C.; Bai, Q.; Lyu, X.; Yang, R. Biosynthesis and biotechnological production of salidroside from Rhodiola genus plants. Phytochem. Rev. 2022, 21, 1605–1626. [Google Scholar] [CrossRef]
  91. Lütken, H.; Meropi-Antypa, N.; Kemp, O.; Hegelund, J.N.; Müller, R. Hairy Root Cultures of Rhodiola rosea to Increase Valuable Bioactive Compounds. In Production of Plant Derived Natural Compounds through Hairy Root Culture, 1st ed.; Malik, S., Ed.; Springer International Publishing AG: Cham, Switzerland, 2017; pp. 65–88. [Google Scholar] [CrossRef]
  92. Chen, Y.; Tang, M.; Yuan, S.; Fu, S.; Li, Y.; Li, Y.; Wang, Q.; Cao, Y.; Liu, L.; Zhang, Q. Rhodiola rosea: A therapeutic candidate on cardiovascular diseases. Oxidative Med. Cell. Longev. 2022, 2022, 1348795. [Google Scholar] [CrossRef]
  93. Zhou, X.; Wu, Y.; Wang, X.; Liu, B.; Xu, H. Salidroside production by hairy roots of Rhodiola sachalinensis obtained after transformation with Agrobacterium rhizogenes. Biol. Pharm. Bull. 2007, 30, 439–442. [Google Scholar] [CrossRef] [Green Version]
  94. Yu, H.-S.; Ma, L.-Q.; Zhang, J.-X.; Shi, G.-L.; Hua, Y.-H.; Wang, Y.-N. Characterization of glycosyltransferases responsible for salidroside biosynthesis in Rhodiola sachalinensis. Phytochemistry 2011, 72, 862–870. [Google Scholar] [CrossRef]
  95. Lan, X.; Chang, K.; Zeng, L.; Liu, X.; Qiu, F.; Zheng, W.; Quan, H.; Liao, Z.; Chen, M.; Huang, W.; et al. Engineering salidroside biosynthetic pathway in hairy root cultures of Rhodiola crenulata based on metabolic characterization of tyrosine decarboxylase. PLoS ONE 2013, 8, e75459. [Google Scholar] [CrossRef] [PubMed]
  96. Liu, X.; Tang, Y.; Zeng, J.; Qin, J.; Lin, M.; Chen, M.; Liao, Z.; Lan, X. Biochemical characterization of tyrosine aminotransferase and enhancement of salidroside production by suppressing tyrosine aminotransferase in Rhodiola crenulate. Ind. Crops Prod. 2021, 173, 114075. [Google Scholar] [CrossRef]
  97. Grech-Baran, M.; Sykłowska-Baranek, K.; Giebułtowicz, J.; Wroczyński, P.; Pietrosiuk, A. Tyrosol glucosyltransferase activity and salidroside production in natural and transformed root cultures of Rhodiola kirilowii (Regel) Regel et Maximowicz. Acta Biol. Crac. Ser. Bot. 2013, 55, 126–133. [Google Scholar] [CrossRef]
  98. Grech-Baran, M.; Sykłowska-Baranek, K.; Krajewska-Patan, A.; Wyrwał, A.; Pietrosiuk, A. Biotransformation of cinnamyl alcohol to rosavins by non-transformed wild type and hairy root cultures of Rhodiola kirilowii. Biotechnol. Lett. 2014, 36, 649–656. [Google Scholar] [CrossRef] [Green Version]
  99. Stepanova, A.; Malunova, M.; Salamaikina, S.; Selimov, R.; Solov’eva, A. Establishment of Rhodiola quadrifida hairy roots and callus culture to produce bioactive compounds. Phyton 2021, 90, 543–552. [Google Scholar] [CrossRef]
  100. Clifford, M.N. Chlorogenic acids and other cinnamates—Nature, occurrence, dietary burden, absorption and metabolism. J. Sci. Food Agric. 2000, 80, 1033–1043. [Google Scholar] [CrossRef]
  101. Fu, R.; Zhang, P.; Jin, G.; Wang, L.; Qi, S.; Cao, Y.; Martin, C.; Zhang, Y. Versatility in acyltransferase activity completes chicoric acid biosynthesis in purple coneflower. Nat. Commun. 2021, 12, 1563. [Google Scholar] [CrossRef]
  102. Liu, Q.; Liu, F.; Zhang, L.; Niu, Y.; Liu, Z.; Liu, X. Comparison of chicoric acid, and its metabolites caffeic acid and caftaric acid: In vitro protection of biological macromolecules and inflammatory responses in BV2 microglial cells. Food Sci. Hum. Wellness 2017, 6, 155–166. [Google Scholar] [CrossRef]
  103. Tanyeli, A.; Akdemir, F.N.E.; Eraslan, E.; Güler, M.C.; Nacar, T. Anti-oxidant and anti-inflamatuar effectiveness of caftaric acid on gastric ulcer induced by indomethacin in rats. Gen. Physiol. Biophys. 2019, 38, 175–181. [Google Scholar] [CrossRef]
  104. Peng, Y.; Sun, Q.; Park, Y. The bioactive effects of chicoric acid as a functional food ingredient. J. Med. Food 2019, 22, 645–652. [Google Scholar] [CrossRef] [PubMed]
  105. Abd El-Twab, S.M.; Hussein, O.E.; Hozayen, W.G.; Bin-Jumah, M.; Mahmoud, A.M. Chicoric acid prevents methotrexate-induced kidney injury by suppressing NF-κB/NLRP3 inflammasome activation and up-regulating Nrf2/ARE/HO-1 signaling. Inflamm. Res. 2019, 68, 511–523. [Google Scholar] [CrossRef] [PubMed]
