Sei sulla pagina 1di 53

Biotechnology Advances 20 (2002) 101 153

Plant cell cultures:


Chemical factories of secondary metabolites
S. Ramachandra Raoa,*, G.A. Ravishankarb
a

Laboratory of Biofunctional Materials, School of Materials Science, Japan Advanced Institute of Science and
Technology, 1-1, Asahidai, Tatsunokuchi, Ishikawa 923-1292, Japan
b
Plant Cell Biotechnology Department, Central Food Technological Research Institute, Mysore 570 013, India

Abstract
This review deals with the production of high-value secondary metabolites including
pharmaceuticals and food additives through plant cell cultures, shoot cultures, root cultures and
transgenic roots obtained through biotechnological means. Plant cell and transgenic hairy root cultures
are promising potential alternative sources for the production of high-value secondary metabolites of
industrial importance. Recent developments in transgenic research have opened up the possibility of
the metabolic engineering of biosynthetic pathways to produce high-value secondary metabolites. The
production of the pungent food additive capsaicin, the natural colour anthocyanin and the natural
flavour vanillin is described in detail. D 2002 Elsevier Science Inc. All rights reserved.
Keywords: Secondary metabolites; Pharmaceuticals; Plant cell cultures; Hairy root cultures; Biotransformation;
Immobilization

1. Introduction
For centuries, mankind is totally dependent on plants as source of carbohydrates, proteins
and fats for food and shelter. In addition, plants are a valuable source of a wide range of
secondary metabolites, which are used as pharmaceuticals, agrochemicals, flavours, fragrances, colours, biopesticides and food additives. Over 80% of the approximately 30,000 known
natural products are of plant origin (Phillipson, 1990; Balandrin and Klocke, 1988; Fowler

* Corresponding author. Tel.: +81-761-51-1668; fax: +81-761-51-1665.


E-mail addresses: srrao@jaist.ac.jp, raovan@mailcity.com (S. Ramachandra Rao).
0734-9750/02/$ see front matter D 2002 Elsevier Science Inc. All rights reserved.
PII: S 0 7 3 4 - 9 7 5 0 ( 0 2 ) 0 0 0 0 7 - 1

102

S. Ramachandra Rao, G.A. Ravishankar / Biotechnology Advances 20 (2002) 101153

and Scragg, 1988). The number of known chemical structures is estimated to be nearly
fourfold greater than that in the microbial kingdom. In 1985, of the 3500 new chemical
structures identified, 2600 came from the higher plants. Worldwide, 121 clinically useful
prescription drugs are derived from plants (Payne et al., 1991). The surveys of plant
medicinal usage by the American public have shown an increase from just about 3% of
the population in 1991 to over 37% in 1998 (Brevoort, 1998). The US market sales of plant
medicinals have climbed to about US$3 billion per year (Glaser, 1999). Even today, 75% of
the worlds population relies on plants for traditional medicine. In the US, where chemical
synthesis dominates the pharmaceutical industry, 25% of the pharmaceuticals are based on
plant-derived chemicals (Payne et al., 1991; Farnsworth, 1985). Plants will continue to
provide novel products as well as chemical models for new drugs in the coming centuries,
because the chemistry of the majority of plant species is yet to be characterized (Cox and
Balick, 1994). The advent of chemical analyses and the characterization of molecular
structures have helped in precisely identifying these plants and correlating them with their
activity under controlled experimentation. Despite advancements in synthetic chemistry, we
Table 1
Plant-derived pharmaceuticals of importance
Product

Use

Plant species

Ajmalicine
Artemisinin
Ajmaline
Acinitine
Berberine
Camptothecin
Capsaicin
Castanospermine
Codeine
Colchicine
Digoxin
Diosgenin
Ellipticine
Emetine
Forskolin
Ginsenosides
Morphine
Podophyllotoxin
Quinine
Sanguinarine

Antihypertensive
Antimalarial

Intestinal ailment
Antitumour
Counterirritant
Glycoside inhibitor
Sedative
Antitumour
Heart stimulant
Steroidal precursor
Antitumour

Bronchial asthma
Health tonic
Sedative
Antitumour
Antimalarial
Antiplaque

Shikonin
Taxol
Vincristine
Vinblastine

Antibacterial
Anticancer
Antileukemic
Antileukemic

Cath. roseus
Artemisia annua
Ra. serpentina
Acotinum spp.
C. japonica
Camptotheca acuminata
Ca. frutescens
Castanospermum australe
P. somniferum
Colchium autumnale
Di. lanata
Dioscorea deltoidea
Orchrosia elliptica
Cephaclis ipecaccuanha
Coleus forskolii
Panax ginseng
P. somniferum
Podophyllum petalum
Cinchon. ledgeriana
Sanguinaria canadensis
P. somniferum
L. erythrorhizon
Taxus brevifolia
Cath. roseus
Cath. roseus

n/a: not available. Adapted from Ravishankar and Ramachandra Rao (2000).

Cost
(US$ per kilogram)
37,000
400
75,000
n/a
3250
432,000
750
n/a
17,000
35,000
3000
1000
240,000
1500
n/a
n/a
340,000
n/a
500
4,800
4500
600,000
2,000,000
1,000,000

S. Ramachandra Rao, G.A. Ravishankar / Biotechnology Advances 20 (2002) 101153

103

still depend upon biological sources for a number of secondary metabolites including
pharmaceuticals (Pezzuto, 1995). Their complex structural features are difficult to synthesize.
We have listed some plant-derived pharmaceuticals in Table 1.
Elaborative pathways from basic primary metabolites, which are synthesized immediately
as a result of photosynthetic activity, produce secondary metabolites. Many of them are
unique to the plant kingdom and are not produced by microbes or animals. However, with the
advancement of transgenic research, it is possible to produce compounds and molecules,
which were also not originally synthesized in plants.

2. Need for a biotechnological approach


Biotechnology offers an opportunity to exploit the cell, tissue, organ or entire organism by
growing them in vitro and to genetically manipulate them to get desired compounds. Many
facets of biotechnological approaches can be envisaged (Table 2). Since the world population
is increasing rapidly, there is extreme pressure on the available cultivable land to produce
food and fulfill the needs. Therefore, for other uses such as production of pharmaceuticals and
chemicals from plants, the available land should be used effectively. Hence, it is appropriate
to develop modern technologies leading to plant improvement for better utilization of the land
to meet the requirements.
The development of micropropagation methods for a number of medicinal plant species
has been already reported and needs to be adopted (Naik, 1998). Cryopreservation of cells is
an area of importance in the conservation of medicinal plants. It has already been used in
many plant species. The development and adoption of plant cell culture and organ culture
methods have lead to the production of plant products on a large scale. This has been possible
Table 2
Aspects of biotechnological approaches to plant-derived secondary metabolites
1. Plant cell tissue and organ cultures
Cell culture
Shoot culture
Root culture
Scale-up of cultures
2. Transgenic plants/organisms
Metabolic engineering
Heterologous expression
Molecular forming
3. Micropropagation of medicinal plants
Endangered plants
High-yielding varieties
Metabolically engineered plants
4. Newer sources
Algae
Other photosynthetic marine forms
5. Safety considerations

104

S. Ramachandra Rao, G.A. Ravishankar / Biotechnology Advances 20 (2002) 101153

by the combined efforts of cell biologists and chemical engineers. Chemical engineers are
currently developing improved and appropriate bioreactors for the improvement of production systems by adopting techniques of growth and metabolite production coupled with
downstream processing of the products. The improvements in molecular biological research
have given a new dimension to in vitro culture as well as for plant improvement, enhancing
the yields of the product and resulting in multiple products or producing novel products from
genetically engineered plants. Moreover, the need for safer drugs without side effects has led
to the use of natural ingredients with proven safety. These factors have laid emphasis on the
use of biotechnological methods to enhance the production of pharmaceuticals and food
additives, both in quality and quantity.

3. Plant cell culture as a source of secondary metabolites


Plant cell cultures are an attractive alternative source to whole plant for the production of
high-value secondary metabolites (Ravishankar et al., 1999; Dornenburg and Knorr, 1997;
Scragg, 1997; Alfermann and Petersen, 1995; DiCosmo and Misawa, 1995; Stockigt et al.,
1995; Endress, 1994; Ravishankar and Venkataraman, 1990) (Tables 3 and 4). Plant cells
are biosynthetically totipotent, which means that each cell in culture retains complete
genetic information and hence is able to produce the range of chemicals found in the parent
plant. The advantages of this technology over the conventional agricultural production are
as follows.
It is independent of geographical and seasonal variations and various environmental
factors.
It offers a defined production system, which ensures the continuous supply of products,
uniform quality and yield.
It is possible to produce novel compounds that are not normally found in parent plant.
Table 3
Groups of natural products that were so far isolated from tissue and suspension cultures of higher plants
Phenylpropanoids

Alkaloids

1.
2.
3.
4.

Anthocyanins
1. Acridines
Coumarins
2. Betalaines
Flavonoids
3. Quinolizidines
Hydroxycinnamoyl 4. Furonoqui nones
derivatives
5. Harringtonines
5. Isoflavonoids
6. Isoquinolines
6. Lignans
7. Indoles
7. Phenolenones
8. Purines
8. Proanthocyanidins
9. Pyridines
9. Stilbenes
10. Tropane
10. Tanins
alkaloids
Adapted from Stockigt et al. (1995).

Terpenoids
1.
2.
3.
4.
5.

Quinones

Steroids

Carotenes
1. Anthroquinones
1. Cardiac glycosides
Monoterpenes 2. Benzoquinones
2. Pregnenolone
Sesquiterpenes 3. Naphthoquinones derivatives
Diterpenes
Triterpenes

S. Ramachandra Rao, G.A. Ravishankar / Biotechnology Advances 20 (2002) 101153

105

Table 4
Food additives from plant cell cultures
Product type
Colours
Anthocyanins

Betalaines
Crocin
Crocetins
Carotenoids
Anthraquinones
Naphthoquinones
Flavours
Vanillin
Garlic
Onion
Basmati
Citrus flavour
Cocoa flavour
Pungent food additive
Capsaicin
Sweeteners
Stevioside
Glycyrrhizin
Thaumatin
Essential oils
Mint oil
Chamomile oil
Jasmine oil
Aniseed oil

Plant species

Reference

V. vinifera
Euphorbia spp.
D. carota
Pe. frutescens
B. vulgaris
Cheno. rubrum
Crocus sativus
Gardenia jasminoides
Lycopersicon esculentum
Cinchon. ledgeriana
M. citrifolia
L. erythrorhizon

Pepin et al. (1995)


Yamamoto et al. (1982)
Rajendran et al. (1994)
Zhong and Yoshida (1995)
Klebnikov et al. (1995)
Berlin et al. (1986)
Sujata et al. (1990)
George and Ravishankar (1995)
Fosket and Radin (1983)
Robbins and Rhodes (1986)
Kieran et al. (1993)
Sim and Chang (1993)

Va. planifolia
Allium sativum
Allium cepa
Oryza sativa
Citrus spp.
Theobromo cacao

Dornenburg and Knorr (1996)


Ohsumi et al. (1993)
Collin and Masker (1988)
Suvarnalatha et al. (1994)
Cresswell (1990)
Townsley (1972)

Ca. frutescens
Ca. annuum

Lindsey and Yeoman (1984)


Johnson et al. (1990)

Stevia rebaudiana
Glycyrrhiza glabra
Thaumatococcus danielli

Swanson et al. (1992)


Hayashi et al. (1988)
van der Wel and Ledeboer (1989)

Mentha piperata
Ma. chamomilla
Jasmine officinale
Pim. anisum

Chung et al. (1994)


Kireeva et al. (1978)
Banthrope (1994)
Ernst (1989)

Adapted from Ramachandra Rao (1998).

It is independent of political interference.


Efficient downstream recovery and product.
Rapidity of production.
In addition, plant cell can perform stereo- and regiospecific biotransformations for the
production of novel compounds from cheap precursors.
There are a number of plant cell cultures producing a higher amount of secondary
metabolites than in intact plants (Table 5). However, there are still problems in the
production of metabolites by cell cultures resulting from the instability of cell lines, low
yields, slow growth and scale-up problems (Ravishankar and Venkataraman, 1993).

106

S. Ramachandra Rao, G.A. Ravishankar / Biotechnology Advances 20 (2002) 101153

Table 5
High yields of secondary products
Product

Plant species

Yield (% D.W.)

Reference

Rosmarinic acid
Rosmarinic acid
Anthroquinones
Shikonin
Berberine
Jatrorhizine
Anthocyanins
Berberine
Diosgenin
Sanguinarine
Serpentine

Sa. officinalis
Col. blumei
M. citrifolia
L. erythrorhizon
Th. minus
Berberis wilsonae
Pe. frutescens
C. japonica
Diosc. deltoidea
P. somniferum
Cath. roseus

36.0
21.4
18.0
12.4
10.6
10.0
8.9
7.5
3.8
2.5
2.2

Hippolyte et al. (1992)


Ulbrich et al. (1985)
Zenk et al. (1975)
Fujita (1988)
Kobayashi et al. (1988)
Breuling et al. (1985)
Zhong et al. (1994)
Matsubara et al. (1989)
Sahai and Knuth (1985)
Park et al. (1992)
Zenk et al. (1977)

Adapted from Ravishankar and Ramachandra Rao (2000).

Several strategies have been adopted for the enhancement of metabolites (Table 6).
There has been tremendous success in the production of high-value secondary metabolites such as shikonin from cell cultures of Lithospermum erythrorhizon, berberine from
Coptis japonica and sanguinarine from Papaver somniferum. These are produced at
industrial levels.

4. Strategies to increase secondary metabolite production in plant cell cultures


During the past decade, a considerable progress has been made to stimulate formation
and accumulation of secondary metabolites using plant cell cultures (Ravishankar and
Ramachandra Rao, 2000; Dixon, 1999; Ravishankar and Venkataraman, 1993; Buitelaar
and Tramper, 1992; Fowler and Stafford, 1992; Payne et al., 1991). The adopted
strategies for enhancing the secondary metabolites of plant cell cultures have been
described in detail.

Table 6
Strategies to enhance production of secondary metabolites in plant cell cultures
1. Obtaining efficient cell lines for growth
2. Screening of high-growth cell line to produce metabolites of interest
a. Mutation of cells
b. Amenability to media alterations for higher yields
3. Immobilization of cells to enhance yields of extracellular metabolites and to facilitate biotransformations
4. Use of elicitors to enhance productivity in a short period of time
5. Permeation of metabolites to facilitate downstream processing
6. Adsorption of the metabolites to partition the products from the medium and to overcome feedback inhibition
7. Scale-up of cell cultures in suitable bioreactors

S. Ramachandra Rao, G.A. Ravishankar / Biotechnology Advances 20 (2002) 101153

107

4.1. Screening and selection of highly productive cell lines


Strain improvement begins with the choice of a parent plant with high contents of the
desired products for callus induction to obtain high-producing cell lines. Then, a screening of
the heterogeneous population for variant cell clones containing the highest levels of desired
product was employed.
The heterogeneity in the biochemical activity existing within a population of cells has been
exploited to obtain highly productive cell lines (Ogino et al., 1978). Selection can be easily
achieved if the product of interest is a pigment. For example, in cultures of L. erythrorhizon,
extensive screening of a number of clones resulted in a 1320-fold increase in shikonin
production (Fujita et al., 1984). Enhanced anthocyanin production by clonal selection and
visual screening has been reported in Euphorbia milli and Daucus carota (Yamamoto et al.,
1982; Dougall, 1980). Other techniques such as HPLC and RIA were also used to screen for
high-yielding cell lines (Matsumoto et al., 1980; Zenk, 1978).
Mutation strategies have also been employed in order to obtain overproducing cell lines.
The use of selective agents has been employed as an alternative approach to select highyielding cell lines (Rhodes et al., 1988). In this method, a large population of cells is exposed
to a toxic (or cytotoxic) inhibitor or environmental stress and only cells that are able to resist
the selection procedures will grow. p-Fluorophenylalanine (PFP), an analogue of phenylalanine, was extensively used to select high-yielding cell lines with respect to phenolics
(Berlin, 1980). Increased capsaicin and rosmarinic acid in PFP cell lines of Capsicum
annuum and Anchusa were reported (Salgado-Garciglia and Ochoa-Alejo, 1990; Quesnell
and Ellis, 1989). Other selective agents such as 5-methyltryptophan, glyphosate and biotin
have also been used to select high-yielding cell lines (Amrhein et al., 1985; Wataneba et al.,
1982; Widholm, 1974).
4.2. Manipulation of nutrients to improve yield
Manipulation of the culture environment must be effective in increasing the product
accumulation. The expression of many secondary metabolite pathways is easily altered by
external factors such as nutrient levels, stress factors, light and growth regulators. Many of the
constituents of plant cell culture media are important determinants of growth and accumulation of secondary metabolites (Stafford et al., 1986; Misawa, 1985).
4.2.1. Sugar levels
Plant cell cultures are usually grown heterotrophically using simple sugars as carbon
source and inorganic supply of other nutrients. The level of sucrose has been shown to affect
the productivity of secondary metabolite-accumulating cultures. Thus, sucrose concentrations of 2.5% (w/v) and 7.5% (w/v) in Coleus blumei media brought about rosmarinic acid
yields of 0.8 and 3.3 g/l, respectively (Misawa, 1985). For indole alkaloid accumulation in
cell culture as of Catharanthus roseus, 8% (w/v) sucrose was found to be optimal in the
tested concentration range of 4 12% (w/v) (Knobloch and Berlin, 1980). Yields of
benzophenanthridine alkaloids from suspension cultures of Eschscholtzia californica were

108

S. Ramachandra Rao, G.A. Ravishankar / Biotechnology Advances 20 (2002) 101153

increased 10-fold to around 150 mg/l by increasing the sucrose concentration to 8% (w/v)
(Berlin et al., 1983). The osmotic stress created by sucrose alone and with other osmotic
agents were found to regulate anthocyanin production in Vitis vinifera cell suspension
cultures (Do and Cormier, 1990). A dual role of sucrose as carbon source and osmotic agent
was observed in Solanum melongena (Mukherjee et al., 1991). However, higher concentrations of sucrose at 5% (w/v) reduced the anthocyanin production in cell suspension
cultures of Aralia cordata, where 3% (w/v) favoured the anthocyanin accumulation
(Sakamoto et al., 1993).
4.2.2. Nitrate levels
Nitrogen concentration was found to affect the level of proteinaceous or amino acid
products in cell suspension cultures. The plant tissue culture medium such as MS, LS or B5
has both nitrate and ammonium as sources of nitrogen. However, the ratio of the ammonium/
nitratenitrogen and overall levels of total nitrogen have been shown to markedly affect the
production of secondary plant products. For example, reduced levels of NH4 + and increased
levels of NO3 promoted the production of shikonin and betacyanins, whereas higher ratios
of NH4 + /NO3 increased the production of berberine and ubiquinone (Bohm and Rink,
1988; Nakagawa et al., 1984; Fujita et al., 1981; Ikeda et al., 1977). Reduced levels of total
nitrogen improved the production of capsaicin in Capsicum frutescens, anthraquinones in
Morinda citrifolia and anthocyanins in Vitis species (Yamakawa et al., 1983; Yeoman et al.,
1980; Zenk et al., 1975). However, complete elimination of nitrate in cultures of Chrysanthemum cinerariaefolium induced twofold increases in pyrethrin accumulation in the
second phase of culture (Rajasekaran et al., 1991).
4.2.3. Phosphate levels
The phosphate concentration in the medium can have a major effect on the production of
secondary metabolites in plant cell cultures. Higher levels of phosphate were found to
enhance the cell growth, where it had negative influence on secondary product accumulation.
Sasse et al. (1982) have given a number of examples to show that a medium limited in
phosphate either induces or stimulates both the product and the levels of key enzymes leading
to the product. Thus, reduced phosphate levels induced the production of ajmalicine and
phenolics in Cath. roseus, of caffeoyl putrescines in Nicotiana tabacum and of harman
alkaloids in Peganum harmala. Similar results were obtained in a separate study for the
production of betacyanins in callus cultures of Beta vulgaris (Bohm and Rink, 1988). In
contrast, increased phosphate was shown to stimulate synthesis of digitoxin in Digitalis
purpurea and of betacyanin in Chenopodium rubrum and Phytolacca americana (Bohm and
Rink, 1988; Hagimori et al., 1982a,b).
4.2.4. Growth regulators
Growth regulator concentration is often a crucial factor in secondary product accumulation
(DiCosmo and Towers, 1984; Deus and Zenk, 1982). The type and concentration of auxin or
cytokinin or the auxin/cytokinin ratio alters dramatically both the growth and the product
formation in cultured plant cells (Mantell and Smith, 1984). The growth regulator 2,4-

S. Ramachandra Rao, G.A. Ravishankar / Biotechnology Advances 20 (2002) 101153

109

dichlorophenoxyacetic acid (2,4-D) has been shown to inhibit the production of secondary
metabolites in a large number of cases. In such cases, elimination of 2,4-D or replacement of
2,4-D by naphthalene acetic acid (NAA) or indole acetic acid (IAA) has been shown to
enhance the production of anthocyanins in suspensions of Populus and D. carota, of
betacyanins in suspensions of Portulaca, of nicotine in suspensions of N. tabacum, of
shikonin in suspensions of L. erythrorhizon and of anthraquinones in M. citrifolia (Rajendran
et al., 1992; Seitz and Hinderer, 1988; Tabata, 1988; Sahai and Shuler, 1984; Bohm and Rink,
1988; Zenk et al., 1975). However, stimulation by 2,4-D has been observed in carotenoid
biosynthesis in suspensions of D. carota (Mok et al., 1976) and in anthocyanin production in
callus cultures of Oxalis linearis (Meyer and van Staden, 1995).
Cytokinins have different effects depending on the type of metabolite and species
concerned. Thus, kinetin stimulated the production of anthocyanin in Haplopappus gracilus
but inhibited the formation of anthocyanins in Populus cell cultures (Seitz and Hinderer, 1988;
Mok et al., 1976). Gibberellic acid and abscisic acid are reported to suppress production of
anthocyanins in a number of cultures (Bohm and Rink, 1988; Seitz and Hinderer, 1988).
4.2.5. Precursor feeding
Precursor feeding has been an obvious and popular approach to increase secondary
metabolite production in plant cell cultures. The concept is based upon the idea that any
compound, which is an intermediate, in or at the beginning of a secondary metabolite
biosynthetic route, stands a good chance of increasing the yield of the final product. Attempts
to induce or increase the production of plant secondary metabolites, by supplying precursor or
intermediate compounds, have been effective in many cases. The addition of phenylalanine as
a precursor led to improvement in rosmarinic acid yield in Col. blumei cell cultures (Ibrahim,
1987). Addition of phenylalanine to Salvia officinalis suspension cultures stimulated the
production of rosmarinic acid and decreased the production time as well (Ellis and Towers,
1970). Phenylalanine is also the precursor of the N-benzoylphenylisoserine side chain of
taxol, and supplementation of Taxus cuspidata cultures with phenylalanine resulted in
increased yields of taxol (Fett-Neto et al., 1994, 1993). Use of the distant precursor
phenylalanine and a near precursor such as isocapric acid resulted in enhanced capsaicin
content in cell cultures of Ca. frutescens (Lindsey and Yeoman, 1985; Yeoman et al., 1980).
Feeding of ferulic acid to cultures of Vanilla planifolia resulted in increase in vanillin
accumulation (Romagnoli and Knorr, 1988). Similarly, anthocyanin synthesis in carrot
cultures was restored by the addition of a dihydroquarcetin (naringen). Furthermore, addition
of leucine led to enhancement of volatile monoterpenes a- and b-pinine in cultures of Perilla
frutescens, whereas addition of geraniol to rose cell cultures led to accumulation of nerol and
citronellol (Mulder-Krieger et al., 1988).
4.3. Optimizing the culture environment
Culture environmental conditions such as light, temperature, medium pH and oxygen
have been examined for their effect upon secondary metabolite accumulation in many types
of cultures.

