You are on page 1of 14

Algal Research 25 (2017) 76–89

Contents lists available at ScienceDirect

Algal Research
journal homepage: www.elsevier.com/locate/algal

Review article

Microalgae and cyanobacteria as enzyme biofactories MARK


a a
Bruno dos Santos Alves Figueiredo Brasil , Félix Gonçalves de Siqueira ,
Thaís Fabiana Chan Saluma, Cristina Maria Zanetteb,c, Michele Rigon Spierb,⁎
a
Embrapa Agroenergy, Brasília, Federal District, Brazil
b
Federal University of Paraná, Technology Sector, Chemical Engineering Department, Post-Graduation Program in Food Engineering, Curitiba, Parana, Brazil
c
Midwestern State University (UNICENTRO), Food Engineering Department, Guarapuava, Parana, Brazil

A R T I C L E I N F O A B S T R A C T

Keywords: Microalgae consist of a diverse group that includes prokaryotic cyanobacteria and eukaryotic photosynthetic
Photosynthetic microorganisms microorganisms that inhabit freshwater and marine habitats. Microalgae can be used in different industries,
Enzymes including as food for human consumption, as animal feed, in aquaculture, in cosmetics and as biofuels. Because
Recombinant proteins they are photoautotrophs, with minimal nutritional requirements, microalgae have advantages compared to
Bioactive compounds
other microbial cells. An overview of the great potential of these cells to synthesize enzymes for several
Biomanufacturing
industrial applications is provided. Photosynthetic microorganism-derived enzymes include cellulases, galacto-
sidases, proteases, lipases, phytases, laccases, amylases, antioxidant enzymes and enzyme associated with
carbohydrate accumulation and the carbon concentration. Furthermore, recent reports on microalgae genomics
are revealing a variety of novel genes that should be investigated for biotechnological applications. Exploring
algal genetic diversity will also enable the efficient use of photosynthetic microorganisms as recombinant
enzyme biofactories that will be useful to industry.

1. Introduction year respectively), each with approximate global production values of


USD 40 million/year [8].
Microalgae are microscopic organisms that contain chlorophyll and The potential uses of microalgae extend beyond the products studied to
are found in freshwater and marine habitats [1]. The term “microalgae” date. Recent studies emphasize the potential of algal biomass as an
refers to prokaryotic cyanobacteria and eukaryotic photosynthetic alternative to fossil fuels as raw material for the production of biofuels
microorganisms primarily found within the taxa Chlorophyta, Rhodo- such as biodiesel, bioethanol, biohydrogen, and biomethane [9–11]. Several
phyta, Glaucocystophyta, Euglenophyta, Chlorarachniophyta, Heterokonta, reports have established that microalgae can be sources of biomass for the
Haptophyta, Cryptophyta and Alveolata [2]. production of fine chemicals production including chlorophylls, β-carotene,
Therefore, microalgae consist of a very diverse group with a astaxanthin, phycocyanin [4] and omega -3 fatty acids [12,13]. There are
significant unexplored genetic potential; the number of known species also numerous potential commercial applications of microalgae, including
represents only 1% of what is believed to exist [3–5]. Moreover, recent in the paper industry as fiber/polymer composites and as a filler material
reports on microalgae genomics are finding a variety of novel genes and [14,15], in cosmetics formulations [16], for soil restoration, as supplements
a large functional genetic diversity that may be investigated for in animal feed [17], for phycoremediation [4,18], and in therapeutic
biotechnological use [6]. supplements for their polysaccharides (β-glucan) [1], essential amino acids
Microalgae have been studied for more than 100 years, and they are [19], lipids (tri- and diglycerides, phospholipids, glycolipids), alkenes,
currently being cultivated in large scale, mainly in open pond systems. alginates (from brown algae), agar, and carragenates (from red algae)
The annual worldwide production is approximately 7,000 tons of dry [20]. Additionally, microalgae are reported to efficiently adsorb and recover
algal biomass, and the global algae biomass market is worth between heavy metals [21] and can be used in wastewater treatment in integrated
USD 3.8 billion and USD 5.4 billion. The health food sector accounts for processes [22–24].
USD 1.5 billion, and aquaculture applications account for USD 0.51 Although some reviews focus on the utilization of microalgae for
billion [7]. Dietary supplements containing Chlorella and Arthospira value products and biotechnological applications [1,9,21,25,26], none
species, which are used as dried whole microalgae, represent the largest address the potential of microalgae to be used as producers of
production volumes worldwide (2,000 and 5,000 tons of dry matter/ industrially important enzymes.


Corresponding author at: Chemical Engineering Department, Post-Graduation Program in Food Engineering, P.O. Box 19011, Zip Code: 81531-980, Curitiba, Paraná, Brazil.
E-mail address: spier@ufpr.br (M.R. Spier).

http://dx.doi.org/10.1016/j.algal.2017.04.035
Received 15 August 2016; Received in revised form 6 February 2017; Accepted 28 April 2017
2211-9264/ © 2017 Elsevier B.V. All rights reserved.
B.d.S.A.F. Brasil et al. Algal Research 25 (2017) 76–89

Microalgae have several advantages compared to other microbial prokaryotic cyanobacteria clade [46,48,49]. Using conservative ap-
cells in industrial enzyme synthesis due to their minimal nutritional proaches, the number of eukaryotic algal species was estimated to be
requirements (natural or artificial light, CO2, water, nitrogen source, 72,500 [50] and the number of cyanobacteria species was estimated at
and some salts), which reduce cost. Nevertheless, microalgae can have approximately 6,000 [51].
disadvantages for enzyme production, such as longer cultivation time Therefore, simple algal morphologic features hide a huge polyphy-
compared to bacteria, yeast or fungi and additional stages downstream letic genetic reservoir, orders of magnitude larger than that found
to recover intracellular enzymes. Furthermore, the majority of biomo- within animals or plant taxa [52–54]. For instance, while Homo sapiens,
lecules are produced intracellularly in small quantities, which makes Mus musculus and Danio rerio present 52.8% of their gene sets as
analysis and industrial scale-up difficult. However, recent advances in orthologs, only 16.3% of orthologous genes are found among Chlamy-
cell lysis techniques have provided a high extraction yield of several domonas reinhardtii, Thalassiosira pseudonana and Cyanidioschyzon mer-
metabolites from microalgae [27,28]. olae genomes [6].
This review focuses on the great capacity of microalgae cells to Uncovering algal genetic diversity will enable access to novel genes,
synthesize enzymes for different industrial applications. The enzymes including industrially valuable enzyme-encoding genes, for future
reported included cellulases, amylases, galactosidases, proteases, li- biotechnological exploration. There are approximately 14,500 anno-
pases, phytases, laccases, antioxidant enzymes, and enzymes involved tated sequences from enzyme-coding genes derived from algae species
in carbohydrate accumulation and carbon concentration. Bioprocesses available in GenBank at present (Figure 1). Furthermore, considerable
for enzyme production are addressed. Separation processes are also advances in algal genomics and in silico predictive capacity are being
discussed, microalgae cell walls are rigid and require specific unit made [55]. There are over 60 eukaryotic microalgae and more than 40
operations to ensure lysis (cell breaking) while preserving the integrity cyanobacteria genome sequencing projects either published or under-
of the released cytosolic or membrane-associated enzymes. Finally, way (www.ncbi.nlm.nih.gov/genomes; http://genome.jgi.doe.gov)
recent advances in recombinant enzyme expression in microalgae and [6,55,56]. Bioinformatic tools, such as Kyoto Encyclopedia of Genes
cyanobacteria are discussed, including the model species and genetic and Genomes (www.genome.jp/kegg), BioModels databases (www.ebi.
tools available. ac.uk/biomodels/), CpBase (Chloroplast Genome Database), MitoBase
(Mitochondria Genome Database), and MMETSP (Marine Microbial
2. Parameters for microalgae cultivation Eukaryote Transcriptome Sequencing Project) are enabling the predic-
tion and characterization of gene regulatory pathways and the annota-
The primary factors of importance in microalgae cultivation process tion of de novo genomes of algal species. In addition, an increasing
are light, nutrients, aeration, temperature and pH. For maximal biomass number of transcriptomics, proteomics, and other systems biology
yield, the microalgae culture must be supplied with macronutrients and studies are being reported [25,57]. There are also several in silico
micronutrients in adequate amounts. Macronutrient requirements analysis platforms, such as The Greenhouse (https://greenhouse.lanl.
include carbon, nitrogen, phosphorus, sulfur, potassium (also silicon gov/greenhouse/), pico-PLAZA (http://bioinformatics.psb.ugent.be/
for diatoms), and trace elements as minerals (e.g., Co, Mo, Mn-enzymes pico-plaza/), Phytozome (https://phytozome.jgi.doe.gov/pz/portal.
cofactors) and vitamins (cyanocobalamin, thiamin and biotin) [29,30]. html), IMG - Integrated Microbial Genomes (https://img.jgi.doe.gov/
As the carbon source in photoautotrophic microalgae metabolism, ), CyVerse (http://www.cyverse.org/), KBase (https://kbase.us/),
most prefer to utilize free CO2. In an alkaline culture medium, some EDGE bioinformatics (https://bioedge.lanl.gov/edge_ui/), Galaxy
microalgae such as Chlorella, Dunaliella and Arthospira can assimilate (https://galaxyproject.org/) and Ergatis (http://ergatis.sourceforge.
bicarbonate [30]. In heterotrophic growth, microalgae grow in the net/), that can allow the identification of novel algal encoded variants
dark, and cells obtain energy exclusively from organic carbon in the (i.e., gene orthologs) from known bacterial or fungal encoded enzymes.
media. In mixotrophic growth, microalgae can obtain energy from both These approaches could also help to identify key biosynthetic genes
light and different organic carbonic sources including glucose, glycerol, for the production of high-value products in microalgae, which could in
acetate and fructose [31,32]. turn be altered using molecular tools [55,56,58,59]. An example is the
Few studies have estimated the influence of medium composition on genetic transformation of the carotenoid-producing microalgae Haema-
enzyme production by microalgae. It was observed in Dunaliella salina tococcus pluvialis with an additional version of the phytoene desaturase
cultures that altering the nitrogen source in the medium can increase gene, which led to a 26% increase of its astanxanthin content [60].
the activity of specific enzymes [32]. Guzmán-Murillo et al. [33] Another example is the expression of the phytoene synthase genes
evaluated different agricultural fertilizers as nitrogen sources to scale derived from Dunaliela salina and Chlorella zoofingensis in transgenic
up the photoautotrophic production of superoxide dismutase (SOD) by Chlamydomonas reinhardtii, which increased lutein content more than 2-
Phaeodactylum tricornutum. The maximum concentration of intracellular fold [61,62].
SOD produced was 79 and 72 U mg− 1 protein using media containing In addition, the advent of flexible and simple new gene-editing
nitrogen from ammonium sulfate and urea, respectively. These results tools, such as CRISPR/CAS9 technology, offers promising opportunities
indicate that an alternative nitrogen source can be used for the in eukaryotic microalgae metabolic engineering. The CRISPR/CAS9
synthesis of SOD at reduced costs. Table 1 summarizes parameters system consists of a specific guide RNA (gRNA) and a non-specific
related to the production of microalgae enzymes described in the CRISPR-associated endonuclease protein (Cas9). The Cas9 endonu-
literature. clease promotes a double strand break (DSB) in the DNA loci targeted
by the gRNA sequence. After the DSB, the cell’s DNA repair mechanism
3. Algae genetic diversity and potential for enzyme production either promotes a non-homologous end joining, which usually leads to
an InDel mutation, or promotes a homology-directed repair that
Photosynthetic organelles (plastids or chloroplasts) were primarily replaces the targeted sequence with a co-transformed exogenous
derived from the endosymbiotic uptake of a cyanobacterium. sequence [63]. Recent studies have reported the successful use of
Subsequently, these primary plastids (such as those found in chlor- CRISPR/CAS9 systems in Phaeodactylum tricornutum, Chlamydomonas
ophytes and rodophytes) were spread to other eukaryotic clades reinhardtii, Nannochloropsis spp. and Thalassiosira pseudonana, establish-
through subsequent rounds of secondary and even tertiary endosym- ing novel paths for genomic sequence editing in microalgae [64–68].
biosis [46,47]. These evolutionary events led to the presence of the
“algal phenotype” characterized as photosynthetic unicellular or plur- 4. Microalgal enzymes
icellular (without tissue organization) organisms found in humid
environments within 12 of the 45 major eukaryotic clades and, in the Enzymes from microorganisms could be used as catalysts in diverse

