Jurnal Internasional PDF

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 21

Biotechnology Advances 30 (2012) 1119–1139

Contents lists available at SciVerse ScienceDirect

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

Production of recombinant proteins by filamentous fungi


Owen P. Ward ⁎
Department of Biology, University of Waterloo, Waterloo, Ontario, Canada N2L3G1

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

Available online 24 September 2011 The initial focus of recombinant protein production by filamentous fungi related to exploiting the extraordi-
nary extracellular enzyme synthesis and secretion machinery of industrial strains, including Aspergillus, Tri-
Keywords: choderma, Penicillium and Rhizopus species, was to produce single recombinant protein products. An early
Aspergillus recognized disadvantage of filamentous fungi as hosts of recombinant proteins was their common ability
Trichoderma to produce homologous proteases which could degrade the heterologous protein product and strategies to
Penicillium
prevent proteolysis have met with some limited success. It was also recognized that the protein glycosylation
Recombinant protein
Heterologous
patterns in filamentous fungi and in mammals were quite different, such that filamentous fungi are likely not
Protease to be the most suitable microbial hosts for production of recombinant human glycoproteins for therapeutic
Genome use. By combining the experience gained from production of single recombinant proteins with new scientific
Filamentous information being generated through genomics and proteomics research, biotechnologists are now poised to
Fungi extend the biomanufacturing capabilities of recombinant filamentous fungi by enabling them to express
Pathogenesis genes encoding multiple proteins, including, for example, new biosynthetic pathways for production of
new primary or secondary metabolites. It is recognized that filamentous fungi, most species of which have
not yet been isolated, represent an enormously diverse source of novel biosynthetic pathways, and that the
natural fungal host harboring a valuable biosynthesis pathway may often not be the most suitable organism
for biomanufacture purposes. Hence it is expected that substantial effort will be directed to transforming
other fungal hosts, non-fungal microbial hosts and indeed non microbial hosts to express some of these
novel biosynthetic pathways. But future applications of recombinant expression of proteins will not be con-
fined to biomanufacturing. Opportunities to exploit recombinant technology to unravel the causes of the del-
eterious impacts of fungi, for example as human, mammalian and plant pathogens, and then to bring forward
solutions, is expected to represent a very important future focus of fungal recombinant protein technology.
Crown Copyright © 2011 Published by Elsevier Inc. All rights reserved.

1. Introduction producers of polysaccharides and biosurfactants. Some filamentous


fungi are food materials in their own right, such as mushrooms, single
Filamentous fungi are extraordinary organisms which impact cell protein/biomass (single cell protein, SCP) or indeed lipid-rich bio-
widely on so many aspects of our lives. These organisms are charac- mass (single cell oil, SCO), whereas other fungi are components in
terized as having branched filamentous structures or hyphae having fermented foods. Some are producers of an array of fungal enzymes,
typical diameters of 2–18 um, with (higher fungi) or without (lower for example amylases, amyloglucosidases, cellulases, pectinases,
fungi) cross-walls or septae. Higher fungi include Aspergillus, laccases/ligninases, phytase, proteases, microbial rennets, lipases and
Penicillium, Trichoderma and Fusarium species. Lower fungi include glucose oxidase. Many intracellular fungal enzymes are also exploited
Rhizopus and Mucor species. Filamentous fungi are chemo-organotrophs as biocatalysts in enzyme biotransformations in bioorganic synthesis
meaning they obtain their energy and carbon by oxidation of organic reactions while others use biodegradative processes in soil bioremedia-
compounds. tion. Some strains are well known pathogens of humans, animals and
In traditional fermentation technology filamentous fungi are dom- plants (Cutler et al., 2007; Maor and Shirasu, 2005; Segal and Walsh,
inant producers of a range of primary metabolites, including organic 2006) while others, for example mycorhizal fungi, have beneficial asso-
acids, such as citric, gluconic, fumaric, kojic, itaconic acid and fatty ciations with plants and/or participate in nutrient recycling in soil.
acids. They also produce important secondary metabolites, especially Some fungi are responsible for food spoilage, wood decay and infesta-
as human therapeutics, for example, penicillin, cephalosporin, ergot tion of damp buildings (Bennett, 2006) and many fungal spores are
alkaloids, griseofulvin, lovastatin, taxol and zeranol. Some are known allergens (Meyer, 2004).
The known high productivity chartacteristics of filamentous fungi
are in part related to their inherent abilities to grow at high rates and
⁎ Tel.: + 1 519 888 4567x32427; fax: + 1 519 746 0614. to high biomass densities supported by low cost substrates in rela-
E-mail address: opward@uwaterloo.ca. tively simple fermenters. For industrial fermentations, filamentous

0734-9750/$ – see front matter. Crown Copyright © 2011 Published by Elsevier Inc. All rights reserved.
doi:10.1016/j.biotechadv.2011.09.012
1120 O.P. Ward / Biotechnology Advances 30 (2012) 1119–1139

fungi may be cultivated using traditional surface culture methods, primary or secondary metabolites, or indeed to facilitate production
where oxygen uptake involves passive exposure of the culture to of novel compounds through introduction of new biosynthetic path-
the atmosphere or in a semi-solid culture where a non-homogeneous ways. The tremendous level of metabolic diversity exhibited by cur-
culture may be aerated through various forced air and/or mixing rently known filamentous fungi, together with the knowledge
strategies. They may also be cultured in intensively mixed stirred that only a small number of the estimated 1.5 million species
tank reactors where the objective is to achieve conditions within (Hawksworth, 2001), which are thought to exist, means filamentous
the reactor which approach homogeneity, thereby facilitating more fungi will continue to supply the biosynthetic tools for synthesis of
sophisticated process control. In addition to the beneficial character- a myriad of novel products for countless years to come. This research
istics described above, fungi are especially interesting targets for pro- is still at an early stage and some example roles of recombinant pro-
duction of recombinant proteins because of their demonstrated teins in metabolic engineering of fungi are addressed in Section 5.
capacities to hyperproduce and secrete enzyme proteins, for example Of course, the enormous diversity of fungal organisms does not
glucoamylase production by Aspergillus with impressive titers of imply that a newly isolated natural host capable of biosynthesizing
greater than 25 g/L. novel new beneficial compounds will be a suitable host for large
Processes for production of mold-modified foods have been scale manufacture. Indeed many novel organisms that will be identi-
implemented for many thousands of years. At least the initial stages fied in the future are likely to be the ones that are hard to cultivate,
of these fermented food processes were promoted by surface culture even unculturable and will require better hosts be they other filamen-
suggesting a likely requirement for air, as a source of oxygen, to sup- tous fungal organisms or non-fungal organisms. Efforts are already
port preferential growth of molds on the medium surfaces. Since the being directed to exploiting the unique nature of some of the en-
raw materials were derived from plants including soybeans, wheat zymes or metabolic pathways or systems of filamentous fungi by
and rice, common logic led early scientists to conclude that the transferring these capabilities to other organisms, and some prelimi-
molds had the capacity to at least partially degrade the principal con- nary examples of expression of fungal proteins in other hosts are dis-
stituents of these plant materials, namely the associated carbohy- cussed in Section 6.
drates (including starches, pectins, celluloses and hemicelluloses), Filamentous fungi also interact with other organisms using a vari-
proteins and lipids. Other molds have long been known to participate ety of extremely complicated mechanisms, many of which are as yet
in processes including pathogenesis of plants, spoilage of fruits and poorly understood and their interactions often have overall beneficial
vegetables and rotting of wood, likewise with the presumed involve- and perhaps more frequently negative societal outcomes. Thus some
ment of mold products which could mediate the biodegradation of of the first filamentous fungal organisms to be sequenced were
major structural constituents of these plant materials. human or agricultural plant pathogens and, as we go forward, geno-
Since the principal structural components of plants, being subjected mic and proteonomic research will provide new insights into the mo-
to biodegradative processes during tradititional food fermentations, lecular mechanisms involved in pathogenesis. These studies will
plant pathogenesis, fruit and vegetable spoilage, wood rotting, and re- include cloning and expression of recombinant proteins in fungi as
lated processes, are polymeric substances, early scientists soon postu- prospective candidate causative proteins in pathogenesis followed
lated that cellular assimilation of breakdown products of these by application of strategies to disrupt these proteins with a view to
polymeric substances might require that the substrates be degraded relating these manipulations to pathogenicity and virulence of the
in the extracellular environment. Indeed simple methods for surface pathogen. While this area of research is in its infancy, some indica-
cultivation of the molds involved, on agar-like media containing indi- tions of its potential are included in Section 7.
vidual substrates, often insoluble, demonstrated zones of hydrolysis Some recent reviews on aspects of this review topic are listed in
around the mold colony, illustrating that depolymerization reactions Table 1, including reference to some informative tables from these re-
had been facilitated or catalyzed. It was soon concluded that these bio- view papers.
degradations were mediated by hydrolytic and other depolymerizing
enzymes which were generally secreted by the molds into the extracel- 2. Filamentous fungi as hosts for production of recombinant proteins
lular medium or sometimes were located/attached on the extracellular
surfaces of the biodegradative fungal organism. Jokichi Takamine, a Many filamentous fungi are natural excellent producers of extra-
Japanese immigrant to the United States, was first to commercialize cellular enzymes and hence are exceptional candidate hosts for the
an isolated microbial enzyme. In 1894, he patented a process for prep- production of recombinant proteins (Iwashita, 2002; Wang et al.,
aration of diastatic enzymes from molds which was marketed as Taka- 2005). Organisms such as Aspergillus and Trichoderma species are no-
diastase. The method involved growth of the fungus on the surface of table in their abilities to produce and secrete very high levels of pro-
solid substrates, such as wheat or bran, clearly based on the traditional teins, with Aspergillus niger being capable of producing 25–30 g/L of
processes for preparation of oriental fermented foods (Ward, 1989). glucoamylase and Trichoderma reesei reported to be capable of pro-
The enzyme and producing organism were later characterized as fun- ducing 100 g/L of extracellular protein (Demain and Vaishnav, 2009).
gal alpha-amylase and Aspergillus oryzae, respectively (Gwynne and Meyer (2008) discussed four common strategies for implementa-
Devchand, 1992). tion of transformations of filamentous fungi: The protoplast-mediated
The development of recombinant technology harnesses the power method involves use of cell wall-degrading enzymes for protoplast
of many of these filamentous fungi as hosts in the production of spe- preparation with subsequent uptake of foreign DNA promoted by addi-
cific recombinant proteins as final products with applications in the tion of polyethylene glycol (PEG) and calcium chloride. Noted disad-
agricultural, food and nutrition, biomedical and pharmaceutical, and vantages of this method are that transformations may vary with
energy and industrial sectors (Schuster et al., 2002). This focus has batch variations in the lytic enzyme, that a regeneration procedure is
represented the principal effort in applied genetic engineering over needed and that the high copy number of insertions of DNA may result
the past 25 years or so and is discussed in Sections 2–4 of this review. in a less controlled transformation. In the Agrobacterium tumefaciens
Further advancements of the core transformation technologies transformation, the A. tumefaciens carries a binary vector containing
combined with progress in the fields of genomics and proteomics is the target DNA between a 24-base pair repeating unit and a virulence
leading to a more complex level of host engineering, whereby recom- region required for DNA transfer. The gene-carrier organism is
binant expression of multiple proteins and enzymes is facilitating en- co-cultured with the filamentous fungus and the transformation is ad-
gineering of blocks of new physiological or metabolic machinery into vantageous in that the low copy number of DNA insertions facilitates a
recombinant hosts. An example of these developments relates to our more targeted integration. Beijersbergen et al. (2001) patented a meth-
ability to engineer metabolic pathways so as to enhance production of od for Agrobacterium-mediated transformation of mold species
O.P. Ward / Biotechnology Advances 30 (2012) 1119–1139 1121

Table 1
Some prior reviews related to the topic of production of recombinant proteins by filamentous fungi.

Review—short title Reference Table Contents

Genetic engineering of filamentous fungi Meyer (2008) 1 Industrially important compounds produced by filamentous fungi
Refining heterologous protein production in filamentous Sharma et al. (2009) 2 Heterologous industrial enzymes
fungi 3 Important heterologous proteins in recombinant Aspergillus
Filamentous fungi as cell factories for heterologous protein Punt et al. (2002) 1 Fungal and yeast hosts for hIL-6;
production 4 Systematics of biotechnologically relevant true fungi
Aspergillus as a host for heterologous expression Lubertozzi and Keasling 1 Bioactice fungal metabolites
(2009) 2 Some fungal conversions
3 Some fungal bioremediations
Transcriptional regulation of plant cell wall degradation by Aro et al. (2005)
filamentous fungi
Proteomics of filamentous fungi Kim et al. (2007) 1 List of fungal proteomics papers
Production of human therapeutic proteins by yeasts and Gerngross (2004)
filamentous fungi
Physiology and biotechnology of Aspergillus Ward et al. (2006) IV Aspergillus genes of industrial interest
V Aspergillus recombinant food enzymes
VI,VII,VIII Important heterologous proteins expressed in Aspergillus
IX Patents on recombinant protein production by Aspergillus
Aspergillus as a cell factory for protein production Braaksma and Punt (2008) 1 Effect of secreted protease activity of protease gene disruption strains
Recombinant protein production systems for Aspergillus Fleissner and Dersch (2010) 1,2 Recombinant protein production by aspergilla including hosts and
promoters
Genomics of folding, secretion, glycosylation in aspergilli Geysens et al. (2009) 1 Genes involved in protein-folding, unfolded protein response,
glycosylation
Biotechnology of Trichoderma Schuster and Schmoll
(2010)
Bioconversion of lignocellulose biomass Kumar et al. (2008)
Engineering of Penicillium chrysogenum Harris et al. (2009)
Engineered biosynthesis of peptide antibiotics Stachelhaus et al. (1996) 1 Non-ribosomally synthesized antibiotics and producing hosts
Engineering primary metabolic pathways of industrial Kern et al. (2007)
micro-organisms
Taxol-producing endophytic fungi Zhou et al. (2010) 1 Taxol-producing strains
2 Common transformation methods for filamentous fungi

belonging the Ascomycotina, Basidiomycotina, Deuteromycotina, Mas- A. awamori and A. oryzae, appeared to be better filamentous fungal
tigomycotina and Zygomycotina and illustrated example transforma- hosts for recombinant protein production than some other filamen-
tions for Aspergillus awamori, Aspergillus nidulans, A. niger, tous fungi. The Mucor rennin gene under the control of a suitable
Colletotrichum gloeosporiodes, Fusarium solani, Fusarium graminearum, alpha-amylase promoter, introduced into A. oryzae, resulted in pro-
Neurospora crassa, T. reesei, Pleurotus ostreatus and Agaricus bisporus. duction yields of the heterologous protein of 3.3 g/L (Christensen et
In applying Agrobacterium-mediated transformation strategies to fila- al., 1988).
mentous fungi, for example A. bisporus, Romaine (2002) observed Some perceived or suggested disadvantages of filamentous fungi as
that it was preferable to co-cultivate the bacterium with fruit body tis- heterologous protein hosts relate to their relatively low frequencies of
sue rather than with spores. A third transformation method, which transformation, potential morphological defects, and observed protein
often requires protoplast preparation, involves electric pulse-mediated modifications due to protease activity or low pH (Kinghorn and Unkles,
reversible membrane permeabilization to promote DNA uptake. In con- 1994; Radzio and Kueck, 1997). It was observed that production levels
trast, the fourth more specialized method, which can be implemented of most non-fungal recombinant proteins (mammalian, bacterial,
without cell wall removal, involves shooting DNA-coated metal parti- avian, plant etc.) in filamentous fungi were generally lower as com-
cles at high speed into cells. More specific molecular transformations pared to those of homologous proteins and with likely bottlenecks at
involve targeting of recombinant genes to a specific position in the ge- the level of transcription and translation, secretion, with possible limi-
nome which will enhance transcription of newly introduced DNA tations also at the post-translational level (i.e., inefficient translocation,
and/or deletion of genes with potential to reduce the positive effects folding, transport, processing, or secretion) (Broekhuijsen et al., 1993;
of the desired transformation, be it production of a specific recombi- Gouka et al., 1997a; Jeenes et al., 1994).
nant protein or insertion of multiple enzymes or proteins participating A general model for fungal protein synthesis and secretion, based
in a specific metabolic pathway or other physiological event. on Aspergillus species has been summarized by Fleissner and Dersch
In addition to DNA-based methods, introduction of RNA-based (2010). During synthesis, proteins are directed into the endoplasmic
methods such as antisense RNA, hammerhead ribozymes and RNA in- reticulum where folding takes place and glycosylation is initiated. In
terference approaches have been found to be very useful for silencing Aspergillus species, protein disulfide isomerase (Pdi) assists in the
particular genes in filamentous fungi (Fulci and Macino, 2007; folding and maturation of secretory proteins and the ability of PdiA
Hammond and Keller, 2005; Muller et al., 2006; Yamada et al., 2007). to catalyze the refolding of denatured and reduced RNase has been
Detailed classical physiological and biochemical knowledge is demonstrated (Ngiam et al., 2000). Improperly folded or glycosylated
available for many of the candidate hosts and molecular techniques, proteins are sent to the proteosome or vacuoles for degradation. Fur-
including genome sequencing and annotation strategies. These are ther modification, including glycosylation, occurs in the golgi bodies.
providing data to support efforts in optimizing expression and secre- SNARE proteins facilitate vesicle-mediated trafficking of the proteins
tion of recombinant proteins in filamentous fungi. Filamentous fungi, to the hyphal tip for extracellular secretion. In an interesting experi-
especially well studied Aspergillus species, have also been shown to ment, Gordon et al. (2000) fused a green fluorescent protein sGFP
efficiently implement posttranslational modifications such that heter- (S65T) to truncated A. niger Gla (Gla:499) which was successfully in-
ologous eukaryotic proteins are expressed in a correctly folded form tegrated into the A. niger genome. Confocal fluorescence microscopy
(Kinghorn and Unkles, 1994). Aspergillus species, especially A. niger, confirmed that GFP was partially localized within the hyphal cell
1122 O.P. Ward / Biotechnology Advances 30 (2012) 1119–1139

wall and that protein secretion occurred at the apical or subapical hy- in Table 2. For further information, go to the GRAS website at http://
phal regions. www.accessdata.fda.gov/scripts/fcn/fcnNavigation.cfm?rpt=grasListing.
Limitations at the transcriptional level can be due to low steady- The availability of genomic data, combined with other methods in-
state mRNA levels resulting from a low transcription initiation rate cluding proteomics (deOliveira and deGraaf, 2011) and metabolo-
or more likely from a reduced mRNA stability. It has been suggested mics, is and will continue to support strain development strategies
that at least five structural components may influence mRNA stability. for production of recombinant proteins through use of molecular
In the case of hil6 and aglA transcripts in Aspergillus species, glaA methods for industrial fermentations. For example, comparative ge-
fusions appeared to stabilize mRNA levels (Gouka et al., 1997b). nomic studies among Aspergillus species suggest that A. oryzae, is
Jeenes et al. (1994) reported similar results for fusions of egg-white enriched with genes which participate in the degradation of biomass
lysozyme with glucoamylase. In many cases, low levels of production and in primary and secondary metabolism (Kobayashi et al., 2007).
of recombinant proteins are due to post-translational secretion Also in A. oryzae when cDNA microarrays and expressed sequence
bottlenecks rather than transcription (Conesa et al., 2001; van den
Hombergh et al., 1997).
Table 2
After secretion, a major known problem for heterologous proteins
Examples of GRAS notices filed since 1998 relating to filamentous fungi.
is their degradation by high extracellular enzyme producing filame-
tous fungi, perhaps most notably Aspergillus species which secrete a GRN Substance
diversity of extracellular proteases (van den Hombergh et al., 1997). no.