  106. Lu, Q.-B.; Wan, M.-Y.; Wang, P.-Y.; Zhang, C.-X.; Xu, D.-Y.; Liao, X.; Sun, H.-J. Chicoric acid prevents PDGF-BB-induced VSMC dedifferentiation, proliferation and migration by suppressing ROS/ NF-κB/mTOR/P70S6K signaling cascade. Redox Biol. 2018, 14, 656–668. [Google Scholar] [CrossRef] [PubMed]
  107. Qu, Y.; Shen, Y.; Teng, L.; Huang, Y.; Yang, Y.; Jian, X.; Fan, S.; Wu, P.; Fu, Q. Chicoric acid attenuates tumor necrosis factor-α-induced inflammation and apoptosis via the Nrf2/HO-1, PI3K/AKT and NF-κB signaling pathways in C28/I2 cells and ameliorates the progression of osteoarthritis in a rat model. Int. Immunopharmacol. 2022, 111, 109129. [Google Scholar] [CrossRef]
  108. Wang, Y.; Diao, Z.; Li, J.; Ren, B.; Zhu, D.; Liu, Q.; Liu, Z.; Liu, X. Chicoric acid supplementation ameliorates cognitive impairment induced by oxidative stress via promotion of antioxidant defense system. RSC Adv. 2017, 7, 36149–36162. [Google Scholar] [CrossRef] [Green Version]
  109. Liu, Q.; Chen, Y.; Shen, C.; Xiao, Y.; Wang, Y.; Liu, Z.; Liu, X. Chicoric acid supplementation prevents systemic inflammation-induced memory impairment and amyloidogenesis via inhibition of NF-kB. FASEB J. 2017, 31, 1494–1507. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Liu, Q.; Fang, J.; Chen, P.P.; Die, Y.; Wang, J.; Liu, Z.; Liu, X. Chicoric acid improves neuron survival against inflammation by promoting mitochondrial function and energy metabolism. Food Funct. 2019, 10, 6157–6169. [Google Scholar] [CrossRef]
  111. Abbasi, B.H.; Saxena, P.K.; Murch, S.J.; Liu, C.-Z. Echinacea biotechnology: Challenges and opportunities. In Vitro Cell. Dev. Biol. Plant 2007, 43, 481–492. [Google Scholar] [CrossRef]
  112. Parsons, J.L.; Cameron, S.I.; Harris, C.S.; Smith, M.L. Echinacea biotechnology: Advances, commercialization and future considerations. Pharm. Biol. 2018, 56, 485–494. [Google Scholar] [CrossRef] [Green Version]
  113. Murthy, H.N.; Kim, Y.-S.; Park, S.-Y.; Paek, K.-Y. Biotechnological production of caffeic acid derivatives from cell and organ cultures of Echinacea species. Appl. Microbiol. Biotechnol. 2014, 98, 7707–7717. [Google Scholar] [CrossRef]
  114. Evidente, A.; Masi, M. Natural bioactive cinnamoyltyramine alkylamides and co-metabolites. Biomolecules 2021, 11, 1765. [Google Scholar] [CrossRef] [PubMed]
  115. Trypsteen, M.; Van Lijsebettens, M.; Van Severen, R.; Van Montagu, M. Agrobacterium rhizogenes-mediated transformation of Echinacea purpurea. Plant Cell Rep. 1991, 10, 85–89. [Google Scholar] [CrossRef] [PubMed]
  116. Liu, C.-Z.; Abbasi, B.H.; Gao, M.; Murch, S.J.; Saxena, P.K. Caffeic Acid Derivatives Production by Hairy Root Cultures of Echinacea purpurea. J. Agric. Food. Chem. 2006, 54, 8456–8460. [Google Scholar] [CrossRef] [PubMed]
  117. Abbasi, B.H.; Tian, C.-L.; Murch, S.J.; Saxena, P.K.; Liu, C.-Z. Light-enhanced caffeic acid derivatives biosynthesis in hairy root cultures of Echinacea purpurea. Plant Cell Rep. 2007, 26, 1367–1372. [Google Scholar] [CrossRef]
  118. Abbasi, B.H.; Stiles, A.R.; Saxena, P.K.; Liu, C.-Z. Gibberellic acid increases secondary metabolite production in Echinacea purpurea hairy roots. Appl. Biochem. Biotechnol. 2012, 168, 2057–2066. [Google Scholar] [CrossRef]
  119. Abbasi, B.H.; Liu, R.; Saxena, P.K.; Liu, C.-Z. Cichoric acid production from hairy root cultures of Echinacea purpurea grown in amodified airlift bioreactor. J. Chem. Technol. Biotechnol. 2009, 84, 1697–1701. [Google Scholar] [CrossRef]
  120. Liu, R.; Sun, W.; Liu, C.-Z. Computational fluid dynamics modeling of mass-transfer behavior in a bioreactor for hairy root culture. II. Analysis of ultrasound-intensified process. Biotechnol. Prog. 2011, 27, 1672–1678. [Google Scholar] [CrossRef]
  121. Liu, R.; Li, W.; Sun, L.-Y.; Liu, C.-Z. Improving root growth and cichoric acid derivatives production in hairy root culture of Echinacea purpurea by ultrasound treatment. Biochem. Eng. J. 2012, 60, 62–66. [Google Scholar] [CrossRef]
  122. Abdoli, M.; Moieni, A.; Naghdi Badi, H. Influence of KNO3, CaCl2 and MgSO4 concentrations on growth and cichoric acid accumulation in hairy root culture of purple coneflower (Echinacea purpurea L.). J. Med. Plants 2013, 12, 75–84. Available online: http://jmp.ir/article-1-111-en.html (accessed on 31 March 2023).