77
B.d.S.A.F. Brasil et al.

Table 1
Culture conditions and disruption methods for recovering intracellular enzymes from microalgae

Microalgae/cyanobacteria Enzyme Culture conditions Disruption method References

Synechococcus lividus SKP50, S. lividus DSK74, S. Phytase Cultivation at 50 °C under illumination of 2,500 lux. Approximately Not described Klanbut and
bigranulatus Skuja and Chroococcidiopsis phosphate-free medium containing 0.1% (w/v) calcium phytate as the Peerapornpisarn [34]
thermalis geitler sole phosphorus source supplemented with 0.5% (w/v) glucose and 0.02
% (w/v) casamino acids
Poterioochromonas malhamensis peterfi α-galactosidase Nutrient medium containing glucose (10 mg/mL), aerated at 27 °C, and French press and sonicator Dey and Kauss [35]
illuminated with one daylight fluorescent tube at a distance of
approximately 35 cm (2 klux) for approximately 1 h
Anabaena variabilis ATCC 29413 Protease Photoautotrophic with N2 as the nitrogen source Thawed in a buffer and passed through a French press Lockau et al. [36]
at 140 MPa
Anabaena variabilis Prolylendopeptidase Photoautotrophic with N2 or NH4NO3 as nitrogen source in continuously Sonication and centrifugation at 48, 000g for 30 min Strohmeier et al. [37]
illuminated batch culture
Anabaena variabilis Aminopeptidase Grown at ambient temperature under constant illumination at 1,300 lux, Thawed in a buffer and passed through a French press Niven [38]
and cultures were bubbled with air. at 80 MPa
Arthrospira platensis Protease Stirring at 4 °C overnight in buffer pH = 8, Nanni et al. [39]

78
Grown at 30 °C in Erlenmeyer flasks, continuously shaken and aerated
with CO2-enriched (5%) air, irradiance of 70 mmol photon m–2 s–1 centrifugation at 30,000g for 1 h
A. platensis Protease Not described Citrate buffer 50 mM, pH 5 at room temperature for Yada et al. [40]
1 h followed by centrifugation at 14, 000g for 30 min
Porphyridium cruentum SOD Photoautotrophic in salt medium under aeration Thawed in a buffer and passed through a French press Misra and Fridovich [41]
at 20,000 psi
Anabaena cylindrica SOD Grown aerobically in nitrogen-free BG 11 medium at 25 °C and at a Cells were resuspended in potassium phosphate buffer Canini et al. [42]
photon fluence rate of 25 μmol m− 2 s− 1 (pH 7.8) containing EDTA and sonicated in an ice-
bath.
A. platensis SOD Grown at 25 °C under the light intensity of 28 μmol photons m− 2 s− 1 in Sonication operated with Gunes et al. [43]
Pseudanabeana sp. glass bottles for 10 days 80 % power output for 10 min over 8 pulsation cycles
Synechococcus nidulans
Botryococcus sudeticus Lipase Grown in various oils (canola, corn, palm and olive) in modified Bold’s None (lipase is extracellular) Yong et al. [44]
basal medium at ambient temperature with 24 h of synthetic white light.
Nannochloropsis oceanica Lipase Grown in seawater enriched with F/2 medium nutrients at 120 rpm, 20 °C Sonication at 0 °C in 5 1-min cycles, with a cooling Savvidou et al. [45]
and pH 8.0. Continuous illumination at 100 μmol photon m− 2 s− 1 on the time of 1 min between cycles. Centrifugation at 4 °C
culture surface. and 7,000g for 10 min
Algal Research 25 (2017) 76–89
B.d.S.A.F. Brasil et al. Algal Research 25 (2017) 76–89

Figure 1. Number of enzyme-coding genes from algal species. Graphics represent the numbers of predicted enzyme-coding genes sequences from eukaryotic algae and cyanobacteria
deposited in GenBank (http://www.ncbi.nlm.nih.gov/gene/) by November, 2016. The six major enzyme classes (hydrolases, oxidoreductases, transferases, ligases, isomerases and lyases)
are represented by colored bars.

industrial processes. The search for new sources of microbial enzymes is Anabaena strains were reported as potential organisms that produce
ongoing and requires sustainable solutions [69]. In 2014, the global hydrolytic enzymes with possible fungicidal activity [77,80,81]. One
market for industrial enzymes was estimated at USD 4.2 billion and is research group identified chitosanase homologs in A. iyengarii and A.
expected to reach nearly USD 7.1 billion by 2018 [70]. laxa. The cyanobacterial strain A. laxa showed putative genes (end 1
Although there is no industrial production of enzymes from micro- and end 2) encoding β-1,4-endoglucanase, which is in the peptidase
algae, several reports show the great capacity of microalgae cells to family, and glycoside hydrolase [77]. The algal polysaccharidases
synthesize enzymes. Different classes of enzymes were reported, (marine carbohydrase in CAZymes classification) identified to date
including hydrolases, oxidoreductases and lyases. (agarases, carrageenases, and alginate lyases) display unique structures
and biochemical features and are distantly related to the currently
4.1. Cellulases known glycoside hydrolases [82]. These findings highlight the potential
for new and original biotechnological uses of these enzymes.
Cellulases, hemicellulases and pectinases can be collectively re-
ferred to as holocellulases; these enzymes are capable of degrading the 4.2. Amylolytic enzymes
cell wall carbohydrate polymers of plants or algae [71]. This enzyme
group is usually found in microbial decomposers of plant and algal Amylases are a group of enzymes that promote the hydrolysis of
organic matter, e.g., filamentous fungi, on soil and in fresh or salt starch, oligosaccharides, and polysaccharides. The best-known amy-
water. Cellulose degradation by fungi, for example, occurs via the lases are α-amylase (EC 3.2.1.1), β-amylase (EC 3.2.1.2), glucoamylase
synergistic activity of three types of cellulases: endoglucanases (EC (E.C 3.2.1.3), isoamylase (EC 3.2.1.68) and glucosidases (EC 3.2.1.20)
3.2.1.4; endo-β-1,4-glucanases), cellobiohydrolases (EC 3.2.1.91) and [83].
β-glucosidases (EC 3.2.1.21). The main potential applications of Levis and Gibbs [84] reported α-amylase activity in C. reinhardtii
cellulases are in the food (to break down non-digestible carbohydrates), grown photoautotrophically in 12-h/12-h light/dark cycles. Amylase
animal feed (enhancing fiber degradation), textile (biostoning for jeans reached maximum activity after 4 h of darkness but was active through
and biopolishing for cotton), biofuel (cellulosic ethanol), and chemical the entire cell cycle.
industries [71,72]. Rismani- Yazdi et al. [85] and Shang et al. [86] studied genes that
Algae and microalgae are the major contributors to the production encode enzymes involved in the catabolism of starch in the Dunaliella
of primary energy in the aquatic environment. Some organisms (e.g., genus. Two different pathways were identified, a phosphorolytic and a
Euglena) produce polysaccharides for carbon storage such as paramylon hydrolytic pathway. In D. tertiolecta, the enzymes associated with
and require β-1,3-glucan hydrolase to metabolize the accumulated hydrolytic metabolism are α-amylase and oligo-1,6-glucosidase. α-
carbohydrates [73]. Amylase and β-amylase were identified in D. parva. These three
Aquatic microorganisms are the primary decomposers of polysac- enzymes are responsible for starch metabolism to produce D-glucose.
charides-storage by algae. The degradation of these sugars are per- For the phosphorolytic degradation of starch, glycogen phosphorylase
formed by enzymatic-path microorganisms (CAZymes), thereby pro- was found in D. parva, and starch phosphorylase was found in D.
moting a major carbon cycling processes that is established in the tertiolecta.
Earth’s ecological cycles [74]. Cellulases comprise a group of enzymes
known to exhibit features of synthesis (chitosan) or the degradation of 4.3. Galactosidases
poly-oligo-dimers saccharides [75,76]. However, few manuscripts re-
port the production of cellulases, hemicellulases, and pectinases by α-D-galactoside galactohydrolase (EC 3.2.1.22), also known as α-
microalgae to degrade these polysaccharides in aquatic environments, galactosidase, hydrolyzes α-1,6-linked galactose residues found in
even though microalgae and algae possess genes encoding these oligosaccharides and galactomannan polysaccharides and releases α-
enzymes [77–79]. D-galactose [87]. α-Galactosidases are useful in the paper and pulp,

79
B.d.S.A.F. Brasil et al. Algal Research 25 (2017) 76–89

sugar, and food and animal feed industries [88]. tri-peptides, whereas the smaller enzyme was capable of hydrolyzing
Intracellular α-galactosidase (α-gal) activity has been detected in longer oligopeptides.
Poterioochromonas malhamensis, unicellular golden-brown algae that Protease production from Arthospira platensis was reported by Nanni
exhibit a mechanism of osmotic regulation. P. malhamensis α-gal et al. [39] and Yada et al. [40]. In the first study, a novel serine protease
presents optimal activity at pH 7.0 in cell-free extracts. The intracellular associated with the selective proteolysis of phycobiliproteins was
enzyme activity increases under conditions of high external osmotic isolated, purified and partially characterized. Yada et al. [40] purified
pressure. This condition causes cell shrinkage within a few minutes. As a new arginine-specific protease (Sp-protease) that hydrolyzed gelatin
a mechanism of recovery, the cell biosynthesizes intracellular isoflor- and fibrin, however, phycocyanin was not hydrolyzed. Both enzymes
idoside (α-galactosylglycerol). Higher external osmotic pressure in- from A. platensis had a molecular weight of 80 kDa and act indepen-
duces the production of α-galactosidase, which then reduces the dently of Ca+ 2.
intracellular concentration of isofloridoside (O-α-D-galactopyranosyl- Because studies have reported the ability of microalgae to produce
(1 → 1) glycerol), thus releasing glycerol and galactose. Some of the proteases and because production is associated with the availability and
carbon released is incorporated into β-glucans reserves in the algal cell nature of the nitrogen source [99], future studies should evaluate
or as wall components [35]. Isofloridoside apparently acts as a changes in the nitrogen source in the medium, to induce protease
substrate to induce α-gal catalysis when the osmotic pressure outside activity in microalgae.
the cell causes a rapid decrease in its internal levels. The formed
products are used by the cell as osmoregulators or possibly as carbon 4.5. Lipases
sources. Galactose and glycerol accumulate within the cell as an
osmoregulatory mechanism to maintain vital cellular activities. Lipases (triacylglycerol acyl hydrolases, E.C. 3.1.1.3) are enzymes
β-Galactosidase (β-D-galactohydrolase, EC 3.2.1.23), also called that naturally hydrolyze triglyceride into fatty acids and glycerol.
lactase, hydrolyzes D-galactosyl residues from oligosaccharides, poly- However, in aqueous-restricted environments, they can catalyze ester-
mers and secondary metabolites [89]. The main industrial application ification, transesterification, inter-esterification, and aminolysis
has been the hydrolysis of lactose in milk and dairy products. More [100–102].
recently, β-galactosidases with transgalactosylation activities have been Lipases are among the most important enzymes for biotechnological
extensively exploited for the synthesis of galactooligosaccharides, applications. They are useful in the detergent, food, flavor, pharma-
which are used as probiotic food ingredients [90,91]. ceutical, chemical, agrochemical, and cosmetics industries [103].
Davies et al. [92] tested macro and microalgae species for β- Few microalgal lipases and genes encoding lipases have been
galactosidase activity. Eight of nine species from the Chlorophyceae investigated, compared to bacterial, fungal, animal and plant lipases.
and Cyanophyceae families presented some β-D-galactosidase activity. A lipase from the photosynthetic cyanobacterium A. platensis was
High enzyme activity was shown in Cosmarium sp. and Acutodesmus isolated and characterized for the first time by Demir and Tukel
obliquus. Girard et al. [93] tested cheese whey permeate as a partial [104]. The lipase presented a pNPP hydrolyzing activity of 45 U
substitute for culture medium in the mixotrophic cultivation of A. mg− 1 protein after being purified 375-fold, and the protein is mono-
obliquus. Lactose was metabolized by microalgae, thus promoting a meric and specific for the 3-position in the ester bond. Godet et al.
reduction in lactose concentration and an accumulation of glucose and [105] isolated a new gene from the microalgae Isochrysis galbana that
galactose in the culture medium and, indicating the extracellular encode a 49-kDa lipase of 457 amino acids. The deduced protein shares
production of β-galactosidase by microalgae. similarities with known lipases. Phylogenetic analyses showed simila-
Several studies reported α-galactosidase [94,95] and β-galactosidase rities to fungal lipase sequences.
production from yeast, molds and bacteria using whey and other agro- In the past ten years, the main microalgae species that has been used
industrial wastes as substrates. These agro-industrial byproducts could as a model to understand lipid metabolism in green microalgae is
be potential inducers of galactosidase synthesis by microalgae. α- Chlamydomonas reinhardtii. Many putative lipases are encoded in the
Galactosidase inducers included D-galactose, melibiose, stachyose and Chlamydomonas genome. As with many other algae, the photosynthetic
raffinose. The β-galactosidase inducer is lactose. Future work is microalgae C. reinhardtii accumulates triacylglycerols (TAGs) when
required to increase knowledge about the importance of α- and β- deprived of nitrogen, a condition that causes genome-wide transcrip-
galactosidases in the metabolism of microalgae. tional changes. Among the genes regulated by nitrogen deprivation are
numerous genes encoding lipases [106]. Li et al. [107] described the
4.4. Proteases identification of a mutant from C. reinhardtii with a lesion in a
galactoglycerolipid lipase-encoding gene. This gene is one of the
Proteases constitute a wide group of enzymes that catalyze peptide- lipase-encoding genes that is upregulated when microalgae are de-
bond cleavage in proteins and peptides [96]. Proteases have a wide prived of nitrogen. Disrupting a galactoglycerolipid lipase-encoding
range of metabolic functions and industrial applications, primarily in gene caused a reduction of TAG content and galactoglycerolipid
the detergent, pharmaceutical and food industries [97]. hydrolysis. Mansfeldt et al. [108] also confirmed that a galactoglycer-
In microalgae metabolism, protease activity increases during envir- olipid lipase gene from a Chlorella sp. strain is upregulated in these
onmental stress such as light or nutrient limitation and cell apoptosis. conditions. In C. reinhardtii, one of the downregulated genes was
Proteolysis also plays a role in the control of organelle senescence and recently shown to encode a lipase implicated in TAG hydrolysis
heterocyst formation in cyanobacteria [98]. [109]. They identified the lipase-encoding gene CrLIP1, and they
Lockau et al. [36] described a calcium-dependent serine protease suggested that the genes repressed during TAG accumulation might
from the cyanobacteria Anabaena variabilis. The enzyme is a trypsin-like encode lipases that hydrolyze TAG, DAG (diacylglycerol) or MAG
protease that, requires calcium for full activity. Using A. variabilis (monoacylglycerol). When the quantity of the CrLIP1 transcript was
mutant IM 141, which lacks this calcium-dependent protease, Stroh- reduced, there was a delay in TAG hydrolysis upon nitrogen resupply,
meier et al. [37] showed that A. variabilis contains a second soluble confirming that CrLIP1 is involved in Chlamydomonas TAG lipolysis.
trypsin-like protease, a prolylendopeptidase. The data obtained suggest that the lipase encoded by CrLIP1 hydrolyses
The first report of the purification of aminopeptidases (EC the DAG formed from TAG lipolysis, facilitating TAG turnover.
3.4.11.15) from cyanobacteria was performed by Niven using A. Recently, Savvidou et al. [45] produced and characterized a lipase
variabilis (1995). Two enzymes (188 kDa and 59k Da) were separated from Nannochloropsis oceanica. The microalga was grown in seawater
by gel filtration chromatography and showed different substrate enriched with nutrients. The cultures were incubated at 20 °C and pH 8
specificity profiles. The larger enzyme showed specificity for di- and with 24 h of synthetic light. Lipase was extracted by sonication and