Proteases have been shown to be responsible for degradation of 8 Pectin esterase derived from Aspergillus oryzae carrying a gene encoding
many recombinant proteins (Broekhuijsen et al., 1993; Roberts et pectin esterase from Aspergillus aculeatus
10 Exopeptidase derived from Aspergillus oryzae carrying a gene encoding a
al., 1992).
leucine aminopeptidase from Aspergillus sojae
Traditional fermentations for production of extracellular enzymes 32 Pectin lyase derived from Trichoderma reesei carrying a gene encoding
by filamentous fungi were based on fermented food processes. These pectin lyase from Aspergillus niger
processes which involved surface or semi-solid media are 34 Aspartic proteinase derived from Aspergillus oryzae carrying a gene
non-homogeneous, making fine process control impossible. This mo- encoding aspartic proteinase from Rhizomucor miehei
43 Lipase derived from Aspergillus oryzae carrying a gene encoding lipase
tivated desires to produce fungal extracellular enzymes in submerged from Thermomyces lanuginosus
culture in stirred tank reactors. Some early challenges with respect to 54 Xylanase derived from Fusarium venenatum carrying a gene encoding
growing filamentous fungi in submerged culture related to the high xylanase from Thermomyces lanuginosus
viscosities which developed in the media caused by the increased 75 Lipase derived from Aspergillus oryzae carrying a gene encoding lipase
from Fusarium oxysporum
concentrations of filamentous biomass, making mass transfer and es-
89 Five enzyme preparations from Aspergillus niger: Carbohydrase enzyme
pecially aeration of these oxygen-requiring organisms more challeng- preparation, catalase enzyme preparation, glucose oxidase enzyme
ing and generally leading to early cessation of growth and limitation preparation, pectinase enzyme preparation, and protease enzyme
of desired protein product yields. These problems were addressed in preparation
various ways at the engineering, physiological and molecular levels. 90 Carbohydrase enzyme preparation from Aspergillus oryzae, protease
enzyme preparation from Aspergillus oryzae, and carbohydrase enzyme
Improvements in fermenter design were directed towards increasing preparation from Rhizopus oryzae
aeration while controlling mycelial shearing effects. High growth and 103 Lipase enzyme preparation from Aspergillus oryzae carrying a gene
product formation rates were achieved by manipulation of fungal constructed from a modified Thermomyces lanuginosus lipase gene and a
morphology, generally to reduce mycelial strand length and promote portion of the Fusarium oxysporum lipase gene
106 Glucose oxidase enzyme preparation from Aspergillus oryzae carrying a
formation of highly branched mycelia. An example of a manipulation
gene encoding a glucose oxidase from Aspergillus niger
at the molecular level was provided by Akin et al. (2003) who maxi- 111 Lipase enzyme preparation from Aspergillus niger
mized heterologous protein production by transforming the cells 113 Lipase enzyme preparation from Aspergillus oryzae
with cotA-encoding nucleic acids controlled by a regulatable promot- 122 Laccase enzyme preparation produced by Aspergillus oryzae expressing
er. Some of these strategies are discussed in more detail elsewhere in the gene encoding a laccase from Myceliophthora thermophila
132 Lactase enzyme preparation from Aspergillus niger
this paper. 142 Phospholipase enzyme preparation from Aspergillus oryzae expressing the
gene encoding a phospholipase A1 from Fusarium venenatum
149 Beta-glucanase enzyme preparation from Trichoderma harzianum
3. Survey of principal players 150 Glucosamine hydrochloride prepared from chitin obtained from
Aspergillus niger
158 Lipase preparation from Aspergillus niger expressing a gene encoding a
Some of the principal organisms involved in food fermentation lipase from Candida antartica
processes were Aspergillus and Rhizopus species. For example, the ini- 183 Phospholipase A2 enzyme preparation from Aspergillus niger expressing a
tial stage of production of soy sauce involves predominant growth of gene encoding a porcine phospholipase A2
A. oryzae strains on a mixture of soybeans and wheat, while produc- 195 Mixed beta-glucanase and xylanase enzyme preparation from Humicola
insolens
tion of tempeh involves cultivation of Rhizopus oligosporus on cooked 201 Asparaginase enzyme preparation from Aspergillus oryzae expressing the
soybean mash. Not surprisingly, these organisms have also been asparaginase gene from A. oryzae
prime candidate hosts for production of recombinant proteins. A 214 Asparaginase enzyme preparation from Aspergillus niger expressing the
combination of our historical knowledge and experience of the per- asparaginase gene from A. niger
230 Chymosin enzyme preparation from Trichoderma reesei expressing the
formance of GRAS (Generally Regarded As Safe) strains with new ge-
bovine prochymosin B gene
nomic information has been used to facilitate the design of a new 238 Lipase enzyme preparation derived from Hansenula polymorpha
generation of genetically modified strains capable of efficient produc- expressing a gene encoding a lipase from Fusarium heterosporum
tion of beneficial recombinant proteins (van Dijck et al., 2003). 296 Lipase enzyme preparation from a genetically modified strain of
The GRAS food additives list of the United States Food and Drug Aspergillus niger
315 Transglucosidase enzyme preparation from Trichoderma reesei expressing
Administration includes enzyme products from A. niger and A. oryzae, the gene encoding transglucosidase from Aspergillus niger
Endothia parasitica, Mucor miehei, Mucor pusillus and others. Since 345 Carboxypeptidase enzyme preparation from modified Aspergillus niger
1998, the FDA has published an inventory of notifications it has re- 333 Acid fungal protease enzyme preparation from Trichoderma reesei
ceived regarding applications for GRAS recognition/exemption. The expressing the gene encoding acid fungal protease from T. reesei
372 Glucoamylase (GA) enzyme preparation from Trichoderma reesei
majority of these notices relating to products of filamentous fungi in-
expressing the gene encoding the GA from T. reesei
volved recombinant proteins. Examples of these listings are included
O.P. Ward / Biotechnology Advances 30 (2012) 1119–1139 1123

tags were used to characterize transcriptional activity associated with niger and its awamori variant. Detailed genetic analyses have also
energy catabolism and hydrolytic enzyme production, transcription been carried out on the human pathogen, Aspergillus fumigatus, but
levels of most catabolic genes of the EM and TCA pathways were ob- because of the severe toxigenic nature of this organism as a producer
served to be higher in glucose-rich conditions as compared with of the highly toxic aflatoxins, it is not relevant as a host for biotech-
glucose-depleted conditions (Maeda et al., 2004). As will be discussed nology production processes. A. nidulans is of particular interest as a
later, interesting studies have also been implemented in traditional in- model filamentous fungal organism for studies of cell biology and
dustrial solid-phase media. For example, rich gene-expression profiles gene regulation. It is also related to A. niger and A. oryzae, which are
for hydrolytic enzymes were observed in wheat-bran media, which the best natural filamentous fungal production hosts. While A.
exhibited lowest expression of catabolic genes. This suggested the latter nidulans is the best genetically characterized Aspergillus species, it is
poor expression may have released catabolite repression of hydrolytic also deemed to be unsuitable as a recombinant host for biotechnology
enzyme synthesis. Gene arrays, gene deletion and insertion strategies processes, because it produces sterigmatocystin, which is also a toxin,
and other emerging molecular techniques will undoubtedly receive albeit much less severe than aflatoxins. A.niger and A. oryzae are also
widespread application as a means to better grasp and exploit the recognized potential broad based recombinant hosts for the biophar-
mechanisms of industrial product formation, regulation, and secretion maceutical industry for production of recombinant proteins.
by Aspergillus species and other filamentous fungi (Akao et al., 2002;
Bautista et al., 2000; Moralejo et al., 2002; Ngiam et al., 2000; Sims et 3.1.1. A. nidulans
al., 2004; Zarrin et al., 2005). Conventional matings lead to the identification of greater than 900
Because of the resource intensive nature of genome sequencing genes in A. nidulans (Brody et al., 1991). The whole genome has a size
and functional analysis, in the case of filamentous fungi, priority at- of 30.1 Mb, with eight well marked chromosomes containing approx-
tention has focussed on the most important strains based on industri- imately 10,000 genes. Physical and chromosomal linkage maps, and
al and/or agricultural productivity considerations or on strains which genome sequence have been described in detail (Archer and Dyer,
represent important human, animal and plant pathogens. In addition 2004; Clutterbuck, 1997; Galagan et al., 2005; Lubertozzi and
some starting species were selected for genomic characterization, for Keasling, 2009; Monsanto, 2001; Sims et al., 2004). Research is con-
example A. nidulans, where substantial beneficial prior physiological tinuing with the ultimate objective of describing and relating expres-
and genetic knowledge had already been established. Consequently, sion patterns, cellular roles and functions of all genes.
pioneering genomic research was implemented on industrial and
pathogenic Aspergillus species. More recent interest in developing
more efficient systems for bioconversion of biomass to energy provid- 3.1.2. A. niger
ed the impetus to characterize the genome of the high cellulose and Sequencing of a derivative of the enzyme-producing strain A. niger
hemicellulase producer, T. reesei. In the case of other industrial extra- NRRL 3122 (ATCC 22343, CBS 115989) indicated a genome size is
cellular enzyme producers which may also be excellent candidate fil- 35.9 Mb, containing 14,097 predicted genes (Archer and Dyer,
amentous fungal hosts for production of recombinant proteins, for 2004). The sequence data for A. niger ATCC strain 9029 is held at the
example certain Penicillium, Rhizopus, Fusarium and Mucor strains, as Pacific Northwest National Laboratory (PNL) and is available to re-
well as some thermophilic fungi, there was insufficient interest searchers upon request. Genencor has access to the A. niger genome
and/or resources to substantially characterize these strains genetical- sequence data of Integrated Genomics (Machida, 2002). The Joint
ly with respect to their enzyme production, secretion function and Genome Institute (JGI) initiated a sequencing program for the citrate-
potential recombinant protein production potential. In some producing A. niger ATCC 1015, in 2004 as part of the United States
of the latter cases, for example in the case of Penicillium and DOE Genome Program, with participation of PNL and Oakridge National
Mucor/Fusarium, detailed sequencing and functional genomic studies Laboratory. Sequencing information on this strain was made available at
have been directed at the highest profile application of these organ- http://www.jgi.doe.gov/aspergillus. The genome sequence and analysis
isms, namely to penicillin production by Penicillium chrysogenum of the ancestor of a current A. niger enzyme production strains, A. niger
and to the plant pathogenic properties of F. graminearum. Mention CBS 513.88, indicates a genome size of 33.9 Mb (Pel et al., 2007).
is made of this research below, in case some of the genomic and Among the 14,165 open reading frames identified, strong functions
functional findings from these studies become relevant and applica- were predicted for 6505 of them. This paper and supplementary infor-
ble to extracellular enzyme-producing strains as potential candidate mation referenced on line includes detailed functional genomic analyses
recombinant protein-producing hosts. In addition, molecular biolo- of protein secretion, carbohydrases and proteases as well as correspond-
gists are applying recombinant technologies to investigate the unique ing comparative analyses among the different Aspergillus species
metabolic properties of these organisms with the expectation that a (A. niger, A. nidulans, A. oryzae and A. fumigatus).
fuller understanding will lead to beneficial societal outcomes. Tsang et al. (2009) combined analysis of proteins secreted by A.
A discussion follows highlighting progress in genomics research as niger with genomic predictions of signal-peptide containing proteins
it pertains to some of the more important industrial filamentous fun- to confirm that the presumed secreted proteins were in fact secreted
gal strains. Most filamentous fungi have estimated genomic sizes of and not the result of cell autolysis. Combining gene expression and
30–40 Mb, encoding 9000–13,000 genes (Machida, 2002). proteomic data for A. niger overproducing strains of lipid, protein
and carbohydrate-degrading enzymes, facilitated identification of
3.1. Aspergillus 898 proteins and demonstrated that the strains exhibited upregula-
tion of proteins participating in carbon- and N-metabolism, as well
The genus Aspergillus consists of more than 180 officially recog- as protein folding and protein degradation. The data enabled re-
nized species, most of which degrade plant polysaccharides (de searchers to manipulate the system incuding overexpression of a
Vries, 2003), and they are particularly important industrial filamen- putative protein glycosylation gene and to increase secretion of a
tous fungi for the large-scale production of both homologous and het- specified enzyme (Jacobs et al., 2009).
erologous enzymes (Fawole and Odunfa, 2003; Wang et al., 2003). A.
oryzae and A. niger are on the Generally Recognized as Safe (GRAS) 3.1.3. A. oryzae
list of the Food and Drug Administration (FDA) in the United States The Japanese National Institute of Technology and Evaluation
(Tailor and Richardson, 1979). completed sequencing of the A. oryzae genome which consists of
Molecular and genetic studies of Aspergillus species most relevant eight chromosomes ranging from 2.8 to 7.0 Mb (Kitamoto et al.,
to recombinant protein production deal with A. nidulans, A. oryzae, A. 1994; Machida, 2002). The total genome size was estimated to be
1124 O.P. Ward / Biotechnology Advances 30 (2012) 1119–1139

36.8 Mb with the number of genes being about 12,074 (Machida et al., aggressive mutation programs, could produce as much as 100 g/L ex-
2005). tracellular protein, with up to 60% as the major cellulase Cel7a (CBHI)
Fleissner and Dersch (2010) recently reviewed the range of re- and 20% of Cel 6a (CBHII).
combinant protein products produced by Aspergillus species. The The complete pattern of proteins related to expression of cellulase
principal host species identified were: A. niger, A. awamori, A. oryzae, and hemicellulase genes in T. reesei was characterized by Ouyang et
A. nidulans and A. terreus. The predominant promoters used for re- al. (2006). Cultivation of T. reesei on cellulose, xylan, a mixture of
combinant protein production were: adhA, alcA, alcC, aldA, amdS, plant polysaccharides or indeed lactose promotes high levels of ex-
amdS, amyA, amyB, aphA, exlA, gdhA, glaA, glaA1, gpdA, oliC, pression of cellulase and hemicellulase genes (Mach and Zeilinger,
pkiA, sodM, sucA, tef1 and tpiA. Recombinant products from humans 2003; Seiboth et al., 2007). Sophorose is thought to be the natural cel-
included: alpha1-proteinase inhibitor, antigen-binding (Fab′) frag- lulase inducer (Sternberg and Mandels, 1979; Vaheri et al., 1979).
ment, corticosteroid binding globulin, epithelial growth factor, granu- That notwithstanding, genomic analysis casts little mechanistic light
locyte macrophage colony stimulating factor, growth hormone, on its enormous protein secretion capacity. Despite its effectiveness
humanized IgG1(kappa) antibodies, interferon-alpha-2, interleukin-6, in degrading plant polysaccharides, suggesting it should contain ex-
lactoferrin, lysozyme, mucus proteinase inhibitor, parathyroid hor- pansions of genes encoding enzymes capable of digesting plant cell
mone, single-chain variable region fragment (scFv) anti lysozyme con- walls, T. reesei contains fewer genes encoding glycoside hydrolases
struct, superoxide dismutase and tissue plasminogen activator. (total 200) than other phytopathogens such as F. graminearum
Recombinant products originating from other animals included: por- (total 243) and Magnaporthe grisea (total 231), A. oryzae (total 285)
cine pancreatic phospholipase A2 and prochymosin; bovine chymosin, or A nidulans (247). It was also noted that while plant polysaccharases
prochymosin and prochymosin B; hen egg-white lysozyme and llama often contain a carbohydrate-binding molecule (CBM) within its re-
antibodies. Recombinant plant proteins expressed in Aspergillus lated group of fungal genomes (N. crassa, F. graminearum, M. grisea
include Thaumatococcus daniellii thaumatin and Cyamosis tetragonoloba and T. reesei), they had the smallest number of CBM-containing
alpha-galactosidase. Recombinant bacterial proteins expressed in proteins.
Aspergillus species included Cellulomonas fimi endoglucanase, Clostridium First efforts to produce heterologous proteins in T. reesei focussed
thermocellum dockerin, Eschericia coli enterotoxin subunit B, beta- on calf chymosin (Harkki et al., 1989; Uusitalo et al., 1991) after
galactosidase, beta-glucuronidase and Thermobifida fusca hydrolase. Re- which Nyyssonen et al. (1993) reported use of this host to produce
combinant proteins from other fungal genera expressed in Aspergillus in- antibody fragments. It was observed that higher production of recom-
cluded: Agaricus meleagris pyranose dehydrogenase, M. miehei binant proteins was generally observed when the original source of
triglyceride lipase and aspartyl protease, Phanerochaete chrysospor- the gene encoding the protein was taxonomically related to the re-
ium lignin peroxidase H8 and manganese peroxidase H4, Pleurotus combinant host. Cellulase gene promoters are most often incorporat-
eryngii peroxidase, Pycnoporus cinnabarinus laccase, Thermomyces ed into cassettes for production of recombinant proteins by
lanuginosus lipase and Trametes versicolor laccase. Many recombi- Trichoderma (Penttila, 1998; Schmoll and Kubicek, 2003), most fre-
nant proteins from one species of Aspergillus have also been quently the signal peptide of Cel 7a (CBHI) which mediates efficient
expressed in another Aspergillus species. The very interesting recombinant protein secretion. This topic was reviewed by Schuster
swollenin-like protein from A. fumigatus which, like swollenin and Schmoll (2010).
from T. reesei, disrupts cellulosic materials and has similarities to the Three recombinant endoxylanases from Chaetomium thermophilum
plant proteins (expansins) which have a cell wall loosening effect, were expressed in T. reesei with a view to facilitating their production
was produced as a recombinant protein in A. oryzae (Chen et al., for application in biobleaching of kraft pulp (Mantyla et al., 2007).
2010). While the protein exhibits no apparent enzyme activity, in The expression cassettes utilized the strong T. reesei cel 7A promoter.
the presence of cellulases, it promoted efficient saccharification of The host was a low protease producer where deletions in the endoglu-
crystalline cellulose. canase I, endoglucanase II and cellobiohydrolase I genes rendered it the
desired low cellulase producer for applications in kraft pulp treatment.
3.2. Trichoderma It was demonstrated that a commercially viable recombinant thermo-
stable xylanase can be produced by T. reesei. Recently, the industrially
The genome sequence of the commercially important high pro- interesting biocatalyst cinnanoyl esterase from an unsuitable host, the
ducer of cellulases and hemicellulases, T. reesei, has been published anaerobic fungus Piromyces equi, was successfully expressed in recom-
(Martinez et al., 2008) while analysis and annotation of the genomes binant T. reesei as a more suitable producing host (Poidevin et al.,
of two biocontrol species, Trichoderma altroviride and Trichoderma 2009).
vireus are proceeding. T. reesei is a soft-rot ascomycete filamentous Substantial effort has focussed on transforming fuel ethanol yeast
fungus with a long and safe track record as a producer of commercial strains with cellulolytic genes from Trichoderma species to facilitate
cellulases, initially with applications in food processing (Nevalainen their ability to ferment cellulose to ethanol. In a recent example,
et al., 1994). Studies aimed at understanding and optimizing factors Huang et al. (2010) described cloning and expression of the endoglu-
affecting productivity and catalytic efficiency of cellulases are funda- canase gene egVIII from Trichoderma viride into Saccharomyces
mental to overcoming the major biomass pre-treatment obstacle to cerevisiae.
commercialization of processes for production of bioenergy from lig-
nocellulose biomass. Applications of its cellulase and hemicellulase 3.3. Penicillium
compliment in the pulp and paper and textile industries are also im-
portant (Buchert et al., 1998; Galanti et al., 1998). T. reesei represents Limited genomic sequencing information appears to be available
a principal target cellulase host in the quest to replace gasoline with on potential recombinant protein-producing filamentous fungi other
cellulose-derived ethanol. than Aspergillus and Trichoderma species. As selected Penicillium spe-
cies, for example Penicillium purpurogenum, Penicillium funiculosum
3.2.1. T. reesei and Penicillium (Talaromyces) emersonii are high producers of cellu-
T. reesei has a genome size of 33 Mb and seven chromosomes lases, hemicellulases and pectinases, they may have considerable po-
(http://genome.jgf-psf..org/Trire2/Trire2.home.html). The predicted tential as recombinant protein-producing hosts. Chavez et al. (2010)
number of genes in the genome was 9129 (Martinez et al., 2008). carried out transformation studies and demonstrated high transfor-
T. reesei has an extraordinary ability to secrete proteins. Cherry and mation frequencies in two cheese ripening fungi, Penicillium
Fidantsef (2003) reported that some industrial strains following camemberti and Penicillium roqueforti, which exhibit low protease
O.P. Ward / Biotechnology Advances 30 (2012) 1119–1139 1125

