Sunteți pe pagina 1din 15

JBA-06956; No of Pages 15

Biotechnology Advances xxx (2015) xxx–xxx

Contents lists available at ScienceDirect

Biotechnology Advances

journal homepage: www.elsevier.com/locate/biotechadv

Research review paper

Endophytes as in vitro production platforms of high value plant


secondary metabolites
Aarthi Venugopalan, Smita Srivastava ⁎
Department of Biotechnology, Indian Institute of Technology Madras, Chennai 600 036, India

a r t i c l e i n f o a b s t r a c t

Article history: Many reports have been published on bioprospecting of endophytic fungi capable of producing high value bioac-
Received 9 February 2015 tive molecules like, paclitaxel, vincristine, vinblastine, camptothecin and podophyllotoxin. However, commercial
Received in revised form 27 June 2015 exploitation of endophytes for high value-low volume plant secondary metabolites remains elusive due to widely
Accepted 16 July 2015
reported genomic instability of endophytes in the axenic culture. While most of the endophyte research focuses
Available online xxxx
on screening endophytes for novel or existing high value biomolecules, very few reports seek to explore the
Keywords:
possible mechanisms of production of host–plant associated or novel secondary metabolites in these organisms.
Endophytic fungi With an overview of host–endophyte relationship and its possible impact on the secondary metabolite
Host–endophyte relationship production potential of endophytes, the review highlights the evidence reported for and against the presence
Secondary metabolite of host-independent biosynthetic machinery in endophytes. The review aims to address the question, why
In vitro production should and how can endophytes be exploited for large scale in vitro production of high value phytochemicals?
Bioprocess optimization In this regard, various bioprocess optimization strategies that have been applied to sustain and enhance the prod-
Axenic culture instability uct yield from the endophytes have also been described in detail. Further, techniques like mixed fermentation/
Mixed fermentation
co-cultivation and use of epigenetic modifiers have also been discussed as potential strategies to activate cryptic
Epigenetic modifiers
gene clusters in endophytes, thereby aiding in novel metabolite discovery and overcoming the limitations
associated with axenic culture of endophytes.
© 2015 Elsevier Inc. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
2. What are endophytes? — definition, discovery and classification . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
3. Host–endophyte relationship — role in host plant evolution, defense and secondary metabolite production . . . . . . . . . . . . . . . . . . . . 0
3.1. Production of plant secondary metabolites in endophytes — the possibility . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
4. Using endophytes as in vitro production platforms for plant secondary metabolites . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
4.1. Limitations of producing bioactive secondary metabolites from plants . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
4.2. Alternative methods for in vitro production of plant secondary metabolites . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
4.3. Host-independent biosynthesis of plant secondary metabolites in endophytes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
4.3.1. Case studies on taxol production . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
4.3.2. Case studies on camptothecin production . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
5. Strategies for sustainable and enhanced production of secondary metabolites in endophytes . . . . . . . . . . . . . . . . . . . . . . . . . . 0
5.1. Strain improvement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
5.1.1. Mutagenesis in endophytes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
5.1.2. Methods for genetic transformations in endophytes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
5.2. Bioprocess optimization strategies for enhanced secondary metabolite production in endophytes . . . . . . . . . . . . . . . . . . . . . 0
5.2.1. Optimization of fermentation parameters . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
5.2.2. Elicitor/inhibitor addition . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
5.2.3. Precursor feeding . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
5.2.4. Use of adsorbent resins/solid supports . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0

⁎ Corresponding author.
E-mail address: smita@iitm.ac.in (S. Srivastava).

http://dx.doi.org/10.1016/j.biotechadv.2015.07.004
0734-9750/© 2015 Elsevier Inc. All rights reserved.

Please cite this article as: Venugopalan, A., Srivastava, S., Endophytes as in vitro production platforms of high value plant secondary metabolites,
Biotechnol Adv (2015), http://dx.doi.org/10.1016/j.biotechadv.2015.07.004
2 A. Venugopalan, S. Srivastava / Biotechnology Advances xxx (2015) xxx–xxx

6. Strategies to unlock the cryptic/silenced gene clusters in endophytes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0


6.1. Co-cultivation and mixed fermentation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
6.2. Use of epigenetic modifiers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
7. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0

1. Introduction new bioactive molecule reported from a plant source, there follows re-
ports of endangered status or even extinction of a medicinally impor-
Fungi have been known to be a rich repository of medicinally tant plant due to over-harvesting. Hence, the focus turned toward
important compounds since the discovery of penicillin. Today, the fungi namely the “endophytes”, which reside within these medicinally
range of drugs derived from fungi stretch from antibiotics to immune- important plants and thus may have acquired their medicinal abilities.
suppressants to anti-cholesterol drugs (statins). While plants still The landmark in this area of endophyte bioprospecting was undoubted-
remain the major source of drugs or their lead molecules, with every ly the discovery of Taxomyces andreanae (Stierle et al., 1993), the first

Fig. 1. Structures of various bioactive molecules produced by endophytes. (a) Taxol, (b) camptothecin (c), vincristine (d), vinblastine (e), huperzine (f), podophyllotoxin (g), diosgenin and
(h) azadirachtin.

Please cite this article as: Venugopalan, A., Srivastava, S., Endophytes as in vitro production platforms of high value plant secondary metabolites,
Biotechnol Adv (2015), http://dx.doi.org/10.1016/j.biotechadv.2015.07.004
A. Venugopalan, S. Srivastava / Biotechnology Advances xxx (2015) xxx–xxx 3

endophyte reported to produce taxol, the billion dollar anti-cancer lead 2. What are endophytes? — definition, discovery and classification
molecule (Fig. 1a). The number of patents and publications that follow-
ed this discovery triggered high hopes of a sustainable alternative The term endophyte was coined by Heinrich Anton de Bary in 1884 to
production route using endophytic fungi for almost all the major originally define any organism occurring within plant tissues (Hyde and
life-saving drugs, without having to harvest the trees to meet the Soytong, 2008). Broadly this definition includes bacteria (Kobayashi and
ever-increasing market demands. Indeed the initial discovery was Palumbo, 2000), fungi (Stone et al., 2000), algae (Trémouillaux-Guiller
followed by a plethora of different endophytes reported to produce et al., 2002), insects (Tooker and Hanks, 2004) and other vascular plants
various other bioactive molecules including camptothecin, vincristine, (Marler et al., 1999). However, majority of the endophyte research has fo-
vinblastine, huperzine, podophyllotoxin, diosgenin and azadirachtin cused on endophytic fungi (Arnold and Lewis, 2005), almost making
(Zhao et al., 2011b) (Fig. 1b–h). However, despite numerous reports them synonymous with the term ‘endophyte’. These endophytes have
documenting the secondary metabolites from endophytes in the last been differentiated from mycorrhizae based on the three hallmarks of a
two decades (Table 1), there have been no major breakthroughs in mycorrhizal association which includes the absence of a localized inter-
terms of commercial exploitation of any endophyte as a source of bioac- face of specialised hyphae (present in most mycorrhiza), the absence of
tive molecules. Reports related to inconsistent production of many of synchronized plant–fungus development, and the lack of plant benefits
these biomolecules in the axenic culture of endophytes upon isolation with respect to nutrient transfer (Brundrett, 2004).
from the host plant have raised doubts on commercial feasibility The earliest records of the presence of endophytic fungi have come
of endophytes as sustainable production platforms. In this review, from the 400 million year old fossils of the early Devonian Rhynie
literature evidence available in support as well as in opposition of the chert deposits (Krings et al., 2007) which suggest that endophyte–
presence of host-independent biosynthetic machinery in endophytic plant associations may have evolved along with the evolution of higher
fungi has been presented. Recent advances in the bioprocessing plants. Modern day studies on endophytic fungi can be traced back to
of endophytes post the taxol boom have also been discussed. This re- mid-19th century. Based on differences in evolutionary relatedness,
view aims at highlighting the need for greater research on the complex taxonomy, plant hosts, and ecological functions, the endophytic
ecological interactions between endophytes and their host plant as well fungi are classified as the clavicipitaceous or grass endophytes
as with other endophytic microbiota in the host. A deeper understand- (C-endophytes) and the non-clavicipitaceous endophytes (NC-
ing of these interactions, when combined with rational bioprocess endophytes) of non-vascular plants, ferns, conifers, and angiosperms
optimization, will enable the provision of a culture environment (Rodriguez et al., 2009), with the latter being further separated into
which is conducive to sustainable and maximum production of high three functional classes based on host colonization patterns, mechanism
value secondary metabolites, during in vitro cultivation of potential of transmission between host generations, in planta biodiversity levels,
endophytes. and ecological function. Interest in the NC-endophytes has now

Table 1
Examples of high value secondary metabolites isolated from endophytes.

Compound Bioactivity Endophyte strain Referencea

Azadirachtin A and B Natural pesticide Eupenicillium parvum Kusari et al. (2012b)


Berberine Antibiotic, anti-diabetic, anti-inflammatory, anti-cancer. S6 Gao et al. (2008)
Camptothecin Anti-cancer Several strains Zhao et al. (2010)
Cytochalasins Anti-cancer, antibiotic Rhinocladiella sp., Strobel and Daisy (2003)
Digoxin glycoside Cardiotonic, diuretic, anti-epileptic Several strains Kaul et al. (2012)
Diosgenin Progesterone precursor, cholesterol lowering activity Cephalosporium sp. Zhao et al. (2010)
Paecilomyces sp.
Emodin Anti-diabetic, anti-viral, anti-cancer Thielavia subthermophila Zhao et al. (2010)
INFU/Hp/KF/34B
Gentiopicrin Anti-malarial fungicide and larvicide QJ16 and QJ18 Yin et al. (2009)
Ginkgolide B Used in treatment of cardiovascular diseases, cerebrovascular diseases Fusarium oxysporum SYP0056 Cui et al. (2012)
and migraines
Gymnemagenin Anti-diabetic Penicillium oxalicum Parthasarathy and Sathiyabama (2014)
Huperzine A Potential treatment for neurodegenerative diseases Several strains Zhao et al. (2010)
Hypericin Anti-depressant, anti-inflammatory, anti-microbial, anti-oxidant, anti-viral T. subthermophila Zhao et al. (2010)
INFU/Hp/KF/34B
Kaempferol Anti-oxidant, anti-cancer Fusarium chlamydosporum Chaturvedi et al. (2014)
Lapachol Anticancer Aspergillus niger Nirupama et al. (2011)
Alternaria alternata
Piperine Antimicrobial, antidepressant, anti-inflammatory, anti-oxidative, Periconia sp. Verma et al. (2011)
anti-cancer
Podophyllotoxin Anti-cancer Several strains Zhao et al. (2010)
Quercetin glycoside Anti-hypertensive, anti-cancer, anti-inflammatory, anti-oxidant Penicillium sp. Gh01 Nair and Padmavathy (2014)
Rhein Antitumor, anti-inflammatory, antimicrobial and hemostatic Fusarium solani R13 You et al. (2013)
Rohitukine Anti-inflammatory, immuno-modulatory, anti-cancer Fusarium proliferatum (MTCC 9690) Kumara et al. (2012)
Sanguinarine Anti-cancer F. proliferatum BLH51 Wang et al. (2014b)
Sipeimine Antitussive and expectorant Fritillaria ussuriensis Fu7 Yin and Chen (2008)
Subglutinols A and B Immune-suppressants Fusarium subglutinans Lee et al. (1995)
Tanshinone I and IIA Cardiotonic, anti-inflammatory Emericella foeniculicola TR21 Ma et al. (2011)
Trichoderma atroviride D16 Ming et al. (2012)
Taxol Anti-cancer Several strains Zhao et al. (2010); Strobel and Daisy (2003);
Strobel et al. (2001); Strobel et al. (1999)
Toosendanin Digestive tract-parasiticide, selective presynaptic blocker, effective O-L-5, O-SC II-4, O-RC-3 Zhao et al. (2010)
anti-botulismic agent; agricultural insecticide
Vincamine Vasodilator, cerebral stimulant XM-J2 Yin and Sun (2011)
Vincristine & vinblastine Anti-cancer Several strains Zhao et al. (2010)
a
A single representative reference for each compound is provided — more examples exist.