80
B.d.S.A.F. Brasil et al. Algal Research 25 (2017) 76–89

characterized. This lipase showed hydrolysis activity against pNP esters coccoid green algae T. aeria produces an extracellular laccase-like
of fatty acids with different chain sizes, and the optimum pH was 7. enzyme that can oxidize phenolic substances and the anthraquinonic
Yong et al. [44] also reported the production of lipase from a microalga dye Abu62 [121]. Confirmation of this enzyme as a laccase indicates
strain. They produced, purified, and characterized an extracellular that algal laccases might contribute to the bio-transformation of several
lipase from Botryococcus sudeticus. They studied the effect of different natural and xenobiotic aromatic compounds [126]. Species of the
oils (palm, canola, corn and olive) in the culture medium and the effect “Moewusinia” clade, including Chlamydomonas moewusii and T. aeria,
of agitation on lipase production. Continuous illumination was main- secrete putative laccases that might help algae survive in harsh
tained with synthetic white light and cultures were kept at room environments [127].
temperature. Olive oil in the medium and agitation provided the best The laccases of green algae have been reported as biomolecules
conditions to produce lipases. The lipase was purified and presented a capable of breaking down phenolic pollutants in contaminated surface
molecular mass of 120 kDa and maximal activity at 50 °C and pH 10. waters, biodegrading various xenobiotics and textile dyes from water
The lipase also has good thermal stability, showing its potential to be [128–131]. In addition, algae have been shown to effectively treat
applied in different industries. wastewaters from the paper industries without requiring co-substrates
[132].
4.6. Phytases
4.8. Antioxidant enzymes
Phytases (myo-inositol hexakisphosphate phosphohydrolase) initi-
ate the removal of phosphate residues from phytate, the principal Reactive oxygen species (ROS) are generated in aerobic organism as
molecule for organic phosphorus (P) storage in grains and plant seeds result of respiration and substrate oxidation. Environmental stresses
[110]. Phytases can be added to animal feed to increase the bioavail- such as intense light, heavy metals, herbicides, UV radiation, high salt
ability of P, and recent studies have shown the potential use of phytases concentrations, and extreme temperatures stimulate ROS production.
in food processing [111,112]. Phytases are classified into the following Consequently, microalgae possess antioxidant defense mechanisms that
three types depending on the position of phytate dephosphorylation combat ROS cell damage. Enzymatic antioxidant defenses include SOD,
initiation of the phytate: 3-phytases (E.C 3.1.3.8), 6-phytases (E.C glutathione reductase, catalase and peroxidase [133].
3.1.3.26) and 5-phytases (E.C 3.1.3.72) [113].
Klanbut and Peerapornpisarn [34] found phytase activity in the
intracellular fraction of the following four species of thermotolerant 4.8.1. Superoxide dismutase
blue-green algae isolated in Thailand: Synechococcus lividus SKP50, S. Superoxide dismutase (EC 1.15.1.1) are metalloenzymes that con-
lividus DSK74, S. bigranulatus Skuja, and Chroococcidiopsis thermalis vert superoxide radicals (O2•) into oxygen (O2) and hydrogen peroxide
Geitler. The maximum enzyme activity was 1.83 mU mL− 1, which was (H2O2). The applications of SOD include therapeutic and prophylactic
achieved by S. lividus SKP50. Phytase activity was not found in the applications in humans, in the preservation of biological materials
extracellular culture broth. (organs for transplantation and sperm), in the preservation of perish-
Further studies could be performed to assess whether microalgae able materials such as foodstuffs and vaccination agents and as an
are capable of producing phytase and whether the enzyme is extra- antigenic agent for the serodiagnosis of pathogens [134].
cellular or cell disruption is necessary for recovery. In addition, medium SODs are present in prokaryotic and eukaryotic cells, but relatively
composition can be assessed in future studies to induce the synthesis of little information is available regarding SOD activity in microalgae
enzymes using specific substrates (such as the organic phosphorus [135]. Misra and Fridovich [41] purified a Mn-SOD from the micro-
source). It has been reported that phytic acid stimulates microbial algae Porphyridium cruentum. The enzyme had a molecular weight of 40
phytase synthesis [114]. kDa and was stable during freezing and thawing.
Canini et al. [42] report the purification of Fe-SOD from the
4.7. Laccases nitrogen-fixing cyanobacteria Anabaena cylindrica. Gunes et al. [43]
investigated and compared SOD activities in S. nidulans, A. platensis and
Typical laccases (EC 1.10.3.2) are extracellular monomeric glyco- Pseudanabeana sp and found the specific activities of 50.4 U mg− 1,
proteins that belong to the blue multicopper oxidase family that 30.0 U mg− 1, and 18.4 U mg− 1, respectively. Guzmán-Murillo et al.
includes ascorbate oxidase, bilirubin oxidase, ceruloplasmin, and others [33] tested different nitrogen medium composition in the scaled up
[115]. Laccases can promote the oxidation of complex polymeric production of SOD by Phaeodactylum tricornutum and concluded that
structures such as lignin into phenolic compounds [116]. These SOD production by microalgae is comparable to that by the yeast
enzymes are considered ideal “green catalysts” since they can oxidize Debaryomyces hansenii.
a wide variety of compounds using ambient O2 from the air and Because SOD is a promising and potent antioxidant enzyme, future
releasing H2O as the only byproduct [115]. studies should evaluate SOD synthesis in microalgae.
The natural functions of laccases include the degradation and
biosynthesis of lignin [117]. They have great relevance as models for 4.9. Enzymes involved in carbohydrate accumulation
structure/function relationships and as sustainable tools for industrial
processes, demonstrating many applications in the food (brewing, color Microalgae show the ability to metabolize fatty acids into carbohy-
enhancement in tea, cork modification), textile (denim bleaching and drates, which is a useful metabolic advantage. González-Fernández and
finishing), and pulp and paper industries (paper pulp delignification Ballesteros [136] reported the main enzymes for carbohydrate storage
[118]. Laccases are involved in morphogenesis processes, detoxification in microalgae. The enzymes responsible for this conversion are
(soil), immunity and plant pathogenicity and bioremediation processes isocitrate lyase (EC 4.1.3.1), which is also known as isocitrate glyox-
[119]. ylate-lyase (succinate-forming) or isocitrase. This enzyme participates
Laccases and laccase-like enzymes are found in insects, plants, in the decarboxylation step of the tricarboxylic acid cycle and is used by
microorganism (white-rot fungi), prokaryotes and lichens [120]. Ad- bacteria, fungi, plants and microalgae. This enzyme acts by cleaving
ditionally, laccase activity has been described in the soil algae isocitrate into succinate and glyoxylate. Another enzyme involved in
Tethacystis aeria [121] and Shewanella xiamenensis, which show laccase carbohydrate accumulation is malate synthase, also known as acetyl-
activity on the substrate 2′-azino-bis-3-ethylbenzthiazoline-6-sulfonic CoA: glyoxylate C-acetyltransferase. It catalyzes the reaction of glyox-
acid (ABTS) [122]. The Shewanella genus is capable of dissimilatory ylate and acetyl-CoA to form malate. After malate enters the cytosol,
manganese and iron oxide reduction ([123–125], apud [122]). The subsequent reactions occur to produce glucose [136].