activity. They concluded that these species had all of the right strain pathogenic strains and indeed the first of the zygomycetes to be
characteristics as suitable hosts for production of recombinant pro- fully sequenced was Rhizopus oryzae which is the primary cause of
teins. Gonzalez-Vogel et al. (2011) recently identified a number of the potentially lethal angioinvasive mucormycosis infection (Ibrahim
protein complexes containing enzymes including arabinofuranosi- et al., 2003; Kwon-Chung and Bennett, 1992). Related Mortierella spe-
dases, beta-glucosidase, xylanases, acetyl esterases and ferulyl ester- cies are of great interest in the area of lipid production and molecular
ases, in the soft rot fungus P. purpurogenum using a proteomics transformations involving these species are being investigated (Mac-
strategy. Guais et al. (2010) prepared a partial DNA library for P. Kenzie et al., 2000). Nevertheless, as is indicated below, some of the
funiculosum and sequenced genes encoding four GH54 α-L- genomic information is directly relevant to the long established abil-
arabinofuranosidases. This organism has been used to produce com- ities of this and related strains to produce hydrolytic enzymes.
mercial mixtures of enzymes degrading complex agricultural residues
containing cellulose, hemicellulose, arabinoxylan, arabinogalactan, 3.4.1. Rhizopus oryzae
proteases etc., with applications as a feed additive to enhance feed di- The genome sequence of R. oryzae strain 99–880, isolated from a
gestibility. The enzyme mixture contained more than fifty separate fatal infection of mucormycosis, has recently been published (Ma et
proteins (Guasi et al., 2008). The strong gpdA promoter from A. al., 2009). Total length of the R. oryzae genome was found to be
nidulans was used to promote overexpression of pectin lyase by Pen- 45.26 Mb while total number of protein-encoding genes was 17,467.
icillium griseoroseum in submerged fermentation production systems Evidence was provided that there is whole-genome duplication in
(Cardoso et al., 2010). Penicillium canescens was transformed with a this strain mainly attributed to an ancestral duplication event. Of spe-
vector encoding the laccase of Trametes hirsute under control of an ef- cific interest to diagnostic and therapeutic treatment of mucormyco-
ficient promoter of the bgaS gene of P. canescens and efficiently sis is the genomic characterization of expanded families of cell-wall
expressed and secreted the recombinant protein (Abianova et al., synthesis enzymes required for fungal cell wall metabolism but
2010). which are not present in mammalian hosts and hence which may
As might be expected, the predominant effort related to sequenc- be targeted by novel future drugs. Of interest both to therapy as
ing and annotation of Penicillium has been directed at the principal well as to use of R. oryzae and related species as hosts for recombinant
producer of penicillins, P. chrysogenum. While antibiotic-producing protein production are the annotated expanded gene families of se-
strains are generally not considered as suitable hosts for production creted proteases characterized, especially aspartic proteases and sub-
of natural or recombinant enzymes or other proteins for use in tilases. It was suggested that these proteases may mediate the
foods or pharmaceuticals, some of the P. chrysogenum genomic infor- pathogenic infection process, as these enzymes have previously
mation may be applicable to development of non-antibiotic- been thought to be associated with virulence of pathogenic Rhizopus
producing Penicillium strains as recombinant protein-producing species (Schoen et al., 2002; Spreer et al., 2006). In this case, these
hosts. Promoters of the genes encoding glutamate dehydrogenase, proteases may mediate penetration of hyphae through decaying or-
β-acetylhexosaminidase and gamma-actin from P. chrysogenum may ganic matter (Ma et al., 2009).
be used to construct potent vectors for expression and secretion of
homologous and heterologous proteins in these strains and also in 3.5. White rot fungi
other hosts (Barredo Fuente et al., 2001).
White rot fungi are basidiomycetes that are of great interest as en-
3.3.1. P. chrysogenum zyme producers as they produce unique extracellular oxidative en-
The complete genome sequence of the penicillin producer P. zymes that degrade lignin which surrounds and protects cellulose
chtysogenum Wisconsin 54–1255 strain (ATCC 28089, see Elander, microfibrils of plant cell walls, especially woody plants. The white
1983) was published in 2008. Genome size was 32.19 Mb, compara- rot fungi are particularly important because they degrade the lignin
ble with that of other filamentous fungi, and the total gene number while not attacking the cellulose. These filamentous fungi are the
was 12,943 (van den Berg et al., 2008). In addition to cellular func- only microbes capable of efficient depolymerization and mineraliza-
tional characterization of the P. chrysogenum genes, particular atten- tion of lignin. P. chrysosporium has been the most intensively studied
tion was paid to the penicillin biosynthetic genes. This information white rot fungus. White rot fungi secrete an array of peroxidases and
may provide more general direction for manipulation/engineering oxidases that attack lignin non-specifically by producing lignin-free
of metabolic pathways to increase production of natural target me- radicals, which subsequently facilitate spontaneous cleavage reac-
tabolites or indeed to facilitate production of wholly novel metabo- tions (Kirk and Farrel, 1987). These enzymes also participate in deg-
lites in filamentous fungi. The transcriptomes of the sequenced radation of organic pollutants in bioremediation. Recently,
strain and a high penicillin-producing strain were compared and high-resolution two dimensional electrophoresis-based proteomics
as might have been expected, many of the genes involved in synthe- coupled to LC-MS/MS was used to monitor enzyme expression and
sis of the penicillin precursors, valine, cysteine and α-aminoadipic chemical products present during the process of degradation of aro-
acid were observed to be increased in the high penicillin-producing matic substrates by P. chrysosporium, as a means of gaining a better
strain. Some genes were identified which control β-lactam output insight into the process of lignin degradation (Matsuzaki et al.,
and genes with predicted roles as transporters appeared to be upre- 2008). Not surprisingly, the first basidiomycete genome to be se-
gulated under penicillin-producing conditions. Culmination of this quenced was the white rot fungus P. chrysosporium.
work clearly represents a milestone for future metabolic engineer-
ing strategies, which of course may involve participation or use of 3.5.1. P. chrysosporium
recombinant proteins. Its thirty million base-pair genome was sequenced using a whole
genome shotgun method. The genome length was 29.9 Mb, similar
3.4. Rhizopus in size to most of the other sequenced filamentous fungi genomes.
The genome contains 11,777 protein coding genes. Analysis of the ge-
A number of important extracellular industrial and medical en- nome indicates an array of genes which encode secreted enzymes, in-
zymes are produced by the zygomycetes, including the important mi- cluding oxidases, peroxidases and hydrolytic enzymes which are
crobial rennets produced by Rhizomucor miehei and Rhizomucor known to co-operatively cause wood decay (Martinez et al., 2004).
pusillus and digestive lipases, proteases and amylases are produced Recombinant proteins have been expressed in a variety of basidio-
by Rhizopus arrhizus. However, the major fungal genomics resources mycetes. For example, a vector encoding interleukin-32, the human
related to this group of filamentous fungi have been directed to cytokine associated with some inflammatory and autoimmune
1126 O.P. Ward / Biotechnology Advances 30 (2012) 1119–1139

diseases, was successfully introduced and expressed in the edible 3.7. N. crassa
mushroom P. eryngii, via an A. tumefaciens transformation (Chung et
al., 2011). There is continuing interest in expressing the diverse While N. crassa is not recognized as an important industrial host, it
degrading enzymes from basidiomycetes in more conventional indus- is included in this discussion as a powerful model filamentous fungal
trial work-horse hosts. For example ligninolytic basidiomycetes con- system which has been characterized biochemically and genetically.
tain a sugar oxidoreductase (pyranose dehydrogenase) that has very This host can be grown at high growth rates in simple defined
broad substrate specificity towards breakdown constituents of ligno- media and can produce high amounts of recombinant proteins. The
cellulose. In order to extend the biodegradative capability of more approximate genome size is 40 Mb, and it contains about 10,000 pro-
conventional industrial strains, this enzyme from A. meleagris was tein-coding genes and many of the genes involved in interesting aspects
heterologously expressed in A. nidulans and A. niger (Pisanelli et al., of Neurospora biology, including its secondary metabolism have been
2010). The white rot fungus, T. versicolor produces two groups of lac- annotated (Colot et al., 2006; Galagan et al., 2003). Up-to-date informa-
cases with several isoforms. Two of these laccases were expressed as tion may be obtained online from http://www.fgsc.net.
recombinant enzymes in A. oryzae and the recombinant enzymes Tian et al. (2009) applied microarray and shotgun proteomics
exhibited catabolic degradative activity against hydroxylated PCBs analysis on strains of a cellulolytic N. crassa fungus grown in different
(Fujihiro et al., 2009). Rodgers et al. (2010) noted that while basidio- media in order to combine data from gene expression and the proteome/
mycetes are the predominant sources of laccases with potential large secretome in an attempt to better understand the cellulose-degrading
applications in delignification, basidiomycetes are in general not as system and the principal genes involved.
versatile or suitable as industrial fermentation producers as com- Recently, N. crassa has been used as a host for production or re-
pared to ascomycetes and consequently much effort has focussed on combinant subunit vaccines including influenza hemagglutinin (HA)
transforming the more suitable fermentation hosts to produce recom- and neuraminidase antigens (NA) (Allgaier et al., 2009). High molec-
binant basidiomycetes laccase. However, there have been problems in ular weight particles containing NA could be generated in a hetero-
achieving production of recombinant laccases in good fermentation karyon expression system facilitating downstream processing on the
hosts primarily due to glycosylation deficiencies and these challenges one hand but also enables mixtures of different antigens to be co-
are currently being addressed with a view to mass producing effective expressed together, thereby facilitating tailoring of a vaccine directed
laccases. at a particular pathogen target or variant.
Sakaki and Munetsuna (2010) have surveyed the various enzymes
which could co-operate to degrade complex pollutants such as poly- 3.8. Selected key genomic resources
chlorinated dibenzo-dioxins and furans, including angular dioxygenase,
cytochrome P450 (CYP), lignin peroxidase, manganese-dependent per- A variety of institutional and online resources are available to re-
oxidase and dehalogenase and concluded that combinations of distinct searchers with interests in genomic aspects of filamentous fungi and
enzymes could have significant application in these biodegradations. are clearly relevant to the topic of recombinant protein production
Given that white rot fungi already produce lignin and Mn-dependent by these hosts. Reference is made to some of these below:
peroxidases and CYPs it was concluded that supplementing this host http://www.aspgd.org/ “is the home of the Aspergillus Genome Da-
by adding additional recombinant capability would make this organism tabase, a resource for genomic sequence data and gene and protein in-
a very powerful bioremediation strain. While the risks associated with formation for Aspergillus species. AspGD is based on the Candida
releasing genetically engineered organisms to the environment were Genome Database and is funded by the National Institute of Allergy
recognized, it was suggested this could be addressed by creating suicid- and Infectious Diseases at the US National Institutes of Health”. Subsites
al engineered strains (Paul et al., 2005). deal with the annotated Aspergillus genomes of strains of A. fumigatus,
A. clavatus, A. nidulans, A. niger, A. oryzae and Aspergillus terreus.
The aim of the JGI Fungal Genomics Program is “to scale up sequenc-
3.6. Fusarium ing and analysis of fungal genomes to explore the diversity of fungi in
DOE mission areas and to develop the Genomic Encyclopedia of Fungi
While a high profile Fusarium species, F. graminearum is the caus- in the areas of: Plant feedstock health (mycorrhizal symbiosis, plant
ative agent of some important plant diseases, other Fusarium strains pathogenicity, biocontrol); Biorefinery (lignocellulose degradation,
are used in fermentations processes, including production of single sugar fermentation, industrial organisms) and Fungal diversity” (http://
cell protein approved for human consumption and some of these jgi-psf.org/programs/fungi/about-programs.jsf). Subsites deal with im-
strains may have potential for production of recombinant proteins. portant filamentous hosts including Aspergillus carbonarius, P. chrysos-
Nevertheless, the predominant scientific research to date has fo- porium, Sporotrichum thermophile, Thielavia terrestris, T. versicolor and
cussed on F. graminearum which causes plant diseases of substantial T. reesei.
economic importance including Fusarium ear root of maize and The Fungal Genome Initiative (FGI) of the Broad Institute of MIT
head blight of cereals. In addition F. graminearum produces myco- and Harvard “produces and analyzes sequence data from fungal or-
toxins in infected plants which, if they find their way into food and ganisms that are important to medicine, agriculture and industry.
feed products, constitute a health risk. Over 50 fungi have been sequenced or are being sequenced, including
human and plant pathogens as well as fungi that serve as basic
models for molecular and cellular biology. In partnership with the
3.6.1. F. graminearum wider fungal research community, organisms are selected for se-
The sequencing and annotation of F. graminearum was reported by quencing as part of a cohesive strategy that considers not only the
Cuomo et al. (2007) and gene annotation information was revisited value of data from each organism given their role in basic research,
by Wong et al. (2011). Updated resource information may be health, agriculture, and industry, but also their value in comparative
assessed at http://mips.gsf.de/genre/proj/FGDB/. genomics”. It includes databases on R. oryzae and on the Fusarium
The Cuomo et al. paper indicates a genome size of 36.1 Mb includ- Comparative project. (http://broadinstitute.org/scientific-community/
ing 32 genes being predicted plant cell-degrading enzymes, including science/projects/fungal-genome-initiative/fungal-genome-initiative).
xylanases, pectate lyase and cutinases which were postulated to func- The Fungal Genetics Stock Center (http://www.fgsc.net) “is a re-
tion in pathogenesis by facilitating plant tissue penetration and mac- source available to the Fungal Genetics research community and to
eration and nutrient provision for the invading organism. The recent educational and research organizations in general. The FGSC is funded
annotated information indicated a set of 13,718 protein coding genes. largely by a grant from the National Science Foundation (Award
O.P. Ward / Biotechnology Advances 30 (2012) 1119–1139 1127

Number 0235887) of the United States of America and to a lesser extent levels of a variety of secreted recombinant proteins including bovine
by the payments made by researchers who use our services. Most fungal prochymosin (Ward et al., 1990), hIL-6 (Broekhuijsen et al., 1993),
strains in the FGSC collection are listed in the online searches. Specific hen egg-white lysozyme (Jeenes et al., 1993), human lactoferrin
groups of materials are listed by category include: Neurospora, (Ward et al., 1992, 1995), and phytases from A. awamori (Martin et
Aspergillus, Fusarium and Magnaporthe, Ustilago, Cryptococcus, other al., 2003).In the case of chymosin and lactoferrin production, gram
Fungi”. The FGSC together with other organizations is a major sponsor to multigram quantities of recombinant product were produced per
of the Fungal Genetics Conference (http://www.fgsc.net/26thFGC/ liter when the high-level-production strains were put through a mu-
index.htm). tation program (Dunn-Coleman et al., 1991; Ward et al., 1995). En-
The above websites in turn provide links to other resources. hancements to this approach involved use of the catalytic domain of
glucoamylase rather than the complete enzyme (Gouka et al.,
4. Improving recombinant protein expression in filamentous fungi 1997b). To facilitate subsequent cleavage of the two protein ele-
ments, a linker proteolytic processing site is incorporated between
4.1. Molecular strategies the carrier moiety component and the protein of interest. The linker
region is designed to allow the catalytic domain and the rest of the fu-
A primary practical motivation for studying gene expression in fil- sion protein to fold independently. The N-terminal fungal protein ap-
amentous fungal hosts is to understand the molecular mechanisms of pears to serve as a carrier, improving translocation of the
transcription regulation in these organisms and to improve recombi- recombinant protein into the ER, as well as its folding, is mediated
nant protein expression, especially by the study of DNA sequences by the N-terminal fungal protein. Subsequently in most cases the fu-
participating in transcription initiation and/or regulation and selec- sion protein is cleaved facilitating secretion of the separate proteins
tion of strong promoters. Transcriptional regulation of extracellular by a KEX2-like endopeptidase at a KEX2 recognition site introduced
plant cell wall-degrading enzymes produced by filamentous fungi specifically into the fusion protein as a linker, as indicated above
has been reviewed by Aro et al. (2005). (Broekhuijsen et al., 1993; Punt et al., 2002; Ward et al., 1990, 1995).
The promoter regions of the Aspergillus amylase genes consist of Fidelity of cleavage of the KEX2 processing site: Sometimes, aber-
four highly conserved sequences, one of which (region IIIa) is essen- rant forms of the recombinant product are observed when gene
tial for high-level expression and another of which (Region IIIb) con- fusion strategies are employed. When a part of the fungal glucoamy-
tains sequences thought to enhance expression in combination with lase protein (GAM), linked via a KEX2 processing site, was also used
region IIIa (Minetoki et al., 1998). A sequence of CCAAT present in in a gene-fusion strategy in A. niger to produce extracellular bovine
the promoter region of the A. nidulans amdS (encoding acetamidase) pancreatic trypsin inhibitor (BPTI), aberrant forms of the recombi-
is required for high-level expression of amdS, and related CCAAT se- nant protein were attributed to possible variations in A. niger
quences are present in the promoter regions of a number of other A. KEX-2-like endoprotease point of attack of the GAM-BPTI fusion pro-
nidulans genes (Papagiannopoulos et al., 1996). One of the most tein or indeed involved another endoprotease (MacKenzie et al.,
strongly expressed genes in A. oryzae, the enolase gene (enoA), con- 1998). For example, while the desired recombinant protein is normal-
tains a15-bp element with a sequence essential for transcription reg- ly linked to the glucoamylase via a Lys-Arg KEX2-like cleavage site in
ulation of the gene (Toida et al., 2000). The melO promoter appears to A. niger, the fidelity of cleavage to release mature protein is not al-
be effective as a mediator of strong synthesis of recombinant proteins ways observed to be consistent and appears to be also influenced by
in Aspergillus hosts (Ishida et al., 2001). The A. oryzae TAKA-amylase sequences immediately downstream and upstream of the KEX2 site
promoter preceded by its upstream activating sequences, was found (Spencer et al., 1998).
to be suitable for expression of protein products in Aspergillus species The protein, neoculin (NCL), naturally produced in the fruits of the
(Boel et al., 1996). Berka et al. (2002) patented novel vectors contain- tropical plant Curculigo latifolia, is about 500 times sweeter than
ing polyadenylation sequences linked to the 3′ terminus of the DNA sugar. It is a heterodimer consisting of an N-glycosylated acidic subu-
sequence encoding the heterologous protein and which may include nit (NAS) and a basic subunit (NBS) linked by disulphide bonds. Re-
promoter and signal sequences for promotion of expression and se- combinant neoculin (rNCL) was produced in A. oryzae by using
cretion of heterologous proteins in filamentous fungi. Schmoll et al. separate NAS and NBS constructs, each fused to the A. oryzae
(2010) described the construct used to produce class 1 hydrophobin α-amylase via KEX2 cleavage sites (Nakajima et al., 2006). The NAS
from A nidulans in T. reesei. When the class II hydrophobin-encoding component was properly N-glycosylated and the sweetness proper-
promoter from T. reesei, hfb2, was used with lactose as carbon source, ties of the rNCL were comparable with the native NCL.
the majority of the recombinant protein was secreted into the medi- Gene fusion strategies are also exploited to produce expressed
um by T. reesei. In contrast when the T. reesei cel7A promoter was proteins containing a tag that may facilitate product extraction during
used, the recombinant protein was not secreted into the medium, downstream processing. By way of example, Collen et al.(2001) ge-
but remained cell wall-bound. High expression of the fumR gene, netically engineered endoglucanase (Cel7B) from T. viride with a pep-
which encodes fumarase in a high fumaric acid producing strain of tide extension containing non-polar tryptophan-proline residues
R. oryzae, was observed under good fumaric acid-producing condi- which facilitated preferential partitioning of the protein into the less
tions (high sugar, low N) and the regulation of this gene may be of in- polar phase of an aqueous two phase model system.
terest for production of recombinant proteins and metabolic
engineering in Rhizopus species. Gene expression was primarily regu- 4.1.2. Overproduction of foldases and chaperones
lated at the level of transcription. Foldases catalyze the isomerizations and disulfide bond forma-
tions and molecular chaperones, which are non-catalytic, mediate
4.1.1. Gene-fusions strategies folding of the nascent polypeptides into functional proteins and pre-
Some early recombinant research on filamenous fungal sought to vent non-productive protein–protein interactions (Conesa et al.,
produce recombinant proteins by ‘coat-tailing’ a hyper-produced 2000). Chaperones may act in diverse ways such as identifying defec-
and secreted homologous protein with subsequent cleavage of result- tive proteins in the ER, inducing synthesis of folding enzymes or in-
ing fused proteins. Thus, techniques involving fusing the target gene deed ER-associated protein degradation responses for degradation
to the 3′ end of a homologous gene encoding glucoamylase improved of defective proteins.
production of recombinant proteins, for example, of mammalian pro- It has been postulated that hyper-production of recombinant pro-
teins, by filamentous fungi (Gouka et al., 1997a, b). Fusions to the glu- teins into the ER has the potential to overload the folding, assembly
coamylase gene of A. niger/A. awamori promoted production of high and secretion machinery of filamentous fungi. Therefore, the effects
1128 O.P. Ward / Biotechnology Advances 30 (2012) 1119–1139