Please cite this article as: Venugopalan, A., Srivastava, S., Endophytes as in vitro production platforms of high value plant secondary metabolites,
Biotechnol Adv (2015), http://dx.doi.org/10.1016/j.biotechadv.2015.07.004
4 A. Venugopalan, S. Srivastava / Biotechnology Advances xxx (2015) xxx–xxx

increased due to their diversity, ecological roles, potential applications metabolic machinery of endophytes, the example of camptothecin
and the ability to switch between endophytic and other lifestyles. producing endophytic fungi will be discussed here.
The evolutionary role of camptothecin and its derivatives include
3. Host–endophyte relationship — role in host plant evolution, protecting plants against heat shock (Zu et al., 2003) and defending
defense and secondary metabolite production them against insect and pathogen attack by binding and inhibiting
the DNA topoisomerase I (Sirikantaramas et al., 2009). While the
All the plant species, including non-vascular plants, ferns, conifers camptothecin biosynthetic pathway in plants has not been completely
and angiosperms, are believed to be in symbiotic relationship with elucidated, many of the rate-limiting enzymes have been identified
endophytic fungi (Rodriguez et al., 2009). Many plants harbor as (Sun et al., 2011). In turn, a host-dependent cross species biosynthetic
many as hundreds of endophytes within apparently healthy tissues pathway for camptothecin production in endophytic fungi has been
(Stone et al., 2000). The co-evolution of fungi and land plants may proposed by Kusari et al. (2011b), where some of the fungal biosynthet-
have begun much earlier than the early Devonian period. It has been ic enzymes were shown to be homologous to their host counterparts
hypothesized that the early mutualistic symbiosis between an alga which could be the result of horizontal gene transfer (HGT) during co-
and fungal partners catalyzed the evolution of the modern land plants evolution. Both the host plant and its endophytes have also developed
from their aquatic ancestors with the fungal partner enabling the several intrinsic as well as co-evolutionary resistance mechanisms in-
survival of the ancestors of land plants against drought, mineral volving mutations in the Top1 gene in order to protect themselves
deficiencies, UV radiation and temperature fluctuations which charac- against endogenously produced camptothecin (Kusari et al., 2011a).
terized the early Paleozoic land (Selosse and Le Tacon, 1998). Indeed, Thus, it can be said that every aspect of growth, development and
endophytic fungi are known to confer upon their host plant adaptation survival of the endophytes and that of their plant hosts is dependent
to stresses like salinity, tolerance to drought, metals, disease, heat, and on their co-evolution which is characterized by a balanced antagonism,
herbivory, and also promote growth via biosynthesis of plant hormones the hallmark of endophytism (Schulz et al., 1999).
and nutrient acquisition (Rodriguez et al., 2009). For example, the The fact that endophytic fungi are found to mimic the host plant
symbiotic dependence of Dichanthelium lanuginosum plants on their secondary metabolite profile also led to a possibility that these plant
endophytic Curvularia sp. has been reported for thermoprotection, and metabolites could in fact be a product of their respective endophytes
that of the grass species Leymus mollis with its endophyte Fusarium (Kusari et al., 2012a). This has been demonstrated in the case of
culmorum has been reported for salinity tolerance (Rodriguez et al., ansamitocin, a potent antitumor maytansinoid originally isolated from
2009), with the endophyte also benefitting due to the symbiotic plants belonging to Celastraceae, Rhamnaceae, and Euphorbiaceae
association in both cases. families and later from the actinomycetes Actinosynnema pretiosum
One of the tools used by the endophytic fungi for aiding in the ssp. pretiosum and A. pretiosum ssp. auranticum. Although initially
survival of their host plants is the production of secondary metabolites. isolated from the shrub Maytenus serrata, unlike other plant metabo-
Two theories have been proposed to explain how endophytes may be lites, detailed characterization of the ansamitocin biosynthetic gene
assisting the host plants in chemical defense (Kusari et al., 2012a). The clusters has been carried out only in the actinomycete A. pretiosum
mosaic theory (Carroll, 1991) proposes that endophytes create a ssp. auranticum rather than in the plant by Yu et al. (2002). Further,
heterogenous chemical composition within and among otherwise ge- the lack of evidence for the production of maytansinoids from cultured
netically uniform plant organs thus varying their worth for herbivores plant cells (callus and suspension) and the absence of a key
or susceptibility to infection by pathogens, while a parallel theory maytansinoid biosynthetic gene in plants substantiated the possibility
proposed by Arnold et al. (2003) suggests that endophytes might act that the true biosynthetic source of the maytansinoid backbone could
as “acquired immune systems” in their host plants. Examples of second- be a bacterial endophyte and symbiosis could have triggered the
ary metabolites produced by endophytes, with known role in deter- production in plants (Cassady et al., 2004).
rence of herbivory, include peramine, ergot and indole diterpene
alkaloids, lolines and swainsonine — a trihydroxyindolizidine alkaloid 4. Using endophytes as in vitro production platforms for plant
(Panaccione et al., 2014). A recent report has found a direct correlation secondary metabolites
between the variation in taxol content among different parts of a Taxus
plant and the quantity of a resident taxol producing fungal endophyte, 4.1. Limitations of producing bioactive secondary metabolites from plants
wherein depletion of the endophyte via treatment with fungicides re-
sulted in decreased expression of taxol biosynthesis genes in the plant Several statistics point at how indispensable plants are as secondary
and hence in reduced plant taxol accumulation (Soliman et al., 2013). metabolite sources. Twenty five percent of the drugs prescribed world-
wide originate from plants, with plant-based drugs accounting for 11 %
3.1. Production of plant secondary metabolites in endophytes — the of the 252 basic and essential drugs recognized by the WHO (Dubey
possibility et al., 2012). At least 120 distinct plant-based active compounds are
being used as important drugs in one or more countries (Taylor,
Although endophytic fungi have been researched for more than a 2000). Natural products, especially plant-derived, have contributed to
century now, it is only in the last twenty years that they have been 47% of the anticancer drugs in the market (Newman and Cragg, 2007).
reported as being capable of mimicking the secondary metabolite reper- However, the indispensability of plant-based bioactive molecules unfor-
toire of the host plant. The “xenohormesis” hypothesis (Howitz and tunately brings with it several disadvantages — (i) extremely low yields
Sinclair, 2008) states that “heterotrophs”, due to evolutionary selection which vary with environmental conditions, (ii) limited supply leading
pressures, have evolved the ability to sense signaling and stress-induced to over-harvesting, and (iii) extremely complex structures which
molecules from plants, with this sensing ability being retained even if make total synthesis and semi-synthesis very challenging. The current
the capacity to biosynthesize these compounds is lost over time. This demand–supply statistics for various high value phytochemicals are
lends credence to the idea that the stress signaling in plants is also listed in Table 2. Over-harvesting of trees to meet the increasing market
sensed by the endophytic fungi which in turn mount defense responses demand has led to their endangerment and even extinction in some
similar to the host plant. Hence, it naturally follows that conserved gene cases. While the tropical rainforests, which are some of the richest and
clusters would exist across plants and their endophytes, which might be yet untapped sources of high value metabolites, are rapidly plummeting
activated by suitable associations leading to the production of similar on one hand from 14% to a mere 6%, less than 1% of these trees and
secondary metabolites in both plant and fungi (Kusari et al., 2012a). In plants have been subject to bioprospecting so far, essentially implying
order to explain how interactions with the host plant shape the that hundreds of valuable medicines could be lost to us forever before

Please cite this article as: Venugopalan, A., Srivastava, S., Endophytes as in vitro production platforms of high value plant secondary metabolites,
Biotechnol Adv (2015), http://dx.doi.org/10.1016/j.biotechadv.2015.07.004
A. Venugopalan, S. Srivastava / Biotechnology Advances xxx (2015) xxx–xxx 5

being discovered (Taylor, 2005). Hence, search for alternative and the fermentation period shorter with lesser risk of contamination in
sustainable sources of these high value plant secondary metabolites is comparison to that with slow growing plant cells/tissue cultivations.
the need of the hour. Optimization, regulation of fermentation parameters and subsequent
scale-up is relatively easier in microbial fermentations in comparison
4.2. Alternative methods for in vitro production of plant secondary to that in plant cell/tissue cultivations in bioreactors. Moreover, micro-
metabolites bial cultures are easily amenable to yield enhancement strategies like
strain improvement, precursor feeding, elicitor addition, appending in-
In vitro plant cell/tissue cultures, including callus and hairy root hibitors and co-cultivation (Zhao et al., 2010). Microbial fermentation of
cultures, have been extensively reviewed for mass production of plant endophytes can thus relieve the threat of over-harvesting of plants
secondary metabolites (Mulabagal and Tsay, 2004; Srivastava and while ensuring a steady supply of important plant secondary metabo-
Srivastava, 2007). However, limitations with plant cell/tissue culture lites, irrespective of environmental conditions.
technology include genomic instability, low yields, and scale-up difficul-
ties resulting in only few success stories at the industrial level, like taxol 4.3. Host-independent biosynthesis of plant secondary metabolites in
production by Phyton biotech and production of shikonin, ginseng and endophytes
berberine by Mitsui Chemicals (Linden, 2006).
Total chemical synthesis, another alternative, has been explored While there are many reports available on the isolation of endophyt-
for most of the important metabolites with success in some cases ic fungi capable of producing high value plant derived drugs, no break-
(e.g. vanillin — most of its demand is met through chemical synthesis) throughs have been reported yet on the application of these strains for
and not so in most other cases due to the sterical complexity (e.g. mor- the commercial production of high value phytochemicals. A widely ac-
phine has five chiral centers, which makes its chemical synthesis knowledged (and reported) problem of subsequent attenuation in the
complex and uneconomical) and structural complexity (e.g. chemical product yield with subculture of endophytes under axenic conditions
synthesis of paclitaxel involves 40 reactions with harsh solvent and is one of the major limitations in the commercialization of this
low product yield which makes its total synthesis impractical) bioprocess. Scientific investigators have questioned the existence of
(Wilson and Roberts, 2012). host-independent biosynthetic machinery in endophytes due to the in-
The discovery of endophytic fungi capable of mimicking the second- consistent production of secondary metabolites observed in vitro. On
ary metabolite repertoire of their host in most of the medicinally or oth- the contrary, sustainable production of metabolites like tanshinone IIA
erwise industrially valued plants has given an impetus to the possibility and taxol by the axenic culture of endophytic fungi up to 10 generations
of using microbial fermentation to meet the ever-growing demand for has also been reported (Ma et al., 2011; Zhao et al., 2013b). Thus, there
several life-saving drugs. Moreover, endophytes have been known to exists evidence for both supporting the claims of a host-independent
evolve novel resistance mechanisms to the toxic metabolites produced machinery and for opposing it. Three schools of thought exist on the or-
by them and the host as a co-evolutionary adaptation (Kusari et al., igins of secondary metabolism in plants, all of which emphasize on the
2011a; Mu et al., 1999), which simplifies bioprocessing of these organ- role of endophytes — 1.) Both plants and endophytic microbes may have
isms without the risk of growth inhibition due to self-toxicity of the co-evolved parallel pathways to produce these natural products;
product. 2.) Horizontal gene transfer (HGT) may have taken place between
Microbial fermentation has certain inherent advantages that make plants and microbes during their long period of association; 3.) Either
this strategy more robust than others. The culture medium for fungal plants or endophytic fungi may produce these secondary metabolites
cells is relatively simple and inexpensive, largely consisting of industrial and transfer them to the other symbiont (Karuppusamy, 2009). Further,
by-products/wastes like corn steep liquor and molasses, whereas plant the xenohormesis hypothesis supports the idea that endophytes might
cell culture medium requires expensive additives like phytohormones have the machinery to synthesize the host plant metabolites (Kusari
which increase the production cost. Faster growth rate of fungi makes et al., 2012a). Recent studies have reported the presence of key

Table 2
Demand–supply statistic for various high value metabolites derived from plants.

Compound Uses Demand and supply statistics Reference

Taxol Antitumor • Annual sales in 2006 = 3.7 billion USD Malik et al. (2011)
• Annual demand = 600 kg
• Treatment per patient consumes about eight 60-year-old yew trees
• 10,000 kg of Taxus bark or 3000 yew trees = 1 kg of taxol
Camptothecin Antitumor • Supplied from two plants — Camptotheca acuminata and Nothapodytes foetida — both critically endangered Patwardhan (2006)
• ~1000–1500 tons of wood chips = 1 ton of camptothecin
• Annual sales in 2003 = 1 billion USD
• Annual biomass demand in India in 2006 = 500–700 metric tons
• Annual biomass demand in Japan in 2006 = 1000 metric tons
• Global camptothecin demand in 2011 required 100 million trees
Vinca alkaloids Antitumor • Very low yield from plant (0.0003% DW) Hendrawati et al.
• Annual demand = 0.3 tons (2012)
• Annual sales = 200 million USD
• Production via plant cell cultures unsuccessful so far
Digitalis Cardiotonic • Annual demand = 1000 metric tons plant material Mangathayaru (2013)
glycosides • Patients require 1 mg/day
• Worldwide drug demand = several thousand kilograms per year
• India — annual demand ≥30 quintals
Diosgenin Progesterone • Accounts for 2/3rds of the total world consumption of steroids Dangi et al. (2014)
precursor • Annual demand = 3000 tons
• India — annual demand = 150 tons.
• India total production = 30 tons
Podophyllotoxin Antitumor • Only 2 plant sources known — Podophyllum hexandrum and Podophyllum peltatum — both endangered Alam et al. (2009)
• Annual demand ≥100 metric tons
• Annual supply = 50–80 tons

Please cite this article as: Venugopalan, A., Srivastava, S., Endophytes as in vitro production platforms of high value plant secondary metabolites,
Biotechnol Adv (2015), http://dx.doi.org/10.1016/j.biotechadv.2015.07.004
6 A. Venugopalan, S. Srivastava / Biotechnology Advances xxx (2015) xxx–xxx