81
B.d.S.A.F. Brasil et al. Algal Research 25 (2017) 76–89

4.10. Enzymes involved in carbon concentration insect expression systems usually exhibit scaling-up technical difficul-
ties and prohibitive costs for industrial enzymes production [153].
Carbonic anhydrase (EC 4.2.1.1) is a zinc-dependent metalloenzyme Plant-based recombinant protein expression systems are attractive
that catalyzes the formation of carbonic acid from CO2 and H2O [137]. because of their scalability and low cost. However, their long-life cycles
The importance of carbonic anhydrase in carbon fixation metabolism is and issues related to product recovery from plant tissues/organs make this
being intensely investigated in photosynthetic organisms to increase method non-ideal for heterologous enzymes expression [148,154,155].
biofuel production [138]. A study reported microalgal isolates from the However, photosynthetic microorganisms combine features of both micro-
genera Desmodesmus, Kirchneriella and Acutodesmus, which exhibit bial- and plant-based systems that are desirable for recombinant protein
constant high carbonic anhydrase activity during cultivation in closed expression, including high growth rate, ease of cultivation, rapid scalability
photobioreactors at ambient temperature with 0.03% CO2. In higher at the pilot level, protein secretion, and the absence of human pathogens.
levels of CO2, the activity decreases significantly [139]. The authors Furthermore, eukaryotic algae species are able to perform post-translational
observed that CO2 levels modulate the carbonic anhydrase activity. The modifications [58,146,149,154–159].
increase in enzyme activity correlates with the decrease in free CO2. To date, diverse proteins of industrial, nutritional and medical use
Moreover, the increase in carbonic anhydrase activity also correlates have been successfully expressed in photosynthetic microorganisms
with a decrease in bicarbonate, suggesting its conversion to CO2 by this (Table 3). Highlights include the use of algae to produce therapeutic
enzyme. The study also suggests that a CO2-rich environment increases proteins. Human antibodies, for example, are complex polypeptides
the availability of dissolved inorganic carbon (free CO2 and HCO3-) and that require disulfide bond formation and proper folding, which cannot
modulates the activity of carbonic anhydrase [139]. be achieved in prokaryotic systems. In addition, issues related to
hyperglycosylation have hampered advances in antibody production
5. Disruption methods for enzymes recovery in yeast [176]. Therefore, algae appear to be attractive alternatives to
the costly mammalian cell cultures. Accordingly, the successful expres-
The steps involved in the production of intracellular metabolites sion of functional human antibodies against Herpes Simplex Virus and
from microalgae start with cell cultivation and biomass recovery Anthrax in C. reinhardtii chloroplasts has been shown [172,173].
(flocculation, centrifugation, and/or filtration) and are followed by Immunotoxins have also been expressed in C. reinhardtii chloro-
downstream processing, which includes cell lysis, extraction and plasts [174,175]. These therapeutic proteins are formed by recombi-
product purification [140]. The downstream recovery of microalgal nant antibodies attached to small toxin molecules/proteins, which
products is responsible for 57% of the cost of the final product [141]. target and kill cancer cells. Usually, these toxins cannot be produced
Different disruption methods have been used for microalgae, such as in mammalian expression platforms since they inhibit cell metabolism.
mechanical and chemical methods and enzymatic hydrolysis. Therefore, algae chloroplasts are an adequate environment for the
Mechanical methods are preferred since they prevent chemical con- expression of these antibody–drug conjugates since they possess
tamination, are easy to scale up and preserves the functionality of prokaryotic-like cell machinery and yet can properly fold human
bioproducts [142]. However, these methods usually have higher energy antibodies. The C. reinhardtii chloroplast is also uniquely suitable for
requirements than chemical or enzymatic methods. Because heat is the expression of surface proteins of the malarial parasite, Plasmodium
released during mechanical disruption, a cooling system is needed for spp., which are not glycosylated but have several disulfide bonds
thermolabile products [143]. High-pressure homogenization, ultrasoni- [169–171].
cation, high-speed centrifugation and bead milling are the most Furthermore, there is a growing body of studies reporting the
extensively used methods for enzyme recovery on a laboratory scale, expression of recombinant enzymes for industrial use in microalgae
as shown in Table 1. Serive et al. [28] tested nine mechanical disruption [160–162,177]. For example, α-galactosidases have been expressed
techniques on microalgae cells, and a mixer mill showed the most from genes derived from Lactobacillus acidophilus and Trichoderma reesei
efficient cell disruption and recovery of metabolites. inserted into the chloroplastic genomes of Dunaliella tertiolecta and
Chlamydomonas reinhardtii [161]. Additionally, two phytase-coding
6. Algae and cyanobacteria as recombinant enzyme expression genes, from Escherichia coli and Caulobacter crescentus have been
systems expressed in the chloroplasts of D. tertiolecta and C. reinhardtii
[161,162].
The characterization of new algal genomes coupled with genetic Yoon et al. [162] demonstrated that the use of whole cells of
tool development has opened paths for engineering these organisms transgenic C. reinhardtii expressing the E. coli phytase gene as direct
into platforms for recombinant protein production, including enzymes. feed additives for young broiler chicks resulted in higher phosphorous
The use of genetically modified organisms to produce industrial adsorption. Therefore, even though recombinant proteins expressed
enzymes was the main factor driving the expansion of this market from from plastid genomes remain confined within this organelle, the costs
USD 1.6 billion in 1998 to USD 5.1 billion in 2009 [195]. Currently, of enzyme recovery and purification can be reduced in some cases. The
~ 50% of industrial enzymes are produced using recombinant DNA gene encoding β-1,4-endoxylanase fromTrichoderma reesei has been
technology [144]. Although conventional microbial systems (e.g., E. expressed in the chloroplasts of D. tertiolecta and C.reinhardtii [161]
coli, S. cerevisiae and P. pastoris) and animal cells have well established and in the nuclear genome of C. reinhardtii [160]. Rasala and
molecular tools and have been used for several years to produce collaborators (2012) have developed a plasmid construct that allowed
recombinant proteins, they exhibit significant drawbacks mainly re- high expression from the nuclear genome by fusing the xylanase gene to
lated to protein processing, solubility and production costs (Table 2) the selection marker gene with the foot-and-mouth-disease-virus 2A
[144,146,149]. self-cleavage peptide coding sequence between these. In this same
For example, although bacterial host systems are cheap and easily study, the secretion signal sequence from the Arylsulfatase I gene
operated, they lack eukaryotic post-translational modification machin- (ARS1) of C. reinhardtii was fused to the xylanase gene leading to the
ery and may require laborious and costly processing steps for protein release of the bioactive enzyme in the surrounding media. In addition,
purification from inclusion bodies [150]. Although yeast and filamen- Synechocystis sp. 6803 has been used for the expression of a D-lactate
tous fungi based systems have basic eukaryotic machinery and the dehydrogenase-coding gene derived from E. coli [178].
capacity for protein secretion, recombinant protein inactivity due to
issues with fungi-specific post-translational patterns, high concentra- 7. Eukaryotic microalgae as recombinant protein biofactories
tions of intracellular proteases, and insufficient secreted protein yields
have been reported [146,151,152]. In addition, mammalian cell and Eukaryotic microalgae hosts offer three distinct contexts for recom-

82
B.d.S.A.F. Brasil et al.

Table 2
Comparison of eukaryotic microalgae and cyanobacteria with the most commonly used systems for recombinant protein production

Taxa Cell growth rate Production cost Expression level Advantages Disadvantages

a,b
Eukaryotic algae High (6 h) Very low Nucleus: Scalability; Simple media requirements; GRAS. Low availability of molecular tools.
Low Nucleus: Eukaryotic gene expression machinery; Post-translational modifications (i.e., Nucleus: Low expression efficiency
Chloroplast: disulfide bond formation, phosphorylation, glycosylation); Protein secretion. Chloroplast: Absence of glycosylation; Protein remains in the
Low-high Chloroplast: Post-translational modifications (i.e.; disulfide bond formations, chloroplast.
phosphorylation).
Cyanobacteriaa,b High (4 h) Very low Low-moderate Scalability; Simple media requirements; GRAS; Post-translation modifications (i.e.; Low availability of molecular tools; Absence of glycosylation and some
disulfide bond formations, phosphorylation). types of post-translational modifications; Issues with protein solubility.
Bacteria Very high Moderate Low-high Easy operation; Simple media requirements; Disulfide bond formation (periplasm); Absence of post-translational modifications; Issues with protein
(i.e., E. coli)3,4,5 (30 min) Well-developed molecular tools; solubility; Culture contamination.
Yeastc,d Very high Moderate Low-high Easy operation; Simple media requirements; GRAS; Eukaryotic gene expression Culture contamination; Distinct glycosylation patterns/hyper-
(90 min) machinery; Post-translational modifications; Protein secretion; Well-developed glycosylation.
molecular tools.
Filamentous fungic,d Very high Moderate Low-high Easy operation; Simple media requirements; Eukaryotic gene expression machinery; High intracellular concentration of proteases; Culture contamination;

83
(90 min) Post-translational modifications; Protein secretion. Distinct glycosylation patterns.
c,e
Insect cells Moderate (18 h) High Low-high Eukaryotic gene expression machinery; Post-translational modifications. Demanding culture conditions; Simple glycosylation pattern (no sialic
acid);
Mammalian cellsc,d Low (> 24 h) High Low-moderate Eukaryotic protein processing; Post-translational modifications; Well-developed Demanding culture conditions; Culture contamination; Cell line
molecular tools. instability.
Plantsc,f Low (> 24 h) Low Nucleus: Scalability; Tissue/organ-specific expression. Long life-cycles; Issues with recombinant protein recovery from plant
Low Nucleus: Eukaryotic gene expression machinery; Post-translational modifications organs/tissues
Chloroplast: (disulfide bond formation, phosphorylation, glycosylation); Protein secretion Chloroplast: Absence of glycosylation; Protein remains in the
Low-high Chloroplast: Post-translational modifications (disulfide bond formations, chloroplast.
phosphorylation)

Partly from:
a
Rasala and Mayfield [144];
b
Wang et al., [56];
c
Yin et al., [145];
d
Corchero et al., [146];
e
Assenberg et al. [147];
f
Fischer et al., [148].
Algal Research 25 (2017) 76–89
B.d.S.A.F. Brasil et al. Algal Research 25 (2017) 76–89

Table 3
Examples of recombinant proteins expressed in algae.

Field Application Recombinant protein Host species References

Industrial Enzyme for food and paper Xylanase Dunaliella tertiolecta; Rasala et al. [160]; Georgiana et al.
manufacturing Chlamydomonas reinhardtii [161]
Enzyme for animal feed (increases Phytase D. tertiolecta; C. reinhardtii Yoon et al. [162]; Georgiana et al.
phosphorous uptake) [161]
Enzyme for animal feed (increases α-galactosidase D. tertiolecta; C. reinhardtii Georgiana et al. [161]
oligosaccharide hydrolysis)

Nutritional Supplementation of organic selenium Selenoprotein C. reinhardtii Hou et al. [163]


Fish dietary supplement and growth Flounder Growth hormone Chlorella elipsoidea Kim et al. [164]
promoter

Medical Anti-bacterial and anti-fungal peptide NP-1 peptide C. elipsoidea Chen et al. [165]
Stimulates angiogenesis Vascular endothelial growth factor (VEGF) C. reinhardtii Rasala et al. [166]
Increases resistance to UV-induced stress Metallothionein C. reinhardtii Zhang et al. [167]
Anemia treatment Erythropoietin C. reinhardtii Eichler-Stahlberg et al. [168]
Wound healing High Mobility group Protein 1B (HMGB1) C. reinhardtii Rasala et al. [166]
Anti-malarial vaccines Plasmodium surface proteins (AMS-1, MSP1, C. reinhardtii Dauvillée et al. [169]; Gregory et al.
Pfs25, Pfs28, Pfs48/45, Pfs25) [170]; Jones et al. [171]
Antibody against anthrax Anti-PA 83 anthrax IgG1 C. reinhardtii Tran et al. [172]
Antibody against Herpes Simplex Virus Anti-HSV glycoprotein D lsc C. reinhardtii Mayfield et al. [173]
Imunotoxins against B-cell lymphoma Anti-CD22-gelonin sc C. reinhardtii Tran et al. [174]; Tran et al. [175];
Anti-CD22-ETA sc

The list is not exhaustive. Partly from: Barrera and Mayfield [154]; Rasala and Mayfield [144]; Specht et al. [176].