of overexpression of genes for several ER chaperones and foldases in may induce an immune response in the patient being treated, reduc-
filamentous fungi, including bipA (from a family of binding proteins, ing treatment efficacy. Engineering humanized glycosylation path-
BiP), pdiA (from a family of protein disulfide isomerase) and a family ways into filamentous fungi, including trimming the branches of
of calnexins on overproduction of recombinant proteases have been high mannose-containing glycoproteins, has been found to be very
evaluated (Conesa et al., 2001; Jeenes et al., 1997; Ngiam et al., complex (Gerngross, 2004).
2000; van Gemeren et al., 1997). It was found that filamentous Antitrypsin, the human a1-proteinase inhibitor (a1-PI), is the
fungi overproducing specific proteins, both homologous and heterol- most abundant inhibitor of serine proteases in plasma (Brantly et
ogous, exhibited increased levels of bipA transcription, whereas direct al., 1991). Progressive emphysema develops in antitrypsin-deficient
interventions to overexpress bipA overexpression appeared not to af- patients ultimately leading to death (Crystal, 1996). Conventional
fect yields of secreted proteins (Punt et al., 1998). Overproduction of antitrypsin-inhibitor replacement therapy uses a limited plasma de-
fungal proteins generally increased bipA mRNA levels in A. niger. In rived source which has created momentum for production of the re-
the case of two transformed A. niger strains which produced HEWL, combinant form. While several hosts have been tested for efficacy of
a twofold induction in bipA mRNA levels was observed (Ngiam et production, altered glycosylation patterns or complete absence of gly-
al., 2000). BiP overexpression did not increase secreted levels of cosylation in the recombinant product reduced in vitro stability of the
hIL-6 in Aspergillus (Gouka et al., 1997a) and pdiA overexpression inhibitor and resulted in its rapid removal from the circulation system
did not increase secreted yields of HEWL in A. niger (Ngiam et al., (Karnaukhova et al., 2006).
2000). Disruption of a vacuolar protein sorting receptor gene in A. A mature and biologically active glycosylated recombinant a1-PI pro-
oryzae, which targets aberrant and recombinant proteins for vacuolar duced by A. niger exhibited improved stability over a non-glycosylated
degradation, enhanced production and secretion of the bovine chy- recombinant product produced by E. coli (Karnaukhova et al., 2007).
mosin and human lysozyme heterologous proteins (Yoon et al., The recombinant protein was fused to a well secreted native fungal pro-
2010). tein with a KEX2 recognition site at the fusion junction which was
cleaved in vivo by a KEX2-type protease. Implementation of strategies
4.1.3. Glycosylation for increasing glycosylation in Aspergillus resulted in increased pro-
Glycosylation patterns from filamentous fungi are more similar to duction of the recombinant protein chymosin (van den Brink et al.,
those of mammals than the patterns observed in common yeast hosts 2006). In one case, a poorly used glycosylation site within the chy-
(Maras et al., 1999a, 1999b; Nevalainen et al., 2005). The two main mosin molecule was improved resulting in much more efficient pro-
glycosylation processes common to eukaryotes involve N- and duction of the glycosylated chymosin. In the second case, when the
O-glycosylation, whereby oligosaccharides attach to the beta-amide N-glycosylation site was located away from the native chymosin at-
moiety of asparagine residues and mainly to serine and threonine tached via a linker, a substantial increase in recombinant protein
β-hydroxy groups. N-glycosylation involves transfer of pre- was observed.
assembled glycosyl precursors to specific asparagine residues of the
nascent polypeptide chain after which glycosidase- and glycosyl 4.1.4. Other molecular strategies
transferase-mediated modifications of the oligosaccharide occur The following are miscellaneous examples of molecular strategies
resulting in production of a common trimannosyl-chitobiose core used to enhance production of recombinant proteins by filamentous
with branched N-acetylglucosamine residues generating the high fungi:
mannose N-glycans characteristic of filamentous fungi and yeasts.
O-glycosylation in fungi starts in the endoplasmic reticulum, and in- – Hastrup et al. (1997) proposed production of a proenzyme, in
volves O-mannosylations resulting in the sequential build up of the cases where the enzyme was unstable or harmful to the producing
O-glucosyl structure. Geysens et al. (2009) has recently used analysis host, which could be proteolytically activated after secretion.
of the genome sequences to review folding, secretion and glycosyla- – An activator protein binding site containing the CCAAT sequence
tion, especially the N-glycosylation processes while Goto (2007) has was identified within the cis regulatory region of the A. niger
described the O-glycosylation process, both in Aspergillus. glaA gene. Insertion of multiple copies of this binding site into
Filamentous fungi have two distinct alpha-1,2-mannosidases, one the promoter of transformed recombinant plasmid sequence en-
of which is similar to the mammalian Golgi alpha-1,2-mannosidases hanced promoter production of the heterologous protein (Liu et
that trim 3 mannose moieties off Man8GlcNAc2 to form Man5GlcNAc2 al., 2003).
as substrate for GlcNAc transferase 1, and another distinct fungal – Berka et al. (2002, 2003) disclosed constructed novel vectors,
alpha-1,2-mannosidase (Ichishima et al., 1999; Yoshida et al., 2000). which encoded the desired heterologous polypeptide and a secre-
However, the mammalian-like enzyme is neither well expressed nor tory sequence functional in the filamentous fungus secretory
secreted such that very little of the lower mannosylated moiety gets system.
transferred (Maras et al., 1997). N-glycans from fungi also differ – A. oryzae produces two predominant proteases, serine-type car-
from mammalian N-glycans in having terminal altered substituents boxypeptidase (CPase) and aspartic endopeptidase under acidic
such as glucose, galactose or phosphoesters (De Pourcq et al., 2010). conditions (Takuchi and Ichishima, 1986).
Maras et al. (1997) employed recombinant mammalian A typical antisense control strategy, whereby vectors are created
beta-1,4-galactosyl transferase and alpha-2,6-sialyltransferase to to express a high level of the antisense RNA complementary to
make T. reesei cellobiohydrolase 1 more mammalian-like with respect the RNA transcript of a target gene, used to inhibit fungal gene ex-
to its glycosylation pattern. Recombinant human β-1,2-GlcNAc trans- pression, was used to isolate an low CPase-producing A. oryzae
ferase was subsequently overexpressed in Trichoderma, thereby en- mutant expressing high and stable levels of lysozyme (Zheng et
hancing its GlcNAc transfer capability (Maras et al., 1999a, 1999b) al., 1998).
and similar transformations with the corresponding rat GlcNAc trans- – Researchers had limited success in striving for overproduction of
ferase were implemented in A. nidulans (Kasajima et al., 2006). Kainz manganese peroxidase in its natural host, P. chrysogenum (Cullen,
et al. (2008) has carried out other molecular stratefies to successfully 1997). However, a combination of strategies including use of a
produce lower mannosylated Man3GlcNAc2 N-glycans in recombi- strong glucoamylase promoter, a protease-deficient A. niger host,
nant Aspergillus strains. culture pH manipulation and incorporation of hemin into the cul-
For production of therapeutic proteins, glycoform is very impor- ture medium facilitated strong recombinant enzyme production
tant as incorrectly glycosylated proteins, for example recombinant (Broekhuijsen et al., 1993; Conesa., 2001; Conesa et al., 2000;
human therapeutic glycoproteins produced by filamentous fungi, Punt et al., 2002; Stewart et al., 1996).
O.P. Ward / Biotechnology Advances 30 (2012) 1119–1139 1129

– Promoters of the genes encoding glutamate dehydrogenase, media, typically induces protease production by A. niger (Ahamed et
beta-acetylhexosaminidase and gamma-actin from P. chryso- al., 2005) and productivity of secreted egg lysozyme by a recombinant
genum may be used to block expression of undesired genes strain of A. niger was reduced in such rich media (Archer et al., 1990).
through anti-sense construction (Barredo Fuente et al., 2001). Double disruption of the two protease genes in A. oryzae, tppA and
pepE, facilitated an increase of 63% in the production level of human ly-
4.2. Protease-deficient strategies sozyme (Jin et al., 2007). Combination strategies of using non-protease
inducing medium and use of the aspartyl protease inhibitor pepstatin
Production, properties and classification of microbial proteases represent an alternative strategy to minimizing the impacts of proteases
have recently been reviewed (Ward., 2011; Ward et al., 2009). In on fungal recombinant protein activity (Ahamed et al., 2005). In two re-
addition to the observed variabilities in processing of fusion proteins cent proteomic studies involving A. niger, it was observed that under
by KEX-like endoproteases in Aspergillus discussed above recombi- conditions of culture starvation, resulting from depletion of carbon
nant protein-degrading fungal proteases have long been known to source, proteases were found to predominate in the secretome and
be problematic (Ward et al., 2006). Braaksma and Punt (2008) hence these conditions should be avoided to minimize protease secre-
reviewed various strategies for controlling protease activity as a tion during production of recombinant proteins (Adav et al., 2010;
means of supporting recombinant protein production. Methods in- Braaksma et al., 2010).
cluded classical selection of protease mutants, molecular genetic
methods to construct protease mutants, targeted to protease genes 4.3. Manipulations of morphology
and protease regulators, manipulation of fermentation conditions,
specifically pH, control of metabolites/catabolites such as carbon, nitro- Vegetative growth involves hyphal extension and occurs at the
gen, sulfur and phosphorus, induction of proteases and physiological hyphal tip. Branching leads to new hyphal extension units. The hy-
and morphological effects. phal tip is the principal region of protoplasmic activity, protein pro-
With enzyme-overproducing industrial strains, one approach was duction and extracellular protein secretion and hence this is the
to partially inactivate some of the more prominent extracellular pro- principal locus for biological process-related recombinant protein
teases, for example the alkaline proteases and the metallo-proteases production. Further back from the tip, protoplasmic compartments
(Christensen and Lehmbeck, 2000). Buxton and Gabor (1997) patent- become more vacuolated. It follows that a greater degree of branching
ed a sequence encoding the vacuolar PEPA aspartic protease and will increase rates of fungal growth, protein synthesis and extracellu-
methods for transforming strains to produce the protease and per- lar protein secretion. Morphology of the mycelium is strongly influ-
haps more importantly for development of Aspergillus mutants defec- enced by the surrounding environment and other factors including
tive in the production of aspartic protease. Given that filamentous inoculum size and type (vegetative, spores, etc.). On the surface of
fungi can contain as many as 80 proteolytic genes of varying known solid media, filamentous fungi grow as mycelial mats. In submerged
and unknown function, researchers are cautioned against trying to cultures, fungi may attach to suspended particles if present or grow
develop mutants deficient in multiple proteases (Machida, 2002). Im- as diffuse filamentous mycelia or as dense pellets, which may develop
pacts on recombinant protein production of constructing stable A. to different sizes. Morphological form influences rate of growth and
niger recombinants containing up to three disrupted protease genes product formation. Predominant growth and metabolism of fungi in
were characterized (Van den Hombergh et al., 1997). Specific mu- pelleted form occurs at the pellet surface where there is maximum
tants of A. nidulans, deficient in the aspartic protease gene, exhibited access to nutrients and oxygen. Inside the pellet, inward diffusion of
the ability to produce chymosin as well as other recombinant proteins nutrients and outward diffusion of product become limiting and
(Berka et al., 2003). When the alkaline protease gene of a strain of A. vacuolization and lysis are frequently observed. Recently, Driouch et
oryzae was transformed to produce heterologous endoglucanase, en- al. (2010) described a novel approach, involving use of silicate micro-
hanced production and stability of the recombinant protein was ob- particles, to engineering different morphology states in A. niger to im-
served in shake flask cultures (Lehmbeck., 2001). prove enzyme production.
Antisense RNA may be used to reduce expression of particular Because of morphological problems noted for Aspergillus species in
genes, including proteases in recombinant hosts. PEPB protein, re- fermenters which result in rheology and viscosity problems leading
cently characterized as a member of the glutamic proteases, was to mass transfer limitations, Jensen (1997) proposed use of alterna-
thought to be the causative agent in degradation of recombinant tive thermophilic fungal hosts for production of recombinant pro-
thaumatin in A. awamori containing a disrupted pepA gene producing teins. It was observed that when thermophilic fungal strains
inactive PEPA. Thaumatin production was improved by expression of including Acremonium, Corynascus, Thielavia, Myceliophthora, Thermo-
pepB antisense RNA, but results indicated antisense mRNA had only ascus and Chaetomium species, were grown in batch fermentations
partially silenced pepB gene expression. A substantial further increase under the same conditions used to culture A. oryzae, medium viscos-
in thaumatin production was achieved by disruption of the pepB gene ities observed were much lower.
(Fujinaga et al., 2004; Moralejo et al., 2002). Impact of morphology changes, as they effect recombinant protein
Disruption of some protease regulator genes has been effective in production, may be at least partially related to protease production or
substantially reducing protease activity in Aspergillus species. For ex- release. Growth of the A. niger mycelium as large pellets was associat-
ample, disruption of the prtT gene which is a regulatory gene which ed with lower specific protease activities and increased specific glu-
encodes a member of the Zn-binuclear cluster family appears to elim- coamylase activities were found when A. niger was cultured in
inate two Aspergillus proteases from the medium including PEPA and media which generated large pellets (Papagianni and Young, 2002).
reduces total protease activity by 80% (Punt et al., 2008). Yoon et al. In general, fungal pelleted growth mediates greater lysis in fungi,
(2011) reported on experiments which demonstrated how successive for example, in Aspergillus species, and this results in the presence
disruption of ten protease genes in A. oryzae was effective in enhanc- of higher levels of proteolytic activity in filtrates of pelleted cultures,
ing heterologous production of human lysozyme and bovine chymo- as compared to filamentous growth (Ahamed et al., 2005). While the
sin production. greater proteolytic activity in pellet cultures is likely to be partly due
Manipulation of fungal culture pH away from the optimal pH for to intrapellet cell lysis, differential expression may also be a factor.
activity and implementation of cultivation strategies which prevent Dai et al. (2004) has reported that one of seven genes that were dif-
release of intracellular proteases via mycelial cell lysis have been shown ferentially expressed in A. niger pellets encoded a pepsin-type prote-
to reduce proteolysis of secreted recombinant proteases (Denison, ase. pH could be manipulated to cause morphological mutant
2000; O'Donnell et al., 2001; Wang et al., 2005). Use of peptide-rich formation and recombinant glucoamylase production in A niger
1130 O.P. Ward / Biotechnology Advances 30 (2012) 1119–1139

(Swift et al., 1998). In studies of an A. niger strain containing a gene non-glycosylated recombinant proteins, assuming for a particular
for the Gla-GFP fusion protein, protease activity in pelleted growth product that problems related to recombinant protein degradation by
was only one-third of that observed with filamentous growth (Xu et proteases of the host producing strain have been resolved. It is also
al., 2000). clear that where the interest relates to recombinant glycosylated pro-
Morphology of the filamentous fungus, Chrysosporium lucknowense, teins, especially recombinant proteins for human therapeutic use, the
has been manipulated genetically to produce a non-filamentous form, quite different native protein glycosylation patterns observed in fungi
thereby reducing viscosity of culture systems. The non-filamentous render them unsuitable hosts for recombinant human glycoprotein
form also exhibits low protease activity and is capable of producing production, and some other hosts, for example Pichia species, may be
very high yields of heterologous proteins (Verdoes et al., 2007). more suitable microbial hosts to be engineered for mammalian glyco-
General aspects of the relationships between morphology and protein production. Nevertheless, research is continuing to be aimed
productivity in filamentous fungi have been reviewed by Grimm et at engineering the appropriate human glycosylation machinery into fil-
al. (2005). amentous fungi.

4.4. Solid-state fermentation approaches 5. Recombinant proteins in metabolic engineering of fungi

While the mechanisms have not been fully elucidated, variations In recombinant protein production, the initial priority targets
between solid-state and submerged culture fermentation conditions were individual proteins and glycoproteins for beneficial industrial,
can alter the expression of genes and rates of enzyme production agricultural, food or pharmaceutical use. However, the development
(Iwashita, 2002). During growth of A. oryzae on solid substrates, re- of genomic and proteomic capabilities combined with molecular
gions of differentiation of filamentous mycelia exist, from aerial my- tools provides the tools to manipulate hosts with respect to more
celium to medium-associated mycelium, and the different regions of complex combinations of proteins, with diverse functions, be they
filamentous growth mediate differential levels of gene expression catalytic, regulatory or with other physiological roles. As other
and product formation (Masai et al., 2005). In some cases, the con- organisms, the filamentous fungi offer potential opportunities for im-
trasting activities reflect whether the activity is cell wall-associated proving desired metabolite product yields, reducing or eliminating
or excreted into the culture medium: in a case in point in submerged undesired metabolite bi-products, extending substrate ranges and in-
cultures, some enzyme activities were associated with the cell wall, troducing new enzymes or pathways leading to production of new
whereas in solid-state cultures, they were secreted into the medium products (Kern et al., 2007). For example, hosts may be transformed
(Wang et al., 2005). In solid state culture, wheat bran media caused to produce the multiple enzymes associated as part or all of a new
lowest expression of catabolic genes, which were thought to have re- metabolite biosynthesis sequence. Alternatively, an existing pathway
leased catabolite repression of synthesis of hydrolytic enzymes, may be engineered to address bottlenecks and maximize rate of me-
thereby mediating rich hydrolytic enzyme gene expression. Oda et tabolite biosynthesis or indeed to alter specificity of one or more reac-
al. (2006) concluded that certain enzymes were selectively secreted tions to facilitate expansion of product range. Approaches invariably
under conditions of solid-state culture or in submerged culture, inde- involve genetic modifications, including expression of recombinant
pendent of the composition of the medium. In a different but a partly enzymes and manipulation of host transcription and/or translation
analogous situation, proteome differences were observed when F. machinery. The opportunities to exploit filamentous fungi for produc-
graminearum was cultured in vitro and in planta, with some proteins tion of a vast range of new products, most of which are not yet con-
being expressed in only one of the two conditions (Paper et al., 2007). ceived, is enormous and derives from the diverse nature of fungal
Imanaka et al. (2010) compared cultivation of A. oryzae, using species. With an estimated 1.5 million species (Hawksworth, 2001),
three different culture methods, i.e., shake-flasks, agar-plate and only a small number of which has been sampled by genomic and pro-
membrane surface liquid culture, and observed differences in growth, teomic methods, and with a likely average of 10,000 genes per spe-
secretion of proteases and alpha-amylase, secreted protein level and cies, the resource is almost unlimited. Some of these concepts, as
gene transcriptional profile by DNA microarray analysis. Protease ac- they apply to primary and secondary metabolites, are briefly dis-
tivities, especially oryzin (alkaline protease) and alpha-amylase, were cussed in this section through use of selected examples.
much higher in agar-plate and membrane surface liquid culture than
in shake flasks. Transcriptional gene profiles from the agar-plate and
membrane surface liquid culture manifested somewhat similar pat- 5.1. Primary metabolites
terns but were quite different from the shake flask profiles.
In some cases, solid state culture promoted production of a more 5.1.1. Citric acid
homogeneous glycosylated recombinant product. Submerged cul- With respect to primary metabolites, A niger has been investigated
tures by A. oryzae transformed to produce the important heterologous as a target model for metabolic engineering of filamentous fungi, es-
antigenic protein Pre–S2 of human hepatitis B virus, resulted in pro- pecially with respect to citric acid production with a view to increas-
duction of a partially degraded heterologously glycosylated protein, ing metabolic fluxes through the glycolytic pathway leading to citrate
whereas in solid-state culture a homogeneous glycosylated form of production (Ruijter et al., 2002). Seven or more enzymes were select-
the whole fusion protein was obtained (Maruyama et al., 2000). Use ed as principal targets and two genes encoding regulatory enzymes in
of wheat bran (2%) in solid state culture mediated a 500-fold increase the pathway were overexpressed (Ruijter et al., 1997; Torres et al.,
in production of recombinant chymosin as compared with submerged 1996).
culture (Tsuchiya et al., 1994). te Biesebeke et al. (2002) found that
pelleted growth in submerged fermentations and semi-solid fermen- 5.1.2. Biodiesel
tations showed different patterns of gene expression and protein Biodiesel, originating from some form of biological host, arguably
secretion. represents part of the “biosolution” to the world's energy problems
(Demain, 2009). The products of fungi and algae are currently being
4.5. Concluding comments used as beneficial food and feed additives and nutritional products.
While it is still unclear if fungal and algal strains are appropriate for
Because of their extraordinarily high productivities and simple production of microbial oils for biodiesel production, they do contain
growth requirements, filamentous fungi can compete or outcompete the machinery for bio-oil biosynthesis and the tools for transforming
most other recombinant hosts in processes for production of their biosynthetic enzymes to other hosts, which may be deemed
O.P. Ward / Biotechnology Advances 30 (2012) 1119–1139 1131

more suitable for energy production, are well on the way to being including filamentous fungi, contain complex peptide synthetase
established. templates which mediate synthesis of a variety of bioactive peptides,
Several filamentous fungi accumulate very high amounts of micro- more than 300 of which are known, without ribosomal involvement
bial lipids, especially highly unsaturated lipids (Ward and Singh, (Stachelhaus et al., 1996).The bioactive products include antibiotics,
2005). The concept of using filamentous fungi as producers as lipids toxins, enzyme inhibitors, antiviral agents, antitumor agents and im-
was introduced during World War 1 in Germany and processes munosuppressants. These non-ribosomal large protein templates
using a variety of strains including Mortierella isabellina, Mortierella contain protein domains which catalyze synthesis of a wide range of
alpina and Mucor vinacea have been scaled up to produce commercial low molecular weight linear, branched and cyclic peptides which
quantities of lipids (Ratledge, 1978). While the primary emphasis was may be further transformed by acylation, glycosylation and other re-
on the nutritional benefit of unsaturated fatty acids, scientists in the actions (Marahiel, 1992). Research is ongoing related to manipulation
Soviet Union considered the Mucorales as potential candidate hosts of the participating genes encoding synthetases with altered sub-
for production of lipids for use in biofuels (Feofilova et al., 2010). If strate specificities, using gene disruption and reconstitution strate-
momentum in this area continues, strategies will be needed for opti- gies, with a view to determining the potential to engineer synthesis
mization of processes which will include detailed examination of the of novel peptides, including novel antibiotics and other therapeutics
lipid synthetic pathways, in order to determine metabolic bottle- (Kleinkauf and von Döhren, 1990; Stachelhaus and Marahiel, 1995).
necks. In addition, sequencing and annotation of the genes encoding
the participating enzymes will provide opportunities for transforma- 5.2.2. Antibiotics
tion of hosts to produce recombinant enzymes. These biochemical Biochemistry of non-ribosomal peptide synthesis has been inves-
and molecular approaches may be combined with fermentation opti- tigated for many years as it relates, for example, to production of pep-
misation strategies, both to optimize production rates and yields and tide antibiotics, including beta-lactams by the filamentous fungi P.
to produce products of varying lipid compositions. chrysogenum and A. nidulans and production of cyclosporin A by Toly-
pocladium niveum (Weber et al., 1994; Aharonowitz et al., 1993;
5.1.3. Higher unsaturated fatty acids (HUFAs) Smith et al., 1990; Gutierrez et al., 1991).
M. alpina produces the C-20 highly unsaturated fatty acids Numerous genes involved in biosynthesis of antibiotics and other
dihomo-gamma-linolenic acid, arachidonic acid and eicosapentaenoic secondary metabolites have been cloned, including genes involved in
and has been used for industrial production of arachidonic acid the synthesis of β-lactam antibiotics by the filamentous fungi, P.
(Shimizu et al., 1997). In mammals, γ-linolenic acid (GLA) is an inter- chrysogenum, Cephalosporium acremonium and A. nidulans (Martín and
mediate of the (n−6) pathway conversion of linolenic acid to arachidonic Liras, 1989). As with other antibiotics, genes involved in penicillin,
acid and a direct precursor of dihomo-gamma-linolenic acid. Certain cephalosporin and cephamycin biosynthesis are organized in clusters
condi tions (aging, stress, some infections, diabetes, eczema) may be and pathway-specific regulatory genes positively or negatively modu-
manifested as causing a deficiency of Δ6-desaturation and of GLA, and late genes expression in these clusters and hence regulate antibiotic
hence there is considerable interest in investigating production of re- biosynthesis (Loder and Abraham, 1971; Martín and Liras, 1989;
combinant forms of Δ6-desaturase and the efficient enzyme from M. Nuesch et al., 1987). Positive regulatory elements also impact the inter-
alpina as a target of interest. GLA is further metabolized to produce related processes of secondary metabolism and differentiation in these
anti-inflammatory eicosanoids, such as prostaglandins and leukotrienes, organisms. Clustering of biosynthetic enzymes is facilitating research
and GLA and its metabolites also regulate expression levels of various into the molecular mechanisms which mediate expression of genes in-
gene products. As a first step, this enzyme was cloned from M. alpina volved in antibiotic synthesis, which will enable gene expression to be
and expressed in A. oryzae under the control of the amyB promoter. positively regulated and optimized to systematically maximize antibiot-
While GLA was not produced by the control A. oryzae strain, GLA ic formation in industrial fermentation processes (Martin, 1992).
constituted 25.2% of total fatty acids in the recombinant host Routes to production of penicillin G, cephalosporins and cephamycins
transformed to express Δ6-desaturase (Sakuradani et al., 1999). share a common early pathway to synthesis of the isopenicillin-N inter-
Similar enzymes have now been cloned from a range of filamentous mediate. The ‘natural’ cephalosporins produced by A. chrysogenum have
fungi including Cunninghamella echinulata, R. arrhizus, Rhizopus nigricans, relatively low clinical efficacy and are produced semi-synthetically from
Rhizopus stolonifer, Mucor rouxii and Pythium irregulare. The gene encod- intermediate building blocks, 7-aminodeacetoxycephalosporanic acid
ing the enzyme from R. stolonifer was recently expressed in Pichia (7-ADCA) and 7-aminocephalosporanic acid (7-ACA). While P.
pastoris, enabling the transformed Pichia strain to produce about 22.4% chrysogenum cannot produce cephalosporins and cephamycins, it is an
of its fatty acid content as GLA (Wan et al., 2011). attractive potential recombinant host because of its hyper capacity to
produce penicillin, via the common precursor isopenicillin N and in-
5.2. Secondary metabolites deed the host was successfully engineered to synthesize cephalosporins
and 7-ADCA (Crawford et al., 1995; Robin et al., 2001; 2003a,b; van de
Some of the recent exciting developments in fungal secondary Sandt and de Vroom, 2000). The principal transformations of P.
metabolite production relate to the continuing developments in our chrysogenum involved introduction of the gene cefE from Streptomyces
knowledge of mechanisms for production of non-ribosomal peptides clavuligerus, encoding production of the expandase enzyme deacetoxy-
(NRPs) and polyketides (PKSs). While the A. nidulans genome indi- cephalosporin-C synthase, and the genes cefEF and cefG from P.
cates the presence of 26 PKSs and twenty four NRPSs, only six chrysogenum, encoding the dual expandase/hydroxylase and acetyl-
PKSs (synthesizing monodictyphenone, asperfuranone, orsellinic transferas, respectively. Other recombinant proteins, for example the
acid/F9775, asperthicin, napthopyrone, sterigmatocystin) and four cmcH gene from S. clavuligerus, encoding deacetylcephalosporin
NRPSs responsible for the biosynthesis of terrequinone, aspyridones, O-carbamoyltransferase, have been successfully expressed in P.
emericellamide and penicillin, are reported to have been character- chrysogenum with a view to producing other novel cephem precursors
ized in A. nidulans (Balibar et al., 2007; Bergmann et al., 2007; Chiang (Harris et al., 2009). This work demonstrates how metabolic engineer-
et al., 2010; Evans et al., 2011; Sanchez et al., 2009; Szewczyk et al., ing strategies involving production of recombinant enzymes into strains
2008; von Dohren, 2009). already improved through classical mutation programs can lead to de-
velopment of multipurpose host platforms for production of non-native
5.2.1. Engineered biosynthesis of novel peptides secondary metabolites.
In addition to the predominant and ubiquitous ribosomal machin- In related work, two dimensional electrophoresis-based proteo-
ery for synthesis of peptides and proteins, certain organisms, mics was used to compare protein maps, with up to 950 proteins
1132 O.P. Ward / Biotechnology Advances 30 (2012) 1119–1139