110

S. Ramachandra Rao, G.A. Ravishankar / Biotechnology Advances 20 (2002) 101153

4.3.1. Temperature
A temperature range of 1725 C is normally used for the induction of callus tissues and
growth of cultured cells. However, each plant species may favour a different temperature.
Toivonen et al. (1992) found that lowering the cultivation temperature increased the total
fatty acid content per cell in dry weight. When the temperature was maintained at 19 C,
biotransformation of digitoxin to digoxin is favoured, whereas 32 C favours purpureaglycoside A formation in Digitalis lanata cell cultures (Kreis and Reinhard, 1992). Ikeda et al.
(1977) observed a higher yield of ubiquinone in tobacco cell cultures at 32 C when
compared to either 24 or 28 C. Courtois and Guren (1980) reported a 12-fold higher
production of crude alkaloids in cell cultures of Cath. roseus at 16 C as compared to the
normal 27 C.
4.3.2. Illumination
Accumulation of anthocyanin was strongly stimulated by light in cell cultures of D. carota
and Vitis hybrids (Seitz and Hinderer, 1988). Illumination was found to affect the composition
of sesquiterpenes in callus cultures of Marticaria chamomilla (Mulder-Krieger et al., 1988).
Exclusion of light in callus cultures of Citrus limon prompted the accumulation of
monoterpenes (Mulder-Krieger et al., 1988).
4.3.3. Medium pH
The medium pH is usually adjusted between 5 and 6 before autoclaving and extremes of
pH are avoided. The concentration of hydrogen ions in the medium changes during the
development of the culture. The medium pH decreases during ammonia assimilation and
increases during nitrate uptake (McDonald and Jackman, 1989). Photoautotrophic cell
suspension cultures of Cheno. rubrum showed that the increase in the external pH from
4.5 to 6.3 increased the cytosolic pH by 3.0 units and the vacuolar pH by about 1.3 units
(Husemann et al., 1992).
4.3.4. Agitation and aeration
The importance of aeration and agitation is crucial for large-scale production. Kreis and
Reinhard (1989) described that the influence of dissolved oxygen levels of 50% allowed an
alkaloid yield of around 3-g/l culture after 20 days of growth in an airlift bioreactor. Higher
aeration rates produced a dramatic decrease in alkaloid productivity. It is evident that airlift
and stirred tank bioreactors can allow similar secondary product levels in cultured plant
cells. However, in stirred tank vessels, the characteristics of the stirrer may be critical
(Kreis and Reinhard, 1989).
Ambid and Fallot (1981) investigated the effect of the composition of the gaseous
environment on production of volatiles by fruit suspension cultures. They reported that
the addition of carbon dioxide stimulated the synthesis of monoterpenes by Muscat grape
suspensions and induced the formation of linalool. Kobayashi et al. (1991) reported that
the use of carbon dioxide at the 2% level was critical to prevent cell browning and to
sustain berberine production in suspension cultures of Thalictrum minus in bubble
column reactors.

S. Ramachandra Rao, G.A. Ravishankar / Biotechnology Advances 20 (2002) 101153

111

4.4. Elicitation
Plants produce secondary metabolites in nature as a defense mechanism against attack by
pathogens. Plants have been found to elicit the same response as the pathogen itself when
challenged by compounds of pathogenic origin (elicitors). Elicitors are signals triggering
the formation of secondary metabolites. Secondary pathways are activated in response to
stress. Biotic and abiotic elicitors are used to stimulate secondary metabolite product
formation in plant cell cultures, thereby reducing the process time to attain high product
concentrations and increased culture volumes (Barz et al., 1988; Eilert, 1987; DiCosmo and
Tallevi, 1985). Elicitors of fungal, bacterial and yeast origin, viz. polysaccharides,
glycoproteins, inactivated enzymes, purified curdlan, xanthan and chitosan, and salts of
heavy metals were reported for the production of various secondary metabolites (Ramachandra Rao et al., 1996a,b; Rajendran et al., 1994; Guo et al., 1992; Furze et al., 1991;
Johnson et al., 1991; Robbins et al., 1991; DiCosmo et al., 1987; Funk et al., 1987).
Treatment of P. somniferum cell suspensions with a homogenate of Botrytis mycelium
resulted in a remarkable accumulation of sanguinarine of up to 3% of the cell dry weight
(Constabel, 1990). The treatment of root cultures of Datura stramonium with copper and
cadmium salts has been found to induce the rapid accumulation of high levels of
sesquiterpenoid defensive compounds (Furze et al., 1991).

4.5. Permeabilization
In most cases, the products formed by plant cell cultures are stored in the vacuoles. In
order to release the products from vacuoles of plant cells, two membrane barriers plasma
membrane and tonoplast have to be penetrated. Cell permeabilization depends on the
formation of pores in one or more of the membrane systems of the plant cell, enabling the
passage of various molecules into and out of the cell. The permeability of the cells can be
monitored by measuring the activity of enzymes of the primary metabolism, viz.
hexokinase, glucose 6-phosphate dehydrogenase, isocitrate dehydrogenase, malic and
citrate synthetase (Brodelius, 1988b). Attempts have been made to permeabilize the plant
cells transiently, to maintain the cell viability and to have short time periods of increased
mass transfer of substrate and metabolites to and from the cell (Parr et al., 1987; Brodelius
and Nilsson, 1983). A wide variety of permeabilizing agents are used to enhance the
accessibility of enzymes or to provoke release of intracellular stored product (Berlin et al.,
1989; Knorr et al., 1985; Parr et al., 1984; Felix, 1982). Organic solvents such as
isopropanol, dimethylsulfoxide (DMSO) and polysaccharides like chitosan have been used
as permeabilizing agents (Van Uden et al., 1990; Beaumont and Knorr, 1987; Knorr and
Teutonico, 1986). Other permeabilization methods include ultrasonication, electroporation
and ionophoretic release, in which the cells are subjected to a low current in a specially
designed device (Brodelius et al., 1988). In addition, using high electric field pulses and
ultrahigh pressure (Dornenburg and Knorr, 1993) has been reported for the recovery of
secondary metabolites.

112

S. Ramachandra Rao, G.A. Ravishankar / Biotechnology Advances 20 (2002) 101153

4.6. In situ product removal


A low accumulation of secondary compounds in cell cultures in a number of cases
may not be due to a lack of key biosynthetic enzymes but rather due to feedback
inhibition, enzymatic or nonenzymatic degradation of the product in the medium or
volatility of substances produced. In such cases, it should be possible to increase the net
production by the addition of an artificial site for product accumulation, for example, by
use of second solid or liquid phase introduced into the aqueous medium. Such threephase systems accumulate traces of secondary metabolites from the culture medium.
The use of in situ product removal of metabolites has a number of key potential
advantages beyond promoting secretion. The removal and sequestering of the product in
a nonbiological compartment may increase its total production (Beiderbeck and
Knoop, 1988).
Robbins and Rhodes (1986) reported that addition of amberlite XAD-7 resin stimulated
the production of anthraquinones by 15 times compared to a medium without adsorbent.
Addition of charcoal led to 2060-fold improvements in yields of coniferyl aldehyde in
Ma. chamomilla suspensions (Beiderbeck and Knoop, 1987). The addition of Miglyol or
silica gel RP8 stimulated ethanol production in cell cultures of Pimpinella anisum
(Mulder-Krieger et al., 1988). The addition of XAD-4 increased the vanilla flavour
production in Vanilla fragrans cell suspension cultures (Knuth and Sahai, 1991).

4.7. -Cyclodextrins
Many plant cell cultures hardly convert precursors in the presence of organic phases,
often as a result of dramatic decrease of cell viability. Therefore, the enzymatic activities
are reduced in these systems. A new approach to solve this problem of bioconversion of
water-insoluble precursors is to combine the advantages of apolar systems (higher
solubility if the substrate) and aqueous systems (maintenance of cell viability) by carrying
out bioconversions in the presence of clathering agents such as cyclodextrins. Cyclodextrins have the ability to form stable inclusion complexes with natural spices and
flavour substances in their cyclodextrin cavity (Haggin, 1992). Cyclodextrins may be
modified through substituting various functional compounds on the primary or secondary
phase of the molecule. The chemically modified cyclodextrins are more water soluble
than native cyclodextrins (Eastburn and Tao, 1994; Szejtli, 1988). Through complexation,
the physical properties of ligands are changed, including their solubility in aqueous media
(Qi and Hedges, 1995; Van Uden et al., 1994; Szejtli, 1986, 1982). The hydroxylation of
b-estradiol to 4-hydroxy and 2-hydroxy b-estradiol was enhanced in the presence of
b-cyclodextrin in Mucuna pruriens cell cultures (Woerdenbag et al., 1990a) and multistep
conversion of coniferyl alcohol into podophyllotoxin was demonstrated in Podophyllum
hexandrum cell suspension cultures (Woerdenbag et al., 1990b). Cyclodextrin complexation of flavours provides protection against the damaging factors of the environment (Qi
and Hedges, 1995).

S. Ramachandra Rao, G.A. Ravishankar / Biotechnology Advances 20 (2002) 101153

113

5. Organ cultures as a source of pharmaceuticals


Since production of secondary metabolites is generally higher in differentiated tissues,
there are attempts to cultivate shoot cultures and root cultures for the production of
medicinally important compounds. These organ cultures are relatively more stable (Roja,
1994). There are a number of medicinal plants whose shoot cultures have been studied for
metabolites (Table 7). Similarly, root cultures are valuable sources of medicinal compounds
(Table 8). Root systems of higher plants generally exhibit slower growth and are difficult to
harvest. Hence, alternative methods need to be found for root-derived compounds. Until now,
however, there is no commercial process as an alternative for root-derived compounds, except
in case of utilizing hairy root culture systems, which has been described in this review.

6. Hairy root cultures for pharmaceuticals


The ability of Agrobacterium rhizogenes to induce hairy roots in a range of host plants has
lead to studies on it as a source of root-derived pharmaceuticals (Flores et al., 1999). Tepfer
(1990) summarized 116 plants belonging to 30 dicotyledonous families wherein hairy roots
have been induced. Hairy roots are induced by transfer of T-DNA from the plasmid of Agr.
rhizogenes (Ambros et al., 1986; Petit et al., 1983) to host tissue, resulting in root formation
by virtue of auxin synthesis genes coded by bacterial DNA. The Ri plasmid of Agr.
rhizogenes also elicits the synthesis of opines such as agropine or mannopine. The transformed nature of the roots is genetically checked by opine detection or Southern analysis.
The interest in hairy roots is mainly due to their ability to grow fast without needing an
external supply of auxins. Many times, they do not need incubation under light. They are
fairly well stable in metabolite yield due to their genetic stability. Because of these
Table 7
Shoot cultures of medicinal plants
Plant species

Product

Reference

Artemi. annua
Atro. belladona
Begonia spp.
Cath. roseus
Cinchona spp.

Artemesinin
Atropine

Vindoline
Vinblastine
Quinine
Cardenolides
Cardenolides
Essential oils
Kutkin
Steviosides
Withanolides
Indirubin
Alkaloids

Park et al. (1989)


Benjamin et al. (1987)
Takayama and Misawa (1981)
Staba and Chung (1981)
Krueger et al. (1982)
Hirata et al. (1987)
Lui and Staba (1979)
Hagimori et al. (1982a,b)
Charlwood and Moustou (1988)
Upadhyay et al. (1989)
Akita et al. (1994)
Heble (1985)
Shim et al. (1998)
Konishi et al. (1998)

Di. lanata
Di. purpurea
Pelargonium tomentosum
Picrprrhiza kurroa
Ste. rebaudiana
Withania somniferum
Polygonum tinctorium
Decentra pergrina

Adapted from Ravishankar and Ramachandra Rao (2000).

114

S. Ramachandra Rao, G.A. Ravishankar / Biotechnology Advances 20 (2002) 101153

Table 8
Root cultures of medicinal plants
Plant species

Product

Reference

B. vulgaris
Bidens alba
Calystegia sepium
Coreopsis tinctoria
Hyoscamus albus
Hyoscamus muticus
Hemidesmus indicus
Artemisia absynthium
Polygonum tinctorium

Betalaines
Polyacetylenes
Tropane alkaloids
Phenylpropanoids
Hyoscyamine
Hyoscyamine
2-hydroxy-4-methoxybenzaldehyde
Volatile oils
Indigo, Indirubin

Hamill et al. (1986)


Norton and Towers (1986)
Jung and Tepfer (1987)
Thron et al. (1989)
Hashimoto and Yamada (1986)
Flores et al. (1987)
Sreekumar et al. (1998)
Kennedy et al. (1993)
Shim et al. (1998)

Adapter from Payne et al. (1991).

advantages, many of the root-derived plant products once not considered feasible for
production by cell culture are being reinvestigated for production using the hairy root culture
technology. A number of such examples are given in Table 9. Several factors influence the
yield of secondary metabolites of pharmaceutical interest in hairy root cultures. These are
nutrients, elicitors and biotransformations of precursors to products and genetic manipulation
through the Ri plasmid of Agr. rhizogenes.
Several hairy roots have been put to scale-up studies in bioreactors. However, due to their
structural features and metabolite localization characteristics, they need different type of
reactors than the ones used for plant cell cultures. Hairy roots need anchorage for growth
Table 9
Hairy root cultures producing pharmaceutical products of interest
Plant species

Product

Reference

Bidens spp.
Cinchon. ledgeriana
Cichorium intybus
Datura spp.
Cassia spp.
Duboisia leichhardtii
Echinacea purpurea
Glycyrrhiza uralensis
Hy. albus
Panax ginseng
Salvia miltorrhiza
Artemi. absynthium
L. erythrorhizon
Ra. serpentina
Rubia cordifolia
Gl. glabra
Panax ginseng
Hy. muticus

Polyacetylenes
Quinolene alkaloids
Esculetin
Tropane
Anthroquinones
Tropane alkaloids
Alkaloids
Glycyrrhizin
Alkaloids
Saponin
Diterpenes
Volatile oil
Shikonin
Ajmaline, serpentine
Anthroquinones
Isoprenylated flavonoids
Ginsenoside
Hyoscyamine

McKinely et al. (1993)


Hamill et al. (1987)
Bais et al. (1999)
Rhodes (1989)
Ko et al. (1988)
Mano et al. (1989)
Trypsteen et al. (1991)
Ko et al. (1989)
Shimomura et al. (1991)
Yoshikawa and Furuya (1987)
Hu and Alfermann (1993)
Kennedy et al. (1993)
Shimomura et al. (1986)
Benjamin et al. (1994)
Shin and Kim (1996)
Asada et al. (1998)
Kunshi et al. (1998)
Sevon et al. (1998)

Adapted from Ravishankar and Ramachandra Rao (2000).

S. Ramachandra Rao, G.A. Ravishankar / Biotechnology Advances 20 (2002) 101153

115

since they are highly branched (Fig. 1). Various configurations of hairy root bioreactors are
given in Fig. 2. Certain hairy roots are tolerant to submerged cultivations. Taya et al. (1989)
found that stirred tank reactors are poor for cultivation of B. vulgaris. In our experience, hairy
roots of B. vulgaris do not produce betalaines when submerged totally. In shake flask cultures, they proliferate fast and produce betalaines after achieving a stationary phase of growth.
Air-sparging without impeller-driven agitation may also be helpful as in the case of hairy
roots of Nicotiana rustica (Rodriguez-Mendiola et al., 1991). We have found that use of airsparging system is suitable for chicory hairy roots for esculin and esculetins production (Bais
et al., 1999). Therefore, any single design of reactor will not be suitable for all hairy roots.
Most remarkable developments of scale-up in large vessels have been in the cultivation of
Panax ginseng hairy root biomass in a 20-ton cultivation tank (Scheidegger, 1990).
Hairy roots also produce valuable compounds by biotransformations (Table 10). Recently,
an innovative approach of coculture of hairy roots of Atropa belladona and shooty teratomas
of scopolamine-rich Duboisia plants was done, wherein hyoscyamine released by the former
was bioconverted to scopolamine by the latter tissues (Subroto et al., 1996). However, these
systems need to be studied carefully within parameters of uptake mechanisms, biosynthetic
potential of the tissue and, if possible, the extracellular release of the final product. Thus,
hairy root technology will be useful in not only producing root-derived compounds but also
to produce novel compounds by biotransformations.
The ability to engineer the genomic DNA of hairy root through tailormade Ri plasmid will
be a tremendous use in producing novel compounds. Using this systems, it may be possible to

Fig. 1. Highly branched hairy root cultures of Cichorium intybus.

116

S. Ramachandra Rao, G.A. Ravishankar / Biotechnology Advances 20 (2002) 101153

Fig. 2. Configuration of hairy root bioreactors.

produce more than one product with a single type of hairy root. There already are attempts in
this direction as exemplified by the introduction of cDNA of ornithine decarboxylase (ODC)

S. Ramachandra Rao, G.A. Ravishankar / Biotechnology Advances 20 (2002) 101153

117

Table 10
Biotransformations using hairy root cultures for production of pharmaceuticals
Plant species

Substrate

Product

Reference

Hy. niger

Hyoscyamine

Scopolamine

Cinchon. ledgeriana
Nicotiana spp.

Tryptophan
Lysine, cadaverine

Quinine
Nicotine

Putrescine, agmatine
Putrescine
Spermidine
Cadaverine
2-phenylpropanoic acid
Digitoxigenin
Hyoscyamine
Hyoscyamine
18b-glycyrrhetinic acid
Butylated hydroxytoluene
Phenolic acid

Anabasine
Scopolamine
Hyoscyamine

Hashimoto and Yamada


(1983)
Hay et al. (1986)
Walton and Belshaw
(1988)
Walton et al. (1988)
Yoshioka et al. (1989)

Duboisia myoporoides

Panax ginseng
Panax ginseng
Atro. belladona
Dubo. leichhardtii
Panax ginseng
Bidens sulphureus
Panax ginseng

Sugar esters
Digitoxin
Scopolamine
Scopolamine
Glucosides
Stilbene quinones
Glycosylated phenolic
compounds

Furuya et al. (1989)


Kawaguchi et al. (1990)
Subroto et al. (1996)
Subroto et al. (1996)
Asada et al. (1993)
Flores et al. (1994)
Ushiyama and Furuya
(1989)

from yeast into N. rustica. ODC enhances the formation of alkaloid nicotine since it is the
first step of pyrrolidine alkaloid biosynthesis. Hyoscyamine 6b-hydroxylase (H6H) enzyme
that catalyzes epoxidative conversion of hyoscyamine to scopolamine is an important step in
the biogenetic pathway (Yamada and Hashimoto, 1988), which has been induced in Atro.
belladona by the H6H gene from Hyoscyamus niger. The hairy roots of Atro. belladona
produced high amounts of scopolamine (Hashimoto et al., 1993). Such approaches would
enhance the utility of hairy roots to produce novel compounds. Most importantly, these
compounds are produced at shorter periods of time than in intact plants since the growth cycle
in vitro is shorter than in vivo conditions.