binant protein expression: the nuclear, the plastid and the mitochon- [187], functional antibodies [172,188] and other proteins of biotech-
drial genomes [55,144,154]. All three algal genomes have been nological interest, including enzymes, has been achieved in C. reinhard-
successfully engineered for recombinant gene expression using trans- tii ([189–191]).
formation methods such as biolistic (nuclear, mitochondrial, and However, C. reinhardtii presents limited potential for protein
chloroplastic genomes), glass bead agitation (nuclear and chloroplastic production in large-scale cultivation systems due primarily to limita-
genomes), electroporation, and Agrobacterium-mediated gene transfer tions in the maximal cell density achieved in stationary-phase cultures
and silicon carbide whiskers (nuclear genome) (reviewed by [55]). The [185,196]. Alternative species, such as Dunalliela salina, Haematococcus
gene products expressed from the nucleus can be directed to subcellular pluvialis, Phaeodactylum tricornutum, Nannochloropsis gaditana and Chlor-
locations or secreted and are subjected to proper folding and post- ella sorokiniana are being tested for recombinant protein expression.
translational modifications (i.e.; disulfide bond formations, phosphor- Dunalliela salina and Haematococcus pluvialis have been cultivated for
ylation, glycosylation) by the eukaryotic cell machinery of microalgal decades at large scale to produce high-value carotenoids (β-carotene
hosts (Table 2). and astaxanthin, respectively) using hypersaline media, a condition
The possibility of secreting the recombinant products expressed that aids contamination management in open pond systems. Both the
from the nuclear genome into the surrounding media drastically nuclear and the chloroplastic genomes have been transformed in these
reduces the costs of downstream processing, making this genome an two species (Table 4); however, the absence of complete genomic
attractive option for the expression of enzyme-coding genes. However, sequences and an established molecular toolkit hampers their further
recombinant nuclear genes frequently shows silencing behavior leading development as recombinant protein hosts.
to low protein expression levels [179]. Furthermore, the diatom species Phaeodactylum tricornutum has a
In contrast, expression levels within the chloroplastic compartment fully sequenced genome and a significant set of molecular tools
are usually higher, accounting for up to 10.5% of the total soluble available, including genetic transformation methods, the availability
protein content [159]. The plastid genome is amenable to homologous of RNAi vectors, and the possibility of gene editing through transcrip-
recombination, and post-translational modifications such as disulfide tion activator-like effector nucleases (TALENs). In addition, proteins of
bond formation and phosphorylation can take place in the chloroplast. therapeutic interest such as monoclonal antibodies and the Hepatitis B
However, proteins expressed in this compartment cannot be glycosy- virus surface antigen immunogen (HBsAg) have been successfully
lated or targeted to secretion or other organelles (Table 2). expressed in P. tricornutum [192].
The mitochondrial genome also offers an alternative option for The species Chlorella sorokiniana and Nannochloropsis gaditana are
heterologous gene expression, but engineering of this genome has been ranked among the most promising for biomass and biofuel production
poorly explored to date. There are few reports regarding recombinant in freshwater and saltwater based media, respectively ([193]; NAABB
protein expression within this organelle in the green microalgae species Final Report, 2014; [185]). Both present high growth rates, high oil
Chlamydomonas reinhardtii [180–182]. content, and robustness and can achieve high densities in large-scale
Species from the Chlamydomonas genus have been studied as a cultures. Recent advances in genomic sequencing, the characterization
model for photosynthetic organisms for decades [183]. In addition, the of promoters, and genetic transformation methods have been reported
ease of C. reinhardtii transgene insertion [184], the availability of fully for both species [185,193,194]. The increasing interest in genetically
sequenced nuclear, chloroplast, and mitochondrial genomes [155], and engineering these organisms for biofuels production has led to an
advanced molecular tools such as well-characterized chloroplast and expansion in the knowledge about these species genetic features and
nuclear promoter elements [158] make this species the model of choice their molecular toolkits.
for genetic studies and recombinant protein expression in microalgae It is important to highlight, that competing with bacterial/fungal
(Table 4). Furthermore, the availability of cell wall-deficient strains systems for the production of industrial enzymes is not possible given
coupled with the haploid nature and a well-known sexual life cycle of C. the current available knowledge. Advances regarding the optimization
reinhardtii species make strain engineering relatively straightforward of large-scale phototrophic growth systems, and the development of
[144]. Expression at economically viable levels of recombinant vaccines powerful genetic tools to drive high levels of enzyme expression pose

84
B.d.S.A.F. Brasil et al.

Table 4
Selected eukaryotic microalgae and cyanobacteria species with potential for recombinant protein production

Taxon Species Genome sequence Transformation methods available Engineered Key features
available genomes

Chlorophyta Chlamydomonas 121 Mb (N, Chl, M) Agrobacterium; Biolistics; Electroporation; Glass N, C, M Well-developed molecular and bioinformatics tools; Cell wall-deficient mutants
reinhardtiia,b,c,h,i bead agitation; Silicon carbide whiskers. available; Metabolic pathways available. Gene targeting by CRISPR/Cas9, TALENs, and
ZFNs available.
Chlorophyta Chlorella sorokinianac,d,e 60 Mb (N1, Chl, M) Biolistics. N Amenable to large-scale culturing; Fast growth rate.
Chlorophyta Dunalliela salinaa,c,e – Biolistics. N, C Resistant to high salinity; Amenable to large-scale culturing; Lack of rigid cell wall.
Chlorophyta Haematococcus pluvialisc,e,f – Biolistics; Agrobacterium. N, C Resistant to high salinity; Amenable to large-scale culturing.
Eustigmatophyceae Nannochloropsis gaditanac 34 Mb (N, Chl, M) Electroporation. N DNA integration in the nuclear genome by homologous recombination; Amenable to
large-scale culturing.
Bacillariophyceae Phaeodactylum 27 Mb (N, Chl, M) Biolistics. N Well-developed molecular tools. Gene targeting by CRISPR/Cas9 and TALENs available.
tricornutumc,e,j
Cyanobacteria Synechococcus elongatesg 2,7 Mb (Chr, P) Electroporation, conjugation. Chr, P DNA integration in the genome by homologous recombination; Well-developed

85
molecular tools.
g
Cyanobacteria Synechocystis sp. PCC6803 3,6 Mb (Chr, P) Electroporation, ultrasonic transformation and Chr, P DNA integration in the genome by homologous recombination; Well-developed
conjugation. molecular tools.

N: nucleus; Chl, chloroplast; M: mithocondria; chr: Chromosome; P: plasmid; TALENs: transcription activator-like effector nucleases: ZFN: zinc-finger nucleases.
Partly from:
a
Rasala and Mayfield [144];
b
Rosales-Mendoza et al. [158];
c
Stephens et al. [55];
d
Barry et al. [185];
e
Gong et al. [58];
f
Saei et al. [186];
g
Wang et al. [56];
h
Shin et al. [67];
i
Baek et al. [64];
j
Hopes et al. [65].
Algal Research 25 (2017) 76–89
B.d.S.A.F. Brasil et al. Algal Research 25 (2017) 76–89

major hurdles that must be overcome. Even the expression of algal the European Union, Luxembourg, 2014, pp. 19–28.
[8] Algaemax, The Algae Market, a new Opportunity for European SMEs, Ateknea
enzyme-coding genes will most likely happen in other systems until, at Solutions Catalonia, S.A., 2013 Available in http://www.algaemax.eu/more-in-
some point in the future, algae genetic engineering becomes cost detail.html (Accessed in: 15 Jul 2015).
effective. However, the ability of microalgae to mimic mammalian [9] S. Behera, R. Singh, R. Arora, et al., Scope of algae as third generation biofuels,
Front. Bioeng. Biotechnol. 90 (2015) 1–13.
post-translational modifications (e.g., glycosylation) promises to accel- [10] B.S.A.F. Brasil, F.C.P. Silva, F.G. Siqueira, Microalgae biorefineries: the Brazilian
erate the development of algal systems for the production of recombi- scenario in perspective, New Biotechnol. (2016) (In Press, Corrected Proof).
nant proteins for biomedical applications. There is a growing body of [11] C. Yang, R. Nie, J. Fu, et al., Production of aviation fuel via catalytic hydrothermal
decarboxylation of fatty acids in microalgae oil, Bioresour. Technol. 146 (2013)
data regarding the expression of antibody-derived drugs, hormones, 569–573.
growth factors, anti-microbial peptides, and vaccines (Table 3). There- [12] E. Molina Grima, J.A. Sánchez Pérez, F. Garcia Camacho, et al., The production of
fore, the market demand for these high-value products might foster the polyunsaturated fatty acids by microalgae: from strain selection to product
purification, Process Biochem. 30 (1995) 711–719.
development of efficient recombinant protein production in photosyn-
[13] E. Ryckebosch, C. Bruneel, R. Termote-Verhalle, et al., Nutritional evaluation of
thetic microorganisms. Another promising approach for reducing the microalgae oils rich in omega-3 long chain polyunsaturated fatty acids as an
costs of algal enzyme production is the concomitant use of the residual alternative for fish oil, Food Chem. 160 (2014) 393–400.
algal biomass and lipids in an integrated biorefinery approach [10]. The [14] Y.W. Chen, H.V. Lee, J.C. Juan, et al., Production of new cellulose nanomaterial
from red algae marinebiomass Gelidium elegans, Carbohydr. Polym. 151 (2016)
exploitation of the whole algal biomass for the production of high- 1210–1219.
added-value products, such as carotenoids and enzymes, together with [15] Z. Xiang, W. Gao, L. Chen, et al., A comparison of cellulose nanofibrils produced
low added value products, such as animal feed and/or energy, would from Cladophora glomerata algae and bleached eucalyptus pulp, Cellulose 23
(2016) 493–503.
certainly improve the economical sustainability of the process. [16] M.A. Borowitzka, High-value products from microalgae—their development and
commercialisation, J. Appl. Phycol. 25 (2013) 743–756.
8. Future perspectives [17] P. Spolaore, C. Joannis-Cassan, E. Duran, et al., Commercial applications of
microalgae, J. Biosci. Bioeng. 101 (2006) 87–96.
[18] J.M. Girard, R. Tremblay, N. Faucheux, M. Heitz, J. Deschênes, Phycoremediation
The variety of enzymes found among microalgal species demon- of cheese whey permeate using directed commensalism between Scenedesmus
strates the importance of these versatile microorganisms as cellular obliquus and Chlorella protothecoides, Algal Res. 22 (2017) 122–126.
[19] C. Posten, C. Walter, Microalgal Biotechnology: Integration and Economy, Walter
factories. Although a large part of microalgae genetic resources remain
de Gruyter, Berlin, 2012.
unexplored, a growing number of enzymes with potential applications [20] M. Plaza, A. Cifuentes, E. Ibanez, In the search of new functional food ingredients
in the food, pharmaceutical, and chemical industry have been reported. from algae, Trends Food Sci. Technol. 19 (2008) 31–39.
[21] K.S. Kumar, H.U. Dahms, E.J. Won, et al., Microalgae – a promising tool for heavy
The development of new strategies and methods to induce and optimize
metal remediation, Ecotoxicol. Environ. Saf. 113 (2015) 329–352.
microalgae enzyme production, including biomanufacturing, is an [22] A.P. Batista, L. Ambrosano, S. Graça, et al., Combining urban waste water
important approach for the scale-up and industrialization of potential treatment with biohydrogen production – an integrated microalgae-based ap-
enzymes. Some of the most important challenges will be increasing the proach, Bioresour. Technol. 184 (2015) 230–235.
[23] Ichsan, H. Hadiyanto, R. Hendroko, Integrated biogas-microalgae from waste
concentration of enzymes and to reducing the cost of downstream waters as the potential biorefinery sources in Indonesia, Energy Procedia 47
operations. Genetic engineering and synthetic biology advances, (2014) 143–148.
coupled with the use of the whole algal biomass to produce several [24] M. Kouhia, H. Holmberg, P. Ahtila, Microalgae-utilizing biorefinery concept for
pulp and paper industry: converting secondary streams into value-added products,
bioproducts in a biorefinery strategy, offer promising possibilities to Algal Res. 10 (2015) 41–47.
overcome these limitations and achieve the cost-effective production of [25] M. Hlavová, Z. Turóczy, K. Bišová, Improving microalgae for biotechnology —
industrial enzymes in microalgal cells in the future. from genetics to synthetic biology, Biotechnol. Adv. 33 (2015) 1194–1203.
[26] M. Koller, A. Muhr, G. Braunegg, Microalgae as versatile cellular factories for
valued products, Algal Res. 6 (2014) 52–63.
Declaration of interest [27] G.J. Gil-Chávez, J.A. Villa, J.F. Ayala-Zavala, et al., Technologies for extraction
and production of bioactive compounds to be used as nutraceuticals and food
ingredients: an overview, Compr. Rev. Food Sci. Food Saf. 12 (2013) 5–23.
The authors report no declarations of interest. [28] B. Serive, R. Kaas, J.B. Bérard, et al., Selection and optimisation of a method for
efficient metabolites extraction from microalgae, Bioresour. Technol. 124 (2012)
Acknowledgements 311–320.
[29] L. Brennan, P. Owende, Biofuels from microalgae- a review of technologic
production, processing and extractions of bio-fuels and co-products, Renew. Sust.
The authors are grateful to the Brazilian Agricultural Research Energ. Rev. 14 (2010) 557–577.
Corporation (EMBRAPA) (grant number 03.12.11.006.00.00) and [30] J. De la Noue, N. Pauw, The potential of microalgal biotechnology: a review of
National Council for Scientific and Technological Development production and uses of microalgae, Biotechnol. Adv. 6 (1988) 725–770.
[31] Y. Alkhamis, J.G. Qin, Cultivation of Isochrysis galbana in phototrophic, hetero-
(CNPq) (grant numbers 310424/2015-1 and 477891/2013-6) for trophic and mixotropic conditions, Biomed. Res. Int. 2013 (2013) 1–9.
supporting this work. [32] O. Perez-Garcia, F.M.E. Escalante, L.E. De-Bashan, et al., Heterotrophic cultures of
microalgae: metabolism and potential products, Water Res. 45 (2011) 11–36.
[33] M.A. Guzmán-Murillo, C.C. López-Bolaños, T. Ledesma-Verdejo, Effects of fertili-
References zer-based culture media on the production of exocellular polysaccharides and
cellular superoxide dismutase by Phaeodactylum tricornutum (Bohlin), J. Appl.
[1] I. Priyadarshani, B. Rath, Commercial and industrial applications of microalgae – a Phycol. 19 (2007) 33–41.
review, J. Algal Biomass Utln. 3 (2012) 89–100. [34] K. Klanbut, Y. Peerapornpisarn, Khanongnuch, et al., Phytase from some strains of
[2] J. Masojídek, G. Torzillo, Mass cultivation of freshwater microalgae, Reference thermophilic blue-green algae, Jpn. J. Phycol. S2 (2002) 57–60.
Module in Earth Systems and Environmental Sciences, Elsevier, 2014, pp. 1–13, , [35] P.M. Dey, H. Kauss, α-galactosidase of Poterioochromonas malhamensis,
http://dx.doi.org/10.1016/B978-0-12-409548-9.09373-8. Phytochemistry 20 (1981) 45–48.
[3] R.A. Anderson, M. Kawacbi, R.A. Anderson, Traditional microalgae isolation [36] W. Lockau, B. Massalsky, A. Dirmeier, Purification and partial characterization of
techniques in algal culturing techniques, Algal Culturing Techniques, Elsevier a calcium-stimulated protease from the cyanobacterium, Anabaena variabilis, Eur.
Academic Press, Amsterdam, 2005, pp. 83–101. J. Biochem. 172 (1988) 433–438.
[4] L. Collins, D. Alvarez, A. Chauhan, Phycoremediation coupled with generation of [37] U. Strohmeier, C. Gerdes, W. Lockau, Proteolysis in heterocyst-forming cyano-
value-added products, in: S. Das (Ed.), Microbial Biodegradation and bacteria: characterization of a further enzyme with trypsin-like specificity, and of
Bioremediation, Elsevier, London, 2014, pp. 341–387. a prolylendopeptidase from Anabaena variabilis, Z. Naturforsch. C 49 (1994)
[5] S.I.I.A. Hadi, H. Santana, P.P.M. Brunale, et al., DNA barcoding green microalgae 70–78.
isolated from neotropical inland waters, PLoS One 11 (2) (2016) e0149284. [38] G.W. Niven, The characterization of two aminopeptidase activities from the
[6] J.A. Gimpel, E.A. Specht, D.R. Georgianna, et al., Advances in microalgae cyanobacterium Anabaena flos-aquae, Biochim. Biophys. Acta 1253 (1995)
engineering and synthetic biology applications for biofuel production, Curr. Opin. 193–198.
Chem. Biol. 17 (2013) 489–495. [39] E. Nanni, E. Balestreti, E. Dainese, et al., Characterisation of a specific
[7] C. Enzing, M. Ploeg, M. Barbosa, et al., Current markets, products and future Phycocyanin-hydrolysing protease purified from Spirulina platensis, Microbiol. Res.
developments for micro-algae, in: M. Vigani, et al. (Ed.), Microalgae-based 156 (2001) 259–266.
products for the food and feed sector: an outlook for Europe, Publications Office of [40] E. Yada, H.N. Nagata, Y. Noguchi, et al., Arginine specific protease from Spirulina