being identified, from a three P. chrysogenum strains, a high and low oils in plants. A bifunctional Δ12- and Δ15-desaturase from Fusarium
penicillin producer and a wild-type strain (Jami et al., 2010). The cys- moniliforme, co-expressed in both soybean and yeast (Yersinia
teine biosynthesis, pentose phosphate and some stress response pro- lipolytica) with the primary LC-PUFA biosynthetic enzymes, enhanced
teins predominated in the high penicillin producing strain while levels of eicosapentaenoic acid (EPA) production (Damude et al.,
pathways for synthesis of other metabolites were reduced. 2006). Enzymes with similar biofunctional properties and potential
Peroxisomes participate in important functional aspects of second- are produced by Claviceps purpurea (Meesapyodsuk et al., 2007) and
ary metabolite synthesis in filamentous fungi, including the final steps Coprinus cinereus (Zhang et al., 2007). A recent review on metabolic
of penicillin synthesis, mediated by isopenicillin N-acetyltransferase engineering of oil-seed crops to produce omega-3 long chained fatty
(Muller et al., 1991;1992). acids shows where recombinant HUFA biosynthetic enzymes from fil-
amentous fungi play their part (Venegas-Caleron et al., 2010).
5.2.3. Taxol Plants and filamentous fungi such as Ascomycota, have different
The tetracyclic diterpene lactam, taxol, is a low toxicity broad pivotal amino acid linked processes for ammonium assimilation. In
spectrum anticancer drug with beneficial application against breast, filamentous fungi, NADP(H)-dependant glutamate dehydrogenase
uterine and other cancers (Wani et al., 1971). Taxol is present natu- (GDH) and glutamine synthetase participate in ammonium assimila-
rally at low concentrations of 0.01–0.05% in the bark, roots and tion whereas in plants glutamine synthetase alone is primarily re-
branches of the western yew, Taxus brevifolia, an endangered species. sponsible for assimilation. When the gene encoding GDH, gdhA,
In the past few years, significant progress has been made in our un- from A. niger was expressed in the cytoplasm of rice plants, positive
derstanding of the general biology and taxol biosynthetic pathway changes in metabolism were observed which lead to enhanced am-
of taxol-producing endophytic fungi, with a view to producing taxol monium assimilation and better plant growth (Abiko et al., 2010).
by microbial fermentation (Lin et al., 2003; Zhou et al., 2008, 2010). Hydrophobins, surface-active proteins obtained from filamentous
Some of the 20 or so genes encoding enzymes of the pathway have fungi, have significant potential applications in genetically-
been sequenced and some genetic transformation systems have engineered plants and hydrophobin-fusion technology is predicted
been established which included development of some fungal promo- to become a very useful tool in plant biotechnology. By way of exam-
ter-containing expression cassettes, ultimately directed at developing ple, when the hydrophobin HFBI sequence from T. reesei was fused to
efficient taxol-producing recombinant fungal hosts (Guo et al., 2006; green fluorescent protein (GFP) and transiently expressed in
Wang et al., 2007a,b,c). Nicotiana benthamiana plants mediated by A. tumefaciens, the fusion
enhanced GFP accumulation (Joensuu et al., 2010). The fusion protein
6. Expressing fungal multi-protein complexes or pathways in constituted more than 50% of total soluble protein and delayed leaf
other hosts necrosis.

While much effort has been fruitfully invested in transforming 6.2. Expression of fungal systems in bacteria and yeast
hosts to produce individual recombinant proteins and to optimizing
recombinant product formation, the evolution in our understanding 6.2.1. Polyketide biosynthetic machinery
of these systems combined with newly acquired knowledge of geno- Polyketides result from biosyntheses involving polymerization of
mics and proteomics is facilitating more advanced engineering of acetyl and propionyl subunits in a manner analogous to fatty acid
hosts with whole new pathways or functions. The huge diversity of synthesis. Polyketide products, generated by different PKS types, in-
fungal species makes fungi a rich reservoir for mining novel biosyn- clude erythromycin, tacrolimus, epothilone, tetracycline, tetraceno-
thetic capabilities and clearly the native fungal hosts of new path- mycin, daunorubicin, 6-methyl salicylic acid, aflatoxin B1 and
ways, deemed to be of value, may not be suitable hosts for optimal lovastatin (Crawford et al., 2006; Fujii, 2010; Kennedy et al., 1999;
expression of the pathway, or synthesis and exploitation of the de- Moriguchi et al., 2010; Shen, 2003; Watanabe et al., 1996). Fungal
sired metabolic products. In this section, a small number of fungal polyketide synthases (PKSs) utilize a variety of acyl-CoA substrates
complex systems expressed in non fungal hosts will be briefly dis- to catalyze carbon to carbon bond forming reactions to synthesize
cussed. It is hoped that the examples point to the great potential to the well known complex fungal polyketides which exhibit a range
exploit fungal diversity in part through transfer of fungal machinery of bioactivities. Many gene clusters, thought to encode the enzymes
to other hosts. responsible for synthesizing polyketides such as bikaverin, zearale-
none and hypothemycin, including genes encoding PKSs, have been
6.1. Expression of fungal systems in plants characterized. Recent reports indicate that these recombinant fungal
enzymes have been expressed in E. coli rendering the bacterial host
There is considerable interest in metabolic engineering of fatty capable of production of the corresponding secondary metabolite
acids into plants to produce new oil seed crops. Since filamentous (Saruwatari et al., 2011). The aromatic polyketide, 6-methylsalicylic
fungi are among the best producers of highly unsaturated fatty acids acid, is synthesized by ATX, encoding gene atX, present in A. terreus
and since higher plants do not produce these fatty acid, there is inter- and Penicillium patulum, and this gene has successfully been
est in transforming plants with some of these key highly unsaturated expressed in E. coli, S. cerevisiae and Streptomyces species and an en-
fatty acids (HUFA) biosynthetic enzymes, with M. alpina being recog- hanced production of 6-methylsalicylic acid was noted in the case of
nized as one of the most useful gene sources (Drexler et al., 2003; S. cerevisiae (Bedford et al., 1995; Kealey et al., 1998). The huge fungal
Huang et al., 1999; Knutzon et al., 1998; Michaelson et al., 1998). Ini- gene responsible for bikaverin (aromatic polyketide anticancer drug)
tial strategies involve enhancing the very long chain fatty acid con- biosynthesis, PKS4 originating in Giberella fujikuroi, was successfully
tent of current plant oils. Some of the early promising steps expressed in E. coli with production of the polyketide (Ma et al.,
involved seed-specific co-expression of an oleate delta12 desaturase 2007). Ma et al. (2009) also reported that the lovastatin nonaketide
and a Δ6 desaturase, both from Mortierella species, in canola (Liu et synthase (LovB) from A. terreus, involved in synthesis of the choles-
al., 2001). The fifth generation of field trials contained 44% γ-linolenic terol lowering drug, lovastatin, could be efficiently expressed by a re-
acid (GLA) not present in rapeseed. Similar high proportions of GLA combinant S. cerevisiae host, research work which is contributing
were accumulated when a Δ6 desaturase from P. irregulare was substantially to determining the complex set of functions and struc-
expressed in Brassica juncea (Hong et al., 2002). Some other fungal tures of the polyketide synthases. Mixing of domains of different
long chain polyunsaturated fatty acid (LC-PUFA)-synthesizing en- PKS genes, including domains originating from fungi and bacteria,
zymes are also being used as part of the strategy to produce HUFA generated different combinations of biosynthetic enzymes and
O.P. Ward / Biotechnology Advances 30 (2012) 1119–1139 1133

demonstrated the potential to vary the synthezing machinery to pro- transformed from gametocytes into infective sporozoites. Sporozoite
duce wholly novel metabolites not produced naturally by the native count reductions mediated by the expressed SM1 and scorpine were
strains (Saruwatari et al., 2011; Zhang et al., 2008). 71 and 90%, respectively. The SMI peptide binds to the surface of the
salivary gland of Anopheles gambiae, the principal malaria vector,
6.2.2. Penicillin biosynthetic machinery and blocks entry of the parasite into the mosquito vector. The scorpion
While current industrial production of penicillin is mediated by P. antimicrobial scorpine, is a hybrid of two toxins, a cecropin and a
chrysogenum, the intrinsic growth limitations of fungi in large fermen- defensin which is two orders of magnitude more toxic against Plasmo-
ters are recognized and there is interest in developing new hosts, for dium. A recombinant M. anisopliae expressing an [SM1]sub8:scorpine
example yeasts, for production of new beta lactams and perhaps fusion protein, reduced sporozite counts by 98% (Fang et al., 2011).
other bioactive compounds such as peptides. An industrial precedent The infective action of the filamentous fungus, B. bassiana, towards
exists for changing from a filamentous fungal host to a yeast host in insects, via their cuticle, is generally a slow process. However, lepi-
the case of citric acid production when the Pfizer Company modified dopteran pests are rapidly killed by ingestion of the Vip3A insecticidal
their process by introducing a Candida guilliermondii yeast strain in proteins produced by Bacillus thuriengensis. A recombinant strain of B.
place of the conventional Aspergillus producer, thereby increasing cit- bassiana, which highly expressed one of the Vip3A toxins, Vip3Aa1 in
rate production rate (Ward, 1989; 1991). In P. chrysogenum, production its conidial cytoplasm, when sprayed on cabbage leaves was shown to
of penicillin is compartmentalized in peroxisomes and the cytosol. The dramatically reduce larval numbers feeding on the cabbage leaves (by
yeast Hansenula polymorpha has a number of features that make it po- 26.2 fold) (Qin et al., 2010). The mid gut of the infected larvae con-
tentially an attractive host as an alternative producer of beta-lactams, tained a digested form of the toxin and the larvae exhibited the
including the availability of some very strong and regulatable pro- expected symptoms of toxin action, namely shrinkage and palsy.
moters. Furthermore, penicillin production in Penicillium is significant- Methods have also been established whereby larvae of organisms
ly mediated by peroxisomal enzymes and it is known that peroxisomes such as the silk worm have been directly transformed to produce re-
of H. polymorpha may be hyperinduced. Gidijala et al.(2009) success- combinant fungal proteins. For example, the 1320-bp endoglucanase
fully engineering H. polymorpha to produce and efficiently secrete 1 gene from T. viride was cloned and expressed in silkworm larvae
penicillin: a) by introducing the P. chrysogenum gene encoding and in a silkworm cell line using a mutant baculovirus expression sys-
the non-ribosomal peptide synthase (NRPS) δ-(L-aminoadipyl)- tem (Li et al., 2010).
L-cysteinyl-D-valine synthetase (ACVS) into H. polymorpha resulting
in production of active ACVS enzyme; b) co-expression of the B. subtilis 7.2. Plant pathogens
stp gene encoding the enzyme phosphopantetheinyl transferase which
activates ACVS; and c) co-expression of P. chrysogenum genes encoding Molecular methods are also being used to develop a better under-
cytosolic isopenicillin N synthase and the peroxisomal enzymes, isope- standing of the mechanisms related to fungal pathogenicity of plants.
nicillin N acyl transferase (IAT) and phenylacetyl CoA ligase (PCL). Re- In addition to having an important role in secondary metabolite pro-
combinant fungal ACVS has also been expressed in baker’s yeast duction, as indicated above, peroxisomes are also involved in filamen-
(Siewers et al., 2009). tous fungal pathogenicity, for example, of cucumber by Colletotrichum
orbiculare and the rice blast pathogen, Magnaporthe oryzae (Bhambra
7. Engineering of pathogenic fungi et al., 2006; Wang et al., 2007d) and in production of host selective
AK-protein toxins by Alternaria alternata which mediate its pathoge-
A very large number of filamentous fungi are pathogenic to other nicity to multiple plant species, including Japanese pear (Imanaka et
organisms, including animals and humans, plants and insects and, in al., 2010). Imazaka et al. (2010) confirmed the peroxisome localiza-
the case of animals and plants, the resulting diseases have vast nega- tion of AK-toxins by developing and expressing recombinant
tive economic and social impacts. Molecular techniques, including GFP-tagged AK-protein toxins in A. alternata and then used mutant
strategies involving production of recombinant proteins may be recombinant A. alternata strains, with AK-toxin disruptions, to con-
used to facilitate elucidation of the causes of pathogenesis and to pro- firm the role of AK-toxins in pathogenicity.
vide direction on solutions on pathogenesis problems. The following It was noted earlier (3.4.1) that the genome of the human R.
brief examples are offered to illustrate potential opportunities. oryzae pathogen contained expanded gene families of encoding se-
creted proteases and that these proteases are involved in the infective
7.1. Entomopathogens process and are characteristic of the virulence of Rhizopus pathogens
(Ma et al., 2009). It was also noted in the same section that R. miehei,
Recombinant fungal technology offers potential to create recombi- which causes fungal infections in humans and animals, can resist
nant hosts which interact with other organisms. Examples would in- higher concentrations of statins than is observed in Mucor
clude transformed fungi which produce molecular species such as circinelloides, where statins inhibit HMG-CoA and ultimately lead to
herbicides, insecticides or biological inhibitors against undesired tar- Mucor cell death. A recombinant M. circinelloides strain expressing
gets. Recently published examples include a recombinant fungus the gene hmgR encoding the HMG-CoA of R. miehei, was rendered
which kills human malaria parasites in mosquitoes, mediated by se- more tolerant to statins. Clearly, our knowledge of fungal pathogene-
creted recombinant toxic peptides and enhancement of infectivity of sis will greatly expand over the next few years as the genomes of
a recombinant entomopathogenic Beauveria bassiana towards larvae more plant pathogens are sequenced and their gene functions are
of the oriental leafworm moth. annotated.
While there have been effective chemical pesticides against malar-
ia and its mosquito carrier, especially pyrethroids, the progress to- 8. Engineering of biocontrol fungi
wards eradication has been hampered by the development of
pyrethroid-resistant mosquitoes. Several fungi, including Metarhizium Filamentous fungi can also have beneficial roles in plant disease
anisopliae, are pathogenic to adult mosquitoes but the infective pro- management as will be illustrated here by focusing on Trichoderma
cess through contact with the cuticle is slow (12–14 days). Recombi- species. Many strains of Trichoderma are highly competent in coloniz-
nant strains of M. anisopliae expressing the antimicrobial toxin, ing plant root surfaces and thrive and proliferate in association with
scorpine, or a salivary gland and mid-gut peptide 1 (SM1) reduced healthy root surfaces (Sriram and Ray, 2005). Many other fungi lack
entry and/or development of Plasmodium falciparum, the parasitic this ability and Trichoderma species have evolved a variety of mecha-
causative agent of malaria, into the Anopheles gut where it gets nisms to attack and parasitize these other fungi and benefit from
1134 O.P. Ward / Biotechnology Advances 30 (2012) 1119–1139