7. Production of foreign proteins in transgenic plants


Recently, transgenic plants are considered to be economically competitive production
systems for the manufacture of variety of foreign proteins. These include recombinant
antibodies and antibody fragments (James and Lee, 2001; Sharp and Doran, 2001; Doran,
2000; Smith and Glick, 2000; Wongsamuth and Doran, 1997; Tavladoraki et al., 1993; Hein
et al., 1991; Hiatt et al., 1989), enzymes such as b-glucuronidase (Kurata et al., 1998) and
invertase (Verdelhan des Molles et al., 1999) and proteins of therapeutic value such as human
interleukin (IL)-2 and IL-4 (Magnuson et al., 1998), ribosome-inactivating proteins (Remi
shih et al., 1998; Francisco et al., 1997), ricin (Sehnke and Ferl, 1999) and human
a1-antitrypsin (Terashima et al., 1999a,b). The most commonly used host species for protein
synthesis in suspension cultures is tobacco (Sharp and Doran, 1999; Liu and Lee, 1999;
Kurata et al., 1998; Wongsamuth and Doran, 1997), although rice cell suspension cultures

118

S. Ramachandra Rao, G.A. Ravishankar / Biotechnology Advances 20 (2002) 101153

have also been used (Terashima et al., 1999a,b). Hiatt et al. (1989) estimated that plantderived antibodies are likely to cost less than the ones derived from hybridoma cells and that
plant cell media are composed of simple sugars and salts less complex than mammalian
media. Consequently, purification of secreted protein is simpler and more economical. Plant
cell-derived proteins are likely to be safer than those derived from other systems, since plant
cell pathogens are not harmful to humans. The expression of these heterologous proteins in
plants has an advantage over their expression in the microbial system, since full-length
antibodies have been produced in plants (De Wilde et al., 1996). On the other hand,
Escherichia coli produced only a monovalent Fab fragment, the largest antibody found in
a microbe (Smith, 1996).
Hairy roots of tobacco (N. tabacum) were used to produce full-length murine IgG1
monoclonal antibody and the maximum production of 18 mg/l was reported in 21-day grown
shake flasks, and up to 14% of the antibody was secreted into the medium. The decline in
antibody production was found after 14 days of cultivation and mainly due to its degradation.
Antibody production by transgenic hairy roots has a negligible effect on growth compared
with hairy roots of wild-type tobacco (Wongsamuth and Doran, 1997). The loss of protein
due to product instability during plant cell culture and subsequent purification significantly
influences the product yields due to occurrence of unfavourable environmental conditions.
Protein stability following secretion improved with the addition of appropriate chemical
stabilizers. Increased recovery of a mouse monoclonal antibody heavy chain by the addition
of DMSO, gelatin and polyvinylpyrrolidone (Magnuson et al., 1998; Wongsamuth and
Doran, 1997) was reported. The other new protein stabilizer, bacitracin, enhanced the cell
growth (Sharp and Doran, 1999) and inhibit the degradation of plant-derived proteins
(Bateman et al., 1997).
Full-length antibodies were first expressed by Hiatt et al. (1989). They have expressed a
monoclonal antibody 6D4 from a mouse hybridoma cell line in tobacco. The k and g
chains of the IgG molecule was expressed with or without mammalian leader sequence.
The transgenic lines upon crossing produced plants with expression of both the chains.
However, the plants without leader sequence did not show assembled gk complexes and
hence lacked functional characteristics. The use of plant-derived endoplasmic reticulum
(ER) targeting sequence has been studied by During et al. (1990), with demonstration of
accumulation of antibody in ER of transgenic tobacco. De Wilde et al. (1996) found that
both full-length IgG molecules and Fab fragments accumulated in the intercellular spaces of
mesophyll cells in leaves of the transgenic Arabidopsis thaliana. Their study with
immunolocalization confirms the production of full-length antibody and its secretion by
plant cells. Moreover, in plants, single cells are able to assemble secretory antibodies,
whereas two different cell types are required in mammals. Ma et al. (1995) have obtained
transgenic N. tabacum that expressed a murine monoclonal antibody k chain, a hybrid
IgAIgG heavy chain, a murine joining in chain and a rabbit secretory component. These
chains were assembled into functional, high-molecular-weight secretory immunoglobulins
that recognized the native Streptococcal antigen I/II, a surface adhesion molecule. Plant
cells also possess the requisite mechanism for the assembly and expression of other
complex recombinant protein molecules. The ability to produce monoclonal antibodies in

S. Ramachandra Rao, G.A. Ravishankar / Biotechnology Advances 20 (2002) 101153

119

plants also led to the opportunity to develop an inexpensive method of mucosal


immunoprotection against genital herpes (Zetlin et al., 1998). The expression of the
production of a full-length functional antibody in algal forms has also been reported
(Conrad and Fiedler, 1994). Smith (1996) states that the folding of full-size immunoglobulins in algae is possibly due to its less reducing cytoplasm. This cytoplasm contains
chaperonins, which facilitate folding.
Ma et al. (1994) have studied the prospects of using plant-produced antibodies in passive
immunization against the cell surface antigen of Streptomyces mutans, which cause tooth
decay. The fact that plant-derived antibodies have been approved for clinical trials (Ma and
Hein, 1995) is an encouraging development. However, R&D of large-scale antibody
production in plants should be pursued by developing systems of higher expression using
easy and efficient downstream processing and the large-scale cultivation of transgenic plants
in controlled greenhouses to drive this exciting research area into the realm of application.
The storage of antibodies for extended periods can be made possible by expressing them in
seeds (Fiedler and Conrad, 1995).
7.1. Vaccines
The ability to express genes encoding the antigens of bacterial and viral pathogens in
plants, with retention of immunogenic properties, is bound to revolutionize vaccine
production. The concept of edible vaccine stemmed from the landmark discovery made by
Mason et al. (1992). Plant systems will be a choice, which may be relatively less expensive
for production as well as storage of antigens.
Strategies for candidate vaccine production are of two types: (1) stable genomic
integration with foreign DNA introduced either by Agrobacterium T-DNA vectors or by
microprojectile bombardment and (2) transient expression using viral vectors (Mason and
Arntzen, 1995). The first system affords stable transformation, which may be heritable.
Furthermore, the expression could be targeted to specific loci using appropriate promoters.
The second system involves the use of viral vectors for transient expression in plants, a
potentially useful means of producing high levels of recombinant antigens. Foreign protein
expression could be achieved either by foreign gene transcription using a subgenomic
promoter or the fusion of foreign proteins to peptides, which contain the capsid protein
that normally coats the virus (Mason and Arntzen, 1995). High levels of a-trichosanthin,
an antiviral protein in transformed plants (Kumagai et al., 1993), have been obtained by
the foreign gene transcription method. Turpen et al. (1995) have developed a method of
expressing the coat promoter of the tobacco mosaic virus (TMV) using peptides of
epitopes derived from malarial sporozoites. The antigenicity measured by ELISA and
Western blot demonstrate that the recombinant coat protein recognized appropriate
monoclonal, antimalarial antibodies. Similarly, Cowpea mosaic virus has also been used
for the expression of foreign peptides such as antigens from the rabies virus and HIV-1
(Yusibov et al., 1997). Stable genomic transformation encoding foreign antigens has been
demonstrated in case of hepatitis B surface antigens as well as E. coli and Vibrio cholerae
(Thanavalla et al., 1995).

120

S. Ramachandra Rao, G.A. Ravishankar / Biotechnology Advances 20 (2002) 101153

One can produce edible vaccines by cloning the genes of fruit plants and expressing the
vaccines in the edible parts of the fruit. However, it is necessary to address the problem
such as oral tolerance (Mahon et al., 1998) and the standardization of dosage requirement
for the scale of protection. These topics need to be studied along with ecological
considerations on a case-by-case basis to get approval for each vaccine production and
administration to target groups.

8. Immobilization of plant cells for the production of secondary metabolites


Improvement in the secondary metabolite production of cell cultures is often associated
with the organization and differentiation of plant cells. The concept of organization and
differentiation led to the use of immobilization technology, which has long been used for
microbes and enzymes. Immobilization is defined as a technique, which confines a
catalytically active enzyme or cells on a fixed support and prevents its entry into liquid
phase (Yeoman et al., 1990; Yeoman, 1987; Fowler, 1986; Lindsey and Yeoman, 1985,
1983a,b). Immobilized plant cells have been used for single and multistep biotransformations of precursors to desired products as well as for the de novo biosynthesis of
secondary metabolites.
Ever since, Brodelius et al. (1979) described the entrapment of viable cells of Cath. roseus,
M. citrifolia and Di. lanata in calcium alginate gel and this technique has received much
attention. Extensive reviews on various aspects of plant cell immobilization are available
(Williams and Mavituna, 1992; Payne et al., 1991; Scragg, 1991; Hulst and Tramper, 1989;
Brodelius 1988a; Hall et al., 1988; Lindsey and Yeoman, 1987).
Immobilization of plant cells has distinct advantages as biocatalyst over the immobilized
enzyme system. It is necessary to provide the immobilized enzyme with proper pH, the flow
of reaction mixture temperature and as supply of cofactors. Immobilized enzymes are
generally applied to single-step reactions. Furthermore, there will be loss in activity during
isolation of enzymes from the organism. The advantage of immobilized enzyme is the high
rate of activity. In contrast to immobilized enzymes, immobilized cells have distinct
advantages: (a) it can carry out multienzyme operations; (b) by selecting highly biosynthetic
cells, catalytic activity can be enhanced; (c) there is no need to provide cofactors since cells
themselves produce them and (d) immobilized cells can be easily handled as compared to
immobilized enzymes. Thus, immobilized cells are gaining much importance as biocatalysts.
The following prerequisites are essential to adopt immobilization for secondary metabolite
production (Payne et al., 1991).
The product of interest should not be strictly growth associated.
Growth of cells should be suppressed to prevent disintegration of the immobilization
matrix, which may lead to disruption of the process.
Immobilized cells should maintain prolonged viability and biosynthetic capacity with high
rates of sustainable secondary metabolite production.
The product should leach out of the cells and beads into the medium.

S. Ramachandra Rao, G.A. Ravishankar / Biotechnology Advances 20 (2002) 101153

121

The most widely used technique involves the entrapment of cells, in some kind of gel or
combination of gels, which are allowed to polymerize around them (Novais, 1988).
Calcium alginate is the most widely used matrix. Besides this, other gels such as agar,
agarose, gelatin, carrageenan and polyacrylamide have also been used (Nilsson et al.,
1983). However, gels of alginate are most widely used because of their simplicity and
relatively lack of toxicity. The other alternative supports are polyurethane foam and hollowfibre membranes.
Table 11 gives a number of examples of the systems of immobilization, which have been
used with plant cells together with the associated plant species and their products.

Table 11
Immobilized plant cell systems used for production of secondary metabolites
Plant species

Substrate/precursor Product

Immobilization method Reference

Bioconversions
Cath. roseus
Di. lanata
D. carota
N. tabacum
P. somniferum

Cathenamine
Digitoxin
Digitoxigenin
Keto esters
Codeinone

Ajmalicine
Digoxin
Periplogenin
Hydroxy esters
Codeine

Agarose
Alginate
Alginate
Alginate
Polyurethane foam

L-Tyrosine
Ferulic acid,
vanillylamine

L-DOPA
Vanillin,
capsaicin

Alginate
Alginate

Felix et al. (1981)


Brodelius et al. (1979)
Jones and Veliky (1981)
Naoshima and Akakabe (1989)
Furuya et al. (1984)
Furusaki et al. (1988)
Wichers et al. (1983)
Johnson et al. (1996)

Ajmalicine

Alginate, agarose

Brodelius et al. (1979)

Capsaicin

Polyurethane foam

Brodelius and Nilsson (1980)


Lindsey and Yeoman (1983b)
Lindsey and Yeoman (1984)

Caffeine
Caffeoyl
putrescine

Membrane
Alginate

Lang et al. (1990)


Berlin et al. (1989)

Capsaicin
Ajmalicine
Phenolics
Pigments
Diosgenin
Berberine
Thiophenes
Anthraquinones

Polyurethane foam
Alginate, agarose
Hollow fibres
Polyurethane foam
Polyurethane foam
Alginate
Alginate
Alginate

Johnson (1993)
Brodelius and Nilsson (1980)
Prenosil and Pedersen (1983)
Lindsey and Yeoman (1984)
Ishida (1988)
Kobayashi et al. (1987)
Ketel et al. (1987)
Brodelius et al. (1979)

Muc. pruriens
Capsicum spp.

Synthesis from precursors


Cath. roseus
Tryptamine,
secologanin
Ca. frutescens
Isocapric acid,
vanillylamine,
valine and
ferulic acid
Coffea arabica
N. tabacum

Theobromine
Phenylalanine

De novo synthesis
Ca. frutescens
Cath. roseus
Glycine max
Lavandula vera
Diosc. deltoidea
Th. minus
Tagetes patula
M. citrifolia

122

S. Ramachandra Rao, G.A. Ravishankar / Biotechnology Advances 20 (2002) 101153

Table 12
Dramatic effects of immobilization on secondary metabolite production in plant cell cultures
Plant species

Product

Fold change

Ca. frutescens
Ca. frutescens
Tag. patula
Coffea arabica
Cath. roseus
Cath. roseus

Capsaicin
Capsaicin
Thiophenes
Methylxanthin
Ajamlicine
Total alkaloids

Muc. pruriens

L-DOPA

Salvia miltiorrhiza

Cryptotanshinone

> 100 (I)


> 100 (I)
Ca 20 (I)
13 (I)
3.5 (I)
No de novo
synthesis
No de novo
synthesis
2.5 (D)

Type of
immobilization

Reference

Foam
Gel
Natural glass
Gel
Gel
Membrane

Lindsey and Yeoman (1984)


Ravishankar et al. (1988)
Hulst and Tramper (1989)
Haldimann and Brodelius (1987)
Asada and Shuler (1989)
Payne et al. (1988)

Gel

Wichers et al. (1983)

Gel

Miyasaka et al. (1986)

(I): increase, (D): decrease. Adapted from Payne et al. (1991).

Immobilization can have a dramatic impact on cellular physiology and secondary product
formation. The more dramatic responses are summarized in Table 12.

9. Biotransformations for the production of secondary metabolites


Biotransformation can be defined as a process through which the functional groups of
organic compounds are modified either stereo- or regiospecifically by living cultures,
entrapped enzymes or permeabilized cells to a chemically different product.
The production of high-value food metabolites, fine chemicals and pharmaceuticals can
be achieved by biotransformations using biological catalysts in the form of enzymes and
whole cells (Ravishankar and Ramachandra Rao, 2000; Krings and Berger, 1998; Meyer
et al., 1997; Scragg, 1997; Berger, 1995; Cheetham, 1995). The range of flavour metabolites and pharmaceuticals produced by plant cell cultures through biotransformation is
shown in Table 13.
Cell suspension cultures, immobilized cells, enzyme preparations and hairy root cultures
can be applied for the production of food additives or pharmaceuticals by biotransformation
process (Table 14). There are two main reasons to choose plant cells for biotransformation
purposes. Firstly, these cells are generally able to catalyze the reactions stereospecifically,
resulting in chirally pure products. Secondly, they can perform regiospecific modifications
that are not easily carried out by chemical synthesis or by microorganisms (Hamada and
Furuya, 1996; Pras et al., 1995; Stockigt, 1993; Pras, 1992; Stepan-Sarkissan, 1991). These
reactions include reduction, oxidation, hydroxylation, acetylation, esterification, glucosylation, isomerization, methylation, demethylation, epoxidation, etc. The presence of biotransformation potential in plant cells is a necessary condition for practical application.
However, for a successful and viable process, the following prerequisites must be met
(Steck and Constabel, 1974).

S. Ramachandra Rao, G.A. Ravishankar / Biotechnology Advances 20 (2002) 101153

123

Table 13
Biotransformation of flavour compounds by plant cell culture systems
Plant species

Substrate

Product

Reference

Citrus limon
Citrus paradisi
N. tabacum
Mentha spp.

Valencene
Valencene
Linalool
Pulegone
Menthol
Steviol
Geranial
Neral
Citronellal
Ferulic acid

Nootkatone
Nootkatone
8-hydroxylinalool
Isomenthone
Neomenthol
Stevioside
Geraniol
Nerol
Citronellol
Vanillin

Drawert et al. (1984)


Drawert et al. (1984)
Suga and Hirata (1990)
Aviv and Galun (1978)
Aviv et al. (1981)
Furuya (1978)
Lappin et al. (1987)

G. jasminoides

Ferulic acid
Isoeugenol
and eugenol
Acetophenone

Vanillin
Isoeugenyl b-rutinoside and
eugenyl b-glucoside
(S)-aromatic alcohol

Curcuma zedoaria
Gl. glabra
Coffea arabica
Papaver bracteatum

Germacrone
Glycyrrhitinic acid
Vanillin
Linalyl acetate

Achillea millefolium
N. tabacum

Borneol, menthol
thymol and farnesol
Carvone

Guaiane-type sesquiterpenes
Hydroxyglycoside esters
Vanillin glucosides
Linalool, a-terpineol
Geraniol
Many products

Cath. roseus
N. tabacum
Cath. roseus
Cath. roseus
Coffea arabica
Euc. perriniana
Euc. perriniana
Euc. perriniana
Cymbidium spp.

Piperitone
Pulegone
Geraniol
Glycyrrhizin
Capsaicin
Menthol
Camphor
Borneol
Menthyl acetate

Ste. rebaudiana
Lavandula angustifolia

Va. planifolia
Ca. frutescens
Eucalyptus perriniana

Dihydrocarvone
neodihydrocareol
Hydroxypiperitone
Menthone 4-hydroxymenthone
10-hydroxygeraniol
Glycyrrhetic acid
Capsaicin glucoside
Menthol 3-O-b-gentiobiosides
Camphor glucosides
( ) Borneol b-gentiobioside
Menthol

Romagnoli and Knorr


(1988)
Johnson et al. (1996)
Orihara et al. (1992)
Akakabe and Naoshima
(1993)
Sakui et al. (1992)
Hayashi et al. (1992)
Kometani et al. (1993a)
Hook et al. (1990)
Figueiredo et al. (1996)
Hirata et al. (1982)
Hamada and Furuya (1996)
Hamada and Furuya (1996)
Hamada and Furuya (1996)
Hamada and Nakata (1992)
Kometani et al. (1993b)
Orihara et al. (1991)
Orihara et al. (1994)
Orihara and Furuya (1993)
Mironowicz et al. (1987)

Adapted from Ramachandra Rao (1998).

(1) The culture must have the necessary enzymes. (2) The substrate or precursor must not
be toxic to the culture. (3) The substrate must reach the cellular compartment of the cell. (4)
The rate of product formation must be faster than its further metabolism.
9.1. Advantages of biotransformations
The advantages include the production of novel compounds, enhancement in the
productivity of desired compound and overcoming the problems associated with chemical
synthesis. Importantly, the studies on biotransformation lead to basic information to elucidate

124

S. Ramachandra Rao, G.A. Ravishankar / Biotechnology Advances 20 (2002) 101153

Table 14
Biotransformations using plant cell cultures for production of pharmaceuticals
Plant species

Substrate

Product

Reference

P. somniferum
Di. purpurea
Di. lanata
Euc. perriniana
Podo. hexandrum
Muc. pruriens
Cath. roseus

Codeine
Digoxin
Digoxin
Taxol derivatives
Podophyllotoxin
DOPA
Vincrisitne

Wilhelm and Zenk (1997)


Alfermann et al. (1980)
Alfermann et al. (1980)
Hamada et al. (1996)
Van Uden et al. (1995)
Pras et al. (1993)
DiCosmo and Misawa (1995)

Capsaicin, vanillin
Capsaicin, vanillin

Johnson et al. (1996)

Ca. frutescens

Thebaine
Digitoxin
Digitoxin
Taxol
Coniferyl alcohol
Tyrosine
Vindoline
Vinblastine
Ferulic acid
Vanillylamine
Protocatechuic aldehyde
Caffeic acid
Digitoxin

Spirulina platensis
Ca. frutescens

Codeine
Isoeugenol

Ca. frutescens
Ca. frutescens

Digoxin,
purpureaglycoside A
Morphine
Vanillin, capsaicin

Ramachandra Rao and


Ravishankar (2000a)
Ramachandra Rao et al.
(2002)
Ramachandra Rao et al. (1999)
Ramachandra Rao and
Ravishankar (1999)

the biosynthetic pathway, and catalysis can be carried out under mild conditions, thus
reducing undesired by-products, energy, safety and costs.