  123. Demirci, T.; Akçay, U.C.; Baydar, N.G. Effects of 24-epibrassinolide and l-phenylalanine on growth and caffeic acid derivative production in hairy root culture of Echinacea purpurea L. Moench. Acta Physiol. Plant. 2020, 42, 66. [Google Scholar] [CrossRef]
  124. Baque, M.A.; Moh, S.-H.; Lee, E.-J.; Zhong, J.-J.; Paek, K.-Y. Production of biomass and useful compounds from adventitious roots of high-value added medicinal plants using bioreactor. Biotechnol. Adv. 2012, 30, 1255–1267. [Google Scholar] [CrossRef] [PubMed]
  125. Stojakowska, A.; Malarz, J.; Szewczyk, A.; Kisiel, W. Caffeic acid derivatives from a hairy root culture of Lactuca virosa. Acta Physiol. Plant. 2012, 34, 291–298. [Google Scholar] [CrossRef] [Green Version]
  126. Yi, T.G.; Park, Y.; Park, J.-E.; Park, N.I. Enhancement of phenolic compounds and antioxidative activities by the combination of culture medium and methyl jasmonate elicitation in hairy root cultures of Lactuca indica L. Nat. Prod. Commun. 2019, 14, 1934578X19861867. [Google Scholar] [CrossRef] [Green Version]
  127. Abrankó, L.; Clifford, M.N. An unambiguous nomenclature for the acyl-quinic acids commonly known as chlorogenic acids. J. Agric. Food Chem. 2017, 65, 3602–3608. [Google Scholar] [CrossRef] [PubMed]
  128. Magaña, A.A.; Kamimura, N.; Soumyanath, A.; Stevens, J.F.; Maier, C.S. Caffeoylquinic acids: Chemistry, biosynthesis, occurrence, analytical challenges, and bioactivity. Plant J. 2021, 107, 1299–1319. [Google Scholar] [CrossRef]
  129. Ciaramelli, C.; Palmioli, A.; De Luigi, A.; Colombo, L.; Sala, G.; Riva, C.; Zoiac, C.P.; Salmona, M.; Airoldi, C. NMR-driven identification of anti-amyloidogenic compounds in green and roasted coffee extracts. Food Chem. 2018, 252, 171–180. [Google Scholar] [CrossRef]
  130. Pérez-Jiménez, J.; Fezeu, L.; Touvier, M.; Arnault, N.; Manach, C.; Hercberg, S.; Galan, P.; Scalbert, A. Dietary intake of 337 polyphenols in French adults. Am. J. Clin. Nutr. 2011, 93, 1220–1228. [Google Scholar] [CrossRef] [Green Version]
  131. Di Lorenzo, C.; Colombo, F.; Biella, S.; Stockley, C.; Restani, P. Polyphenols and human health: The role of bioavailability. Nutrients 2021, 13, 273. [Google Scholar] [CrossRef]
  132. Domínguez-Fernández, M.; Young, P.; Yang, T.; Ludwig, I.A.; Clifford, M.N.; Cid, C.; Rodriguez-Mateos, A. In vivo study of the bioavailability and metabolic profile of (poly)phenols after sous-vide artichoke consumption. Food Chem. 2022, 367, 130620. [Google Scholar] [CrossRef]
  133. Liang, N.; Kitts, D.D. Role of chlorogenic acids in controlling oxidative and inflammatory stress conditions. Nutrients 2016, 8, 16. [Google Scholar] [CrossRef] [Green Version]
  134. Shin, H.S.; Satsu, H.; Bae, M.-J.; Zhao, Z.; Ogiwara, H.; Totsuka, M.; Shimizu, M. Anti-inflammatory effect of chlorogenic acid on the IL-8 production in Caco-2 cells and the dextran sulphate sodium-induced colitis symptoms in C57BL/6 mice. Food Chem. 2015, 168, 167–175. [Google Scholar] [CrossRef] [PubMed]
  135. Han, J.; Miyamae, Y.; Shigemori, H.; Isoda, H. Neuroprotective effect of 3,5-di-O-caffeoylquinic acid on SH-SY5Y cells and senescence-accelerated-prone mice 8 through the up-regulation of phosphoglycerate kinase-1. Neuroscience 2010, 169, 1039–1045. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Kwon, S.-H.; Lee, H.-K.; Kim, J.-A.; Hong, S.-I.; Kim, H.-C.; Jo, T.-H.; Park, Y.-I.; Lee, C.-K.; Kim, Y.-B.; Lee, S.-Y.; et al. Neuroprotective effects of chlorogenic acid on scopolamine-induced amnesia via anti-acetylcholinesterase and anti-oxidative activities in mice. Eur. J. Pharmacol. 2010, 649, 210–217. [Google Scholar] [CrossRef]
  137. Jang, H.; Ahn, H.R.; Jo, H.; Kim, K.-A.; Lee, E.H.; Lee, K.W.; Jung, S.H.; Lee, C.Y. Chlorogenic Acid and Coffee Prevent Hypoxia-Induced Retinal Degeneration. J. Agric. Food Chem. 2014, 62, 182–191. [Google Scholar] [CrossRef] [PubMed]
  138. Kim, J.; Jeong, I.-H.; Kim, C.-S.; Lee, Y.M.; Kim, J.M.; Kim, J.S. Chlorogenic acid inhibits the formation of advanced glycation end products and associated protein cross-linking. Arch. Pharm. Res. 2011, 34, 495–500. [Google Scholar] [CrossRef]