86
B.d.S.A.F. Brasil et al. Algal Research 25 (2017) 76–89

platensis, Mar. Biotechnol. 7 (2005) 474–480. [74] J.H. Hehemann, A.B. Boraston, M. Czjzek, A sweet new wave: structures and
[41] H.P. Misra, I. Fridovich, Purification and properties of superoxide dismutase from mechanisms of enzymes that digest polysaccharides from marine algae, Curr.Opin.
red alga Porphyridium cruentum, J. Biol. Chem. 252 (1977) 6421–6423. Struct. Biol. 28 (2014) 77–86.
[42] A. Canini, P. Civitareale, S. Marini, et al., Purification of iron superoxide dismutase [75] M. Ike, Y. Ko, K. Yokoyama, et al., Cellobiohydrolase I (Cel7A) from Trichoderma
from the cyanobacterium Anabaena cylindrica Lemm. and localization of the reesei has chitosanase activity, J. Mol. Cat. B: Enzymatic. 47 (2007) 159–163.
enzyme in heterocysts by immunogold labeling, Planta 187 (1992) 438–444. [76] J. Liu, W.S. Xia, Characterization of chitosanase from cellulose produced by
[43] S. Gunes, S. Tamburaci, E. Imamoglu, et al., Determination of superoxide Trichodermaviride, Chin. J. Biochem. Mol. Biol. 21 (2005) 713–716.
dismutase activities in different cyanobacteria for scavenging of reactive oxygen [77] V. Gupta, C. Natarajan, K. Kumar, et al., Identification and characterization of
species, J. Biologically Active Prod. Nat. 5 (2015) 25–32. endoglucanases for fungicidal activity in Anabaena laxa (Cyanobacteria), J. Appl.
[44] S.K. Yong, B.H. Lim, S. Saleh, et al., Optimisation, purification and characterisa- Phycol. 23 (2011) 73–81.
tion of extracellular lipase from Botryococcus sudeticus (UTEX 2629), J. Mol. [78] M. Martin, S. Bivera, S. Steels, et al., Functional screening of a metagenomic
Catal. B-Enzym. 126 (2016) 99–105. library of seaweed-associated microbiota: identification and characterization of a
[45] M.G. Savvidou, T.G. Sotiroudis, F.N. Kolisis, Cell surface and cellular debris- halotolerant, cold-active marine endo-ß-1,4-endoglucanase, Appl. Enviorn.
associated heat-stable lipolytic enzyme activities of the marine alga Microbial. 80 (2014) 4958–4967.
Nannochloropsis oceanica, Biocatal. Biotransformation 34 (1) (2016) 24–32. [79] G. Michel, M. Czjzek, Polysaccharide - degrading enzymes from marine bacteria,
[46] S.B. Gould, Evolutionary genomics: algae's complex origins, Nature 492 (2012) in: A. Trincone (Ed.), Marine Enzymes for Biocatalysis: Sources, Biocatalytic
46–48. Characteristic and Bioprocesses of Marine Enzymes, Woodhead Publishing
[47] P.J. Keeling, The number, speed, and impact of plastid endosymbioses in Limited, Cambridge, 2013, pp. 429–445.
eukaryotic evolution, Annu. Rev. Plant Biol. 64 (2013) 583–607. [80] R. Prasanna, N. Lata, R. Tripathi, et al., Evaluation of fungicidal activity of
[48] B.A. Curtis, G. Tanifuji, F. Burki, et al., Algal genomes reveal evolutionary extracellular filtrates of cyanobacteria—possible role of hydrolytic enzymes, J.
mosaicism and the fate of nucleomorphs, Nature 492 (2012) 59–65. Basic Microbiol. 48 (2008) 186–194.
[49] L.A. Katz, Origin and diversification of eukaryotes, Annu. Rev. Microbiol. 66 [81] R. Prasanna, A. Sood, P. Jaiswal, et al., Rediscovering cyanobacteria as valuable
(2012) 411–427. sources of bioactive compounds, Appl. Biochem.Microbiol. 46 (2010) 133–147.
[50] M.D. Guiry, How many species of algae are there? J. Phycol. 48 (2012) [82] M. Martin, D. Portetelle, G. Michel, et al., Microorganisms living on macroalgae:
1057–1063. diversity, interactions and biotechnological applications, Appl. Microbiol.
[51] J.C. Nabout, B. da Silva Rocha, F.M. Carneiro, et al., How many species of Biotechnol. 98 (2014) 2917–2935.
Cyanobacteria are there? Using a discovery curve to predict the species number, [83] M. Vihinen, P. Mantsala, Microbial amylolytic enzymes, Crit. Rev. Biochem. Mol.
Biodivers. Conserv. 22 (2013) 2907–2918. Biol. 24 (1989) 329–418.
[52] S. Kaur, M. Sarkar, R.B. Srivastava, et al., Fatty acid profiling and molecular [84] C. Levis, M. Gibbs, Starch degradation in synchronously grown Chlamydomonas
characterization of some freshwater microalgae from India with potential for reinhardtii and characterization of amylase, Plant Physiol. 74 (1984) 459–463.
biodiesel production, New Biotechnol. 29 (3) (2012) 332–344. [85] H. Rismani- Yazdi, B. Haznedaroglu, K. Bibby, et al., Transcriptome sequencing
[53] D.K. Lim, S. Garg, M. Timmins, et al., Isolation and evaluation of oil-producing and annotation of the microalgae Dunaliella tertiolecta: pathway description and
microalgae from subtropical coastal and brackish waters, PLoS One 7 (2012) gene discovery for production of next-generation biofuels, BMC Genomics 12
e40751. (2011) 1–17.
[54] T. Mutanda, D. Ramesh, S. Karthikeyan, et al., Bioprospecting for hyper-lipid [86] C. Shang, G. Bi, Z. Yuan, et al., Discovery of genes for production of biofuels
producing microalgal strains for sustainable biofuel production, Bioresour. through transcriptome sequencing of Dunaliella parva, Algal Res. 13 (2016)
Technol. 102 (2011) 57–70. 318–326.
[55] E. Stephens, J. Wolf, M. Oey, et al., Genetic engineering for microalgae strain [87] P. Katriola, E. Rajashekhara, Q. Yan, et al., Biotechnological potential of microbial
improvement in relation to biocrude production systems, in: N.R. Moheimani, α-galactosidases, Crit. Rev. Biotechnol. 34 (4) (2013) 307–317.
et al. (Ed.), Biomass and Biofuels from Microalgae, Springer, Heidelberg, 2015, pp. [88] L. Weignerová, P. Simerská, V. Kren, α-galactosidases and their applications in
191–249. biotransformations, Biocatal. Biotransfor. 27 (2009) 79–89.
[56] B. Wang, J. Wang, W. Zhang, et al., Application of synthetic biology in [89] Q. Husain, β-galactosidases and their potential applications: a review, Crit. Rev.
cyanobacteria and algae, Front. Microbiol. 3 (2012) 344. Biotechnol. 30 (2010) 41–62.
[57] M.J. Reijnders, R.G. van Heck, C.M. Lam, et al., Green genes: bioinformatics and [90] Z. Mlichová, M. Rosenberg, Current trends of β-galctosidase application in food
systems-biology innovations drive algal biotechnology, Trends Biotechnol. 32 technology, J. Food Nutr. Res. 45 (2006) 47–54.
(2014) 617–626. [91] C. Oliveira, P.M.R. Guimarães, L. Domingues, Recombinant microbial systems for
[58] Y. Gong, H. Hu, Y. Gao, et al., Microalgae as platforms for production of improved β-galactosidase production and biotechnological applications,
recombinant proteins and valuable compounds: progress and prospects, J. Ind. Biotechnol. Adv. 29 (2011) 600–609.
Microbiol. Biotechnol. 38 (2011) 1879–1890. [92] C.M. Davies, S.C. Apte, S.M. Peterson, et al., Plant and algal interference in
[59] R.H. Wijffels, O. Kruse, K.J. Hellingwerf, Potential of industrial biotechnology bacterial β-D-galactosidase and β-glucuronidase assays, Appl. Environ. Microbiol.
with cyanobacteria and eukaryotic microalgae, Curr. Opin. Biotechnol. 24 (3) 60 (1994) 3959–3964.
(2013) 405–413. [93] J.M. Girard, M.L. Roy, M.B. Hafsa, et al., Mixotrophic cultivation of green
[60] J. Steinbrenner, G. Sandmann, Transformation of the green alga Haematococcus microalgae Scenedesmus obliquus on cheese whey permeate for biodiesel produc-
pluvialis with a phytoene desaturase for accelerated astaxanthin biosynthesis, tion, Algal Res. 5 (2014) 241–248.
Appl. Environ. Microbiol. 72 (12) (2006) 7477–7484. [94] S.M. Kotwal, M.M. Gote, S.R. Sainkar, et al., Production of α-galactosidase by
[61] B.F. Cordero, I. Couso, R. León, et al., Enhancement of carotenoids biosynthesis in thermophilic fungus Humicola sp. in solid state fermentation and its applications in
Chlamydomonas reinhardtii by nuclear transformation using a phytoene synthase soyamilk hydrolysis, Process Biochem. 33 (1998) 337–343.
gene isolated from Chlorella zofingiensis, Appl. Microbiol. Biotechnol. 91 (2) (2011) [95] C.T.N. Sanada, S.G. Karp, M.R. Spier, et al., Utilization of soybean vinasse for α-
341–351. galactosidase production, Food Res. Int. 42 (2009) 476–483.
[62] I. Couso, M. Vila, H. Rodriguez, et al., Overexpression of an exogenous phytoene [96] D. Zhu, Q. Wu, N. Wang, Industrial enzymes, in: M. Moo-Young (Ed.),
synthase gene in the unicellular alga Chlamydomonas reinhardtii leads to an Comprehensive Biotechnology, Academic Press, Burlington, 2011, pp. 3–13.
increase in the content of carotenoids, Biotechnol. Prog. 27 (1) (2011) 54–60. [97] M.B. Rao, A.M. Tanksale, M.S. Ghatge, et al., Molecular and biotechnological
[63] P.D. Hsu, E.S. Lander, F. Zhang, Development and applications of CRISPR-Cas9 for aspects of microbial protease, Microbiol. Mol. Biol. Rev. 62 (1998) 597–635.
genome engineering, Cell 157 (6) (2014) 1262–1278. [98] J.L. Pérez-Lloréns, E. Benítez, J.J. Vergara, et al., Characterization of proteolityc
[64] K. Baek, D.H. Kim, J. Jeong, et al., DNA-free two-gene knockout in enzyme activities in macroalgae, Eur. J. Phycol. 38 (2003) 55–64.
Chlamydomonas reinhardtii via CRISPR-Cas9 ribonucleoproteins, Sci. Report. 6 [99] P.M. Souza, M.L. Bitterncourt, C.C. Caprara, et al., A biotechnology perspective of
(2016). fungal proteases, Braz. J. Microbiol. 46 (2015) 337–346.
[65] A. Hopes, V. Nekrasov, S. Kamoun, T. Mock, Editing of the urease gene by CRISPR- [100] A.M. Baron, M.I. Sarquis, M. Baigori, et al., A comparative study of the synthesis of
Cas in the diatom Thalassiosira pseudonana, Plant Methods 12 (1) (2016) 49. n-butyl-oleate using a crude lipolytic extract of Penicillum coryophilum in water-
[66] M. Nymark, A.K. Sharma, T. Sparstad, A.M. Bones, P. Winge, A CRISPR/Cas9 restricted environments, J. Mol. Catal. B Enzym. 34 (2005) 25–32.
system adapted for gene editing in marine algae, Sci. Report. 6 (2016). [101] M. Perignon, J. Lecomte, M. Pina, et al., Activity of immobilized Thermomyces
[67] S.-E. Shin, J.-M. Lim, H.G. Koh, et al., CRISPR/Cas9-induced knockout and knock- lanuginosus and Candida antarctica B lipases in interesterification reactions: effect
in mutations in Chlamydomonas reinhardtii, Sci. Report. 6 (2016). of the Aqueous microenvironment, J. Am. Oil Chem. Soc. 90 (8) (2013)
[68] Q. Wang, Y. Lu, Y. Xin, et al., Genome editing of model oleaginous microalgae 1151–1156.
Nannochloropsis spp. by CRISPR/Cas9, Plant J. 88 (6) (2016) 1071–1081. [102] T.F.C. Salum, P. Villeneuve, B. Barea, et al., Synthesis of biodiesel in column fixed-
[69] J.L. Adrio, A. Demain, Microbial enzymes: tools for biotechnological process, bed bioreactor using the fermented solid produced by Burkholderia cepacia
Biomol. Ther. 4 (2014) 117–139. LTEB11, Process Biochem. 45 (2010) 1348–1354.
[70] BBC Research, Report BIO030H F Enzymes in Industrial Applications: Global [103] F. Hasan, A.A. Shah, A. Hameed, Industrial applications of microbial lipases,
Markets, BBC Research, Wellesley, 2014. Enzyme Microb. Tech. 39 (2006) 235–251.
[71] F.G. Siqueira, E.X. Ferreira-Filho, Plant cell wall as a substrate for the production [104] B.S. Demir, S.S. Tukel, Purification and characterization of lipase from Spirulina
of enzymes with industrial applications, Mini-Rev. Org. Chem. 7 (2010) 54–60. platensis, J. Mol. Catal. B Enzym. 64 (2010) 123–128.
[72] M.K. Bhat, S. Bhat, Cellulose degrading enzymes and their potential industrial [105] S. Godet, J. Herault, G. Pencreac'h, et al., Isolation and analysis of a gene from the
applications, Biotechnol. Adv. 15 (1997) 583–620. marine microalga Isochrysis galbana that encodes a lipase-like protein, J. Appl.
[73] T. Takeda, Y. Nakano, M. Takahashi, et al., Identification and enzymatic Phycol. 24 (2012) 1547–1553.
characterization of an endo-1,3-beta-glucanase from Euglena gracilis, [106] R. Miller, G. Wu, R.R. Deshpande, et al., Changes in transcript abundance in
Phytochemistry 116 (2015) 21–27. Chlamydomonas reinhardtii following nitrogen deprivation predict diversion of