10. Scale-up of plant cell cultures


Since plant cells produce unique pharmaceuticals, which can be harnessed, they need to be
produced in large-scale bioreactors. Configuration of bioreactors used for microbial cells
cannot always be utilized directly for plant cells, owing to distinctive features, which are not
favourable for plant cell cultivation. Plant cells are less stable in productivity, highly shear
sensitive, exhibit low oxygen requirements (ca. 1-mmol O2 at 10 6 cells) slow growth
(doubling time 25110 h) and often occur as cell clumps of 24-mm diameter. Different
Table 15
Bioreactor types used for plant cell cultures
Reactor type

Plant species

Reference

Stirred tank

Cath. roseus
N. tabacum
N. tabacum
M. citrifolia
L. erythrorhizon
Cath. roseus
Cath. roseus
Ruta vulgaris

Drapeau et al. (1986)


Kato et al. (1977)
Noguchi et al. (1977)
Wagner and Vogelmann (1977)
Tanaka (1987)
Morris et al. (1984)
Shuler et al. (1983)
Shuler and Hallsby (1985)

Bubble column
Airlift stirred
Aerated
Fluidized bed
Tubular membrane
Disc turbine

S. Ramachandra Rao, G.A. Ravishankar / Biotechnology Advances 20 (2002) 101153

125

Table 16
Large-scale suspension cultures reactors for plant cell cultures
Plant species

Product

Bioreactor capacity

Reference

Cath. roseus
Col. blumei
L. erythrorhizon
N. tabacum

Serpentine
Rosmarinic acid
Shikonin
Biomass
Biomass
Saponins
Biomass
Biomass
Biomass

100 l airlift
300 l airlift
750 l agitated
20,000 l agitated
1500 l bubble column
20,000 l agitated
750 75,000 l agitated
750 75,000 l agitated
750 75,000 l agitated

Smart and Fowler (1981)


Rosevear (1984)
Tabata and Fujita (1985)
Kato et al. (1976)
Noguchi et al. (1977)
Ushiama et al. (1986)
Ritterhaus et al. (1990)
Ritterhaus et al. (1990)
Ritterhaus et al. (1990)

Panax ginseng
E. purpurea
Ra. serpentina
Panax ginseng

Adapted from Sahai (1994).

configuration of bioreactors has been adopted depending on the nature of cells. Few
representative references are made in Table 15. Several studies are available for large-scale
cultivation of plant cells (Table 16).
Cell/tissue types such as cell suspension cultures, immobilized cells and hairy roots
have been very ideal for scale-up. However, the configuration of reactors will be different
for different cell or tissue types. No one design could be recommended as a common
one. The most important contribution, however, is to produce not only the biomass but
also the metabolite in an economical manner. In the present state of art, any compound
less than US$ 1000 per kilogram is difficult to produce. Hence, it is advantageous
to utilize bioreactor technology for compounds, which are costlier and has a higher
market demand.
10.1. Scale-up of biotransformations
So far, a few reports are available on scale-up of biotransformation. In the food area, the
development of a process for making high-fructose corn syrup by immobilized glucose
isomerase was reported. In the pharmaceutical industry, the first biotransformation was the
production of modified penicillin by immobilized penicillin acylase using single-step
biotransformation (West, 1996). Biotransformation of 12b-hydroxylation of methyldigitoxin
to methyldigoxin and other metabolites such as deacetayllanatoside C by Di. lanata cell
Table 17
Biotransformation of b-methyldigitoxin to b-methyldigoxin by Di. lanata cells in 20-l reactor
Precursor

Productivity (g)

% Conversion

b-methyldigitoxin added
Unconverted b-methyldigitoxin
b-methyldigoxin formed
By-product
Yield

17.24
2.04
14.36
0.28

100
11.8
81.7
1.4
94.9

Adapted from DiCosmo and Misawa (1995).

126

S. Ramachandra Rao, G.A. Ravishankar / Biotechnology Advances 20 (2002) 101153

cultures was reported (Kreis and Reinhard, 1992; 1990; Reinhard et al., 1989). A
biotransformation process for the production of digoxin was developed using Di. lanata
in cell suspension cultures using 20- and 300-l airlift bioreactors. The results of the study
indicated that maximum digoxin was reached in 8% (w/v) glucose medium, and about 80%
of the digoxin produced was found in the medium (Table 17). Panda et al. (1992)
demonstrated the alkaloid production by precursor feeding using stirred tank bioreactors
in plant cell cultures of Holarrhena antidysentrica.

11. Food additive production from plant cell cultures studied in the authors laboratory
11.1. Capsaicin
Capsaicin (Fig. 3) is a pungent principle of green pepper (Capsicum spp.). Capsaicin
preparations are used as a counterirritant in lumbago, neuralgia and rheumatic disorders.
Taken internally, Capsicum preparation has a tonic and carminative action especially useful
in dyspepsia. Capsicum oleoresin is added to tannin or rose gargles for pharyngitis and
relaxed sore throat. Capsaicin is administered as a bacteriostatic (Gal, 1965) or fungistatic
compound in the form of powder, tincture, liniment, plaster, ointment and medicated wool. In
the US, two commercial creams, viz. Zostrix and Axsain, are formulated with purified
capsaicin for alleviation of arthritis pain (Anonymous, 1991). Commercial production of
capsaicin by separation from Capsicum oleoresin and its subsequent purification would
involve several steps.
Capsaicin can be produced by immobilized cell cultures, which leach out to the medium,
facilitating easy separation and purification. The cultivation of the Capsicum crop takes 45
months and therefore cannot offer a continuous production procedure. In contrast, immobilized cell cultures of Capsicum offer a continuous method of producing capsaicin under in
vitro controlled conditions. There are several reports of capsaicin production in immobilized
cell cultures (Johnson et al., 1990, 1991; Ravishankar et al., 1988; Mavituna et al., 1987;
Lindsey and Yeoman, 1984). Our attempts to enhance the level of capsaicin by treating
immobilized cell cultures of Ca. frutescens with elicitors (Johnson et al., 1990) and
permeability agents (Johnson et al., 1991) or by manipulating culture conditions (Johnson,
1993) have been successful. An innovative method of capsaicin production by immobilizing
the placenta, the site of synthesis of capsaicin (Iwai et al., 1979), was reported by Johnson and
Ravishankar (1996). Capsaicin production increased several folds higher in immobilized

Fig. 3. Chemical structure of capsaicin.

S. Ramachandra Rao, G.A. Ravishankar / Biotechnology Advances 20 (2002) 101153

127

placenta than in chilly fruit. With precursor biotransformation using coumaric acid, it
registered an 11-fold increase (Johnson and Ravishankar, 1996).
Here, we provide capsaicin production in 2-l airlift culture vessel using immobilized cells/
placenta. An airlift culture vessel of cylindrical shape was constructed with Corning glass of
the configuration given in Fig. 4.
11.2. Inputs of operation
1. 20-day-old Capsicum cells to placenta (100-g fresh weight).
2. Alginate and calcium chloride solution required to envelop 100-g cells/placenta in
beads. One litre of 2.5% (w/v) sodium alginate with cells extruded into 2 l of 0.9% (w/v)
calcium chloride dihydrate.
3. Beads washed with water were transferred into the vessel containing 1 l of MS
medium supplemented with 3% sucrose, 2-mg/l 2,4-D and 0.5-mg/l kinetin [standard
medium (SM)].
4. Airflow (2:1 mixture of CO2 + air for the initial 7 days of culture and 4:1 for the latter
7 days of production) at a rate of 4 V.V.M.
5. Incubation at 25 2 C under continuous light of 2000 lx.
6. pH adjustment to 5.8 during culture.
7. Replenishment of entire medium after 7 days with fresh medium.
8. Capsaicin recovery and analysis in 2 weeks of culture with two harvests at 7-day intervals.

Fig. 4. Schematic representation of column reactor process for capsaicin production using immobilized cell
cultures of Ca. frutescens.

128

S. Ramachandra Rao, G.A. Ravishankar / Biotechnology Advances 20 (2002) 101153

11.3. Yield components


The airlift culture vessel was run with immobilized cells or placenta for 2 weeks under
three different conditions using (1) SM, (2) SM + elicitor or (3) SM + precursor ( p-coumaric
acid). The results are presented below.
11.3.1. Yields in SM
1. Immobilized cells: 35 mg/100-mg fresh cells/15 days or 0.4% on dry weight basis
2. Immobilized placenta: 300 mg/100-g fresh placenta/15 days or 0.1% on dry
weight basis
11.3.2. Yields in SM+elicitor
Elicitors used were curdlan at 8-mg/l concentration for immobilized cells and Rhizopus
oligosporus mycelial extract equivalent to 2.5 g of mycelium/l of medium administered for
placental immobilized cultures.
1. Immobilized cells: 76 mg/100-g fresh cells/15 days or 0.95% on dry weight basis
2. Immobilized placenta: 300 mg/100-g fresh placenta/15 days or 1.25% on dry
weight basis
11.3.3. Yields in SM+precursor (2.5-mM coumaric acid)
1. Immobilized cells: not done
2. Immobilized placenta: 1148 mg/100-g fresh placenta/15 days or 7.6% on dry
weight basis
By our method, we are able to get a maximum production of 1.15 g/l. However, increases
can be obtained using continuous cultivation with adsorption of capsaicin and recycling of
medium to minimize the cost. The plant may not be viable if it is less than 10,000-l capacity.
We have recently studied capsaicin production in bubble column and packed-bed reactor at
1-l level, which needs further scale-up and its performance to be evaluated vis-a`-vis alginate
immobilized system (Table 18).
Apart from the production of capsaicin from immobilized cell cultures of Ca. frutescens,
attempts have been made in our laboratory to produce vanilla flavour metabolites from
Capsicum-immobilized cultures upon feeding of phenylpropanoid intermediates protocatechuic aldehyde, caffeic acid (Ramachandra Rao and Ravishankar, 2000a), ferulic acid,
coniferyl aldehyde and veratraldehyde (Ramchandra Rao and Ravishankar, 1998). The
feeding of phenylpropanoid precursors not only produced the vanilla flavour metabolites
but also increased the yields of capsaicin (Ramachandra Rao, 1998; Ramachandra Rao and
Ravishankar, 2000a). Feeding of clove principle, isoeugenol, to immobilized cultures of Ca.
frutescens showed formation of vanilla flavour metabolites (Ramachandra Rao and
Ravishankar, 1999).

S. Ramachandra Rao, G.A. Ravishankar / Biotechnology Advances 20 (2002) 101153

129

Table 18
Comparison of continuous production of capsaicin from bubble column and packed-bed reactor
Bioreactor

Productivity

Bubble column
Biomass density
Average productivity
Average specific productivity
Maximum duration of continuous operation
Total production
Packed-bed reactor
Biomass density
Average productivity
Average specific productivity
Maximum duration of continuous operation
Total production
Production under fungal elicitation

100 g fresh weight/l


1.7 mg/l/day
17 mg/g fresh weight/day
21 days
35 mg/l in 21 days
380 g/l
7.4 mg/l/day
19.5 mg/g fresh weight/day
28 days
207 mg/l in 28 days
330 mg/l in 28 days

Adapted from Madhusudhan (1998).

11.4. Vanillin
Vanillin (Fig. 5) is the most universally accepted aroma chemical used in processed foods,
pharmaceutical products and perfumeries (Prince and Gunson, 1994; Anonymous, 1993;
Webster, 1995). It occurs in the vanilla bean at a level of 2% dry weight, and it is associated
with many other compounds. Approximately 12,000 tons of vanillin is consumed annually,
from which only 20 tons are extracted from the vanilla beans (Dornenburg and Knorr, 1996;
Feron et al., 1996; Berger, 1995). The cost of the pure natural vanillin flavour is priced at US$
4000 per kg, while the synthetic equivalent costs about 12 US$ per kg (Krings and Berger,
1998; Berger, 1995). Other than flavour qualities, use of vanillin as antimicrobial against molds
and yeast has been described (Lopez-Malo et al., 1995; Cerrutti et al., 1997; Cerrutti and
Alzamora, 1996). Vanillin was found to be an antioxidant (Burri et al., 1989) and will be useful
in the development of health foods. Vanillin is also known for its antimutagenic activity
(Kometani et al., 1993a) and glucosylvanillin has been found to impart distinctive flavour note,
hence would be useful in tonics and syrups. Interestingly, at the 8th FAOBMB Congress,
Iekhsan et al. (1998) reported the discovery of use of vanillin as antidote for neutralizing the
toxic effect of toxin from Chinorex fleckeri (boxjellyfish). There are a number of cases of
poisoning by boxjellyfish, and often, it is fatal to individuals. The findings of Iekhsan et al.
(1998) hence provide newer utility of vanillin as a pharmaceutically important compound.

Fig. 5. Chemical structure of vanillin.

130

S. Ramachandra Rao, G.A. Ravishankar / Biotechnology Advances 20 (2002) 101153

Production of vanillin by conventional method by cultivating Vanilla plants is tedious and


expensive. Though vanilla flavour has its own utility as flavouring, the use of vanillin
obtained through biotechnology assumes significance since there is worldwide demand for
natural compounds. In this context, vanillin and its other flavour metabolites are being
produced through biotransformation to provide alternate method of production of biovanillin
through cheaper substrates (Table 19). The increase in production of vanillin and its other
flavour metabolites has been made possible by several factors using optimization by
enhanced substrate availability mediated by cyclodextrin (Ramachandra Rao and Ravisha-

Table 19
Biotransformations leading to vanillin formation through microbial and plant cell cultures
Organism bioconverting the substrate
Ferulic acid
Aspergillus niger
Pycnoporous cinnabarinus
Pseudomonas fluorescens
Pseudomonas fluorescens AN 103
Corynebacterium glutamicum
Ca. frutescens
Spir. platensis
Haemotococcus pluvialis
Va. planifolia
Va. planifolia
Eugenol
Corynebacterium glutamicum
Pseudomomnas spp.
Arthrobacter globiformis
Spir. platensis
Isoeugenol
Enterobacter spp.
Serratia spp.
Spir. platensis
Ca. frutescens
Vanillylamine
Asper. niger
E. coli
Spir. platensis
Ca. frutescens
Coniferyl aldehyde
Ca. frutescens
Spir. platensis
Coniferyl aldehyde
Vanillyl alcohol
Ca. frutescens
Spir. platensis
Penicillum simplicissimum
Adapted from Ravishankar and Ramachandra Rao (2000).

Reference
Lesage-Messen et al. (1996)
Andreoni et al. (1995)
Narbad and Gasson (1998)
Labuda et al. (1993)
Ramachandra Rao (1998)
Ramachandra Rao et al. (1996a)
Usha et al. (1999)
Romagnoli and Knorr (1988)
Funk and Brodelius (1990)
Tadasa and Kayahara (1983)
Rabenhorst (1996)
Cooper (1987)
Ramachandra Rao et al. (1996a)
Rabenhorst (1991)
Ramachandra Rao et al. (1996a)
Ramachandra Rao and Ravishankar (1999)
Yoshida et al. (1997)
Ramachandra Rao (1998)
Sudhakar Johnson et al. (1996)
Ramachandra Rao (1998)
Ramachandra Rao et al. (1996a)
Markus et al. (1992)
Ramachandra Rao and Ravishankar (1998)
Ramachandra Rao et al. (1996a)
Fraaije et al. (1997)

S. Ramachandra Rao, G.A. Ravishankar / Biotechnology Advances 20 (2002) 101153

131

nakar, 1998), elicitation of bioconversion process using microbial elicitors (Ramachandra


Rao, 1998) and in situ adsorption of products using adsorbents, viz. activated charcoal (Knuth
and Sahai 1991; Westcott et al., 1994) and amberlite XAD-4 and XAD-7 (Ramachandra Rao
and Ravishankar, 2000b), to overcome the feedback inhibition, thereby enhancing yield and
recovery. The formation of vanilla flavour metabolites from various phenylpropanoids in cell
cultures of C. frutescens is shown in Fig. 6
11.5. Anthocyanin
Anthocyanins are the most ubiquitous pigments seen in nature, widely distributed in the
pericarps of several fruits, flowers and vegetables. They are glycosylated polyhydroxy and
polymethoxy derivatives of flavylium (2-phenylbenzopyrrolium) salts belonging to the group

Fig. 6. Vanilla flavour metabolites formation from various phenylpropanoid precursors in Ca. frutescens
cell cultures.

132

S. Ramachandra Rao, G.A. Ravishankar / Biotechnology Advances 20 (2002) 101153

Table 20
Effect of antioxidants on lipid peroxidation
Compound

IC50

Butylated hydroxyanisole
Butylated hydroxytoluene
a-tocopherol
Anthocyanin from carrot cell cultures

0.125
0.250
0.500
0.005

Adapted from Narayan et al. (1999).

of two or three portions the aglycone base, a group of sugars and a group of acyl acids. The
aglycone moiety is referred to as anthocyanidin. There are more than 15 anthocyanidins
(Francis, 1982). Anthocyanins are widely used a colourants. However, they are known to
have several pharmacological attributes such as antiinflammatory, antiulcer and woundhealing properties (Koichi and Hasashi, 1990; Vega et al., 1987). We have recently
established the antioxidant properties of anthocyanin obtained through cell culture (Narayan
et al., 1999). Culture-derived anthocyanin effectively prevented autooxidation of lipids as
well as lipid peroxidation in biological systems (Table 20). Our studies have shown high
anthocyanin production in mutated cell cultures of D. carota (Table 21). Chromatographic
analyses of anthocyanin extract showed the presence of cyanidin xylosyl galactose, cyanidin
monoglucose and cyanidin galactose in the molar ratio of 5:3:2. The chemical structure of
cyanidine-3-glucoside and delphinidin-3-glucoside is shown in Fig. 7.
Presently, the commercial source of anthocyanins is the grape peel in view of its quoted
price of US$ 12001500 per kilogram (Ilker, 1987) and estimated market of US$ 200
million. Plant cell cultures are an alternative source of anthocyanin. The possibility of
obtaining novel anthocyanin with two to three times greater stability at slightly acidic pH has
been demonstrated using D. carota cells (Vunsh et al., 1986) Research in our laboratory has
shown that anthocyanin production in carrot cultures is stimulated by indole-3-acetic acid
Table 21
Growth and production of anthocyanin in D. carota cell cultures
Parameters
Growth
Specific growth rate
Doubling time
Growth index
Maximum cell density
Dry weight/fresh weight ratio
Packed cell volume
Anthocyanin production
Average content
Specific productivity ( qp)
Total production
Average productivity
Adapted from Madhusudhan (1998).

Value
0.217/day
3.2 days
12.2
18.92 1.17 g/l
0.085
0.95 ml/g fresh weight
15.58% (w/w) dry mass
10.52 mg/g/day
3.1 g/l in 15 days
207 mg/l/day

S. Ramachandra Rao, G.A. Ravishankar / Biotechnology Advances 20 (2002) 101153

133

Fig. 7. Chemical structure of anthocyanin.

(Rajendran et al., 1992). Elicitation of anthocyanin by fungal elicitor has been useful to
enhance the yields by 1.25-fold using Aspergillus flavus mycelial extract (Rajendran et al.,
1994). Similarly, its elicitation by phycocyanin was also reported (Ramachandra Rao et al.,
1996a,b). The cell aggregate size of 500850 mm was critical for maximum anthocyanin
production (Madhusudhan and Ravishankar, 1996).
The high yields obtained by several researchers have resulted in exciting possibility of
scale-up of anthocyanin production systems. In the future, anthocyanins will be used for
antioxidant effect as well as their value as colours. Developments in genetic engineering
of anthocyanin production have been elegantly reviewed by Guttason (1994). There is a
possibility of obtaining plants with flowers of different hues. This aspect may be
adopted for cell culture production. However, to date, no enhancement of hue has
been demonstrated.

12. Metabolic engineering of biochemical pathways


It is envisaged that the use of plant cell culture approach for pharmaceutically important
compounds can be made industrially applicable only upon the overproduction of metabolites
or the production of novel compounds that too with the expression of more than one
compounds of interest in a plant. This is not realized by routine cell line selection and other
parameters of growth/production media development. Therefore, there is a need to study the
metabolic pathways and its regulatory steps for overexpressing the regulatory genes for ratelimiting steps. The recent advancement in plant cell and tissue culture techniques and the
genetic transformation of plants using Agrobacterium-mediated or direct gene transfer make
possible the incorporation of foreign genes into plants producing new genotypes of desirable
traits. Thus, genetically engineered or modified plants can be used as green bioreactors for
the production of value-added chemicals (Saito et al., 1992).
The progress of genetic manipulation largely depends on (a) the ability to obtain genes
encoding desirable traits, (b) the proper alignment of the expressed gene with its endogenous
substrate and the compartmentation/transport of its product, (c) the successful transformation
and reproducible regeneration of transgenic plants, (d) the proper site of gene integration
combined with the level and patterns of gene expression and (e) the evaluation of the altered
metabolic pathways.

134

S. Ramachandra Rao, G.A. Ravishankar / Biotechnology Advances 20 (2002) 101153

12.1. Molecular strategies


Strategies for overproduction of metabolites by genetic manipulation can be as follows.

1.
2.
3.
4.
5.

Overproduction of precursors of secondary metabolites (e.g. A).