  139. Skała, E.; Makowczyńska, J.; Wieczfinska, J.; Kowalczyk, T.; Sitarek, P. Caffeoylquinic acids with potential biological activity from plant in vitro cultures as alternative sources of valuable natural products. Curr. Pharm. Des. 2020, 26, 2817–2842. [Google Scholar] [CrossRef]
  140. Kodoma, M.; Wada, H.; Otani, H.; Kohmoto, K.; Kimura, Y. 3,5-Di-O-caffeoylquinic acid, an infection-inhibiting factor from Pyrus pyrifolia induced by infection with Alternaria alternata. Phytochemistry 1998, 47, 371–373. [Google Scholar] [CrossRef]
  141. Leiss, K.A.; Maltese, F.; Choi, Y.H.; Verpoorte, R.; Klinkhamer, P.G.L. Identification of chlorogenic acid as a resistance factor for thrips in Chrysanthemum. Plant Physiol. 2009, 150, 1567–1575. [Google Scholar] [CrossRef] [Green Version]
  142. Yadav, R.; Mehrotra, M.; Singh, A.K.; Niranjan, A.; Singh, R.; Sanyal, I.; Lehri, A.; Pande, V.; Amla, D.V. Improvement in Agrobacterium-mediated transformation of chickpea (Cicer arietinum L.) by the inhibition of polyphenolics released during wounding of cotyledonary node explants. Protoplasma 2017, 254, 253–269. [Google Scholar] [CrossRef]
  143. Hook, I. Secondary metabolites in hairy root cultures of Leontopodium alpinum Cass. (Edelweiss). Plant Cell Tissue Org. Cult. 1994, 38, 321–326. [Google Scholar] [CrossRef]
  144. El-Esawi, M.A.; Elkelish, A.; Elansary, H.O.; Ali, H.M.; Elshikh, M.; Witczak, J.; Ahmad, M. Genetic Transformation and Hairy Root Induction Enhance the Antioxidant Potential of Lactuca serriola L. Oxidative Med. Cell. Longev. 2017, 2017, 5604746. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Malarz, J.; Stojakowska, A.; Kisiel, W. Long-term cultured hairy roots of chicory—A rich source of hydroxycinnamates and 8-deoxylactucin glucoside. Appl. Biochem. Biotechnol. 2013, 171, 1589–1601. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Bernard, G.; Alves Dos Santos, H.; Etienne, A.; Samaillie, J.; Neut, C.; Sahpaz, S.; Hilbert, J.-L.; Gagneul, D.; Jullian, N.; Tahrioui, A.; et al. MeJA elicitation of chicory hairy roots promotes efficient increase of 3,5-diCQA accumulation, a potent antioxidant and antibacterial molecule. Antibiotics 2020, 9, 659. [Google Scholar] [CrossRef] [PubMed]
  147. Maciel, G.; Lopes, A.A.; Cantrell, C.L.; de Castro França, S.; Bertoni, B.W.; Lourenço, M.V. Jasmonates promote enhanced production of bioactive caffeoylquinic acid derivative in Eclipta prostrata (L.) L. hairy roots. Plant Cell Tissue Org. Cult. 2022, 149, 363–369. [Google Scholar] [CrossRef] [PubMed]
  148. Lopes, A.A.; Souza, G.R.S.; de Castro França, S.; Lourenço, M.V. Biosynthetic studies through feeding experiments in Eclipta prostrata (L.) L. hairy roots. Plant Cell Tissue Org. Cult. 2022, 151, 215–219. [Google Scholar] [CrossRef]
  149. Fu, X.; Yin, Z.-P.; Chen, J.-G.; Shangguan, X.-C.; Wang, X.; Zhang, Q.-F.; Peng, D.-Y. Production of chlorogenic acid and its derivatives in hairy root cultures of Stevia rebaudiana. J. Agric. Food Chem. 2015, 63, 262–268. [Google Scholar] [CrossRef] [PubMed]
  150. Skała, E.; Kicel, A.; Olszewska, M.A.; Kiss, A.K.; Wysokińska, H. Establishment of hairy root cultures of Rhaponticum carthamoides (Willd.) Iljin for the production of biomass and caffeic acid derivatives. BioMed Res. Int. 2015, 2015, 181098. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  151. Skała, E.; Sitarek, P.; Toma, M.; Szemraj, J.; Radek, M.; Nieborowska-Skorska, M.; Skorski, T.; Wysokińska, H.; Śliwiński, T. Inhibition of human glioma cell proliferation by altered Bax/Bcl-2-p53 expression and apoptosis induction by Rhaponticum carthamoides extracts from transformed and normal roots. J. Pharm. Pharmacol. 2016, 68, 1454–1464. [Google Scholar] [CrossRef]
  152. Śliwińska, A.; Figat, R.; Zgadzaj, A.; Wileńska, B.; Misicka, A.; Nałęcz-Jawecki, G.; Pietrosiuk, A.; Sykłowska-Baranek, K. Polyscias filicifolia (Araliaceae) hairy roots with antigenotoxic and anti-photogenotoxic activity. Molecules 2022, 27, 186. [Google Scholar] [CrossRef]
  153. Tusevski, O.; Vinterhalter, B.; Krstić Milošević, D.; Soković, M.; Ćirić, A.; Vinterhalter, D.; Zdravković Korać, S.; Petreska Stanoeva, J.; Stefova, M.; Gadzovska Simic, S. Production of phenolic compounds, antioxidant and antimicrobial activities in hairy root and shoot cultures of Hypericum perforatum L. Plant Cell Tissue Organ Cult. 2017, 128, 589–605. [Google Scholar] [CrossRef]