biosynthetic genes in taxol and camptothecin producing endophytic complexes with the endophyte cell wall structures that would persist
fungal strains (Kusari et al., 2011b, 2014). Whole genome sequencing for up to a few passages of the fungal cultures. However, a very recent
of an endophytic Shiraia sp. Slf14, previously reported to produce both study reported by Yang et al. (2014c) on the whole genome sequencing
huperzine A and hypocrellin A, has also revealed the presence of a puta- of taxol producing endophyte Penicillium aurantiogriseum NRRL 62431
tive huperzine A (HupA) biosynthetic gene cluster (Yang et al., 2014a). directly contradicts the findings of Heinig et al. (2013). When the fungal
These findings provide strong indications that endophytic fungi are genome was scanned via multiple sequence alignments for potential
secondary metabolite factories by themselves. To further elaborate on paclitaxel biosynthesis genes, putative genes showing low homology
the various contradictory evidences available, the taxol and with the Taxus genes but with distinct conserved amino acid sites
camptothecin producing endophytic fungi have been discussed below emerged, supporting the independent evolution theory over HGT. Out
as useful case-studies. of 13 known paclitaxel genes, only 7 potential homologs were identified
in NRRL 62431, which supports the divergence of the two biosynthetic
4.3.1. Case studies on taxol production pathways with conservation of only specific enzyme sites important
The list of taxol producing endophytic fungi is an ever-growing one, for the activity rather than the whole protein structure. The
with variable taxol yields (from nanogram to milligram per liter media) P. aurantiogriseum strain NRRL 62431 continued to produce taxol for
and loss in productivity with subculture. This has prompted some more than twelve passages without any contact with the host plant,
workers to question whether the source of the fungal taxol is in fact thus ruling out the possibility of passive taxane accumulation in fungal
the host plant pool. However, the isolation of these endophytes from cell walls. Moreover, the isolation of taxol producing endophytes from
non-taxol producing trees has also been reported (Flores-Bustamante non-taxol producing plants like Citrus medica and Taxodium distichum
et al., 2010; Li et al., 1998a), which contradicts the idea of fungal taxol leaves the debate open on how these endophytic fungi acquired the
being a host plant adduct. In addition, de-novo synthesis of paclitaxel ability to produce taxol independently.
in axenic culture of a fungal endophyte was demonstrated by Stierle
et al. (1993) by feeding of [1–14C] acetic acid and L-[U-14C] phenylala- 4.3.2. Case studies on camptothecin production
nine as precursors. Several studies have reported the presence of taxol The key enzymes identified in the biosynthetic pathway of
biosynthetic genes like ts, dbat and bapt in these endophytes using camptothecin so far include tryptophan decarboxylase (TDC), geraniol
primers designed based on published sequences from Taxus trees. A 10-hydroxylase (G10H), secologanin synthase (SLS) and strictosidine
comprehensive list of taxol genes identified in endophytes has been synthase (STR). In the camptothecin-producing endophytic fungal
documented by Kusari et al. (2014). In most cases, the fungal genes strain of Fusarium solani INFU/Ca/KF/3, Kusari et al. (2011b) reported
identified have been found to be homologous to their plant counter- the presence of TDC, G10H and SLS genes by using primers designed
parts (with N96% sequence similarity) (Staniek et al., 2009; Zhang from corresponding plant gene sequences. These fungal genes were
et al., 2009a, b; Zhou et al., 2007), which strongly supports the horizon- highly homologous with their plant counterparts. However, no amplifi-
tal gene transfer (HGT) theory. cation product was obtained with any STR gene-specific primer and so it
However, the sheer complexity of the taxol biosynthetic pathway, was argued that the endophyte might be using the host STR pool for
which involves more than 20 steps localized to different subcellular camptothecin biosynthesis. However, neither artificial reconstitution
compartments with genes possibly scattered over different plant into the host nor supplementing with various host plant tissue extracts
chromosomes, has raised doubts over the possibility of transfer of the could restore the camptothecin biosynthesis in the strain which showed
entire pathway from the host plant to the endophyte (Heinig et al., complete product attenuation after seven generations of subculture. To
2013). Further, the occurrence of horizontal gene transfer between explain the yield attenuation observed upon sub-culturing, the strain in
distantly-related organisms is a rarity, constrained by the amount of its seventh generation subculture was probed for the presence of the
genetic information transferred and other genetic barriers, which biosynthetic genes. While STR was not detected, TDC, G10H and SLS
contradicts the observation of taxol biosynthesis in many different gene products were obtained, but the homology with the plant counter-
endophytic fungi (Kurland et al., 2003). However, some studies offer parts was much lesser indicating random mutations. It was therefore
support to the idea that the microbial taxol gene cluster exists indepen- proposed that the lack of in planta selection pressure under axenic
dent of the plant genes having co-evolved with the plant machinery conditions caused immediate degradation of the partial camptothecin
rather than being acquired through HGT. This has been well established biosynthetic machinery possessed by the fungus, thus completely
in the case of gibberellin biosynthetic pathways in fungi and higher impairing its ability to synthesize camptothecin even when the host–
plants which show several differences and have evolved independently endophyte association was restored. Contrary to these observations,
rather than by HGT (Hedden et al., 2001; Tudzynski, 2005). Similar find- there exist reports on other endophytic strains that have been shown
ings have been reported in case of taxol producing endophytes where to sustain camptothecin production in axenic cultures up to several
the fungal ts and bapt genes have been found to have low similarities generations (Pu et al., 2013).
to the corresponding plant genes (Xiong et al., 2013; Yang et al., When whole genome sequences of 53 fungi (including plant and
2014c), which suggests and supports co-evolution theory rather than animal fungi) were analyzed, there was no detection of STR or any
HGT theory. other terpene indole alkaloid (TIA) pathway downstream genes includ-
In a detailed study conducted on various taxol producing endophyte ing those involved in vincristine and vinblastine synthesis or even the
strains including T. andreanae, Heinig et al. (2013) found no fungal taxol biosynthesis genes (Sachin et al., 2013). This finding is supported
genomic sequence related to known taxane-specific sequences from by studies on the evolution of STR and strictosidine synthase-like pro-
yew trees, thus ruling out the possibility of horizontal gene transfer. In teins (SSLs), where the latter has been reported from plants, algae and
order to test the other possibility, which is the existence of an indepen- cyanobacteria but rarely from fungi (Sohani et al., 2009). Three different
dently evolved biosynthetic pathway, shotgun sequencing of the full hypotheses have been proposed by Sachin et al. (2013) to explain
genome of the strains T. andreanae and EF0021 was done, revealing camptothecin production by endophytic fungi and attenuation on sub-
only certain putative terpene synthases which were neither similar to culturing with specific reference to STR. The first hypothesis is that
each other nor homologous with plant terpene synthases. However, a STR function in fungi is carried out by a new SSL or a totally different
significant degree of sequence similarity would be expected of enzymes protein, which would explain why conventional attempts at detecting
that catalyze reactions as complex as taxadiene synthesis even if the STR gene or its orthologs would fail. If such a gene did exist, both the
evolution of the plant and fungal biosynthetic pathways happened production of camptothecin and its attenuation could be explained,
independently. Hence, the authors concluded that fungal taxane was the latter due to gene silencing or methylation process under axenic
in fact residual plant taxane which, due to its lipophilicity, formed conditions. The second hypothesis argues for the presence of the STR

Please cite this article as: Venugopalan, A., Srivastava, S., Endophytes as in vitro production platforms of high value plant secondary metabolites,
Biotechnol Adv (2015), http://dx.doi.org/10.1016/j.biotechadv.2015.07.004
A. Venugopalan, S. Srivastava / Biotechnology Advances xxx (2015) xxx–xxx 7

gene on certain extra-chromosomal elements (ECEs) in the fungal cyto- taxol producing endophytes. Table 3 lists examples of usage of muta-
plasm while the third hypothesis proposes that the STR gene is carried genesis for increasing yield of various metabolites from endophytes.
in a plasmid within endohyphal bacteria with the genes being acquired Mutagenesis on mycelium has the disadvantage of genetic
via HGT in both cases. Product attenuation in these two cases is ex- separation in off-springs and it is difficult to control and optimize the
plained by the loss of the ECEs including plasmids and the endohyphal conditions for mutagenesis in the case of spores, while mutagenesis
bacteria upon repeated sub-culturing of the axenic fungal culture. Re- on protoplasts does not have these limitations (Kai et al., 2009). Further,
ports on the presence of plasmids carrying secondary metabolic gene the lack of a cell wall makes protoplasts more environment-sensitive
clusters in antibiotic producing Streptomyces species give strong support and so protoplast mutagenesis is a very useful strategy for yield en-
to the latter two hypotheses (Medema et al., 2010; Mochizuki et al., hancement in endophytic fungi. Most of the studies so far have focussed
2003). However, evidences from endophytes to prove these hypotheses on optimization of the conditions for protoplast preparation and regen-
are yet to emerge. eration as factors including pH, temperature, time, enzyme combina-
tions, osmotic stabilizers, pre-treatment, regeneration medium and
5. Strategies for sustainable and enhanced production of secondary method significantly affect the success rate (Xu et al., 2006b; Zhao
metabolites in endophytes et al., 2004; Zhou et al., 2008). There are a few reports on product
yield enhancement achieved via protoplast mutagenesis and regenera-
In order to realize and completely tap the secondary metabolite ar- tion (Kai et al., 2009; Zhang et al., 2011b; Zhao et al., 2005b; Zhou
senal of endophytic fungi on an industrial scale, yield and productivity et al., 2005). Protoplast fusion is another technology which has been
enhancement strategies at several levels are required. A combination applied to endophytes for enhancing product yields. A high-level taxol
of genetic/metabolic and bioprocess engineering may be used to sustain producing strain HDF-68 was obtained by inactivated protoplast fusion
and enhance production of high value secondary metabolites from of the two mutant strains UV40–19 and UL50–6 of Nodulisporium
endophytic fungi. While the potential of endophytes as sources of high sylviforme (Zhao et al., 2013b). Compared to the parent strains, taxol
value phytochemicals and novel natural products is enormous, the production increased to 468.62 μg/L i.e. 24.51% and 19.35% compared
yields are insufficient to be commercially exploited. Hence, the develop- with UV40–19 and UL50–6, respectively.
ment of high yielding endophytic fungal strains (via strain improve-
ment techniques) which are able to synthesize metabolites at 5.1.2. Methods for genetic transformations in endophytes
commercially feasible levels is required. These improved strains can Most studies on the genetic engineering of endophytic fungi are
further be subjected to various bioprocess optimization strategies still at a preliminary stage, focusing on the development of efficient
well known in microbial fermentations for further enhancement in methods for fungal DNA transformation. However, the success of
secondary metabolite yield and productivity. these studies provides a strong platform for future research on the
insertion of key biosynthetic and/or regulatory genes via utilization of
improved genetic transformation methods for sustaining and enhanc-
5.1. Strain improvement ing secondary metabolite production from the axenic culture of the
endophytes. Currently, most of the available reports on genetic manip-
Strain improvement involves not just product yield enhancement ulation of endophytes are on taxol producing endophytes. Long et al.
but also improvement of other characteristics including removal of un- (1998) used a protoplast transformation protocol in the endophytic
wanted co-metabolites, improved utilization of inexpensive carbon and fungus, Pestalotiopsis microspora while developing molecular genetic
nitrogen sources and alteration of cellular morphology for improved ox- techniques to elucidate the fungal taxol biosynthetic pathway.
ygen transfer in the fermenter or for ease in downstream processing Unexpectedly, the majority of P. microspora transformants contained
(Barrios-Gonzalez et al., 2003). Two strategies are used either separate- extrachromosomal DNAs with terminal telomeric repeats which can
ly or in concert — 1) mutagenesis followed by random/rational play an important role in adaptive and developmental implication. A
screening and protoplast fusion techniques and 2) genetic engineering procedure for PEG-mediated transformation of the taxol producing en-
that includes secondary metabolite gene cluster amplification, gene dophytic fungal strain Ozonium sp. EFY-21 was established by Wei et al.
disruption, and cloning of regulatory genes. (2010). Yamin et al. (2012) successfully used this protocol to construct a
fungal expression vector (pV2+-TS-pAN7-1) containing the taxadiene
5.1.1. Mutagenesis in endophytes synthase gene (ts), encoding for a rate-limiting enzyme in taxol
This technique involves the use of mutagens to induce changes in biosynthesis pathway, and used it to generate a five times higher
the genetic characteristics of organisms. Mutagens employed may be taxol-yielding mutant of the endophyte, Ozonium sp. EFY-21. Liu et al.
physical (ultraviolet (UV), X-ray, γ-rays, laser, microwave, ion beams (2013) established an Agrobacterium tumefaciens mediated genetic
etc.) or chemical (ethyl methyl sulfonate (EMS), nitrosoguanidine transformation (ATMT) method for a taxol-producing fungus, Ozonium
(NTG), sodium nitrite (NaNO2), diethyl sulfate (DES)). Selection of sp. EFY21, which greatly enhanced the transformation efficiency com-
improved mutants is done either through random screening for the pared to the traditional PEG-mediated protoplast transformation. A
desired trait or through rational screening which involves selection for similar ATMT protocol was developed for the genetic transformation
a particular characteristic of the desired genotype that is different of a taxol-producing Cladosporium cladosporioides strain MD2 (Zhang
from the one of final interest (Barrios-Gonzalez et al., 2003) et al., 2011a). Yet another method of transformation, restriction
e.g. hygromycin resistant mutants are frequently selected to screen fur- enzyme-mediated integration (REMI) was used for transformation of
ther for increased taxol production (Kai et al., 2009). Compound muta- a taxol-producing endophytic Ozonium sp. strain BT2 (Wang et al.,
tions are also induced by using two or more mutagens in order or using 2007). Compared to conventional transformation methods, REMI
only one mutagen but repeatedly or using two or more mutagens at the increases transformation rate and the frequency of single-copy integra-
same time in order to utilize the interaction between mutagens (Zhou tion events (Bӧlker et al., 1995; Manivasakam et al., 2001). Spiering et al.
et al., 2010). While the exact mechanism behind the observed increase (2005) have reported the application of gene silencing technique like
in the production of secondary metabolites in microbes upon mutation RNAi in fungal endophytes.
is unclear, genetic studies indicate that mutation may be causing chang- Apart from taxol producing endophytes, other microorganisms too
es in regulatory genes, thereby leading to phenotype changes as well as have been utilized for taxol production via gene overexpression.
increased production of metabolites (Adrio and Demain, 2006). Examples include preparation of taxadiene by overexpression of genes
Mutagenesis in fungal endophytes can be carried out using protoplasts, encoding isopentenyl diphosphate isomerase, geranylgeranyl diphos-
mycelium or spores. This strategy has been very successfully applied to phate synthase, and taxadiene synthase in Escherichia coli (Ajikumar

Please cite this article as: Venugopalan, A., Srivastava, S., Endophytes as in vitro production platforms of high value plant secondary metabolites,
Biotechnol Adv (2015), http://dx.doi.org/10.1016/j.biotechadv.2015.07.004
8 A. Venugopalan, S. Srivastava / Biotechnology Advances xxx (2015) xxx–xxx

Table 3
Examples on usage of mutagenesis for improvement in product yield from endophytes.