87
B.d.S.A.F. Brasil et al. Algal Research 25 (2017) 76–89

metabolism, Plant Physiol. 154 (2010) 1737–1752. 491–515.


[107] X. Li, E.R. Moellering, B. Liu, et al., A galactoglycerolipid lipase is required for [142] D. Wang, Y. Li, X. Hu, et al., Combined enzymatic and mechanical cell disruption
triacylglycerol accumulation and survival following nitrogen deprivation in and lipid extraction of green alga Neochloris oleoabundans, Int. J. Mol. Sci. 16
Chlamydomonas reinhardtii, Plant Cell 24 (2012) 4670–4686. (2015) 7707–7722.
[108] C.B. Mansfeldt, L.V. Richter, B.A. Ahner, et al., Use of de novo transcriptome [143] A.K. Lee, D.M. Lewis, P.J. Ashman, Disruption of microalgal cells for the extraction
libraries to characterize a novel oleaginous marine chlorella species during the of lipids for biofuels: processes and specific energy requirements, Biomass
accumulation of triacylglycerols, PLoS One 11 (2) (2016). Bioenergy 46 (2012) 89–101.
[109] X. Li, C. Benning, M.H. Kuo, Rapid triacylglycerol turnover in Chlamydomonas [144] B.A. Rasala, S.P. Mayfield, Photosynthetic biomanufacturing in green algae;
reinhardtii requires a lipase with broad substrate specificity, Eukaryot. Cell 11 production of recombinant proteins for industrial, nutritional, and medical uses,
(2012) 1451–1462. Photosynth. Res. 123 (2014) 227–239.
[110] X.G. Lei, C.H. Stahl, Biotechnological development of effective phytases for [145] J. Yin, G. Li, X. Ren, et al., Select what you need: a comparative evaluation of the
mineral nutrition and environmental protection, Appl. Microbiol. Biotechnol. 57 advantages and limitations of frequently used expression systems for foreign
(2001) 474–481. genes, J. Biotechnol. 127 (2007) 335–347.
[111] R. Greiner, U. Konietzny, Phytase for food application, Food Technol. Biotechnol. [146] J.L. Corchero, B. Gasser, D. Resina, et al., Unconventional microbial systems for
44 (2006) 125–140. the cost-efficient production of high-quality protein therapeutics, Biotechnol. Adv.
[112] P. Shah, K. Bhavsar, S.K. Soni, et al., Strain improvement and up scaling of phytase 31 (2013) 140–153.
production by Aspergillus niger NCIM 563 under submerged fermentation condi- [147] R. Assenberg, P.T. Wan, S. Geisse, et al., Advances in recombinant protein
tions, J. Ind. Microbiol. Biotechnol. 36 (2009) 373–380. expression for use in pharmaceutical research, Curr. Opin. Struct. Biol. 23 (2013)
[113] D.E.S.C. Rao, K.V. Rao, T.P. Reddy, et al., Molecular characterization, physico- 393–402.
chemical properties, kwon and potential applications of phytases: an overview, [148] R. Fischer, E. Stoger, S. Schillberg, et al., Plant-based production of biopharma-
Crit. Rev. Biotechnol. 29 (2009) 182–198. ceuticals, Curr. Opin. Plant Biol. 7 (2004) 152–158.
[114] M.R. Spier, R. Greiner, J.A. Rodriguez-Léon, et al., Phytase production using citric [149] J.A. Gimpel, H.H. Nour-Eldin, M.A. Scranton, et al., Refactoring the six-gene
pulp and other residues of the agroindustry in SSF by fungal isolates, Food Photosystem II core in the chloroplast of the green algae Chlamydomonas
Technol. Biotechnol. 46 (2008) 178–182. reinhardtii, ACS Synth. Biol. 5 (2016) 589–596.
[115] D. Maté, E. Gárcia-Ruiz, S. Camarero, et al., Directed evolution of fungal laccases, [150] S. Sahdev, S.K. Khattar, K.S. Saini, Production of active eukaryotic proteins
Curr. Genomics 12 (2011) 113–122. through bacterial expression systems: a review of the existing biotechnology
[116] P. Baldrian, Fungal laccases—occurrence and properties, FEMS Microbiol. Rev. 30 strategies, Mol. Cell. Biochem. 307 (2008) 249–264.
(2006) 215–242. [151] D. Porro, M. Sauer, P. Branduardi, et al., Recombinant protein production in
[117] A. Piscitelli, C. Del Vecchio, V. Faraco, et al., Fungal laccases: versatile tools for yeasts, Mol. Biotechnol. 31 (2005) 245–259.
lignocellulose transformation, C. R. Biol. 334 (2011) 789–794. [152] P.J. Punt, N. van Biezen, A. Conesa, et al., Filamentous fungi as cell factories for
[118] P. Giardina, G. Sannia, Laccases: old enzymes with a promising future, Cell. Mol. heterologous protein production, Trends Biotechnol. 20 (2002) 200–206.
Life Sci. 72 (2015) 855–856. [153] L. Ikonomou, Y.J. Schneider, S. Agathos, Insect cell culture for industrial
[119] H. Claus, Laccases: structure, reactions, distribution, Micron 35 (2004) 93–96. production of recombinant proteins, Appl. Microbiol. Biotechnol. 62 (2003) 1–20.
[120] A. Lisov, A. Zavarzina, A. Zavarzin, et al., Dimeric and monomeric laccases of soil- [154] D.J. Barrera, S.P. Mayfield, High-value recombinant protein production in
stabilizing lichen Solorinacrocea: purification, properties and reactions with humic microalgae, in: A. Richmond (Ed.), Handbook of Microalgal Culture: Applied
acids, Soil Biol. Biochem. 45 (2012) 161–167. Phycology and Biotechnology, Wiley, New York, 2013, pp. 532–544.
[121] B. Otto, D. Schlosser, W. Reisser, First description of a laccase-like enzyme in soil [155] E. Specht, S. Miyake-Stoner, S. Mayfield, Micro-algae come of age as a platform for
algae, Arch. Microbiol. 192 (2010) 759–768. recombinant protein production, Biotechnol. Lett. 32 (2010) 1373–1383.
[122] I.S. Ng, F.X. Xu, X. Zhang, et al., Enzymatic exploration of catalase from a [156] K.J. Lauersen, H. Berger, J.H. Mussgnug, et al., Efficient recombinant protein
nanoparticle producing and biodecolorizing algae Shewanella xiamenensis BC01, production and secretion from nuclear transgenes in Chlamydomonas reinhardtii, J.
Bioresour. Technol. 184 (2015) 429–435. Biotechnol. 167 (2) (2013) 101–110.
[123] J.K. Fredrickson, M.F. Romine, A.S. Beliaev, et al., Towards environmental [157] S. Purton, J. Szaub, T. Wannathong, et al., Genetic engineering of algal
systems biology of Shewanella, Nat. Rev. Microbiol. 6 (2008) 592–603. chloroplasts: progress and prospects, Russ. J. Plant Physiol. 60 (2013) 491–499.
[124] K.H. Nealson, A. Belz, B. McKee, Breathing metals as a way of life: geobiology in [158] S. Rosales-Mendoza, L.M.T. Paz-Maldonado, R.E. Soria-Guerra, Chlamydomonas
action, Antonie Van Leeuwenhoek 81 (2002) 215–222. reinhardtii as a viable platform for the production of recombinant proteins: current
[125] L. Shi, K.M. Rosso, T.A. Clarke, et al., Molecular underpinnings of Fe (III) oxide status and perspectives, Plant Cell Rep. 31 (2012) 479–494.
reduction by Shewanella oneidensis MR-1, Front. Microbiol. 3 (2012) 50. [159] R. Surzycki, K. Greenham, K. Kitayama, et al., Factors effecting expression of
[126] B. Otto, D. Schlosser, First laccase in green algae: purification and characterization vaccines in microalgae, Biologicals 37 (2009) 133–138.
of an extracellular phenol oxidase from Tetracystisaeria, Planta 240 (2014) [160] B.A. Rasala, P.A. Lee, Z. Shen, et al., Robust expression and secretion of Xylanase1
1225–1236. in Chlamydomonas reinhardtii by fusion to a selection gene and processing with the
[127] B. Otto, C. Beuchel, C. Liers, et al., Laccase-like enzyme activities from chlor- FMDV 2A peptide, PLoS One 7 (2012) e43349.
ophycean green algae with potential for bioconversion of phenolic pollutants, [161] D.R. Georgiana, M.J. Hannon, M. Marcuschi, et al., Production of recombinant
FEMS Microbiol. Lett. 362 (2015). enzymes in the marine alga Dunaliella tertiolecta, Algal Res. 2 (2013) 2–9.
[128] E. Acuner, F.B. Dilek, Treatment of tectilon yellow 2G by Chlorella vulgaris, Process [162] S.M. Yoon, S.Y. Kim, K.F. Li, et al., Transgenic microalgae expressing Escherichia
Biochem. 39 (2004) 623–631. coli AppA phytase as feed additive to reduce phytate excretion in the manure of
[129] N. Daneshvar, M. Ayazloo, A.R. Khataee, et al., Biological decolourization of dye young broiler chicks, Appl. Microbiol. Biotechnol. 91 (2011) 553–563.
solution containing malachite green by microalgae Cosmarium sp, Bioresour. [163] Q. Hou, S. Qiu, Q. Liu, J. Tian, Z. Hu, J. Ni, Selenoprotein-transgenic
Technol. 98 (2007) 1176–1182. Chlamydomonas reinhardtii, Nutrients 5 (3) (2013) 624–636.
[130] S.A.C. Lima, P.M.L. Castro, R. Morais, Biodegradation of p-nitrophenol by [164] D.-H. Kim, Y.T. Kim, J.J. Cho, J.-H. Bae, S.-B. Hur, I. Hwang, T.-J. Choi, Stable
microalgae, J. Appl. Phycol. 15 (2003) 137–142. integration and functional expression of flounder growth hormone gene in
[131] E. Safonova, K. Kvitko, P. Kuschk, et al., Biodegradation of phenanthrene by the transformed microalga, Chlorella ellipsoidea, Mar. Biotechnol. 4 (1) (2002) 63–73.
green alga Scenedesmus obliquus ES-55, Eng. Life Sci. 5 (2005) 234–239. [165] Y. Chen, Y. Wang, Y. Sun, et al., Highly efficient expression of rabbit neutrophil
[132] E. Tarlan, F.B. Dilek, U. Yetis, Effectiveness of algae in the treatment of a wood- peptide-1 gene in Chlorella ellipsoidea cells, Curr. Genet. 39 (5) (2001) 365–370.
based pulp and paper industry wastewater, Bioresour. Technol. 84 (2002) 1–5. [166] B.A. Rasala, M. Muto, P.A. Lee, et al., Production of therapeutic proteins in algae,
[133] N. Mallick, F.H. Mohn, Reactive oxygen species: response of algal cells, J. Plant analysis of expression of seven human proteins in the chloroplast of
Physiol. 157 (2000) 183–193. Chlamydomonas reinhardtii, Plant Biotechnol. J. 8 (6) (2010) 719–733.
[134] A. Bafana, S. Dutt, Baldrian S. Kumar, et al., Superoxide dismutase: an industrial [167] Y.K. Zhang, G.F. Shen, B.G. Ru, Survival of human metallothionein-2 transplas-
perspective, Crit. Rev. Biotechnol. 31 (2011) 65–76. tomic Chlamydomonas reinhardtii to ultraviolet B exposure, Biochim. Biophys
[135] P.J. Janknegt, M.D. Graaff, W.H.V. De Poll, et al., Short-term antioxidative Sinica. 38 (3) (2006) 187–193.
responses of 15 microalgae exposed to excessive irradiance during ultraviolet [168] A. Eichler-Stahlberg, W. Weisheit, O. Ruecker, M. Heitzer, Strategies to facilitate
radiation, Eur. J. Phycol. 44 (2009) 525–539. transgene expression in Chlamydomonas reinhardtii, Planta 229 (4) (2009)
[136] C. González-Fernández, M. Ballesteros, Linking microalgae and cyanobacteria 873–883.
culture conditions and key-enzymes for carbohydrate accumulation, Biotechnol. [169] D. Dauvillée, S. Delhaye, S. Gruyer, C. Slomianny, S.E. Moretz, C. d'Hulst,
Adv. 30 (2012) 1655–1661. C.A. Long, S.G. Ball, S. Tomavo, Engineering the chloroplast targeted malarial
[137] J.V. Moroney, S.G. Bartlett, G. Samuelsson, Carbonic anhydrase in plants and vaccine antigens in Chlamydomonas starch granules, PLoS One 5 (12) (2010)
algae, Plant Cell Environ. 24 (2001) 141–153. e15424.
[138] J.M. González, S.Z. Fisher, Carbonic anhydrases in industrial applications, in: [170] J.A. Gregory, F. Li, L.M. Tomosada, C.J. Cox, A.B. Topol, J.M. Vinetz, S. Mayfield,
S.C. Frost, R. McKenna (Eds.), Carbonic Anhydrase: Mechanism, Regulation, Links Algae-produced Pfs25 elicits antibodies that inhibit malaria transmission, PLoS
to Disease, and Industrial Applications, Springer, Netherlands, 2014, pp. 405–426. One 7 (5) (2012) e37179.
[139] G.V. Swarnalatha, N.S. Hegde, V.S. Chauhan, et al., The effect of carbon dioxide [171] C.S. Jones, T. Luong, M. Hannon, M. Tran, J.A. Gregory, Z. Shen, S.P. Briggs,
rich environment on carbonic anhydrase activity, growth and metabolite pro- S.P. Mayfield, Heterologous expression of the C-terminal antigenic domain of the
duction in indigenous freshwater microalgae, Algal Res. 9 (2015) 151–159. malaria vaccine candidate Pfs48/45 in the green algae Chlamydomonas rein-
[140] K.Y. Show, D.J. Lee, J.H. Tay, et al., Microalgal drying and cell disruption – recent hardtii, Appl. Microbiol. Biotechnol. 97 (5) (2013) 1987–1995.
advances, Bioresour. Technol. 184 (2015) 258–266. [172] M. Tran, B. Zhou, P.L. Pettersson, et al., Synthesis and assembly of a full-length
[141] M.E. Grima, E.H. Belarbi, F.G.A. Fernandéz, et al., Recovery of microalgal biomass human monoclonal antibody in algal chloroplasts, Biotechnol. Bioeng. 104 (2009)
and metabolites: process options and economics, Biotechnol. Adv. 20 (2003) 663–673.