Overexpression of the gene product, which limits the specific pathway (e.g. B).
Creating a new branch for a preexisting pathway (e.g. F to C).
Down-regulation of existing reaction using antisense methods (blocking, e.g. C to E).
Manipulation of regulatory genes whose products may bind DNA and function as
transcriptional activators or repressors.
6. Selection of regulatory mutants with increased tissue-independent expression for in
vitro production of metabolites.
7. Sometimes, the tissue/organ-specific expression by using appropriate promoters is
required to target the metabolite production. This would result in metabolic energy
saving by localized production of metabolites.
Studies related to the importance of plants by metabolic engineering or transgenic
protocols are presented here, where success has been obtained for enhancement of yields.
12.2. Tryptophan decarboxylase (EC 4.1.1.28)
Tryptophan decarboxylase catalyses the decarboxylation of L-tryptophan to tryptamine.
Tryptamine is a substrate for strictosidine synthase (Stockigt and Zenk, 1977). Interestingly,
cDNA clone from Cath. roseus encoding tryptophan decarboxylase has been expressed in
tobacco plants (Songstad et al., 1991), which resulted in the increased levels of tryptamine
and tyramine as predicted. This study has been extended to metabolically engineer Brassica
napus, wherein tryptophan, which is generally bioconverted to glucosinolate (an undesirable
product of oil seed cake, which renders the product unpalatable by cattle), is diverted away by
expressing tryptophan decarboxylase gene of Cath. roseus. In such a situation, transgenic B.
napus produced low amount of glucosinolates, making the oil seed cake palatable to cattle
(Chavadej et al., 1994).
12.3. Strictosidine synthase (EC 4.3.3.2)
Strictosidine synthase catalyzes the stereospecific condensation of primary amino group
of tryptamine and aldehyde group of iridoid glycoside secologanin to form the indole

S. Ramachandra Rao, G.A. Ravishankar / Biotechnology Advances 20 (2002) 101153

135

alkaloid strictosidine. Strictosidine synthase is of biotechnological interest in the biomimetic


synthesis of monoterpenoid indole alkaloids because the reaction product is a mixture of
the diastereomers.
The cDNA encoding strictosidine synthase was isolated from Rauwolfia serpentina and it
has been functionally expressed in E. coli, Saccharomyces cerevisiae and insect cells using a
baculovirus vector (Kutchan, 1989; Kutchan et al., 1988). Strictosidine synthase from Cath.
roseus has been expressed in tobacco (McNight et al., 1990).
12.4. H6H (EC 1.14.11.11)
The biosynthesis of scopolamine from hyoscyamine is catalyzed by H6H. This enzyme
catalyzed hydroxylation and epoxidation reactions (Robbins et al., 1994).
The cDNA encoding H6H from Hy. niger was introduced into Atropa belladona using
either Agrobacterium tumefaciens- or Agr. rhizogenes-mediated transformation. The resultant
Atropa transgenic plants contained high levels of scopolamine (Yun et al., 1992). This is a
first example of metabolic engineering to produce medicinally useful compound.
12.5. Berberine bridge enzyme (EC 1.5.3.9)
Berberine bridge enzyme catalyses the stereospecific conversion of N-methyl group
(S)-reticuline into (S)-scoulerine (Kutchan, 1995). Direct conversion of N-methyl group into
a methylene bridge moiety is not easily achievable presently by synthetic chemistry. It is of
interest to note that berberine bridge enzyme is an elicitor-induced yeast cell wall (Schumacher et al., 1987), thereby providing possibility of overproduction of berberine in cell
cultures. Moreover, cDNA encoding the berberine bridge enzyme has been isolated by
Dittrich and Kutchan (1991) in elicited cell suspensions of Es. californica (California poppy)
and expressed heterologously into insect cultures using baculovirus expression vector, which
resulted in the production of sufficient quantities.
12.6. Berbamunine synthase
Berbamunine synthase (EC 1.1.3.34) catalyzes the formation of the ether linkage between
one molecule of (R)-N-methylcoclaurine and one molecule of (S)-N-methylcoclaurine to form
the bisbenzylisoquinoline alkaloid berbamunine. The cDNA encoding berbamurine synthase
was isolated from B. stolonifera cell suspension cultures and expressed in insect culture using
a baculovirus expression vector (Kraus and Kutchan, 1995).
12.7. Polyphenol oxidase (PPO)
PPO is ubiquitous in higher plants and its regulation provides several advantages. Downregulation of PPO increases the quality attributes of crop plant-derived commodities,
whereas overexpression reduces danger from pests. It has been concluded that no phenotypic

136

S. Ramachandra Rao, G.A. Ravishankar / Biotechnology Advances 20 (2002) 101153

effects are visible in transgenic tomato or tobacco plants with high or low PPO levels
(Steffens et al., 1994).
12.8. p-Hydroxycinnamoyl-CoA hydratase/lyase (HCHL)
Higher plants possess the phenylpropanoid pathway for the production of lignin and some
secondary phenolic compounds. Coumaric acid and ferulic acids and their CoA esters are
intermediates in these routes. Expression of the bacterial gene for the side chain cleavage
enzyme in plants may offer the potential to generate vanillin and related flavour compounds
in a range of crops by diversion of phenylpropanoid metabolism. At IFR, attempts have been
made to produce vanillin in hairy root cultures of Dat. stramonium by expressing the HCHL
gene from Pseudomonas fluorescens AN103. The expression of the HCHL gene in Dat.
stramonium hairy root cultures caused substantial changes in the content of p-hydroxybenzoic acid in the form of glucosides and glucose esters and along with traces of glucosides of
p-hydroxybenzyl alcohol and vanillic acid. The potential of this line to biotransform added
ferulic acid to vanillin is being studied (Mitra et al., 1999).
12.9. 4-coumarate-CoA ligase (4CL)
4CL (EC 6.2.1.12) catalyses a branch-point reaction (conversion of hydroxycinnamic acids
into the corresponding CoA esters) between the central phenylpropanoid pathway and
pathways leading to various secondary metabolites such as flavonoids, lignans, lignin and
related phenylpropanoid compounds (Holton and Cornish, 1995; Douglas, 1996). Brodelius
and Xue (1997) isolated and characterized 4CL from Va. planifolia cell suspension cultures.
Based on studies using the inhibitors of 4CL (Funk and Brodelius, 1990), they concluded that
the expression of antisense mRNA for 4CL gene would redirect the flow of phenylpropanoid
precursors from lignin biosynthesis into flavour compound biosynthesis in Va. planifolia.
Thus, genetic engineering for metabolic alteration or overexpression of pathway has much
to offer. There are exciting possibilities to produce high-value, low-volume pharmaceutical
products such as the ones described here. Moreover, one can imaginatively produce novel
compounds by expression of foreign genes. The questions regarding environmental study of
transgenic plants and regulatory considerations of administration of the phytopharmaceuticals
derived from them has not been dealt since it is beyond the scope of this review.

13. Conclusion
Though plant cell cultures can produce a whole range of secondary metabolites, there are
few success stories of production at commercial scale. The prospect of production of highcost, low-volume products such as anti-HIV and anti-cancer compounds is very high, thus
putting this technology to a position of being able to make a commercial impact in a few
selected pharmaceuticals. The advancement of knowledge in phytochemistry, regulation of
secondary pathways and ability to express desired traits by transgenics is expected to drive

S. Ramachandra Rao, G.A. Ravishankar / Biotechnology Advances 20 (2002) 101153

137

technology to produce a range of pharmaceutical and healthcare products. Vaccines and


antibodies from plants are already fast-advancing fields, with investments from multinational
companies. They aim to provide protection against diseases of all sections of society, since
the poor have been deprived of expensive immunization schedules.
Use of transgenic plants for the production of pharmaceuticals will be bound by the IPR
protection. Those who can invest in the R&D will benefit in the era of processes and patents.
Developing countries, which are still behind in recombinant DNA research, may not be able
to develop their own complete technologies. Nations rich in biodiversity and knowledge base
of their local vegetation may protect their plants and develop cultivation methods. The
technique of micropropagation by in vitro culture methods will be a handy tool, which does
not need high levels of biotechnology. The R&D of production of secondary metabolites
through biotechnology has made a beginning and is expected to provide disease-remedial or
disease-preventive molecules at affordable costs for the benefit of mankind.

Acknowledgments
The authors would like to thank the Council of Scientific and Industrial Research (CSIR),
Department of Science and Technology (DST) and the Department of Biotechnology (DBT),
Government of India, New Delhi for sponsoring the research grants on plant cell cultures for
secondary metabolites. They also like to thank B. Suresh for his help in the preparation of
this review.

References
Akakabe Y, Naoshima Y. Mechanistic pathway of biotransformation of acetophenone by immobilized cell cultures
of Gardenia. Phytochemicals 1993;32:1189 91.
Akita M, Shigeoka T, Kozumi Y, Kuwamura M. Mass propagation of shoots of Stevia rebaudiana using a large
scale bioreactor. Plant Cell Rep 1994;13:180 3.
Alfermann AW, Petersen M. Natural products formation by plant cell biotechnology. Plant Cell Tissue Org Cult
1995;43:199 205.
Alfermann AW, Schuller I, Reinhard E. Biotransformation of cardiac glycosides by immobilized cell cultures of
Digitalis lanata L. Planta Med 1980;40:218 23.
Ambid C, Fallot J. Role of the gaseous environment on volatile compound production by fruit cell suspension
cultured in vitro. In: Schreier P, editor. Flavour 81. Berlin: de Gruyter, 1981. pp. 529 38.
Ambros PF, Matzke AJM, Matzyke MA. Localization of Agrobacterium rhizogenes T-DNA in plant chromosomes
by in situ hybridization. EMBO J 1986;5:2073 7.
Amrhein N, Johanning D, Smart CC. A glyphosate-tolerant plant tissue cultures. In: Neumann KH, Barz W,
Reinhard E, editors. Primary and secondary metabolism of plant cell cultures. Berlin: Springer-Verlag, 1985.
pp. 355 61.
Andreoni A, Bernasconi S, Besetti G. Biotransformation of ferulic acid and related compounds by mutant strains
of Pseudomonas fluorescens. Appl Microbiol Biotechnol 1995;42:830 5.
Anonymous. Pain killing chilli. The Week. 1991 p. 57 (Sep 8).
Anonymous. Vanilla the queen of spices. H&R Contact 1993;59:15 9.

138

S. Ramachandra Rao, G.A. Ravishankar / Biotechnology Advances 20 (2002) 101153

Asada M, Shuler ML. Stimulation of ajmalicine production and excretion from Catharanthus roseus: effects of
adsorption in situ, elicitors, and alginate immobilization. Appl Microbiol Biotechnol 1989;30:475 81.
Asada Y, Saito H, Yoshikawa T, Sakamoto K, Furuya T. Biotransformation of 18b-glycyrrhetinic acid by ginseng
hairy root culture. Phytochemicals 1993;34:1049 52.
Asada Y, Li W, Yoshikawa T. Isoprenylated flavonoids from hairy root cultures of Glycyrrhiza glabra. Phytochemicals 1998;47:389 92.
Aviv D, Galun E. Biotransformation of monoterpenes by Mentha cell lines: conversion of pulegone to isomenthone. Planta Med 1978;33:70 7.
Aviv D, Krochmal E, Dantes E, Galun E. Biotransformation of monoterpenes by Mentha cell lines: conversion of
menthone to neomenthol. Planta Med 1981;42:236 43.
Bais HP, George J, Ravishankar GA. Production of esculin by hairy root cultures of Cichorium intybus L. CV.
Lucknow Local. Indian J Exp Biol 1999;37:269 73.
Balandrin MJ, Klocke JA. Medicinal, aromatic and industrial materials from plants. In: Bajaj YPJ, editor.
Biotechnology in agriculture and forestry. Medicinal and aromatic plant, vol. 4. Berlin: Springer-Verlag,
1988. pp. 1 36.
Banthrope DV. Secondary metabolism in plant tissue culture: scope and limitations. J Nat Prod Rep 1994;11:
303 28.
Barz W, Daniel S, Hinderer W, Jaques U, Kessmann H, Koster J, Tiemann K. Elicitation and metabolism of
phytoalexins in plant cell cultures. In: Pais M, Mavituna F, Novais J, editors. Plant cell biotechnology. NATO
ASI Series. Berlin: Springer-Verlag, 1988. pp. 211 30.
Bateman KS, Congiu M, Tregear GW, Clarke AE, Anderson MA. Bacitracin significantly reduces degradation of
peptides in plant cell cultures. Biotechnol Bioeng 1997;53:226 31.
Beaumont MD, Knorr D. Effects of immobilizing agents and procedures on viability of cultured celery (Apium
graveolens) cells. Biotechnol Lett 1987;9:377 82.
Beiderbeck R, Knoop B. Two-phase culture. In: Constable F, Vasil I, editors. Cell culture and somatic cell genetics
of plants, vol. 4. San Diego: Academic Press, 1987. pp. 255 66.
Beiderbeck R, Knoop B. Enhanced production of secondary substances: addition of artificial accumulation sites to
cultures. In: Bajaj YPS, editor. Biotechnology in agriculture and forestry. Medicinal and aromatic plants, vol. 4.
Berlin: Springer-Verlag, 1988. pp. 123 35.
Benjamin BD, Roja G, Heble MR, Chadha MS. Multiple shoot cultures of Atropa belladona: effects of physicochemical factors on growth and alkaloid formation. J Plant Physiol 1987;129:129 35.
Benjamin BD, Roja G, Heble MR. Alkaloid synthesis by root cultures of Rauwolfia serpentina transformed by
Agrobacterium rhizogenes. Phytochemicals 1994;35:381 3.
Berger RG. Aroma biotechnology. Berlin: Springer-Verlag, 1995.
Berlin J. Para-flurophenylalanine resistant cell lines of tobacco. Zeit-fur pflan Physiol 1980;97:317 24.
Berlin J, Forche E, Wray V, Hammer J, Hosel W. Formation of benzophenanthiridine alkaloids by suspension
cultures of Eschscholtzia californica. Z Naturforsch 1983;38:346 52.
Berlin J, Sieg S, Strack D, Bokern M, Harns H. Production of betalaines by suspension cultures of Chenopodium
rubrum L. Plant Cell Tissue Org Cult 1986;5:163 74.
Berlin J, Martin B, Nowak J, Witte L, Wray V, Strack D. Effects of permeabilization on the biotransformation of
phenylalanine by immobilized tobacco cell cultures. Z Naturforsch 1989;44c:249 54.
Bohm H, Rink E. Betalaines. In: Constabel F, Vasil I, editors. Cell culture and somatic cell genetics of plants,
vol. 5. New York: Academic Press, 1988. pp. 449 63.
Breuling M, Alfermann AW, Reinhard E. Cultivation of cell cultures of Berberis wisoniae in 20 L airlift bioreactors. Plant Cell Rep 1985;4:220 3.
Brevoort P. The blooming U.S. botanical market: a new overview. Herbalgram 1998;44:33 46.
Brodelius P. Immobilization of plant cells as a source of biochemicals. In: Moo-Young M, editor. Bioreactor
immobilized enzymes and cells. Essex: Elsevier, 1988a. pp. 167 96.
Brodelius P. Permeabilization of plant cells for release of intracellularly stored products: viability studies. Appl
Microbiol Biotechnol 1988b;27:561 6.

S. Ramachandra Rao, G.A. Ravishankar / Biotechnology Advances 20 (2002) 101153

139

Brodelius P, Nilsson K. Entrapment of plant cells in different matrices. FEBS Lett 1980;122:312 6.
Brodelius P, Nilsson K. Permeabilization of immobilized plant cells resulting in release of intracellularly stored
products with preserved cell viability. Eur J Appl Microbiol Biotechnol 1983;17:275 80.
Brodelius PE, Xue ZT. Isolation and characterization of a cDNA from cell suspension cultures of Vanilla
planifolia encoding 4-coumarate: coenzyme A ligase. Plant Physiol Biochem 1997;35:497 506.
Brodelius P, Deus B, Mosbach K, Zenk MH. Immobilized plant cells for the production of natural products. FEBS
Lett 1979;103:93 7.
Brodelius P, Funk C, Shillito RD. Permeabilization of cultured plant cells by electroporation for the release of
intracellularly stored secondary products. Plant Cell Rep 1988;7:186 8.
Buitelaar RM, Tramper J. Strategies to improve the production of secondary metabolites with plant cell cultures: a
literature review. J Biotechnol 1992;23:111 43.
Burri J, Graf M, Lambelet P, Loeliger J. Vanillin: more than a flavouring agent a potent antioxidant. J Sci Food
Agric 1989;48:49 56.
Cerrutti P, Alzamora SM. Inhibitory effects of vanillin on some food spoilage yeasts in laboratory media and fruit
purees. Int J Food Microbiol 1996;29:379 86.
Cerrutti P, Alzamora SM, Vidales SL. Vanillin as an antimicrobial for producing shelf-stable strawberry puree.
J Food Sci 1997;62:608 10.
Charlwood BV, Moustou C. Essential oil accumulation in shoot-proliferation cultures of Pelargonium spp. In:
Robins RJ, Rhodes MJC, editors. Manipulating secondary metabolism in culture. Cambridge: Cambridge
Univ. Press, 1988. pp. 187 94.
Chavadej S, Brisson N, McNeil JN, de Luca V. Redirection of tryptophan leads to production of low indole
glucosinolate Conola. Proc Natl Acad Sci USA 1994;91:2166 70.
Cheetham PSJ. Biotransformations: new routes to food ingredients. Chem Ind. 1995 pp. 265 8 (April).
Chung IS, Kang YM, Oh JM, Kim T, Lee HJ, Chae YA. Continuous suspended cultures of Mentha piperata in cell
recycled airlift bioreactor. Biotechnol Tech 1994;8:789 92.
Collin HA, Masker D. Allium compounds. In: Constabel F, Vasil I, editors. Cell culture and somatic cell genetics
of plants, vol. 5. San Diego: Academic Press, 1988. pp. 475 93.
Conrad U, Fiedler U. Expression of engineered antibodies in plant cells. Plant Mol Biol 1994;26:1023 30.
Constabel F. Medicinal plant biotechnology. Planta Med 1990;56:421 5.
Cooper B. Method and microorganisms for the preparation of coniferyl aldehyde. DE 3 604874 A1, 1987.
Courtois D, Guren J. Temperature response of Catharanthus roseus cells cultivated in liquid medium. Plant Sci
Lett 1980;17:473 82.
Cox PA, Balick MJ. The ethanobotanical approach to drug discovery. Sci Am. 1994 pp. 82 7 (June).
Cresswell R. The production of flavour components from citrus tissue cultures. Abstracts from the 7th
International Congress of Plant Cell and Tissue Culture. International Association for Plant Cell Culture,
Amsterdam, 1990.
Deus NBS, Zenk MH. Exploitation of plant cells for the production of alkaloids in Catharanthus roseus cell
suspension cultures. Planta Med 1982;50:427 31.
De Wilde C, De Neve M, De Rycke R, Bruyns A-M, De Larger G, Van Montagu M, Depicker A, Engler G. Intact
antigen-binding MAK33 antibody and Fab fragment accumulate on intercellular spaces of Arabidopsis thaliana. Plant Sci 1996;114:133 41.
DiCosmo F, Misawa M. Plant cell and tissue culture: alternatives for metabolite production. Biotechnol Adv
1995;13:425 35.
DiCosmo F, Tallevi SG. Plant cell cultures and microbial insult: interactions with biotechnological potential.
Trends Biotechnol 1985;3:110 1.
DiCosmo F, Towers GHN. Stress and secondary metabolism in cultured plant cells. In: Timmerman BN, Steelink
FA, Loewus FA, editors. Recent advances in phytochemistry, vol. 18. New York: Plenum, 1984. pp. 97 175.
DiCosmo F, Quesne A, Misawa M, Tallevi SG. Increased synthesis of ajmalicine and catharanthine by cell
suspension cultures of Catharanthus roseus in response to fungal culture filtrates. Appl Microbiol Biotechnol
1987;14:101 6.

140

S. Ramachandra Rao, G.A. Ravishankar / Biotechnology Advances 20 (2002) 101153

Dittrich H, Kutchan TM. Molecular expression and induction of berberine bridge enzyme, an enzyme essential to
the formation of benzophenanthridine alkaloids in the response of plants to pathogenic attack. Proc Natl Acad
Sci USA 1991;88:9963 73.
Dixon RA. Plant Natural products: the molecular genetic basis of biosynthetic diversity. Curr Opin Biotechnol
1999;10:192 7.
Do CB, Cormier F. Accumulation of anthocyanins enhanced by a high osmotic potential in grape (Vitis vinifera L.)
cell suspensions. Plant Cell Rep 1990;9:143 6.
Doran PM. Foreign protein production in plant tissue cultures. Curr Opin Biotechnol 2000;11:199 204.
Dornenburg H, Knorr D. Cellular permeabilization of cultured tissues by high electric field pulses or ultra high
pressure for the recovery of secondary metabolites. Food Biotechnol 1993;7:35 48.
Dornenburg H, Knorr D. Production of the phenolic flavour compounds with cultured cells and tissues of Vanilla
planifolia species. Food Biotechnol 1996;10:75 92.
Dornenburg H, Knorr D. Challenges and opportunities for metabolite production from plant cell and tissue
cultures. Food Technol 1997;51:47, 48, 50 52, 54.
Dougall DK. Nutrition and metabolism. In: Staba E, editor. Plant tissue culture as a source of biochemicals. Boca
Raton, FL: CRC Press, 1980. pp. 21 58.
Douglas CJ. Phenylpropanoid metabolism and lignin biosynthesis: from weeds to trees. Trends Plant Sci
1996;1:171 8.
Drapeau D, Blanch HW, Wilke CR. Growth kinetics of Dioscorea deltoidea and Catharanthus roseus in batch
culture. Biotechnol Bioeng 1986;28:1555 63.
Drawert F, Berger RG, Godelmann R. Regioselective biotransformation of valencene in cell suspension cultures of
Citrus spp. Plant Cell Rep 1984;3:37 40.
During K, Hippe S, Kreuzaler F, Schell J. Synthesis and self assembly of a functional monoclonal antibody in
transgenic Nicotiana tabacum. Plant Mol Biol 1990;15:281 93.
Eastburn SD, Tao BY. Applications of modified cyclodextrins. Biotechnol Adv 1994;12:335 79.
Eilert U. Elicitation: methodology and aspects of application. In: Constabel F, Vasil I, editors. Cell culture and
somatic cell genetics of plants, vol. 4. San Diego: Academic Press, 1987. pp. 153 96.
Ellis BE, Towers GHN. Biogenesis of rosmarinic acid in Mentha. J Biochem 1970;118:291 7.
Endress R. Plant cell biotechnology. Berlin: Springer-Verlag, 1994.
Ernst D. Pimpinella anisum L. (Anise). Cell culture somatic embryogenesis and the production of anise oil. In:
Bajaj YPS, editor. Biotechnology in agriculture and forestry, vol. 1. Berlin: Springer, 1989. p. 381.
Farnsworth NR. The role of medicinal plants in drug development. In: Kroogsgard-Larsen P, Brogger Christenses
S, editors. Natural products and drug development. Denmark: Munsgaard Copenhagen, 1985. pp. 17 30.
Felix HR. Permeabilized cells. Anal Biochem 1982;120:211 34.
Felix H, Brodelius P, Mosbach K. Enzyme activities of the primary and secondary metabolism of simultaneously
permeabilized and immobilized plant cells. Anal Biochem 1981;116:462 72.
Feron G, Bonnarme P, Durand A. Prospects for the microbial production of food flavours. Trends Food Sci
Technol 1996;7:285 93.
Fett-Neto AG, Melanson SJ, Sakata K, DiCosmo F. Improved growth and taxol yield in developing callus of
Taxus cuspidata by medium composition modification. Bio/Technology 1993;11:731 4.
Fett-Neto AG, Melanson SJ, Nicholson SA, Pennington JJ, DiCosmo F. Improved taxol yield by aromatic
carboxylic and amino acid feeding to cell cultures of Taxus cuspidata. Biotechnol Bioeng 1994;44:
967 71.
Fiedler U, Conrad U. High level production and long-term storage of engineered antibodies in transgenic tobacco
seeds. Bio/Technology 1995;13:1090 3.
Figueiredo AC, Almendra MJ, Barroso JG, Scheffer JJC. Biotransformation of monoterpenes and sesquitepenes
by cell suspension cultures of Achillea millefolium L. sp. Millefolium. Biotechnol Lett 1996;18:863 8.
Flores HE, Hoy MW, Puckard JJ. Secondary metabolites from root cultures. Trends Biotechnol 1987;5:64 9.
Flores HE, Dai Y-R, Freyer AJ, Michaels PJ. Biotransformation of butylated hydroxytoluene in hairy root cultures.
Plant Physiol Biochem 1994;32:511 9.