  154. Petersen, M.; Simmonds, M.S.J. Rosmarinic acid. Phytochemistry 2003, 62, 121–125. [Google Scholar] [CrossRef]
  155. Petersen, M.; Abdullah, Y.; Benner, J.; Eberle, D.; Gehlen, K.; Hücherig, S.; Janiak, V.; Kim, K.H.; Sander, M.; Weitzel, C.; et al. Evolution of rosmarinic acid biosynthesis. Phytochemistry 2009, 70, 1663–1679. [Google Scholar] [CrossRef] [PubMed]
  156. Ho, J.H.-C.; Hong, C.-Y. Salvianolic acids: Small compounds with multiple mechanisms for cardiovascular protection. J. Biomed. Sci. 2011, 18, 30. Available online: http://www.jbiomedsci.com/content/18/1/30 (accessed on 31 March 2023). [CrossRef] [PubMed] [Green Version]
  157. Ma, L.; Tang, L.; Yi, Q. Salvianolic Acids: Potential Source of Natural Drugs for the Treatment of Fibrosis Disease and Cancer. Front. Pharmacol. 2019, 10, 97. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Swamy, M.K.; Sinniah, U.R.; Ghasemzadeh, A. Anticancer potential of rosmarinic acid and its improved production through biotechnological interventions and functional genomics. Appl. Microbiol. Biotechnol. 2018, 102, 7775–7793. [Google Scholar] [CrossRef]
  159. Pandey, V.; Swami, R.K.; Narula, A. Harnessing the potential of roots of traditional power plant: Ocimum. Front. Plant. Sci. 2021, 12, 765024. [Google Scholar] [CrossRef]
  160. Tada, H.; Murakami, Y.; Omoto, T.; Shimomura, K.; Ishimaru, K. Rosmarinic acid and related phenolics in hairy root cultures of Ocimum basilicum. Phytochemistry 1996, 42, 431–434. [Google Scholar] [CrossRef]
  161. Bais, H.P.; Walker, T.S.; Schweizer, H.P.; Vivanco, J.M. Root specific elicitation and antimicrobial activity of rosmarinic acid in hairy root cultures of Ocimum basilicum. Plant Physiol. Biochem. 2002, 40, 983–995. [Google Scholar] [CrossRef]
  162. Srivastava, S.; Conlan, X.A.; Adholeya, A.; Cahill, D.M. Elite hairy roots of Ocimum basilicum as a new source of rosmarinic acid and antioxidants. Plant Cell Tissue Organ Cult. 2016, 126, 19–32. [Google Scholar] [CrossRef]
  163. Srivastava, S.; Conlan, X.A.; Cahill, D.M.; Adholeya, A. Rhizophagus irregularis as an elicitor of rosmarinic acid and antioxidant production by transformed roots of Ocimum basilicum in an in vitro co-culture system. Mycorrhiza 2016, 26, 919–930. [Google Scholar] [CrossRef]
  164. Kwon, D.Y.; Kim, Y.B.; Kim, J.K.; Park, S.U. Production of rosmarinic acid and correlated gene expression in hairy root cultures of green and purple basil (Ocimum basilicum L.). Prep. Biochem. Biotechnol. 2021, 51, 35–43. [Google Scholar] [CrossRef] [PubMed]
  165. Kochan, E.; Wysokińska, H.; Chmiel, A.; Grabias, B. Rosmarinic acid and other phenolic acids in hairy roots of Hyssopus officinalis. Z. Naturforsch. 1999, 54, 11–16. [Google Scholar] [CrossRef]
  166. Fattahi, M.; Nazeri, V.; Torras-Claveria, L.; Sefidkon, F.; Cusido, R.M.; Zamani, Z.; Palazon, J. A new biotechnological source of rosmarinic acid and surface flavonoids: Hairy root cultures of Dracocephalum kotschyi Boiss. Ind. Crops Prod. 2013, 50, 256–263. [Google Scholar] [CrossRef]
  167. Weremczuk-Jeżyna, I.; Skała, E.; Olszewska, M.A.; Kiss, A.K.; Balcerczak, E.; Wysokińska, H.; Kicel, A. The identification and quantitative determination of rosmarinic acid and salvianolic acid B in hairy root cultures of Dracocephalum forrestii W.W. Smith. Ind. Crops Prod. 2016, 91, 125–131. [Google Scholar] [CrossRef]
  168. Nourozi, E.; Hosseini, B.; Maleki, R.; Mandoulakani, B.A. Iron oxide nanoparticles: A novel elicitor to enhance anticancer flavonoid production and gene expression in Dracocephalum kotschyi hairy-root cultures. J. Sci. Food Agric. 2019, 99, 6418–6430. [Google Scholar] [CrossRef]
  169. Fraga, B.M.; González-Coloma, A.; Alegre-Gómez, S.; López-Rodríguez, M.; Amador, L.J.; Díaz, C.E. Bioactive constituents from transformed root cultures of Nepeta teydea. Phytochemistry 2017, 133, 59–68. [Google Scholar] [CrossRef]
  170. Lee, S.Y.; Lee, C.Y.; Eom, S.H.; Kim, Y.K.; Park, N.I.; Park, S.U. Rosmarinic acid production from transformed root cultures of Nepeta cataria L. Sci. Res. Essays 2010, 5, 1122–1126. Available online: https://academicjournals.org/journal/SRE/article-full-text-pdf/371F3FF19194 (accessed on 31 March 2023).