Product Wild-type strain Mutant strain Mutagen Yield enhancement Reference

Taxol HD1-3 UN25-3 UV, NTG and UV + NTG 1.4 fold Kai et al. (2008)
Taxol HD1-3 UD (14–1) UV + DES 1.34 fold Zhao et al. (2011e)
Taxol Nodulisporium sylviforme HQD33 N. sylviforme NCEU-1 UV, EMS, 60Co (60Co-γ-ray) and NTG 2.5 fold Zhou et al. (2001)
Taxol Fusarium maire strain Y1117 F. maire K178 UV + DES 8.6 fold Xu et al. (2006a)
Berberine S6 S-NU-302 UV, X-rays and NaNO2 170% Gao et al. (2008)
Tanshinone IIA Emericella foeniculicola TR21 E. foeniculicola NU152 UV + NaNO2 1.46 fold Ma et al. (2011)
Vincamine XM-J2 XM-UL-7-5 UV-laser 2.1 fold (62.3% increase in biomass) Yin and Sun (2011)

et al., 2010) and expression of eight taxoid biosynthetic genes in yeast Ravishankar, 2002). Strategies like media manipulation, culture
from episomal vectors (Dejong et al., 2006). However, endophytic condition optimization, elicitor addition, precursor feeding and use of
fungi from the host plant offer certain advantages over other adsorbent resins have been adopted in axenic cultures of endophytic
microorganisms where genetic manipulation for product enhancement fungi for enhancing the yield of high value secondary metabolites.
is concerned. Specifically, the sheer complexity of biosynthetic pathways Some of these strategies used in literature are discussed below.
(e.g. taxol biosynthesis involves almost 20 enzymatic reactions) makes
expression of all the genes in a foreign microorganism a very tedious 5.2.1. Optimization of fermentation parameters
process. On the other hand, the existence of at least partial biosynthetic The importance of providing the right combination of culture condi-
enzyme machinery in endophytes makes the genetic manipulation of tions for enhancing the production of secondary metabolites via
endophytic fungi easier. fermentation technology is well known. By optimizing and controlling
fermentation parameters like medium composition, pH, temperature,
5.2. Bioprocess optimization strategies for enhanced secondary metabolite agitation, and photoperiod, a sustainable, economic and environment
production in endophytes friendly bioprocess with consistent product quality and quantity
independent of the environmental variations can be developed for
Use of various bioprocess optimization strategies for yield enhance- industrial scale-up. Bode et al. (2002) coined the term “one strain
ment of secondary metabolites has been successful with plant cell/ many compounds” (OSMAC), to describe how a single microbial strain
tissue cultivations and has been extensively reviewed (Chattopadhyay can be induced to produce many compounds by simply varying the cul-
et al., 2002; Mulabagal and Tsay, 2004; Ramachandra Rao and ture parameters. In some cases, the OSMAC approach has even led to the

Table 4
Examples of culture parameter optimization for improvement in product yield from endophytes.

Product Strain Optimized parameters Methodology Yield enhancement Reference

Taxol Fusarium mairei UH23 Initial pH, inoculation volume, working Single factor 10.2% Dai and Tao (2008)
volume, temperature, carbon and
nitrogen source, fermentation period
Taxol Mutant F. maire K178 Nitrogen source and trace elements Plackett Burman design 1.3 fold Xu et al. (2006a)
and RSM
Taxol Nodulisporium sylviforme pH, temperature, agitation rate, Single factor 1.15 fold (including Zhao et al. (2011d)
UV40–19 fermentation period elicitors)
Taxol Pestalotiopsis microspora Monobasic sodium phosphate Single factor 2.2 fold (at 1 μg/mL) Li et al. (1998b)
Ne32
Palmarumycin C13 Berkleasmium sp. Dzf12 Metal ions RSM 6 fold Mou et al. (2013)
Palmarumycin C13 Berkleasmium sp. Dzf12 Carbon and nitrogen sources Plackett Burman design 2.5 fold Zhao et al. (2013a)
and RSM
Exo polysaccharide (EPS) Berkleasmium sp. Dzf12 Carbon and nitrogen sources, trace Fractional factorial design 6.29 fold Li et al. (2012a)
elements and RSM
Zofimarin Xylaria sp. Acra L38 Carbon and nitrogen sources Orthogonal array design, 8 fold Chaichanan et al.
Plackett Burman design (2014)
and RSM
Beauvericin Fusarium redolens Dzf2 Carbon and nitrogen sources, initial pH Plackett Burman design 1.27 fold Xu et al. (2010)
and RSM
Huperzine Colletotrichum Temperature, pH, agitation rate, Single factor 28.58% Zhao et al. (2013c)
gloeosporioides ES026 fermentation period
Carbon source 51.89%
Berberine Mutant strain S-NU-3-2 Carbon and nitrogen sources, L16 (43) orthogonal 47.2% Hong and Yang
illumination condition, temperature design (2010)
Mycoepoxydiene Phomopsis sp. Hant25 Complex media — Czapek yeast Single factor 10.3 fold Thammajaruk et al.
autolysate broth (CzYB), Malt Czapek (2011)
broth (MCzB) and modified M1D
medium (MM1D); shaking/static period
Camptothecin Fusarium oxysporum NFX06 Glucose, peptone and MgSO4 RSM 1.02 fold Musavi et al. (2014)
Camptothecin Trichoderma atroviride Medium composition, fermentation time, Single factor 50 to 75 fold (including Pu et al. (2013)
LY357 pH, temperature, agitation rate elicitor and adsorbent
addition)
Camptothecin Entrophospora infrequens Medium composition Single factor 503 ± 25 μg/100 g dry cell Amna et al. (2012)
RJMEF001 mass (in Sabouraud broth)
Vincamine XM-J2 Medium composition and pH L16 (43) orthogonal 1.4 fold (1.5 fold increase Yin and Sun (2011)
design in biomass)
Sipeimine Fritillaria ussuriensis Fu7 Medium composition and temperature L16 (44) orthogonal design 77% increase (155% Yin and Chen (2011)
increase in biomass)

Please cite this article as: Venugopalan, A., Srivastava, S., Endophytes as in vitro production platforms of high value plant secondary metabolites,
Biotechnol Adv (2015), http://dx.doi.org/10.1016/j.biotechadv.2015.07.004
A. Venugopalan, S. Srivastava / Biotechnology Advances xxx (2015) xxx–xxx 9

Table 5
Examples of elicitation strategy for improvement in product yield from endophytes.

Product Organism Elicitor Yield enhancement Reference

Taxol Nodulisporium sylviforme UV40–19Serine, SA, silver nitrate, ammonium acetate 1.15 fold (including culture parameters) Zhao et al. (2011d)
Taxol Periconia sp. Serinol, p-hydroxy benzoic acid, β-resorcyclic acid, 8 fold (0.01 mM benzoic acid) — all Li et al. (1998a)
gallic acid, Benzoic acid other elicitors had positive effect
Camptothecin Trichoderma atroviride LY357 Salicylic acid 3.4 fold Pu et al. (2013)
Methyl jasmonate 2.2 fold
Ca2+, Cu2+, Mn2+ Slight increase
Li+ Slight decrease
10-Hydroxy camptothecin Xylaria sp. Ce3+, Cr3+, La3+, MJ, SA, Cu2+, Fe2+, Se5+, Mn2+, 2.7 fold (0.1 mM SA) — except MJ all Liu et al. (2010)
Ca2+, Li+ elicitors had positive effect
Palmarumycin C13 Berkleasmium sp. Dzf12 Yeast extract 3.2 fold Zhao et al. (2011c)
Palmarumycin C13 Berkleasmium sp. Dzf12 Water extracted polysaccharide from host plant 2.69 fold Li et al. (2012b)
(Dioscorea zingiberensis) rhizomes
Palmarumycin C12 Berkleasmium sp. Dzf12 Crude oligosaccharide prepared by acid hydrolysis 9.83 fold Li et al. (2012c)
Palmarumycin C13 of the water extracted polysaccharide 3.24 fold
Huperzine Colletotrichum gloeosporioides ES026 Ethanol and methanol 51.89% (with ethanol) Zhao et al. (2013c)

discovery of novel metabolites (Grond et al., 2002; Wang et al., 2014a), for improved product yields in endophytic fungi have been listed in
thus highlighting the crucial role of culture parameter optimization in Table 5.
microbial fermentation for desired results. Optimization of medium In addition to elicitors, use of metabolic inhibitors in order to divert
and fermentation parameters can be done either by changing one factor the flux toward the product of interest has also been documented.
at a time or by varying several factors at the same time and looking for Specifically, in the case of a taxol producing endophyte, which also pro-
interactions using statistical analysis. Statistical design of experiments is duced large amounts of ergosterol, use of sterol biosynthesis inhibitors
an organized approach that gives more reliable information per to divert the geranyl-geranyl pyrophosphate pool toward taxol biosyn-
experiment as it enables the visualization of the interactions among thesis resulted in the enhancement of the taxol yield up to 50 fold
experimental variables, leading to predictions of the response data in (Li et al., 1998b). Examples of inhibitor addition for improved product
areas not directly covered by experimentation (Thiry and Cingolani, yields in endophytic fungi are listed in Table 6.
2002). Both statistical as well as single factor optimization studies
have been reported in the fermentation of endophytic fungi. Examples 5.2.3. Precursor feeding
of culture parameter optimization carried out in endophytic fungi for Precursor feeding is the strategy of exogenously supplying a biosyn-
increasing the product yield have been listed in Table 4. thetic precursor or other intermediates in the biosynthetic pathway to
the culture medium in order to increase the desired product yield
5.2.2. Elicitor/inhibitor addition (Mulabagal and Tsay, 2004). It is a widely used strategy in plant cell/
Elicitors are signaling molecules which trigger the formation of tissue cultivation and is now being explored in the fermentation of
secondary metabolites in cell cultures by inducing plant defense, hyper- endophytic fungi also. It is feasible in cases where the precursors or in-
sensitive response and/or pathogenesis related proteins (Baldi et al., termediates are inexpensive. While a high concentration of precursors
2009). They are classified as biotic and abiotic based on their origin, may usually increase reaction rates, the toxicity to cells may also
where the former includes polysaccharides, proteins, glycoproteins, increase with increase in concentration, and so the concentration of pre-
cell-wall fragments etc. derived from fungi, bacteria and even plants cursor to be added to an endophytic fungal culture has to be optimized
and the latter includes metal ions, UV light, chemically defined carefully (Gaosheng and Jingming, 2012). Precursor feeding studies
compounds and any other stresses which are of non-biological origin. have also played a major part in the elucidation of biosynthetic
The success of elicitation as an effective strategy for improving second- pathways of various metabolites including taxol and camptothecin
ary metabolite production in plant cell cultures has prompted many (Guerra-Bubb et al., 2012; Yamazaki et al., 2004). Stierle et al. (1993)
reviews (Namdeo, 2007; Radman et al., 2003) and studies on the mech- used isotope-labeled precursors to establish the existence of taxol
anisms of elicitor action (Vasconsuelo and Boland, 2007; Zhao et al., biosynthetic machinery in the fungal culture, T. andreanae. Feeding of
2005a). Though the strategy has been used predominantly in plant sys- the precursor leucine to the camptothecin producing Entrophospora
tems, application of elicitation to microbial systems is also known and is infrequens was used to establish that leucine rather than mevalonic
becoming increasingly popular, especially with endophytic fungal acid is the precursor for the terpenoid moiety of the camptothecin mol-
systems where host plant signaling plays a very important role in ecule (Amna et al., 2012). Examples related to the effect of precursor
secondary metabolism. Interestingly, in the case of endophytic fungi, feeding on the product yield in endophytic fungi have been listed in
even small molecules like ethanol have caused significant enhancement Table 7.
in the product yield (Venugopalan and Srivastava, 2015; Zhao et al.,
2013c), which supports the idea that elicitation can become a cost- 5.2.4. Use of adsorbent resins/solid supports
effective, yield and productivity enhancement strategy for sustainable In industrial processes, downstream processing is easier if the
metabolite production in endophytic fungal fermentations at large fermentation product is extracellular rather than being stored inside
scale. Further examples related to the application of elicitation strategy the cell. Further, in the case of self-toxic metabolites immediate removal

Table 6
Examples of inhibitor addition for improvement in product yield from endophytes.

Product Organism Inhibitor Effect Reference

Taxol Pestalotiopsis microspora Ribonuclease inhibitors (vanadyl sulfate, lauryl sulfate, bentonite, and polyvinyl sulfate) Positive (vanadyl sulfate) Li et al. (1998b)
Ne32 Sterol biosynthesis inhibitors (paclobutrazol, tebuconazole, triadimenol, lovastatin, triadimefon, 50 fold yield increase (triadimefon)
2-chloroethylphosphonic acid, arachidonic acid, 1-aminobenzotriazole, fluconazole)

Please cite this article as: Venugopalan, A., Srivastava, S., Endophytes as in vitro production platforms of high value plant secondary metabolites,
Biotechnol Adv (2015), http://dx.doi.org/10.1016/j.biotechadv.2015.07.004
10 A. Venugopalan, S. Srivastava / Biotechnology Advances xxx (2015) xxx–xxx

Table 7
Examples of precursor feeding for improvement in product yield from endophytes.

Product Organism Precursor Effect Reference

Taxol Pestalotiopsis microspora Ne32 Sodium benzoate Positive Li et al. (1998b)


Camptothecin Trichoderma atroviride LY357 Tryptamine Positive Pu et al. (2013)
Secologanin Negative
Camptothecin Entrophospora infrequens RJMEF001 Tryptophan Positive Amna et al. (2012)
Tryptamine Positive
Geraniol No camptothecin
Citral No camptothecin
Mevalonic acid + tryptophan Negative
Leucine + tryptophan Positive
Geraniol + tryptophan No camptothecin
Citral + tryptophan No camptothecin