S. Ramachandra Rao, G.A. Ravishankar / Biotechnology Advances 20 (2002) 101153

141

Flores HE, Vivanco JM, Loyola-Vorgas M. Radicle biochemistry: the biology of root-specific metabolism. Trends
Plant Sci 1999;4:220 6.
Fosket D, Radin D. Induction of carotegenesis in cultured cells of Lycopersicon esculentum. Plant Sci Lett
1983;30:165 75.
Fowler MW. Process strategies for plant cell cultures. Trends Biotechnol 1986;4:214 8.
Fowler MW, Scragg AH. Natural products from higher plants and plant cell culture. In: Pais MSS, Mavituna
F, Novais JM, editors. Plant cell biotechnology. NATO ASI Series, vol. 18. Berlin: Springer-Verlag, 1988.
pp. 165 77.
Fowler MW, Stafford A. Plant cell culture process systems and product synthesis. In: Flowler MW, Warren GS,
editors. Plant biotechnology. Oxford: Pergamon, 1992. pp. 79 98.
Fraaije MW, Pikkemaat M, Van Berkel WJH. Enigmatic gratituous induction of the covalent flavoprotein vanillyl
alcohol oxidase in Penicillum simplicissimum. Appl Environ Microbiol 1997;63:435 9.
Francis FJ. Analysis of anthocyanins. In: Marakakil M, editor. Anthocyanins as food colours. New York: Academic Press, 1982. pp. 181 207.
Francisco JA, Gawlak SL, Miller M, Bathe J, Russell D, Chace D, Mixan B, Zhao L, Fell HP, Siegall CB.
Characterization of bryodin 1 and bryodin 1-based single chain immunotoxin from tobacco cell culture.
Bioconjugate Chem 1997;8:708 13.
Fujita Y. Industrial production of shikonin and berberine. Applications of plant cell and tissue culture. Ciba
Foundation Symposium 137. Chichester: Wiley, 1988. pp. 228 38.
Fujita Y, Hara Y, Ogino T, Suga C. Production of shikonin derivatives by cell suspension cultures of Lithospermum erythrorhizon: I. Effects of nitrogen sources on the production of shikonin derivatives. Plant Cell Rep
1981;1:59 60.
Fujita Y, Takahashi S, Yamada Y. Selection of cell lines with high productivity of shikonin derivatives through
protoplasts of Lithospermum erythrorhizon. Proc Euro Congr Biotechnol, 3rd 1984;1:161 6.
Funk C, Brodelius P. Phenylpropanoid metabolism in suspension cultures of Vanilla planifolia Andr: II. Effects of
precursor feeding and metabolic inhibitors. Plant Physiol 1990;94:95 101.
Funk C, Gugler R, Brodelius P. Increased secondary metabolite formation in plant cell suspension cultures after
treatment with a yeast carbohydrate preparation (elicitor). Phytochemicals 1987;26:401 5.
Furusaki S, Nozawa T, Isohara T, Furuya T. Influence of substrate transport on the activity of immobilized
Papaver somniferum cells. Appl Microbiol Biotechnol 1988;29:437 41.
Furuya T. Biotransformation by plant cell cultures. In: Thorpe TA, editor. Frontiers of plant tissue cultures.
Calgary: The International Association for Plant Tissue Culture, 1978. pp. 191 200.
Furuya T, Yoshikawa T, Taira M. Biotransformation of codeinone to codeine by immobilized cells of Papaver
somniferum. Phytochemicals 1984;23:999 1001.
Furuya T, Ushiyama M, Asada Y, Yoshikawa T. Biotransformation of 2-phenylpropionic acid in root culture of
Panax ginseng. Phytochemicals 1989;28:483 7.
Furze JM, Rhodes MJC, Parr AJ, Robins RJ, Whithehead IM, Threlfall DR. Abiotic factors elicit sesquiterpenoid
phytoalexin production but not alkaloid production in transformed root cultures of Datura stramonium. Plant
Cell Rep 1991;10:111 4.
Gal IF. Neuorc angaben uber Capscidin. Experentia 1965;21:383.
George PS, Ravishankar GA. Induction of crocin and crocetins in callus cultures of Gardenia jasminoides Ellis.
Food Biotechnol 1995;9:29 38.
Glaser V. Billion-dollar market blossoms as botanicals take root. Nat Biotechnol 1999;17:17 8.
Guo YG, Li HJ, Cai YX, Zhong Y, Peng ZY. Studies on the production of secondary metabolites in plant cell
cultures. In: Furusaki S, Endo I, Matsuno R, editors. Biochemical engineering for 2001. Tokyo: SpringerVerlag, 1992. pp. 242 5.
Guttason NC. The biologists palette: genetic engineering of anthocyanin biosynthesis and flower colour. In: Ellis
BE, Kuroki GW, Stafford HA, editors. Genetic engineering of plant secondary metabolism. New York:
Plenum, 1994. pp. 93 124.
Haggin J. Cyclodextrin research focuses on variety of applications. Chem Eng News 1992;70:25 6.

142

S. Ramachandra Rao, G.A. Ravishankar / Biotechnology Advances 20 (2002) 101153

Hagimori M, Matsumoto T, Obi Y. Studies on the production of Digitalis cardenolides by plant tissue cultures.
Plant Cell Physiol 1982a;23:1205 11.
Hagimori M, Matsumoto T, Obi Y. Studies on the production of Digitalis purpurea cardenolides by plant tissue
culture. Plant Physiol 1982b;69:653 6.
Haldimann D, Brodelius P. Redirecting cellular metabolism of cultured plant cells: a model study with Coffea
arabica. Phytochemicals 1987;26:1431 4.
Hall RD, Holden MA, Yeoman MM. In: Bajaj YPS, editor. Immobilization of plant cells. Berlin: Springer-Verlag,
1988. pp. 136 56.
Hamada H, Furuya T. Recent advances in plant biotransformations. Plant Tissue Cult Biotechnol 1996;2:
52 9.
Hamada H, Nakata S. Biotransformation of glycyrrhizin by buffered cell suspension cultures of Catharanthus
roseus. Plant Tissue Cult Lett 1992;9:32 3.
Hamada H, Sanada K, Furuya T, Kawabe S, Jaziri M. Nat Prod Lett 1996;9:47.
Hamill JD, Parr AJ, Robins RJ, Rhodes MJC. Secondary product formation by cultures of Beta vulgaris and
Nicotiana rustica transformed with Agrobacterium rhizogenes. Plant Cell Rep 1986;5:111 4.
Hamill JD, Parr AJ, Rhodes MJC, Robins RJ, Walton NJ. New routes to plant secondary products. Biotechnology
1987;5:800 4.
Hashimoto T, Yamada Y. Tropane alkaloid production in Hyoscyamus root cultures. J Plant Physiol 1986;124:
61 75.
Hashimoto T, Yamada Y. Scopolamine production in suspension cultures and redifferentiated roots of Hyoscyamus
niger. J Med Plant Res 1983;47:195 9.
Hashimoto T, Yun D-Z, Yamada Y. Production of tropane alkaloids in genetically engineered root cultures.
Phytochemicals 1993;32:713 8.
Hay CA, Anderson LA, Roberts MF, Phillipson JD. In vitro cultures of Cinchona species. Plant Cell Rep
1986;5:1 4.
Hayashi H, Fukui H, Tabata M. Examination of triterpenoids produced by callus and cell suspension cultures of
Glycyrrhiza glabra. Plant Cell Rep 1988;7:508 11.
Hayashi H, Yamada K, Fukui H, Tabata M. Examination of triterpenoids produced by callus and cell suspension
cultures of Glycyrrhiza glabra. Phytochemicals 1992;3:2729 33.
Heble MR. Multiple shoot cultures: a viable alternative in vitro system for the production of known and new
biologically active plant constituents. In: Neumann K-H, Barz W, Reinhard E, editors. Primary and secondary
metabolism of plant cell cultures. Berlin: Springer-Verlag, 1985. pp. 281 5.
Hein MB, Tang Y, McLeod DA, Janda KD, Hiatt A. Evaluation of immunoglobulins from plant cells. Biotechnol
Prog 1991;7:455 61.
Hiatt A, Cafferkey R, Bowdish K. Production of antibodies in transgenic plants. Nature 1989;342:76 8.
Hippolyte I, Marin B, Baccou JC, Jonard R. Growth and rosmarinic acid production in cell suspension cultures of
Salvia officinalis L. Plant Cell Rep 1992;11:109 12.
Hirata T, Hamada H, Aoki T, Suga T. Stereoselectivity of the reduction of carvone and dihydrocarvone by
suspension cells of Nicotiana tabacum. Phytochemicals 1982;21:2209 12.
Hirata K, Yamanaka A, Kurano N, Miyamoto K, Miura Y. Production of indole alkaloids in multiple shoot
cultures of Catharanthus roseus (L.) G. Don. Agric Biol Chem 1987;51:1311 7.
Holton TA, Cornish EC. Genetics and biochemistry of anthocyanin biosynthesis. Plant Cell 1995;7:1071 83.
Hook I, Lecky R, Mckenna B, Sheridan H. Biotransformation of linalyl acetate by suspension cultures of Papaver
bracteatum. Phytochemicals 1990;29:2143 4.
Hu BZ, Alfermann AW. Diterpenoid production in hairy root cultures of Saliva miltiorrhiza. Phytochemicals
1993;32:699 703.
Hulst AC, Tramper J. Immobilized plant cells: a literature survey. Enzyme Microb Technol 1989;11:546 58.
Husemann W, Callies R, Leibfritz D. External pH modifies the intracellular pH and the mode of photosynthetic
CO2 assimilation in photoautotrophic cell suspension cultures of Chenopodium rubrum L. Bot Acta 1992;
105:116.

S. Ramachandra Rao, G.A. Ravishankar / Biotechnology Advances 20 (2002) 101153

143

Ibrahim RK. Regulation of synthesis of phenolics. In: Constabel F, Vasil I, editors. Cell culture and somatic cell
genetics of plants, vol. 4. San Diego: Academic Press, 1987. pp. 77 95.
Iekhsan O, Jaya V, Mustaffa MA, Rais MM. Antitoxin compounds from the plant extracts of Ipomea pes-caprae
(L.) R. Br. In: 8th FAOBMB Congress. Kuala Lumpur, 1998. p. 20 (Abstract volume).
Ikeda T, Matsumoto T, Noguchi M. Effects of inorganic nitrogen source and physical factors on the formation of
ubiquinone by tobacco plant cells in suspension culture. Agric Biol Chem 1977;41:1197 201.
Ilker R. In vitro pigment production an alternative to colour synthesis. Food Technol 1987;41:70 2.
Ishida B. Improved diosgenin production in Dioscorea deltoidea cell cultures by immobilized in polyurethane
foam. Plant Cell Rep 1988;7:270 3.
Iwai K, Suzuki T, Fujiwake H. Formation and accumulation of pungent principle of hot pepper fruits, capsaicin
and its analogues in Capsicum annuum L. var. annuum. cv. Karayatsubusa at different stages of flowering.
Agric Biol Chem 1979;43:2493 8.
James E, Lee JM. The production of foreign proteins from genetically modified plant cells. Adv Biochem Eng/
Biotechnol 2001;72:127 56.
Johnson TS. Studies on production of capsaicin in immobilized cells and placental tissues of Capsicum annuum
and Capsicum frutescens. PhD Thesis, University of Mysore, Mysore, 1993.
Johnson TS, Ravishankar GA. Precursor biotransformation in immobilized placental tissues of Capsicum frutescens Mill: I. Influence of feeding intermediate metabolites of the capsaicinoid pathway on capsaicin and
dihydrocapsaicin accumulation. J Plant Physiol 1996;147:481 5.
Johnson TS, Ravishankar GA, Venkataraman LV. In vivo capsaicin production by immobilized cells and placental
tissues of Capsicum annuum L. grown in liquid medium. Plant Sci 1990;70:223 9.
Johnson TS, Ravishankar GA, Venkataraman LV. Elicitation of capsaicin production in freely suspended and
immobilized cell cultures of Capsicum frutescens Mill. Food Biotechnol 1991;5:197 205.
Johnson TS, Ravishankar GA, Venkataraman LV. Biotransformation of ferulic acid and vanillylamine to capsaicin
and vanillin in immobilized cell cultures of Capsicum frutescens. Plant Cell Tissue Org Cult 1996;44:
117 21.
Jones A, Veliky IA. Effect of medium constituents on the viability of immobilized plant cells. Can J Bot
1981;59:2095 101.
Jung G, Tepfer D. Use of genetic transformation by the T-DNA of Agrobacterium rhizogenes to stimulate biomass
and tropane alkaloid production in Atropa belladona and Calystegia sepium roots grown in vitro. Plant Sci
1987;50:145 51.
Kato A, Kawazoke S, Iizima M, Shimizu Y. Continuous culture of tobacco cells. J Ferment Technol 1976;54:
82 7.
Kato A, Shiozawa Y, Yamada A, Nishida K, Noguchi M. A jar fermenter culture of Nicotiana tabacum L. cell
suspensions. Agric Biol Chem 1977;36:899 904.
Kawaguchi K, Hirotani M, Yoshikawa T, Furuya T. Biotransformation of digitoxigenin in ginseng hairy root
cultures. Phytochemicals 1990;29:837 43.
Kennedy AI, Deans SG, Svoboda KP, Waterman PG. Volatile oils from normal and transformed root of Artemisia
absynthium. Phytochemicals 1993;32:1449 51.
Ketel D, Hulst A, Gruppen H, Breteler H, Tramper J. Effects of immobilization and environmental stress on
growth and production of nonpolar metabolites of Tagetes minuta cells. Enzyme Microb Technol 1987;9:
303 7.
Kieran PM, Malone DM, Macloughlin PF. Variation of aggregate size in plant suspension batch and semi
continuous cultures. Trans Inst Chem Eng 1993;71:40 6.
Kireeva SA, Melinikov VN, Reznikov SA, Meshcheryakova NI. Essential oil accumulation in a peppermint callus
cultures. Fiziol Rast (Moscow) 1978;25:564 70.
Klebnikov A, Dubuis B, Kut OM, Prenosil JE. Growth and production of Beta vulgaris cell culture in fluidized
bed reactors. Bioprocess Eng 1995;14:51 6.
Knobloch KH, Berlin J. Influence of medium composition on the formation of secondary compounds in cell
suspension cultures of Catharanthus roseus L. G. Don. Z Naturforsch 1980;35C:551 6.

144

S. Ramachandra Rao, G.A. Ravishankar / Biotechnology Advances 20 (2002) 101153

Knorr D, Teutonico RA. Chitosan immobilization and permeabilization of Amaranthus tricolor cells. J Agric Food
Chem 1986;34:582 5.
Knorr D, Miazga SM, Teutonica RA. Immobilization and permeabilization of cultured plant cells. Food Technol
1985;39:139 42.
Knuth ME, Sahai OP. Flavour composition and method. United States Patent No. 5,068, 184, 1991.
Ko KS, Ebizyka Y, Noguchi H, Sanakawa U. Secondary metabolites production by hairy roots and regenerated
plants transformed by Ri plasmids. Chem Pharm Bull 1988;36:417 20.
Ko KS, Noguchi H, Ebizyka Y, Sanakawa U. Oligoside production by hairy root cultures transformed by Ri
plasmids. Chem Pharm Bull 1989;37:245 8.
Kobayashi Y, Fukui H, Tabata M. An immobilized cell culture system for berberine production by Thalictrum
minus cells. Plant Cell Rep 1987;6:185 6.
Kobayashi Y, Fukui H, Tabata M. Berberine production by batch and semi-continuous cultures of immobilized
Thalictrum cells in an improved bioreactor. Plant Cell Rep 1988;7:249 53.
Kobayashi Y, Fukui H, Tabata M. Effect of carbon dioxide and ethylene on berberine production and cell
browning in Thalictrum minus cell cultures. Plant Cell Rep 1991;9:496 9.
Koichi N, Hasashi K. Pharmacology of anthocyanosides from wild blue berries. Part 3. Shokuhin Kogyo
1990;33:57 62.
Kometani T, Tanimoto H, Nishimura T, Okada S. Glucosylation of vanillin by cultured plant cells. Biosci,
Biotechnol, Biochem 1993a;57:1290 3.
Kometani T, Tanimoto H, Nishimura T, Kanbara I, Okada S. Glucosylation of capsaicin by cell suspension
cultures of Coffea arabica. Biosci, Biotechnol, Biochem 1993b;57:2192 3.
Konishi T, Konishi K, Takemura T, Matuda N, Konoshima T, Kiyosawa S. Alkaloids from tissue cultures of
Decentra peregrina. Nat Med 1998;52:47 53.
Kraus PFX, Kutchan TM. Molecular cloning and heterologous expression of cDNA encoding berbamunine
synthase, a C O phenol-coupling p-450 from the higher plant Berberis stolonifera. Proc Natl Acad Sci
USA 1995;92:2071 5.
Kreis W, Reinhard E. The production of secondary metabolites by plant cells cultivated in bioreactors. Planta Med
1989;55:409 16.
Kreis W, Reinhard E. Two-stage cultivation of Digitalis lanata cells: semicontinuous production of deacetyllanatoside C in 20 L airlift bioreactors. J Biotechnol 1990;16:123 36.
Kreis W, Reinhard E. 12b-hydroxylation of digitoxin by suspension cultured Digitalis lanata cells: production of
digoxin in 20 L and 300 L airlift reactors. J Biotechnol 1992;26:257 73.
Krings U, Berger RG. Biotechnological production of flavours and fragrances. Appl Microbiol Biotechnol
1998;49:1 8.
Krueger RJ, Carew DP, Lu JHC, Staba EJ. Initiation, maintenance and alkaloid content of Catharanthus roseus
leaf organ cultures. Planta Med 1982;45:56 7.
Kumagai MH, Turpen TH, Weinzettl N, Delta-Cioppa G, Turpen AM, Donson J, Hilf ME, Grantham GL, Dawson
WO, Chow TP, Piatak JRM, Grill LK. Rapid, high-level expression of biologically active a-trichosanthin in
transfected plants by RNA viral vector. Proc Natl Acad Sci USA 1993;90:427 30.
Kunshi M, Shimomura K, Takida M, Kitanaka S. Growth and ginsenoside production of adventitious and hairy
root cultures in an interspecific hybrid ginseng (Panax ginseng  P. quinquefolium). Nat Med 1998;52:1 4.
Kurata H, Takemura T, Furusakai S, Kado CI. Light controlled expression of a foreign gene using the chalcone
synthase promoter in tobacco BY-2 cells. J Ferment Bioeng 1998;86:317 23.
Kutchan TM. Expression of enzymatically active cloned strictosidine synthase from the higher plant Rauwolfia
serpentina in Escherichia coli. FEBS Lett 1989;257:127 30.
Kutchan TM. Alkaloid biosynthesis the basis of metabolic engineering of medicinal plants. Plant Cell
1995;7:1059 70.
Kutchan TM, Hampp N, Lottspeich F, Beyreuther K, Zenk MH. The cDNA clone for strictosidine synthase from
Rauwolfia serpentina: DNA sequence determination and expression in Escherichia coli. FEBS Lett 1988;
257:40 4.