nutrients from these other fungi as they support and enhance plant biosynthetic pathway related to the production of current or new pri-
growth as biocontrol agents (Harman et al., 2004). Biocontrol agents mary or secondary metabolites. While this activity is still at an early
utilize several antagonistic strategies against pathogens and all of stage, it will clearly gain momentum and will become a more impor-
these mechanisms are exploited by Trichoderma (Dennis and tant pursuit in the field of industrial microbiology in the years ahead.
Webster, 1971a,b,c). These mechanisms include: competition by lim- It has also been recognized that fungi represent an enormously
iting access to the pathogen's nutritional needs; production of diverse range of organisms and hence a huge resource for novel bio-
inhibitory metabolites or other toxins including trichothecin, sesqui- synthetic systems, especially since the vast majority of fungal species
terpene and certain enzymes; and parasitism. In the case of T. harzia- on the planet have yet to be cultured, identified and characterized. It
num, it was noted that combined effect of production of an antibiotic is clear that many of the fungi naturally harboring novel biosynthetic
and production of hydrolytic enzymes had a synergistic effect on the machinery will likely not be the best hosts for large-scale exploitation
pathogens (Schirmböck et al., 1994). Enzymes thought beneficial to of these capabilities. It is expected that in many cases, the target bio-
Trichoderma species in attacking pathogens such as Fusarium oxy- synthetic abilities will be better utilized by transformation into other
sporum and Botrytis cinerea include the lytic enzymes, chitinase, fungal hosts, perhaps also to other microbial hosts, and in some cases,
beta-1,3-glucanases, and protease (Cherif and Benhamou, 1990; to non-microbial hosts, for example, to plants.
Elad and Kapat, 1999; Geremia et al., 1993; Tronsmo et al., 1993). One of the most exciting new aspects of the study of recombinant
When a recombinant chitinase from a soil bacterium Serratia proteins expression in fungi is the realization that the future applica-
marcescens used to control Sclerotium rolfsii was introduced into T. tions will not be confined to the biomanufacturing capacities of these
harzianum under control of a constitutive promoter, the constitutive organisms. Some of the natural beneficial impacts and especially the
production of chitinase by the Trichoderma biocontrol agent enabled deleterious impacts of fungi relate to their extremely complex inter-
it to overgrow the S. rolfsii pathogen (Haran et al., 1993). An alkaline actions with other organisms, for example through mutualism, com-
protease producing transformant of T. harzianum was thought to en- mensalisms and especially pathogenesis. About 300 fungi are
hance its mycoparasitic activity (Sriram and Ray, 2005). known to cause diseases in humans (Monk and Goffeau, 2008) and
Of course, another approach to protecting plants against fungal many of these organisms exhibit multidrug resistance mediated by
plant pathogens is to directly arm the plants with the requisite biode- multidrug efflux pumps (Balzi et al., 1994). A myriad of fungal patho-
fense machinery and indeed several genes encoding the Trichoderma gens attack plants and a quantum shift is currently taking place in the
enzymes mediating Trichoderma's parasitic role have been expressed advancement of our knowledge of the plant–microbe interactions in-
in plants, rendering them more resistant to fungal plant pathogens volved in these diseases (Boller and He, 2009). The underlying causa-
(Bolar et al., 2000; Lorito et al., 1998; St Leger et al., 1996). tive mechanisms involved in these attacks are now being
characterized using modern genomic and molecular strategies. Put
9. Conclusions simply, the economic and social costs associated with the impacts of
fungal pathogenesis are enormous. The opportunities to exploit re-
The principal focus over the past 25 years of recombinant protein combinant technology to unravel the causes and then bring forward
research efforts as applied to filamentous fungi related to the evalua- solutions will represent a very important future focus of recombinant
tion and development of the workhorse fungi, especially the industri- protein technology as it relates to fungi.
al extracellular enzyme-producing fungi, as hosts for recombinant
protein production. The special advantages of these organisms, name-
ly their abilities to grow at high rates and to high densities in com- References
mercial fermenters and their natural abilities to secrete high levels
Abianova AR, Chulkin AM, Vavilova EA, Federova TV, Loginov DS, Koroleva OV, Benevo-
of homologous enzymes, are widely recognized. In exploiting these lenskii SV. A heterologous production of the Trametes hirsuta laccase in the fungus
advantages for production of recombinant proteins, researchers Penicillium canescens. Prikl Biokhim Mikrobiol 2010;46:342–7.
have generally used the most effective regulatory, expression and se- Abiko T, Wakayama M, Kawakami A, Obara M, Kisaka H, Miwa T, et al. Changes in ni-
trogen assimilation, metabolism and growth in transgenic rice plants expressing
cretion machinery identified in these organisms and hooked it up to a fungal NADP(H)-dependent glutamate dehydrogenase (gdhA). Planta
the genes encoding the desired recombinant protein, an approach 2010;232:299–311.
which has met with considerable commercial success. Adav SS, Li AA, Manavalan A, Punt P, Sze SK. Quantitative iTRAQ secretome analysis of
Aspergillus niger reveals nivel hydrolytic enzymes. J Proteome Res 2010;9:3932–40.
Filamentous fungi also have several disadvantages as hosts for Ahamed A, Singh A, Ward OP. Culture-based strategies for minimization of protease ac-
heterologous protein production, perhaps the most serious being tivity in filtrates from Aspergillus niger. World J Microbiol Biotechnol 2005;21:
their abilities also to produce and secrete homologous proteases 1577–83.
Aharonowitz Y, Bergmeyer J, Cantoral JM, Cohen G, Demain AL, Fink U, et al. δ-(L
which may degrade the desired recombinant product. In addition to
-α-Aminoadipyl)-L-cysteinyl-D-valine synthetase, the multienzyme integrating
the secreted proteases, filamentous fungi may be subject to different the four primary reactions in β-lactam biosynthesis, as a model peptide synthe-
degrees of lysis during the fermentation process, the extent of tase. Biotechnology 1993;11:807–10.
which will vary with other aspects of fungal physiology including Akao T, Gomi K, Goto K, Okazaki N, Akita O. Subtractive cloning of cDNA from Aspergil-
lus oryzae differentially regulated between solid-state and liquid (submerged) cul-
morphology, such that intracellular proteases may be released ture. Curr Genet 2002;41:275–81.
which can attack the recombinant protein. In addition, there are sig- Akin AR, Bodie, EA, Burrow, S, Dunn-Coleman N, Turner G, Ward M. Regulatable
nificant differences in the mechanisms of protein glycosylation ob- growth of filamentous fungi. United States Patent Application No. 20030045697.
2003.
served in fungi and mammalian cells. While considerable research is Allgaier S, Taylor RD, Brudnaya Y, Jacobson DJ, Cambareri E, Stuart WD. Vaccine pro-
being invested in modifying the glycosylation machinery in fungi to duction in Neurospora crassa. Biologicals 2009;37:128–32.
use them as hosts for production hosts of more human-like glycopro- Archer DB, Dyer PS. From genomics to post-genomics in Aspergillus. Curr Opin Micro-
biol 2004;7:499–504.
teins, it is widely accepted that yeasts such as Pichia, may be better Archer DB, Jeens DJ, MacKenzie DA, Brightwell G, Lambert N, Lorne G, et al. Hen egg
candidate hosts for production of recombinant human therapeutic white lysozyme expressed and secreted from A. niger is correctly processed and
glycoproteins. folded. Biotechnology 1990;8:741–5.
Aro N, Pakula T, Penttila M. Transcriptional regulation of plant cell wall degradation by
The vast amounts of scientific information being generated filamentous fungi. FEMS Microbiol Rev 2005;29:719–39.
through advances in genomics and proteomics are expanding the op- Balibar CJ, Howard-Jones AR, Walsh CT. Terrequinone A biosynthesis through
portunities for new applications for recombinant filamentous fungi. It l-tryptophan oxidation, dimerization and bisprenylation. Nat Chem Biol 2007;3:
584–92.
is facilitating a shift from simply inserting a gene encoding a single
Balzi E, Wang S, Leterme S, Van Dyck L, Goffeau A. PDR5, a novel yeast multidrug con-
protein product to inserting machinery encoding multiple genes, for ferring transporter controlled by a transcription regulator PDR1. J Biol Chem
example encoding metabolic enzymes for a new or improved 1994;269:2206–14.
O.P. Ward / Biotechnology Advances 30 (2012) 1119–1139 1135

Barredo Fuente JL, Rodriguez Saiz M, Collados De La Vieja AJ, Moreno Valle MA, Salto Conesa A, Punt PJ, Luijk N, van den Hondel CAMJJ. The secretion pathway in filamen-
Maldonado F, Diez Garcia B. Promoters of the genes glutamate dehydrogenase tous fungi: a biotechnological view. Fungal Genet Biol 2001;33:155–71.
beta-N-acetylhexosaminidase and γ-actin and their use in filamentous fungi ex- Conesa A, van den Hondel CAMJJ, Punt PJ. Studies on the production of fungal peroxi-
pression, secretion and antisense systems. United States Patent 6,300,095. 2001. dases in Aspergillus niger. Appl Environ Microbiol 2000;66:3016–23.
Bautista LF, Aleksenko A, Hentzer M, Santerre-Henriksen A, Nielsen J. Antisense silenc- Conesa A. Overproduction of fungal peroxidases in filamentous fungi (Thesis). Univer-
ing of the CreA gene in Aspergillus nidulans. Appl Environ Microbiol 2000;66: sity of Leiden; 2001.
4579–81. Crawford JM, Dancy BCR, Hill EA, Udwary DW, Townsend CA. Identification of a starter
Bedford DJ, Schweizer E, Hopwood DA, Khosla C. Expression of a functional fungal poly- unit acyl-carrier protein transacylase domain in an iterative type I polyketide
ketide synthase in the bacterium Streptomyces coelicolor A3(2). J Bacteriol synthase. Proc Natl Acad Sci USA 2006;103:16728–33.
1995;177:4544–8. Crawford L, Stepan AM, McAda PC, Rambosek JA, Conder MJ, Vinci VA, et al. Production
Beijersbergen AGM, Bundock P, Gouka RJ, de Groot MJA, Hooykaas PJJ. Agrobacterium of cephalosporin intermediates by feeding adipic acid to recombinant Penicillium
mediated transformation of moulds, in particular those belonging to the genus chrysogenum strains expressing ring expansion activity. Bio-Technology 1995;13:
Aspergillus. United States Patent 6255115. 2001. 58–62.
Bennett JW. The molds of Katrina. Update (NY Acad Sci) 2006; Jan/Feb: 6–9. Crystal R. α1-Antitrypsin deficiency. Marcel Dekker Inc; 1996.
Bergmann S, Schumann J, Scherlach K, Lange C, Brakhage AA, Hertweck C. Genomics-driven Cullen D. Recent advances on the molecular genetics of ligninolytic fungi. J Biotechnol
discovery of PKS–NRPS hybrid metabolites from Aspergillus nidulans. Nature Chem 1997;53:273–89.
Biol 2007;3:213–7. Cuomo CA, Guldener U, Xu J-R, Trail F, Turgeon BG, Di Pietro A, et al. The Fusarium
Berka RM, Cullen D, Gray GL, Gregory Lawrence H, Hayenga KJ, Lawlis VB. Heterologous graminearum genome reveals a link between localized polymorphism and patho-
polypeptides expressed in filamentous fungi, processes for making same, and vec- gen specialization. Science 2007;317:1400–2.
tors for making same. United States Patent 6,379,928. 2002. Cutler JE, Deepe GS, Klein BS. Advances in combating fungal diseases: vaccines on the
Berka RM, Cullen D, Gray GL, Hayenga KJ, and Lawlis VB. Heterologous polypeptides threshold. Nat Rev Microbiol 2007;5:13–28.
expressed in filamentous fungi, processes for making same, vectors for making Dai Z, Mao X, Magnuson JK, Lasure LL. Identification of genes associated with morphol-
same. United States Patent Application 20030224482. 2003. ogy in Aspergillus niger by using suppression subtractive hybridization. Appl Envi-
Bhambra GK, Wang Z-Y, Soanes DM, Wakley GE, Talbot NJ. Peroxisome carnithine ace- ron Microbiol 2004;70:2474–85.
tyl transferase is required for elaboration of penetration hyphae during plant infec- Damude HG, Zhang H, Farrall L, Ripp KG, Tomb J-F, Hollerbach D, et al. Identification of
tion by Magnaporthe grisea. Mol Microbiol 2006;72:6345–54. bifunctional 12/3 fatty acid desaturases for improving the ratio of 3–6 fatty acids in
Boel E, Christensen T, Woldike H. Process for production of protein products in microbes and plants. Proc Natl Acad Sci USA 2006;103:9446–51.
Aspergillus. United States Patent 5,536,661. 1996. De Pourcq K, De Schutter K, Callewaert N. Engineering of glycosylation in yeast and
Bolar JP, Norelli JL, Wong KW, Hayes CK, Harman GE, Aldwinckle HS. Expression of other fungi: current state and perspectives. Appl Microbiol Biotechnol 2010;87:
endochitinase from Trichoderma harzianum in transgenic apple increases resis- 1617–31.
tance to apple scab and reduces vigor. Phytopathology 2000;90:72–7. de Vries RP. Regulation of Aspergillus genes encoding plant cell wall polysaccharide-
Boller T, He SY. Innate immunity in plants: an arms race between pattern recognition degrading enzymes; relevance for industrial production. Appl Microbiol Biotechnol
receptors in plants and effectors in microbial pathogens. Science 2009;324:742–4. 2003;61:10–20.
Braaksma M, Martens-Uzonova EM, Punt PJ, Schaap PJ. An inventory of the Aspergillus Demain AL, Vaishnav P. Production of recombinant proteins by microbes and higher
niger secretome by combining in silico predictions with shotgun proteomics data. organisms. Biotechnol Advances 2009;27:297–306.
BMC Genome 2010;11:584–95. Demain AL. Biosolutions to the energy problem. J Indust Microbiol 2009;36:319–32.
Braaksma M, Punt PJ. Aspergillus as a cell factory for protein production: controlling Denison SH. pH regulation of gene expression. Fungal Genet Biol 2000;29:61–71.
protease activity in fungal production. In: Goldman GH, Osmani SA, editors. The As- Dennis C, Webster J. Antagonistic properties of species-groups of Thrichoderma. 1. Pro-
pergilli: genomics, medical aspects, biotechnology and research methods. Boca duction of non-volatile antibiotics. Trans Brit Mycol Soc 1971a;57:25–39.
Raton: CRC Press; 2008. p. 441–55. Dennis C, Webster J. Antagonistic properties of species-groups of Thrichoderma. 2. Pro-
Brantly ML, Wittes JT, Vogelmeier CF, Hubbard RC, Fells GA, Crystal RG. Use of highly duction of volatile antibiotics. Trans Brit Mycol Soc 1971b;57:41–8.
purified alpha 1-antitrypsin standard to establish ranges for the common normal Dennis C, Webster J. Antagonistic properties of species-groups of Thrichoderma. 3. Hy-
and deficient alpha 1-antitrypsin phenotypes. Chest 1991;100:703–8. phal interaction. Trans Brit Mycol Soc 1971c;57:363–9.
Brody H, Griffith J, Cuticchia AJ, Arnold J, Timberlake WE. Chromosome-specific recombinant deOliveira JMPF, deGraaf LH. Proteomics of industrial fungi: trends and insights. Appl
DNA libraries from the fungus Aspergillus nidulans. Nucleic Acids Res 1991;19:3105–9. Microbiol Biotechnol 2011;89:225–37.
Broekhuijsen MP, Mattern IE, Contreras R, Kinghorn JR, van den Hondel CAMJJ. Secre- Drexler H, Spiekermann P, Meyer A, Domergue F, Zank T, Sperling P, et al. Metabolic en-
tion of heterologous proteins by Aspergillus niger: Production of active human in- gineering of fatty acids for breeding of new oilseed crops: strategies, problems and
terleukin-6 in a protease deficient mutant by KEX2-like processing of a first results. J Plant Physiol 2003;160:779–802.
glucoamylase-hIL6 fusion protein. J Biotechnol 1993;31:135–45. Driouch H, Sommer B, Wittmann C. Morphology engineering of Aspergillus niger for im-
Buchert T, Oksanen T, Pere J, Siika-Aho M, Suurnakki A, Viikari L. Applications of proved enzyme production. Biotech Bioeng 2010;105:1058–68.
Trichoderma reesei enzymes in the pulp and paper industry. In: Harman GE, Kubi- Dunn-Coleman NS, Bloebaum P, Barka M, Bodie E, Robinson N, Armstrong G, et al. Commer-
cek CP, editors. Trichoderma and Gliocladium, 2. London: Taylor and Francis; 1998. cial levels of chymosin production by Aspergillus. Mol Gen Genet 1991;230:288–94.
p. 343–63. Elad Y, Kapat A. The role of Trichoderma harzianum protease in the biocontrol of Botrytis
Buxton FJ, Gabor V, Jacob NL. Aspergillus niger vacuolar aspartyl protease. United States cinerea. Eur J Plant Pathol 1999;105:177–89.
Patent 5674728. 1997. Elander R. Strain improvement and preservation of beta-lactam producing microor-
Cardoso PG, Teixeira JA, de Queiroz MV, de Araujo EF. Pectin lyase production by re- ganisms. In: Demain AL, Solomon N, editors. Antibiotics containing the β-lactam
combinant Penicillium griseoroseum. Can J Microbiol 2010;56:831–7. structure I. New York: Springer-Verlag; 1983. p. 97-146.
Chavez R, Roa A, Navarrete K, Trebotich J, Espinosa Y, Vaca I. Evaluation of properties of Evans BS, Robinson SJ, Kelleher NL. Surveys of non-ribosomal peptide and polyketide
several cheese-ripening fungi for potential biotechnological applications. assembly lines in fungi and prospects for their analysis in vitro and in vivo. Fungal
Mycoscience 2010;51:84–7. Genet Biol 2011;48:49–61.
Chen XA, Ishida N, Todaka N, Nakamura R, Maruyama JI, Takahashi H, et al. Promotion Fang W, Vega-Rodriguez J, Ghosh AK, Jacobs-Lorena M, Kang A, St Leger RJ. Develop-
of efficient saccharification of crystalline cellulose by Aspergillus fumigatus Swo1. ment of transgenic fungi that kill human malaria parasites in mosquitoes. Science
Appl Environ Microbiol 2010;76:2556–61. 2011;331:1074–7.
Cherif M, Benhamou N. Cytochemical aspects of chitin breakdown during the parasitic Fawole OB, Odunfa SA. Some factors affecting production of pectic enzymes by
action of a Trichoderma sp. on Fusarium oxysporum f.sp. radicislycopersici. Phytopa- Aspergillus niger. Int Biodeter Biodegr 2003;52:223–7.
thology 1990;80:1406–14. Feofilova EP, Dergeeva YE, Inashechkin AA. Biodiesel-fuel: content, production, producers,
Cherry J, Fidantsef A. Directed evolution of industrial enzymes: an update. Curr Opin contemporary biotechnology (review). Appl Biochem Microbiol 2010;46:369–78.
Biotechnol 2003;14:438–43. Fleissner A, Dersch P. Expression and export: recombinant protein production systems
Chiang YM, Szewczyk E, Davidson AD, Entwistle R, Keller NP, Wang CC, et al. Character- for Aspergillus. Appl Microbiol Biotechnol 2010;87:1255–70.
ization of the Aspergillus nidulans monodictyphenone gene cluster. Appl Environ Fujihiro S, Higuchi R, Hisamatsu S, Sonoki S. Metabolism of hydroxylated PCBs by
Microbiol 2010;76:2067–74. cloned laccase isoforms. Appl Microbiol Biotechnol 2009;82:853–60.
Christensen T, Lehmbeck J. Fungus wherein the areA, pepC and/or pepE genes have been Fujii I. Functional analysis of fungal polyketide biosynthesis genes. J Antibiot 2010;63:
inactivated. United States Patent 6013452. 2000. 207–18.
Christensen T, Woeldike H, Boel E, Mortensen SB, Hjortshoej K, Thim L, et al. High level ex- Fujinaga M, Cherney MM, Oyama H, Oda K, James MNG. The molecular structure and
pression of recombinant genes in Aspergillus oryzae. Bio-Technology 1988;6:1419–22. catalytic mechanism of a novel carboxyl peptidase from Scytalidium lignicolum.
Chung SJ, Kim S, Sapkota K, Choi BS, Shin C, Kim SJ. Expression of recombinant interleu- Proc Natl Acad Sci USA 2004;101:3364–9.
kin-32 in Pleurotus eryngii. Ann Microbiol 2011;61:331–8. Fulci V, Macino G. Quelling: post-translational gene silencing guided by small RNAs in
Clutterbuck AJ. The validity of the Aspergillus nidulans linkage map. Fungal Genet Biol Neurospora crassa. Curr Opin Microbiol 2007;10:199–203.
1997;21:267–77. Galagan JE, Calvo SE, Borkovich KA, Selker EU, Read ND, Jaffe D. The genome sequence
Collen A, Ward M, Tjerneld F, Stalbrand H. Genetic engineering of the Trichoderma of the filamentous fungus Neurospora crassa. Nature 2003;422:859–68.
reesei endoglucanase I (Cel7B) for enhanced partitioning in aqueous two-phase Galagan JE, Calvo SE, Cuomo C, Ma L-J, Wortman JR, Batzoglou S, et al. Sequencing of
systems containing thermoseparating ethylene oxide-propylene oxide copoly- Aspergillus nidulans and comparative analysis with A. fumigatus and A. oryzae. Na-
mers. J Biotechnol 2001;87:179–91. ture 2005;438:1105–15.
Colot HV, Park G, Turner GE, Ringelberg C, Crew CM, Litvinkova L, et al. A Galanti YM, De Conti A, Monteverdi R. Applications of Trichoderma enzymes in the tex-
high-throughput gene knockout procedure for Neurospora reveals functions for tile industry. In: Harman GE, Kubicek CP, editors. Trichoderma and Gliocladium,
multiple transcription factors. Proc Natl Acad Sci USA 2006;103:10352–7. Vol 2. London: Taylor and Francis; 1998. p. 311–26.
1136 O.P. Ward / Biotechnology Advances 30 (2012) 1119–1139