of the product is necessary to sustain higher yields, as the growth of the 6. Strategies to unlock the cryptic/silenced gene clusters
organism is affected by the release of its toxic metabolites (Singh et al., in endophytes
2010). In situ product removal (ISPR) is an integrated bioprocess
involving simultaneous production and separation. It confers several While the application of bioprocess optimization strategies on
advantages on the fermentation process including improved product endophytic fungi in axenic culture has led to increased product yields
recovery, elimination of feedback inhibition, overcoming product deg- in many cases, most often genomic instability or silencing of the
radation, avoiding product auto-toxicity, reducing cost and improving biosynthetic genes in axenic culture hinders attempts of bioprocess
the product yield efficiently (Luo et al., 2014; Schügerl and Hubbuch, development using newly isolated strains. Hence, the challenges associ-
2005). Two ways of employing ISPR are extraction with a liquid solvent ated with stabilizing the productive capabilities of the axenic culture of
or adsorption with a solid sorbent added to the culture medium (Xu endophytes can be addressed via the use of strategies like co-cultivation
et al., 2009). The latter strategy, which has the advantage of lower risk and epigenetic modifier addition. A rational approach toward exploiting
of toxicity as inert polymeric resins are employed as opposed to reactive the axenic cultures of endophytes for secondary metabolite production
organic solvents, has been employed extensively in endophytic fungal has been depicted in Fig. 2.
fermentation (Table 8). Several parameters like the type, concentration,
time of addition and incubation period of resin need to be selected and 6.1. Co-cultivation and mixed fermentation
optimized for improving metabolite production in the suspension
culture of the endophytes. Recently, addition of adsorbent resins Currently, studies with respect to bioprospecting of endophytes and
has been explored as a potential strategy to trigger the differential exploitation of endophytes for known high value metabolites have been
expression of secondary metabolites in fungal strains, which could done using the classical approach of screening several different endo-
lead to the discovery of novel bioactive molecules (González- phytes from a host plant and identifying competent endophytes with
Menéndez et al., 2014). the desired trait i.e. the ability to produce a specific metabolite while
Another strategy of yield enhancement or even novel metabolite the remaining strains are discarded without further explorations
discovery is the use of inert solid supports like polypropylene, cellulose, (Kusari et al., 2012a). Further, the identified endophytes are maintained
absorbent cotton gauze, polyester–cellulose, or polyurethane in solid- as axenic monocultures for fermentation of the desired product. On one
state and submerged fermentation (Bigelis et al., 2006). Culturing hand, in axenic culture, the endophyte no longer has access to the spe-
fungi on static supports in liquid medium enable exploitation of various cialized microenvironment of the host plant which it is attuned to,
environmental stimuli that influence metabolism, morphogenesis, and which may lead to silencing of its secondary metabolite genes, and on
differentiation thus affecting their metabolite profile. The presence of the other hand standard culture conditions may not be sufficient to
the solid support may mimic natural fungal habitats thus triggering trigger expression of the cryptic biosynthetic gene clusters present in
changes in the growth and physiology of the culture (Bigelis et al., fungal endophytes leading to a redundancy in terms of novel metabolite
2006). The physicochemical characteristics of the support system and discovery. An important finding via the use of whole-genome sequenc-
its interaction with available water, oxygen, and medium components ing strategies is that the number of genes encoding the biosynthetic
and other factors like availability of the gaseous phase in interstitial enzymes in various fungi and bacteria is greater than the known sec-
spaces and aeration, nutrient and waste product absorbance and ondary metabolites of these microbes (Scherlach and Hertweck,
diffusion, induce environmental conditions which mimic those found 2009). Slight changes in culture parameters are known to significantly
in nature. This strategy has been successfully applied to the endophytic affect the metabolic processes in microbes, especially in the case of en-
strain of Phomopsis sp. using absorbent cotton gauze and cellulose dophytes whose survival is defined by a delicately balanced relationship
paper for yield enhancement of mycoepoxydiene by 1 and 1.2 fold, with the host plant featuring complex multipartite interactions with
respectively (Thammajaruk et al., 2011). other endophytes. Examples exist to indicate the sensitivity of the

Table 8
Use of adsorbent resins for improvement in product yield from endophytes.

Product Organism Resin type Yield enhancement Reference

Beauvericin Fusarium redolens Dzf2 Polystyrene resin (X-5) 1.8 fold Xu et al. (2009)
Palmarumycin C13 Berkleasmium sp. Dzf12 Polystyrene resin (AB-8) 1.4 fold Zhao et al. (2011a)
Botrallin Hyalodendriella sp. ponipodef12 Polystyrene resin DM-301 2.29 fold Luo et al. (2014)
TMC-264 Hyalodendriella sp. ponipodef12 Polystyrene resin DM-301 11.76 fold Luo et al. (2014)
Camptothecin Trichoderma atroviride LY357 HP20 11 fold Pu et al. (2013)
XAD16 4.4 fold

Please cite this article as: Venugopalan, A., Srivastava, S., Endophytes as in vitro production platforms of high value plant secondary metabolites,
Biotechnol Adv (2015), http://dx.doi.org/10.1016/j.biotechadv.2015.07.004
A. Venugopalan, S. Srivastava / Biotechnology Advances xxx (2015) xxx–xxx 11

Fig. 2. A rational approach toward the exploitation of axenic cultures of endophytes for secondary metabolite production.

metabolic machinery that endophytes exhibit to the slightest changes novel co-culture systems which mimic the host microenvironment by
in culture conditions. Paranagama et al. (2007) reported the production providing the endophyte with the same kind of in planta selection
of six new secondary metabolites by the endophyte, Paraphaeosphaeria pressure under which it thrives can prove to be beneficial with
quadriseptata when distilled water was used in the medium in respect to host-associated and novel secondary metabolite yields in
place of tap water. Similarly, when the medium was changed from endophytes. This requires a complete understanding of the complex
solid to liquid, Chaetomium chiversii produced radicicol instead of ecological interactions between endophytes and plants and also
chaetochromin A. among the various endophytes that co-exist within the same host.
Apart from screening of individual culture parameters, co- Co-cultivation/mixed fermentation strategy has been employed and re-
cultivation or mixed fermentation is another OSMAC strategy that can ported for novel product isolation and as a yield enhancement strategy
enable simulation of the natural habitat of endophytes. Designing (Table 9).

Table 9
Use of co-cultivation/mixed fermentation strategy for improvement in product yield and novel product discovery from endophytes.

Compound Co-culture/mixed fermentation system Outcome Reference

Taxol Paraconiothyrium strain Alternaria sp. 2.7 fold yield increase Soliman and Raizada
SSM001 Phomopsis sp. 3.8 fold yield increase (2013)
Alternaria sp. and Phomopsis sp. 7.8 fold yield increase
Marinamide and marinamide methylether Two unidentified mangrove-derived endophytic fungi Novel alkaloids with potent cytotoxicity Zhu and Lin (2006)
6-Methylsalicylic acid Two unidentified mangrove-derived endophytic fungi Induced only in co-culture Zhu et al. (2007)
Cyclo-(phe–phe) dipeptide
Subenniatins A and B Fusarium tricinctum Fusarium begonia Novel linear depsipeptides Wang et al. (2013)
Acremostatins A, B and C Acremonium sp. Mycogone rosea Novel lipoaminopeptide Degenkolb et al. (2002)
Lateropyrone F. tricinctum Bacillus subtilis 1.4 fold increase Ola et al. (2013)
Enniatin A1 78 fold increase
Enniatin B 9 fold increase
Enniatin B1 36.3 fold increase
Fusaristatin A 7.9 fold increase
Macrocarpon C Novel substituted pyranone
N-(carboxymethyl)-anthranilic acid Novel starting material for indigo synthesis
(−)-Citreoisocoumarin Novel metabolite
(−)-Citreoisocoumarinol Novel metabolite
Enniatins A1, B1, and B (cyclic depsipeptides) F. tricinctum Streptomyces lividans Induced only in co-culture Ola et al. (2013)
Fusaristatin A — lipopeptide Induced only in co-culture
Red resin (traditional Chinese medicine) Dracaena cochinchinensis Fusarium moniliforme 9568D Resin accumulation in inoculated sites Jiang et al. (2003)
Taxol Taxus chinensis var. maireiFusarium mairei 38 fold yield increase Li et al. (2009)
Taxol Taxus cuspidata F. mairei 4.3 fold yield increase Li and Tao (2009)
Podophyllotoxin Linum album Piriformospora indicaa 3.4 fold yield increase Baldi et al. (2010)
Sebacina vermiferaa 3.8 fold yield increase
6-Methoxy podophyllotoxin L. album P. indicaa 4.9 fold yield increase Baldi et al. (2010)
S. vermiferaa 8.7 fold yield increase
Stemphyperylenol (anti-fungal polyketide) Alternaria tenuissima Nigrospora sphaerica Induced only in co-culture Chagas et al. (2013)
13-Oxo-9,11-octadecadienoic acid (oxylipin) Endophyte Paraconiothyrium Phytopathogen Fusarium Decreased the mycotoxin production by Combès et al. (2012)
variabile oxysporum F. oxysporum
a
Arbuscular mycorrhizae-like fungi.

Please cite this article as: Venugopalan, A., Srivastava, S., Endophytes as in vitro production platforms of high value plant secondary metabolites,
Biotechnol Adv (2015), http://dx.doi.org/10.1016/j.biotechadv.2015.07.004
12 A. Venugopalan, S. Srivastava / Biotechnology Advances xxx (2015) xxx–xxx

Table 10
Use of epigenetic modifiers for novel product discovery from endophytes.

Epigenetic modifier used Organism Effect Reference

Suberoylanilide hydroxamic acid (SBHA) Endophytic Fusarium Production of novel fusaric acid derivative: Chen et al. (2013)
(HDAC inhibitor) oxysporum strain R1 • 5-Butyl-6-oxo-1,6-dihydropyridine-2-carboxylic acid
• 5-(But-9-enyl)-6-oxo-1,6-dihydropyridine-2-carboxylic acid
Sodium butyrate (HDAC inhibitor) Leucostoma persoonii • Novel compound — cytosporone R Beau et al. (2012)
• Enhanced production of known cytosporones B (360%), C (580%) and E (890%)
5-Azacytidine (DNMT inhibitor) and/or Endophytic Alternaria sp. Production of mycotoxins including alternariol, alternariol-5-O-methyl ether, Sun et al. (2012)
suberoylanilide hydroxamic acid 3′-hydroxyalternariol-5-O-methyl ether, altenusin, tenuazonic acid, and altertoxin II.
(HDAC inhibitor)
Suberoylanilide hydroxamic acid (HDAC Pestalotiopsis acacia Production of 3 novel aromatic Yang et al. (2013)
inhibitor) + 5-azacytidine Compounds:
(DNMT inhibitor) • 20-Hydroxy-60-hydroxymethyl-40-methylphenyl-2,6-dihydroxy-3-(2-isopentenyl)benzoate
• 4,6-Dihydroxy-7-hydroxymethyl-3-methylcoumarin
• 4,6-dihydroxy-3,7-dimethylcoumarin and five known polyketides:
Endocrocin
Pestalotiollide B
Pestalotiopyrone G
Scirpyrone A
7-Hydroxy-2-(2-hydroxypropyl)-5-methylchromone
5-Azacytidine (DNMT inhibitor) Pestalotiopsis crassiuscula Production of 7 metabolites: Yang et al. (2014b)
• 4,6-Dihydroxy-7-hydroxymethyl-3-methoxymethylcoumarin (novel metabolite)
• 4,6-Dihydroxy-7-hydroxymethyl-3-methylcoumarin
• 4,6-Dihydroxy-3,7-dimethylcoumarin
• Pestalotiopyrone G
• Scirpyrone A
• Scirpyrone B
• 2′-Hydroxy-6′-hydroxymethyl-4′-methylphenyl 2,6-dihydroxy-3-(2-isopentenyl) benzoate
Nicotinamide (an NAD+-dependent Graphiopsis chlorocephala Production of 6 novel benzophenones: cephalanones A–F and a known metabolite: Asai et al. (2013)
HDAC inhibitor) 2-(2,6-dihydroxy-4-methylbenzoyl)-6-hydroxybenzoic acid

Designing a successful co-cultivation system involves several regulation of secondary metabolite synthesis has resulted in several
challenges. An optimum inoculum ratio of the various species being novel approaches for the isolation of new secondary metabolites from
co-cultured should be established such that the competition between filamentous fungi. The proposed strategies include over-expression of
the species is balanced and the growth of one does not overpower a cluster-specific transcription factor (TF) gene, exchange of promoters
the other. Culture conditions and medium composition have to be opti- for biosynthesis genes or TF genes with inducible promoters such as the
mized to accommodate the different growth morphologies being used alcohol dehydrogenase A gene promoter, and over-expression or
in the co-culture system, while also maximizing productivity of the deletion of a global regulator gene (Brakhage, 2013; Scharf and
desired product by one of the organisms. Reactor design is also an im- Brakhage, 2013). These methods can be applied to endophytic fungi as
portant factor to be considered (Weathers et al., 2010). When these well to modulate their secondary metabolite machinery.
challenges are addressed, co-cultivation/mixed fermentation becomes
a powerful strategy as the competition or antagonism experienced
7. Conclusions
during co-cultivation may lead to a significantly enhanced production
of constitutively present compounds and/or to an accumulation of
The vast amount of knowledge that is already available and the
cryptic compounds that are not detected in axenic culture of the select-
newer pieces of the puzzle that are emerging in endophyte research
ed strain (Marmann et al., 2014).
only make it obvious that endophytic fungi will be one of the major
sources of secondary metabolites in the near future. In addition, consid-
6.2. Use of epigenetic modifiers
ering the various yield enhancement strategies available that can be
tailored to endophytic fungi, the future potential of endophytic fungi
Fungal biosynthetic gene clusters, which are often located in the
research for the production of high value natural products looks
distal regions of chromosomes, exist in a heterochromatin state. These
extremely promising. However, a much greater understanding of the
genes are often transcriptionally controlled by epigenetic regulation
host plant-endophyte dynamics at the molecular level is essential to
such as histone deacetylation and DNA methylation (Pettit, 2010).
exploit the promising potential of these extraordinary organisms at
Histone deacetylation and DNA methylation operate to modify the chro-
commercial level. This will also enable rational optimization of the
matin and thus regulate gene expression or silencing in fungi and other
bioprocess for high and sustainable production of high value secondary
organisms. Hence, the possibility of using epigenetic modifiers like DNA
metabolites from endophytes.
methyltransferase (DNMT) and histone deacetylase (HDAC) inhibitors
to induce chromatin remodeling has been proposed as a strategy for
unlocking cryptic biosynthetic gene clusters (Brakhage and Schroeckh, References
2011). Commonly used DNMT inhibitors include 5-azacytidine and Adrio, J.L., Demain, A.L., 2006. Genetic improvement of processes yielding microbial prod-
5-aza-20-deoxycytidine, while HDAC inhibitors include hydroxamic- ucts. FEMS Microbiol. Rev. 30, 187–214.
acid containing compounds or cyclic peptides such as trichostatin A Ajikumar, P.K., Xiao, W.H., Tyo, K.E.J., Wang, Y., Simeon, F., Leonard, E., et al., 2010. Iso-
prenoid pathway optimization for taxol precursor overproduction in Escherichia
and trapoxin B (Cichewicz, 2010). Table 10 lists the examples available
coli. Science 330, 70–74.
on the usage of epigenetic modifiers for novel product discovery in Alam, A., Naik, P.K., Mishra, G.P., 2009. Congruence of RAPD and ISSR markers for evalua-
endophytes. tion of genomic relationship among 28 populations of Podophyllum hexandrum Royle
There are many literature reviews available on the regulatory from Himachal Pradesh, India. Turk. J. Bot. 33, 1–12.
Amna, T., Amina, M., Sharma, P.R., Puri, S.C., Al-Youssef, H.M., Al-Taweel, A.M., et al., 2012.
elements that modulate the transcription of secondary metabolite Effect of precursors feeding and media manipulation on production of novel antican-
genes (Brakhage, 2013; Keller et al., 2005). An understanding of the cer pro-drug camptothecin from endophytic fungus. Braz. J. Microbiol. 43, 1476–1489.