  171. Yang, Y.K.; Lee, S.Y.; Park, W.T.; Park, N.I.; Park, S.U. Exogenous auxins and polyamines enhance growth and rosmarinic acid production in hairy root cultures of Nepeta cataria L. Plant Omics 2010, 3, 190–193. Available online: https://www.pomics.com/park_3_6_2010_190_193.pdf (accessed on 31 March 2023).
  172. Petersen, M.; Szabo, E.; Meinhard, J.; Karwatzki, B.; Gertlowski, C.; Kempin, B.; FuB, E. Biosynthesis and accumulation of rosmarinic acid in suspension cultures of Coleus blumei. Plant Cell Tissue Org. Cult. 1995, 43, 89–92. [Google Scholar] [CrossRef]
  173. Li, W.; Koike, K.; Asada, Y.; Yoshikawa, T.; Nikaido, T. Rosmarinic acid production by Coleus forskohlii hairy root cultures. Plant Cell Tissue Organ Cult. 2005, 80, 151–155. [Google Scholar] [CrossRef]
  174. Bauer, N.; Kiseljak, D.; Jelaska, S. The effect of yeast extract and methyl jasmonate on rosmarinic acid accumulation in Coleus blumei hairy roots. Biol. Plant 2009, 53, 650–656. [Google Scholar] [CrossRef]
  175. Vuković, R.; Bauer, N.; Ćurkovic-Perica, M. Genetic elicitation by inducible expression of β-cryptogein stimulates secretion of phenolics from Coleus blumei hairy roots. Plant Sci. 2013, 199–200, 18–28. [Google Scholar] [CrossRef]
  176. Hücherig, S.; Petersen, M. RNAi suppression and overexpression studies of hydroxyphenylpyruvate reductase (HPPR) and rosmarinic acid synthase (RAS) genes related to rosmarinic acid biosynthesis in hairy root cultures of Coleus blumei. Plant Cell Tissue Organ Cult. 2013, 113, 375–385. [Google Scholar] [CrossRef]
  177. Ru, M.; An, Y.; Wang, K.; Peng, L.; Li, B.; Bai, Z.; Wang, B.; Liang, Z. Prunella vulgaris L. hairy roots: Culture, growth, and elicitation by ethephon and salicylic acid. Eng. Life Sci. 2016, 16, 494–502. [Google Scholar] [CrossRef] [Green Version]
  178. Ru, M.; Wang, K.; Bai, Z.; Peng, L.; He, S.; Wang, Y.; Liang, Z. A tyrosine aminotransferase involved in rosmarinic acid biosynthesis in Prunella vulgaris L. Sci. Rep. 2017, 7, 4892. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  179. Ru, M.; Li, Y.; Guo, M.; Chen, L.; Tan, Y.; Peng, L.; Liang, Z. Increase in rosmarinic acid accumulation and transcriptional responses of synthetic genes in hairy root cultures of Prunella vulgaris induced by methyl jasmonate. Plant Cell Tissue Organ Cult. 2022, 149, 371–379. [Google Scholar] [CrossRef]
  180. Lee, S.Y.; Xu, H.; Kim, Y.K.; Park, S.U. Rosmarinic acid production in hairy root cultures of Agastache rugosa Kuntze. World J. Microbiol. Biotechnol. 2008, 24, 969–972. [Google Scholar] [CrossRef]
  181. Vergara-Martínez, V.M.; Estrada-Soto, S.E.; Valencia-Díaz, S.; Garcia-Sosa, K.; Peña-Rodríguez, L.M.; Arellano-García, J.J.; Perea-Arango, I. Methyl jasmonate enhances ursolic, oleanolic and rosmarinic acid production and sucrose induced biomass accumulation, in hairy roots of Lepechinia caulescens. PeerJ 2021, 9, e11279. [Google Scholar] [CrossRef]
  182. Naliwajski, M.R.; Wileńska, B.; Misicka, A.; Pietrosiuk, A.; Sykłowska-Baranek, K. HPLC-PDA-ESI-HRMS-based profiling of secondary metabolites of Rindera graeca anatomical and hairy roots treated with drought and cold stress. Cells 2022, 11, 931. [Google Scholar] [CrossRef]
  183. Grzegorczyk, I.; Królicka, A.; Wysokińska, H. Establishment of Salvia officinalis L. hairy root cultures for the production of rosmarinic acid. Z. Naturforsch. 2006, 61, 351–356. [Google Scholar] [CrossRef]
  184. Grzegorczyk, I.; Matkowski, A.; Wysokińska, H. Antioxidant activity of extracts from in vitro cultures of Salvia officinalis L. Food Chem. 2007, 104, 536–541. [Google Scholar] [CrossRef]
  185. Grzegorczyk, I.; Wysokińska, H. Antioxidant compounds in Salvia officinalis L. shoot and hairy root cultures in the nutrient sprinkle bioreactor. Acta Soc. Bot. Pol. 2010, 79, 7–10. [Google Scholar] [CrossRef]