88
B.d.S.A.F. Brasil et al. Algal Research 25 (2017) 76–89

[173] S.P. Mayfield, S.E. Franklin, R.A. Lerner, Expression and assembly of a fully active [186] A.A. Saei, P. Ghanbari, A. Barzegari, Haematococcus as a promising cell factory to
antibody in algae, Proc. Natl. Acad. Sci. 100 (2) (2003) 438–442. produce recombinant pharmaceutical proteins, Mol. Biol. Rep. 39 (2012)
[174] M. Tran, R.E. Henry, D. Siefker, et al., Production of anti-cancer immunotoxins in 9931–9939.
algae: Ribosome inactivating proteins as fusion partners, Biotechnol. Bioeng. 110 [187] I.A. Dreesen, G. Charpin-El Hamri, M. Fussenegger, Heat-stable oral alga-based
(11) (2013) 2826–2835. vaccine protects mice from Staphylococcus aureus infection, J. Biotechnol. 145
[175] M. Tran, C. Van, D.J. Barrera, et al., Production of unique immunotoxin cancer (2010) 273–280.
therapeutics in algal chloroplasts, Proc. Natl. Acad. Sci. 110 (1) (2013) E15–E22. [188] S.P. Mayfield, S.E. Franklin, Expression of human antibodies in eukaryotic micro-
[176] E.A. Specht, P.S. Karunanithi, J.A. Gimpel, et al., Host Organisms: Algae, in: algae, Vaccine 23 (2005) 1828–1832.
C. Wittmann, J. Lio (Eds.), Industrial Biotechnology: Microorganisms, Wiley, [189] R. Boehm, Bioproduction of therapeutic proteins in the 21st century and the role
Weinheim, 2017, pp. 605–641. of plants and plant cells as production platforms, Ann. N. Y. Acad. Sci. 1102
[177] J. Zhou, H. Zhang, H. Meng, et al., Discovery of a super-strong promoter enables (2007) 121–134.
efficient production of heterologous proteins in cyanobacteria, Nat. Sci. Rep. 4 [190] S.S. Li, H.J. Tsai, Transgenic microalgae as a non-antibiotic bactericide producer
(2014) 4500. to defend against bacterial pathogen infection in the fish digestive tract, Fish
[178] S.A. Angermayr, K.J. Hellingwerf, On the use of metabolic control analysis in the Shellfish Immunol. 26 (2009) 316–325.
optimization of cyanobacterial biosolar cell factories, J. Phys. Chem. B 117 (2013) [191] J. Voigt, M. Fausel, P. Bohley, et al., Structure and expression of the ornithine
11169–11175. decarboxylase gene of Chlamydomonas reinhardtii, Microbiol. Res. 159 (2004)
[179] H. Cerutti, A.M. Johnson, E. Gillham, et al., A eubacterial gene conferring 403–417.
spectinomycin resistance on Chlamydomonas reinhardtii: integration into the [192] F. Hempel, J. Lau, A. Klingl, et al., Algae as protein factories: expression of a
nuclear genome and gene expression, Genetics 145 (1997) 97. human antibody and the respective antigen in the diatom Phaeodactylum
[180] B.L. Randolph-Anderson, J.E. Boynton, N.W. Gillham, et al., Further character- tricornutum, PLoS One 6 (2011) e28424.
ization of the respiratory deficient dum-1 mutation of Chlamydomonas reinhardtii [193] R. Radakovits, R.E. Jinkerson, S.I. Fuerstenberg, et al., Draft genome sequence and
and its use as a recipient for mitochondrial transformation, Mol. Gen. Genet. 236 genetic transformation of the oleaginous alga Nannochloropsis gaditana, Nat.
(1993) 235–244. Commun. 3 (2012) 686.
[181] C. Remacle, P. Cardol, N. Coosemans, et al., High-efficiency biolistic transforma- [194] F. Li, D. Gao, H. Hu, High-efficiency nuclear transformation of the oleaginous
tion of Chlamydomonas mitochondria can be used to insert mutations in complex I marine Nannochloropsis species using PCR product, Biosci. Biotechnol. Biochem.
genes, Proc. Natl. Acad. Sci. U. S. A. 103 (2006) 4771–4776. 78 (2014) 812–817.
[182] T. Yamasaki, S. Kurokawa, K.I. Watanabe, et al., Shared molecular characteristics [195] S. Sanchez, A.L. Demain, Enzymes and Bioconversions of Industrial,
of successfully transformed mitochondrial genomes in Chlamydomonas reinhardtii, Pharmaceutical, and Biotechnological Significance, Org. Process. Res. Dev. 15
Plant Mol. Biol. 58 (2005) 515–527. (2011) 224–230.
[183] R.A. Lewin, Studies on the flagella of algae. I. General observations on chlamy- [196] NAABB (National Alliance for Advanced Biofuels and Bioproducts), National
domonas moewusii Gerloff, Biol. Bull. (1952) 74–79. Alliance for Advanced Biofuels and Bioproducts - Final Report, US Department of
[184] K.L. Kindle, High-frequency nuclear transformation of Chlamydomonas reinhardtii, energy, 2014 Available at: http://energy.gov/eere/bioenergy/downloads/
Proc. Nat. Acad. Scie. 87 (3) (1990) 1228–1232. national-alliance-advanced-biofuels-and-bioproducts-synopsis-naabb-final ,
[185] A.N. Barry, S.R. Starkenburg, R.T. Sayre, Strategies for optimizing algal biology for (Accessed in: 02 Out 2015).
enhanced biomass production, Front. Energy. Res. 3 (2015) 1–5.

89

You might also like