Please cite this article as: Venugopalan, A., Srivastava, S., Endophytes as in vitro production platforms of high value plant secondary metabolites,
Biotechnol Adv (2015), http://dx.doi.org/10.1016/j.biotechadv.2015.07.004
A. Venugopalan, S. Srivastava / Biotechnology Advances xxx (2015) xxx–xxx 13

Arnold, A.E., Lewis, L.C., 2005. Ecology and evolution of fungal endophytes, and their roles Grond, S., Papastavrou, I., Zeeck, A., 2002. Novel α-L-rhamnopyranosides from a single
against insects. In: Vega, F., Blackwell, M. (Eds.), Insect–Fungal Associations: Ecology strain of Streptomyces by supplement-induced biosynthetic steps. Eur. J. Org. Chem.
and Evolution. Oxford University Press, New York, pp. 74–96. 2002, 3237–3242.
Arnold, A.E., Mejía, L.C., Kyllo, D., Rojas, E.I., Maynard, Z., Robbins, N., et al., 2003. Fungal Guerra-Bubb, J., Croteau, R., Williams, R.M., 2012. The early stages of taxol biosynthesis:
endophytes limit pathogen damage in a tropical tree. Proc. Natl. Acad. Sci. 100, an interim report on the synthesis and identification of early pathway metabolites.
15649–15654. Nat. Prod. Rep. 29, 683–696.
Asai, T., Otsuki, S., Sakurai, H., Yamashita, K., Ozeki, T., Oshima, Y., 2013. Benzophenones Hedden, P., Phillips, A.L., Rojas, M.C., Carrera, E., Tudzynski, B., 2001. Gibberellin biosyn-
from an endophytic fungus, Graphiopsis chlorocephala, from Paeonia lactiflora cultivat- thesis in plants and fungi: a case of convergent evolution? J. Plant Growth Regul.
ed in the presence of an NAD+-dependent HDAC inhibitor. Org. Lett. 15, 2058–2061. 20, 319–331.
Baldi, A., Srivastava, A.K., Bisaria, V.S., 2009. Fungal elicitors for enhanced production of Heinig, U., Scholz, S., Jennewein, S., 2013. Getting to the bottom of Taxol biosynthesis by
secondary metabolites in plant cell suspension cultures. In: Varma, A., Kharkwal, fungi. Fungal Divers. 60, 161–170.
A.C. (Eds.), Symbiotic Fungi. Springer, Berlin Heidelberg, pp. 373–380. Hendrawati, O., Woerdenbag, H.J., Hille, J., Kayser, O., 2012. Metabolic engineering of me-
Baldi, A., Farkya, S., Jain, A., Gupta, N., Mehra, R., Datta, V., et al., 2010. Enhanced produc- dicinal plants and microorganisms for the production of natural products. In: Kayser,
tion of podophyllotoxins by co-culture of transformed Linum album cells with plant O., Warzecha, H. (Eds.), Pharmaceutical Biotechnology: Drug Discovery and Clinical
growth-promoting fungi. Pure Appl. Chem. 82, 227–241. Applications, Second Edition Wiley‐VCH Verlag GmbH & Co. KGaA, pp. 491–526.
Barrios-Gonzalez, J., Fernandez, F.J., Tomasini, A., 2003. Microbial secondary metabolites Hong, Y.I.N., Yang, G.A.O., 2010. Optimization of fermentation conditions of a
production and strain improvement. Indian J. Biotechnol. 2, 322–333. mutagenized berberine-producing strain. J. Fungal Res. 1, 15.
Beau, J., Mahid, N., Burda, W.N., Harrington, L., Shaw, L.N., Mutka, T., et al., 2012. Epigenet- Howitz, K.T., Sinclair, D.A., 2008. Xenohormesis: sensing the chemical cues of other
ic tailoring for the production of anti-infective cytosporones from the marine fungus species. Cell 387–391.
Leucostoma persoonii. Mar. Drugs 10, 762–774. Hyde, K.D., Soytong, K., 2008. The fungal endophyte dilemma. Fungal Divers. 33, 163–173.
Bigelis, R., He, H., Yang, H.Y., Chang, L.P., Greenstein, M., 2006. Production of fungal Jiang, D.F., Ma, P., Yang, J., Wang, X., Xu, K., Huang, Y., et al., 2003. Formation of blood resin
antibiotics using polymeric solid supports in solid-state and liquid fermentation. in abiotic Dracaena cochinchinensis inoculated with Fusarium 9568D. Ying Yong Sheng
J. Ind. Microbiol. Biotechnol. 33, 815–826. Tai Xue Bao 14, 477–478.
Bode, H.B., Bethe, B., Hӧfs, R., Zeeck, A., 2002. Big effects from small changes: possible Kai, Z., Xuan, W., Yushi, S., Ying, W., Wenxiang, P., Dongpo, Z., 2008. Screening of high
ways to explore nature's chemical diversity. Chembiochem 3, 619–627. taxol producing fungi by NTG combining with UV mutagenesis. J. Nat. Sci. Heilong-
Bӧlker, M., Bӧhnert, H.U., Braun, K.H., Gӧrl, J., Kahmann, R., 1995. Tagging pathogenicity jiang Univ. 1, 16.
genes in Ustilago maydis by restriction enzyme-mediated integration (REMI). Mol. Kai, Z., Qingshen, S., Yanjun, Z., Wenxiang, P., Tao, J., Dongpo, Z., 2009. Screening and
Gen. Genet. 248, 547–552. characterization of a high taxol producing fungus by protoplast mutagenesis. High
Brakhage, A.A., 2013. Regulation of fungal secondary metabolism. Nat. Rev. Microbiol. 11, Technol. Lett. 15, 220–226.
21–32. Karuppusamy, S., 2009. A review on trends in production of secondary metabolites from
Brakhage, A.A., Schroeckh, V., 2011. Fungal secondary metabolites—strategies to activate higher plants by in vitro tissue, organ and cell cultures. J. Med. Plants Res. 3,
silent gene clusters. Fungal Genet. Biol. 48, 15–22. 1222–1239.
Brundrett, M., 2004. Diversity and classification of mycorrhizal associations. Biol. Rev. 79, Kaul, S., Ahmed, M., Zargar, K., Sharma, P., Dhar, M.K., 2012. Prospecting endophytic
473–495. fungal assemblage of Digitalis lanata Ehrh. (foxglove) as a novel source of digoxin:
Carroll, G.C., 1991. Beyond pest deterrence — alternative strategies and hidden costs of a cardiac glycoside. Biotech 3, 335–340.
endophytic mutualisms in vascular plants. In: Andrews, J.A., Hirano, S.S. (Eds.), Micro- Keller, N.P., Turner, G., Bennett, J.W., 2005. Fungal secondary metabolism—from biochem-
bial Ecology of Leaves. Springer-Verlag, New York, pp. 358–375. istry to genomics. Nat. Rev. Microbiol. 3, 937–947.
Cassady, J.M., Chan, K.K., Floss, H.G., Leistner, E., 2004. Recent developments in the Kobayashi, D.Y., Palumbo, J.D., 2000. Bacterial endophytes and their effects on plants and
maytansinoid antitumor agents. Chem. Pharm. Bull. 52, 1–26. uses in agriculture. In: Bacon, C.W., White, J.F. (Eds.), Microbial Endophytes. Marcel
Chagas, F.O., Dias, L.G., Pupo, M.T., 2013. A mixed culture of endophytic fungi increases Dekker, New York, pp. 199–236.
production of antifungal polyketides. J. Chem. Ecol. 39, 1335–1342. Krings, M., Taylor, T.N., Hass, H., Kerp, H., Dotzler, N., Hermsen, E.J., 2007. Fungal endo-
Chaichanan, J., Wiyakrutta, S., Pongtharangkul, T., Isarangkul, D., Meevootisom, V., 2014. phytes in a 400-million-yr-old land plant: infection pathways, spatial distribution,
Optimization of zofimarin production by an endophytic fungus, Xylaria sp. Acra and host responses. New Phytol. 174, 648–657.
L38. Braz. J. Microbiol. 45, 287–293. Kumara, P.M., Zuehlke, S., Priti, V., Ramesha, B.T., Shweta, S., Ravikanth, G., et al., 2012.
Chattopadhyay, S., Farkya, S., Srivastava, A.K., Bisaria, V.S., 2002. Bioprocess considerations Fusarium proliferatum, an endophytic fungus from Dysoxylum binectariferum Hook.
for production of secondary metabolites by plant cell suspension cultures. Biotechnol. f, produces rohitukine, a chromane alkaloid possessing anti-cancer activity. Antonie
Bioprocess Eng. 7, 138–149. Van Leeuwenhoek 101, 323–329.
Chaturvedi, P., Gajbhiye, S., Roy, S., Dudhale, R., Chowdhary, A., 2014. Determination of Kurland, C.G., Canback, B., Berg, O.G., 2003. Horizontal gene transfer: a critical view. Proc.
kaempferol in extracts of Fusarium chlamydosporum, an endophytic fungi of Natl. Acad. Sci. 100, 9658–9662.
Tylophora indica (Asclepeadaceae) and its anti-microbial activity. IOSR J. Pharm. Kusari, S., Košuth, J., Čellárová, E., Spiteller, M., 2011a. Survival-strategies of endophytic
Biol. Sci. 9, 51–55. Fusarium solani against indigenous camptothecin biosynthesis. Fungal Ecol. 4,
Chen, H.J., Awakawa, T., Sun, J.Y., Wakimoto, T., Abe, I., 2013. Epigenetic modifier-induced 219–223.
biosynthesis of novel fusaric acid derivatives in endophytic fungi from Datura stramo- Kusari, S., Zühlke, S., Spiteller, M., 2011b. Effect of artificial reconstitution of the interac-
nium L. Nat. Prod. Bioprospect. 3, 20–23. tion between the plant Camptotheca acuminata and the fungal endophyte Fusarium
Cichewicz, R.H., 2010. Epigenome manipulation as a pathway to new natural product solani on camptothecin biosynthesis. J. Nat. Prod. 74, 764–775.
scaffolds and their congeners. Nat. Prod. Rep. 27, 11–22. Kusari, S., Hertweck, C., Spiteller, M., 2012a. Chemical ecology of endophytic fungi: origins
Combès, A., Ndoye, I., Bance, C., Bruzaud, J., Djediat, C., Dupont, J., et al., 2012. Chemical of secondary metabolites. Chem. Biol. 19, 792–798.
communication between the endophytic fungus Paraconiothyrium variabile and the Kusari, S., Verma, V.C., Lamshoeft, M., Spiteller, M., 2012b. An endophytic fungus from
phytopathogen Fusarium oxysporum. PLoS ONE 7, e47313. Azadirachta indica A. Juss. that produces azadirachtin. World J. Microbiol. Biotechnol.
Cui, Y., Yi, D., Bai, X., Sun, B., Zhao, Y., Zhang, Y., 2012. Ginkgolide B produced endophytic 28, 1287–1294.
fungus (Fusarium oxysporum) isolated from Ginkgo biloba. Fitoterapia 83, 913–920. Kusari, S., Singh, S., Jayabaskaran, C., 2014. Rethinking production of Taxol (paclitaxel)
Dai, W.L., Tao, W.Y., 2008. Preliminary study on fermentation conditions of taxol- using endophyte biotechnology. Trends Biotechnol. 32, 304–311.
producing endophytic fungus. Chem. Ind. Eng. Prog. 27, 883–886. Lee, J.C., Lobkovsky, E., Pliam, N.B., Strobel, G., Clardy, J., 1995. Subglutinols A and B: im-
Dangi, R., Misar, A., Tamhankar, S., Rao, S., 2014. Diosgenin content in some Trigonella munosuppressive compounds from the endophytic fungus Fusarium subglutinans.
species. Indian J. Adv. Plant Res. 1, 47–51. J. Org. Chem. 60, 7076–7077.
Degenkolb, T., Heinze, S., Schlegel, B., Strobel, G., Gräfe, U., 2002. Formation of new Li, Y.C., Tao, W.Y., 2009. Interactions of Taxol-producing endophytic fungus with its host
lipoaminopeptides, acremostatins A, B, and C, by co-cultivation of Acremonium sp. Taxus spp. during Taxol accumulation. Cell Biol. Int. 33, 106–112.
Tbp-5 and Mycogone rosea DSM 12973. Biosci. Biotechnol. Biochem. 66, 883–886. Li, J.Y., Sidhu, R.S., Ford, E., Hess, W.M., Strobel, G.A., 1998a. The induction of taxol
Dejong, J.M., Liu, Y., Bollon, A.P., Long, R.M., Jennewein, S., Williams, D., et al., 2006. production in the endophytic fungus — Periconia sp. from Torreya grandifolia. J. Ind.
Genetic engineering of taxol biosynthetic genes in Saccharomyces cerevisiae. Microbiol. 20, 259–264.
Biotechnol. Bioeng. 93, 212–224. Li, J.Y., Sidhu, R.S., Bollon, A., Strobel, G.A., 1998b. Stimulation of taxol production in liquid
Dubey, D., Rath, S., Sahu, M.C., Debata, N.K., Padhy, R.N., 2012. Antimicrobials of plant or- cultures of Pestalotiopsis microspora. Mycol. Res. 102, 461–464.
igin against TB and other infections and economics of plant drugs-introspection. Li, Y.C., Tao, W.Y., Cheng, L., 2009. Paclitaxel production using co-culture of Taxus
Indian J. Tradit. Knowl. 11, 225–233. suspension cells and paclitaxel-producing endophytic fungi in a co-bioreactor. Appl.
Flores-Bustamante, Z.R., Rivera-Orduña, F.N., Martínez-Cárdenas, A., Flores-Cotera, L.B., Microbiol. Biotechnol. 83, 233–239.
2010. Microbial paclitaxel: advances and perspectives. J. Antibiot. (Tokyo) 63, Li, P., Xu, L., Mou, Y., Shan, T., Mao, Z., Lu, S., et al., 2012a. Medium optimization for
460–467. exopolysaccharide production in liquid culture of endophytic fungus Berkleasmium
Gao, Y., Yin, H., Sun, Y., Zhang, Z., Cui, Y., 2008. Mutagenesis of a berberine-producing en- sp. Dzf12. Int. J. Mol. Sci. 13, 11411–11426.
dophytic fungus. J. Fungal Res. 4, 6. Li, Y., Li, P., Mou, Y., Zhao, J., Shan, T., Ding, C., et al., 2012b. Enhancement of diepoxin ζ
Gaosheng, H., Jingming, J., 2012. Production of useful secondary metabolites through reg- production in liquid culture of endophytic fungus Berkleasmium sp. Dzf12 by polysac-
ulation of biosynthetic pathway in cell and tissue suspension culture of medicinal charides from its host plant Dioscorea zingiberensis. World J. Microbiol. Biotechnol. 28,
plants. In: Leva, A., Rinalsi, L.M.R. (Eds.), Recent Advances in Plant in Vitro Culture. 1407–1413.
InTech, pp. 197–210. Li, Y., Shan, T., Mou, Y., Li, P., Zhao, J., Zhao, W., et al., 2012c. Enhancement of
González-Menéndez, V., Asensio, F., Moreno, C., de Pedro, N., Monteiro, M.C., de la Cruz, palmarumycin C12 and C13 production in liquid culture of the endophytic fungus
M., et al., 2014. Assessing the effects of adsorptive polymeric resin additions on fungal Berkleasmium sp. Dzf12 by oligosaccharides from its host plant Dioscorea
secondary metabolite chemical diversity. Mycology 5, 179–191. zingiberensis. Molecules 17, 3761–3773.