Geremia RA, Goldman GH, Jacobs D, Ardrtes W, Vila SB, Van Montagu M, et al. Molec- Iwashita K. Recent studies of protein secretion by filamentous fungi—review. J Biosci
ular characterization of the proteinase-encoding gene, prb1, related to mycopara- Bioeng 2002;94:530–5.
sitism by Trichoderma harzianum. Mol Microbiol 1993;8:603–13. Jacobs DI, Olsthoorn MMA, Maillet I, Akeroyd M, Breestraat S, Donkers S, et al. Effective
Gerngross TU. Advances in the production of human therapeutic proteins yeasts and lead saelection for improved porotein production in Aspergillus niger based on in-
filamentous fungi. Nature Biotechnol 2004;22:1409–14. tegrated genomics. Fungal Genet Biol 2009;46:S141–52.
Geysens S, Whyteside G, Archer DB. Genomics of protein folding in the endoplasmic re- Jami MS, Barreiro C, Garcia Estradfa C, Martin JF. Proteome analysis of the penicillin
ticulum, secretion stress and glycosylation in the aspergilli. Fungal Genet Biol producer Penicillium chrysogenum: characterization of protein changes during in-
2009;46:S121–40. dustrial strain improvement. Mol Cell Proteomics 2010;9:1182–98.
Gidijala L, Kiel JAKW, Douma RD, Seifar RM, van Gulik WM, Bovenberg RA, et al. Amn Jeenes DJ, MacKenzie DA, Archer DB. Transcriptional and post translational events af-
engineered yeast efficiently secreting penicillin. PLoS One 2009;4(12):e8317. fect the production of secreted hen egg white lysozyme by A. niger. Transgen Res
Gonzalez-Vogel A, Eyzaguirre J, Oleas G, Callegari E, Navarette M. Proteomic analysis in 1994;3:297–303.
non-denaturing condition of the secretome reveals the presence of multienzyme Jeenes DJ, Marczinke B, MacKenzie DA, Archer DB. A truncated glucoamylase gene fu-
complexes in Penicillium purpurogenum. Appl Microbiol Biotechnol 2011;89: sion for heterologous protein secretion from A. niger. FEMS Microbiol Lett
145–55. 1993;107:267–71.
Gordon CL, Archer DB, Jeenes DJ, Doonan JH, Wells B, Trinci APJ, et al. A glucoamylase:: Jeenes DJ, Pfaller R, Archer DB. Isolation and characterization of a novel stress inducible
GFP gene fusion to study protein secretion by individual hyphae of Aspergillus PDI family gene from A. niger. Gene 1997;194:151–6.
niger. J Microbiol Meth 2000;42:39–48. Jensen EB, Boominathan KC. Thermophilic fungal expression system. United States Pat-
Goto M. Protein O-glycosylation in fungi: diverse structures and multiple functions. ent 5695985. 1997.
Biosci Biotechnol Biochem 2007:1415–27. Jin FJ, Watanabe T, Juvvadi PR, Maruyama J-I, Arioka M, Kitamoto K. Double dis-
Gouka RJ, Punt PJ, van den Hondel CAMJJ. Efficient production of secreted proteins by ruption of the proteinase genes, tppA and pepE, increase the production
Aspergillus: progress, limitations and prospects. Appl Microbiol Biotechnol level of human lysozyme by Aspergillus oryzae. Appl Gen Mol Biotechnol
1997a;47:1-11. 2007;76:1059–68.
Gouka RJ, Punt PJ, van den Hondel CAMJJ. Glucoamylase gene fusions alleviate limita- Joensuu JJ, Conley AJ, Lienemann M, Brandle JE, Linder MB, Menassa R. Hydrophobin fu-
tions for protein production in Aspergillus awamori at the transcriptional and sions for high-level transient protein expression and purification in Nicotiana
(post) translational levels. Appl Environ Microbiol 1997b;63:488–97. benthamiana. Plant Physiol 2010;152:622–33.
Grimm LH, Kelly S, Krull R, Hempel DC. Morphology and productivity of filamentous Kainz E, Gallmetzer A, Hatzl C, Nett JH, Li H, Schinko T, et al. N-glycan modification in
fungi. Appl Microbiol Biotechnol 2005;69:375–84. Aspergillus species. Appl Environ Microbiol 2008;74:1076–86.
Guais O, Tourrasse O, Dourdoigne M, Parrou JL, Francois JM. Characterization of the Karnaukhova E, Ophir Y, Golding B. Recombinant human alpha-1-proteinase inhibitor
family GH54 α-l-arabinofuranosidases in Penicillium funiculosum, including a towards therapeutic use. Amino Acids 2006;30:317–32.
novel protein bearing a cellulose-binding domain. Applied Microbiol Biotechnol Karnaukhova E, Ophir Y, Trinh L, Dalal N, Punt P, Golding B, et al. Expression of human
2010;87:1007–21. α(I)-proteinase inhibitor in Aspergillus niger. Microb Cell Factories 2007;6(34):
Guasi O, Borderies G, Pichereaux C, Maestracci M, Neugnot V, Rossignol M, et al. Prote- 1-10.
omics analysis of “Rovabio Excel”, a secreted protein cocktail from the filamentous Kasajima T, Yamaguichi M, Hirai N, Ohmachi T, Yoshida T. In vivo expression of
fungus Penicillium funiculosum grown under industrial process fermentation. J UDP-N-acetylglucosamine: α-3-D-mannoside β-1,2-N-acetylglucosyl-transferase
Indust Microbiol Biotechnol 2008;35:1659–68. 1 (GnT-1) in Aspergillus oryzae and effects on the sugar chain of alpha-amylase.
Guo BH, Wang YC, Zhou XW, Hu K, Tan F, Miao ZQ, Tang KX. An endophytic Biosci Biotechnol Biochem 2006;70:2662–8.
taxol-producing fungus BT2 isolated from Taxus chinensis var. mairei. Afr J Biotech- Kealey JT, Liu L, Santi DV, Betlach MC, Barr PJ. Production of a polyketide natural prod-
nol 2006;5:875–7. uct in nonpolyketide-producing prokaryotic and eukaryotic hosts. Proc Natl Acad
Gutierrez S, Diez B, Alvarez E, Barredo JL, Martin JF. Expression of the penDE gene of Sci USA 1998;95:499–505.
Penicillium chrysogenum encoding isopenicillin N acyltransferase in Cephalosporium Kennedy J, Auclair K, Kendrew SG, Park C, Vederas JC, Hutchinson CR. Modulation of
acremonium: production of benzylpenicillin by the transformants. Mol Gen Genet polyketide synthase activity by accessory proteins during lovastatin biosynthesis.
1991;225:56–64. Science 1999;284:1368–72.
Gwynne DI, Devchand M. Expression of foreign proteins in the genus Aspergillus. Bio- Kern A, Tilley E, Hunter IS, Legisa M, Glieder A. Engineering primary metabolic path-
technology 1992;23:203–14. ways of industrial micro-organisms. J Biotechnol 2007;129:6-29.
Hammond TM, Keller NP. RNA silencing in Aspergillus nidulans is independent of Kim Y, Nandakumar MP, Marten MR. Proteomics of filamentous fungi. Trends Biotech-
RNA-dependent RNA polymerases. Genetics 2005;169:607–17. nol 2007;25:395–400.
Haran S, Schickler H, Peer S, Logemann S, Oppenheim A, Chet I. Increased constitutive Kinghorn JR, Unkles SE. Aspergillus. New York: Plenum Press; 1994.
chitinase activity in transformed Trichoderma harzianum. Biological Control Kirk TK, Farrel RL. Enzymatic “combustion”: the microbial degradation of lignin. Ann
1993;3:101–8. Rev Microbiol 1987;41:465–505.
Harkki A, Uusitalo J, Bailey M, Penttila M, Knowles JKC. A novel fungal expression sys- Kitamoto K, Kimura K, Gomi K, Kumagai C. Electrophoretic karyotype and gene assign-
tem: secretion of actice calf chymosin from the filamentous fungus Trichoderma ment to chromosomes of Aspergillus oryzae. Biosci Biotechnol Biochem 1994;58:
reesei. Nature Biotechnol 1989;7:596–603. 1467–70.
Harman GE, Howell CR, Viterbo A, Chet I, Lorito M. Trichoderma species—opportunistic, Kleinkauf H, von Döhren H. Non-ribosomal biosynthesis of peptide antibiotics. Eur J
avirulent plant symbionts. Nature Revs Microbiol 2004;2(1):43–56. Biochem 1990;192:1-15.
Harris DM, Westerlaken I, Schipper D, van der Krogt ZA, Gombert AK, Sutherland J, Knutzon DS, Thurmond JM, Huang Y-S, Chaudhary S, Bobik EG, Chan GM, et al. Identi-
et al. Engineering of Penicillium chrysogenum for fermentative production of a fication of Δ5-desaturase from Mortierella alpina by heterologous expression in
novel carbamoylated cephem antibiotic precursor. Metab Eng 2009;11:125–37. bakers' yeast and canola. J Biol Chem 1998;273:29360–6.
Hastrup S, Branner S, Jorgensen BR, Christensen T, Jorgensen BB, Shuster JR et al. Pro- Kobayashi T, Abe K, Asai K, Gomi K, Juwadi PR, Kitamoto K, et al. Genomics of Aspergil-
cesses for producing an enzyme. United States Patent 5,702,934. 1997. lus oryzae. Biosci Biotechnol Biochem 2007;71:646–70.
Hawksworth DL. The magnitude of fungal diversity: the 1.5 million species estimate Kumar R, Singh S, Singh OV. Bioconversion of lignocellulosic biomass: biochemical and
revisited. Mycol Res 2001;105:1422–32. molecular perspectives. J Indust Microbiol Biotechnol 2008;35:377–91.
Hong H, Datla N, Reed DW, Covello PS, MacKenzie SL, Qiu X. High-level production of Kwon-Chung KJ, Bennett JE. Mucormycosis. Medical mycology. Philadelphia: Lea and
γ-linolenic acid in Brassica juncea using a Δ6 desaturase from Pythium irregulare. Febiger; 1992. p. 524–59.
Plant Physiol 2002:354–62. Lehmbeck J. Alkaline protease deficient filamentous fungi. United States Patent 6,
Huang X-M, Yang Q, Liu Z-H, Fan J-X, Chen X-L, Song J-Z, et al. Cloning and heterolo- 291,209. 2001.
gous expression of a novel endoglucanase gene egVIII from Trichoderma viride in Li X, Wang D, Zhou F, Yang H, Bhaskar R, Hu J, et al. Cloning and expression of a cellu-
Saccharomyces cerevisiae. Appl Biochem Biotechnol 2010;162:103–15. lose gene in the silkworm, Bombyx mori by improved Bac-to-Bac/BmNPV baculo-
Huang Y-S, Chaudhary S, Thurmond JM, Bobik Jr EG, Yuan L, Chan GM, et al. Cloning of virus expression system. Mol Biol Rep 2010;8:3721–8.
Δ12- and Δ6-desaturases from Mortierella alpina and recombinant production of Lin FC, Liu XH, Wang HK, Zhang CL. Recent research and prospect on taxol and its pro-
γ-linolenic acid in Saccharomyces cerevisiae. Lipids 1999;34:649–59. ducing fungi. Acta Microbiol Sin 2003;43:534–8.
Ibrahim AS, Edwards JEJ, Filler SG. Zygomycosis. In: Dismukes WE, Pappas PG, Sobel JD, ed- Liu J-W, Huang Y-S, DeMichele S, Bergana M, Bobik Jr E, Hastilow C, et al. Evaluation of
itors. Clinical mycology. New York: Oxford University Press; 2003. p. 241–51. the seed oils from a canola plant genetically transformed to produce high levels of
Ichishima E, Taya N, Ikeguichi M, Chiba Y, Nakamura M, Kawabata C, et al. Molecular γ-linolenic acid. In: Hunag Y-S, Ziboh VA, editors. γ-Linolenic acid: recent ad-
and enzymatic properties of recombinant 1,2-α-mannosidase from Aspergillus sai- vances in biotechnology and clinical applications. Champaign IL: AOCS Press;
toi overexpressed in Aspergillus oryzae cells. Biochem J 1999;339:589–97. 2001. p. 61–71.
Imanaka H, Tanaka S, Feng B, Imamura K, Nakanishi K. Cultivation characteristics Liu L, Liu J, Qiu RX, Zhu XG, Dong ZY, Tang GM. Improving heterologous gene ex-
and gene expression profiles of Aspergillus oryzae by membrane-surface liquid pression in Aspergillus niger by introducing multiple copies of protein-binding
culture, shaking-flask culture and agar-plate culture. J Biosci Bioeng sequence containing CCAAT to the promoter. Lett Appl Microbiol 2003;36:
2010;109:267–73. 358–61.
Imazaka A, Tanaka A, Harimoto Y, Yamamoto M, Akimitsu K, Park P, et al. Contribution Loder PB, Abraham EB. Isolation and nature of intracellular peptides from a cephalo-
of peroxisomes to secondary metabolism and pathogenicity in the fungal plant sporin C producing Cephalosporium sp. Biochem J 1971;123:471–6.
pathogen Alternaria alternata. Eukaryot Cell 2010;9:682–94. Lorito M, Woo SL, Fernandez IG, Colucci G, Harman GE, Pintor-Toro JA, et al. Genes from
Ishida H, Matsumura K, Hata Y, Kawato A, Suginami K, Abe Y, et al. Establishment of a mycoparasitic fungi as a source for improving plant resistance to fungal pathogens.
hyper-protein production system in submerged Aspergillus oryzae culture under Proc Natl Acad Sci USA 1998;95:7860–5.
tyrosinase-encoding gene (melO) promoter control. Appl Microbiol Biotechnol Lubertozzi D, Keasling JD. Developing Aspergillus as a host for heterologous expression.
2001;57:131–7. Biotechnol Adv 2009;27:53–75.
O.P. Ward / Biotechnology Advances 30 (2012) 1119–1139 1137

Ma LJ, Ibrahim AS, Skory C, Grabherr MG, Burger G, Butler M, et al. Genomic analysis of Muller D, Stahl U, Meyer V. Application of hammerhead ribozymes in filamentous
the basal lineage fungus Rhizopus oryzae reveals a whole-genome duplication au- fungi. J Microbiol Meth 2006;65:585–95.
thor(s): source. PLoS Genet 2009;5(7). Article No: e1000549. Muller WH, Bovenberg RA, Groothuis MH, Kattevilder F, Smaal EB, van der Voort LH,
Ma SM, et al. Enzymatic synthesis of aromatic polyketides using PKS4 from Gibberella et al. Involvement of microbodies in penicillin biosynthesis. Biochim Biophys
fujikuroi. J Am Chem Soc 2007;129:10642–3. Acta 1992;1116:210–3.
Mach RL, Zeilinger S. Regulation of gene expression in industrial fungi: Trichoderma. Muller WH, van der Krift TP, Krouwer AJJ, Wosten HAB, van der Voort L, Smaal EB, et al.
Appl Microbiol Biotechnol 2003;60:515–22. localization of the pathway of the penicillin biosynthesis in Penicillium chryso-
Machida M. Progress of Aspergillus oryzae genomics. Adv Appl Microbiol 2002;51: genum. EMBO 1991;10:489–95.
81-106. Nakajima K, Asakura T, Maruyama J, Morita Y, Oike H, Shimizu-Ibuka A, et al. Extra-
Machida M, Asai K, Sano M, Tanaka T, Kumagai T, Terai G, et al. Genome sequencing and cellular production of neoculin, a sweet-tasting heterodimeric protein with
analysis of Aspergillus oryzae. Nature 2005;438:1157–61. taste-modifying activity by Aspergillus oryzae. Appl Environ Microbiol 2006;72:
MacKenzie DA, Wongwathanarat P, Carter AT, Archer DB. Isolation and use of a homol- 3716–23.
ogous histone H4 promoter and a ribosomal DNA region in a transformation vector Nevalainen H, Souminen P, Taimisto K. On the safety of Trichoderma reesei. J Biotechnol
for the oil-producing fungus Mortierella alpina. Appl Environ Microbiol 2000;66: 1994;37:193–200.
4655–61. Nevalainen KMH, Te'o VS, Bergquist PL. Heterologous protein expression in filamen-
MacKenzie DA, Kraunsoe JAE, Chesshyre JA, Lowe G, Komiyama T, Fuller RS, et al. Aber- tous fungi. Trends Biotechnol 2005;23:468–74.
rant processing of wild-type and mutant bovine pancreatic trypsin inhibitor se- Ngiam C, Jeenes DA, Punt PJ, van den Hondel CAMJJ, Archer DA. Characterization of a
creted by Aspergillus niger. J Biotechnol 1998;63:137–46. foldase, protein disulfide isomerase A in the protein secretory pathway of Aspergil-
Maeda H, Sano M, Maruyama Y, Tanno T, Akao T, Totsuka Y, et al. Transcriptional anal- lus niger. Appl Environ Microbiol 2000;66:775–82.
ysis of genes for energy catabolism and hydrolytic enzymes in the filamentous fun- Nuesch J, Heim J, Treichler H-J. The biosynthesis of sulfur-containing β-lactam antibi-
gus Aspergillus oryzae using cDNA microarrays and expressed sequence tags. Appl otics. Annu Rev Microbiol 1987;41:51–75.
Microbiol Biotechnol 2004;65:74–83. Nyyssonen E, Penttila M, Harkki M, Saloheimo A, Knowles JK, Keranen S. Efficient pro-
Mantyla A, Paloheimo M, Hakola S, Lindberg E, Leskinen S, Kallio J, et al. Production in duction of antibody fragments by the filamentous fungus Trichoderma reesei. Bio-
Trichoderma reesei of three xylanases from Chaetomium thermophilum: a recombi- technology 1993;11:591–5.
nant thermoxylanase for biobleaching of kraft pulp. Appl Microbiol Biotechnol O'Donnell D, Wang L, Xu J, Ridgway D, Gu T, Moo-Young M. Enhanced heterologous
2007;76:377–86. protein production in Aspergillus niger through pH control of extracellular protease
Maor R, Shirasu K. The arms race continues: battle strategies between plants and fungal activity. Biochem Eng 2001;8:187–93.
pathogens. Curr Opin Microbiol 2005;8:399–404. Oda K, Kakizono D, Yamada O, Lefuji H, Akita O, Iwashita K. Proteomic analysis of extra-
Marahiel MA. Multidomain enzymes involved in peptide synthesis. FEBS Lett cellular proteins of Aspergillus oryzae grown under submerged and solid-state cul-
1992;307:40–3. ture conditions. Appl Environ Microbiol 2006;72:3448–57.
Maras M, De Bruyn A, Vervecken W, Uusitalo J, Penttila M, Busson R, et al. In vivo Ouyang J, Yan M, Kong D, Xu L. A complete protein pattern of cellulose and hemicellu-
synthesis of complex N-glycans by expression of human N-acetylglucosamyl- lase genes in the filamentous fungus Trichoderma reesei. Biotechnol J 2006;1:
transferase 1 in the filamentous fungus Trichoderma reesei. FEBS Lett 1266–74.
1999a;452:365–70. Papagianni M, Young MM. Protease secretion in glucoamylase producer Aspergillus
Maras M, Saelens X, Laroy W, Piens K, Claeyssens M, Fiers W, et al. In vitro conversion niger cultures: fungal morphology and inoculum effects. Proc Biochem 2002;l37:
of the carbohydrate moiety of fungal glycoproteins to mammalian-type oligosac- 1271–8.
charides—evidence for N-acetylglucosaminetransferase-1-accepting glycans from Papagiannopoulos P, Andrianopoulos A, Sharp JA, Davis MA, Hynes MJ. The hapC gene
Trichoderma reesei. Eur J Biochem 1997;249:701–7. of Aspergillus nidulans is involved in the expression of CCAAT-containing pro-
Maras M, van Die I, Contreras R, van den Hondel CAMJJ. Filamentous fungi as production moters. Mol Gen Genet 1996;251:412–21.
organisms for glycoproteins of bio-medical interest. Glycoconj J 1999b;16:99-107. Paper JM, Scott-Craig JS, Adhikari ND, Cuomo CA, Walton JD. Comparative proteomics
Martin JA, Murphy RA, Power RFG. Cloning and expression of fungal phytases in genet- of extracellular proteins in vitro and in planta from the pathogenic fungus Fusari-
ically modified strains of Aspergillus awamori. J Ind Microbiol Biotechnol 2003;30: um graminearum. Proteomics 2007;7:3171–83.
568–76. Paul D, Pandey G, Jain RK. Suicidal genetically engineered microorganisms for bioreme-
Martin JF. Clusters of genes for the biosynthesis of antibiotics—regulatory genes and diation: need and perspectives. Bioessays 2005:563–73.
overproduction of pharmaceuticals. J Ind Microbiol Biotechnol 1992;9:73–90. Pel HJ, de Winde JH, Archer DB, Dyer PS, Hofmann G, Schaap PJ, et al. Genome sequenc-
Martín JF, Liras P. Organization and expression of genes involved in the biosynthesis of ing and analysis of the versatile cell factory Aspergillus niger CBS 513.88. Nature
antibiotics and other secondary metabolites. Annu Rev Microbiol 1989;43: Biotechnol 2007;25:221–31.
173–206. Penttila M. Heterologous protein production in Trichoderma. In: Harman GE, Kubicek CP,
Martinez D, Berka RM, Henrissat B, Saloheimo M, Arvas M, Baker SE, et al. Genome se- editors. Trichoderma and Gliocladium. London: Taylor and Francis; 1998. p. 365–82.
quencing and analysis of the biomass-degrading fungus Trichoderma reesei (syn. Pisanelli I, Kujawa M, Gschnitzer D, Spaduit O, Seiboth B, Peterbauer C. Heterologous
Hypocrea jecorina). Nature Biotechnol 2008;26:553–60. expression of an Agaricus meleagris pyranose dehydrogenase-encoding gene in As-
Martinez D, Larrondo LF, Putnam N, Gelpke MD, Huang K, Chapman J, et al. Genome se- pergillus spp. and characterization of the recombinant enzyme. Appl Microbiol Bio-
quence of the lignocellulose degrading fungus Phanerochaete chrysosporium strain technol 2010;86:599–606.
RP78. Nature Biotechnol 2004;22:695–700. Poidevin L, Levasseur A, Paes G, Navarro D, Heiss-Blanquet S, Asther M, et al. Heterol-
Maruyama J, Ohnuma H, Yoshikawa A, Kadokura H, Nakajima H, Kitamoto K. Produc- ogous production of the Piromyces equi cinnamoyl esterase in Trichoderma reesei
tion and product quality assessment of human hepatitis B virus-preS2 antigen in for biotechnological applications. Letts Appl Microbiol 2009;49:673–8.
submerged and solid state cultures of A. oryzae. J Biosci Bioeng 2000;90:118–20. Punt PJ, van Biezen N, Conesa A, Albers A, Mangnus J, van den Hondel C. Filamentous
Masai K, Maruyama J, Nakajima H, Sakamoto K, Akita O, Kitamoto K. Analysis of differ- fungi as cell factories for heterologous protein production. Trends Biotechnol
ential gene expression across regions of the Aspergillus oryzae mycelium. Fungal 2002;20:200–6.
Genet Newslett 2005;52:161. Punt PJ, van Gemeren IA, Drint-Kuijvenhoven A, Hessing JGM, van Muijlwijk-Har-
Matsuzaki F, Shimizu M, Wariishi H. Proteomic and metabolomic analyses of the teveld GM, Beijersbergen A, et al. Analysis of the role of the gene bipA, encod-
white-rot fungus Phanerochaete chrysogenum exposed to exogenous benzoic acid. ing the major endoplasmic reticulum chaperone protein in the secretion of
J Proteome Res 2008;7:2342–50. homologous and heterologous proteins in black Aspergilli. Appl Microbiol
Meesapyodsuk D, Reed DW, Covello PS, Qiu X. Primary structure, regioselectivity, and Biotechnol 1998;50:447–54.
evolution of the membrane-bound fatty acid desaturases of Claviceps purpurea. J Punt PJ, Schuren FHJ, Lehmbeck J, Christens J, Hjort C, van den Hondel C. Characteriza-
Biol Chem 2007;282:20191–9. tion of the Aspergillus niger prtT, a unique regulator of extracellular protease
Meyer HW. Molds in floor dust and building-related symptoms in adolescent school encoding genes. Fungal Genet Biol 2008;45:1591–9.
children. Indoor Air 2004;14:65–72. Qin Y, Ying SH, Chen Y, Shen ZC, Feng MG. Integration of insecticidal Vip3Aa1 into
Meyer V. Genetic engineering of filamentous fungi-progress, obstacles and future Beauveria bassiana enhances fungal virulence to Spodoptera litura larvae by cuticle
trends. Biotechnol Adv 2008;26:177–85. and per os infection. Appl Environ Microbiol 2010;76:4611–8.
Michaelson LV, Lazarus CM, Griffiths G, Napier JA. Isolation of a Δ5-fatty acid desaturase Radzio R, Kueck U. Synthesis of biotechnologically relevant heterologous proteins in fil-
gene from Mortierella alpina. J Biol Chem 1998;273:19055–9. amentous fungi. Proc Biochem 1997;32:529–37.
Minetoki T, Kumagai C, Gomi K, Kitamoto K, Takahashi K. Improvement of promoter ac- Ratledge C. Lipids in fatty acids. In: Rose AH, editor. Primary products of metabolism.
tivity by the introduction of multiple copies of the conserved region III sequence, London: Academic Press; 1978. p. 263–96.
involved in the efficient expression of Aspergillus oryzae amylase-encoding genes. Roberts IN, Jeenes DJ, Mackenzie DA, Wilkinson AP, Summer IG, Archer DB. Heterolo-
Appl Microbiol Biotechnol 1998;50:459–67. gous gene expression in A. niger: A glucoamylase–porcine pancreatic phospholi-
Monk BC, Goffeau A. Outwitting multidrug resistance to antifungals. Science 2008;321: pase A2 fusion protein is secreted and processed to yield mature enzyme. Gene
367–9. 1992;122:155–61.
Monsanto. Microbial sequence database. http://microbial.cereon.com2001. Robin J, Jakobsen M, Beyer M, Noorman H, Nielsen J. Physiological characterisation of Penicil-
Moralejo FJ, Cardoza RE, Gutierrez S, Lombrana M, Fierro F, Martin JF. Silencing of the lium chrysogenum strains expressing the expandase gene from Streptomyces clavuligerus
Aspergillopepsin B (pepB) gene of Aspergillus awamori by antisense RNA expres- during batch cultivations. Growth and adipoyl-7-aminodeacetoxycephalosporanic
sion or protease removal by gene disruption results in a large increase in thauma- acid production. Appl Microbiol Biotechnol 2001;57:357–62.
tin production. Appl Environ Microbiol 2002;68:3550–9. Robin J, Lettier G, McIntyre M, Noorman H, Nielsen J. Continuous cultivations of a Pen-
Moriguchi T, Kezuka Y, Nonaka T, Ebizuka Y, Fujii I. Hidden function of catalytic domain icillium chrysogenum strain expressing the expandase gene from Streptomyces cla-
in 6-methylsalicylic acid synthase for product release. J Biol Chem 2010;285: vuligerus: growth yields and morphological characterization. Biotechnol Bioeng
15637–43. 2003a;83:361–8.
1138 O.P. Ward / Biotechnology Advances 30 (2012) 1119–1139