  186. Khoshsokhan, F.; Babalar, M.; Salami, S.A.; Sheikhakbari-Mehr, R.; Mirjalili, M.H. Rosmarinic acid production in hairy root cultures of Salvia nemorosa L. (Lamiaceae). Biocatal. Agric. Biotechnol. 2022, 45, 102494. [Google Scholar] [CrossRef]
  187. Dowom, S.A.; Abrishamchi, P.; Radjabian, T.; Salami, S.A. Elicitor-induced phenolic acids accumulation in Salvia virgata Jacq. hairy root cultures. Plant Cell Tissue Organ Cult. 2022, 148, 107–117. [Google Scholar] [CrossRef]
  188. Li, J.; Li, B.; Luo, L.; Cao, F.; Yang, B.; Gao, J.; Yan, Y.; Zhang, G.; Peng, L.; Hu, B. Increased phenolic acid and tanshinone production and transcriptional responses of biosynthetic genes in hairy root cultures of Salvia przewalskii Maxim. treated with methyl jasmonate and salicylic acid. Mol. Biol. Rep. 2020, 47, 8565–8578. [Google Scholar] [CrossRef]
  189. Grzegorczyk-Karolak, I.; Kuźma, Ł.; Skała, E.; Kiss, A.K. Hairy root cultures of Salvia viridis L. for production of polyphenolic compounds. Ind. Crops Prod. 2018, 117, 235–244. [Google Scholar] [CrossRef]
  190. Grzegorczyk-Karolak, I. Optimization of culture conditions and cultivation phase for the growth of Salvia viridis transformed roots and polyphenolic compound production. Plant Cell Tissue Organ Cult. 2020, 142, 571–581. [Google Scholar] [CrossRef]
  191. Wojciechowska, M.; Owczarek, A.; Kiss, A.K.; Grąbkowska, R.; Olszewska, M.A.; Grzegorczyk-Karolak, I. Establishment of hairy root cultures of Salvia bulleyana Diels for production of polyphenolic compounds. J. Biotechnol. 2020, 318, 10–19. [Google Scholar] [CrossRef]
  192. Krzemińska, M.; Owczarek, A.; Gonciarz, W.; Chmiela, M.; Olszewska, M.A.; Grzegorczyk-Karolak, I. The antioxidant, cytotoxic and antimicrobial potential of phenolic acids-enriched extract of elicited hairy roots of Salvia bulleyana. Molecules 2022, 27, 992. [Google Scholar] [CrossRef]
  193. Krzemińska, M.; Owczarek, A.; Olszewska, M.A.; Grzegorczyk-Karolak, I. In vitro strategy for the enhancement of the production of bioactive polyphenols in transformed roots of Salvia bulleyana. Int. J. Mol. Sci. 2022, 23, 7771. [Google Scholar] [CrossRef]
Figure 1. Chemical structures of selected simple phenolics and phenolic acids.
Figure 1. Chemical structures of selected simple phenolics and phenolic acids.
Ijms 24 06920 g001
Figure 2. Chemical structures of selected phenylethanoids.
Figure 2. Chemical structures of selected phenylethanoids.
Ijms 24 06920 g002
Figure 3. Chemical structures of caftaric acid, chicoric acid, and echinacoside.
Figure 3. Chemical structures of caftaric acid, chicoric acid, and echinacoside.
Ijms 24 06920 g003
Figure 4. Chemical structures of selected acylquinic acids (CGAs).
Figure 4. Chemical structures of selected acylquinic acids (CGAs).
Ijms 24 06920 g004
Figure 5. Chemical structures of rosmarinic acid and selected salvianolic acids.
Figure 5. Chemical structures of rosmarinic acid and selected salvianolic acids.
Ijms 24 06920 g005
Table 1. Hairy root cultures that produced high yields of simple phenolics and phenolic acids.
Table 1. Hairy root cultures that produced high yields of simple phenolics and phenolic acids.
Plant SpeciesHairy RootsNatural ProductMaximum Content in the BiomassLiterature
Datura stramonium L.Expressing pmHCHLp-Hydroxybenzoic acid O-β-D-glucoside
p-Hydroxybenzyl alcohol O-β-D-glucoside
4.4 mg/g FW
2.8 mg/g FW
[32]
Beta vulgaris L.Expressing pmHCHLp-Hydroxybenzoic acid (4-Hydroxybenzoic acid) glucose ester140 mg/g DW[33]
Momordica charantia L.Wild typeGentisic acid
Salicylic acid
5.5 mg/g DW
2.5 mg/g DW
[45]
Polygonum multiflorum Thunb.Wild typePyrogallol1.4 mg/g DW[48]
Ficus carica L. cv. SabzWild typeGallic acid
Coumaric acid
7.5 mg/g DW
0.9 mg/g DW
[52]
Leonurus sibirica L.Expressing AtPAP1Chlorogenic acid
Caffeic acid
19.4 mg/g DW
11.4 mg/g DW
[56]
Leonotis nepetifolia (L.) R. Br.Wild typep-Coumaric acid
m-Coumaric acid
4.9 mg/g DW
2.1 mg/g DW
[58]
Table 2. Hairy root cultures that produced high yields of phenylethanoids.