Please cite this article as: Venugopalan, A., Srivastava, S., Endophytes as in vitro production platforms of high value plant secondary metabolites,
Biotechnol Adv (2015), http://dx.doi.org/10.1016/j.biotechadv.2015.07.004
14 A. Venugopalan, S. Srivastava / Biotechnology Advances xxx (2015) xxx–xxx

Linden, J.C., 2006. Secondary products from plant tissue culture. Encycl. Life Support Syst. Rodriguez, R.J., White Jr., J.F., Arnold, A.E., Redman, R.S., 2009. Fungal endophytes:
(UNESCO-EOLSS) Biotechnol. 4, 1–9. diversity and functional roles. New Phytol. 182, 314–330.
Liu, K., Ding, X., Deng, B., Chen, W., 2010. 10-Hydroxycamptothecin produced by a new Sachin, N., Manjunatha, B.L., Mohana Kumara, P., Ravikanth, G., Shweta, S.,
endophytic Xylaria sp., M20, from Camptotheca acuminata. Biotechnol. Lett. 32, Suryanarayanan, T.S., et al., 2013. Do endophytic fungi possess pathway genes for
689–693. plant secondary metabolites? Curr. Sci. 104, 178.
Liu, L., Wei, Y.M., Zhou, X.W., Lin, J., Sun, X.F., Tang, K.X., 2013. Agrobacterium tumefaciens- Scharf, D.H., Brakhage, A.A., 2013. Engineering fungal secondary metabolism: a roadmap
mediated genetic transformation of the Taxol-producing endophytic fungus Ozonium to novel compounds. J. Biotechnol. 163, 179–183.
sp EFY21. Genet. Mol. Res. 12, 2913. Scherlach, K., Hertweck, C., 2009. Triggering cryptic natural product biosynthesis in
Long, D.M., Smidmansky, E.D., Archer, A.J., Strobel, G.A., 1998. In vivo addition of telomeric microorganisms. Org. Biomol. Chem. 7, 1753–1760.
repeats to foreign DNA generates chromosomal DNAs in the taxol-producing fungus Schügerl, K., Hubbuch, J., 2005. Integrated bioprocesses. Curr. Opin. Microbiol. 8, 294–300.
Pestalotiopsis microspora. Fungal Genet. Biol. 24, 335–344. Schulz, B., Römmert, A.K., Dammann, U., Aust, H. Jür, Strack, D., 1999. The endophyte–host
Luo, H., Liu, H., Cao, Y., Xu, D., Mao, Z., Mou, Y., et al., 2014. Enhanced production of interaction: a balanced antagonism? Mycol. Res. 103, 1275–1283.
botrallin and TMC-264 with in situ macroporous resin adsorption in mycelial liquid Selosse, M.A., Le Tacon, F., 1998. The land flora: a phototroph–fungus partnership? Trends
culture of the endophytic fungus Hyalodendriella sp. Ponipodef12. Molecules 19, Ecol. Evol. 13, 15–25.
14221–14234. Singh, M.P., Leighton, M.M., Barbieri, L.R., Roll, D.M., Urbance, S.E., Hoshan, L., et al., 2010.
Ma, C., Jiang, D., Wei, X., 2011. Mutation breeding of Emericella foeniculicola TR21 for im- Fermentative production of self-toxic fungal secondary metabolites. J. Ind. Microbiol.
proved production of tanshinone IIA. Process Biochem. 46, 2059–2063. Biotechnol. 37, 335–340.
Malik, S., Cusidó, R.M., Mirjalili, M.H., Moyano, E., Palazón, J., Bonfill, M., 2011. Production Sirikantaramas, S., Yamazaki, M., Saito, K., 2009. A survival strategy: the coevolution of the
of the anticancer drug taxol in Taxus baccata suspension cultures: a review. Process camptothecin biosynthetic pathway and self-resistance mechanism. Phytochemistry
Biochem. 46, 23–34. 70, 1894–1898.
Mangathayaru, K., 2013. Pharmacognosy: An Indian perspective. Pearson Education, Sohani, M.M., Schenk, P.M., Schultz, C.J., Schmidt, O., 2009. Phylogenetic and transcrip-
India. tional analysis of a strictosidine synthase‐like gene family in Arabidopsis thaliana
Manivasakam, P., Aubrecht, J., Sidhom, S., Schiestl, R.H., 2001. Restriction enzymes in- reveals involvement in plant defence responses. Plant Biol. 11, 105–117.
crease efficiencies of illegitimate DNA integration but decrease homologous integra- Soliman, S.S.M., Raizada, M.N., 2013. Interactions between co-habitating fungi elicit
tion in mammalian cells. Nucleic Acids Res. 29, 4826–4833. synthesis of Taxol from an endophytic fungus in host Taxus plants. Front. Microbiol.
Marler, M., Pedersen, D., Mitchell-Olds, T., Callaway, R.M., 1999. A polymerase chain 4, 1–14.
reaction method for detecting dwarf mistletoe infection in Douglas-fir and western Soliman, S.S.M., Trobacher, C.P., Tsao, R., Greenwood, J.S., Raizada, M.N., 2013. A fungal
larch. Can. J. For. Res. 29, 1317–1321. endophyte induces transcription of genes encoding a redundant fungicide pathway
Marmann, A., Aly, A.H., Lin, W., Wang, B., Proksch, P., 2014. Co-cultivation—a powerful in its host plant. BMC Plant Biol. 13, 93.
emerging tool for enhancing the chemical diversity of microorganisms. Mar. Drugs Spiering, M.J., Moon, C.D., Wilkinson, H.H., Schardl, C.L., 2005. Gene clusters for insecticidal
12, 1043–1065. loline alkaloids in the grass-endophytic fungus Neotyphodium uncinatum. Genetics
Medema, M.H., Trefzer, A., Kovalchuk, A., van den Berg, M., Müller, U., Heijne, W., et al., 169, 1403–1414.
2010. The sequence of a 1.8-Mb bacterial linear plasmid reveals a rich evolutionary Srivastava, S., Srivastava, A.K., 2007. Hairy root culture for mass-production of high value
reservoir of secondary metabolic pathways. Genome Biol. Evol. 2, 212–224. secondary metabolites. Crit. Rev. Biotechnol. 27, 29–43.
Ming, Q., Han, T., Li, W., Zhang, Q., Zhang, H., Zheng, C., et al., 2012. Tanshinone IIA and Staniek, A., Woerdenbag, H.J., Kayser, O., 2009. Taxomyces andreanae: a presumed
tanshinone I production by Trichoderma atroviride D16, an endophytic fungus in paclitaxel producer demystified? Planta Med. 75, 1561–1566.
Salvia miltiorrhiza. Phytomedicine 19, 330–333. Stierle, A., Strobel, G., Stierle, D., 1993. Taxol and taxane production by Taxomyces
Mochizuki, S., Hiratsu, K., Suwa, M., Ishii, T., Sugino, F., Yamada, K., et al., 2003. The large andreanae, an endophytic fungus of Pacific yew. Science 260, 214–216.
linear plasmid pSLA2‐L of Streptomyces rochei has an unusually condensed gene Stone, J.K., Bacon, C.W., White, J.F., 2000. An overview of endophytic microbes:
organization for secondary metabolism. Mol. Microbiol. 48, 1501–1510. endophytism defined. In: Bacon, C.W., White, J.F. (Eds.), Microbial Endophytes.
Mou, Y., Luo, H., Mao, Z., Shan, T., Sun, W., Zhou, K., et al., 2013. Enhancement of Marcel Dekker, New York, pp. 3–30.
palmarumycins C12 and C13 production in liquid culture of endophytic fungus Strobel, G., Daisy, B., 2003. Bioprospecting for microbial endophytes and their natural
Berkleasmium sp. Dzf12 after treatments with metal ions. Int. J. Mol. Sci. 14, products. Microbiol. Mol. Biol. Rev. 67, 491–502.
979–998. Strobel, G.A., Ford, E., Li, J.Y., Sears, J., Sidhu, R.S., Hess, W.M., 1999. Seimatoantlerium
Mu, J.H., Bollon, A.P., Sidhu, R.S., 1999. Analysis of β-tubulin cDNAs from taxol-resistant tepuiense gen. nov. A unique epiphytic fungus producing taxol from the Venezuelan
Pestalotiopsis microspora and taxol-sensitive Pythium ultimum and comparison of Guayana. System. Appl. Microbiol. 22, 426–433.
the taxol-binding properties of their products. Mol. Gen. Genet. 262, 857–868. Strobel, G.A., Hess, W.M., Baird, G., Ford, E., Li, J.Y., Sidhu, R.S., 2001. Stegolerium kukenani
Mulabagal, V., Tsay, H.S., 2004. Plant cell cultures-an alternative and efficient source for gen. nov. An endopyhtic, taxol producing fungus from the Roraima and Kukenan
the production of biologically important secondary metabolites. Int. J. Appl. Sci. Eng. Tepuis of Venezuela. Mycotaxon 78, 353–361.
2, 29–48. Sun, Y., Luo, H., Li, Y., Sun, C., Song, J., Niu, Y., et al., 2011. Pyrosequencing of the
Musavi, S.F., Dhavale, A., Balakrishnan, R.M., 2014. Optimization and kinetic modelling of Camptotheca acuminata transcriptome reveals putative genes involved in
cell associated camptothecin production from an endophytic Fusarium oxysporum camptothecin biosynthesis and transport. BMC Genomics 12, 533.
NFX06. Prep. Biochem. Biotechnol. 45, 158–172. Sun, J., Awakawa, T., Noguchi, H., Abe, I., 2012. Induced production of mycotoxins in an
Nair, D.N., Padmavathy, S., 2014. Impact of endophytic microorganisms on plants, endophytic fungus from the medicinal plant Datura stramonium L. Bioorg. Med.
environment and humans. Sci. World J. 2014, 1–11. Chem. Lett. 22, 6397–6400.
Namdeo, A.G., 2007. Plant cell elicitation for production of secondary metabolites: a Taylor, L., 2000. Plant Based Drugs and Medicines. Raintree Nutr Inc., Carson City NV.
review. Pharmacogn. Rev. 1, 69–79. Taylor, L., 2005. The Healing Power of Rainforest Herbs: A Guide to Understanding and
Newman, D.J., Cragg, G.M., 2007. Natural products as sources of new drugs over the last Using Herbal Medicinals. Square One Publishers, New York.
25 years. J. Nat. Prod. 70, 461–477. Thammajaruk, N., Sriubolmas, N., Israngkul, D., Meevootisom, V., Wiyakrutta, S., 2011.
Nirupama, R., Chaithra, K., Govindappa, M., Chandrappa, C.P., Vinay, R.B., 2011. Antimicro- Optimization of culture conditions for mycoepoxydiene production by Phomopsis
bial and antioxidant activities of endophytes from Tabebuia argentea and identifica- sp. Hant25. J. Ind. Microbiol. Biotechnol. 38, 679–685.
tion of anticancer agent (lapachol). J. Med. Plants Res. 5, 3643–3652. Thiry, M., Cingolani, D., 2002. Optimizing scale-up fermentation processes. Trends
Ola, A.R.B., Thomy, D., Lai, D., Brӧtz-Oesterhelt, H., Proksch, P., 2013. Inducing secondary Biotechnol. 20, 103–105.
metabolite production by the endophytic fungus Fusarium tricinctum through cocul- Tooker, J.F., Hanks, L.M., 2004. Trophic position of the endophytic beetle, Mordellistena
ture with Bacillus subtilis. J. Nat. Prod. 76, 2094–2099. aethiops Smith (Coleoptera: Mordellidae). Environ. Entomol. 33, 291–296.
Panaccione, D.G., Beaulieu, W.T., Cook, D., 2014. Bioactive alkaloids in vertically transmit- Trémouillaux-Guiller, J., Rohr, T., Rohr, R., Huss, V.A.R., 2002. Discovery of an endophytic
ted fungal endophytes. Funct. Ecol. 28, 299–314. alga in Ginkgo biloba. Am. J. Bot. 89, 727–733.
Paranagama, P.A., Wijeratne, E.M.K., Gunatilaka, A.A.L., 2007. Uncovering biosynthetic Tudzynski, B., 2005. Gibberellin biosynthesis in fungi: genes, enzymes, evolution, and
potential of plant-associated fungi: effect of culture conditions on metabolite impact on biotechnology. Appl. Microbiol. Biotechnol. 66, 597–611.
production by Paraphaeosphaeria quadriseptata and Chaetomium chiversii. J. Nat. Vasconsuelo, A., Boland, R., 2007. Molecular aspects of the early stages of elicitation of
Prod. 70, 1939–1945. secondary metabolites in plants. Plant Sci. 172, 861–875.
Parthasarathy, R., Sathiyabama, M., 2014. Gymnemagenin-producing endophytic fungus Venugopalan, A., Srivastava, S., 2015. Enhanced camptothecin production by ethanol
isolated from a medicinal plant Gymnema sylvestre R.Br. Appl. Biochem. Biotechnol. addition in the suspension culture of the endophyte, Fusarium solani. Bioresour.
172, 3141–3152. Technol. 188, 251–257.
Patwardhan, D.R.A., 2006. Research and Action in Natural Wealth Administration Verma, V.C., Lobkovsky, E., Gange, A.C., Singh, S.K., Prakash, S., 2011. Piperine production
(RANWA). by endophytic fungus Periconia sp. isolated from Piper longum L. J Antibiot (Tokyo)
Pettit, R.K., 2010. Small‐molecule elicitation of microbial secondary metabolites. Microb. 64, 427–431.
Biotechnol. 4, 471–478. Wang, Y., Guo, B., Miao, Z., Tang, K., 2007. Transformation of taxol‐producing endophytic
Pu, X., Qu, X., Chen, F., Bao, J., Zhang, G., Luo, Y., 2013. Camptothecin-producing endophyt- fungi by restriction enzyme‐mediated integration (REMI). FEMS Microbiol. Lett. 273,
ic fungus Trichoderma atroviride LY357: isolation, identification, and fermentation 253–259.
conditions optimization for camptothecin production. Appl. Microbiol. Biotechnol. Wang, J., Lin, W., Wray, V., Lai, D., Proksch, P., 2013. Induced production of depsipeptides
97, 9365–9375. by co-culturing Fusarium tricinctum and Fusarium begoniae. Tetrahedron Lett. 54,
Radman, R., Saez, T., Bucke, C., Keshavarz, T., 2003. Elicitation of plants and microbial cell 2492–2496.
systems. Biotechnol. Appl. Biochem. 37, 91–102. Wang, W.J., Li, D.Y., Li, Y.C., Hua, H.M., Ma, E.L., Li, Z.L., 2014a. Caryophyllene sesquiter-
Ramachandra Rao, S., Ravishankar, G.A., 2002. Plant cell cultures: chemical factories of penes from the marine-derived fungus Ascotricha sp. ZJ-M-5 by the One Strain-
secondary metabolites. Biotechnol. Adv. 20, 101–153. Many Compounds Strategy. J. Nat. Prod. 77, 1367–1371.