S. Ramachandra Rao, G.A. Ravishankar / Biotechnology Advances 20 (2002) 101153

145

Labuda IM, Keon KA, Goers SK. Microbial conversion process for the production of vanillin. In: Schreier P,
Winterhalter P, editors. Progress in flavour and precursor studies. Carol Stream: Allured Publishers, 1993.
pp. 477 82.
Lang J, Yoon K, Prenosil J. The effects of precursor feeding on alkaloid production in Coffea arabica plant cell
cultures in a novel membrane bioreactor. In: Christiansen C, Mun L, Villadsen J, editors. Proceedings of the
5th European Congress on Biotechnology. Copenhagen: Mundsgaard, 1990. pp. 132 5.
Lappin GJ, Stride JD, Tampion J. Biotransformation of monoterpenoides by suspension cultures of Lavandula
angustifolia. Phytochemicals 1987;26:995 7.
Lesage-Meeseen L, Delttre M, Haon M, Thibaul J-F, Ceccald BC, Brunerie P, Asther M. A two-step bioconversion process for vanillin production from ferulic acid combining Aspergillus niger and Pycnoporus cinnabarinus. J Biotechnol 1996;50:107 13.
Lindsey K, Yeoman MM. Novel experimental systems for studying the production of secondary metabolites by
plant tissue cultures. In: Mantell SH, Smith H, editors. Plant biotechnology. London: Cambridge Univ. Press,
1983a. pp. 39 66.
Lindsey K, Yeoman MM. The relationship between growth rate, differentiation and alkaloid accumulation in cell
cultures. J Exp Bot 1983b;34:1055 65.
Lindsey K, Yeoman MM. The synthetic potential of immobilized cells of Capsicum frutescens Mill. cv. annuum.
Planta 1984;162:495 501.
Lindsey K, Yeoman MM. Immobilized plant cells. In: Yeoman MM, editor. Plant cell culture technology. Berlin:
Springer-Verlag, 1985. pp. 229 67.
Lindsey K, Yeoman MM. Immobilized plant cell culture systems. In: Neumann KH, Barz W, Reinhard E, editors.
Primary and secondary metabolism of plant cell cultures. Berlin: Springer-Verlag, 1987. pp. 304 15.
Liu F, Lee JM. Effect of culture conditions on monoclonal antibody production from genetically modified tobacco
suspension cultures. Biotechnol Bioprocess Eng 1999;4:259 63.
Lopez-Malo A, Alzamora SM, Argaiz A. Effect of natural vanillin on germination time and radial growth of
moulds in fruit-based agar systems. Food Microbiol 1995;12:213 9.
Lui JHC, Staba EJ. Effects of precursors on serially propagated Digitalis lanata leaf and root cultures. Phytochemicals 1979;18:1913 6.
Ma JKC, Hein MB. Plant antibodies for immunotherapy. Plant Physiol 1995;109:341 6.
Ma JKC, Lehner T, Stabila P, Fux CI, Hiatt A. Assembly of monoclonal antibodies with IgGI IgA heavy chain
domains in transgenic tobacco plants. Eur J Immunol 1994;24:131 8.
Ma JKC, Hiatt A, Hein M, Vine ND, Wang F, Stabila P, van Dolleweerd C, Mostov D, Lehner T. Generation and
assembly of secretory antibodies in plants. Science 1995;268:716 9.
Madhusudhan R. Plant cell cultures for the production of phytochemicals anthocyanin and capsaicin in bioreactors. PhD Thesis, University of Mysore, Mysore, 1998.
Madhusudhan R, Ravishankar GA. Gradient of anthocyanin in cell aggregates of Daucus carota in suspension
cultures. Biotechnol Lett 1996;18:1253 6.
Magnuson NS, Linzmaier PM, Reeves R, An G, HayGlass K, Lee JM. Secretion of biologically active human
interleukin-2 and interleukin-4 from genetically modified tobacco cells in suspension culture. Protein Expression Purif 1998;13:45 52.
Mahon BP, Moore A, Johnson PA, Mills KHG. Approaches to new vaccines. Crit Rev Biotechnol 1998;18:
257 82.
Mano Y, Ohkawa H, Yamada Y. Production of tropane alkaloids by hairy root cultures of Duboisia leichhardtii
transformed by Agrobacterium rhizogenes. Plant Sci 1989;59:191 201.
Mantell SH, Smith H. Cultural factors that influence secondary metabolite accumulation in plant cell and tissue
cultures. In: Mantell SH, Smith H, editors. Plant biotechnology. Cambridge: Cambridge Univ. Press, 1984.
pp. 75 108.
Markus PH, Peters ALJ, Roos R. Process for the preparation of phenaldehydes. European Patent, EP 0542348
A2, 1992.
Mason HS, Arntzen CJ. Transgenic plants as vaccine production system. Trends Biotechnol 1995;13:388 92.

146

S. Ramachandra Rao, G.A. Ravishankar / Biotechnology Advances 20 (2002) 101153

Mason HS, Lam DM, Arntzen CJ. Expression of hepatitis B surface antigen in transgenic plants. Proc Natl Acad
Sci USA 1992;89:11745 9.
Matsubara K, Yamada Y, Kitani S, Yoshioka T, Morimoto T, Fujita T. High density culture Coptis japonica cells
increases berberine production. J Chem Technol Biotechnol 1989;46:61 9.
Matsumoto T, Ikeda T, Kanno N, Kisaki T, Noguchi M. Selection of high ubiquinone 10-producing strain of
tobacco cultures by cell cloning technique. Agric Biol Chem 1980;44:967 9.
Mavituna F, Wilkinson AK, Williams PD, Park JMC. Production of secondary metabolites by immobilized plant
cells in novel bioreactors. In: Moody GW, Baker PB, editors. Bioreactors and biotransformations. London:
Elsevier, 1987. pp. 26 37.
McDonald KA, Jackman AP. Bioreactor studies of growth and nutrient utilization in Alfalfa suspension cultures.
Plant Cell Rep 1989;8:455 8.
McKinely TC, Michaels PJ, Flores HE. Is lipoxygenase involved in polyacetylene biosynthesis in Asteraceae?
Plant Physiol Biochem 1993;31:835 53.
McNight TD, Roessner CA, Devagupta R, Scott AI, Nessler CL. Nucleotide sequence of a cDNA encoding the
vacuolar protein strictosidine synthase from Catharanthus roseus. Nucleic Acids Res 1990;18:4939.
Meyer HJ, van Staden J. The in vitro production of an anthocyanin from callus cultures of Oxalis linearis. Plant
Cell Tissue Org Cult 1995;40:55 8.
Meyer HP, Kiener A, Imwinkelried R, Shaw N, Lonza AG. Biotransformations for fine chemical production.
Chimia 1997;51:287.
Mironowicz A, Kukulczanka K, Krasinki K, Siewinski A. Transformation of isoprenoids by orchids in tissue
culture. Phytochemicals 1987;26:1959 60.
Misawa M. Production of useful plant metabolites. In: Fiechter A, editor. Adv Biochem Eng Biotechnol. Berlin:
Springer-Verlag, 1985. pp. 59 88.
Mitra A, Kitamura Y, Mayer MJ, Michael AJ, Narbad AJ, Parr AJ, Walton NJ. p-Hydroxycinnamoyl-CoA
hydratase/lyase a Pseudomonas enzyme expressed in hairy roots of Datura stramonium. Biochem Soc
Trans 1999;27:A51.
Miyasaka H, Masu M, Yamamoto T, Endo Y, Yoneda K. Production of cryptotanshinone and ferruginol by
immobilized culture cells of Salvia miltiorrhiza. Phytochemicals 1986;25:1621 4.
Mok MC, Gabelman WH, Skoog F. Carotenoid synthesis in tissue cultures of Daucus carota. J Am Soc Hortic Sci
1976;101:442 9.
Morris P, Smart NJ, Fowler MW. Fluidized bed vessel for the culture of immobilized plant cells and its applications for the continuous production of fine cell suspension. Plant Cell Tissue Org Cult 1984;2:207 16.
Mukherjee SK, Sabapathi RB, Gupta N. Low sugar and osmotic requirements for shoot regeneration from leaf
pieces of Solanum melongena L.. Plant Cell Tissue Org Cult 1991;25:13 6.
Mulder-Krieger T, Verpoorte R, Svendse A, Scheffer J. Production of essential oils and flavours in plant cell and
tissue cultures. A review. Plant Cell Tissue Org Cult 1988;13:85 114.
Naik GR. Micropropagation studies in medicinal and aromatic plants. In: Khan IA, Khanun A, editors. Role of
biotechnology in medicinal and aromatic plants. Hyderabad: Ukaz Publications, 1998. pp. 50 6.
Nakagawa K, Konagai A, Fukui H, Tabata H. Release and crystallization of berberine in the liquid medium of
Thalictrum minus cell suspension cultures. Plant Cell Rep 1984;3:254 7.
Naoshima Y, Akakabe Y. Biotransformation of some ketoesters through the consecutive reuse of immobilized
Nicotiana tabacum cells. J Org Chem 1989;54:4237 9.
Narayan MS, Naidu A, Ravishankar GA, Srinivas L, Venkataraman LV. Antioxidant effect of anthocyanin on
enzymatic lipid peroxidation. Prostaglandins, Leukotrienes Essent Fatty Acids 1999;60:1 4.
Narbad A, Gasson MJ. Metabolism of ferulic acid via vanillin using a novel CoA-dependent pathway in a newly
isolated strain of Pseudomonas fluorescens. Microbiology 1998;144:1397 405.
Nilsson K, Birnbaum S, Flygare S, Linse L, Schroder U, Jeppsson U, Larsson P, Mosbach K, Brodelius P. A
general method for the immobilization of cells with preserved viability. Eur J Appl Microbiol Biotechnol
1983;17:319 26.
Noguchi M, Matsumoto T, Hirata Y, Yamamoto K, Katsuyama A, Kato A, Azechi S, Kato K. Improvement of

S. Ramachandra Rao, G.A. Ravishankar / Biotechnology Advances 20 (2002) 101153

147

growth rates of plant cell cultures. In: Barz W, Reinhard E, Zenk MH, editors. Plant tissue culture and its
biotechnological applications. New York: Springer-Verlag, 1977. pp. 85 94.
Norton RA, Towers GHN. Factors affecting synthesis of polyacetylenes in root cultures of Bidens alba. J Plant
Physiol 1986;122:41 53.
Novais J. Methods of immobilization of plant cells. In: Pais M, Mavituna F, Novais J, editors. Plant cell
biotechnology. NATO ASI Series. New York: Springer, 1988. pp. 353 63.
Ogino T, Hiraoka N, Tabata M. Selection of high nicotine producing cell lines of tobacco callus by single cell
cloning. Phytochemicals 1978;22:2447 50.
Ohsumi C, Hayashi T, Sano K. Formation of allin in the culture tissues of Allium sativum oxidation of S-allyl-Lcysteine. Phytochemicals 1993;33:107 11.
Orihara Y, Furuya T. Studies on plant tissue culture. Biotransformation of ( )-borneol by cultured cells of
Eucalyptus perriniana. Phytochemicals 1993;34:1045 58.
Orihara T, Miyatake H, Furuya T. Triglucosylation on the biotransformation of (+)-menthol by cultured cells of
Eucalyptus perriniana. Phytochemicals 1991;30:1843 5.
Orihara Y, Furuya T, Hashimoto N, Deguchi Y, Tokoro K, Kanisawa T. Biotransformation of isoeugenol and
eugenol by cultured cells of Eucalyptus perriniana. Phytochemicals 1992;31:827 31.
Orihara T, Noguchi T, Furuya T. Biotransformation of (+)-camphor by cells of Eucalyptus perriniana. Phytochemicals 1994;35:941 5.
Panda AK, Bisaria VS, Mishra S. Alkaloid production by plant cell cultures of Holarrhena antidysentrica: II.
Effect of precursor feeding and cultivation of stirred tank bioreactor. Biotechnol Bioeng 1992;39:1052.
Park JM, Hu W-S, Staba EJ. Cultivation of Artemisia annua L. plantlets in a bioreactor containing a single carbon
and a single nitrogen source. Biotechnol Bioeng 1989;34:1209 13.
Park JM, Yoon SY, Giles KL, Songstad DD, Eppstein D, Novakovski D, Friesen L, Rosevear I. Production of
sanguinarine by suspension culture of Apaver somniferum in bioreactors. J Ferment Bioeng 1992;74:292 6.
Parr AJ, Robins RJ, Rhodes MJC. Permeabilization of Cinchona ledgeriana cells by dimethyl sulfoxide. Effect on
alkaloid release and long term membrane integrity. Plant Cell Rep 1984;3:262 5.
Parr AJ, Robins RJ, Rhodes MJC. Release of secondary metabolites by plant cell cultures. In: Webb C, Mavituna
F, editors. Plant and animal cells: process possibilities. Chichester: Ellos Horwood, 1987. pp. 229 37.
Payne GF, Payne NN, Shuler ML, Asada M. In situ adsorption for enhanced alkaloid production by Catharanthus
roseus. Biotechnol Lett 1988;10:187 92.
Payne GF, Bringi V, Prince C, Shuler ML. Plant cell and tissue culture in liquid systems. Munich: Hanser Publ.,
1991. pp. 1 10.
Pepin M-F, Archambault J, Chavarie C, Cormier F. Growth kinetics of Vitis vinifera cell suspension cultures:
shake flask cultures. Biotechnol Bioeng 1995;47:131 8.
Petit A, David C, Dahl GA, Ellis JG, Guyon P, Casse-Delbast F, Tempe J. Opine concept: plasmids in Agrobacterium rhizogenes cooperate for opine degradation. Mol Gen Genet 1983;190:204 14.
Pezzuto JM. Natural product cancer chemoprotective agents. In: Arnason JT, Mata R, Romeo JT, editors. Recent
advances in phytochemistry. Phytochemistry of medicinal plants, vol. 29. New York: Plenum, 1995. pp. 19 45.
Phillipson JD. Plants as source of valuable products. In: Charlwood BV, Rhodes MJC, editors. Secondary products
from plant tissue culture. Oxford: Clarendon Press, 1990. pp. 1 21.
Pras N. Bioconversion of naturally occurring precursors and related synthetic compounds using plant cell cultures.
J Biotechnol 1992;26:29 62.
Pras N, Woerdenbag HJ, Batterman S, Visser JF, Van Uden W. Mucuna pruriens: improvement of the biotechnological production of the anti-Parkinson drug L-DOPA by plant cell selection. Pharm World Sci 1993;15:263 8.
Pras N, Woerdenbag HJ, Van Uden W. Bioconversion potential of plant enzymes for the production of pharmaceuticals. Plant Cell Tissue Org Cult 1995;43:117 21.
Prenosil JE, Pedersen H. Immobilized plant cell reactors. Enzyme Microb Technol 1983;5:323 31.
Prince RC, Gunson DE. Just plain vanilla? Trends Biochem Sci 1994. p. 521 (December).
Qi ZH, Hedges AR. Uses of cyclodextrins for flavours. In: Ho CT, Tan CT, Tong C-H, editors. Flavour technology. Washington: American Chemical Society, 1995. pp. 231 42.

148

S. Ramachandra Rao, G.A. Ravishankar / Biotechnology Advances 20 (2002) 101153

Quesnell AA, Ellis BE. Comparison of UV-irradiation and p-fluorophenylalanine as selective agents for the
production of aromatic compounds in plant cell culture. J Biotechnol 1989;10:27 38.
Rabenhorst J. Verfahren zur Herstellung von naturichem vanillin. European Patent EP 0405197 A1, 1991.
Rabenhorst J. Production of methoxyphenol-type natural aroma chemicals by biotransformation of eugenol with a
new Pseudomonas spp. Appl Microbiol Biotechnol 1996;46:470 4.
Rajasekaran T, Ravishankar GA, Venkataraman LV. Influence of nutrient stress on pyrethrin production by
cultured cells of pyrethrum (Chrysanthemum cinerariaefolium). Curr Sci 1991;60:705 7.
Rajendran L, Ravishankar GA, Venkataraman LV, Prathiba KR. Anthocyanin production in callus cultures of
Daucus carota L. as influenced by nutrient stress and osmoticum. Biotechnol Lett 1992;14:707 14.
Rajendran L, Suvarnalatha G, Ravishankar GA, Venkataraman LV. Enhancement of anthocyanin production in
callus cultures of Daucus carota L. under influence of fungal elicitors. Appl Microbiol Biotechnol 1994;
42:227 31.
Ramachandra Rao S. Studies on biotransformation to produce phytochemicals of importance using plant cell
cultures. PhD Thesis. University of Mysore, Mysore, 1998.
Ramachandra Rao S, Ravishankar GA. Biotransformation of phenylpropanoid compounds to vanilla flavour
metabolites in immobilized cell cultures of Capsicum frutescens. 4th International Food Convention,
Mysore, 1998. p. 44 (Abstract volume).
Ramachandra Rao S, Ravishankar GA. Biotransformation of isoeugenol to vanilla flavour metabolites and
capsaicin in freely suspended and immobilized cell cultures of Capsicum frutescens: study of the influence
of b-cyclodextrin and fungal elicitor. Process Biochem 1999;35:341 8.
Ramachandra Rao S, Ravishankar GA. Biotransformation of protocatechuic aldehyde and caffeic acid to vanillin
and capsaicin in freely suspended and immobilized cell cultures of Capsicum frutescens. J Biotechnol
2000a;76:137 46.
Ramachandra Rao S, Ravishankar GA. Vanilla flavour: production by conventional and biotechnological routes.
J Sci Food Agric 2000b;80:289 304.
Ramachandra Rao S, Ravishankar GA, Venkataraman LV. An improved process for the preparation of vanillin.
Indian Patent No. 1022/DEL/96, 1996.
Ramachandra Rao S, Sarada R, Ravishankar GA. Phycocyanin, a new elicitor of capsaicin and anthocyanin
accumulation in plant cell cultures. Appl Microbiol Biotechnol 1996b;46:619 21.
Ramachandra Rao S, Usha T, Ravishankar GA. Biotransformation of codeine to morphine in free and immobilized
cell cultures of Spirulina platensis. World J Microbiol Biotechnol 1999;15:465 9.
Ramachandra Rao S, Usha T, Ravishankar GA. Biotransformation of digitoxin to digoxin in cell cultures of
Capsicum frutescens in the presence of b-cyclodextrin. Biocatal Biotransform 2002.
Ravishankar GA, Ramachandra Rao S. Biotechnological production of phyto-pharmaceuticals. J Biochem, Mol
Biol Biophys 2000;4:73 102.
Ravishankar GA, Venkataraman LV. Food applications of plant cell cultures. Curr Sci 1990;59:914 20.
Ravishankar GA, Venkataraman LV. Role of plant cell culture in food biotechnology: current trends, limitations
and future prospects. In: Prakash J, Pierik RLM, editors. Plant biotechnology: commercial prospects and
problems. New Delhi: Oxford IBH Press, 1993. pp. 255 74.
Ravishankar GA, Sarma KS, Venkataraman LV, Kadyam AK. Effect of nutritional stress on capsaicin production
in immobilized cell cultures of Capsicum annuum. Curr Sci 1988;57:381 3.
Ravishankar GA, Bhyalakshmi N, Ramachandra Rao S. Production of food additives. In: Ramawat KG, Merillon
JM, editors. Biotechnology: secondary metabolites. New Delhi: Oxford IBH, 1999. pp. 89 110.
Reinhard E, Kreis W, Barthlen U, Helmbold U. Semicontinuous cultivation of Digitalis lanata cells: production of
b-methyldigitoxin in a 300 L airlift bioreactor. Biotechnol Bioeng 1989;34:502 8.
Remi Shih NJ, McDonald KA, Dandekar AM, Girbes T, Iglesias R, Jackman AP. A novel type-1 ribosome
inactivating protein isolated from the supernatant of transformed suspension cultures of Trichosanthes kirilowii. Plant Cell Rep 1998;17:531 7.
Rhodes MJC. In: Kurz WGW, editor. Primary and secondary metabolism of plant cell cultures. Berlin: SpringerVerlag, 1989. pp. 58 72.