Robin J, Bonneau S, Schipper D, Noorman H, Nielsen J. Influence of the adipate and dis- Swift RJ, Wiebe MG, Robson GD, Trinci APJ. Recombinant glucoamylase production by
solved oxygen concentrations on the beta-lactam production during continuous Aspergillus niger B1 in chemostat and pH-autostat cultures. Fungal Genet Biol
cultivations of a Penicillium chrysogenum strain expressing the expandase gene 1998;25:100–9.
from Streptomyces clavuligerus. Metab Eng 2003b;5:42–8. Szewczyk E, Chiang YM, Oakley CE, Davidson AD, Wang CC, Oakley BR. Identification
Rodgers CJ, Blanford CF, Giddens SR, Skamnioto P, Armstrong FA, Gurr SJ. Designer lac- and characterization of the asperthecin gene cluster of Aspergillus nidulans. Appl
cases: a vogue for high-potential fungal enzymes. Trends Biotechnol 2010;28: Environ Microbiol 2008;74:7607–12.
63–72. Tailor MJ, Richardson T. Application of microbial enzymes in food systems and in bio-
Romaine PC, Chen X. Methods and compositions for highly efficient transformation of technology. Adv Appl Microbiol 1979;25:7-35.
filamentous fungi. United States Patent Application 20020070007. 2002. Takuchi M, Ichishima E. A 155 K acid carboxypeptidase O from Aspergillus oryzae. Agric
Ruijter GJG, Kubicek CP, Visser J. Production of organic acids by fungi. Mycota 2002;10: Biol Chem 1986;50:633–8.
213–20. te Biesebeke R, Ruijter G, Rahardjo YSP, Hoogschagen MJ, Heerikhuisen M, Levin A,
Ruijter GJG, Panneman H, Visser J. Overexpression of phosphofructokinase and pyru- et al. Aspergillus oryzae in solid-state and submerged fermentations. FEMS Yeast
vate kinase in citric acid producing Aspergillus niger. Biochim Biophys Acta Res 2002;2:245–8.
1997;1334:317–26. Tian C, Beeson WT, Iaverone AT, Sun J, Marletta MA, Cate JH, Glass NL. Systems anasly-
Sakaki T, Munetsuna E. Enzyme systems for biodegradation of polychlorinated sis of plant cell wall degradation by the model filamentous fungus Neurospora
dibenzo-p-dioxins. Appl Microbiol Biotechnol 2010;88:23–30. crassa. Proc Natl Acad Sci USA 2009;106:22157–62.
Sakuradani E, Kobayashi M, Shimizu S. Δ6-fatty acid desaturase from an arachidonic Toida J, Fukuzawa M, Kobayashi G, Ito K, Sekiguchi J. Cloning and sequencing of the
acid-producing Mortierella fungus—gene cloning and its heterologous expression triacylglycerol lipase gene of Aspergillus oryzae and its expression in Escherichia
in a fungus, Aspergillus. Gene 1999;238:445–53. coli. FEMS Microbiol Lett 2000;189:159–64.
Sanchez JF, Chiang YM, Szewczyk E, Davidson AD, Ahuja M, Oakley CE, et al. Molecular Torres NV, Voit EO, Gonzalez-Alcon C. Optimization of nonlinear biotechnological pro-
genetic analysis of the orsellinic acid/F9775 gene cluster of Aspergillus nidulans. cesses with linear programming: application to citric acid production by Aspergillus
Mol Biosyst 2009;6:587–93. niger. Biotechnol Bioeng 1996;49:247–58.
Saruwatari T, Praseuth AP, Sato M, Torikai K, Noguchi H, Watanabe K. A comprehensive Tronsmo A, Klemsdal SS, Hayes CK, Lorito M, Harman GE. The role of hydrolytic enzymes
overview on genomically directed assembly of aromatic polyketides and macrolide produced by Trichoderma harzianum in biological control of plant diseases. In: Suomi-
lactones using fungal megasynthases. J Antibiotics 2011;64:9-17. nen P, Reinikainen T, editors. Proc 2nd Tricel Symposium on Trichoderma Cellulases
Schirmböck M, Lorito M, Wang YL, Hayes CK, Arisan-Atac I, Scala F, et al. Parallel forma- and other Hydrolases, 8. Found Biotechnol Indust Fermenation Res; 1993. p. 159–68.
tion and synergism of hydrolytic enzymes and peptaibol antibiotics, molecular Tsang A, Butler G, Powlowski, Panisko EA, Baker SE. Analytical and computational ap-
mechanisms involved in the antagonistic action of Trichoderma harzianum against proaches to define the Aspergillus niger secretome. Fungal Genet Biol
phytopathogenic fungi. Appl Environ Microbiol 1994;60:4364–70. 2009;46(Suppl 1):S153–60.
Schmoll M, Kubicek CP. Regulation of Trichoderma cellulose formation: lessons in mo- Tsuchiya K, Nagasjhiam T, Yamamoto Y, Gomi K, Kitamoto K, Umagai C. High level se-
lecular biology from an industrial fungus. A review. Acta Microbiol Immunol Hung cretion of calf chymosin using a glucoamylase prochymosin fusion gene in Asper-
2003;50:125–45. gillus oryzae. Biosci Biotechnol Biochem 1994;58:895–9.
Schmoll M, Seibel C, Kotlowski C, Vendt FWG, Liebmann B, Kubicek C. Recombinant Uusitalo JM, Nevalainen KM, Harkki AM, Knowles JK, Penttila ME. Enzyme production
production of an Aspergillus bidulans class 1 hydrophobin (DewA) in Hypocrea by recombinant Trichoderma reesei strains. J Biotechnol 1991;17:35–49.
jecorina (Trichoderma reesei) is promoter dependent. Appl Microbiol Biotechnol Vaheri MP, Vaheri MEO, Kauppinen VS. Formation and release of cellulolytic enzymes
2010;88:95-103. during growth of Trichoderma reesei on cellobiose and glycerol. Eur J Appl Micro-
Schoen C, Reichard U, Monod M, Kratzin HD, Ruchel R. Molecular cloning of an extra- biol Biotechnol 1979;8:73–80.
cellular aspartic proteinase from Rhizopus microsporus and evidence for its expres- van de Sandt EJAX, de Vroom E. Innovations in cephalosporin and penicillin produc-
sion during infection. Med Mycol 2002;40:61–71. tion: painting the antibiotics industry green. Chim Oggi-Chem Today 2000;18:
Schuster A, Schmoll M. Biology and biotechnology of Trichoderma. Appl Microbiol Bio- 72–5.
technol 2010;87:787–99. van den Berg M, Albang R, Albermann K, Badger JH, Daran J-M, Driessen AJM, et al. Ge-
Schuster E, Dunn-Coleman N, Frisvad JC, van Dijck PWM. On the safety of Aspergillus nome sequencing and analysis of the filamentous fungus Penicillium chrysogenum.
niger—a review. Appl Microbiol Biotechnol 2002;59:426–35. Nature Biotechnol 2008;26:1161–8.
Segal BH, Walsh TJ. Current approaches to diagnosis and treatment of invasive asper- van den Brink HJ, Petersen SG, Rahbek-Nielsen H, Hellmuth K, Harboe M. Increased
gillosis. Am J Respir Crit Care Med 2006;173:707–17. production of chymosin by glycosylation. J Biotechnol 2006;125:304–10.
Seiboth B, Gamauf C, Pail M, Hartl L, Kubicek CP. The D-xylose reductase of Hypocrea van den Hombergh JPTW, van de Vondervoort PJI, Fraissinet L, Visser J. Aspergillus as a
jecorina is the major aldose reductase in pentose and D-galactose catabolism and host for heterologous protein production: the problem of proteases. Trends Bio-
necessary for beta-galactosidase and cellulase induction by lactose. Mol Microbiol technol 1997;15:256–63.
2007;66:890–900. Van Dijck PWM, Selten GCM, Hempenius RA. On the safety of a new generation of DSM
Sharma R, Katoch M, Srivastava P, Qazi G. Approaches for refining heterologous protein Aspergillus niger enzyme production strains. Regulat Toxicol Pharmacol 2003;38:
production in filamentous fungi. World J Microbiol Biotechnol 2009;25:2083–94. 27–35.
Shen B. Polyketide biosynthesis beyond the type I, II and III polyketide synthase para- van Gemeren IA, Punt PJ, Drint-Kuyvenhoven A, Broekhuijsen MP, Hoog AV, Beijersber-
digms. Curr Opin Chem Biol 2003;7:285–95. gen A, et al. The ER chaperone encoding bipA gene of black Aspergilli is induced by
Shimizu S, Ogawa J, Kataoka M, Kobayashi M. Screening of novel microbial enzymes for heat shock and unfolded proteins. Gene 1997;198:43–52.
the production of biologically and chemically useful compounds. Adv Biochem Eng Venegas-Caleron M, Sayanova O, Napier JA. An alternative to fish oils: metabolic engi-
Biotech 1997;58:45–87. neering of oil-seed crops to produce omega-3 long chain polyunsaturated fatty
Siewers V, Chen X, Huang L, Zhang J, Nielsen J. Heterologous production of non-ribosomal acids. Prog Lipid Res 2010;49:108–19.
peptide LLD-ACV in Saccharomyces cerevisiae. Metab Eng 2009;11:391–7. Verdoes JC, Punt PJ, Burlingame R, Bartels J, Van Dijk R, Slump E, et al. A dedicated vec-
Sims AH, Robson GD, Hoyle DC, Oliver SG, Turner G, Prade RA, et al. Use of expressed tor for efficient library construction and high throughput screening in the hyphal
sequence tag analysis and cDNA microarrays of the filamentous fungus Aspergillus fungus Chrysosporium lucknowense. Ind Biotechnol 2007;3:48–57.
nidulans. Fungal Genet Biol 2004;41:199–212. von Dohren H. A survey of nonribosomal peptide synthetase (NRPS) genes in Aspergil-
Smith DJ, Earl AJ, Turner G. The multifunctional peptide synthetase performing the first lus nidulans. Fungal Genet Biol 2009;46(Suppl 1):S45–52.
step of penicillin biosynthesis in Penicillium chrysogenum is a 421 073 dalton protein Wan X, Zhang YB, Wang P, Jiang M. Molecular cloning and expression analysis of a
similar to Bacillus brevis peptide antibiotic synthetases. EMBO J 1990;9:2743–50. delta 6-fatty acid desaturase gene from Rhizopus stolonifer strain YF6 which can ac-
Spencer JA, Jeenes DJ, MacKenzie DA, Hyanie DT, Archer DB. Determinants of the fidel- cumulate high levels of gamma-linolenic acid. J Microbiol 2011;49:151–4.
ity of processing glucoamylase-lysozyme fusions by Aspergillus niger. Eur J Biochem Wang JW, Zheng LP, Tan RX. Involvement of nitric oxide in cerebroside-induced de-
1998;258:107–12. fense responses and taxol production in Taxus yunnanensis suspension cells. Appl
Spreer A, Ruchel R, Reichard U. Characterization of an extracellular subtilisin protease Microbiol Biotechnol 2007a;75:1183–90.
of Rhizopus microsporus and evidence for its expression during invasive rhinoorbi- Wang L, Darin R, Tingyue G, Moo-Young M. Effects of process parameters on heterolo-
tal mycosis. Med Mycol 2006;44:723–31. gous protein production in Aspergillus niger fermentation. J Chem Technol Biotech-
Sriram S, Ray RC. Trichoderma: systematics, molecular taxonomy and agricultural and nol 2003;78:1259–66.
industrial applications. In: Ray RC, editor. Microbial Biotechnology in Agriculture Wang L, Ridgway D, Gu T, Moo-Young M. Bioprocessing strategies to improve heterol-
and Aquaculture, Vol 1. Enfield: Science Publishers; 2005. p. 335–76. ogous protein production in filamentous fungal fermentations. Biotechnol Ad-
St Leger RJ, Joshi L, Bidochka MJ, Roberts DW. Construction of an improved vances 2005;23:115–29.
mycoinsecticide overexpressing a toxic protease. Proc Natl Acad Sci USA Wang SW, Ma X, Ping WX, Zhou DP. Research advances on taxol production by microbe
1996;93:6349–54. fermentation. Microbiology 2007b;34:561–5.
Stachelhaus T, Marahiel MA. Modular structure of genes encoding multifunctional pep- Wang YC, Guo BH, Miao ZQ, Tang KX. Transformation of taxol-producing endophytic
tide synthetases required for non-ribosomal peptide synthesis. FEMS Microbiol fungi by restriction enzyme-mediated integration (REMI). FEMS Microbiol Lett
Lett 1995;125:3-14. 2007c;273:253–9.
Stachelhaus T, Schneider A, Marahiel MA. Engineered biosynthesis of peptide antibi- Wang Z-Y, Soanes DM, Kershaw MJ, Talbot NJ. Functional analysis of lipid metabolism
otics. Biochem Pharmacol 1996;52:177–86. in Magnaporthe grisea reveals a requirement for peroxisomal fatty acid β-oxidation
Sternberg D, Mandels GR. Induction of cellulolytic enzymes in Trichoderma reesei by during appressorium-mediated plant infection. Mol Plant Microbe Interact
sophorose. J Bacteriol 1979;139:761–9. 2007d;20:475–91.
Stewart P, Whitwam RE, Kersten PJ, Cullen D, Tien M. Efficient expression of a Phaner- Wani MC, Taylor HL, Wall ME, Coggon P, McPhail AT. Plant antitumor agents. VI. The
ochaete chrysosporium manganese peroxidase gene in Aspergillus oryzae. Appl isolation and structure of taxol, a novel antileukemic and antitumor agent from
Microbiol Biotechnol 1996;62:860–4. Taxus brevifolia. J Am Chem Soc 1971;93:2325–7.
O.P. Ward / Biotechnology Advances 30 (2012) 1119–1139 1139

Ward M, Wilson LJ, Kodama KH, Rey MW, Berka RM. Improved production of chymosin Yamada O, Ikeda R, Ohkita Y, Hayashi R, Sakamoto K, Akita O. Gene silencing by RNA
in Aspergillus by expression as a glucoamylase-chymosin fusion. Bio-Technology interference in the koji mold Aspergillus oryzae. Biosci Biotechnol Biochem
1990;8:435–40. 2007;71:138–44.
Ward OP, Qin WM, Dhanjoon J, Ye J, Singh A. Physiology and biotechnology of Aspergil- Yoon J, Aishan T, Maruyama J, Kitamoto K. Enhanced production and secretion of het-
lus. Adv Appl Microbiol 2006:1-75. erologous proteins by the filamentous fungus Aspergillus oryzae via disruption of
Ward OP, Rao MB, Kulkarni A. Proteases. In: Schaechter M, editor. Encyclopedia of mi- the vacuolar protein sorting receptor gene Aovps10. Appl Environ Microbiol
crobiology Vol 1. Oxford: Elsevier; 2009. p. 495–511. 2010;76:5718–27.
Ward OP, Singh A. Omega-3/6 fatty acids: alternative sources of production. Process Yoon J, Maruyama J, Kitamoto K. Disruption of ten protease genes in the filamentous
Biochem 2005;40:3627–52. fungus Aspergillus oryzae highly improves production of heterologous proteins.
Ward OP. Bioprocessing. Milton Keynes: Open University Press; 1991. Appl Microbiol Biotechnol 2011;89:747–59.
Ward OP. Fermentation biotechnology: principles, processes and products. Milton Key- Yoshida T, Kato Y, Asada Y, Nakajima T. Filamentous fungus Aspergillus oryzae has two
nes: Open University Press; 1989. types of alpha-1,2-mannosidase, one of which is a microsomal enzyme that
Ward OP. Proteases. Comprehensive Biotechnology 2nd ed. Moo-Young M, editor. Vol. 3. removes a single mannose residue from Man9GlcNAc2. Glycoconj J 2000;17:745–8.
2011. pp 571–582. Zarrin M, Leeder AC, Turner G. A rapid method for promoter exchange in Aspergillus
Ward PP, May GS, Headon DR, Conneely OM. An inducible expression system for the nidulans using recombinant PCR. Fungal Genet Biol 2005;42:1–8.
production of human lactoferrin in A. nidulans. Gene 1992;122:219–23. Zhang S, Sakuradani E, Ito K, Shimizu S. Identification of a novel bifunctional 12/15 fatty
Ward PP, Peddington CS, Cunnighham GA, Zhou X, Wyatt RD, Conneely OM. A system acid desaturase from a basidiomycete, Coprinus cinereus TD#822–2. FEBS Lett
for production of commercial quantities of human lactoferrin: a broad spectrum 2007;581:315–9.
natural antibiotic. Biotechnology 1995;13:498–503. Zhang W, Li Y, Tang Y. Engineered biosynthesis of bacterial aromatic polyketides in
Watanabe CM, Wilson D, Linz JE, Townsend CA. Demonstration of the catalytic roles Escherichia coli. Proc Natl Acad Sci USA 2008;105:20683–8.
and evidence for the physical association of type I fatty acid synthases and a poly- Zheng XF, Kobayashi Y, Takeuchi A. Construction of a low-serine-
ketide synthase in the biosynthesis of aflatoxin B1. Chem Biol 1996;3:463–9. typecarboxypeptidase-producing mutant of Aspergillus oryzae by the expression
Weber G, Schörgendorfer K, Schneider-Scherzer E, Leitner E. The peptide synthetase of antisense RNA and its use as a host for heterologous protein secretion. Appl
catalyzing cyclosporin production in Tolypocladium niveum is encoded by a giant Microbiol Biotechnol 1998;49:39–44.
45.8-kilobase open reading frame. Curr Genet 1994;26:120–5. Zhou X, Zhu H, Liu L, Lin J, Tang K. A review: recent advances and future prospects of
Wong P, Walter M, Lee W, Mannhaupt G, Munsterkotter M, Mewes H-W, et al. FGDB: taxol-producing endophytic fungi. Appl Microbiol Biotechnol 2010;86:1707–17.
revisiting the genome annotation of the plant pathogen Fusarium graminearum. Zhou XW, Wei YM, Zhu HF, Wang ZN, Lin J, Liu L, Tang KX. Protoplast formation, regen-
Nucleic Acid Res 2011;39:D637–9. eration and transformation from the taxol-producing fungus Ozonium sp. Afr J Bio-
Xu J, Wang L, Ridgway D, Gu T, Moo-Young M. Increased heterologous protein produc- technol 2008;7:2017–24.
tion in Aspergillus niger fermentation through extracellular proteases inhibition by
pelleted growth. Biotechnol Prog 2000;16:222–7.

You might also like