Please cite this article as: Venugopalan, A., Srivastava, S., Endophytes as in vitro production platforms of high value plant secondary metabolites,
Biotechnol Adv (2015), http://dx.doi.org/10.1016/j.biotechadv.2015.07.004
A. Venugopalan, S. Srivastava / Biotechnology Advances xxx (2015) xxx–xxx 15

Wang, X.J., Min, C.L., Ge, M., Zuo, R.H., 2014b. An endophytic sanguinarine-producing Zhang, W., Wang, M., Huang, Y., Chea, S.K.P., Zheng, Z., Qian, X., et al., 2011b. New and
fungus from Macleaya cordata, Fusarium proliferatum BLH51. Curr. Microbiol. 68, highly efficient methodology for screening high-yield strains of cytotoxic
336–341. deacetylmycoepoxydiene (DAM). Lett. Appl. Microbiol. 52, 441–447.
Weathers, P.J., Towler, M.J., Xu, J., 2010. Bench to batch: advances in plant cell culture for Zhao, K., Zhou, D.P., Ping, W.X., Ge, J., 2004. Study on the preparation and regeneration of
producing useful products. Appl. Microbiol. Biotechnol. 85, 1339–1351. protoplast from taxol-producing fungus Nodulisporium sylviforme. Nat. Sci. 2, 52–59.
Wei, Y., Zhou, X., Liu, L., Lu, J., Wang, Z., Yu, G., et al., 2010. An efficient transformation Zhao, J., Davis, L.C., Verpoorte, R., 2005a. Elicitor signal transduction leading to production
system of taxol-producing endophytic fungus EFY-21 (Ozonium sp.). Afr. J. Biotechnol. of plant secondary metabolites. Biotechnol. Adv. 23, 283–333.
9, 1726–1733. Zhao, K., Ping, W., Ma, X., Liu, J., Zhou, D., 2005b. Breeding of high-yield strain of taxol by
Wilson, S.A., Roberts, S.C., 2012. Recent advances towards development and commercial- mutagenesis of protoplast and primary discussion of genetic differences between
ization of plant cell culture processes for the synthesis of biomolecules. Plant mutants and their parent strain. Wei Sheng Wu Xue Bao 45, 355–358.
Biotechnol. J. 10, 249–268. Zhao, J., Zhou, L., Wang, J., Shan, T., Zhong, L., Liu, X., et al., 2010. Endophytic fungi for pro-
Xiong, Z.Q., Yang, Y.Y., Zhao, N., Wang, Y., 2013. Diversity of endophytic fungi and screen- ducing bioactive compounds originally from their host plants. Curr. Res. Technol.
ing of fungal paclitaxel producer from Anglojap yew, Taxus × media. BMC Microbiol. Educ. Trop. Appl. Microbiol. Microb. Biotechnol. 1, 567–576.
13, 71. Zhao, J., Li, Y., Shan, T., Mou, Y., Zhou, L., 2011a. Enhancement of diepoxin ζ production
Xu, F., Tao, W., Cheng, L., Guo, L., 2006a. Strain improvement and optimization of the with in situ resin adsorption in mycelial liquid culture of the endophytic fungus
media of taxol-producing fungus Fusarium maire. Biochem. Eng. J. 31, 67–73. Berkleasmium sp. Dzf12 from Dioscorea zingiberensis. World J. Microbiol. Biotechnol.
Xu, F., Tao, W.Y., Cheng, L., Guo, L.J., 2006b. Formation and regeneration of protoplasts of 27, 2753–2758.
taxol-producing endophytic fungus Fusarium maire. J. Food Sci. Biotechnol. 25, 20–24. Zhao, J., Shan, T., Mou, Y., Zhou, L., 2011b. Plant-derived bioactive compounds produced
Xu, L.J., Liu, Y.S., Zhou, L.G., Wu, J.Y., 2009. Enhanced beauvericin production with in situ by endophytic fungi. Mini Rev. Med. Chem. 11, 159–168.
adsorption in mycelial liquid culture of Fusarium redolens Dzf2. Process Biochem. Zhao, J., Zheng, B., Li, Y., Shan, T., Mou, Y., Lu, S., et al., 2011c. Enhancement of diepoxin ζ
44, 1063–1067. production by yeast extract and its fractions in liquid culture of Berkleasmium-like
Xu, L.J., Liu, Y.S., Zhou, L.G., Wu, J.Y., 2010. Optimization of a liquid medium for beauvericin endophytic fungus Dzf12 from Dioscorea zingiberensis. Molecules 16, 847–856.
production in Fusarium redolens Dzf2 mycelial culture. Biotechnol. Bioprocess Eng. 15, Zhao, K., Li, Z., Ge, N., Li, X., Wang, X., Zhou, D., 2011d. Investigation of fermentation con-
460–466. ditions and optimization of medium for taxol production from taxol-producing fungi.
Yamazaki, Y., Kitajima, M., Arita, M., Takayama, H., Sudo, H., Yamazaki, M., et al., 2004. Bio- J. Med. Plants Res. 5, 6528–6535.
synthesis of camptothecin. In silico and in vivo tracer study from [1–13C] glucose. Zhao, K., Sun, L., Wang, X., Li, X., Zhou, D., 2011e. Screening of high taxol producing fungi
Plant Physiol. 134, 161–170. by mutagenesis and construction of subtracted cDNA library by suppression
Yamin, W., Lu, L., Xuanwei, Z., Juan, L., Xiaofen, S., Kexuan, T., 2012. Engineering taxol subtracted hybridization for differentially expressed genes. Wei Sheng Wu Xue Bao
biosynthetic pathway for improving taxol yield in taxol-producing endophytic fungus 51, 923–933.
EFY-21 (Ozonium sp.). Afr. J. Biotechnol. 11, 9094–9101. Zhao, J., Wang, X., Sun, W., Mou, Y., Peng, Y., Zhou, L., 2013a. Medium optimization for
Yang, X.L., Awakawa, T., Wakimoto, T., Abe, I., 2013. Induced production of novel palmarumycin C13 production in liquid culture of endophytic fungus Berkleasmium
prenyldepside and coumarins in endophytic fungi Pestalotiopsis acaciae. Tetrahedron sp. Dzf12 using response surface methodology. Electron. J. Biotechnol. 16, 16.
Lett. 54, 5814–5817. Zhao, K., Sun, L., Ma, X., Li, X., Wang, X., Ping, W., et al., 2013b. Improved taxol production
Yang, H., Wang, Y., Zhang, Z., Yan, R., Zhu, D., 2014a. Whole-genome shotgun assembly in Nodulisporium sylviforme derived from inactivated protoplast fusion. Afr.
and analysis of the genome of Shiraia sp. strain Slf14, a novel endophytic fungus J. Biotechnol. 10, 4175–4182.
producing huperzine A and hypocrellin A. Genome Announc. 2, e00011–e00014. Zhao, X.M., Wang, Z.Q., Shu, S.H., Wang, W.J., Xu, H.J., Ahn, Y.J., et al., 2013c. Ethanol and
Yang, X.L., Huang, L., Ruan, X.L., 2014b. Epigenetic modifiers alter the secondary metabo- methanol can improve huperzine A production from endophytic Colletotrichum
lite composition of a plant endophytic fungus, Pestalotiopsis crassiuscula obtained gloeosporioides ES026. PLoS ONE 8, e61777.
from the leaves of Fragaria chiloensis. J. Asian Nat. Prod. Res. 16, 412–417. Zhou, D.P., Sun, J.Q., Yu, H.Y., Ping, W.X., Zheng, X.Q., 2001. Nodulisporium, a genus new to
Yang, Y., Zhao, H., Barrero, R.A., Zhang, B., Sun, G., Wilson, I.W., et al., 2014c. Genome se- China. Mycosystema 20, 277–278.
quencing and analysis of the paclitaxel-producing endophytic fungus Penicillium Zhou, D., Zhao, K., Ping, W., Ge, J., Ma, X., Jun, L., 2005. Study on the mutagenesis of
aurantiogriseum NRRL 62431. BMC Genomics 15, 69. protoplasts from taxol-producing fungus Nodulisporium sylviforme. J. Am. Sci. 1, 62.
Yin, H., Chen, J.L., 2008. Sipeimine-producing endophytic fungus isolated from Fritillaria Zhou, X., Wang, Z., Jiang, K., Wei, Y., Lin, J., Sun, X., et al., 2007. Screening of taxol-
ussuriensis. Z. Naturforsch. C 63, 789. producing endophytic fungi from Taxus chinensis var. mairei. Appl. Biochem.
Yin, H., Chen, J., 2011. The fermentation conditions of a sipeimine producing endophytic Microbiol. 43, 439–443.
fungus isolated from Fritillaria ussuriensis. J. Northwest Univ. (Nat. Sci. Ed.) 2, 18. Zhou, X., Wei, Y., Zhu, H., Wang, Z., Lin, J., Liu, L., et al., 2008. Protoplast formation,
Yin, H., Sun, Y.H., 2011. Vincamine-producing endophytic fungus isolated from Vinca regeneration and transformation from the taxol-producing fungus Ozonium sp. Afr.
minor. Phytomedicine 18, 802–805. J. Biotechnol. 7, 2017–2024.
Yin, H., Zhao, Q., Sun, F.M., An, T., 2009. Gentiopicrin-producing endophytic fungus Zhou, X., Zhu, H., Liu, L., Lin, J., Tang, K., 2010. A review: recent advances and future
isolated from Gentiana macrophylla. Phytomedicine 16, 793–797. prospects of taxol-producing endophytic fungi. Appl. Microbiol. Biotechnol. 86,
You, X., Feng, S., Luo, S., Cong, D., Yu, Z., Yang, Z., et al., 2013. Studies on a rhein-producing 1707–1717.
endophytic fungus isolated from Rheum palmatum L. Fitoterapia 85, 161–168. Zhu, F., Lin, Y., 2006. Marinamide, a novel alkaloid and its methyl ester produced by the
Yu, T.W., Bai, L., Clade, D., Hoffmann, D., Toelzer, S., Trinh, K.Q., et al., 2002. The biosynthetic application of mixed fermentation technique to two mangrove endophytic fungi
gene cluster of the maytansinoid antitumor agent ansamitocin from Actinosynnema from the South China Sea. Chin. Sci. Bull. 51, 1426–1430.
pretiosum. Proc. Natl. Acad. Sci. U. S. A. 99, 7968–7973. Zhu, F., Lin, Y., Ding, J., Wang, X., Huang, L., 2007. Secondary metabolites of two marine-
Zhang, P., Zhou, P., Yu, L., 2009a. An endophytic taxol‐producing fungus from Taxus × derived mangrove endophytic fungi (strain nos. 1924# and 3893#) by mixed
media, Aspergillus candidus MD3. FEMS Microbiol. Lett. 293, 155–159. fermentation. Chem. Ind. For. Prod. 27, 8–10.
Zhang, P., Zhou, P.P., Yu, L.J., 2009b. An endophytic taxol-producing fungus from Taxus Zu, Y.G., Tang, Z.H., Yu, J.H., Liu, S.G., Wang, W., Guo, X.R., 2003. Different responses of
media, Cladosporium cladosporioides MD2. Curr. Microbiol. 59, 227–232. camptothecin and 10-hydroxycamptothecin to heat shock in Camptotheca acuminata
Zhang, P., Liu, T.T., Zhou, P.P., Li, S.T., Yu, L.J., 2011a. Agrobacterium tumefaciens-mediated seedlings. Acta Bot. Sin. 45, 809–814.
transformation of a taxol-producing endophytic fungus, Cladosporium cladosporioides
MD2. Curr. Microbiol. 62, 1315–1320.

Please cite this article as: Venugopalan, A., Srivastava, S., Endophytes as in vitro production platforms of high value plant secondary metabolites,
Biotechnol Adv (2015), http://dx.doi.org/10.1016/j.biotechadv.2015.07.004

S-ar putea să vă placă și