S. Ramachandra Rao, G.A. Ravishankar / Biotechnology Advances 20 (2002) 101153

149

Rhodes MJC, Hamill J, Parr AJ, Robins RJ, Walton NJ. In: Robins RJ, Rhodes MJC, editors. Manipulating
secondary metabolism in culture. Oxford: Cambridge Univ. Press, 1988. pp. 83 93.
Ritterhaus E, Ulrich J, Westphal K. Large scale production of plant cell cultures. Int Assoc Plant Tissue Cult News
Lett 1990;61:2 10.
Robbins RJ, Rhodes MJC. The stimulation of anthraquinone production by Cinchona ledgeriana cultures with
polymeric adsorbents. Appl Microbiol Biotechnol 1986;24:35 41.
Robbins MP, Hartnoll J, Morris P. Phenylpropanoid defence responses in transgenic Lotus corniculatus: 1.
Glutathione elicitation of isoflavan phytoalexins in transformed root cultures. Plant Cell Rep 1991;10:
59 62.
Robbins RJ, Walton NJ, Parr AJ, Lindsey E, Arid H, Rhodes MJC, Hamill JD. Progress in genetic engineering of
the pyridine and tropane alkaloid biosynthetic pathways of solanaceous plants. In: Ellis BE, Kuroki GW,
Stafford HA, editors. Recent advances in phytochemistry, vol. 28. Genetic engineering of plant secondary
metabolism. New York: Plenum, 1994. pp. 1 33.
Rodriguez-Mendiola MA, Stafford A, Cressuel R, Arias-Castr C. Bioreactors for growth of plant roots. Enzyme
Microb Technol 1991;13:697 702.
Roja G. Biotechnology of indigenous medicinal plants. PhD Thesis, Bombay University, Bombay, 1994.
Romagnoli LG, Knorr D. Effects of ferulic acid treatment on growth and flavour development of cultured Vanilla
planifolia cells. Food Biotechnol 1988;2:93 104.
Rosevear A. Putting a bit of color into the subject. Trends Biotechnol 1984;2:145 6.
Sahai OM. Plant tissue culture. In: Gabelman A, editor. Bioprocess production of flavour and fragrance and color
ingredients. New York: Wiley, 1994. pp. 239 75.
Sahai O, Knuth M. Commercializing plant tissue culture processes: economics, problem and prospects. Biotechnol
Prog 1985;1:1 9.
Sahai OP, Shuler ML. Environmental parameters influencing phenolics production by batch cultures of Nicotiana
tabacum. Biotechnol Bioeng 1984;26:111 20.
Saito K, Yamazaki M, Murakoshi I. Transgenic medicinal plants: Agrobacterium-mediated foreign gene transfer
and production of secondary metabolites. J Nat Prod 1992;55:149 62.
Sakamoto K, Iida K, Sawamura K, Hajiro K, Asada Y, Yoshikawa T, Furuya T. Effects of nutrients on anthocyanin
production in cultured cells of Aralia cordata. Phytochemicals 1993;33:357 60.
Sakui N, Kuroyanagi M, Ishitobi Y, Sato M, Ueno A. Biotransformation of sesquiterpenes by cultured cells of
Curcuma zedoaria. Phytochemicals 1992;31:143 7.
Salgado-Garciglia R, Ochoa-Alejo N. Increased capsaicin content in PFP resistant cells of chilli pepper (Capsicum
annuum L.). Plant Cell Rep 1990;8:617 20.
Sasse F, Knobloch K, Berlin J. Induction of secondary metabolism in cell suspension cultures of Catharanthus
roseus, Nicotiana tabacum and Peganum harmala. In: Fujiwara A, editor. Proceedings of the 5th International
Congress of Plant Tissue and Cell Culture. Tokyo: Abe Photo Printing, 1982. pp. 343 4.
Scheidegger A. Plant biotechnology goes commercial in Japan. Trends Biotechnol 1990;8:197 8.
Schumacher H-M, Gundlach H, Fiedler F, Zenk MH. Elicitation of benzophenanthridine alkaloid biosynthesis in
Eschscholtzia cell cultures. Plant Cell Rep 1987;6:410 3.
Scragg AH. Plant cell bioreactors. In: Stafford A, Warren GS, editors. Plant cell and tissue culture. Milton Keynes:
Open Univ. Press, 1991. pp. 221 39.
Scragg AH. The production of aromas by plant cell cultures. In: Schepier T, editor. Adv Biochem Eng Biotechnol,
vol. 55. Berlin: Springer-Verlag, 1997. pp. 239 63.
Sehnke PC, Ferl RJ. Processing of preproricin in transgenic tobacco. Protein Expression Purif 1999;15:188 95.
Seitz HU, Hinderer W. Anthocyanins. In: Constabel F, Vasil I, editors. Cell culture and somatic cell genetics of
plants, vol. 5. San Diego: Academic Press, 1988. pp. 49 76.
Sevon N, Hiltunen R, Caldentey KMO. Somoclonal variation in transformed roots and protoplast-derived hairy
root cultures of Hyoscyamus muticus. Planta Med 1998;64:37 41.
Sharp JM, Doran PM. Effect of bacitracin on growth and monoclonal antibody production by tobacco hairy roots
and cells suspensions. Biotechnol Bioprocess Eng 1999;4:253 8.

150

S. Ramachandra Rao, G.A. Ravishankar / Biotechnology Advances 20 (2002) 101153

Sharp JM, Doran PM. Characterization of monoclonal antibody fragments produced by plant cells. Biotechnol
Bioeng 2001;73:338 46.
Shim JY, Chang YJ, Kim SU. Indigo and indirubin derivatives from indoles in Polygonum tinctorium tissue
cultures. Biotechnol Lett 1998;20:1139 43.
Shimomura K, Satake M, Kamada H. Production of useful secondary metabolites by hairy roots transformed
with Ri-plasmid. In: Somers D, Gengenbach BG, Biesboer DD, Hackett WP, Green CE, editors. Proceedings
of the 5th International Congress of Plant Tissue and Cell Culture. Minneapolis: University of Minneapolis,
1986. p. 250.
Shimomura K, Sauerwein M, Ishimaru K. Tropane alkaloids in the adventitious and hairy root cultures of
solanaceous plants. Phytochemicals 1991;30:2275 8.
Shin SW, Kim YS. Production of anthroquinones derivatives by hairy root cultures of Rubia cordifolia var.
pratensis. Korean J Pharmacogn 1996;27:301 8.
Shuler ML, Hallsby GA. Bioreactor considerations for chemical production from plant cell cultures. In: Zaitlin M,
Day P, Hollaender A, editors. Biotechnology in plant science relevance to agriculture in the eighties. New
York: Academic Press, 1985. pp. 191 205.
Shuler ML, Sarkar OP, Hallsby GA. Entrapped plant cell tissue cultures. Ann NY Acad Sci 1983;413:373 82.
Sim SJ, Chang HN. Increased shikonin production by hairy roots of Lithospermum erythrorhizon in two-phase
bubble column reactor. Biotechnol Lett 1993;15:145 50.
Smart NJ, Fowler MW. Effect of aeration on large scale culture of plant cells. Biotechnol Lett 1981;3:171 6.
Smith MD. Antibody production in plants. Biotechnol Adv 1996;14:267 81.
Smith MD, Glick BR. The production of antibodies in plants: an idea whose time has come? Biotechnol Adv
2000;18:85 9.
Songstad DD, Kurz WGW, Nessler CL. Tyramine accumulation in Nicotiana tabacum transformed with a
chimeric tryptophan decarboxylase gene. Phytochemicals 1991;30:3245 6.
Sreekumar S, Seeni S, Pushpangadhan P. Production of 2-hydroxy 4-methoxy benzaldehyde using root cultures of
Hemidesmus indicus. Biotechnol Lett 1998;20:631 5.
Staba EJ, Chung AC. Quinine and quinidine production by Cinchona leaf, root and unorganized culture. Phytochemicals 1981;20:84 9.
Stafford A, Morris P, Fowler MW. Plant cell biotechnology: a perspective. Enzyme Microb Technol 1986;8: 19 23.
Steck W, Constabel F. Biotransformation in plant cell cultures. Lloydia 1974;37:181 91.
Steffens JC, Darel E, Hunt MD. Polyphenoloxidase. In: Ellis BE, Kuroki GW, Stafford HA, editors. Genetic
engineering of plant secondary metabolism. New York: Plenum, 1994. pp. 275 312.
Stepan-Sarkissan G. Biotransformations by plant cell cultures. In: Stafford A, Warren GS, editors. Plant cell and
tissue culture. Milton Keynes: Open Univ. Press, 1991. pp. 163 204.
Stockigt J. Biotransformations with cultivated plant cells. Agro-Food-Ind Hi-Tech 1993;4:25 8.
Stockigt J, Zenk MH. Strictosidine: the key intermediate in the biosynthesis of monoterpenoid indole alkaloids.
J Chem Soc, Chem Commun. 1977;912 4.
Stockigt J, Obitz P, Flakenhagen H, Lutterbach R, Endress R. Natural products and enzymes from plant cell
cultures. Plant Cell Tissue Org Cult 1995;43:914 20.
Subroto MA, Kwok K, Hamill JD, Doran PM. Coculture of genetically transformed roots and shoots for synthesis,
translocation and biotransformation of secondary metabolites. Biotechnol Bioeng 1996;49:481 94.
Suga T, Hirata T. Biotransformation of exogenous substrates by plant cell cultures. Phytochemicals 1990;29:
2393 406.
Sujata V, Ravishankar GA, Venkataraman LV. Induction of crocin, crocetin, picrocrocin and safranal synthesis in
callus cultures of saffron (Crocus sativus L.). Biotechnol Appl Biochem 1990;12:336 40.
Suvarnalatha G, Narayan MS, Ravishankar GA, Venkataraman LV. Flavour production in plant cell cultures of
Basmati rice (Oryza sativa L.). J Sci Food Agric 1994;66:439 42.
Swanson SM, Mahady GB, Beecher CWW. Stevioside biosynthesis by callus, root, shoot and rooted shoot
cultures in vitro. Plant Cell Tissue Org Cult 1992;28:151 7.
Szejtli J. Cyclodextrins and their inclusion complexes. Budapest: Academia Kiado, 1982.

S. Ramachandra Rao, G.A. Ravishankar / Biotechnology Advances 20 (2002) 101153

151

Szejtli J. Cyclodextrins in biotechnology. Starch/Staerke 1986;38:388 90.


Szejtli J. Cyclodextrin technology. Dordrecht: Kluwer Academic Publishing, 1988.
Tabata M. Naphthoquinones. In: Constael F, Vasil I, editors. Cell culture and somatic cell genetics of plants, vol. 5.
San Diego: Academic Press, 1988. pp. 99 111.
Tabata M, Fujita Y. Production of shikonin by plant cell cultures. In: Zaitlin M, Day P, Hollaender A, editors.
Biotechnology in plant science. Orlando: Academic Press, 1985. pp. 207 18.
Tadasa K, Kayahara H. Initial steps in eugenol degradation pathway of a microorganism. Agric Biol Chem
1983;47:2639 40.
Takayama S, Misawa M. Mass propagation of Begonia  biemalis plantlets by shake culture. Plant Cell Physiol
1981;22:211 4.
Tanaka H. Large scale cultivation of plant cells at higher cell densities: a review. Process Biochem 1987;22:
106 13.
Tavladoraki P, Benvenuto E, Trinca S, DeMartinis D, Cattaneo A, Galeffi P. Transgenic plants expressing a
functional single-chain Fv antibody are specifically protected from virus attack. Nature 1993;366:469 72.
Taya M, Yoyama A, Kando O, Kobayashi T, Matsui C. Growth and characteristics of plant hairy roots and their
cultures in bioreactors. J Chem Eng Jpn 1989;22:74 89.
Tepfer D. Genetic transformation using Agrobacterium rhizogenes. Physiol Plant 1990;79:14 6.
Terashima M, Murai Y, Kawamura M, Nakanishi S, Stoltz T, Chen L, Drohan W, Rodriguez RL, Katoh S.
Production of functional human a1-antitrypsin by plant cell culture. Appl Microbiol Biotechnol 1999a;52:
516 23.
Terashima M, Ejiri Y, Hashikawa N, Yoshida H. Effect of osmotic pressure on human a1-antitrypsin production by
plant cell culture. Biochem Eng J 1999b;4:31 6.
Thanavalla Y, Yang YF, Lyons P, Mason HS, Arntzen CJ. Immunogenicity of a transgenic plant derived hepatitis
B surface antigen. Proc Natl Acad Sci USA 1995;92:3358 61.
Thron U, Maresch L, Beiderbeck R, Reichling J. Accumulation of unusual phenylpropanoids in transformed and
non-transformed root cultures of Coreopsis tinctora. Z Naturforsch 1989;44C:573 7.
Toivonen L, Laakso S, Rosenqvist H. The effect of temperature on hairy root cultures of Catharanthus roseus:
growth, indole alkaloid accumulation and membrane lipid composition. Plant Cell Rep 1992;11:395 9.
Townsley PM. Chocolate from plant cells. J Inst Can Sci Technol Aliment 1972;7:76 8.
Trypsteen M, Van Lijsebettens M, Van Severen R, Van Montagu M. Agrobacterium rhizogenes mediated transformation of Echinacea purpurea. Plant Cell Rep 1991;10:85 9.
Turpen TH, Reinl SJ, Charoenvit Y, Hoffman SL, Fallarme V, Grill LK. Malarial epitopes expressed on the surface
of recombinant Tobacco Mosaic Virus. Bio/Technology 1995;13:53 7.
Ulbrich B, Wiesner W, Arens H. Large scale production of rosmarinic acid from plant cell cultures of Coleus
blumei Benth. In: Deus-Neumann B, Barz W, Reinhard E, editors. Secondary metabolism of plant cell culture.
Berlin: Springer-Verlag, 1985. pp. 293 303.
Upadhyay R, Arumugam N, Bhozwani SS. In vitro propagation of Picrorhiza kurroa royle Ex Benth an
endangered species of medicinal importance. Phytomorphology 1989;39:235 42.
Usha T, Ramachandra Rao S, Ravishankar G. A process for the production of vanilla flavour metabolites through
biotransformation. Indian Patent No. 1193/DEL/99, 1999.
Ushiyama M, Furuya T. Glycosylation of phenolic compounds by root cultures of Panax ginseng. Phytochemicals
1989;28:3009 13.
Ushiama K, Oda H, Westphal K. Large scale tissue culture of Panax ginseng root. In: Somers B, Genbach D,
Biesboer D, Hackett W, Green C, editors. Sixth International Congress of Plant Tissue and Cell Culture.
Minneapolis: University of Minnesota, 1986 (Abstract No. 252).
van der Wel H, Ledeboer AM. Thaumatins. In: Stumpf PK, Conn EE, editors. The biochemistry of plants a
comprehensive treatise, vol. 15. San Diego: Academic Press, 1989. pp. 379 91.
Van Uden W, Pras N, Malingre ThM. On the improvement of the podophyllotoxin production by phenylpropanoid precursor feeding to cell cultures of Podophyllum hexandrum Royle. Plant Cell Tissue Org Cult
1990;23:217 24.

152

S. Ramachandra Rao, G.A. Ravishankar / Biotechnology Advances 20 (2002) 101153

Van Uden W, Woerdenbag HJ, Pras N. Cyclodextrins as a useful tool for bioconversions in plant cell biotechnology. Plant Cell Tissue Org Cult 1994;38:103 13.
Van Uden W, Bouma AS, Waker JFS, Middel O, Wichers HJ, Waard P, Woerdenbag H, Kellog RM, Pras N.
The production of podophyllotoxin and its 5-methoxy derivatives through bioconversion of cyclodextrin
complexed desoxypodophyllotoxin by plant cell cultures. Plant Cell Tissue Org Cult 1995;42:73 9.
Vega FA, Ramos MF, Fernandez M, Santesteban M, Galar A. Pharmacological activity of anthocyanins. Cienc Ind
Farm 1987;6:205 8.
Verdelhan des Molles E, Gomord V, Bastin M, Faye L, Courtois D. Expression of carrot invertase gene in tobacco
suspension cells cultivated in batch and continuous culture conditions. J Biosci Bioeng 1999;87:302 6.
Vunsh R, Matilsky MB, Keren ZM, Robinfeld B. Production of a natural red colour by carrot cell suspension
cultures. In: Samers DA, Gengnebach BG, Biesboer DD, Hackett WP, Green CE, editors. VI International
Congress of Plant Tissue and Cell Cultures. Minneapolis: University of Minneapolis, 1986. p. 119.
Wagner F, Vogelmann H. Cultivation of plant tissue culture in bioreactors and the function of secondary metabolites. In: Barz W, et al, editor. Plant tissue culture and its biotechnological applications. Berlin: SpringerVerlag, 1977. pp. 245 52.
Walton NJ, Belshaw NJ. The effect of cadaverine on the formation of anabasine from lysine in hairy root cultures
of Nicotiana hesperis. Plant Cell Rep 1988;7:115 8.
Walton NJ, Robins RJ, Belshaw NJ, Parr AJ, Hamill JD, Rhodes MJC. Perturbation of alkaloid biosynthesis in
transformed root cultures of solanaceous plants by metabolic precursor and their analogues. In: Robins RJ,
Rhodes MJC, editors. Manipulating secondary metabolism in culture. Cambridge: Cambridge Univ. Press,
1988. pp. 73 8.
Wataneba K, Yano SI, Yamada Y. Selection of cultured plant cell lines producing high levels of biotin. Phytochemicals 1982;21:513 6.
Webster TM. New perspectives on vanilla. Cereals Food World 1995;40:198 200.
West S. Chemical biotransformations. In: Godfrey T, West S, editors. Industrial enzymology. London: Macmillan,
1996. pp. 157 75.
Westcott RJ, Cheetham PSJ, Barraclough AJ. Use of organized Vanilla plant aerial roots for the production of
vanillin. Phytochemicals 1994;35:135 8.
Wichers HJ, Malingre ThM, Huizing HJ. The effect of some environmental parameters on the production of
L-DOPA by alginate entrapped cells of Mucuna pruriens. Planta 1983;158:482 6.
Widholm JM. Evidence for compartmentation of tryptophan in cultured plant tissues. Free tryptophan levels and
inhibition of anthranilate synthetase. Physiol Plant 1974;30:323 6.
Wilhelm R, Zenk MH. Biotransformation of thebaine by cell cultures of Papaver somniferum and Mohonia
nervosa. Phytochemicals 1997;46:701 8.
Williams PD, Mavituna F. Immobilized plant cells. In: Fowler MW, Warren GS, editors. Plant biotechnology.
Oxford: Pergamon, 1992. pp. 63 78.
Woerdenbag HJ, Pras N, Frijling HW, Lerk CF, Malingre ThM. Cyclodextrin facilitated bioconversion of 17bestradiol by a phenoloxidase from Mucuna pruriens cell cultures. Phytochemicals 1990a;29:1551 4.
Woerdenbag HJ, Pras N, Van Uden W, Frijling HW, Pras N, Malingre ThM. Increased podophyllotoxin production
in Podophyllum hexandrum cell suspension cultures after feeding coniferyl alcohol as a b-cyclodextrin complex. Plant Cell Rep 1990b;9:97 100.
Wongsamuth R, Doran PM. Production of monoclonal antibodies by tobacco hairy roots. Biotechnol Bioeng
1997;54:401 15.
Yamada Y, Hashimoto T. Biosynthesis of tropane alkaloids. In: Bock G, Marsh J, editors. Applications of plant
cell and tissue culture. Chichester: Wiley, 1988. pp. 199 212.
Yamakawa T, Kato S, Ishida K, Kodama T, Minoda Y. Production of anthocyanins by Vitis cells in suspension
culture. Agric Biol Chem 1983;47:2185 91.
Yamamoto T, Mizuguchi R, Yamada Y. Selection of a high stable pigment producing strain in cultured Euphorbia
milli cells. Theor Appl Genet 1982;16:113 6.
Yeoman MM. Techniques, characteristics, properties, and commercial potential of immobilized plant cells. In:

S. Ramachandra Rao, G.A. Ravishankar / Biotechnology Advances 20 (2002) 101153

153

Constabel F, Vasil I, editors. Cell culture and somatic cell genetics of plants, vol. 4. San Diego: Academic
Press, 1987. pp. 197 215.
Yeoman MM, Meidzybrodzka MB, Lindsey K, McLauchlan WR. The synthetic potential of cultured plant cells.
In: Sala F, Parisi B, Cella R, Cifferi O, editors. Plant cell cultures: results and perspectives. Amsterdam:
Elsevier, 1980. pp. 327 43.
Yeoman MM, Holden MA, Corchet P, Holden PR, Goy JG, Hobbs MC. Exploitation of disorganized plant
cultures for the production of secondary metabolites. In: Charlwood BV, Rhodes MJC, editors. Secondary
products from plant tissue culture. Oxford: Clarendon Press, 1990. pp. 139 66.
Yoshida A, Takenaka Y, Tamaki H, Frebort I, Adachi O, Kumagai H. Vanillin formation by microbial amine
oxidase from vanillylamine. J Ferment Bioeng 1997;84:603 5.
Yoshikawa T, Furuya T. Saponin production by cultures of Panax ginseng transformed with Agrobacterium
rhizogenes. Plant Cell Rep 1987;6:449 53.
Yoshioka K, Yamagata H, Ito H, Deno Y, Fujita Y, Yamada Y. Effects of exogenous polyamines on tropane
alkaloid production by a root culture of Duboisia myoporoides. Planta Med 1989;55:523 4.
Yun D-J, Hashimoto T, Yamada Y. Metabolic engineering of medicinal plants: transgenic Atropa belladona with
an improved alkaloid composition. Proc Natl Acad Sci USA 1992;89:11799 803.
Yusibov V, Modelska A, Slepleuski K, Agadjanyam M, Weiner D, Hooper DC, Koprowski H. Antigens production in plants by infection with chimeric plant viruses immunize against rabies and HIV-1. Proc Natl Acad Sci
USA 1997;94:5784 8.
Zenk MH. The impact of plant cell cultures on Industry. In: Thorpe EA, editor. Frontiers of plant tissue culture.
Calgary: The International Association of Plant Tissue Culture, 1978. pp. 1 14.
Zenk MH, EI-Shagi H, Schulte U. Anthraquinone production by cell suspension cultures of Morinda citrifolia.
Planta Med Suppl 1975;79 101.
Zenk MH, EI-Shagi E, Arens H, Stockigt J, Weiler EW, Deus B. Formation of the indole alkaloids serpentine and
ajmalicine in suspension cultures of Catharanthus roseus. In: Barz W, Reinhard E, Zenk MH, editors. Plant
tissue culture and its biotechnological applications. Berlin: Springer-Verlag, 1977. pp. 27 43.
Zetlin L, Olmsted SS, Moench TR, Co MS, Martinell BJ, Paradkar VM, Russell DR, Queen C, Cone RA, Whaley
KJ. A humanized monoclonal antibody produced in transgenic plants for immunoprotection of the vagina
against genital herpes. Nat Biotechnol 1998;16:1361 4.
Zhong JJ, Konstantinov KB, Toshida T. Computer-aided on-line monitoring of physiological variables in suspended cell cultures of a Perilla frutescens in a bioreactor. J Ferment Bioeng 1994;77:445 7.
Zhong J-J, Yu J-T, Yoshida T. Recent advances in plant cell cultures in bioreactors. World J Microbiol Biotechnol
1995;11:461 7.

Potrebbero piacerti anche