Table 2. Hairy root cultures that produced high yields of phenylethanoids.
Plant SpeciesHairy RootsNatural ProductMaximum Content in the BiomassLiterature
Paulownia tomentosa Steud.Wild typeVerbascoside94.9 mg/g DW[73]
Plantago lanceolata L.Wild typePlantamajoside30–80 mg/g DW[75]
Verbascum xanthophoeniceum Griseb.Wild typeVerbascoside23.3 mg/g DW[86]
Rhodiola sachalinensis
Boriss.
Overexpressing
UGT72B14
Salidroside19.8 mg/g DW[94]
Table 3. Hairy root cultures that produced high yields of caffeoyltartaric acids and echinacoside.
Table 3. Hairy root cultures that produced high yields of caffeoyltartaric acids and echinacoside.
Plant SpeciesHairy RootsNatural ProductMaximum Content in the BiomassLiterature
Echinacea purpurea
(L.) Moench
Wild typeChicoric acid27 mg/g DW[117]
Echinacea purpurea
(L.) Moench
Wild type, supplemented with 0.025 μM GA3Chicoric acid
Caftaric acid
36 mg/g DW
7.5 mg/g DW
[118]
Echinacea purpurea
(L.) Moench
Wild type, supplemented with 1 mg/L
of 24-epibrassinolide
Chicoric acid
Caftaric acid
Echinacoside
24.1 mg/g DW
6.9 mg/g DW
4.3 mg/g DW
[124]
Echinacea purpurea
(L.) Moench
Wild type, supplemented with 500 μM L-phenylalanineChicoric acid
Caftaric acid
Echinacoside
17.4 mg/g DW
6.3 mg/g DW
5.4 mg/g DW
[124]
Table 4. Hairy root cultures that produced high yields of acylquinic acids.
Table 4. Hairy root cultures that produced high yields of acylquinic acids.
Plant SpeciesHairy RootsNatural ProductMaximum Content in the BiomassLiterature
Lactuca virosa L.Wild type3,5-di-O-caffeoylquinic acid25.8 mg/g DW[125]
Cichorium intybus L.Wild type3,5-di-O-caffeoylquinic acid
5-O-caffeoylquinic acid
55.7 mg/g DW
9.4 mg/g DW
[145]
Cichorium intybus L.
var Orchies
Wild type, elicited with 150 μM MeJa3,5-di-O-caffeoylquinic acid120 mg/g DW[146]
Eclipta prostrata (L.) L.Wild type, elicited with 100 μM of jasmonic acid3,5-di-O-caffeoylquinic acid44.7 mg/g DW[147]
Eclipta prostrata (L.) L.Wild type, elicited with 140 μM MeJa3,5-di-O-caffeoylquinic acid41.6 mg/g DW[147]
Stevia rebaudiana Bertoni
var. FengNong 3
Wild type5-O-caffeoylquinic acid
3,5-di-O-caffeoylquinic acid
39.4 mg/g DW
48.1 mg/g DW
[149]
Table 5. Hairy root cultures that produced high yields of polyphenolic antioxidants (RA, lithospermic acid B, and salvianolic acid K).
Table 5. Hairy root cultures that produced high yields of polyphenolic antioxidants (RA, lithospermic acid B, and salvianolic acid K).
Plant SpeciesHairy RootsNatural ProductMaximum Content in the BiomassLiterature
Ocimum basilicum L.Wild typeRosmarinic acid120 mg/g DW[158]
Ocimum basilicum L.Wild type, elicited
with 2% of Phytophthora cinnamoni cell wall extract
Rosmarinic acid81 mg/g FW[159]
Ocimum basilicum L.Wild type,
cocultivated with
Rhizophagus irregularis
Rosmarinic acid140.5 mg/g DW[161]
Agastache rugosa KuntzeWild typeRosmarinic acid116 mg/g DW[178]
Rindera graeca (A.DC.) Boiss. & Heldr.Wild typeLithospermic acid B106.1 mg/g DW[180]
Salvia przewalskii Maxim.Wild typeRosmarinic acid
Lithospermic acid B
67.1 mg/g DW
21.4 mg/g DW
[186]
Salvia bulleyana DielsWild typeRosmarinic acid110.2 mg/g DW[190]
Salvia bulleyana DielsWild typeRosmarinic acid
Salvianolic acid K
90 mg/g DW
10 mg/g DW
[191]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Malarz, J.; Yudina, Y.V.; Stojakowska, A. Hairy Root Cultures as a Source of Phenolic Antioxidants: Simple Phenolics, Phenolic Acids, Phenylethanoids, and Hydroxycinnamates. Int. J. Mol. Sci. 2023, 24, 6920. https://doi.org/10.3390/ijms24086920

AMA Style

Malarz J, Yudina YV, Stojakowska A. Hairy Root Cultures as a Source of Phenolic Antioxidants: Simple Phenolics, Phenolic Acids, Phenylethanoids, and Hydroxycinnamates. International Journal of Molecular Sciences. 2023; 24(8):6920. https://doi.org/10.3390/ijms24086920

Chicago/Turabian Style

Malarz, Janusz, Yulia V. Yudina, and Anna Stojakowska. 2023. "Hairy Root Cultures as a Source of Phenolic Antioxidants: Simple Phenolics, Phenolic Acids, Phenylethanoids, and Hydroxycinnamates" International Journal of Molecular Sciences 24, no. 8: 6920. https://doi.org/10.3390/ijms24086920

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop